Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 218
Filter
1.
Heliyon ; 10(9): e30192, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38707352

ABSTRACT

Objective: Although the parietal cortex is related to consciousness, the dorsolateral prefrontal and primary motor cortices are the usual targets for repetitive transcranial magnetic stimulation (rTMS) for prolonged disorders of consciousness (pDoC). Herein, we applied parietal rTMS to patients with pDoC, to verify its neurobehavioral effects and explore a new potential rTMS target. Materials and methods: Twenty-six patients with pDoC were assigned to a rTMS or sham group. The rTMS group received 10 sessions of parietal rTMS; the sham group received 10 sessions of sham stimulation. The Coma Recovery Scale-Revised (CRS-R) and event-related potential (ERP) were collected before and after the 10 sessions or sham sessions. Results: After the 10 sessions, the rTMS group showed: a significant CRS-R score increase; ERP appearance of a P300 waveform and significantly increased Fz amplitudes; increased potentials on topographic mapping, especially in the left prefrontal cortex; and an increase in delta and theta band powers at Fz, Cz, and Pz. The sham group did not show such changes in CRS-R score or ERP results statistically. Conclusion: Parietal rTMS shows promise as a novel intervention in the recovery of consciousness in pDoC. It showed neurobehavioral enhancement of residual brain function and may promote frontal activity by enhancing frontal-parietal connections. The parietal cortex may thus be an alternative for rTMS therapy protocols.

2.
CNS Neurosci Ther ; 30(5): e14757, 2024 May.
Article in English | MEDLINE | ID: mdl-38747078

ABSTRACT

BACKGROUND: With the improvement of emergency techniques, the survival rate of patients with severe brain injury has increased. However, this has also led to an annual increase in the number of patients with prolonged disorders of consciousness (pDoC). Hence, recovery of consciousness is an important part of treatment. With advancing techniques, noninvasive neuromodulation seems a promising intervention. The objective of this review was to summarize the latest techniques and provide the basis for protocols of noninvasive neuromodulations in pDoC. METHODS: This review summarized the advances in noninvasive neuromodulation in the treatment of pDoC in the last 5 years. RESULTS: Variable techniques of neuromodulation are used in pDoC. Transcranial ultrasonic stimulation (TUS) and transcutaneous auricular vagus nerve stimulation (taVNS) are very new techniques, while transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS) are still the hotspots in pDoC. Median nerve electrical stimulation (MNS) has received little attention in the last 5 years. CONCLUSIONS: Noninvasive neuromodulation is a valuable and promising technique to treat pDoC. Further studies are needed to determine a unified stimulus protocol to achieve optimal effects as well as safety.


Subject(s)
Consciousness Disorders , Transcranial Direct Current Stimulation , Transcranial Magnetic Stimulation , Vagus Nerve Stimulation , Humans , Consciousness Disorders/therapy , Transcranial Magnetic Stimulation/methods , Transcranial Magnetic Stimulation/trends , Transcranial Direct Current Stimulation/methods , Vagus Nerve Stimulation/methods , Vagus Nerve Stimulation/trends , Transcutaneous Electric Nerve Stimulation/methods , Transcutaneous Electric Nerve Stimulation/trends
4.
Front Biosci (Landmark Ed) ; 29(3): 100, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38538277

ABSTRACT

BACKGROUND: As a dedifferentiated tumor, small cell endometrial neuroendocrine tumors (NETs) are rare and frequently diagnosed at an advanced stage with a poor prognosis. Current treatment recommendations are often extrapolated from histologically similar tumors in other sites or based on retrospective studies. The exploration for diagnostic and therapeutic markers in small cell NETs is of great significance. METHODS: In this study, we conducted single-cell RNA sequencing on a specimen obtained from a patient diagnosed with small cell endometrial neuroendocrine carcinoma (SCNEC) based on pathology. We revealed the cell map and intratumoral heterogeneity of the cancer cells through data analysis. Further, we validated the function of ISL LIM Homeobox 1 (ISL1) in vitro in an established neuroendocrine cell line. Finally, we examined the association between ISL1 and tumor staging in small cell lung cancer (SCLC) patient samples. RESULTS: We observed the significant upregulation of ISL1 expression in tumor cells that showed high expression of the neuroepithelial markers. Additionally, in vitro cell function experiments demonstrated that the high ISL1 expression group exhibited markedly higher cell proliferation and migration abilities compared to the low expression group. Finally, we showed that the expression level of ISL1 was correlated with SCLC stages. CONCLUSIONS: ISL1 protein in NETs shows promise as a potential biomarker for diagnosis and treatment.


