Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.453
Filter
1.
Int J Nanomedicine ; 19: 9035-9053, 2024.
Article in English | MEDLINE | ID: mdl-39253060

ABSTRACT

Background: Ischemic preconditioning-induced serum exosomes (IPC-exo) protected rat heart against myocardial ischemia/reperfusion injury. However, whether IPC-exo regulate replacement fibrosis after myocardial infarction (MI) and the underlying mechanisms remain unclear. MicroRNAs (miRs) are important cargos of exosomes and play an essential role in cardioprotection. We aim to investigate whether IPC-exo regulate post-MI replacement fibrosis by transferring cardioprotective miRs and its action mechanism. Methods: Exosomes obtained from serum of adult rats in control (Con-exo) and IPC groups were identified and analyzed, subsequently intracardially injected into MI rats following ligation. Their miRs profiles were identified using high-throughput miR sequencing to identify target miRs for bioinformatics analysis. Luciferase reporter assays confirmed target genes of selected miRs. IPC-exo transfected with selected miRs antagomir or NC were intracardially administered to MI rats post-ligation. Cardiac function and degree of replacement fibrosis were detected 4 weeks post-MI. Results: IPC-exo exerted cardioprotective effects against excessive replacement fibrosis. MiR sequencing and RT-qPCR identified miR-133a-3p as most significantly different between IPC-exo and Con-exo. MiR-133a-3p directly targeted latent transforming growth factor beta binding protein 1 (LTBP1) and protein phosphatase 2, catalytic subunit, alpha isozyme (PPP2CA). KEGG analysis showed that transforming growth factor-ß (TGF-ß) was one of the most enriched signaling pathways with miR-133a-3p. Comparing to injection of IPC-exo transfected with miR-133a-3p antagomir NC, injecting IPC-exo transfected with miR-133a-3p antagomir abolished protective effects of IPC-exo on declining excessive replacement fibrosis and cardiac function enhancement, while increasing the messenger RNA and protein expression of LTBP1, PPP2CA, and TGF-ß1in MI rats. Conclusion: IPC-exo inhibit excessive replacement fibrosis and improve cardiac function post-MI by transferring miR-133a-3p, the mechanism is associated with directly targeting LTBP1 and PPP2CA, and indirectly regulating TGF-ß pathway in rats. Our finding provides potential therapeutic effect of IPC-induced exosomal miR-133a-3p for cardiac repair.


Subject(s)
Exosomes , MicroRNAs , Myocardial Infarction , Protein Phosphatase 2 , Animals , MicroRNAs/blood , MicroRNAs/genetics , Myocardial Infarction/blood , Myocardial Infarction/therapy , Myocardial Infarction/genetics , Exosomes/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Male , Rats , Rats, Sprague-Dawley , Fibrosis , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/therapy , Myocardium/metabolism , Ischemic Preconditioning/methods , Ischemic Preconditioning, Myocardial/methods
2.
J Cancer Res Clin Oncol ; 150(9): 416, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249161

ABSTRACT

BACKGROUND: Gastric cancer (GC), a prevalent malignant tumor which is a leading cause of death from malignancy around the world. Peritoneal metastasis accounts for the major cause of mortality in patients with GC. Despite hyperthermia intraperitoneal chemotherapy (HIPEC) improves the therapeutic effect of GC, it's equivocal about the mechanism under HIPEC. METHODS: MiR-183-5p expression was sifted from miRNA chip and detected in both GC patients and cell lines by qRT-PCR. Gene interference and rescue experiments were performed to identified biological function in vitro and vivo. Next, we affirmed PPP2CA as targeted of miR-183-5p by dual luciferase reporter assay. Finally, the potential relationship between HIPEC and miR-183-5p was explored. RESULTS: MiR-183-5p is up-regulated in GC and associated with advanced stage and poor prognosis. MiR-183-5p accelerate GC migration in vitro which is influenced by miR-183-5p/PPP2CA/AKT/GSK3ß/ß-catenin Axis. HIPEC exerts migration inhibition via attenuating miR-183-5p expression. CONCLUSION: MiR-183-5p can be used as a potential HIPEC biomarker in patients with CC.


Subject(s)
Cell Movement , Glycogen Synthase Kinase 3 beta , Hyperthermia, Induced , MicroRNAs , Proto-Oncogene Proteins c-akt , Stomach Neoplasms , beta Catenin , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , MicroRNAs/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Hyperthermia, Induced/methods , beta Catenin/metabolism , beta Catenin/genetics , Proto-Oncogene Proteins c-akt/metabolism , Mice , Animals , Male , Female , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Cell Line, Tumor , Mice, Nude , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , Prognosis , Middle Aged , Mice, Inbred BALB C , Antineoplastic Agents, Phytogenic/pharmacology
3.
Cell Commun Signal ; 22(1): 391, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113090

ABSTRACT

BACKGROUND: Approximately 25-30% of patients with acute myeloid leukemia (AML) have FMS-like receptor tyrosine kinase-3 (FLT3) mutations that contribute to disease progression and poor prognosis. Prolonged exposure to FLT3 tyrosine kinase inhibitors (TKIs) often results in limited clinical responses due to diverse compensatory survival signals. Therefore, there is an urgent need to elucidate the mechanisms underlying FLT3 TKI resistance. Dysregulated sphingolipid metabolism frequently contributes to cancer progression and a poor therapeutic response. However, its relationship with TKI sensitivity in FLT3-mutated AML remains unknown. Thus, we aimed to assess mechanisms of FLT3 TKI resistance in AML. METHODS: We performed lipidomics profiling, RNA-seq, qRT-PCR, and enzyme-linked immunosorbent assays to determine potential drivers of sorafenib resistance. FLT3 signaling was inhibited by sorafenib or quizartinib, and SPHK1 was inhibited by using an antagonist or via knockdown. Cell growth and apoptosis were assessed in FLT3-mutated and wild-type AML cell lines via Cell counting kit-8, PI staining, and Annexin-V/7AAD assays. Western blotting and immunofluorescence assays were employed to explore the underlying molecular mechanisms through rescue experiments using SPHK1 overexpression and exogenous S1P, as well as inhibitors of S1P2, ß-catenin, PP2A, and GSK3ß. Xenograft murine model, patient samples, and publicly available data were analyzed to corroborate our in vitro results. RESULTS: We demonstrate that long-term sorafenib treatment upregulates SPHK1/sphingosine-1-phosphate (S1P) signaling, which in turn positively modulates ß-catenin signaling to counteract TKI-mediated suppression of FLT3-mutated AML cells via the S1P2 receptor. Genetic or pharmacological inhibition of SPHK1 potently enhanced the TKI-mediated inhibition of proliferation and apoptosis induction in FLT3-mutated AML cells in vitro. SPHK1 knockdown enhanced sorafenib efficacy and improved survival of AML-xenografted mice. Mechanistically, targeting the SPHK1/S1P/S1P2 signaling synergizes with FLT3 TKIs to inhibit ß-catenin activity by activating the protein phosphatase 2 A (PP2A)-glycogen synthase kinase 3ß (GSK3ß) pathway. CONCLUSIONS: These findings establish the sphingolipid metabolic enzyme SPHK1 as a regulator of TKI sensitivity and suggest that combining SPHK1 inhibition with TKIs could be an effective approach for treating FLT3-mutated AML.


Subject(s)
Glycogen Synthase Kinase 3 beta , Leukemia, Myeloid, Acute , Phosphotransferases (Alcohol Group Acceptor) , Protein Phosphatase 2 , beta Catenin , fms-Like Tyrosine Kinase 3 , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , beta Catenin/metabolism , beta Catenin/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Mice , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/antagonists & inhibitors , Cell Line, Tumor , Sorafenib/pharmacology , Apoptosis/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Cell Proliferation/drug effects , Drug Synergism , Xenograft Model Antitumor Assays , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics
4.
Cell Signal ; 123: 111352, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39173855

ABSTRACT

Cellular signaling pathways rely on posttranslational modifications (PTMs) to finely regulate protein functions, particularly transcription factors. The Hedgehog (Hh) signaling cascade, crucial for embryonic development and tissue homeostasis, is susceptible to aberrations that lead to developmental anomalies and various cancers. At the core of Hh signaling are Gli proteins, whose dynamic balance between activator (GliA) and repressor (GliR) states shapes cellular outcomes. Phosphorylation, orchestrated by multiple kinases, is pivotal in regulating Gli activity. While kinases in this context have been extensively studied, the role of protein phosphatases, particularly Protein Phosphatase 2A (PP2A), remains less explored. This study unveils a novel role for the B″gamma subunit of PP2A, PPP2R3C, in Hh signaling regulation. PPP2R3C interacts with Gli proteins, and its disruption reduces Hedgehog pathway activity as measured by reduced expression of Gli1/2 and Hh target genes upon Hh signaling activation, and reduced growth of a Hh signaling-dependent medulloblastoma cell line. Moreover, we establish an antagonistic connection between PPP2R3C and MEKK1 kinase in Gli protein phosphorylation, underscoring the intricate interplay between kinases and phosphatases in Hh signaling pathway. This study sheds light on the previously understudied role of protein phosphatases in Hh signaling and provides insights into their significance in cellular regulation.


Subject(s)
Hedgehog Proteins , Protein Phosphatase 2 , Signal Transduction , Zinc Finger Protein GLI1 , Hedgehog Proteins/metabolism , Protein Phosphatase 2/metabolism , Humans , Zinc Finger Protein GLI1/metabolism , Phosphorylation , HEK293 Cells , Animals , Cell Line, Tumor , Zinc Finger Protein Gli2/metabolism , Mice
5.
Cell Death Dis ; 15(8): 621, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187513

ABSTRACT

Despite advancements in chemotherapy and the availability of novel therapies, the outcome of adult patients with B-cell acute lymphoblastic leukemia (B-ALL) remains unsatisfactory. Therefore, it is necessary to understand the molecular mechanisms underlying the progression of B-ALL. Brahma-related gene 1 (BRG1) is a poor prognostic factor for multiple cancers. Here, the expression of BRG1 was found to be higher in patients with B-ALL, irrespective of the molecular subtype, than in healthy individuals, and its overexpression was associated with a poor prognosis. Upregulation of BRG1 accelerated cell cycle progression into the S phase, resulting in increased cell proliferation, whereas its downregulation facilitated the apoptosis of B-ALL cells. Mechanistically, BRG1 occupies the transcriptional activation site of PPP2R1A, thereby inhibiting its expression and activating the PI3K/AKT signaling pathway to regulate the proto-oncogenes c-Myc and BCL-2. Consistently, silencing of BRG1 and administration of PFI-3 (a specific inhibitor targeting BRG1) significantly inhibited the progression of leukemia and effectively prolonged survival in cell-derived xenograft mouse models of B-ALL. Altogether, this study demonstrates that BRG1-induced overactivation of the PPP2R1A/PI3K/AKT signaling pathway plays an important role in promoting the progression of B-ALL. Therefore, targeting BRG1 represents a promising strategy for the treatment of B-ALL in adults.


Subject(s)
DNA Helicases , Disease Progression , Nuclear Proteins , Protein Phosphatase 2 , Transcription Factors , Animals , Female , Humans , Male , Mice , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Helicases/metabolism , DNA Helicases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic/drug effects
6.
J Virol ; 98(9): e0104724, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194238

ABSTRACT

Little is known regarding the molecular mechanisms that highly pathogenic Marburg virus (MARV) utilizes to transcribe and replicate its genome. Previous studies assumed that dephosphorylation of the filoviral transcription factor VP30 supports transcription, while phosphorylated VP30 reduces transcription. Here, we focused on the role of the host protein phosphatase 2A (PP2A) for VP30 dephosphorylation and promotion of viral transcription. We could show that MARV NP interacts with the subunit B56 of PP2A, as previously shown for the Ebola virus, and that this interaction is important for MARV transcription activity. Inhibition of the interaction between PP2A and NP either by mutating the B56 binding motif encoded on NP, or the use of a PP2A inhibitor, induced VP30 hyperphosphorylation, and as a consequence a decrease of MARV transcription as well as viral growth. These results suggest that NP plays a key role in the dephosphorylation of VP30 by recruiting PP2A. Generation of recombinant (rec) MARV lacking the PP2A-B56 interaction motif on NP was not possible suggesting an essential role of PP2A-mediated VP30 dephosphorylation for the MARV replication cycle. Likewise, we were not able to generate recMARV containing VP30 phosphomimetic mutants indicating that dynamic cycles of VP30 de- and rephosphorylation are a prerequisite for an efficient viral life cycle. As the specific binding motifs of PP2A-B56 and VP30 within NP are highly conserved among the filoviral family, our data suggest a conserved mechanism for filovirus VP30 dephosphorylation by PP2A, revealing the host factor PP2A as a promising target for pan-filoviral therapies. IMPORTANCE: Our study elucidates the crucial role of host protein phosphatase 2A (PP2A) in Marburg virus (MARV) transcription. The regulatory subunit B56 of PP2A facilitates VP30 dephosphorylation, and hence transcription activation, via binding to NP. Our results, together with previous data, reveal a conserved mechanism of filovirus VP30 dephosphorylation by host factor PP2A at the NP interface and provide novel insights into potential pan-filovirus therapies.


Subject(s)
Marburgvirus , Protein Phosphatase 2 , Transcription, Genetic , Marburgvirus/physiology , Marburgvirus/genetics , Marburgvirus/metabolism , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Humans , Phosphorylation , Virus Replication , HEK293 Cells , Animals , Host-Pathogen Interactions , Viral Proteins/metabolism , Viral Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Marburg Virus Disease/virology , Marburg Virus Disease/metabolism , Protein Binding , Cell Line
7.
Cell Mol Life Sci ; 81(1): 292, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38976080

ABSTRACT

Cisplatin resistance is a major challenge for systemic therapy against advanced bladder cancer (BC). Little information is available on the regulation of cisplatin resistance and the underlying mechanisms require elucidation. Here, we detected that downregulation of the tumor suppressor, PPP2R2B (a serine/threonine protein phosphatase 2 A regulatory subunit), in BC promoted cell proliferation and migration. What's more, low PPP2R2B expression was correlated with cisplatin resistance. In vitro and in vivo experiments verified that PPP2R2B could promote BC sensitivity to cisplatin. In terms of mechanism, we identified a novel function of PPP2R2B as a nucleocytoplasmic transport molecule. PPP2R2B promoted ISG15 entry into the nucleus by mediating binding of IPO5 with ISG15. Nuclear translocation of ISG15 inhibited DNA repair, further increasing ISG15 expression through activation of the STING pathway. Besides, PPP2R2B was down-regulated by SUV39H1-mediated histone 3 lysine 9 trimethylation, which could be restored by the SUV39H1-specific inhibitor, chaetocin. Our data suggest that PPP2R2B expression level is a potential biomarker for chemotherapy response and that chemotherapy in combination with chaetocin may be a feasible treatment strategy for patients with BC.


Subject(s)
Cisplatin , Cytokines , Drug Resistance, Neoplasm , Protein Phosphatase 2 , Ubiquitins , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Humans , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Ubiquitins/metabolism , Ubiquitins/genetics , Cytokines/metabolism , Animals , Cell Line, Tumor , Mice , Cell Proliferation/drug effects , Mice, Nude , Cell Nucleus/metabolism , Antineoplastic Agents/pharmacology , Mice, Inbred BALB C , Gene Expression Regulation, Neoplastic/drug effects , Cell Movement/drug effects , Female , Nerve Tissue Proteins
8.
Adv Sci (Weinh) ; 11(36): e2404080, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39041921

ABSTRACT

The molecular mechanism underlying abnormal osteoclastogenesis triggering subchondral bone remodeling in osteoarthritis (OA) is still unclear. Here, single-cell and bulk transcriptomics sequencing analyses are performed on GEO datasets to identify key molecules and validate them using knee joint tissues from OA patients and rat OA models. It is found that the catalytic subunit of protein phosphatase 2A (PP2Ac) is highly expressed during osteoclastogenesis in the early stage of OA and is correlated with autophagy. Knockdown or inhibition of PP2Ac weakened autophagy during osteoclastogenesis. Furthermore, the ULK1 expression of the downstream genes is significantly increased when PP2Ac is knocked down. PP2Ac-mediated autophagy is dependent on ULK1 phosphorylation activity during osteoclastogenesis, which is associated with enhanced dephosphorylation of ULK1 Ser637 residue regulating at the post-translational level. Additionally, mTORC1 inhibition facilitated the expression level of PP2Ac during osteoclastogenesis. In animal OA models, decreasing the expression of PP2Ac ameliorated early OA progression. The findings suggest that PP2Ac is also a promising therapeutic target in early OA.


Subject(s)
Autophagy-Related Protein-1 Homolog , Autophagy , Disease Models, Animal , Mechanistic Target of Rapamycin Complex 1 , Osteoarthritis , Osteogenesis , Protein Phosphatase 2 , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Rats , Autophagy/genetics , Autophagy/physiology , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Humans , Male , Osteoclasts/metabolism
9.
EMBO J ; 43(17): 3752-3786, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009675

ABSTRACT

Cytokinetic abscission marks the final stage of cell division, during which the daughter cells physically separate through the generation of new barriers, such as the plasma membrane or cell wall. While the contractile ring plays a central role during cytokinesis in bacteria, fungi and animal cells, the process diverges in Apicomplexa. In Toxoplasma gondii, two daughter cells are formed within the mother cell by endodyogeny. The mechanism by which the progeny cells acquire their plasma membrane during the disassembly of the mother cell, allowing daughter cells to emerge, remains unknown. Here we identify and characterize five T. gondii proteins, including three protein phosphatase 2A subunits, which exhibit a distinct and dynamic localization pattern during parasite division. Individual downregulation of these proteins prevents the accumulation of plasma membrane at the division plane, preventing the completion of cellular abscission. Remarkably, the absence of cytokinetic abscission does not hinder the completion of subsequent division cycles. The resulting progeny are able to egress from the infected cells but fail to glide and invade, except in cases of conjoined twin parasites.


Subject(s)
Cytokinesis , Protein Phosphatase 2 , Protozoan Proteins , Toxoplasma , Toxoplasma/enzymology , Toxoplasma/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Cell Membrane/metabolism , Animals , Cell Division , Humans
10.
ACS Appl Mater Interfaces ; 16(30): 38880-38892, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39016239

ABSTRACT

Post-translational modification, mitochondrial abruptions, neuroinflammation, and α-synuclein (α-Syn) aggregation are considered as major causes of Parkinson's disease (PD) pathogenesis. The recent literature highlights neuroimmune cross talk and the negative role of immune effector T (Teff) and positive regulation by regulatory T (Treg) cells in PD treatment. Herein, a strategy to endow Treg action paves the path for development of PD treatment. Thus, we explored the neuroprotective efficiency of the immunomodulator and PP2A (protein phosphatase 2) activator, FTY720 nanoparticles in in vivo experimental PD models. Repurposing of FTY720 for PD is known due to its protective effect by reducing PD and its camouflaged role in endowing EZH2-mediated epigenetic regulation of PD. EZH2-FOXP3 interaction is necessary for the neuroprotective Treg cell activity. Therefore, we synthesized FTY720 nanoparticles to improve FTY720 protective efficacy in an in vivo PD model to explore the PP2A mediated signaling. We confirmed the formation of FTY720NPs, and the results of the behavioral and protein expression study showed the significant neuroprotective efficiency of our nanoformulations. In the exploration of neuroprotective mechanism, several lines of evidence confirmed FTY720NPs mediated induction of PP2A/EZH2/FOXP3 signaling in the induction of Treg cells effect in in vivo PD treatment. In summary, our nanoformulations have novel potential to alleviate PD by inducing PP2A-induced epigenetic regulation-mediated neuroimmunomodulation at the clinical setup.


Subject(s)
Fingolimod Hydrochloride , Nanoparticles , Neuroprotective Agents , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Nanoparticles/chemistry , Mice , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/therapeutic use , Mice, Inbred C57BL , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Male , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Forkhead Transcription Factors/metabolism , Humans , Parkinsonian Disorders/drug therapy
11.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999976

ABSTRACT

Tumor angiogenesis, the formation of new blood vessels to support tumor growth and metastasis, is a complex process regulated by a multitude of signaling pathways. Dysregulation of signaling pathways involving protein kinases has been extensively studied, but the role of protein phosphatases in angiogenesis within the tumor microenvironment remains less explored. However, among angiogenic pathways, protein phosphatases play critical roles in modulating signaling cascades. This review provides a comprehensive overview of the involvement of protein phosphatases in tumor angiogenesis, highlighting their diverse functions and mechanisms of action. Protein phosphatases are key regulators of cellular signaling pathways by catalyzing the dephosphorylation of proteins, thereby modulating their activity and function. This review aims to assess the activity of the protein tyrosine phosphatases and serine/threonine phosphatases. These phosphatases exert their effects on angiogenic signaling pathways through various mechanisms, including direct dephosphorylation of angiogenic receptors and downstream signaling molecules. Moreover, protein phosphatases also crosstalk with other signaling pathways involved in angiogenesis, further emphasizing their significance in regulating tumor vascularization, including endothelial cell survival, sprouting, and vessel maturation. In conclusion, this review underscores the pivotal role of protein phosphatases in tumor angiogenesis and accentuate their potential as therapeutic targets for anti-angiogenic therapy in cancer.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Phosphoprotein Phosphatases , Signal Transduction , Humans , Neovascularization, Pathologic/metabolism , Neoplasms/blood supply , Neoplasms/metabolism , Neoplasms/pathology , Animals , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2/metabolism , Tumor Microenvironment , Phosphorylation , Angiogenesis
12.
Curr Pharm Biotechnol ; 25(12): 1585-1601, 2024.
Article in English | MEDLINE | ID: mdl-39034837

ABSTRACT

BACKGROUND: Cantharidin (CTD), a natural toxic compound from blister beetle Mylabris, has been used for cancer treatment for millenary. CTD and its analogs have become mainstream adjuvant drugs with radiotherapy and chemotherapy in clinical applications. However, the detailed pharmacology mechanism of CTD was not fully elucidated. METHODS: Publications of CTD were collected from the Web of Science Core Collection database from 1991 to 2023 using CiteSpace, VOSviewer, and Scimago Graphica software. RESULTS: A total of 1,611 publications of CTD were mainly published in China and the United States. The University of Newcastle has published the most researches. Mcclusey, Adam, Sakoff, Jennette, and Zhang, Yalin had the most CTD publications with higher H. Notably, CTD researches were mainly published in Bioorganic & Medicinal Chemistry Letters and the Journal of Biological Chemistry. Cluster profile results revealed that protein phosphatase 2A (PP2A), human gallbladder carcinoma, Aidi injection, and cell apoptosis were the hotspots. Concentration on the pharmacology function of PP2A subunit regulation, hepatotoxicity, nephrotoxicity, and cardiotoxicity mechanism should be strengthened in the future. CONCLUSION: Bibliometric analysis combined with a systemic review of CTD research first revealed that PP2A and CTD analogs were the knowledge base of CTD, and PP2A subunit regulation and toxic mechanism could be the frontiers of CTD.


Subject(s)
Bibliometrics , Cantharidin , Cantharidin/therapeutic use , Humans , Animals , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects
13.
Cell Death Dis ; 15(7): 497, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997271

ABSTRACT

Helicobacter pylori (HP) infection initiates and promotes gastric carcinogenesis. ONECUT2 shows promise for tumor diagnosis, prognosis, and treatment. This study explored ONECUT2's role and the specific mechanism underlying HP infection-associated gastric carcinogenesis to suggest a basis for targeting ONECUT2 as a therapeutic strategy for gastric cancer (GC). Multidimensional data supported an association between ONECUT2, HP infection, and GC pathogenesis. HP infection upregulated ONECUT2 transcriptional activity via NFκB. In vitro and in vivo experiments demonstrated that ONECUT2 increased the stemness of GC cells. ONECUT2 was also shown to inhibit PPP2R4 transcription, resulting in reduced PP2A activity, which in turn increased AKT/ß-catenin phosphorylation. AKT/ß-catenin phosphorylation facilitates ß-catenin translocation to the nucleus, initiating transcription of downstream stemness-associated genes in GC cells. HP infection upregulated the reduction of AKT and ß-catenin phosphorylation triggered by ONECUT2 downregulation via ONECUT2 induction. Clinical survival analysis indicated that high ONECUT2 expression may indicate poor prognosis in GC. This study highlights a critical role played by ONECUT2 in promoting HP infection-associated GC by enhancing cell stemness through the PPP2R4/AKT/ß-catenin signaling pathway. These findings suggest promising therapeutic strategies and potential targets for GC treatment.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Neoplastic Stem Cells , Proto-Oncogene Proteins c-akt , Stomach Neoplasms , Animals , Female , Humans , Male , Mice , beta Catenin/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stomach Neoplasms/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics
14.
Cancer Lett ; 598: 217110, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38986733

ABSTRACT

PP2A B55α, encoded by PPP2R2A, acts as a regulatory subunit of the serine/threonine phosphatase PP2A. Despite a frequent loss of heterozygosity of PPP2R2A in cases of non-small cell lung cancer (NSCLC), research on PP2A B55α's functions remains limited and controversial. To investigate the biological roles of PP2A B55α, we conducted bulk RNA-sequencing to assess the impact of PPP2R2A knockdown using two shRNAs in a NSCLC cell line. Gene set enrichment analysis (GSEA) of the RNA-sequencing data revealed significant enrichment of the epithelial-mesenchymal transition (EMT) pathway, with SNAI2 (the gene encoding Slug) emerging as one of the top candidates. Our findings demonstrate that PP2A B55α suppresses EMT, as PPP2R2A deficiency through knockdown or homozygous or hemizygous depletion promotes EMT and metastatic behavior in NSCLC cells, as evidenced by changes in EMT biomarkers, invasion and migration abilities, as well as metastasis in a tail vein assay. Mechanistically, PP2A B55α inhibits EMT by downregulating SNAI2 expression via the GSK3ß-ß-catenin pathway. Importantly, PPP2R2A deficiency also slows cell proliferation by disrupting DNA replication, particularly in PPP2R2A-/- cells. Furthermore, PPP2R2A deficiency, especially PPP2R2A-/- cells, leads to an increase in the cancer stem cell population, which correlates with enhanced resistance to chemotherapy. Overall, the decrease in PP2A B55α levels due to hemizygous/homozygous depletion heightens EMT and the metastatic or stemness/drug resistance potential of NSCLC cells despite their proliferation disadvantage. Our study highlights the significance of PP2A B55α in EMT and metastasis and suggests that targeting EMT/stemness could be a potential therapeutic strategy for treating PPP2R2A-deficient NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Protein Phosphatase 2 , Snail Family Transcription Factors , Epithelial-Mesenchymal Transition/genetics , Humans , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Cell Movement , Cell Line, Tumor , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , A549 Cells , Mice , Neoplasm Invasiveness
15.
Cell Death Dis ; 15(7): 514, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025841

ABSTRACT

Prostate cancer exhibits high prevalence and accounts for a high number of cancer-related deaths. The discovery and characterization of molecular determinants of aggressive prostate cancer represents an active area of research. The Immediate Early Response (IER) family of genes, which regulate Protein Phosphatase 2A (PP2A) activity, has emerged among the factors that influence cancer biology. Here, we show that the less studied member of this family, Immediate Early Response 5 like (IER5L), is upregulated in aggressive prostate cancer. Interestingly, the upregulation of IER5L expression exhibits a robust association with metastatic disease in prostate and is recapitulated in other cancer types. In line with this observation, IER5L silencing reduces foci formation, migration and invasion ability in a variety of human and murine prostate cancer cell lines. In vivo, using zebrafish and immunocompromised mouse models, we demonstrate that IER5L-silencing reduces prostate cancer tumor growth, dissemination, and metastasis. Mechanistically, we characterize the transcriptomic and proteomic landscapes of IER5L-silenced cells. This approach allowed us to identify DNA replication and monomeric G protein regulators as downstream programs of IER5L through a pathway that is consistent with the regulation of PP2A. In sum, we report the alteration of IER5L in prostate cancer and beyond and provide biological and molecular evidence of its contribution to tumor aggressiveness.


Subject(s)
Disease Progression , Prostatic Neoplasms , Protein Phosphatase 2 , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Humans , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Zebrafish , Cell Movement/genetics , Cell Proliferation
16.
EBioMedicine ; 106: 105232, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991381

ABSTRACT

BACKGROUND: Abdominal obesity increases the risk for non-alcoholic fatty liver disease (NAFLD), now known as metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS: To elucidate the directional cell-type level biological mechanisms underlying the association between abdominal obesity and MASLD, we integrated adipose and liver single nucleus RNA-sequencing and bulk cis-expression quantitative trait locus (eQTL) data with the UK Biobank genome-wide association study (GWAS) data using colocalization. Then we used colocalized cis-eQTL variants as instrumental variables in Mendelian randomization (MR) analyses, followed by functional validation experiments on the target genes of the cis-eQTL variants. FINDINGS: We identified 17 colocalized abdominal obesity GWAS variants, regulating 17 adipose cell-type marker genes. Incorporating these 17 variants into MR discovers a putative tissue-of-origin, cell-type-aware causal effect of abdominal obesity on MASLD consistently with multiple MR methods without significant evidence for pleiotropy or heterogeneity. Single cell data confirm the adipocyte-enriched mean expression of the 17 genes. Our cellular experiments across human adipogenesis identify risk variant -specific epigenetic and transcriptional mechanisms. Knocking down two of the 17 genes, PPP2R5A and SH3PXD2B, shows a marked decrease in adipocyte lipidation and significantly alters adipocyte function and adipogenesis regulator genes, including DGAT2, LPL, ADIPOQ, PPARG, and SREBF1. Furthermore, the 17 genes capture a characteristic MASLD expression signature in subcutaneous adipose tissue. INTERPRETATION: Overall, we discover a significant cell-type level effect of abdominal obesity on MASLD and trace its biological effect to adipogenesis. FUNDING: NIH grants R01HG010505, R01DK132775, and R01HL170604; the European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (Grant No. 802825), Academy of Finland (Grants Nos. 333021), the Finnish Foundation for Cardiovascular Research the Sigrid Jusélius Foundation and the Jane and Aatos Erkko Foundation; American Association for the Study of Liver Diseases (AASLD) Advanced Transplant Hepatology award and NIH/NIDDK (P30DK41301) Pilot and Feasibility award; NIH/NIEHS F32 award (F32ES034668); Finnish Diabetes Research Foundation, Kuopio University Hospital Project grant (EVO/VTR grants 2005-2021), the Academy of Finland grant (Contract no. 138006); Academy of Finland (Grant Nos 335443, 314383, 272376 and 266286), Sigrid Jusélius Foundation, Finnish Medical Foundation, Finnish Diabetes Research Foundation, Novo Nordisk Foundation (#NNF20OC0060547, NNF17OC0027232, NNF10OC1013354) and Government Research Funds to Helsinki University Hospital; Orion Research Foundation, Maud Kuistila Foundation, Finish Medical Foundation, and University of Helsinki.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Obesity, Abdominal , Quantitative Trait Loci , Humans , Obesity, Abdominal/genetics , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Polymorphism, Single Nucleotide , Sequence Analysis, RNA , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Cell Nucleus/metabolism , Cell Nucleus/genetics , Adipogenesis/genetics , Single-Cell Analysis , Gene Expression Regulation
17.
Nat Commun ; 15(1): 5776, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38982062

ABSTRACT

The Ser/Thr protein phosphatase 2 A (PP2A) regulates the dephosphorylation of many phosphoproteins. Substrate recognition are mediated by B regulatory subunits. Here, we report the identification of a substrate conserved motif [RK]-V-x-x-[VI]-R in FAM122A, an inhibitor of B55α/PP2A. This motif is necessary for FAM122A binding to B55α, and computational structure prediction suggests the motif, which is helical, blocks substrate docking to the same site. In this model, FAM122A also spatially constrains substrate access by occluding the catalytic subunit. Consistently, FAM122A functions as a competitive inhibitor as it prevents substrate binding and dephosphorylation of CDK substrates by B55α/PP2A in cell lysates. FAM122A deficiency in human cell lines reduces the proliferation rate, cell cycle progression, and hinders G1/S and intra-S phase cell cycle checkpoints. FAM122A-KO in HEK293 cells attenuates CHK1 and CHK2 activation in response to replication stress. Overall, these data strongly suggest that FAM122A is a short helical motif (SHeM)-dependent, substrate-competitive inhibitor of B55α/PP2A that suppresses multiple functions of B55α in the DNA damage response and in timely progression through the cell cycle interphase.


Subject(s)
Amino Acid Motifs , Interphase , Protein Phosphatase 2 , Humans , Cell Cycle Checkpoints/genetics , Cell Proliferation , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 2/metabolism , Checkpoint Kinase 2/genetics , HEK293 Cells , Phosphorylation , Protein Binding , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics
18.
Cell Rep ; 43(7): 114494, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39003739

ABSTRACT

Cell cycle progression is regulated by the orderly balance between kinase and phosphatase activities. PP2A phosphatase holoenzymes containing the B55 family of regulatory B subunits function as major CDK1-counteracting phosphatases during mitotic exit in mammals. However, the identification of the specific mitotic roles of these PP2A-B55 complexes has been hindered by the existence of multiple B55 isoforms. Here, through the generation of loss-of-function genetic mouse models for the two ubiquitous B55 isoforms (B55α and B55δ), we report that PP2A-B55α and PP2A-B55δ complexes display overlapping roles in controlling the dynamics of proper chromosome individualization and clustering during mitosis. In the absence of PP2A-B55 activity, mitotic cells display increased chromosome individualization in the presence of enhanced phosphorylation and perichromosomal loading of Ki-67. These data provide experimental evidence for a regulatory mechanism by which the balance between kinase and PP2A-B55 phosphatase activity controls the Ki-67-mediated spatial organization of the mass of chromosomes during mitosis.


Subject(s)
Ki-67 Antigen , Mitosis , Protein Phosphatase 2 , Animals , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Mice , Ki-67 Antigen/metabolism , Phosphorylation , Chromosomes, Mammalian/metabolism , Chromosomes, Mammalian/genetics , Chromosomes/metabolism
19.
Int Immunopharmacol ; 139: 112680, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39018689

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a common disease with high global morbidity and mortality. Macrophages release IL-1ß and orchestrate airway inflammation in COPD. Previously, we explored the role of a new lncRNA, LincR-PPP2R5C, in regulating Th2 cells in asthma. Here, we established a murine model of COPD and explored the roles and mechanisms by which LincR-PPP2R5C regulates IL-1ß in macrophages. LincR-PPP2R5C was highly expressed in pulmonary macrophages from COPD-like mice. LincR-PPP2R5C deficiency ameliorated emphysema and pulmonary inflammation, as characterized by reduced IL-1ß in macrophages. Unexpectedly, in both lung tissues and macrophages, LincR-PPP2R5C deficiency decreased the expression of the IL-1ß protein but not the IL-1ß mRNA. Furthermore, we found that LincR-PPP2R5C deficiency increased the level of ubiquitinated IL-1ß in macrophages, which was mediated by PP2A activity. Targeting PP2A with FTY720 decreased IL-1ß and improved COPD. In conclusion, LincR-PPP2R5C regulates IL-1ß ubiquitination by affecting PP2A activity in macrophages, contributing to the airway inflammation and emphysema in a murine model of COPD. PP2A and IL-1ß ubiquitination in macrophages might be new therapeutic avenues for COPD therapy.


Subject(s)
Disease Models, Animal , Interleukin-1beta , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive , RNA, Long Noncoding , Ubiquitination , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Interleukin-1beta/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Protein Phosphatase 2/metabolism , Macrophages/immunology , Macrophages/metabolism , Humans , Male , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/immunology , Pulmonary Emphysema/pathology , Pulmonary Emphysema/genetics , Lung/pathology , Lung/immunology , Mice, Knockout
20.
Virology ; 597: 110143, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38917692

ABSTRACT

Merkel Cell Carcinoma (MCC) is a rare neuroendocrine skin cancer. In our previous work, we decoded genes specifically deregulated by MCPyV early genes as opposed to other polyomaviruses and established functional importance of NDRG1 in inhibiting cellular proliferation and migration in MCC. In the present work, we found the SET protein, (I2PP2A, intrinsic inhibitor of PP2A) upstream of NDRG1 which was modulated by MCPyV early genes, both in hTERT-HK-MCPyV and MCPyV-positive (+) MCC cell lines. Additionally, MCC dermal tumour nodule tissues showed strong SET expression. Inhibition of the SET-PP2A interaction in hTERT-HK-MCPyV using the small molecule inhibitor, FTY720, increased NDRG1 expression and inhibited cell cycle regulators, cyclinD1 and CDK2. SET inhibition by shRNA and FTY720 also decreased cell proliferation and colony formation in MCPyV(+) MCC cells. Overall, these results pave a path for use of drugs targeting SET protein for the treatment of MCC.


Subject(s)
Carcinoma, Merkel Cell , Cell Movement , Cell Proliferation , Merkel cell polyomavirus , Protein Phosphatase 2 , Humans , Merkel cell polyomavirus/physiology , Merkel cell polyomavirus/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/genetics , Carcinoma, Merkel Cell/virology , Carcinoma, Merkel Cell/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Fingolimod Hydrochloride/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Histone Chaperones/metabolism , Histone Chaperones/genetics , Polyomavirus Infections/virology , Skin Neoplasms/virology , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL