Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.781
Filter
1.
Front Biosci (Landmark Ed) ; 29(5): 169, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38812304

ABSTRACT

Transforming growth factor-ß (TGF-ß) activated kinase 1 (TAK1), also named mitogen-activated protein kinase 7 (MAPK7), forms a pivotal signaling complex with TAK1-binding proteins (TAB1, TAB2, and TAB3), orchestrating critical biological processes, including immune responses, cell growth, apoptosis, and stress responses. Activation of TAK1 by stimuli, such as tumor necrosis factor α (TNFα), interleukin-1ß (IL-1ß), and Toll-like receptors (TLRs), underscores its central role in cellular signaling. Given the critical role of the TAK1-binding protein (TAK1-TAB) complex in cellular signaling and its impact on various biological processes, this review seeks to understand how ubiquitination thoroughly regulates the TAK1-TAB complex. This understanding is vital for developing targeted therapies for diseases where this signaling pathway is dysregulated. The exploration is significant as it unveils new insights into the activity, stability, and assembly of the complex, underscoring its therapeutic potential in disease modulation.


Subject(s)
Adaptor Proteins, Signal Transducing , MAP Kinase Kinase Kinases , Signal Transduction , Ubiquitination , Humans , MAP Kinase Kinase Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals
2.
Discov Med ; 36(184): 936-945, 2024 May.
Article in English | MEDLINE | ID: mdl-38798253

ABSTRACT

BACKGROUND: Inflammation is a key pathological process in bacterial meningitis, and the transforming growth factor-beta-activated kinase 1 (TAK1)/nuclear factor-kappa B (NF-κB) pathway is implicated in the activation of microglia and the production of inflammatory factors. Interleukin (IL)-10 is an anti-inflammatory cytokine acting in an autocrine fashion in macrophages to limit inflammatory responses by decreasing the production of pro-inflammatory cytokines. This paper investigates how IL-10 can inhibit microglia activation and reduce the inflammatory response of nervous system diseases. METHODS: This study used a pneumococcal-induced in Pneumococcal meningitis (PM) C57BL/6 mice and BV-2 cells model of microglial activation, assessing the effects of IL-10 on the TAK1/NF-κB pathway. The impact of IL-10 on microglial autophagy was investigated through western blot and immunofluorescence. The effects of IL-10 were evaluated by examining cellular activation markers and the activity of molecular signaling pathways (such as phosphorylation levels of TAK1 and NF-κB). RESULTS: Pneumococcus induced the activation of microglia and reduced IL-10. IL-10 inhibited the TAK1/NF-κB pathway, reducing the pneumococcal-induced inflammatory response in microglia. IL-10 ameliorated pneumococcal infection-induced microglial injury by inhibiting autophagy. Animal experiment results also showed that IL-10 inhibited inflammation and autophagy during Pneumococcal meningitis in mice. CONCLUSION: Our study demonstrates that IL-10 reduces the inflammatory response of microglia by inhibiting the TAK1/NF-κB pathway. Additionally, IL-10 ameliorates pneumococcal infection-induced microglial injury by inhibiting the process of autophagy. These results provide a new theoretical basis and offer new insights for developing strategies to treat bacterial meningitis.


Subject(s)
Interleukin-10 , MAP Kinase Kinase Kinases , Meningitis, Pneumococcal , Mice, Inbred C57BL , Microglia , NF-kappa B , Animals , Interleukin-10/metabolism , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Mice , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/immunology , Meningitis, Pneumococcal/pathology , NF-kappa B/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Inflammation/pathology , Autophagy/drug effects , Disease Models, Animal , Cell Line , Streptococcus pneumoniae
3.
Aging (Albany NY) ; 16(9): 7915-7927, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38728237

ABSTRACT

OBJECTIVE: This research aimed to explore IL-21/miR-361-5p/MAP3K9 expression in shoulder arthritis and identify its regulatory pathways. METHODS: We established a rat shoulder arthritis model, then quantified IL21 and miR-361-5p in synovial fluid using ELISA and monitored the arthritis development. Additionally, IL21's effect on miR-361-5p levels in cultured human chondrocytes (HC-a) was assessed. Chondrocyte cell cycle status and apoptosis were measured via flow cytometry. Interactions between miR-361-5p and MAP3K9 were confirmed through dual-luciferase reporting and bioinformatic scrutiny. Protein levels of MAP3K9, p-ERK1/2, p-NF-κB, MMP1, and MMP9 were analyzed by Western blots. RESULTS: IL21 levels were elevated, while miR-361-5p was reduced in the synovial fluid from arthritic rats compared to healthy rats. IL21 was shown to suppress miR-361-5p in chondrocytes leading to hindered cell proliferation and increased apoptosis. Western blots indicated that miR-361-5p curbed MAP3K9 expression, reducing MMP activity by attenuating the ERK1/2/NF-κB pathway in chondrocytes. CONCLUSION: IL21 upregulation and miR-361-5p downregulation characterize shoulder arthritis, resulting in MAP3K9 overexpression. This chain of molecular events boosts MMP expression in chondrocytes and exacerbates the condition's progression.


Subject(s)
Chondrocytes , MAP Kinase Kinase Kinases , MicroRNAs , Animals , Humans , Male , Rats , Apoptosis/genetics , Cell Proliferation/genetics , Chondrocytes/metabolism , Disease Progression , Interleukins/metabolism , Interleukins/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Rats, Sprague-Dawley
4.
Nat Commun ; 15(1): 4216, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760394

ABSTRACT

Antimicrobial peptides (AMPs), ancient scavengers of bacteria, are very poorly induced in macrophages infected by Mycobacterium tuberculosis (M. tuberculosis), but the underlying mechanism remains unknown. Here, we report that L-alanine interacts with PRSS1 and unfreezes the inhibitory effect of PRSS1 on the activation of NF-κB pathway to induce the expression of AMPs, but mycobacterial alanine dehydrogenase (Ald) Rv2780 hydrolyzes L-alanine and reduces the level of L-alanine in macrophages, thereby suppressing the expression of AMPs to facilitate survival of mycobacteria. Mechanistically, PRSS1 associates with TAK1 and disruptes the formation of TAK1/TAB1 complex to inhibit TAK1-mediated activation of NF-κB pathway, but interaction of L-alanine with PRSS1, disables PRSS1-mediated impairment on TAK1/TAB1 complex formation, thereby triggering the activation of NF-κB pathway to induce expression of AMPs. Moreover, deletion of antimicrobial peptide gene ß-defensin 4 (Defb4) impairs the virulence by Rv2780 during infection in mice. Both L-alanine and the Rv2780 inhibitor, GWP-042, exhibits excellent inhibitory activity against M. tuberculosis infection in vivo. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses its own alanine dehydrogenase to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.


Subject(s)
Alanine , Antimicrobial Peptides , Macrophages , Mycobacterium tuberculosis , NF-kappa B , Tuberculosis , Mycobacterium tuberculosis/pathogenicity , Mycobacterium tuberculosis/metabolism , Animals , Mice , NF-kappa B/metabolism , Humans , Macrophages/microbiology , Macrophages/metabolism , Macrophages/immunology , Alanine/metabolism , Antimicrobial Peptides/metabolism , Antimicrobial Peptides/genetics , Tuberculosis/microbiology , Tuberculosis/immunology , Alanine Dehydrogenase/metabolism , Alanine Dehydrogenase/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Signal Transduction , Mice, Inbred C57BL , RAW 264.7 Cells , Female
5.
Biol Direct ; 19(1): 34, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698487

ABSTRACT

BACKGROUND: LncRNA PCED1B-AS1 is abnormally expressed in multiple cancers and has been confirmed as an oncogene. Our study aimed to investigate the regulatory mechanism of lncRNA PCED1B-AS1 in gastric cancer. METHODS: TCGA database was used to analyze the abnormal expression of lncRNA PCED1B-AS1 in gastric cancer. By database prediction and mass spectrometric analysis, miR-3681-3p and MAP2K7 are potential downstream target molecules of lncRNA PCED1B-AS1 and verified by dual-luciferase report assay. RT-qPCR analysis and western blot were performed to detect the expressions of PCED1B-AS1 and MAP2K7 in gastric cancer cell lines and tissues. CCK-8 kit was applied to measure the cell viability. Wound healing and Transwell experiment were used to detect the migration and invasion. Western blot and immunohistochemical staining were performed to detect the expressions of EMT-related proteins in tissues. The changes of tumor proliferation were detected by xenograft experiment in nude mice. RESULTS: PCED1B-AS1 expression was higher but miR-3681-3 expression was lower in gastric cancer cell lines or tissues, compared to normal group. Function analysis verified PCED1B-AS1 promoted cell proliferation and inhibited cell apoptosis in gastric cancer cells in vitro and in vivo. LncRNA PCED1B-AS1 could bind directly to miR-3681-3p, and MAP2K7 was found to be a downstream target of miR-3681-3p. MiR-3681-3p mimics or si-MAP2K7 could partly reverse the effect of PCED1B-AS1 on gastric cancer cells. CONCLUSION: PCED1B-AS1 accelerated cell proliferation and inhibited cell apoptosis through sponging miR-3681-3p to upregulate MAP2K7 expression in gastric cancer, which indicated PCED1B-AS1/miR-3681-3p/MAP2K7 axis may serve as a potential therapeutic target for gastric cancer.