Subject(s)
Carcinoma, Neuroendocrine , Neuroendocrine Tumors , Female , Humans , Transcription Factors/genetics , Retrospective Studies , Single-Cell Gene Expression Analysis , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/analysis , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Endometrium/chemistry , Endometrium/metabolism , Endometrium/pathology , Carcinoma, Neuroendocrine/diagnosis , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/therapy
5.
J Exp Clin Cancer Res ; 43(1): 54, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383447

ABSTRACT

BACKGROUND: LATS1/2 are frequently mutated and down-regulated in endometrial cancer (EC), but the contributions of LATS1/2 in EC progression remains unclear. Impaired antigen presentation due to mutations or downregulation of the major histocompatibility complex class I (MHC-I) has been implicated in tumor immune evasion. Herein, we elucidate the oncogenic role that dysregulation of LATS1/2 in EC leads to immune evasion through the down-regulation of MHC-I. METHODS: The mutation and expression as well as the clinical significance of LATS1/2 in EC was assessed in the TCGA cohort and our sample cohort. CRISPR-Cas9 was used to construct knockout cell lines of LATS1/2 in EC. Differentially expressed genes were analyzed by RNA-seq. The interaction between LATS1/2 and STAT1 was verified using co-immunoprecipitation and GST pull-down assays. Mass spectrometry, in vitro kinase assays, ChIP-qPCR, flow cytometry, immunohistochemistry, immunofluorescence and confocal microscopy were performed to investigate the regulation of LATS1/2 on MHC-I through interaction with and phosphorylate STAT1. The killing effect of activated PBMCs on EC cells were used to monitor anti-tumor activity. RESULTS: Here, we demonstrate that LATS1/2 are frequently mutated and down-regulated in EC. Moreover, LATS1/2 loss was found to be associated with a significant down-regulation of MHC-I, independently of the Hippo-YAP pathway. Instead, LATS1/2 were found to directly interact with and phosphorylate STAT1 at Ser727, a crucial transcription factor for MHC-I upregulation in response to interferon-gamma (IFN-γ) signaling, to promote STAT1 accumulating and moving into the nucleus to enhance the transcriptional activation of IRF1/NLRC5 on MHC-I. Additionally, the loss of LATS1/2 was observed to confer increased resistance of EC cells to immune cell-mediated killing and this resistance could be reversed by over-expression of MHC-I. CONCLUSION: Our findings indicate that dysregulation of LATS1/2 in EC leads to immune evasion through the down-regulation of MHC-I, leading to the suppression of infiltrating activated CD8 + T cells and highlight the importance of LATS1/2 in IFN-γ signaling-mediated tumor immune response, suggesting that LATS1/2 is a promising target for immune checkpoint blockade therapy in EC.


Subject(s)
Endometrial Neoplasms , Tumor Escape , Female , Humans , Histocompatibility Antigens Class I , Antigen Presentation , Protein Serine-Threonine Kinases/genetics , Endometrial Neoplasms/genetics , Intracellular Signaling Peptides and Proteins
6.
Oncogenesis ; 13(1): 3, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191478

ABSTRACT

Endometrial cancer (EC) stands as one of the most prevalent malignancies affecting the female genital tract, witnessing a rapid surge in incidence globally. Despite the well-established association of histone methyltransferase SMYD3 with the development and progression of various cancers, its specific oncogenic role in endometrial cancer remains unexplored. In the present study, we report that the expression level of SMYD3 is significantly upregulated in EC samples and associated with EC progression. Through meticulous in vivo and in vitro experiments, we reveal that depletion of SMYD3 curtails cell proliferation, migration, and invasion capabilities, leading to compromised non-homologous end joining repair (NHEJ) and heightened sensitivity of EC cells to radiation. Furthermore, our pathway enrichment analysis underscores the pivotal involvement of the DNA damage repair pathway in regulating EC progression. Mechanistically, in response to DNA damage, SMYD3 is recruited to these sites in a PARP1-dependent manner, specifically methylating LIG4. This methylation sets off a sequential assembly of the LIG4/XRCC4/XLF complex, actively participating in the NHEJ pathway and thereby fostering EC progression. Notably, our findings highlight the promise of SMYD3 as a crucial player in NHEJ repair and its direct correlation with EC progression. Intriguingly, pharmacological intervention targeting SMYD3 with its specific inhibitor, BCI-121, emerges as a potent strategy, markedly suppressing the tumorigenicity of EC cells and significantly enhancing the efficacy of radiotherapy. Collectively, our comprehensive data position SMYD3 as a central factor in NHEJ repair and underscore its potential as a promising pharmacological target for endometrial cancer therapy, validated through both in vitro and in vivo systems.

7.
J Membr Biol ; 257(1-2): 25-36, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38285125

ABSTRACT

Concerted robust opening of cardiac ryanodine receptors' (RyR2) Ca2+ release 1oplasmic reticulum (SR) is fundamental for normal systolic cardiac function. During diastole, infrequent spontaneous RyR2 openings mediate the SR Ca2+ leak that normally constrains SR Ca2+ load. Abnormal large diastolic RyR2-mediated Ca2+ leak events can cause delayed after depolarizations (DADs) and arrhythmias. The RyR2-associated mechanisms underlying these processes are being extensively studied at multiple levels utilizing various model animals. Since there are well-described species-specific differences in cardiac intracellular Ca2+ handing in situ, we tested whether or not single RyR2 function in vitro retains this species specificity. We isolated RyR2-rich heavy SR microsomes from mouse, rat, rabbit, and human ventricular muscle and quantified RyR2 function using identical solutions and methods. The single RyR2 cytosolic Ca2+ sensitivity was similar across these species. However, there were significant species differences in single RyR2 mean open times in both systole and diastole-like solutions. In diastole-like solutions, single rat/mouse RyR2 open probability and frequency of long openings (> 6 ms) were similar, but these values were significantly greater than those of either single rabbit or human RyR2s. We propose these in vitro single RyR2 functional differences across species stem from the species-specific RyR2 regulatory environment present in the source tissue. Our results show the single rabbit RyR2 functional attributes, particularly in diastole-like conditions, replicate those of single human RyR2 best among the species tested.


Subject(s)
Myocytes, Cardiac , Ryanodine Receptor Calcium Release Channel , Mice , Rats , Humans , Rabbits , Animals , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Heart Ventricles , Mammals/metabolism , Calcium/metabolism
8.
Circ Arrhythm Electrophysiol ; 17(1): e012150, 2024 01.
Article in English | MEDLINE | ID: mdl-38126205

ABSTRACT

BACKGROUND: MicroRNA-1 (miR1), encoded by the genes miR1-1 and miR1-2, is the most abundant microRNA in the heart and plays a critical role in heart development and physiology. Dysregulation of miR1 has been associated with various heart diseases, where a significant reduction (>75%) in miR1 expression has been observed in patient hearts with atrial fibrillation or acute myocardial infarction. However, it remains uncertain whether miR1-deficiency acts as a primary etiological factor of cardiac remodeling. METHODS: miR1-1 or miR1-2 knockout mice were crossbred to produce 75%-miR1-knockdown (75%KD; miR1-1+/-:miR1-2-/- or miR1-1-/-:miR1-2+/-) mice. Cardiac pathology of 75%KD cardiomyocytes/hearts was investigated by ECG, patch clamping, optical mapping, transcriptomic, and proteomic assays. RESULTS: In adult 75%KD hearts, the overall miR1 expression was reduced to ≈25% of the normal wild-type level. These adult 75%KD hearts displayed decreased ejection fraction and fractional shortening, prolonged QRS and QT intervals, and high susceptibility to arrhythmias. Adult 75%KD cardiomyocytes exhibited prolonged action potentials with impaired repolarization and excitation-contraction coupling. Comparatively, 75%KD cardiomyocytes showcased reduced Na+ current and transient outward potassium current, coupled with elevated L-type Ca2+ current, as opposed to wild-type cells. RNA sequencing and proteomics assays indicated negative regulation of cardiac muscle contraction and ion channel activities, along with a positive enrichment of smooth muscle contraction genes in 75%KD cardiomyocytes/hearts. miR1 deficiency led to dysregulation of a wide gene network, with miR1's RNA interference-direct targets influencing many indirectly regulated genes. Furthermore, after 6 weeks of bi-weekly intravenous tail-vein injection of miR1 mimics, the ejection fraction and fractional shortening of 75%KD hearts showed significant improvement but remained susceptible to arrhythmias. CONCLUSIONS: miR1 deficiency acts as a primary etiological factor in inducing cardiac remodeling via disrupting heart regulatory homeostasis. Achieving stable and appropriate microRNA expression levels in the heart is critical for effective microRNA-based therapy in cardiovascular diseases.


Subject(s)
MicroRNAs , Mice , Humans , Animals , MicroRNAs/genetics , Proteomics , Ventricular Remodeling , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac , Action Potentials , Mice, Knockout , Homeostasis
9.
BMC Infect Dis ; 23(1): 794, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37964239

ABSTRACT

OBJECTIVE: To investigate the correlation between colorectal polyps (CRP) and Helicobacter pylori (H. pylori) infection, and the correlation between CRP and the expression of phosphorylated ribosomal protein S6 kinase (p-S6K1). Besides, its related influencing factors were determined in the present study. METHODS: A total of 191 subjects who underwent colonoscopy in our hospital from January 2020 to February 2022 were selected for this study. Among them, 141 patients were diagnosed with CRP, and the other 50 subjects were no significant colorectal abnormalities. 141 CRP patients were divided into H. pylori-positive group (n = 89) and H. pylori-negative group (n = 52) according to the results of the H. pylori test. The expression of p-S6K1 in CRP tissue was detected. The relationship between the p-S6K1 expression and the clinicopathological characteristics of CRP patients was analyzed. The logistic analysis of factors influencing the occurrence of CRP was performed. RESULTS: There were significant differences in pathological type, site of disease, the number and size of polyps between the H. pylori negative group and the H. pylori positive group (P < 0.001, P = 0.037, P = 0.042 and P = 0.039). The percentage of the p-S6K1 positive expression in polyp tissues was higher than that in normal tissue and parapolyp tissues (P < 0.001). The p-S6K1 negative group showed significant difference in the number and pathological type of polyps and the presence or absence of a pedicle as compared with the p-S6K1 positive group (P = 0.006, P < 0.001 and P = 0.012). Logistic multifactor analysis showed that BMI, H. pylori infection, smoking history, ApoB, Lp(a) and the p-S6K1 positive expression were all risk factors for the development of CRP (P = 0.025, P = 0.020, P = 0.010, P = 0.005, P = 0.043 and P < 0.001). CONCLUSION: H. pylori infection was closely related to the pathological type, location, and the number and size of CRP. p-S6K1 was highly expressed in CRP, and was positively related to the number, the pathological type and pedicle of polyps. H. pylori infection and the positive p-S6K1 expression were independent risk factors for CRP. By exploring the association between H. pylori infection as well as p-S6K1 and CRP, it is hoped that it will help to formulate a more rigorous colorectal cancer screening program for H. pylori-positive individuals, and at the same time find a new direction for the prevention of CRP and colorectal cancer, and provide some help for future research.


Subject(s)
Colonic Polyps , Colorectal Neoplasms , Helicobacter Infections , Helicobacter pylori , Humans , Colonic Polyps/complications , Colonic Polyps/epidemiology , Colonic Polyps/pathology , Helicobacter Infections/epidemiology , Risk Factors
10.
Circ Res ; 133(8): 658-673, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37681314

ABSTRACT

BACKGROUND: Cardiac conduction is understood to occur through gap junctions. Recent evidence supports ephaptic coupling as another mechanism of electrical communication in the heart. Conduction via gap junctions predicts a direct relationship between conduction velocity (CV) and bulk extracellular resistance. By contrast, ephaptic theory is premised on the existence of a biphasic relationship between CV and the volume of specialized extracellular clefts within intercalated discs such as the perinexus. Our objective was to determine the relationship between ventricular CV and structural changes to micro- and nanoscale extracellular spaces. METHODS: Conduction and Cx43 (connexin43) protein expression were quantified from optically mapped guinea pig whole-heart preparations perfused with the osmotic agents albumin, mannitol, dextran 70 kDa, or dextran 2 MDa. Peak sodium current was quantified in isolated guinea pig ventricular myocytes. Extracellular resistance was quantified by impedance spectroscopy. Intercellular communication was assessed in a heterologous expression system with fluorescence recovery after photobleaching. Perinexal width was quantified from transmission electron micrographs. RESULTS: CV primarily in the transverse direction of propagation was significantly reduced by mannitol and increased by albumin and both dextrans. The combination of albumin and dextran 70 kDa decreased CV relative to albumin alone. Extracellular resistance was reduced by mannitol, unchanged by albumin, and increased by both dextrans. Cx43 expression and conductance and peak sodium currents were not significantly altered by the osmotic agents. In response to osmotic agents, perinexal width, in order of narrowest to widest, was albumin with dextran 70 kDa; albumin or dextran 2 MDa; dextran 70 kDa or no osmotic agent, and mannitol. When compared in the same order, CV was biphasically related to perinexal width. CONCLUSIONS: Cardiac conduction does not correlate with extracellular resistance but is biphasically related to perinexal separation, providing evidence that the relationship between CV and extracellular volume is determined by ephaptic mechanisms under conditions of normal gap junctional coupling.


Subject(s)
Connexin 43 , Dextrans , Animals , Guinea Pigs , Dextrans/metabolism , Connexin 43/metabolism , Myocytes, Cardiac/metabolism , Sodium/metabolism , Gap Junctions/metabolism , Albumins/metabolism , Mannitol/pharmacology , Mannitol/metabolism , Action Potentials
11.
Brain Sci ; 13(9)2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37759896

ABSTRACT

Frontal transcranial direct current stimulation (tDCS) and parietal tDCS are effective for treating disorders of consciousness (DoC); however, the relative efficacies of these techniques have yet to be determined. This paper compares the neuromodulation effects of frontal and parietal tDCS on DoC. Twenty patients with DoC were recruited and randomly assigned to two groups. One group received single-session frontal tDCS and single-session sham tDCS. The other group received single-session parietal tDCS and single-session sham tDCS. Before and after every tDCS session, we recorded coma recovery scale-revised (CRS-R) values and an electroencephalogram. CRS-R was also used to evaluate the state of consciousness at 9-12-month follow-up. Both single-session frontal and parietal tDCS caused significant changes in the genuine permutation cross-mutual information (G_PCMI) of local frontal and across brain regions (p < 0.05). Furthermore, the changes in G_PCMI values were significantly correlated to the CRS-R scores at 9-12-month follow-up after frontal and parietal tDCS (p < 0.05). The changes in G_PCMI and CRS-R scores were also correlated (p < 0.05). Both frontal tDCS and parietal tDCS exert neuromodulatory effects in DoC and induce significant changes in electrophysiology. G_PCMI can be used to evaluate the neuromodulation effects of tDCS.

12.
BMJ Open ; 13(6): e069042, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37280028

ABSTRACT

OBJECTIVE: Sedentary behaviour is associated with increased cancer risk. We aim to assess the associations of domain-specific and total sedentary behaviour with risk of endometrial cancer, with additional attention paid to potential differences in adjustment strategy for obesity and physical activity. DESIGN: A systematic review and meta-analysis was conducted in accordance with the guidelines of Preferred Reporting Items for Systematic Reviews (PRISMA) and Meta-Analyses and the Meta-analysis of Observational Studies in Epidemiology (MOOSE). DATA SOURCES: PubMed, Embase and MEDLINE databases were searched up to 28 February 2023, supplemented by grey literature searches. ELIGIBILITY CRITERIA FOR SELECTING STUDIES: Observational human studies evaluating the association between sedentary behaviour and endometrial cancer. DATA EXTRACTION AND SYNTHESIS: Two reviewers extracted data and conducted the quality assessment based on Newcastle-Ottawa Scale (NOS) independently. We used a random-effects model with inverse variance approach to pool the estimates. The extent of heterogeneity was quantified with the I 2 statistics. RESULTS: Sixteen studies were included in the systematic review. Fourteen studies involving 882 686 participants were included in the meta-analysis. The pooled relative risks (RRs) for high versus low level of overall sedentary behaviour was 1.28 (95% CI: 1.14 to 1.43; I2 =34.8%). The increased risk regarding specific domains was 1.22 (95% CI: 1.09 to 1.37; I2=13.4%, n=10) for occupational domain, 1.34 (95% CI: 0.98 to 1.83; I2=53.7%, n=6) for leisure-time domain and 1.55 (95% CI: 1.27 to 1.89; I2=0.0%, n=2) for total sedentary behaviour. Larger pooled RRs were observed among studies with adjustment for physical activity and studies without adjustment for body mass index. CONCLUSIONS: Higher levels of sedentary behaviour, total and occupational sedentary behaviour in particular, increase the risk of endometrial cancer. Future studies are needed to verify domain-specific associations based on objective quantification of sedentary behaviour, as well as the interaction of physical activity, adiposity and sedentary time on endometrial cancer.


Subject(s)
Endometrial Neoplasms , Sedentary Behavior , Humans , Female , Obesity/epidemiology , Exercise , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/etiology
13.
Sci Adv ; 9(24): eade6624, 2023 06 16.
Article in English | MEDLINE | ID: mdl-37315132

ABSTRACT

DNA damage repair (DDR) is a double-edged sword with different roles in cancer susceptibility and drug resistance. Recent studies suggest that DDR inhibitors affect immune surveillance. However, this phenomenon is poorly understood. We report that methyltransferase SMYD2 plays an essential role in nonhomologous end joining repair (NHEJ), driving tumor cells adaptive to radiotherapy. Mechanically, in response to DNA damage, SMYD2 is mobilized onto chromatin and methylates Ku70 at lysine-74, lysine-516, and lysine-539, leading to increased recruitment of Ku70/Ku80/DNA-PKcs complex. Knockdown of SMYD2 or its inhibitor AZ505 results in persistent DNA damage and improper repair, which sequentially leads to accumulation of cytosolic DNA, and activation of cGAS-STING pathway and triggers antitumor immunity via infiltration and activation of cytotoxic CD8+ T cells. Our study reveals an unidentified role of SMYD2 in regulating NHEJ pathway and innate immune responses, suggesting that SMYD2 is a promising therapeutic target for cancer treatment.


Subject(s)
CD8-Positive T-Lymphocytes , DNA End-Joining Repair , Histone-Lysine N-Methyltransferase , Ku Autoantigen , Chromatin , Lysine , Ku Autoantigen/metabolism , Histone-Lysine N-Methyltransferase/metabolism
14.
Nat Cancer ; 4(5): 716-733, 2023 05.
Article in English | MEDLINE | ID: mdl-37012401

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP1) is essential for the progression of several types of cancers. However, whether and how PARP1 is stabilized to promote genomic stability in triple-negative breast cancer (TNBC) remains unknown. Here, we demonstrated that the deubiquitinase USP15 interacts with and deubiquitinates PARP1 to promote its stability, thereby stimulating DNA repair, genomic stability and TNBC cell proliferation. Two PARP1 mutations found in individuals with breast cancer (E90K and S104R) enhanced the PARP1-USP15 interaction and suppressed PARP1 ubiquitination, thereby elevating the protein level of PARP1. Importantly, we found that estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) inhibited USP15-mediated PARP1 stabilization through different mechanisms. ER bound to the USP15 promoter to suppress its expression, PR suppressed the deubiquitinase activity of USP15, and HER2 abrogated the PARP1-USP15 interaction. The specific absence of these three receptors in TNBC results in high PARP1 levels, leading to increases in base excision repair and female TNBC cell survival.


Subject(s)
Poly (ADP-Ribose) Polymerase-1 , Triple Negative Breast Neoplasms , Ubiquitin-Specific Proteases , Female , Humans , Deubiquitinating Enzymes/genetics , Genomic Instability , Poly (ADP-Ribose) Polymerase-1/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Specific Proteases/genetics
15.
Oncogene ; 42(4): 322-334, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36446891

ABSTRACT

Aberrant hyperactivation of the Hippo pathway effector YAP/TEAD complex causes tissue overgrowth and tumorigenesis in various cancers, including endometrial cancer (EC). The transcription factor SOX17 (SRY [sex-determining region Y]-box 17) is frequently mutated in EC; however, SOX17 mutations are rare in other cancer types. The molecular mechanisms underlying SOX17 mutation-induced EC tumorigenesis remain poorly understood. Here, we demonstrate that SOX17 serves as a tumor suppressor to restrict the proliferation, migration, invasion, and anchorage-independent growth of EC cells, partly by suppressing the transcriptional outputs of the Hippo-YAP/TEAD pathway. SOX17 binds to TEAD transcription factors through its HMG domain and attenuates the DNA-binding ability of TEAD. SOX17 loss by inactivating mutations leads to the malignant transformation of EC cells, which can be reversed by small-molecule inhibitors of YAP/TEAD or cabozantinib, an FDA-approved drug targeting the YAP/TEAD transcriptional target AXL. Our findings reveal novel molecular mechanisms underlying Hippo-YAP/TEAD pathway-driven EC tumorigenesis, and suggest potential therapeutic strategies targeting the Hippo-YAP/TEAD pathway in SOX17-mutated EC.


Subject(s)
Endometrial Neoplasms , YAP-Signaling Proteins , Female , Humans , Transcription Factors/metabolism , Mutation , Endometrial Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
16.
Cell Death Differ ; 30(2): 475-487, 2023 02.
Article in English | MEDLINE | ID: mdl-36481790

ABSTRACT

Blockade of programmed cell death 1 (PD-1)/programmed cell death 1 ligand (PD-L1) has evolved into one of the most promising immunotherapy strategies for cancer patients. Tumor cells frequently overexpress PD-L1 to evade T cell-mediated immune surveillance. However, the specific genetic alterations that drive aberrant overexpression of PD-L1 in cancer cells remain poorly understood. The gene encoding the E3 ubiquitin ligase substrate-binding adaptor SPOP is frequently mutated in endometrial cancer (EC). Here, we report that SPOP negatively regulates PD-L1 expression at the transcriptional level. Wild-type SPOP binds to IRF1, a primary transcription factor responsible for the inducible expression of PD-L1, and subsequently triggers its ubiquitin- proteasomal degradation to suppress IRF1-mediated transcriptional upregulation of PD-L1. In contrast, EC-associated SPOP mutants lose their capacity to degrade IRF1 but stabilize IRF1, and upregulate PD-L1 expression. EC-associated SPOP mutations accelerate xenograft tumor growth partially by increasing IRF1 and PD-L1 expression. Together, we identify SPOP as a negative regulator of the IRF1-PD-L1 axis and characterize the critical roles of IRF1 and PD-L1 in SPOP mutation-driven tumor immune evasion in EC.


Subject(s)
Endometrial Neoplasms , Nuclear Proteins , Tumor Escape , Female , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Endometrial Neoplasms/genetics , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Mutation/genetics , Nuclear Proteins/metabolism
17.
Circ Res ; 132(2): 187-204, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36583388

ABSTRACT

BACKGROUND: NOTCH1 pathogenic variants are implicated in multiple types of congenital heart defects including hypoplastic left heart syndrome, where the left ventricle is underdeveloped. It is unknown how NOTCH1 regulates human cardiac cell lineage determination and cardiomyocyte proliferation. In addition, mechanisms by which NOTCH1 pathogenic variants lead to ventricular hypoplasia in hypoplastic left heart syndrome remain elusive. METHODS: CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 genome editing was utilized to delete NOTCH1 in human induced pluripotent stem cells. Cardiac differentiation was carried out by sequential modulation of WNT signaling, and NOTCH1 knockout and wild-type differentiating cells were collected at day 0, 2, 5, 10, 14, and 30 for single-cell RNA-seq. RESULTS: Human NOTCH1 knockout induced pluripotent stem cells are able to generate functional cardiomyocytes and endothelial cells, suggesting that NOTCH1 is not required for mesoderm differentiation and cardiovascular development in vitro. However, disruption of NOTCH1 blocks human ventricular-like cardiomyocyte differentiation but promotes atrial-like cardiomyocyte generation through shortening the action potential duration. NOTCH1 deficiency leads to defective proliferation of early human cardiomyocytes, and transcriptomic analysis indicates that pathways involved in cell cycle progression and mitosis are downregulated in NOTCH1 knockout cardiomyocytes. Single-cell transcriptomic analysis reveals abnormal cell lineage determination of cardiac mesoderm, which is manifested by the biased differentiation toward epicardial and second heart field progenitors at the expense of first heart field progenitors in NOTCH1 knockout cell populations. CONCLUSIONS: NOTCH1 is essential for human ventricular-like cardiomyocyte differentiation and proliferation through balancing cell fate determination of cardiac mesoderm and modulating cell cycle progression. Because first heart field progenitors primarily contribute to the left ventricle, we speculate that pathogenic NOTCH1 variants lead to biased differentiation of first heart field progenitors, blocked ventricular-like cardiomyocyte differentiation, and defective cardiomyocyte proliferation, which collaboratively contribute to left ventricular hypoplasia in hypoplastic left heart syndrome.


Subject(s)
Hypoplastic Left Heart Syndrome , Induced Pluripotent Stem Cells , Humans , Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/physiology , Myocytes, Cardiac/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
18.
Cell Commun Signal ; 20(1): 177, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36376931

ABSTRACT

BACKGROUND: Loss-of-function (LOF) mutations of JAK1, a member of the JAK kinase family, were frequently observed in EC, indicating that JAK1 may act as a tumor suppressor, at least in EC. However, the mechanism of JAK1 mediated regulation of tumorigenesis remains poorly understood. METHODS: The genetic alterations of JAK1 in EC using latest sequencing dataset of EC deposited in TCGA database. The RNA-Seq dataset of EC and normal endometrial tissues from TCGA cohort was analyzed. The expression of JAK1 in EC and normal endometrial tissues were investigated using immunohistochemistry. The expression levels of genes in endometrial cancer cells were detected by quantitative reverse transcription-PCR (RT-qPCR) and western blotting. JAK1 protein was efficiently depleted by the two shRNAs. HIF1/2-α protein was efficiently depleted by siRNAs. JAK1 overexpressed EC cells were generated by an expressing plasmid. The proliferation and migration ability of cancer cells were evaluated by CCK8, colony formation assays and transwell assays. The global transcriptomic changes in JAK1-depleted KLE cells were investigated using RNA-Seq. Gene Ontology (GO) Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to identify the most significant pathways that were altered in JAK1-depleted KLE cells. The physical association between HIF-1/2α and JAK1 using co-immunoprecipitation (co-IP) assays. RESULTS: In the present study, we found that JAK1 was frequently mutated and downregulated in EC. JAK1 knockdown promotes EC cell proliferation and migration. JAK1 overexpression reduces EC cell proliferation and migration. We examined the transcriptional profiling changes in JAK1-depleted EC cells and unexpectedly found that the hypoxia inducible factor (HIF) pathway was activated. Mechanistically, JAK1 interacts with HIF-1/2α, and reduces HIF1/2-α protein expression under hypoxia. HIF-1/2α knockdown reverses the JAK1 knockdown-induced growth and migration of EC cells under hypoxia. JAK1 knockdown or pharmacological inhibition of JAK1 kinase activity by Ruxolitinib upregulates transcription of HIF target genes under hypoxia. JAK1 overexpression downregulates transcription of HIF target genes under hypoxia. CONCLUSIONS: These findings provide novel insights into the functional link between JAK1 LOF mutations and abnormal HIF pathway activation in EC and suggest that pharmacological inhibition of HIF1/2 represents a promising therapeutic strategy targeting JAK1-mutated ECs. Video Abstract.


Subject(s)
Endometrial Neoplasms , Humans , Female , Janus Kinase 1 , Endometrial Neoplasms/genetics , Signal Transduction , Cell Proliferation , RNA, Small Interfering/metabolism , Hypoxia/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia , Gene Expression Regulation, Neoplastic , Cell Line, Tumor
19.
Life (Basel) ; 12(8)2022 Jul 29.
Article in English | MEDLINE | ID: mdl-36013330

ABSTRACT

Embryonic stem cells (ESCs) are a type of cells capable of self-renewal and multi-directional differentiation. The self-renewal of ESCs is regulated by factors including signaling pathway proteins, transcription factors, epigenetic regulators, cytokines, and small molecular compounds. Similarly, non-coding RNAs, small RNAs, and microRNAs (miRNAs) also play an important role in the process. Functionally, the core transcription factors interact with helper transcription factors to activate the expression of genes that contribute to maintaining pluripotency, while suppressing the expression of differentiation-related genes. Additionally, cytokines such as leukemia suppressor factor (LIF) stimulate downstream signaling pathways and promote self-renewal of ESCs. Particularly, LIF binds to its receptor (LIFR/gp130) to trigger the downstream Jak-Stat3 signaling pathway. BMP4 activates the downstream pathway and acts in combination with Jak-Stat3 to promote pluripotency of ESCs in the absence of serum. In addition, activation of the Wnt-FDZ signaling pathway has been observed to facilitate the self-renewal of ESCs. Small molecule modulator proteins of the pathway mentioned above are widely used in in vitro culture of stem cells. Multiple epigenetic regulators are involved in the maintenance of ESCs self-renewal, making the epigenetic status of ESCs a crucial factor in this process. Similarly, non-coding RNAs and cellular energetics have been described to promote the maintenance of the ESC's self-renewal. These factors regulate the self-renewal and differentiation of ESCs by forming signaling networks. This review focused on the role of major transcription factors, signaling pathways, small molecular compounds, epigenetic regulators, non-coding RNAs, and cellular energetics in ESC's self-renewal.

20.
Mol Ther Nucleic Acids ; 29: 481-497, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36035750

ABSTRACT

The immature phenotype of embryonic stem cell-derived cardiomyocytes (ESC-CMs) limits their application. However, the molecular mechanisms of cardiomyocyte maturation remain largely unexplored. This study found that overexpression of long noncoding RNA (lncRNA)-Cmarr, which was highly expressed in cardiomyocytes, promoted the maturation change and physiological maturation of mouse ESC-CMs (mESC-CMs). Moreover, transplantation of cardiac patch overexpressing Cmarr exhibited better retention of mESC-CMs, reduced infarct area by enhancing vascular density in the host heart, and improved cardiac function in mice after myocardial infarction. Mechanism studies identified that Cmarr acted as a competitive endogenous RNA to impede the repression of miR-540-3p on Dtna expression and promoted the binding of the dystrophin-glycoprotein complex (DGC) and yes-associated protein (YAP), which in turn reduced the proportion of nuclear YAP and the expression of YAP target genes. Therefore, this study revealed the function and mechanism of Cmarr in promoting cardiomyocyte maturation and provided a lncRNA that can be used as a functional factor in the construction of cardiac patches for the treatment of myocardial infarction.

SELECTION OF CITATIONS
SEARCH DETAIL
...