Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21.981
Filter
1.
Methods Mol Biol ; 2854: 153-170, 2025.
Article in English | MEDLINE | ID: mdl-39192127

ABSTRACT

cGAS is a key cytosolic dsDNA receptor that senses viral infection and elicits interferon production through the cGAS-cGAMP-STING axis. cGAS is activated by dsDNA from viral and bacterial origins as well as dsDNA leaked from damaged mitochondria and nucleus. Eventually, cGAS activation launches the cell into an antiviral state to restrict the replication of both DNA and RNA viruses. Throughout the long co-evolution, viruses devise many strategies to evade cGAS detection or suppress cGAS activation. We recently reported that the Dengue virus protease NS2B3 proteolytically cleaves human cGAS in its N-terminal region, effectively reducing cGAS binding to DNA and consequent production of the second messenger cGAMP. Several other RNA viruses likely adopt the cleavage strategy. Here, we describe a protocol for the purification of recombinant human cGAS and Dengue NS2B3 protease, as well as the in vitro cleavage assay.


Subject(s)
Dengue Virus , Nucleotidyltransferases , Viral Nonstructural Proteins , Humans , Viral Nonstructural Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Proteolysis , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Nucleotides, Cyclic/metabolism , Dengue/virology , Dengue/metabolism
2.
Gene ; 932: 148907, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39218412

ABSTRACT

Oculocutaneous albinism (OCA) is a genetically heterogeneous group of autosomal recessive disorders, which presents with decreased or absent pigmentation in the hair, skin, and eyes. OCA1, as a subtype of OCA, is caused by mutations in the tyrosinase gene (TYR). In this study, we performed in vitro functional analysis of eight TYR variants (one frameshift variant: c.929dupC (p.Arg311Lysfs*7); seven missense variants: c.896G>A (p.Arg299His), c.1234C>A (p.Pro412Thr), c.1169A>G (p.His390Arg), c.937C>A (p.Pro313Thr), c.636A>T (p.Arg212Ser), c.623 T>G (p.Leu208Arg), c.1325C>A (p.Ser442Tyr)) identified in Chinese OCA families. TYR plasmids were transfected into HEK 293 T cells to explore the effects of TYR variants on their processing, protein expression, activity, and degradation. The results showed that all eight variants caused TYR to be retained in the endoplasmic reticulum (ER), processing was blocked, and TYR activity almost disappeared; the frameshift variant caused the size of the TYR protein to be reduced by about 30KD, and the protein expression of the remaining seven missense variants was reduced; the ER-associated degradation (ERAD) pathway mediates the degradation of TYR variants that occur on the Tyrosinase copper-binding domain, while the degradation of TYR variants that are not located on that domain may be mediated by a new degradation pathway--ER-to-lysosome-associated degradation (ERLAD). In summary, TYR variants affected their protein processing and activity, and may also induce ER stress and trigger degradation through the ERLAD pathway in addition to the ERAD degradation pathway, providing new insights into the potential pathogenic mechanism for OCA1 caused by TYR variants.


Subject(s)
Albinism, Oculocutaneous , Endoplasmic Reticulum-Associated Degradation , Endoplasmic Reticulum , Monophenol Monooxygenase , Humans , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , HEK293 Cells , Albinism, Oculocutaneous/genetics , Albinism, Oculocutaneous/metabolism , Endoplasmic Reticulum/metabolism , Mutation, Missense , Asian People/genetics , Female , Male , Proteolysis , Pedigree , East Asian People
3.
Proc Natl Acad Sci U S A ; 121(41): e2414618121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39361641