Subject(s)
Epithelial-Mesenchymal Transition , MAP Kinase Kinase Kinases , Mice, Nude , MicroRNAs , RNA, Long Noncoding , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor , Animals , Mice , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Cell Movement , Neoplasm Metastasis
6.
PLoS One ; 19(5): e0303556, 2024.
Article in English | MEDLINE | ID: mdl-38753858

ABSTRACT

Echinatin is an active ingredient in licorice, a traditional Chinese medicine used in the treatment of inflammatory disorders. However, the protective effect and underlying mechanism of echinatin against acute lung injury (ALI) is still unclear. Herein, we aimed to explore echinatin-mediated anti-inflammatory effects on lipopolysaccharide (LPS)-stimulated ALI and its molecular mechanisms in macrophages. In vitro, echinatin markedly decreased the levels of nitric oxide (NO) and prostaglandin E2 (PGE2) in LPS-stimulated murine MH-S alveolar macrophages and RAW264.7 macrophages by suppressing inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) expression. Furthermore, echinatin reduced LPS-induced mRNA expression and release of interleukin-1ß (IL-1ß) and IL-6 in RAW264.7 cells. Western blotting and CETSA showed that echinatin repressed LPS-induced activation of mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways through targeting transforming growth factor-beta-activated kinase 1 (TAK1). Furthermore, echinatin directly interacted with Kelch-like ECH-associated protein 1 (Keap1) and activated the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway to enhance heme oxygenase-1 (HO-1) expression. In vivo, echinatin ameliorated LPS-induced lung inflammatory injury, and reduced production of IL-1ß and IL-6. These findings demonstrated that echinatin exerted anti-inflammatory effects in vitro and in vivo, via blocking the TAK1-MAPK/NF-κB pathway and activating the Keap1-Nrf2-HO-1 pathway.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , MAP Kinase Kinase Kinases , Signal Transduction , Animals , Male , Mice , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/chemically induced , Anti-Inflammatory Agents/pharmacology , Heme Oxygenase-1/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Kelch-Like ECH-Associated Protein 1/metabolism , Macrophages/drug effects , Macrophages/metabolism , MAP Kinase Kinase Kinases/metabolism , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
7.
Science ; 384(6697): 785-792, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38753784

ABSTRACT

In response to excessive DNA damage, human cells can activate p53 to induce apoptosis. Cells lacking p53 can still undergo apoptosis upon DNA damage, yet the responsible pathways are unknown. We observed that p53-independent apoptosis in response to DNA damage coincided with translation inhibition, which was characterized by ribosome stalling on rare leucine-encoding UUA codons and globally curtailed translation initiation. A genetic screen identified the transfer RNAse SLFN11 and the kinase GCN2 as factors required for UUA stalling and global translation inhibition, respectively. Stalled ribosomes activated a ribotoxic stress signal conveyed by the ribosome sensor ZAKα to the apoptosis machinery. These results provide an explanation for the frequent inactivation of SLFN11 in chemotherapy-unresponsive tumors and highlight ribosome stalling as a signaling event affecting cell fate in response to DNA damage.


Subject(s)
Apoptosis , DNA Damage , Protein Biosynthesis , Ribosomes , Tumor Suppressor Protein p53 , Humans , Cell Line, Tumor , Codon/genetics , Leucine/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Ribosomes/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism
8.
Biochemistry ; 63(11): 1474-1492, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38743619

ABSTRACT

Allostery is a fundamental mechanism driving biomolecular processes that holds significant therapeutic concern. Our study rigorously investigates how two distinct machine-learning algorithms uniquely classify two already close-to-active DFG-in states of TAK1, differing just by the presence or absence of its allosteric activator TAB1, from an ensemble mixture of conformations (obtained from 2.4 µs molecular dynamics (MD) simulations). The novelty, however, lies in understanding the deeper algorithmic potentials to systematically derive a diverse set of differential residue connectivity features that reconstruct the essential mechanistic architecture for TAK1-TAB1 allostery in such a close-to-active biochemical scenario. While the recursive, random forest-based workflow displays the potential of conducting discretized, hierarchical derivation of allosteric features, a multilayer perceptron-based approach gains considerable efficacy in revealing fluid connected patterns of features when hybridized with mutual information scoring. Interestingly, both pipelines benchmark similar directions of functional conformational changes for TAK1's activation. The findings significantly advance the depth of mechanistic understanding by highlighting crucial activation signatures along a directed C-lobe → activation loop → ATP pocket channel of information flow, including (1) the αF-αE biterminal alignments and (2) the "catalytic" drift of the activation loop toward kinase active site. Besides, some novel allosteric hotspots (K253, Y206, N189, etc.) are further recognized as TAB1 sensors, transducers, and responders, including a benchmark E70 mutation site, precisely mapping the important structural segments for sequential allosteric execution. Hence, our work demonstrates how to navigate through greater structural depths and dimensions of dynamic allosteric machineries just by leveraging standard ML methods in suitable streamlined workflows adaptive to the specific system and objectives.


Subject(s)
MAP Kinase Kinase Kinases , Machine Learning , Molecular Dynamics Simulation , Allosteric Regulation , MAP Kinase Kinase Kinases/chemistry , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Humans , Protein Conformation , Workflow , Algorithms
9.
Toxicol Appl Pharmacol ; 487: 116954, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705402

ABSTRACT

Dual-specificity phosphatase 26 (DUSP26) acts as a pivotal player in the transduction of signalling cascades with its dephosphorylating activity. Currently, DUSP26 attracts extensive attention due to its particular function in several pathological conditions. However, whether DUSP26 plays a role in kidney ischaemia-reperfusion (IR) injury is unknown. Aims of the current work were to explore the relevance of DUSP26 in kidney IR damage. DUSP26 levels were found to be decreased in renal tubular epithelial cells following hypoxia-reoxygenation (HR) and kidney samples subjected to IR treatments. DUSP26-overexpressed renal tubular epithelial cells exhibited protection against HR-caused apoptosis and inflammation, while DUSP26-depleted renal tubular epithelial cells were more sensitive to HR damage. Upregulation of DUSP26 in rat kidneys by infecting adenovirus expressing DUSP26 markedly ameliorated kidney injury caused by IR, while also effectively reducing apoptosis and inflammation. The mechanistic studies showed that the activation of transforming growth factor-ß-activated kinase 1 (TAK1)-JNK/p38 MAPK, contributing to kidney injury under HR or IR conditions, was restrained by increasing DUSP26 expression. Pharmacological restraint of TAK1 markedly diminished DUSP26-depletion-exacebated effects on JNK/p38 activation and HR injury of renal tubular cells. The work reported a renal-protective function of DUSP26, which protects against IR-related kidney damage via the intervention effects on the TAK1-JNK/p38 axis. The findings laid a foundation for understanding the molecular pathogenesis of kidney IR injury and provide a prospective target for treating this condition.


Subject(s)
Apoptosis , Epithelial Cells , Kidney Tubules , MAP Kinase Kinase Kinases , Rats, Sprague-Dawley , Reperfusion Injury , p38 Mitogen-Activated Protein Kinases , Animals , Reperfusion Injury/pathology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Male , Kidney Tubules/pathology , Kidney Tubules/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/metabolism , Dual-Specificity Phosphatases/metabolism , Dual-Specificity Phosphatases/genetics , Cell Line , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Inflammation/pathology , Inflammation/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/genetics , Signal Transduction/physiology
10.
Int J Biol Sci ; 20(7): 2403-2421, 2024.
Article in English | MEDLINE | ID: mdl-38725848

ABSTRACT

Ciliogenesis-associated kinase 1 (CILK1) plays a key role in the ciliogenesis and ciliopathies. It remains totally unclear whether CILK1 is involved in tumor progression and therapy resistance. Here, we report that the aberrant high-expression of CILK1 in breast cancer is required for tumor cell proliferation and chemoresistance. Two compounds, CILK1-C30 and CILK1-C28, were identified with selective inhibitory effects towards the Tyr-159/Thr-157 dual-phosphorylation of CILK1, pharmacological inhibition of CILK1 significantly suppressed tumor cell proliferation and overcame chemoresistance in multiple experimental models. Large-scale screen of CILK1 substrates confirmed that the kinase directly phosphorylates ERK1, which is responsible for CILK1-mediated oncogenic function. CILK1 is also indicated to be responsible for the chemoresistance of small-cell lung cancer cells. Our data highlight the importance of CILK1 in cancer, implicating that targeting CILK1/ERK1 might offer therapeutic benefit to cancer patients.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Animals , Female , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , MAP Kinase Kinase Kinases , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Proto-Oncogene Proteins
11.
Neoplasia ; 53: 101003, 2024 07.
Article in English | MEDLINE | ID: mdl-38759377

ABSTRACT

Dynamic changes in the endoplasmic reticulum (ER) morphology are central to maintaining cellular homeostasis. Microtubules (MT) facilitate the continuous remodeling of the ER network into sheets and tubules by coordinating with many ER-shaping protein complexes, although how this process is controlled by extracellular signals remains unknown. Here we report that TAK1, a kinase responsive to various growth factors and cytokines including TGF-ß and TNF-α, triggers ER tubulation by activating αTAT1, an MT-acetylating enzyme that enhances ER-sliding. We show that this TAK1/αTAT1-dependent ER remodeling promotes cell survival by actively downregulating BOK, an ER membrane-associated proapoptotic effector. While BOK is normally protected from degradation when complexed with IP3R, it is rapidly degraded upon their dissociation during the ER sheets-to-tubules conversion. These findings demonstrate a distinct mechanism of ligand-induced ER remodeling and suggest that the TAK1/αTAT1 pathway may be a key target in ER stress and dysfunction.


Subject(s)
Endoplasmic Reticulum , MAP Kinase Kinase Kinases , Microtubules , Signal Transduction , Microtubules/metabolism , Endoplasmic Reticulum/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Acetylation , Animals , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Acetyltransferases/metabolism , Acetyltransferases/genetics , Endoplasmic Reticulum Stress , Mice , Microtubule Proteins
12.
BMC Biol ; 22(1): 122, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38807188

ABSTRACT

BACKGROUND: The innate immune system serves as the first line of host defense. Transforming growth factor-ß-activated kinase 1 (TAK1) is a key regulator of innate immunity, cell survival, and cellular homeostasis. Because of its importance in immunity, several pathogens have evolved to carry TAK1 inhibitors. In response, hosts have evolved to sense TAK1 inhibition and induce robust lytic cell death, PANoptosis, mediated by the RIPK1-PANoptosome. PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. While PANoptosis can be beneficial to clear pathogens, excess activation is linked to pathology. Therefore, understanding the molecular mechanisms regulating TAK1 inhibitor (TAK1i)-induced PANoptosis is central to our understanding of RIPK1 in health and disease. RESULTS: In this study, by analyzing results from a cell death-based CRISPR screen, we identified protein phosphatase 6 (PP6) holoenzyme components as regulators of TAK1i-induced PANoptosis. Loss of the PP6 enzymatic component, PPP6C, significantly reduced TAK1i-induced PANoptosis. Additionally, the PP6 regulatory subunits PPP6R1, PPP6R2, and PPP6R3 had redundant roles in regulating TAK1i-induced PANoptosis, and their combined depletion was required to block TAK1i-induced cell death. Mechanistically, PPP6C and its regulatory subunits promoted the pro-death S166 auto-phosphorylation of RIPK1 and led to a reduction in the pro-survival S321 phosphorylation. CONCLUSIONS: Overall, our findings demonstrate a key requirement for the phosphatase PP6 complex in the activation of TAK1i-induced, RIPK1-dependent PANoptosis, suggesting this complex could be therapeutically targeted in inflammatory conditions.


Subject(s)
Phosphoprotein Phosphatases , Receptor-Interacting Protein Serine-Threonine Kinases , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Humans , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/genetics , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Necroptosis , Immunity, Innate
13.
Nat Commun ; 15(1): 4340, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773142

ABSTRACT

Macrophage-orchestrated inflammation contributes to multiple diseases including sepsis. However, the underlying mechanisms remain to be defined clearly. Here, we show that macrophage TP53-induced glycolysis and apoptosis regulator (TIGAR) is up-regulated in murine sepsis models. When myeloid Tigar is ablated, sepsis induced by either lipopolysaccharide treatment or cecal ligation puncture in male mice is attenuated via inflammation inhibition. Mechanistic characterizations indicate that TIGAR directly binds to transforming growth factor ß-activated kinase (TAK1) and promotes tumor necrosis factor receptor-associated factor 6-mediated ubiquitination and auto-phosphorylation of TAK1, in which residues 152-161 of TIGAR constitute crucial motif independent of its phosphatase activity. Interference with the binding of TIGAR to TAK1 by 5Z-7-oxozeaenol exhibits therapeutic effects in male murine model of sepsis. These findings demonstrate a non-canonical function of macrophage TIGAR in promoting inflammation, and confer a potential therapeutic target for sepsis by disruption of TIGAR-TAK1 interaction.


Subject(s)
Apoptosis Regulatory Proteins , Disease Models, Animal , Lipopolysaccharides , MAP Kinase Kinase Kinases , Macrophages , Sepsis , Animals , Sepsis/immunology , Sepsis/drug therapy , Sepsis/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Male , Mice , Macrophages/metabolism , Macrophages/immunology , Macrophages/drug effects , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Inbred C57BL , Phosphorylation , Humans , Ubiquitination , Zearalenone/analogs & derivatives , Zearalenone/pharmacology , Zearalenone/administration & dosage , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Inflammation/metabolism , Inflammation/pathology , Phosphoric Monoester Hydrolases/metabolism , Mice, Knockout , Lactones , Resorcinols
14.
Front Immunol ; 15: 1386780, 2024.
Article in English | MEDLINE | ID: mdl-38756773

ABSTRACT

Introduction: Intracerebral hemorrhage (ICH) often triggers oxidative stress through reactive oxygen species (ROS). Transforming growth factor-ß-activated kinase 1 (TAK1) plays a pivotal role in regulating oxidative stress and inflammation across various diseases. 5Z-7-Oxozeaenol (OZ), a specific inhibitor of TAK1, has exhibited therapeutic effects in various conditions. However, the impact of OZ following ICH and its underlying molecular mechanisms remain elusive. This study aimed to explore the possible role of OZ in ICH and its underlying mechanisms by inhibiting oxidative stress-mediated pyroptosis. Methods: Adult male Sprague-Dawley rats were subjected to an ICH model, followed by treatment with OZ. Neurobehavioral function, blood-brain barrier integrity, neuronal pyroptosis, and oxidative stress markers were assessed using various techniques including behavioral tests, immunofluorescence staining, western blotting, transmission electron microscopy, and biochemical assays. Results: Our study revealed that OZ administration significantly inhibited phosphorylated TAK1 expression post-ICH. Furthermore, TAK1 blockade by OZ attenuated blood-brain barrier (BBB) disruption, neuroinflammation, and oxidative damage while enhancing neurobehavioral function. Mechanistically, OZ administration markedly reduced ROS production and oxidative stress by facilitating nuclear factor-erythroid 2-related factor 2 (NRF2) nuclear translocation. This was accompanied by a subsequent suppression of the NOD-like receptor protein 3 (NLRP3) activation-mediated inflammatory cascade and neuronal pyroptosis. Discussion: Our findings highlight that OZ alleviates brain injury and oxidative stress-mediated pyroptosis via the NRF2 pathway. Inhibition of TAK1 emerges as a promising approach for managing ICH.


Subject(s)
Cerebral Hemorrhage , MAP Kinase Kinase Kinases , NF-E2-Related Factor 2 , Neurons , Oxidative Stress , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Animals , Pyroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Male , Rats , Signal Transduction/drug effects , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Disease Models, Animal , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Injuries/drug therapy , Reactive Oxygen Species/metabolism , Lactones , Resorcinols , Zearalenone/administration & dosage
15.
Brain Behav ; 14(4): e3487, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38648385

ABSTRACT

INTRODUCTION: Demyelination is a key factor in axonal degeneration and neural loss, leading to disability in multiple sclerosis (MS) patients. Transforming growth factor beta activated kinase 1 (TAK1) is a critical molecule involved in immune and inflammatory signaling pathways. Knockout of microglia TAK1 can inhibit autoimmune inflammation of the brain and spinal cord and improve the outcome of MS. However, it is unclear whether inhibiting TAK1 can alleviate demyelination. METHODS: Eight-week-old male c57bl/6j mice were randomly divided into five groups: (a) the control group, (b) the group treated with cuprizone (CPZ) only, (c) the group treated with 5Z-7-Oxozaenol (OZ) only, and (d) the group treated with both cuprizone and 15 µg/30 µg OZ. Demyelination in the mice of this study was induced by administration of CPZ (ig) at a daily dose of 400 mg/kg for consecutive 5 weeks. OZ was intraperitoneally administered at mentioned doses twice a week, starting from week 3 after beginning cuprizone treatment. Histology, rotarod test, grasping test, pole test, Western blot, RT-PCR, and ELISA were used to evaluate corpus callosum demyelination, behavioral impairment, oligodendrocyte differentiation, TAK1 signaling pathway expression, microglia, and related cytokines. RESULTS: Our results demonstrated that OZ protected against myelin loss and behavior impairment caused by CPZ. Additionally, OZ rescued the loss of oligodendrocytes in CPZ-induced mice. OZ inhibited the activation of JNK, p65, and p38 pathways, transformed M1 polarized microglia into M2 phenotype, and increased brain-derived neurotrophic factor (BDNF) expression to attenuate demyelination in CPZ-treated mice. Furthermore, OZ reduced the expression of proinflammatory cytokines and increases anti-inflammatory cytokines in CPZ-treated mice. CONCLUSION: These findings suggest that inhibiting TAK1 may be an effective approach for treating demyelinating diseases.


Subject(s)
Cuprizone , Demyelinating Diseases , Lactones , Mice, Inbred C57BL , Microglia , Resorcinols , Zearalenone/administration & dosage , Animals , Cuprizone/pharmacology , Microglia/drug effects , Microglia/metabolism , Demyelinating Diseases/drug therapy , Demyelinating Diseases/chemically induced , Mice , Male , MAP Kinase Kinase Kinases/metabolism , Zearalenone/pharmacology , Zearalenone/analogs & derivatives , Cell Polarity/drug effects , Corpus Callosum/drug effects , Corpus Callosum/pathology , Corpus Callosum/metabolism , Disease Models, Animal
16.
Cell Death Dis ; 15(4): 273, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38632238

ABSTRACT

Poor survival and lack of treatment response in glioblastoma (GBM) is attributed to the persistence of glioma stem cells (GSCs). To identify novel therapeutic approaches, we performed CRISPR/Cas9 knockout screens and discovered TGFß activated kinase (TAK1) as a selective survival factor in a significant fraction of GSCs. Loss of TAK1 kinase activity results in RIPK1-dependent apoptosis via Caspase-8/FADD complex activation, dependent on autocrine TNFα ligand production and constitutive TNFR signaling. We identify a transcriptional signature associated with immune activation and the mesenchymal GBM subtype to be a characteristic of cancer cells sensitive to TAK1 perturbation and employ this signature to accurately predict sensitivity to the TAK1 kinase inhibitor HS-276. In addition, exposure to pro-inflammatory cytokines IFNγ and TNFα can sensitize resistant GSCs to TAK1 inhibition. Our findings reveal dependency on TAK1 kinase activity as a novel vulnerability in immune-activated cancers, including mesenchymal GBMs that can be exploited therapeutically.


Subject(s)
Apoptosis , Glioblastoma , Glioma , Receptor-Interacting Protein Serine-Threonine Kinases , Humans , Apoptosis/genetics , Cytokines , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/genetics , Glioma/immunology , Glioma/metabolism , Glioma/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha
17.
PeerJ ; 12: e16967, 2024.
Article in English | MEDLINE | ID: mdl-38680890

ABSTRACT

Background: Ovarian cancer (OC) is the most lethal malignancy in women owing to its diagnosis only at the advanced stage. Elucidation of its molecular pathogenesis may help identify new tumor markers and targets for therapy. Circular RNAs (circRNAs) are stable, conserved, and functional biomolecules that can be used as effective biomarkers for various cancers. Methods: In this study, a potential circRNA related to early diagnosis of OC, circMAN1A2, was analyzed. Overexpression/knockdown of circMAN1A2 in OC cells was used to decipher its effects on cell proliferation with a Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine (EdU), cell cycle, clone formation, and wound healing assay. RNA pull-down and Dual luciferase assay were used to explain the underlying mechanism by which circMAN1A2 regulates OC cell proliferation. In vivo, the effect of circMAN1A2 in OC was evaluated using nude mouse xenograft experiments. Results: CircMAN1A2 was highly expressed in OC and promoted proliferation, clone formation, and tumorigenicity of OC cells. In addition, we found that circMAN1A2 acted as a sponge for microRNA (miR)-135a-3p; miR-135a-3p directly targeted the 3' untranslated region of interleukin 1 receptor accessory protein (IL1RAP) in OC cells, thereby regulating the phosphorylation of transforming growth factor-beta activated kinase 1 (TAK1), which resulted in promotion of OC cell growth. Conclusions: CircMAN1A2 promotes OC cell proliferation by inhibiting the miR-135a-3p/IL1RAP/TAK1 axis. In conclusion, circMAN1A2 may be a biomarker for early detection of OC and a target for subsequent therapy.


Subject(s)
Mannosidases , MicroRNAs , Ovarian Neoplasms , RNA, Circular , Signal Transduction , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Signal Transduction/genetics , Mannosidases/genetics
18.
Adv Sci (Weinh) ; 11(21): e2309002, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38569496

ABSTRACT

Preeclampsia (PE) is considered as a disease of placental origin. However, the specific mechanism of placental abnormalities remains elusive. This study identified thrombospondin-1 (THBS1) is downregulated in preeclamptic placentae and negatively correlated with blood pressure. Functional studies show that THBS1 knockdown inhibits proliferation, migration, and invasion and increases the cycle arrest and apoptosis rate of HTR8/SVneo cells. Importantly, THBS1 silencing induces necroptosis in HTR8/SVneo cells, accompanied by the release of damage-associated molecular patterns (DAMPs). Necroptosis inhibitors necrostatin-1 and GSK'872 restore the trophoblast survival while pan-caspase inhibitor Z-VAD-FMK has no effect. Mechanistically, the results show that THBS1 interacts with transforming growth factor B-activated kinase 1 (TAK1), which is a central modulator of necroptosis quiescence and affects its stability. Moreover, THBS1 silencing up-regulates the expression of neuronal precursor cell-expressed developmentally down-regulated 4 (NEDD4), which acts as an E3 ligase of TAK1 and catalyzes K48-linked ubiquitination of TAK1 in HTR8/SVneo cells. Besides, THBS1 attenuates PE phenotypes and improves the placental necroptosis in vivo. Taken together, the down-regulation of THBS1 destabilizes TAK1 by activating NEDD4-mediated, K48-linked TAK1 ubiquitination and promotes necroptosis and DAMPs release in trophoblast cells, thus participating in the pathogenesis of PE.


Subject(s)
MAP Kinase Kinase Kinases , Necroptosis , Nedd4 Ubiquitin Protein Ligases , Pre-Eclampsia , Thrombospondin 1 , Trophoblasts , Ubiquitination , Humans , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Female , Pregnancy , Trophoblasts/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , Necroptosis/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Thrombospondin 1/metabolism , Thrombospondin 1/genetics , Adult , Placenta/metabolism
19.
PLoS One ; 19(4): e0300539, 2024.
Article in English | MEDLINE | ID: mdl-38574058

ABSTRACT

Genetic and pharmacological perturbation of the cytoskeleton enhances the regenerative potential of neurons. This response requires Dual-leucine Zipper Kinase (DLK), a neuronal stress sensor that is a central regulator of axon regeneration and degeneration. The damage and repair aspects of this response are reminiscent of other cellular homeostatic systems, suggesting that a cytoskeletal homeostatic response exists. In this study, we propose a framework for understanding DLK mediated neuronal cytoskeletal homeostasis. We demonstrate that low dose nocodazole treatment activates DLK signaling. Activation of DLK signaling results in a DLK-dependent transcriptional signature, which we identify through RNA-seq. This signature includes genes likely to attenuate DLK signaling while simultaneously inducing actin regulating genes. We identify alterations to the cytoskeleton including actin-based morphological changes to the axon. These results are consistent with the model that cytoskeletal disruption in the neuron induces a DLK-dependent homeostatic mechanism, which we term the Cytoskeletal Stress Response (CSR) pathway.


Subject(s)
Actins , Axons , Axons/metabolism , Nocodazole/pharmacology , Actins/metabolism , Leucine Zippers , Nerve Regeneration/physiology , Cytoskeleton/metabolism , Homeostasis , MAP Kinase Kinase Kinases/genetics
20.
Exp Hematol ; 133: 104205, 2024 May.
Article in English | MEDLINE | ID: mdl-38490577

ABSTRACT

Protein phosphatase 6 (PP6) is a serine/threonine (Ser/Thr) protein phosphatase, and its catalytic subunit is Ppp6c. PP6 forms the PP2A subfamily with PP2A and PP4. The diverse phenotypes observed following small interfering RNA (siRNA)-based knockdown of Ppp6c in cultured mammalian cells suggest that PP6 plays roles in cell growth and DNA repair. There is also evidence that PP6 regulates nuclear factor kappa B (NF-κB) signaling and mitogen-activated protein kinases and inactivates transforming growth factor-ß-activated kinase 1 (TAK1). Loss of Ppp6c causes several abnormalities, including those of T cell and regulatory T cell function, neurogenesis, oogenesis, and spermatogenesis. PP2A has been reported to play an important role in erythropoiesis. However, the roles of PP6 in other hematopoietic cells have not been investigated. We generated Ppp6cfl/fl;Tie2-Cre (Ppp6cTKO) mice, in which Ppp6c was specifically deleted in hematopoietic and vascular endothelial cells. Ppp6cTKO mice displayed embryonic lethality. Ppp6c deficiency increased the number of dead cells and decreased the percentages of erythroid and monocytic cells during fetal hematopoiesis. By contrast, the number of Lin-Sca-1+c-Kit+ cells, which give rise to all hematopoietic cells, was slightly increased, but their colony-forming cell activity was markedly decreased. Ppp6c deficiency also increased phosphorylation of extracellular signal-regulated kinase 1/2 and c-Jun amino (N)-terminal kinase in fetal liver hematopoietic cells.


Subject(s)
Hematopoiesis , Phosphoprotein Phosphatases , Animals , Female , Mice , Embryo Loss/genetics , Embryo Loss/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hematopoietic Stem Cells/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoprotein Phosphatases/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...