Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.546
Filter
1.
Nat Commun ; 15(1): 4883, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849395

ABSTRACT

The human methyltransferase and transcriptional coactivator MLL4 and its paralog MLL3 are frequently mutated in cancer. MLL4 and MLL3 monomethylate histone H3K4 and contain a set of uncharacterized PHD fingers. Here, we report a novel function of the PHD2 and PHD3 (PHD2/3) fingers of MLL4 and MLL3 that bind to ASXL2, a component of the Polycomb repressive H2AK119 deubiquitinase (PR-DUB) complex. The structure of MLL4 PHD2/3 in complex with the MLL-binding helix (MBH) of ASXL2 and mutational analyses reveal the molecular mechanism which is conserved in homologous ASXL1 and ASXL3. The native interaction of the Trithorax MLL3/4 complexes with the PR-DUB complex in vivo depends solely on MBH of ASXL1/2, coupling the two histone modifying activities. ChIP-seq analysis in embryonic stem cells demonstrates that MBH of ASXL1/2 is required for the deubiquitinase BAP1 recruitment to MLL4-bound active enhancers. Our findings suggest an ASXL1/2-dependent functional link between the MLL3/4 and PR-DUB complexes.


Subject(s)
DNA-Binding Proteins , Histone-Lysine N-Methyltransferase , Protein Binding , Repressor Proteins , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Humans , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Enhancer Elements, Genetic , HEK293 Cells , PHD Zinc Fingers , Histones/metabolism
2.
Cancer Immunol Immunother ; 73(8): 156, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834869

ABSTRACT

BACKGROUND: Ubiquitin-specific proteases family is crucial to host immunity against pathogens. However, the correlations between USP21 and immunosurveillance and immunotherapy for colorectal cancer (CRC) have not been reported. METHODS: The differential expression of USP21 between CRC tissues and normal tissues was analyzed using multiple public databases. Validation was carried out in clinical samples through qRT-PCR and IHC. The correlation between USP21 and the prognosis, as well as clinical pathological characteristics of CRC patients, was investigated. Moreover, cell models were established to assess the influence of USP21 on CRC growth and progression, employing CCK-8 assays, colony formation assays, and wound-healing assays. Subsequently, gene set variation analysis (GSVA) was used to explore the potential biological functions of USP21 in CRC. The study also examined the impact of USP21 on cytokine levels and immune cell infiltration in the tumor microenvironment (TME). Finally, the effect of USP21 on the response to immunotherapy and chemotherapy in CRC was analyzed. RESULTS: The expression of USP21 was significantly upregulated in CRC. High USP21 is correlated with poor prognosis in CRC patients and facilitates the proliferation and migration capacities of CRC cells. GSVA indicated an association between low USP21 and immune activation. Moreover, low USP21 was linked to an immune-activated TME, characterized by high immune cell infiltration. Importantly, CRC with low USP21 exhibited higher tumor mutational burden, high PD-L1 expression, and better responsiveness to immunotherapy and chemotherapeutic drugs. CONCLUSION: This study revealed the role of USP21 in TME, response to therapy, and clinical prognosis in CRC, which provided novel insights for the therapeutic application in CRC.


Subject(s)
Colorectal Neoplasms , Tumor Microenvironment , Ubiquitin Thiolesterase , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Tumor Microenvironment/immunology , Prognosis , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Male , Female , Cell Proliferation , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Middle Aged , Immunotherapy/methods
3.
Cell Mol Life Sci ; 81(1): 247, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38829550

ABSTRACT

BACKGROUND: The high degree of intratumoral genomic heterogeneity is a major obstacle for glioblastoma (GBM) tumors, one of the most lethal human malignancies, and is thought to influence conventional therapeutic outcomes negatively. The proneural-to-mesenchymal transition (PMT) of glioma stem cells (GSCs) confers resistance to radiation therapy in glioblastoma patients. POLD4 is associated with cancer progression, while the mechanisms underlying PMT and tumor radiation resistance have remained elusive. METHOD: Expression and prognosis of the POLD family were analyzed in TCGA, the Chinese Glioma Genome Atlas (CGGA) and GEO datasets. Tumorsphere formation and in vitro limiting dilution assay were performed to investigate the effect of UCHL3-POLD4 on GSC self-renewal. Apoptosis, TUNEL, cell cycle phase distribution, modification of the Single Cell Gel Electrophoresis (Comet), γ-H2AX immunofluorescence, and colony formation assays were conducted to evaluate the influence of UCHL3-POLD4 on GSC in ionizing radiation. Coimmunoprecipitation and GST pull-down assays were performed to identify POLD4 protein interactors. In vivo, intracranial xenograft mouse models were used to investigate the molecular effect of UCHL3, POLD4 or TCID on GCS. RESULT: We determined that POLD4 was considerably upregulated in MES-GSCs and was associated with a meagre prognosis. Ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, is a bona fide deubiquitinase of POLD4 in GSCs. UCHL3 interacted with, depolyubiquitinated, and stabilized POLD4. Both in vitro and in vivo assays indicated that targeted depletion of the UCHL3-POLD4 axis reduced GSC self-renewal and tumorigenic capacity and resistance to IR treatment by impairing homologous recombination (HR) and nonhomologous end joining (NHEJ). Additionally, we proved that the UCHL3 inhibitor TCID induced POLD4 degradation and can significantly enhance the therapeutic effect of IR in a gsc-derived in situ xenograft model. CONCLUSION: These findings reveal a new signaling axis for GSC PMT regulation and highlight UCHL3-POLD4 as a potential therapeutic target in GBM. TCID, targeted for reducing the deubiquitinase activity of UCHL3, exhibited significant synergy against MES GSCs in combination with radiation.


Subject(s)
Neoplastic Stem Cells , Radiation Tolerance , Ubiquitin Thiolesterase , Humans , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Radiation Tolerance/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Animals , Mice , Cell Line, Tumor , Glioma/pathology , Glioma/genetics , Glioma/radiotherapy , Glioma/metabolism , Apoptosis/genetics , Apoptosis/radiation effects , Ubiquitination , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Mice, Nude , Phenotype , Gene Expression Regulation, Neoplastic , Prognosis
4.
Arq Neuropsiquiatr ; 82(5): 1-10, 2024 May.
Article in English | MEDLINE | ID: mdl-38843846

ABSTRACT

BACKGROUND: The diagnosis of Hansen disease (HD) can be difficult when acid-fast bacilli are not detected in the patient's skin sample. OBJECTIVE: To demonstrate that detailed morphological analysis of nonspecific inflammatory and/or noninflammatory alterations in dermal nerves as well as skin adnexa in leprosy-suspected biopsy samples could improve the efficacy of histopathological diagnosis. METHODS: Patients with one to five skin lesions were enrolled in the study and classified into three groups by skin histopathology findings: Hansen disease (HD, n = 13), other diseases (OD, n = 11), and inconclusive cases (INC, n = 11). We quantified dermal nerve damage via the nerve lesion index (NLI) and PGP9.5-immunoreactive axon quantitative index in dermal nerves (AQI). We also measured inflammatory involvement of adnexa in cutaneous samples as indirect evidence of HD. RESULTS: We observed a higher median endoneurial inflammatory infiltrate NLI (HD = 0.5; INC = 0; OD = 0; p < 0.001) and more frequent inflammatory involvement of skin adnexa in samples of the HD group compared with those of the INC and OD groups (HD = 7; INC = 1; OD = 0). However, samples from the INC and OD groups also showed inflammatory and noninflammatory damage of dermal nerves, with 2 or more kinds of alterations in nerves in the same sample (respectively: INC = in 1 and 2 samples; OD = in 3 and 5 respectively). The quantification of PGP9.5-immunoreactive axons in dermal nerves revealed no difference between the groups. CONCLUSION: A detailed morphological analysis of cutaneous nerves in lesions with a suspicion of HD enabled us to select patients with nonspecific inflammatory or non-inflammatory lesions in the dermal nerves in the INC and OD groups, so they may be clinically monitored aiming at a possible future diagnosis of the disease. These INC and OD patients cannot have the HD diagnosis definitely excluded, and HD may coexist with another disease as a comorbidity.


ANTECEDENTES: A hanseníase pode ter o seu diagnóstico histopatológico dificultado quando bacilos álcool-ácido resistentes não são encontrados nas amostras de pele dos pacientes. OBJETIVO: Demonstrar que uma análise morfológica detalhada de alterações histopatológicas dos nervos dérmicos pode aumentar a eficácia diagnóstica. MéTODOS: Foram selecionadas amostras de pele de pacientes com uma a cinco lesões suspeitas de hanseníase. Os casos selecionados foram classificados conforme achados histopatológicos: hanseníase (HD, n = 13), casos inconclusivos (INC, n = 11), e outras doenças (OD, n = 11). Quantificamos as lesões dos nervos cutâneos por meio do índice de lesão de nervos (nerve lesion index, NLI, em inglês) e do índice quantitativo de axônios (axon quantitative index, AQI, em inglês) imunorreativos a PGP9.5 nos nervos cutâneos. Também medimos o envolvimento inflamatório dos anexos em amostras de pele como evidência indireta de hanseníase. RESULTADOS: Foram observadas no grupo HD medianas mais altas do NLI com relação a infiltrados inflamatórios endoneurais (HD = 0,5; INC = 0; OD = 0; p < 0,001) e mais alta frequência de acometimento inflamatório de anexos cutâneos (HD = 7; INC = 1; OD = 0). Entretanto, as amostras dos grupos INC e OD também mostraram comprometimento inflamatório e não inflamatório dos nervos cutâneos, com 2 ou mais tipos de alterações de nervos na mesma amostra (respectivamente: INC = 1 e 2; OD = 3 e 5). Não houve diferença significativa na quantidade de axônios endoneurais imunorreativos a PGP9.5 entre os grupos. CONCLUSãO: A análise morfológica detalhada dos nervos cutâneos em lesões suspeitas de hanseníase permitiu selecionar pacientes com lesões inespecíficas inflamatórias ou não inflamatórias nos nervos dérmicos nos grupos INC e OD, para que sejam monitorados clinicamente visando um possível diagnóstico futuro da doença. Esses pacientes INC e OD não podem ter o diagnóstico de HD definitivamente excluído, e a hanseníase pode coexistir com outra doença como uma comorbidade.


Subject(s)
Immunohistochemistry , Leprosy , Skin , Humans , Male , Female , Leprosy/pathology , Leprosy/complications , Middle Aged , Adult , Skin/innervation , Skin/pathology , Biopsy , Aged , Young Adult , Ubiquitin Thiolesterase/analysis , Adolescent , Statistics, Nonparametric
5.
Sci Rep ; 14(1): 13037, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844605

ABSTRACT

The proteasome-associated deubiquitinase USP14 is a potential drug target. Using an inducible USP14 knockout system in colon cancer cells, we found that USP14 depletion impedes cellular proliferation, induces cell cycle arrest, and leads to a senescence-like phenotype. Transcriptomic analysis revealed altered gene expression related to cell division and cellular differentiation. USP14 knockout cells also exhibited changes in morphology, actin distribution, and expression of actin cytoskeletal components. Increased ubiquitin turnover was observed, offset by upregulation of polyubiquitin genes UBB and UBC. Pharmacological inhibition of USP14 with IU1 increased ubiquitin turnover but did not affect cellular growth or morphology. BioGRID data identified USP14 interactors linked to actin cytoskeleton remodeling, DNA damage repair, mRNA splicing, and translation. In conclusion, USP14 loss in colon cancer cells induces a transient quiescent cancer phenotype not replicated by pharmacologic inhibition of its deubiquitinating activity.


Subject(s)
Cell Proliferation , Cellular Senescence , Colorectal Neoplasms , Ubiquitin Thiolesterase , Humans , Cellular Senescence/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Cell Line, Tumor , Phenotype , Proteasome Endopeptidase Complex/metabolism , Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Ubiquitin/metabolism
6.
Front Immunol ; 15: 1379586, 2024.
Article in English | MEDLINE | ID: mdl-38745648

ABSTRACT

Objective: Choroidal neovascularization (CNV) represents the predominant form of advanced wet Age-related Macular Degeneration (wAMD). Macrophages play a pivotal role in the pathological progression of CNV. Meteorin-like (Metrnl), a novel cytokine known for its anti-inflammatory properties in macrophages, is the focus of our investigation into its mechanism of action and its potential to impede CNV progression. Methods: Cell viability was evaluated through CCK-8 and EdU assays following Metrnl treatment. Expression levels of inflammatory cytokines and proteins were assessed using quantitative reverse-transcription polymerase chain reaction(qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and western blot techniques. Protein-protein interactions were identified through protein mass spectrometry and co-immunoprecipitation (Co-IP). Additionally, in vivo and in vitro neovascularization models were employed to evaluate angiogenesis. Results: Our results revealed downregulated Metrnl levels in the choroid-sclera complex of CNV mice, the aqueous humor of wAMD patients, and activated macrophages. Metrnl overexpression demonstrated a reduction in pro-inflammatory cytokine production, influenced endothelial cell function, and suppressed angiogenesis in choroid explants and CNV models. Through protein mass spectrometry and Co-IP, we confirmed Metrnl binds to UCHL-1 to modulate the NF-κB signaling pathway. This interaction inhibited the transcription and expression of pro-inflammatory cytokines, ultimately suppressing angiogenesis. Conclusion: In summary, our findings indicate that Metrnl down-regulates macrophage pro-inflammatory cytokine secretion via the UCHL-1/NF-κB signaling pathway. This mechanism alleviates the inflammatory microenvironment and effectively inhibits choroidal neovascularization.


Subject(s)
Choroidal Neovascularization , NF-kappa B , Signal Transduction , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Choroidal Neovascularization/genetics , Animals , Mice , Humans , NF-kappa B/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Macrophages/metabolism , Macrophages/immunology , Choroid/metabolism , Choroid/pathology , Choroid/blood supply , Male , Wet Macular Degeneration/metabolism , Wet Macular Degeneration/genetics , Wet Macular Degeneration/pathology , Inflammation/metabolism , Cytokines/metabolism
7.
Molecules ; 29(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38731625

ABSTRACT

Upon a variety of environmental stresses, eukaryotic cells usually recruit translational stalled mRNAs and RNA-binding proteins to form cytoplasmic condensates known as stress granules (SGs), which minimize stress-induced damage and promote stress adaptation and cell survival. SGs are hijacked by cancer cells to promote cell survival and are consequently involved in the development of anticancer drug resistance. However, the design and application of chemical compounds targeting SGs to improve anticancer drug efficacy have rarely been studied. Here, we developed two types of SG inhibitory peptides (SIPs) derived from SG core proteins Caprin1 and USP10 and fused with cell-penetrating peptides to generate TAT-SIP-C1/2 and SIP-U1-Antp, respectively. We obtained 11 SG-inducing anticancer compounds from cell-based screens and explored the potential application of SIPs in overcoming resistance to the SG-inducing anticancer drug sorafenib. We found that SIPs increased the sensitivity of HeLa cells to sorafenib via the disruption of SGs. Therefore, anticancer drugs which are competent to induce SGs could be combined with SIPs to sensitize cancer cells, which might provide a novel therapeutic strategy to alleviate anticancer drug resistance.


Subject(s)
Antineoplastic Agents , Sorafenib , Stress Granules , Humans , Sorafenib/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Stress Granules/metabolism , HeLa Cells , Drug Resistance, Neoplasm/drug effects , Peptides/pharmacology , Peptides/chemistry , Cell Survival/drug effects , Ubiquitin Thiolesterase/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Cell Line, Tumor , Cell-Penetrating Peptides/pharmacology , Cell-Penetrating Peptides/chemistry
8.
J Transl Med ; 22(1): 478, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769534

ABSTRACT

BACKGROUND: Miscarriage is a frustrating complication of pregnancy that is common among women of reproductive age. Insufficient decidualization which not only impairs embryo implantation but disturbs fetomaternal immune-tolerance, has been widely regarded as a major cause of miscarriage; however, the underlying mechanisms resulting in decidual impairment are largely unknown. METHODS: With informed consent, decidual tissue from patients with spontaneous abortion or normal pregnant women was collected to detect the expression profile of UCHL1. Human endometrial stromal cells (HESCs) were used to explore the roles of UCHL1 in decidualization and dNK modulation, as well as the mechanisms involved. C57/BL6 female mice (7-10 weeks old) were used to construct pregnancy model or artificially induced decidualization model to evaluate the effect of UCHL1 on mice decidualization and pregnancy outcome. RESULTS: The Ubiquitin C-terminal hydrolase L1 (UCHL1), as a deubiquitinating enzyme, was significantly downregulated in decidua from patients with miscarriage, along with impaired decidualization and decreased dNKs. Blockage of UCHL1 led to insufficient decidualization and resultant decreased expression of cytokines CXCL12, IL-15, TGF-ß which were critical for generation of decidual NK cells (dNKs), whereas UCHL1 overexpression enhanced decidualization accompanied by increase in dNKs. Mechanistically, the promotion of UCHL1 on decidualization was dependent on its deubiquitinating activity, and intervention of UCHL1 inhibited the activation of JAK2/STAT3 signaling pathway, resulting in aberrant decidualization and decreased production of cytokines associated with dNKs modulation. Furthermore, we found that inhibition of UCHL1 also disrupted the decidualization in mice and eventually caused adverse pregnancy outcome. CONCLUSIONS: UCHL1 plays significant roles in decidualization and dNKs modulation during pregnancy in both humans and mice. Its deficiency indicates a poor pregnancy outcome due to defective decidualization, making UCHL1 a potential target for the diagnosis and treatment of miscarriage.


Subject(s)
Abortion, Spontaneous , Decidua , Killer Cells, Natural , Mice, Inbred C57BL , Ubiquitin Thiolesterase , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/deficiency , Female , Decidua/metabolism , Animals , Pregnancy , Abortion, Spontaneous/metabolism , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Adult , Mice , Stromal Cells/metabolism , Signal Transduction
9.
FASEB J ; 38(10): e23653, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38738548

ABSTRACT

Hypoxic preconditioning has been recognized as a promotive factor for accelerating cutaneous wound healing. Our previous study uncovered that exosomal lncRNA H19, derived from adipose-derived stem cells (ADSCs), plays a crucial role in orchestrating cutaneous wound healing. Herein, we aimed to explore whether there is a connection between hypoxia and ADSC-derived exosomes (ADSCs-exos) in cutaneous wound healing. Exosomes extracted from ADSCs under normoxic and hypoxic conditions were identified using transmission electron microscope (TEM) and particle size analysis. The effects of ADSCs-exos on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were evaluated by CCK-8, EdU, wound healing, and tube formation assays. Expression patterns of H19, HIF-1α, and USP22 were measured. Co-immunoprecipitation, chromatin immunoprecipitation, ubiquitination, and luciferase reporter assays were conducted to confirm the USP22/HIF-1α/H19 axis, which was further validated in a mice model of skin wound. Exosomes extracted from hypoxia-treated ADSCs (termed as H-ADSCs-exos) significantly increased cell proliferation, migration, and angiogenesis in H2O2-exposed HUVECs, and promoted cutaneous wound healing in vivo. Moreover, H-ADSCs and H-ADSCs-exos, which exhibited higher levels of H19, were found to be transcriptionally activated by HIF-1α. Mechanically, H-ADSCs carrying USP22 accounted for deubiquitinating and stabilizing HIF-1α. Additionally, H-ADSCs-exos improved cell proliferation, migration, and angiogenesis in H2O2-triggered HUVECs by activating USP22/HIF-1α axis and promoting H19 expression, which may provide a new clue for the clinical treatment of cutaneous wound healing.


Subject(s)
Exosomes , Human Umbilical Vein Endothelial Cells , Hypoxia-Inducible Factor 1, alpha Subunit , RNA, Long Noncoding , Ubiquitin Thiolesterase , Wound Healing , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Exosomes/metabolism , Humans , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Human Umbilical Vein Endothelial Cells/metabolism , Cell Proliferation , Adipose Tissue/metabolism , Adipose Tissue/cytology , Male , Up-Regulation , Stem Cells/metabolism , Cell Movement , Skin/metabolism , Cell Hypoxia , Mice, Inbred C57BL
10.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727276

ABSTRACT

In mammals, hearing loss is irreversible due to the lack of the regenerative capacity of the auditory epithelium. However, stem/progenitor cells in mammalian cochleae may be a therapeutic target for hearing regeneration. The ubiquitin proteasome system plays an important role in cochlear development and maintenance. In this study, we investigated the role of ubiquitin C-terminal hydrolase L1 (UCHL1) in the process of the transdifferentiation of auditory supporting cells (SCs) into hair cells (HCs). The expression of UCHL1 gradually decreased as HCs developed and was restricted to inner pillar cells and third-row Deiters' cells between P2 and P7, suggesting that UCHL1-expressing cells are similar to the cells with Lgr5-positive progenitors. UCHL1 expression was decreased even under conditions in which supernumerary HCs were generated with a γ-secretase inhibitor and Wnt agonist. Moreover, the inhibition of UCHL1 by LDN-57444 led to an increase in HC numbers. Mechanistically, LDN-57444 increased mTOR complex 1 activity and allowed SCs to transdifferentiate into HCs. The suppression of UCHL1 induces the transdifferentiation of auditory SCs and progenitors into HCs by regulating the mTOR pathway.


Subject(s)
Cell Transdifferentiation , Hair Cells, Auditory , Signal Transduction , TOR Serine-Threonine Kinases , Ubiquitin Thiolesterase , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Cell Transdifferentiation/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/cytology , Mice , Labyrinth Supporting Cells/metabolism , Labyrinth Supporting Cells/cytology , Indoles , Oximes
11.
J Clin Invest ; 134(9)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38690732

ABSTRACT

Epigenetic regulatory mechanisms are underappreciated, yet are critical for enteric nervous system (ENS) development and maintenance. We discovered that fetal loss of the epigenetic regulator Bap1 in the ENS lineage caused severe postnatal bowel dysfunction and early death in Tyrosinase-Cre Bap1fl/fl mice. Bap1-depleted ENS appeared normal in neonates; however, by P15, Bap1-deficient enteric neurons were largely absent from the small and large intestine of Tyrosinase-Cre Bap1fl/fl mice. Bowel motility became markedly abnormal with disproportionate loss of cholinergic neurons. Single-cell RNA sequencing at P5 showed that fetal Bap1 loss in Tyrosinase-Cre Bap1fl/fl mice markedly altered the composition and relative proportions of enteric neuron subtypes. In contrast, postnatal deletion of Bap1 did not cause enteric neuron loss or impaired bowel motility. These findings suggest that BAP1 is critical for postnatal enteric neuron differentiation and for early enteric neuron survival, a finding that may be relevant to the recently described human BAP1-associated neurodevelopmental disorder.


Subject(s)
Cell Differentiation , Enteric Nervous System , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Animals , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Mice , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Mice, Knockout , Female , Gastrointestinal Motility/genetics , Humans
12.
Medicina (Kaunas) ; 60(5)2024 May 16.
Article in English | MEDLINE | ID: mdl-38792998

ABSTRACT

Background and Objectives: This study aims to compare the neuromuscular structure of the vagina in women with posterior vaginal wall prolapse with the neuromuscular structure of the vagina in women without prolapse, to determine the difference, and to demonstrate the role of neuromuscular structure in the physiopathology of prolapse. Materials and Methods: In this prospective study, women aged between 40 and 75 years who had not undergone any vaginal surgery and had not undergone any abdominal prolapse surgery were included. Thirty-one women diagnosed with rectocele on examination were included in the study group. Thirty-one patients who underwent vaginal intervention and hysterectomy for reasons other than rectocele (colposcopy, conization, etc.) without anterior or posterior wall prolapse were included in the control group. Biopsy material was obtained from the epithelium of the posterior wall of the vagina, including the fascia that fits the Ap point. Immunohistochemical staining with Protein Gene Product 9.5 and smooth muscle α-actin was performed in the pathology laboratory. The epithelial thickness measurement and smooth muscle density parameters obtained with these immunohistochemical stainings were compared between the two groups. The collected data were analyzed using the SPSS 23 package program. p values less than 0.05 were considered statistically significant. Results: In the control group, muscle thickness and the number of nerves per mm2 of fascia were statistically significantly higher than in the study group (p < 0.05). Conclusions: We found that smooth muscle tissue and the number of nerves per mm2 of fascia were decreased in posterior vaginal wall prolapse compared to the general population. Based on the correlation coefficients, age was the parameter that most affected the degree of prolapse, followed by parity, number of live births, and number of vaginal deliveries.


Subject(s)
Actins , Vagina , Humans , Female , Middle Aged , Vagina/pathology , Adult , Prospective Studies , Aged , Actins/analysis , Uterine Prolapse/pathology , Muscle, Smooth/pathology , Immunohistochemistry/methods , Ubiquitin Thiolesterase
13.
Cell Mol Life Sci ; 81(1): 232, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780644

ABSTRACT

Ubiquitin-proteasome system dysfunction triggers α-synuclein aggregation, a hallmark of neurodegenerative diseases, such as Parkinson's disease (PD). However, the crosstalk between deubiquitinating enzyme (DUBs) and α-synuclein pathology remains unclear. In this study, we observed a decrease in the level of ubiquitin-specific protease 14 (USP14), a DUB, in the cerebrospinal fluid (CSF) of PD patients, particularly females. Moreover, CSF USP14 exhibited a dual correlation with α-synuclein in male and female PD patients. To investigate the impact of USP14 deficiency, we crossed USP14 heterozygous mouse (USP14+/-) with transgenic A53T PD mouse (A53T-Tg) or injected adeno-associated virus (AAV) carrying human α-synuclein (AAV-hα-Syn) in USP14+/- mice. We found that Usp14 deficiency improved the behavioral abnormities and pathological α-synuclein deposition in female A53T-Tg or AAV-hα-Syn mice. Additionally, Usp14 inactivation attenuates the pro-inflammatory response in female AAV-hα-Syn mice, whereas Usp14 inactivation demonstrated opposite effects in male AAV-hα-Syn mice. Mechanistically, the heterodimeric protein S100A8/A9 may be the downstream target of Usp14 deficiency in female mouse models of α-synucleinopathies. Furthermore, upregulated S100A8/A9 was responsible for α-synuclein degradation by autophagy and the suppression of the pro-inflammatory response in microglia after Usp14 knockdown. Consequently, our study suggests that USP14 could serve as a novel therapeutic target in PD.


Subject(s)
Calgranulin A , Calgranulin B , Mice, Transgenic , Parkinson Disease , Ubiquitin Thiolesterase , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Animals , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/deficiency , Humans , Mice , Female , Male , Calgranulin B/metabolism , Calgranulin B/genetics , Calgranulin A/metabolism , Calgranulin A/genetics , Disease Models, Animal , Mice, Inbred C57BL
14.
Biomolecules ; 14(5)2024 May 12.
Article in English | MEDLINE | ID: mdl-38785979

ABSTRACT

The balance between ubiquitination and deubiquitination is instrumental in the regulation of protein stability and maintenance of cellular homeostasis. The deubiquitinating enzyme, ubiquitin-specific protease 36 (USP36), a member of the USP family, plays a crucial role in this dynamic equilibrium by hydrolyzing and removing ubiquitin chains from target proteins and facilitating their proteasome-dependent degradation. The multifaceted functions of USP36 have been implicated in various disease processes, including cancer, infections, and inflammation, via the modulation of numerous cellular events, including gene transcription regulation, cell cycle regulation, immune responses, signal transduction, tumor growth, and inflammatory processes. The objective of this review is to provide a comprehensive summary of the current state of research on the roles of USP36 in different pathological conditions. By synthesizing the findings from previous studies, we have aimed to increase our understanding of the mechanisms underlying these diseases and identify potential therapeutic targets for their treatment.


Subject(s)
Neoplasms , Ubiquitin Thiolesterase , Humans , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/enzymology , Neoplasms/pathology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Animals , Ubiquitination , Inflammation/metabolism , Signal Transduction , Ubiquitin/metabolism
15.
Biomolecules ; 14(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38785989

ABSTRACT

Endometriosis is a gynecological disorder associated with local inflammation and neuroproliferation. Increased nerve bundle density has been attributed to increased expression of nerve growth factor (NGF) and interleukin-1ß (IL-1ß). Immunohistochemical analysis was carried out on 12 patients presenting with all three anatomic subtypes of endometriosis (deep, superficial peritoneal, endometrioma) at surgery, with at least two surgically excised subtypes available for analysis. Immunolocalization for nerve bundle density around endometriosis using protein gene product 9.5 (PGP9.5), as well as NGF and IL-1ß histoscores in endometriosis epithelium/stroma, was performed to evaluate differences in scores between lesions and anatomic subtypes per patient. Intra-individual heterogeneity in scores across lesions was assessed using the coefficient of variation (CV). The degree of score variability between subtypes was evaluated using the percentage difference between mean scores from one subtype to another subtype for each marker. PGP9.5 nerve bundle density was heterogenous across multiple subtypes of endometriosis, ranging from 50.0% to 173.2%, where most patients (8/12) showed CV ≥ 100%. The percentage difference in scores showed that PGP9.5 nerve bundle density and NGF and IL-1ß expression were heterogenous between anatomic subtypes within the same patient. Based on these observations of intra-individual heterogeneity, we conclude that markers of neuroproliferation in endometriosis should be stratified by anatomic subtype in future studies of clinical correlation.


Subject(s)
Endometriosis , Interleukin-1beta , Nerve Growth Factor , Humans , Female , Endometriosis/metabolism , Endometriosis/pathology , Interleukin-1beta/metabolism , Nerve Growth Factor/metabolism , Adult , Ubiquitin Thiolesterase/metabolism , Middle Aged
16.
Nat Commun ; 15(1): 4519, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806474

ABSTRACT

Protein ubiquitination regulates a wide range of cellular processes. The degree of protein ubiquitination is determined by the delicate balance between ubiquitin ligase (E3)-mediated ubiquitination and deubiquitinase (DUB)-mediated deubiquitination. In comparison to the E3-substrate interactions, the DUB-substrate interactions (DSIs) remain insufficiently investigated. To address this challenge, we introduce a protein sequence-based ab initio method, TransDSI, which transfers proteome-scale evolutionary information to predict unknown DSIs despite inadequate training datasets. An explainable module is integrated to suggest the critical protein regions for DSIs while predicting DSIs. TransDSI outperforms multiple machine learning strategies against both cross-validation and independent test. Two predicted DUBs (USP11 and USP20) for FOXP3 are validated by "wet lab" experiments, along with two predicted substrates (AR and p53) for USP22. TransDSI provides new functional perspective on proteins by identifying regulatory DSIs, and offers clues for potential tumor drug target discovery and precision drug application.


Subject(s)
Deubiquitinating Enzymes , Proteome , Ubiquitination , Humans , Proteome/metabolism , Deubiquitinating Enzymes/metabolism , Deubiquitinating Enzymes/genetics , Deep Learning , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/chemistry , Substrate Specificity , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Machine Learning , Protein Binding , Amino Acid Sequence , Thiolester Hydrolases
17.
Eur J Med Chem ; 272: 116468, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38718626

ABSTRACT

High expression of ubiquitin-specific protease 10 (USP10) promote the proliferation of hepatocellular carcinoma (HCC), thus the development of USP10 inhibitors holds promise as a novel therapeutic approach for HCC treatment. However, the development of selective USP10 inhibitor is still limited. In this study, we developed a novel USP10 inhibitor for investigating the feasibility of targeting USP10 for the treatment of HCC. Due to high USP10 inhibition potency and prominent selectivity, compound D1 bearing quinolin-4(1H)-one scaffold was identified as a lead compound. Subsequent research revealed that D1 significantly inhibits cell proliferation and clone formation in HCC cells. Mechanistic insights indicated that D1 targets the ubiquitin pathway, facilitating the degradation of YAP (Yes-associated protein), thereby triggering the downregulation of p53 and its downstream protein p21. Ultimately, this cascade leads to S-phase arrest in HCC cells, followed by cell apoptosis. Collectively, our findings highlight D1 as a promising starting point for USP10-positive HCC treatment, underscoring its potential as a vital tool for unraveling the functional intricacies of USP10.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents , Carcinoma, Hepatocellular , Cell Proliferation , Drug Discovery , Liver Neoplasms , Transcription Factors , Ubiquitin Thiolesterase , YAP-Signaling Proteins , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Structure-Activity Relationship , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , YAP-Signaling Proteins/metabolism , Molecular Structure , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Apoptosis/drug effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/chemical synthesis , Cell Line, Tumor
18.
Oncotarget ; 15: 328-344, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758815

ABSTRACT

GZ17-6.02 has undergone phase I evaluation in patients with solid tumors (NCT03775525). The RP2D is 375 mg PO BID, with an uveal melanoma patient exhibiting a 15% reduction in tumor mass for 5 months at this dose. Studies in this manuscript have defined the biology of GZ17-6.02 in PDX isolates of uveal melanoma cells. GZ17-6.02 killed uveal melanoma cells through multiple convergent signals including enhanced ATM-AMPK-mTORC1 activity, inactivation of YAP/TAZ and inactivation of eIF2α. GZ17-6.02 significantly enhanced the expression of BAP1, predictive to reduce metastasis, and reduced the levels of ERBB family RTKs, predicted to reduce growth. GZ17-6.02 interacted with doxorubicin or ERBB family inhibitors to significantly enhance tumor cell killing which was associated with greater levels of autophagosome formation and autophagic flux. Knock down of Beclin1, ATG5 or eIF2α were more protective than knock down of ATM, AMPKα, CD95 or FADD, however, over-expression of FLIP-s provided greater protection compared to knock down of CD95 or FADD. Expression of activated forms of mTOR and STAT3 significantly reduced tumor cell killing. GZ17-6.02 reduced the expression of PD-L1 in uveal melanoma cells to a similar extent as observed in cutaneous melanoma cells whereas it was less effective at enhancing the levels of MHCA. The components of GZ17-6.02 were detected in tumors using a syngeneic tumor model. Our data support future testing GZ17-6.02 in uveal melanoma as a single agent, in combination with ERBB family inhibitors, in combination with cytotoxic drugs, or with an anti-PD1 immunotherapy.


Subject(s)
Melanoma , Uveal Neoplasms , Xenograft Model Antitumor Assays , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Melanoma/genetics , Uveal Neoplasms/drug therapy , Uveal Neoplasms/metabolism , Uveal Neoplasms/pathology , Uveal Neoplasms/genetics , Humans , Animals , Mice , Cell Line, Tumor , Signal Transduction/drug effects , Autophagy/drug effects , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics
19.
Nat Commun ; 15(1): 4108, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750011

ABSTRACT

MAPK pathway-driven tumorigenesis, often induced by BRAFV600E, relies on epithelial dedifferentiation. However, how lineage differentiation events are reprogrammed remains unexplored. Here, we demonstrate that proteostatic reactivation of developmental factor, TBX3, accounts for BRAF/MAPK-mediated dedifferentiation and tumorigenesis. During embryonic development, BRAF/MAPK upregulates USP15 to stabilize TBX3, which orchestrates organogenesis by restraining differentiation. The USP15-TBX3 axis is reactivated during tumorigenesis, and Usp15 knockout prohibits BRAFV600E-driven tumor development in a Tbx3-dependent manner. Deleting Tbx3 or Usp15 leads to tumor redifferentiation, which parallels their overdifferentiation tendency during development, exemplified by disrupted thyroid folliculogenesis and elevated differentiation factors such as Tpo, Nis, Tg. The clinical relevance is highlighted in that both USP15 and TBX3 highly correlates with BRAFV600E signature and poor tumor prognosis. Thus, USP15 stabilized TBX3 represents a critical proteostatic mechanism downstream of BRAF/MAPK-directed developmental homeostasis and pathological transformation, supporting that tumorigenesis largely relies on epithelial dedifferentiation achieved via embryonic regulatory program reinitiation.


Subject(s)
Carcinogenesis , Proto-Oncogene Proteins B-raf , T-Box Domain Proteins , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Animals , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Mice , Cell Differentiation , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , MAP Kinase Signaling System/genetics , Gene Expression Regulation, Neoplastic , Mice, Knockout , Female , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism
20.
BMJ Open ; 14(5): e083531, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38754888

ABSTRACT

INTRODUCTION: In light of the burden of traumatic brain injury (TBI) in children and the excessive number of unnecessary CT scans still being performed, new strategies are needed to limit their use while minimising the risk of delayed diagnosis of intracranial lesions (ICLs). Identifying children at higher risk of poor outcomes would enable them to be better monitored. The use of the blood-based brain biomarkers glial fibrillar acidic protein (GFAP) and ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1) could help clinicians in this decision. The overall aim of this study is to provide new knowledge regarding GFAP and UCH-L1 in order to improve TBI management in the paediatric population. METHODS AND ANALYSIS: We will conduct a European, prospective, multicentre study, the BRAINI-2 paediatric study, in 20 centres in France, Spain and Switzerland with an inclusion period of 30 months for a total of 2880 children and adolescents included. To assess the performance of GFAP and UCH-L1 used separately and in combination to predict ICLs on CT scans (primary objective), 630 children less than 18 years of age with mild TBI, defined by a Glasgow Coma Scale score of 13-15 and with a CT scan will be recruited. To evaluate the potential of GFAP and UCH-L1 in predicting the prognosis after TBI (secondary objective), a further 1720 children with mild TBI but no CT scan as well as 130 children with moderate or severe TBI will be recruited. Finally, to establish age-specific reference values for GFAP and UCH-L1 (secondary objective), we will include 400 children and adolescents with no history of TBI. ETHICS AND DISSEMINATION: This study has received ethics approval in all participating countries. Results from our study will be disseminated in international peer-reviewed journals. All procedures were developed in order to assure data protection and confidentiality. TRIAL REGISTRATION NUMBER: NCT05413499.


Subject(s)
Biomarkers , Brain Injuries, Traumatic , Glial Fibrillary Acidic Protein , Tomography, X-Ray Computed , Ubiquitin Thiolesterase , Humans , Brain Injuries, Traumatic/diagnostic imaging , Ubiquitin Thiolesterase/blood , Child , Biomarkers/blood , Prospective Studies , Tomography, X-Ray Computed/methods , Glial Fibrillary Acidic Protein/blood , Adolescent , Child, Preschool , Europe , Female , Male , Infant , Multicenter Studies as Topic , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...