ABSTRACT

The transcription factor E2F1 serves as a regulator of the cell cycle and promotes cell proliferation. It is highly expressed in cancer tissues and contributes to their malignant transformation. Degradation by the ubiquitin-proteasome system may help to prevent such overexpression of E2F1 and thereby to suppress carcinogenesis. A detailed understanding of the mechanisms underlying E2F1 degradation may therefore inform the development of new cancer treatments. We here identified SCFFBXW7 as a ubiquitin ligase for E2F1 by comprehensive analysis. We found that phosphorylation of E2F1 at serine-403 promotes its binding to FBXW7 (F-box/WD repeat-containing protein 7) followed by its ubiquitination and degradation. Furthermore, calcineurin, a Ca2+/calmodulin-dependent serine-threonine phosphatase, was shown to stabilize E2F1 by mediating its dephosphorylation at serine-403 and thereby preventing FBXW7 binding. Treatment of cells with Ca2+ channel blockers resulted in downregulation of both E2F1 protein and the expression of E2F1 target genes, whereas treatment with the Ca2+ ionophore ionomycin induced upregulation of E2F1. Finally, the calcineurin inhibitor FK506 attenuated xenograft tumor growth in mice in association with downregulation of E2F1 in the tumor tissue. Impairment of the balance between the opposing actions of FBXW7 and calcineurin in the regulation of E2F1 abundance may therefore play an important role in carcinogenesis.


Subject(s)
Calcineurin , E2F1 Transcription Factor , F-Box-WD Repeat-Containing Protein 7 , F-Box-WD Repeat-Containing Protein 7/metabolism , F-Box-WD Repeat-Containing Protein 7/genetics , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Calcineurin/metabolism , Calcineurin/genetics , Humans , Phosphorylation , Animals , Mice , Ubiquitination , Protein Binding , HEK293 Cells , Tacrolimus/pharmacology , Cell Line, Tumor , Protein Stability , Proteolysis
4.
ACS Chem Neurosci ; 15(19): 3434-3436, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39354828

ABSTRACT

Recent advancements in Alzheimer's disease (AD) research have led to the approval of lecanemab and donanemab, highlighting the effectiveness of amyloid-beta (Aß) degradation as a treatment for AD. The prospect of small molecule Aß degraders as a potential treatment, which utilizes emerging targeted protein degradation technology, is exciting, given their ability to address some of the limitations of current therapies and their promising future in AD treatment. Despite facing challenges, these degraders are poised to become a future treatment option, harnessing scientific breakthroughs for more targeted and effective AD therapy.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Proteolysis , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Proteolysis/drug effects , Amyloid beta-Peptides/metabolism , Animals , Drug Development/methods
5.
Nat Commun ; 15(1): 8695, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379374

ABSTRACT

Targeted protein degradation has emerged as a novel therapeutic modality to treat human diseases by utilizing the cell's own disposal systems to remove protein target. Significant clinical benefits have been observed for degrading many intracellular proteins. Recently, the degradation of extracellular proteins in the lysosome has been developed. However, there have been limited successes in selectively degrading protein targets in disease-relevant cells or tissues, which would greatly enhance the development of precision medicine. Additionally, most degraders are not readily available due to their complexity. We report a class of easily accessible Folate Receptor TArgeting Chimeras (FRTACs) to recruit the folate receptor, primarily expressed on malignant cells, to degrade extracellular soluble and membrane cancer-related proteins in vitro and in vivo. Our results indicate that FRTAC is a general platform for developing more precise and effective chemical probes and therapeutics for the study and treatment of cancers.


Subject(s)
Neoplasms , Proteolysis , Humans , Animals , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Cell Line, Tumor , Mice , Folic Acid/metabolism , Lysosomes/metabolism , Female , Xenograft Model Antitumor Assays , Folate Receptor 1/metabolism , Folate Receptor 1/genetics , Mice, Nude , Folate Receptors, GPI-Anchored/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics
6.
Commun Biol ; 7(1): 1280, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379486

ABSTRACT

D-site binding protein, DBP, is a clock-controlled transcription factor and drives daily rhythms of physiological processes through the regulation of an array of genes harboring a DNA binding motif, D-box. DBP protein levels show a circadian oscillation with an extremely robust peak/trough ratio, but it is elusive how the temporal pattern is regulated by post-translational regulation. In this study, we show that DBP protein levels are down-regulated by the ubiquitin-proteasome pathway. Analysis using 19 dominant-negative forms of E2 enzymes have revealed that UBE2G1 and UBE2T mediate the degradation of DBP. A proteomic analysis of DBP-interacting proteins and database screening have identified Tumor necrosis factor Receptor-Associated Factor 7 (TRAF7), a RING-type E3 ligase, that forms a complex with UBE2G1 and/or UBE2T. Ubiquitination analysis have revealed that TRAF7 enhances K48-linked polyubiquitination of DBP in cultured cells. Overexpression of TRAF7 down-regulates DBP protein level, while knockdown of TRAF7 up-regulates DBP in cultured cells. Knockout of TRAF7 in NIH3T3 cells have revealed that TRAF7 mediates the time-of-the-day-dependent regulation of DBP levels. Furthermore, TRAF7 has a period-shortening effect on the cellular clock. Together, TRAF7 plays an important role in circadian clock oscillation through destabilization of DBP.


Subject(s)
Circadian Rhythm , Ubiquitination , Animals , Mice , NIH 3T3 Cells , Circadian Rhythm/genetics , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Proteolysis , Circadian Clocks/genetics
8.
Biomed Res Int ; 2024: 7919329, 2024.
Article in English | MEDLINE | ID: mdl-39371248

ABSTRACT

Backgrounds: Various physiological functions and reaction cascades, as well as disease progression in the living systems, are controlled by the activity of specific proteolytic enzymes. We conducted the study to evaluate protease activity by assessing peptide fragments from either conserved or labeled red blood cells (RBCs) with aminofluorescein (AF) in the reaction media. Methods: RBCs were incubated in media containing trypsin. Subsequently, the concentration of peptide fragments in the reaction media, resulted by the digestion with trypsin from conserved cells, was estimated by 3-(4-carboxybenzoyl)quinoline-2-carboxaldehyde (CBQCA) as an amine-reactive fluorogenic reagent. In a second approach, we conjugated AF to the conserved RBCs and then exposed AF-labeled RBCs to trypsin. This was followed by directly measuring the fluorescence intensity (FI) in the reaction media to estimate the concentration of AF-labeled peptide fragments resulting from the enzyme's activity. Results: Show a concentration- and time-dependent increase in FIs, reflecting the activity of trypsin as a proteolytic enzyme. The FIs increased significantly by 4 to 5 folds in samples treated with different enzyme concentrations, and by over 11 folds after 2 h incubation in media containing a 50 µL trypsin, as evidenced by CBQCA assays. Conclusion: These fast and affordable approaches could be applied with high reliability for the general estimation of protease activity in samples and customized for diagnostic purposes and prognostic evaluation in various diseases.


Subject(s)
Erythrocytes , Fluoresceins , Proteolysis , Trypsin , Humans , Erythrocytes/metabolism , Trypsin/metabolism , Trypsin/chemistry , Fluoresceins/chemistry , Peptide Fragments/metabolism , Peptide Fragments/chemistry , Fluorescent Dyes/chemistry
9.
Proc Natl Acad Sci U S A ; 121(42): e2406936121, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39388269

