Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32.265
1.
BMC Cancer ; 24(1): 520, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658865

Acute myeloid leukaemia (AML) is a fatal haematopoietic malignancy and is treated with the conventional combination of cytarabine (Ara-C) and daunorubicin (Dau). The survival rate of AML patients is lower due to the cardiotoxicity of daunorubicin. Clinically, homoharringtonine (HHT) plus Ara-C has been reported to be equally effective as Dau plus Ara-C in some types of AML patients with less toxic effects. We utilized the clinical use of homoharringtonine in combination with Ara-C to test its combination mechanism. We found that the insensitivity of AML cells to cytarabine-induced apoptosis is associated with increased Mcl-1 stability and p38 inactivation. HHT downregulates Mcl-1, phosphorylates H2AX and induces apoptosis by activating p38 MAPK. Inactivation of p38 through inhibitors and siRNA blocks apoptosis, H2AX phosphorylation and Mcl-1 reduction. HHT enhances Ara-C activation of the p38 MAPK signalling pathway, overcoming Ara-C tolerance to cell apoptosis by regulating the p38/H2AX/Mcl-1 axis. The optimal ratio of HHT to Ara-C for synergistic lethality in AML cells is 1:4 (M/M). HHT synergistically induces apoptosis in combination with Ara-C in vitro and prolongs the survival of xenografts. We provide a new mechanism for AML treatment by regulating the p38 MAPK/H2AX/Mcl-1 axis to improve cytarabine therapy.


Apoptosis , Cytarabine , Histones , Homoharringtonine , Leukemia, Myeloid, Acute , Myeloid Cell Leukemia Sequence 1 Protein , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases , Humans , Homoharringtonine/pharmacology , Cytarabine/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Apoptosis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Mice , Histones/metabolism , Cell Line, Tumor , Drug Synergism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Phosphorylation/drug effects , Female
2.
Neuromolecular Med ; 26(1): 15, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38653878

Lycium barbarum polysaccharide (LBP) have a certain curative effect on hypoglycemic and neuroprotective effects, but the specific mechanism is unclear and needs to be further explored. This study aimed to clarify the mechanisms of LBP in the treatment of ICV-STZ mice model of AD from the perspectives of insulin resistance, IRS1/PI3K/AKT signaling pathway, and synaptic protein expression. We used male C57BL/6J mice injected with STZ (3 mg/kg) in the lateral ventricle as an AD model. After treatment with LBP, the learning and memory abilities of ICV-STZ mice were enhanced, and the pathological changes in brain tissue were alleviated. LBP can regulate the expression of proteins related to the IRS1/PI3K/AKT signaling pathway and thereby reducing Aß deposition and tau protein phosphorylation in the brain of ICV-STZ mice. In addition, LBP also can up-regulate the expression of synaptic proteins. The results indicated that LBP played a neuroprotective role by regulating the IRS1/PI3K/AKT pathway, inhibiting tau protein hyperphosphorylation and improving the expression levels of synapse-related proteins.


Alzheimer Disease , Disease Models, Animal , Drugs, Chinese Herbal , Insulin Receptor Substrate Proteins , Mice, Inbred C57BL , Neuronal Plasticity , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , tau Proteins , Animals , Insulin Receptor Substrate Proteins/metabolism , Male , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Mice , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Neuronal Plasticity/drug effects , tau Proteins/metabolism , Streptozocin , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Cognition/drug effects , Amyloid beta-Peptides/metabolism , Phosphorylation/drug effects , Synapses/drug effects , Insulin Resistance , Brain/drug effects , Brain/metabolism , Brain/pathology
3.
Dis Model Mech ; 17(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38655653

Steroid myopathy is a clinically challenging condition exacerbated by prolonged corticosteroid use or adrenal tumors. In this study, we engineered a functional three-dimensional (3D) in vitro skeletal muscle model to investigate steroid myopathy. By subjecting our bioengineered muscle tissues to dexamethasone treatment, we reproduced the molecular and functional aspects of this disease. Dexamethasone caused a substantial reduction in muscle force, myotube diameter and induced fatigue. We observed nuclear translocation of the glucocorticoid receptor (GCR) and activation of the ubiquitin-proteasome system within our model, suggesting their coordinated role in muscle atrophy. We then examined the therapeutic potential of taurine in our 3D model for steroid myopathy. Our findings revealed an upregulation of phosphorylated AKT by taurine, effectively countering the hyperactivation of the ubiquitin-proteasomal pathway. Importantly, we demonstrate that discontinuing corticosteroid treatment was insufficient to restore muscle mass and function. Taurine treatment, when administered concurrently with corticosteroids, notably enhanced contractile strength and protein turnover by upregulating the AKT-mTOR axis. Our model not only identifies a promising therapeutic target, but also suggests combinatorial treatment that may benefit individuals undergoing corticosteroid treatment or those diagnosed with adrenal tumors.


Dexamethasone , Models, Biological , Muscle Contraction , Muscular Diseases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Taurine , Proto-Oncogene Proteins c-akt/metabolism , Humans , Taurine/pharmacology , TOR Serine-Threonine Kinases/metabolism , Muscle Contraction/drug effects , Dexamethasone/pharmacology , Muscular Diseases/pathology , Muscular Diseases/drug therapy , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Muscle Strength/drug effects , Proteasome Endopeptidase Complex/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Organ Size/drug effects , Phosphorylation/drug effects , Adrenal Cortex Hormones/pharmacology , Ubiquitin/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Steroids/pharmacology
4.
J Microbiol Biotechnol ; 34(4): 812-827, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38480001

Phloroglucinol (PG) is one of the abundant isomeric benzenetriols in brown algae. Due to its polyphenolic structure, PG exhibits various biological activities. However, the impact of PG on anagen signaling and oxidative stress in human dermal papilla cells (HDPCs) is unknown. In this study, we investigated the therapeutic potential of PG for improving hair loss. A non-cytotoxic concentration of PG increased anagen-inductive genes and transcriptional activities of ß-Catenin. Since several anagen-inductive genes are regulated by ß-Catenin, further experiments were performed to elucidate the molecular mechanism by which PG upregulates anagen signaling. Various biochemical analyses revealed that PG upregulated ß-Catenin signaling without affecting the expression of Wnt. In particular, PG elevated the phosphorylation of protein kinase B (AKT), leading to an increase in the inhibitory phosphorylation of glycogen synthase kinase 3 beta (GSK3ß) at serine 9. Treatment with the selective phosphoinositide 3-kinase/AKT inhibitor, LY294002, restored the increased AKT/GSK3ß/ß-Catenin signaling and anagen-inductive proteins induced by PG. Moreover, conditioned medium from PG-treated HDPCs promoted the proliferation and migration of human epidermal keratinocytes via the AKT signaling pathway. Subsequently, we assessed the antioxidant activities of PG. PG ameliorated the elevated oxidative stress markers and improved the decreased anagen signaling in hydrogen peroxide (H2O2)-induced HDPCs. The senescence-associated ß-galactosidase staining assay also demonstrated that the antioxidant abilities of PG effectively mitigated H2O2-induced senescence. Overall, these results indicate that PG potentially enhances anagen signaling and improves oxidative stress-induced cellular damage in HDPCs. Therefore, PG can be employed as a novel therapeutic component to ameliorate hair loss symptoms.


Glycogen Synthase Kinase 3 beta , Hydrogen Peroxide , Oxidative Stress , Phloroglucinol , Proto-Oncogene Proteins c-akt , Signal Transduction , beta Catenin , Humans , Phloroglucinol/pharmacology , Phloroglucinol/analogs & derivatives , Oxidative Stress/drug effects , Hydrogen Peroxide/metabolism , Signal Transduction/drug effects , beta Catenin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Phosphorylation/drug effects , Hair Follicle/drug effects , Hair Follicle/metabolism , Hair Follicle/cytology , Dermis/cytology , Dermis/metabolism , Dermis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Alopecia/drug therapy , Alopecia/metabolism
5.
Pflugers Arch ; 476(5): 779-795, 2024 May.
Article En | MEDLINE | ID: mdl-38536493

The American Diabetes Association guidelines (2021) confirmed the importance of raising public awareness of diabetes-induced cognitive impairment, highlighting the links between poor glycemic control and cognitive impairment. The characteristic brain lesions of cognitive dysfunction are neurofibrillary tangles (NFT) and senile plaques formed of amyloid-ß deposition, glycogen synthase kinase 3 beta (GSK3ß), and highly homologous kinase tau tubulin kinase 1 (TTBK1) can phosphorylate Tau proteins at different sites, overexpression of these enzymes produces extensive phosphorylation of Tau proteins making them insoluble and enhance NFT formation, which impairs cognitive functions. The current study aimed to investigate the potential contribution of liraglutide and pramlintide in the prevention of diabetes-induced cognitive dysfunction and their effect on the PI3K/AKT/GSK-3ß/TTBK1 pathway in type 2 diabetic (T2D) rat model. T2D was induced by administration of a high-fat diet for 10 weeks, then injection of a single dose of streptozotocin (STZ); treatment was started with either pramlintide (200 µg/kg/day sc) or liraglutide (0.6 mg/kg/day sc) for 6 weeks in addition to the HFD. At the end of the study, cognitive functions were assessed by novel object recognition and T-maze tests. Then, rats were sacrificed for biochemical and histological assessment of the hippocampal tissue. Both pramlintide and liraglutide treatment revealed equally adequate control of diabetes, prevented the decline in memory function, and increased PI3K/AKT expression while decreasing GSK-3ß/TTBK1 expression; however, liraglutide significantly decreased the number of Tau positive cells better than pramlintide did. This study confirmed that pramlintide and liraglutide are promising antidiabetic medications that could prevent associated cognitive disorders in different mechanisms.


Cognitive Dysfunction , Diet, High-Fat , Glycogen Synthase Kinase 3 beta , Liraglutide , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , tau Proteins , Animals , tau Proteins/metabolism , Rats , Glycogen Synthase Kinase 3 beta/metabolism , Liraglutide/pharmacology , Liraglutide/therapeutic use , Diet, High-Fat/adverse effects , Male , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/drug therapy , Islet Amyloid Polypeptide/metabolism , Islet Amyloid Polypeptide/pharmacology , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Rats, Sprague-Dawley , Streptozocin , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy
6.
Cell Death Differ ; 31(4): 405-416, 2024 Apr.
Article En | MEDLINE | ID: mdl-38538744

BH3 mimetics, including the BCL2/BCLXL/BCLw inhibitor navitoclax and MCL1 inhibitors S64315 and tapotoclax, have undergone clinical testing for a variety of neoplasms. Because of toxicities, including thrombocytopenia after BCLXL inhibition as well as hematopoietic, hepatic and possible cardiac toxicities after MCL1 inhibition, there is substantial interest in finding agents that can safely sensitize neoplastic cells to these BH3 mimetics. Building on the observation that BH3 mimetic monotherapy induces AMP kinase (AMPK) activation in multiple acute leukemia cell lines, we report that the AMPK inhibitors (AMPKis) dorsomorphin and BAY-3827 sensitize these cells to navitoclax or MCL1 inhibitors. Cell fractionation and phosphoproteomic analyses suggest that sensitization by dorsomorphin involves dephosphorylation of the proapoptotic BCL2 family member BAD at Ser75 and Ser99, leading BAD to translocate to mitochondria and inhibit BCLXL. Consistent with these results, BAD knockout or mutation to BAD S75E/S99E abolishes the sensitizing effects of dorsomorphin. Conversely, dorsomorphin synergizes with navitoclax or the MCL1 inhibitor S63845 to induce cell death in primary acute leukemia samples ex vivo and increases the antitumor effects of navitoclax or S63845 in several xenograft models in vivo with little or no increase in toxicity in normal tissues. These results suggest that AMPK inhibition can sensitize acute leukemia to multiple BH3 mimetics, potentially allowing administration of lower doses while inducing similar antineoplastic effects.


AMP-Activated Protein Kinases , Aniline Compounds , Myeloid Cell Leukemia Sequence 1 Protein , Pyrimidines , Sulfonamides , bcl-X Protein , Humans , Animals , Aniline Compounds/pharmacology , Sulfonamides/pharmacology , AMP-Activated Protein Kinases/metabolism , Mice , bcl-X Protein/metabolism , bcl-X Protein/antagonists & inhibitors , Cell Line, Tumor , Pyrimidines/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Pyrazoles/pharmacology , bcl-Associated Death Protein/metabolism , Apoptosis/drug effects , Cell Death/drug effects , Leukemia/drug therapy , Leukemia/pathology , Leukemia/metabolism , Phosphorylation/drug effects , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Drug Synergism
7.
FEBS Lett ; 598(8): 945-955, 2024 Apr.
Article En | MEDLINE | ID: mdl-38472156

TG-interacting factor 1 (TGIF1) contributes to the differentiation of murine white preadipocyte and human adipose tissue-derived stem cells; however, its regulation is not well elucidated. Insulin is a component of the adipogenic cocktail that induces ERK signaling. TGIF1 phosphorylation and sustained stability in response to insulin were reduced through the use of specific MEK inhibitor U0126. Mutagenesis at T235 or T239 residue of TGIF1 in preadipocytes led to dephosphorylation of TGIF1. The reduced TGIF1 stability resulted in an increase in p27kip1 expression, a decrease in phosphorylated Rb expression and cellular proliferation, and a reduced accumulation of lipids compared to the TGIF1-overexpressed cells. These findings highlight that insulin/ERK-driven phosphorylation of the T235 or T239 residue at TGIF1 is crucial for adipocyte differentiation.


3T3-L1 Cells , Adipocytes , Adipogenesis , Cell Differentiation , Homeodomain Proteins , Insulin , Animals , Mice , Phosphorylation/drug effects , Insulin/metabolism , Adipocytes/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Humans , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cell Proliferation/drug effects , Butadienes/pharmacology
8.
Mycotoxin Res ; 40(2): 223-234, 2024 May.
Article En | MEDLINE | ID: mdl-38319535

Mycotoxins have been shown to activate multiple mechanisms that may potentially lead to the progression of Alzheimer's disease (AD). Overexpression/aberrant cleavage of amyloid precursor protein (APP) and hyperphosphorylation of tau (P-tau) is hallmark pathologies of AD. Recent advances suggest that the neurotoxic effects of mycotoxins involve c-Jun N-terminal kinase (JNK) and hypoxia-inducible factor-1α (HIF-1α) signaling, which are closely linked to the pathogenesis of AD. Due to the high toxicity and broad contamination of T-2 toxin, we assessed how T-2 toxin exposure alters APP and P-tau formation in BV2 cells and determined the underlying roles of HIF-1α and JNK signaling. The findings revealed that T-2 toxin stimulated the expression of HIF-1α and hypoxic stress factors in addition to increasing the expression of APP and P-tau. Additionally, HIF-1α acted as a "brake" on the induction of APP and P-tau expression by negatively regulating these proteins. Notably, T-2 toxin activated JNK signaling, which broke this "brake" to promote the formation of APP and P-tau. Furthermore, the cytoskeleton was an essential target for T-2 toxin to exert cytotoxicity, and JNK/HIF-1α participated in this damage. Collectively, when the T-2 toxin induces the production of APP and P-tau, JNK might interfere with HIF-1α's protective function. This study will provide clues for further research on the neurotoxicity of mycotoxins.


Amyloid beta-Protein Precursor , Hypoxia-Inducible Factor 1, alpha Subunit , T-2 Toxin , tau Proteins , T-2 Toxin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , tau Proteins/metabolism , Phosphorylation/drug effects , Amyloid beta-Protein Precursor/metabolism , Mice , Animals , Cell Line , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/drug effects
9.
Cell Rep ; 42(12): 113535, 2023 12 26.
Article En | MEDLINE | ID: mdl-38060450

The phosphoinositide 3-kinase p110α is an essential mediator of insulin signaling and glucose homeostasis. We interrogated the human serine, threonine, and tyrosine kinome to search for novel regulators of p110α and found that the Hippo kinases phosphorylate p110α at T1061, which inhibits its activity. This inhibitory state corresponds to a conformational change of a membrane-binding domain on p110α, which impairs its ability to engage membranes. In human primary hepatocytes, cancer cell lines, and rodent tissues, activation of the Hippo kinases MST1/2 using forskolin or epinephrine is associated with phosphorylation of T1061 and inhibition of p110α, impairment of downstream insulin signaling, and suppression of glycolysis and glycogen synthesis. These changes are abrogated when MST1/2 are genetically deleted or inhibited with small molecules or if the T1061 is mutated to alanine. Our study defines an inhibitory pathway of PI3K signaling and a link between epinephrine and insulin signaling.


Protein Serine-Threonine Kinases , Humans , Animals , Mice , Cell Line , Mice, Inbred C57BL , Male , Female , Epinephrine/pharmacology , Enzyme Activation/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Gene Deletion , Colforsin/pharmacology , Insulin/metabolism , Phosphorylation/drug effects , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics
10.
J Biol Chem ; 299(12): 105369, 2023 Dec.
Article En | MEDLINE | ID: mdl-37865311

Cardiac MyBP-C (cMyBP-C) interacts with actin and myosin to fine-tune cardiac muscle contractility. Phosphorylation of cMyBP-C, which reduces the binding of cMyBP-C to actin and myosin, is often decreased in patients with heart failure (HF) and is cardioprotective in model systems of HF. Therefore, cMyBP-C is a potential target for HF drugs that mimic its phosphorylation and/or perturb its interactions with actin or myosin. We labeled actin with fluorescein-5-maleimide (FMAL) and the C0-C2 fragment of cMyBP-C (cC0-C2) with tetramethylrhodamine (TMR). We performed two complementary high-throughput screens (HTS) on an FDA-approved drug library, to discover small molecules that specifically bind to cMyBP-C and affect its interactions with actin or myosin, using fluorescence lifetime (FLT) detection. We first excited FMAL and detected its FLT, to measure changes in fluorescence resonance energy transfer (FRET) from FMAL (donor) to TMR (acceptor), indicating binding. Using the same samples, we then excited TMR directly, using a longer wavelength laser, to detect the effects of compounds on the environmentally sensitive FLT of TMR, to identify compounds that bind directly to cC0-C2. Secondary assays, performed on selected modulators with the most promising effects in the primary HTS assays, characterized the specificity of these compounds for phosphorylated versus unphosphorylated cC0-C2 and for cC0-C2 versus C1-C2 of fast skeletal muscle (fC1-C2). A subset of identified compounds modulated ATPase activity in cardiac and/or skeletal myofibrils. These assays establish the feasibility of the discovery of small-molecule modulators of the cMyBP-C-actin/myosin interaction, with the ultimate goal of developing therapies for HF.


Carrier Proteins , Drug Discovery , Heart Failure , Myofibrils , Small Molecule Libraries , Humans , Actins/metabolism , Drug Discovery/methods , Heart Failure/drug therapy , Heart Failure/metabolism , Myocardium/metabolism , Myosins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Small Molecule Libraries/pharmacology , Drug Evaluation, Preclinical , Myofibrils/drug effects , Carrier Proteins/metabolism , Biosensing Techniques , Adenosine Triphosphatases/metabolism , Muscle, Skeletal/metabolism , Recombinant Proteins/metabolism , Enzyme Activation/drug effects , Fluorescence Resonance Energy Transfer
11.
J Biol Chem ; 299(7): 104914, 2023 07.
Article En | MEDLINE | ID: mdl-37315787

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase (RTK) commonly targeted for inhibition by anticancer therapeutics. Current therapeutics target EGFR's kinase domain or extracellular region. However, these types of inhibitors are not specific for tumors over healthy tissue and therefore cause undesirable side effects. Our lab has recently developed a new strategy to regulate RTK activity by designing a peptide that specifically binds to the transmembrane (TM) region of the RTK to allosterically modify kinase activity. These peptides are acidity-responsive, allowing them to preferentially target acidic environments like tumors. We have applied this strategy to EGFR and created the PET1 peptide. We observed that PET1 behaves as a pH-responsive peptide that modulates the configuration of the EGFR TM through a direct interaction. Our data indicated that PET1 inhibits EGFR-mediated cell migration. Finally, we investigated the mechanism of inhibition through molecular dynamics simulations, which showed that PET1 sits between the two EGFR TM helices; this molecular mechanism was additionally supported by AlphaFold-Multimer predictions. We propose that the PET1-induced disruption of native TM interactions disturbs the conformation of the kinase domain in such a way that it inhibits EGFR's ability to send migratory cell signals. This study is a proof-of-concept that acidity-responsive membrane peptide ligands can be generally applied to RTKs. In addition, PET1 constitutes a viable approach to therapeutically target the TM of EGFR.


Allosteric Regulation , Cell Membrane , ErbB Receptors , Peptides , Humans , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Protein Structure, Secondary/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Allosteric Regulation/drug effects , Cell Membrane/chemistry , Cell Membrane/metabolism , Hydrogen-Ion Concentration , Peptides/pharmacology , Cell Movement/drug effects , Protein Domains/drug effects , Antineoplastic Agents/pharmacology
12.
J Virol ; 97(4): e0016023, 2023 04 27.
Article En | MEDLINE | ID: mdl-36939350

Host-derived cellular pathways can provide an unfavorable environment for virus replication. These pathways have been a subject of interest for herpesviruses, including the betaherpesvirus human cytomegalovirus (HCMV). Here, we demonstrate that a compound, ARP101, induces the noncanonical sequestosome 1 (SQSTM1)/p62-Keap1-Nrf2 pathway for HCMV suppression. ARP101 increased the levels of both LC3 II and SQSTM1/p62 and induced phosphorylation of p62 at the C-terminal domain, resulting in its increased affinity for Keap1. ARP101 treatment resulted in Nrf2 stabilization and translocation into the nucleus, binding to specific promoter sites and transcription of antioxidant enzymes under the antioxidant response element (ARE), and HCMV suppression. Knockdown of Nrf2 recovered HCMV replication following ARP101 treatment, indicating the role of the Keap1-Nrf2 axis in HCMV inhibition by ARP101. SQSTM1/p62 phosphorylation was not modulated by the mTOR kinase or casein kinase 1 or 2, indicating ARP101 engages other kinases. Together, the data uncover a novel antiviral strategy for SQSTM1/p62 through the noncanonical Keap1-Nrf2 axis. This pathway could be further exploited, including the identification of the responsible kinases, to define the biological events during HCMV replication. IMPORTANCE Antiviral treatment for human cytomegalovirus (HCMV) is limited and suffers from the selection of drug-resistant viruses. Several cellular pathways have been shown to modulate HCMV replication. The autophagy receptor sequestosome 1 (SQSTM1)/p62 has been reported to interact with several HCMV proteins, particularly with components of HCMV capsid, suggesting it plays a role in viral replication. Here, we report on a new and unexpected role for SQSTM1/p62, in HCMV suppression. Using a small-molecule probe, ARP101, we show SQSTM1/p62 phosphorylation at its C terminus domain initiates the noncanonical Keap1-Nrf2 axis, leading to transcription of genes under the antioxidant response element, resulting in HCMV inhibition in vitro. Our study highlights the dynamic nature of SQSTM1/p62 during HCMV infection and how its phosphorylation activates a new pathway that can be exploited for antiviral intervention.


Cytomegalovirus Infections , Cytomegalovirus , Virus Replication , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Antiviral Agents/pharmacology , Transcription, Genetic/drug effects , Phosphorylation/drug effects , Antioxidant Response Elements/drug effects , Cell Line , Humans
13.
Biochem Pharmacol ; 210: 115488, 2023 04.
Article En | MEDLINE | ID: mdl-36889445

The 90 kDa ribosomal S6 kinase (RSK) family of proteins is a group of highly conserved Ser/Thr kinases. They are downstream effectors of the Ras/ERK/MAPK signaling cascade. ERK1/2 activation directly results in the phosphorylation of RSKs, which further, through interaction with a variety of different downstream substrates, activate various signaling events. In this context, they have been shown to mediate diverse cellular processes like cell survival, growth, proliferation, EMT, invasion, and metastasis. Interestingly, increased expression of RSKs has also been demonstrated in various cancers, such as breast, prostate, and lung cancer. This review aims to present the most recent advances in the field of RSK signaling that have occurred, such as biological insights, function, and mechanisms associated with carcinogenesis. We additionally present and discuss the recent advances but also the limitations in the development of pharmacological inhibitors of RSKs, in the context of the use of these kinases as putative, more efficient targets for novel anticancer therapeutic approaches.


Antineoplastic Agents , Carcinogenesis , Molecular Targeted Therapy , Neoplasms , Ribosomal Protein S6 Kinases, 90-kDa , Animals , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Enzyme Activation , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
14.
Nature ; 611(7934): 173-179, 2022 11.
Article En | MEDLINE | ID: mdl-36289326

G-protein-coupled receptors (GPCRs), the largest family of signalling receptors, as well as important drug targets, are known to activate extracellular-signal-regulated kinase (ERK)-a master regulator of cell proliferation and survival1. However, the precise mechanisms that underlie GPCR-mediated ERK activation are not clearly understood2-4. Here we investigated how spatially organized ß2-adrenergic receptor (ß2AR) signalling controls ERK. Using subcellularly targeted ERK activity biosensors5, we show that ß2AR signalling induces ERK activity at endosomes, but not at the plasma membrane. This pool of ERK activity depends on active, endosome-localized Gαs and requires ligand-stimulated ß2AR endocytosis. We further identify an endosomally localized non-canonical signalling axis comprising Gαs, RAF and mitogen-activated protein kinase kinase, resulting in endosomal ERK activity that propagates into the nucleus. Selective inhibition of endosomal ß2AR and Gαs signalling blunted nuclear ERK activity, MYC gene expression and cell proliferation. These results reveal a non-canonical mechanism for the spatial regulation of ERK through GPCR signalling and identify a functionally important endosomal signalling axis.


Adrenergic Agents , Endosomes , Extracellular Signal-Regulated MAP Kinases , Receptors, Adrenergic, beta-2 , Adrenergic Agents/metabolism , Adrenergic Agents/pharmacology , Cell Proliferation , Endosomes/drug effects , Endosomes/enzymology , Endosomes/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, myc , GTP-Binding Protein alpha Subunits, Gs/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Oxid Med Cell Longev ; 2022: 8661200, 2022.
Article En | MEDLINE | ID: mdl-35993019

Background: Neuroinflammation-induced phosphorylated Tau (p-Tau) deposition in central nervous system contributes to neurodegenerative disorders. Propofol possesses neuroprotective properties. We investigated its impacts on tumor necrosis factor-α (TNF-α)-mediated p-Tau deposition in neurons. Methods: Mouse hippocampal neurons were exposed to propofol followed by TNF-α. Cell viability, p-Tau, mitophagy, reactive oxygen species (ROS), NOD-like receptor protein 3 (NLRP3), antioxidant enzymes, and p62/Keap1/Nrf2 pathway were investigated. Results: TNF-α promoted p-Tau accumulation in a concentration- and time-dependent manner. TNF-α (20 ng/mL, 4 h) inhibited mitophagy while increased ROS accumulation and NLRP3 activation. It also induced glycogen synthase kinase-3ß (GSK3ß) while inhibited protein phosphatase 2A (PP2A) phosphorylation. All these effects were attenuated by 25 µM propofol. In addition, TNF-α-induced p-Tau accumulation was attenuated by ROS scavenger, NLRP3 inhibitor, GSK3ß inhibitor, or PP2A activator. Besides, compared with control neurons, 100 µM propofol decreased p-Tau accumulation. It also decreased ROS and NLRP3 activation, modulated GSK3ß/PP2A phosphorylation, leaving mitophagy unchanged. Further, 100 µM propofol induced p62 expression, reduced Keap1 expression, triggered the nuclear translocation of Nrf2, and upregulated superoxide dismutase (SOD) and heme oxygenase-1 (HO-1) expression, which was abolished by p62 knockdown, Keap1 overexpression, or Nrf2 inhibitor. Consistently, the inhibitory effect of 100 µM propofol on ROS and p-Tau accumulation was mitigated by p62 knockdown, Keap1 overexpression, or Nrf2 inhibitor. Conclusions: In hippocampal neurons, TNF-α inhibited mitophagy, caused oxidative stress and NLRP3 activation, leading to GSK3ß/PP2A-dependent Tau phosphorylation. Propofol may reduce p-Tau accumulation by reversing mitophagy and oxidative stress-related events. Besides, propofol may reduce p-Tau accumulation by modulating SOD and HO-1 expression through p62/Keap1/Nrf2 pathway.


Mitophagy , NF-E2-Related Factor 2 , Propofol , Transcription Factor TFIIH , Tumor Necrosis Factor-alpha , tau Proteins , Animals , Mice , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Mitophagy/genetics , Mitophagy/physiology , Neurons/drug effects , Neurons/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Propofol/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Superoxide Dismutase/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
Science ; 377(6603): 328-335, 2022 07 15.
Article En | MEDLINE | ID: mdl-35857590

Human NLRP1 (NACHT, LRR, and PYD domain-containing protein 1) is an innate immune sensor predominantly expressed in the skin and airway epithelium. Here, we report that human NLRP1 senses the ultraviolet B (UVB)- and toxin-induced ribotoxic stress response (RSR). Biochemically, RSR leads to the direct hyperphosphorylation of a human-specific disordered linker region of NLRP1 (NLRP1DR) by MAP3K20/ZAKα kinase and its downstream effector, p38. Mutating a single ZAKα phosphorylation site in NLRP1DR abrogates UVB- and ribotoxin-driven pyroptosis in human keratinocytes. Moreover, fusing NLRP1DR to CARD8, which is insensitive to RSR by itself, creates a minimal inflammasome sensor for UVB and ribotoxins. These results provide insight into UVB sensing by human skin keratinocytes, identify several ribotoxins as NLRP1 agonists, and establish inflammasome-driven pyroptosis as an integral component of the RSR.


Inflammasomes , MAP Kinase Kinase Kinases , NLR Proteins , Pyroptosis , Ribosomes , Stress, Physiological , Anisomycin/toxicity , CARD Signaling Adaptor Proteins/metabolism , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Inflammasomes/radiation effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , MAP Kinase Kinase Kinases/metabolism , Mutation , NLR Proteins/genetics , NLR Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Pyroptosis/drug effects , Pyroptosis/radiation effects , Ribosomes/drug effects , Ribosomes/radiation effects , Ultraviolet Rays
17.
J Med Food ; 25(7): 770-777, 2022 Jul.
Article En | MEDLINE | ID: mdl-35834632

Umbilicaria esculenta (UE), an edible lichen, is widespread in northeast Asian countries, including China, Japan, and Korea. In the present study, we examined the antiwrinkle activity of UE. We observed that the UE extract (UEE) suppressed ultraviolet (UV)-induced matrix metalloprotein-1 (MMP-1) expression and reactive oxygen species (ROS) generation in a human keratinocyte cell line (HaCaT) and human skin tissue. In addition, UEE reversed the UV-induced decrease in collagen in the human skin tissue. Excessive and chronic UV exposure is a key factor underlying skin wrinkle formation via MMP-1 expression. As treatment with UEE disrupted the UV-activated mitogen-activated protein kinase (MAPK) signaling pathway, we applied an antibody array to unveil the underlying mechanism of UEE. Interestingly, UEE treatment inhibited ErbB2 phosphorylation, but not epidermal growth factor receptor phosphorylation, a heterodimerization partner with ErbB2. Furthermore, UEE treatment enhanced UV-suppressed phosphatase activity via ROS suppression. Collectively, our findings indicate that UEE enhances ErbB2 dephosphorylation to suppress UV-induced MMP-1 expression.


Ascomycota , Receptor, ErbB-2 , Skin Aging , Skin , Tissue Extracts , Fibroblasts/drug effects , Fibroblasts/metabolism , HaCaT Cells/drug effects , HaCaT Cells/metabolism , Humans , Lichens , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Aging/drug effects , Tissue Extracts/pharmacology , Ultraviolet Rays/adverse effects
18.
J Enzyme Inhib Med Chem ; 37(1): 2004-2016, 2022 Dec.
Article En | MEDLINE | ID: mdl-35844184

Gastric cancer remains a significant health burden worldwide. In continuation of our previous study and development of effective small molecules against gastric cancer, a series of benzochalcone analogues involving heterocyclic molecules were synthesised and biologically evaluated in vitro and in vivo. Among them, the quinolin-6-yl substituted derivative KL-6 inhibited the growth of gastric cancer cells (HGC27, MKN28, AZ521, AGS, and MKN1) with a submicromolar to micromolar range of IC50, being the most potent one in this series. Additionally, KL-6 significantly inhibited the colony formation, migration and invasion, and effectively induced apoptosis of MKN1 cells in a concentration-dependent manner. The mechanistic study revealed that KL-6 could concentration-dependently suppress STAT3 phosphorylation, which may partly contribute to its anticancer activity. Furthermore, in vivo antitumour study on the MKN1 orthotopic tumour model showed that KL-6 effectively inhibited tumour growth (TGI of 78%) and metastasis without obvious toxicity. Collectively, compound KL-6 may support the further development of candidates for gastric cancer treatment.


Chalcones , STAT3 Transcription Factor , Stomach Neoplasms , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/pharmacology , Humans , Molecular Targeted Therapy , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy
19.
J Ethnopharmacol ; 296: 115440, 2022 Oct 05.
Article En | MEDLINE | ID: mdl-35671865

ETHNOPHARMACOLOGICAL RELEVANCE: Phikud Navakot (PN), a mixture of nine herbal plants, is an ancient Thai traditional medicine used for relieving circulatory disorders and dizziness. PN has also shown anti-inflammatory effects in rats with acute myocardial infarction. Moreover, phytochemical-inhibiting neuroinflammation, including gallic acid, vanillic acid, ferulic acid, and rutin were detected in PN extract; however, the anti-neuroinflammatory activity of PN extract and its components in a coculture system of microglia and neuronal cells is limited. OBJECTIVE: To investigate the anti-neuroinflammatory activities of PN on lipopolysaccharide (LPS)-induced inflammation in a coculture system of microglia and neuronal cells. METHODS: ELISA and qRT-PCR were used to assess cytokine expression. The phosphorylation of mitogen-activated protein kinases (MAPKs) was determined by Western blotting. Microglia-mediated neuroinflammation was evaluated using a BV-2 microglia-N2a neuron transwell co-culture. RESULTS: PN extract and its component, gallic acid, decreased LPS-induced the mRNA expression of interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS), as well as IL-6 protein levels in both microglial monoculture and coculture systems. This was accompanied by a reduction in neurodegeneration triggered by microglia in N2a neurons with increased neuronal integrity markers (ßIII tubulin and tyrosine hydroxylase (TH)). These effects were caused by the ability of PN extract to inhibit extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) activation. CONCLUSION: This is the first study to show that PN extract inhibits neurodegeneration in LPS-activated BV-2 microglia by targeting ERK signaling activity.


Lipopolysaccharides , MAP Kinase Signaling System , Microglia , Plant Extracts , Animals , Coculture Techniques , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Microglia/drug effects , Microglia/metabolism , NF-kappa B/metabolism , Neurons/drug effects , Neurons/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Plant Extracts/pharmacology , Rats
20.
Eur J Pharmacol ; 927: 175067, 2022 Jul 15.
Article En | MEDLINE | ID: mdl-35654135

The cell proliferation effect of S-allyl-L-cysteine (SAC) and its mechanisms were examined in primary cultures of adult rat hepatocytes. In serum-free cultivation, SAC (10-6 M)-stimulated hepatocytes showed significant proliferation compared to control at 5-h culture; the effect was dependent on the culture time and the dose of SAC (EC50 value 8.58 × 10-8 M). In addition, SAC-stimulated hepatocytes significantly increased mRNA expression levels of c-Myc and c-Fos at 1 h and cyclin B1 at 3.5 and 4 h, respectively. In contrast, alliin and allicin, structural analogs of SAC, did not show these effects observed with SAC. The SAC-induced hepatocyte proliferation effects were completely suppressed by monoclonal antibodies against growth hormone receptor and insulin-like growth factor type-I (IGF-I) receptor, respectively. Furthermore, the Janus kinase 2 (JAK2) inhibitor TG101209, phospholipase C (PLC) inhibitor U-73122, IGF-I receptor tyrosine kinase (RTK) inhibitor AG538, PI3 kinase inhibitor LY294002, MEK inhibitor PD98059, and mTOR inhibitor rapamycin completely suppressed the SAC-induced hepatocyte proliferation. JAK2 (p125 kDa) phosphorylation in cultured hepatocytes peaked 5 min after SAC stimulation. SAC-induced IGF-I RTK (p95 kDa) and ERK2 (p42 kDa) phosphorylation had slower rises than JAK2, peaking at 20 and 30 min, respectively. These results indicate that SAC promoted cell proliferation by growth hormone receptor/JAK2/PLC pathway activation followed by activation of the IGF-I RTK/PI3K/ERK2/mTOR pathway in primary cultures of adult rat hepatocytes.


Cysteine/analogs & derivatives , Hepatocytes , Insulin-Like Growth Factor I , Janus Kinase 2 , Mitogen-Activated Protein Kinase 1 , Receptor, IGF Type 1 , Animals , Cell Proliferation/drug effects , Cysteine/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Janus Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Rats , Receptor, IGF Type 1/metabolism , Receptors, Somatotropin/metabolism , TOR Serine-Threonine Kinases/metabolism
...