ABSTRACT

Kirsten rat sarcoma virus (KRAS) mutation is associated with malignant tumor transformation and drug resistance. However, the development of clinically effective targeted therapies for KRAS-mutant cancer has proven to be a formidable challenge. Here, we report that tripartite motif-containing protein 21 (TRIM21) functions as a target of extracellular signal-regulated kinase 2 (ERK2) in KRAS-mutant colorectal cancer (CRC), contributing to regorafenib therapy resistance. Mechanistically, TRIM21 directly interacts with and ubiquitinates v-myc avian myelocytomatosis viral oncogene homolog (c-Myc) at lysine 148 (K148) via K63-linkage, enabling c-Myc to be targeted to the autophagy machinery for degradation, ultimately resulting in the downregulation of enolase 2 expression and inhibition of glycolysis. However, mutant KRAS (KRAS/MT)-driven mitogen-activated protein kinase (MAPK) signaling leads to the phosphorylation of TRIM21 (p-TRIM21) at Threonine 396 (T396) by ERK2, disrupting the interaction between TRIM21 and c-Myc and thereby preventing c-Myc from targeting autophagy for degradation. This enhances glycolysis and contributes to regorafenib resistance. Clinically, high p-TRIM21 (T396) is associated with an unfavorable prognosis. Targeting TRIM21 to disrupt KRAS/MT-driven phosphorylation using the antidepressant vilazodone shows potential for enhancing the efficacy of regorafenib in treating KRAS-mutant CRC in preclinical models. These findings are instrumental for KRAS-mutant CRC treatment aiming at activating TRIM21-mediated selective autophagic degradation of c-Myc.


Subject(s)
Autophagy , Colorectal Neoplasms , Phenylurea Compounds , Proto-Oncogene Proteins c-myc , Proto-Oncogene Proteins p21(ras) , Pyridines , Ribonucleoproteins , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Autophagy/drug effects , Phenylurea Compounds/pharmacology , Animals , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Mice , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Xenograft Model Antitumor Assays , Proteolysis/drug effects , Mutation , Mice, Nude
10.
Commun Biol ; 7(1): 1282, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379572

ABSTRACT

Inhibitors of the integrated stress response (ISR) have been used to explore the potential beneficial effects of reducing the activation of this pathway in diseases. As the ISR is in essence a protective response, there is, however, a risk that inhibition may compromise the cell's ability to restore protein homeostasis. Here, we show that the experimental compound ISRIB impairs degradation of proteins by the ubiquitin-proteasome system (UPS) during proteotoxic stress in the cytosolic, but not nuclear, compartment. Accumulation of a UPS reporter substrate that is intercepted by ribosome quality control was comparable to the level observed after blocking the UPS with a proteasome inhibitor. Consistent with impairment of the cytosolic UPS, ISRIB treatment caused an accumulation of polyubiquitylated and detergent insoluble defective ribosome products (DRiPs) in the presence of puromycin. Our data suggest that the persistent protein translation during proteotoxic stress in the absence of a functional ISR increases the pool of DRiPs, thereby hindering the efficient clearance of cytosolic substrates by the UPS.


Subject(s)
Proteasome Endopeptidase Complex , Stress, Physiological , Ubiquitin , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Stress, Physiological/drug effects , Humans , Ribosomes/metabolism , Ribosomes/drug effects , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Puromycin/pharmacology , Cytosol/metabolism , HeLa Cells , Acetamides , Cyclohexylamines
11.
Life Sci Alliance ; 7(12)2024 Dec.
Article in English | MEDLINE | ID: mdl-39389782

ABSTRACT

The human CTLH/GID (hGID) complex emerged as an important E3 ligase regulating multiple cellular processes, including cell cycle progression and metabolism. However, the range of biological functions controlled by hGID remains unexplored. Here, we used proximity-dependent biotinylation (BioID2) to identify proteins interacting with the hGID complex, among them, substrate candidates that bind GID4 in a pocket-dependent manner. Biochemical and cellular assays revealed that the hGIDGID4 E3 ligase binds and ubiquitinates ARHGAP11A, thereby targeting this RhoGAP for proteasomal degradation. Indeed, GID4 depletion or impeding the GID4 substrate binding pocket with the PFI-7 inhibitor stabilizes ARHGAP11A protein amounts, although it carries no functional N-terminal degron. Interestingly, GID4 inactivation impairs cell motility and directed cell movement by increasing ARHGAP11A levels at the cell periphery, where it inactivates RhoA. Together, we identified a wide range of hGIDGID4 E3 ligase substrates and uncovered a unique function of the hGIDGID4 E3 ligase regulating cell migration by targeting ARHGAP11A.


Subject(s)
Cell Movement , GTPase-Activating Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Humans , GTPase-Activating Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Protein Binding , HEK293 Cells , Proteolysis , rhoA GTP-Binding Protein/metabolism
12.
Mol Cancer ; 23(1): 227, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39390584

ABSTRACT

Breast cancer remains a significant health concern, with triple-negative breast cancer (TNBC) being an aggressive subtype with poor prognosis. Epithelial-mesenchymal transition (EMT) is important in early-stage tumor to invasive malignancy progression. Snail, a central EMT component, is tightly regulated and may be subjected to proteasomal degradation. We report a novel proteasomal independent pathway involving chaperone-mediated autophagy (CMA) in Snail degradation, mediated via its cytosolic interaction with HSC70 and lysosomal targeting, which prevented its accumulation in luminal-type breast cancer cells. Conversely, Snail predominantly localized to the nucleus, thus evading CMA-mediated degradation in TNBC cells. Starvation-induced CMA activation downregulated Snail in TNBC cells by promoting cytoplasmic translocation. Evasion of CMA-mediated Snail degradation induced EMT, and enhanced metastatic potential of luminal-type breast cancer cells. Our findings elucidate a previously unrecognized role of CMA in Snail regulation, highlight its significance in breast cancer, and provide a potential therapeutic target for clinical interventions.


Subject(s)
Chaperone-Mediated Autophagy , Epithelial-Mesenchymal Transition , Lysosomes , Protein Stability , Snail Family Transcription Factors , Snail Family Transcription Factors/metabolism , Humans , Female , Cell Line, Tumor , Lysosomes/metabolism , Neoplasm Metastasis , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Proteolysis , HSC70 Heat-Shock Proteins/metabolism , Mice , Autophagy
13.
Nat Commun ; 15(1): 8731, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384759

ABSTRACT

Many disease-causing proteins have multiple pathogenic mechanisms, and conventional inhibitors struggle to reliably disrupt more than one. Targeted protein degradation (TPD) can eliminate the protein, and thus all its functions, by directing a cell's protein turnover machinery towards it. Two established strategies either engage catalytic E3 ligases or drive uptake towards the endolysosomal pathway. Here we describe CYpHER (CatalYtic pH-dependent Endolysosomal delivery with Recycling) technology with potency and durability from a catalytic mechanism that shares the specificity and straightforward modular design of endolysosomal uptake. By bestowing pH-dependent release on the target engager and using the rapid-cycling transferrin receptor as the uptake receptor, CYpHER induces endolysosomal delivery of surface and extracellular targets while re-using drug, potentially yielding increased potency and reduced off-target tissue exposure risks. The TfR-based approach allows targeting to tumors that overexpress this receptor and offers the potential for transport to the CNS. CYpHER function was demonstrated in vitro with EGFR and PD-L1, and in vivo with EGFR in a model of EGFR-driven non-small cell lung cancer.


Subject(s)
ErbB Receptors , Lysosomes , Proteolysis , Receptors, Transferrin , Humans , Proteolysis/drug effects , Receptors, Transferrin/metabolism , Animals , Cell Line, Tumor , ErbB Receptors/metabolism , Lysosomes/metabolism , Mice , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Hydrogen-Ion Concentration , B7-H1 Antigen/metabolism , Female , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Catalysis , Endosomes/metabolism , Xenograft Model Antitumor Assays
14.
Cell Commun Signal ; 22(1): 482, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39385148

ABSTRACT

The Wnt/ß-catenin signaling pathway is crucial for embryonic development and adult tissue homeostasis. Dysregulation of Wnt signaling is linked to various developmental anomalies and diseases, notably cancer. Although numerous regulators of the Wnt signaling pathway have been identified, their precise function during mouse embryo development remains unclear. Here, we revealed that TMEM132A is a crucial regulator of canonical Wnt/ß-catenin signaling in mouse development. Mouse embryos lacking Tmem132a displayed a range of malformations, including open spina bifida, caudal truncation, syndactyly, and renal defects, similar to the phenotypes of Wnt/ß-catenin mutants. Tmem132a knockdown in cultured cells suppressed canonical Wnt/ß-catenin signaling. In developing mice, loss of Tmem132a also led to diminished Wnt/ß-catenin signaling. Mechanistically, we showed that TMEM132A interacts with the Wnt co-receptor LRP6, thereby stabilizing it and preventing its lysosomal degradation. These findings shed light on a novel role for TMEM132A in regulating LRP6 stability and canonical Wnt/ß-catenin signaling during mouse embryo development. This study provides valuable insights into the molecular intricacies of the Wnt signaling pathway. Further research may deepen our understanding of Wnt pathway regulation and offer its potential therapeutic applications.


Subject(s)
Embryonic Development , Low Density Lipoprotein Receptor-Related Protein-6 , Membrane Proteins , Wnt Signaling Pathway , Animals , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Embryonic Development/genetics , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , Protein Stability , Humans , beta Catenin/metabolism , beta Catenin/genetics , Proteolysis
15.
Science ; 385(6716): eadk2536, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39325906

ABSTRACT

Harnessing the microbiome to benefit human health requires an initial step in determining the identity and function of causative microorganisms that affect specific host physiological functions. We show a functional screen of the bacterial microbiota from mice with low intestinal immunoglobulin A (IgA) levels; we identified a Gram-negative bacterium, proposed as Tomasiella immunophila, that induces and degrades IgA in the mouse intestine. Mice harboring T. immunophila are susceptible to infections and show poor mucosal repair. T. immunophila is auxotrophic for the bacterial cell wall amino sugar N-acetylmuramic acid. It delivers immunoglobulin-degrading proteases into outer membrane vesicles that preferentially degrade rodent antibodies with kappa but not lambda light chains. This work indicates a role for symbionts in immunodeficiency, which might be applicable to human disease.


Subject(s)
Bacteria , Gastrointestinal Microbiome , Host Microbial Interactions , Immunoglobulin A , Immunologic Deficiency Syndromes , Intestinal Mucosa , Animals , Mice , Immunoglobulin A/metabolism , Immunologic Deficiency Syndromes/microbiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Symbiosis , Bacteria/classification , Bacteria/enzymology , Bacteria/isolation & purification , Proteolysis , Host Microbial Interactions/immunology
16.
Science ; 385(6716): 1418-1420, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39325916
17.
J Transl Med ; 22(1): 852, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39304928

ABSTRACT

BACKGROUND: The syncytiotrophoblast (SCT) layer in the placenta serves as a crucial physical barrier separating maternal-fetal circulation, facilitating essential signal and substance exchange between the mother and fetus. Any abnormalities in its formation or function can result in various maternal syndromes, such as preeclampsia. The transition of proliferative villous cytotrophoblasts (VCT) from the mitotic cell cycle to the G0 phase is a prerequisite for VCT differentiation and their fusion into SCT. The imprinting gene P57Kip2, specifically expressed in intermediate VCT capable of fusion, plays a pivotal role in driving this key event. Moreover, aberrant expression of P57Kip2 has been linked to pathological placental conditions and adverse fetal outcomes. METHODS: Validation of STK40 interaction with P57Kip2 using rigid molecular simulation docking and co-immunoprecipitation. STK40 expression was modulated by lentivirus in BeWo cells, and the effect of STK40 on trophoblast fusion was assessed by real-time quantitative PCR, western blot, immunofluorescence, and cell viability and proliferation assays. Co-immunoprecipitation, transcriptome sequencing, and western blot were used to determine the potential mechanisms by which STK40 regulates P57Kip2. RESULTS: In this study, STK40 has been identified as a novel interacting protein with P57Kip2, and its expression is down-regulated during the fusion process of trophoblast cells. Overexpressing STK40 inhibited cell fusion in BeWo cells while stimulating mitotic cell cycle activity. Further experiments indicated that this effect is attributed to its specific binding to the CDK-binding and the Cyclin-binding domains of P57Kip2, mediating the E3 ubiquitin ligase COP1-mediated ubiquitination and degradation of P57Kip2. Moreover, abnormally high expression of STK40 might significantly contribute to the occurrence of preeclampsia. CONCLUSIONS: This study offers new insights into the role of STK40 in regulating the protein-level homeostasis of P57Kip2 during placental development.


Subject(s)
Cell Fusion , Cyclin-Dependent Kinase Inhibitor p57 , Protein Serine-Threonine Kinases , Trophoblasts , Ubiquitin-Protein Ligases , Ubiquitination , Female , Humans , Pregnancy , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteolysis , Trophoblasts/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
18.
Sci Adv ; 10(39): eadq0187, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39331704

ABSTRACT

The period of circadian clocks is maintained at close to 24 hours over a broad range of physiological temperatures due to temperature compensation of period length. Here, we show that the quantitative control of the core clock proteins TIMING OF CAB EXPRESSION 1 [TOC1; also known as PSEUDO-RESPONSE REGULATOR 1 (PRR1)] and PRR5 is crucial for temperature compensation in Arabidopsis thaliana. The prr5 toc1 double mutant has a shortened period at higher temperatures, resulting in weak temperature compensation. Low ambient temperature reduces amounts of PRR5 and TOC1. In low-temperature conditions, PRR5 and TOC1 interact with LOV KELCH PROTEIN 2 (LKP2), a component of the E3 ubiquitin ligase Skp, Cullin, F-box (SCF) complex. The lkp2 mutations attenuate low temperature-induced decrease of PRR5 and TOC1, and the mutants display longer period only at lower temperatures. Our findings reveal that the circadian clock maintains its period length despite ambient temperature fluctuations through temperature- and LKP2-dependent control of PRR5 and TOC1 abundance.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Circadian Clocks , Cold Temperature , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Circadian Clocks/physiology , Mutation , Transcription Factors/metabolism , Transcription Factors/genetics , Proteolysis
19.
Dev Growth Differ ; 66(7): 384-393, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39305158

ABSTRACT

The development of new technologies opens new avenues in the research field. Gene knockout is a key method for analyzing gene function in mice. Currently, conditional gene knockout strategies are employed to examine temporal and spatial gene function. However, phenotypes are sometimes not observed because of the time required for depletion due to the long half-life of the target proteins. Protein knockdown using an improved auxin-inducible degron system, AID2, overcomes such difficulties owing to rapid and efficient target depletion. We observed depletion of AID-tagged proteins within a few to several hours by a simple intraperitoneal injection of the auxin analog, 5-Ph-IAA, which is much shorter than the time required for target depletion using conditional gene knockout. Importantly, the loss of protein is reversible, making protein knockdown useful to measure the effects of transient loss of protein function. Here, we also established several mouse lines useful for AID2-medicated protein knockdown, which include knock-in mouse lines in the ROSA26 locus; one expresses TIR1(F74G), and the other is the reporter expressing AID-mCherry. We also established a germ-cell-specific TIR1 line and confirmed the protein knockdown specificity. In addition, we introduced an AID tag to an endogenous protein, DCP2 via the CAS9-mediated gene editing method. We confirmed that the protein was effectively eliminated by TIR1(F74G), which resulted in the similar phenotype observed in knockout mouse within 20 h.


Subject(s)
Indoleacetic Acids , Animals , Mice , Indoleacetic Acids/pharmacology , Indoleacetic Acids/metabolism , Proteolysis/drug effects , Gene Knockdown Techniques , Degrons
20.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337555

ABSTRACT

The discovery of the lysosome, a major cytoplasmic organelle, represents a breakthrough in the understanding of intracellular protein degradation processes-proteolysis [...].


Subject(s)
Lysosomes , Peptide Hydrolases , Protease Inhibitors , Lysosomes/metabolism , Humans , Peptide Hydrolases/metabolism , Protease Inhibitors/pharmacology , Protease Inhibitors/metabolism , Animals , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL