Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 10.176
1.
J Nanobiotechnology ; 22(1): 219, 2024 May 03.
Article En | MEDLINE | ID: mdl-38698419

BACKGROUND: Adipose-derived stem cells (ASCs) represent the most advantageous choice for soft tissue regeneration. Studies proved the recruitment of ASCs post tissue injury was mediated by chemokine CXCL12, but the mechanism by which CXCL12 is generated after tissue injury remains unclear. Migrasomes are newly discovered membrane-bound organelles that could deliver CXCL12 spatially and temporally in vivo. In this study, we sought to investigate whether migrasomes participate ASC-mediated tissue regeneration. METHODS: Discrepant and asymmetrical soft tissue regeneration mice model were established, in which HE staining, immunofluorescent staining, western blot and qPCR were conducted to confirm the role of CXCL12 and migrasomes in ASC-mediated tissue regeneration. Characterization of ASC-derived migrasomes were carried out by confocal microscopy, scanning electron microscopy, transmission electron microscopy as well as western blot analysis. The function and mechanism of migrasomes were further testified by assisting tissue regeneration with isolated migrasomes in vivo and by in vitro transwell combined with co-culture system. RESULTS: Here, we show for the first time that migrasomes participate in soft tissue regeneration. ASCs generate migrasomes enriched with CXCL12 to mediate tissue regeneration. Migrasomes from ASCs could promote stem cells migration by activating CXCR4/RhoA signaling in vivo and in vitro. Chemoattracted ASCs facilitate regeneration, as demonstrated by the upregulation of an adipogenesis-associated protein. This positive feed-back-loop creates a favorable microenvironment for soft tissue regeneration. Thus, migrasomes represent a new therapeutic target for ASC-mediated tissue regeneration. CONCLUSIONS: Our findings reveal a previously unknown function of ASCs in mediating tissue regeneration by generating migrasomes. The ASC-derived migrasomes can restore tissue regeneration by recruiting stem cells, which highlighting the potential application of ASC-derived migrasomes in regenerative medicine.


Adipose Tissue , Chemokine CXCL12 , Receptors, CXCR4 , Regeneration , Stem Cells , rhoA GTP-Binding Protein , Chemokine CXCL12/metabolism , Animals , Receptors, CXCR4/metabolism , Mice , Adipose Tissue/cytology , Adipose Tissue/metabolism , rhoA GTP-Binding Protein/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Mice, Inbred C57BL , Feedback, Physiological , Cell Movement , Cells, Cultured , Male , Signal Transduction
2.
Mol Cancer ; 23(1): 90, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711083

BACKGROUND: Metabolic reprogramming and epigenetic alterations contribute to the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). Lactate-dependent histone modification is a new type of histone mark, which links glycolysis metabolite to the epigenetic process of lactylation. However, the role of histone lactylation in PDAC remains unclear. METHODS: The level of histone lactylation in PDAC was identified by western blot and immunohistochemistry, and its relationship with the overall survival was evaluated using a Kaplan-Meier survival plot. The participation of histone lactylation in the growth and progression of PDAC was confirmed through inhibition of histone lactylation by glycolysis inhibitors or lactate dehydrogenase A (LDHA) knockdown both in vitro and in vivo. The potential writers and erasers of histone lactylation in PDAC were identified by western blot and functional experiments. The potential target genes of H3K18 lactylation (H3K18la) were screened by CUT&Tag and RNA-seq analyses. The candidate target genes TTK protein kinase (TTK) and BUB1 mitotic checkpoint serine/threonine kinase B (BUB1B) were validated through ChIP-qPCR, RT-qPCR and western blot analyses. Next, the effects of these two genes in PDAC were confirmed by knockdown or overexpression. The interaction between TTK and LDHA was identified by Co-IP assay. RESULTS: Histone lactylation, especially H3K18la level was elevated in PDAC, and the high level of H3K18la was associated with poor prognosis. The suppression of glycolytic activity by different kinds of inhibitors or LDHA knockdown contributed to the anti-tumor effects of PDAC in vitro and in vivo. E1A binding protein p300 (P300) and histone deacetylase 2 were the potential writer and eraser of histone lactylation in PDAC cells, respectively. H3K18la was enriched at the promoters and activated the transcription of mitotic checkpoint regulators TTK and BUB1B. Interestingly, TTK and BUB1B could elevate the expression of P300 which in turn increased glycolysis. Moreover, TTK phosphorylated LDHA at tyrosine 239 (Y239) and activated LDHA, and subsequently upregulated lactate and H3K18la levels. CONCLUSIONS: The glycolysis-H3K18la-TTK/BUB1B positive feedback loop exacerbates dysfunction in PDAC. These findings delivered a new exploration and significant inter-relationship between lactate metabolic reprogramming and epigenetic regulation, which might pave the way toward novel lactylation treatment strategies in PDAC therapy.


Carcinoma, Pancreatic Ductal , Gene Expression Regulation, Neoplastic , Glycolysis , Histones , L-Lactate Dehydrogenase , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Humans , Histones/metabolism , Animals , Cell Line, Tumor , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Mice , Feedback, Physiological , Epigenesis, Genetic , Carcinogenesis/metabolism , Carcinogenesis/genetics , Prognosis , Cell Proliferation , Female
4.
Signal Transduct Target Ther ; 9(1): 105, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38679634

Impaired brain glucose metabolism is an early indicator of Alzheimer's disease (AD); however, the fundamental mechanism is unknown. In this study, we found a substantial decline in isocitrate dehydrogenase 3ß (IDH3ß) levels, a critical tricarboxylic acid cycle enzyme, in AD patients and AD-transgenic mice's brains. Further investigations demonstrated that the knockdown of IDH3ß induced oxidation-phosphorylation uncoupling, leading to reduced energy metabolism and lactate accumulation. The resulting increased lactate, a source of lactyl, was found to promote histone lactylation, thereby enhancing the expression of paired-box gene 6 (PAX6). As an inhibitory transcription factor of IDH3ß, the elevated PAX6 in turn inhibited the expression of IDH3ß, leading to tau hyperphosphorylation, synapse impairment, and learning and memory deficits resembling those seen in AD. In AD-transgenic mice, upregulating IDH3ß and downregulating PAX6 were found to improve cognitive functioning and reverse AD-like pathologies. Collectively, our data suggest that impaired oxidative phosphorylation accelerates AD progression via a positive feedback inhibition loop of IDH3ß-lactate-PAX6-IDH3ß. Breaking this loop by upregulating IDH3ß or downregulating PAX6 attenuates AD neurodegeneration and cognitive impairments.


Alzheimer Disease , Isocitrate Dehydrogenase , Mice, Transgenic , Animals , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Mice , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Feedback, Physiological , Male , Female
5.
Cell Rep ; 43(4): 114032, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38568805

N(6)-methyladenosine (m6A) critically regulates RNA dynamics in various biological processes. The m6A demethylase ALKBH5 promotes tumorigenesis of glioblastoma, while the intricate web that orchestrates its regulation remains enigmatic. Here, we discover that cell density affects ALKBH5 subcellular localization and m6A dynamics. Mechanistically, ALKBH5 is phosphorylated by the large tumor suppressor kinase 2 (LATS2), preventing its nuclear export and enhancing protein stability. Furthermore, phosphorylated ALKBH5 reciprocally erases m6A from LATS2 mRNA, thereby stabilizing this transcript. Unexpectedly, LATS2 depletion suppresses glioblastoma stem cell self-renewal independent of yes-associated protein activation. Additionally, deficiency in either LATS2 or ALKBH5 phosphorylation impedes tumor progression in mouse xenograft models. Moreover, high levels of LATS2 expression and ALKBH5 phosphorylation are associated with tumor malignancy in patients with gliomas. Collectively, our study unveils an oncogenic positive feedback loop between LATS2 and ALKBH5, revealing a non-canonical branch of the Hippo pathway for RNA processing and suggesting potential anti-cancer interventions.


Adenosine/analogs & derivatives , AlkB Homolog 5, RNA Demethylase , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , Humans , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Phosphorylation , Mice , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Adenosine/metabolism , Feedback, Physiological , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , HEK293 Cells , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cell Self Renewal
6.
J Cell Sci ; 137(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38563084

Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.


Focal Adhesions , GTPase-Activating Proteins , Mechanotransduction, Cellular , Tumor Suppressor Proteins , YAP-Signaling Proteins , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Movement , Feedback, Physiological , Focal Adhesions/metabolism , Focal Adhesions/genetics , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Mechanotransduction, Cellular/genetics , Neovascularization, Physiologic , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins/metabolism
7.
Redox Biol ; 72: 103145, 2024 Jun.
Article En | MEDLINE | ID: mdl-38583415

Ferroptosis is a nonapoptotic form of regulated cell death that has been reported to play a central role in cardiac ischemia‒reperfusion (I/R) injury. N-acetyltransferase 10 (NAT10) contributes to cardiomyocyte apoptosis by functioning as an RNA ac4c acetyltransferase, but its role in cardiomyocyte ferroptosis during I/R injury has not been determined. This study aimed to elucidate the role of NAT10 in cardiac ferroptosis as well as the underlying mechanism. The mRNA and protein levels of NAT10 were increased in mouse hearts after I/R and in cardiomyocytes that were exposed to hypoxia/reoxygenation. P53 acted as an endogenous activator of NAT10 during I/R in a transcription-dependent manner. Cardiac overexpression of NAT10 caused cardiomyocyte ferroptosis to exacerbate I/R injury, while cardiomyocyte-specific knockout of NAT10 or pharmacological inhibition of NAT10 with Remodelin had the opposite effects. The inhibition of cardiomyocyte ferroptosis by Fer-1 exerted superior cardioprotective effects against the NAT10-induced exacerbation of post-I/R cardiac damage than the inhibition of apoptosis by emricasan. Mechanistically, NAT10 induced the ac4C modification of Mybbp1a, increasing its stability, which in turn activated p53 and subsequently repressed the transcription of the anti-ferroptotic gene SLC7A11. Moreover, knockdown of Mybbp1a partially abolished the detrimental effects of NAT10 overexpression on cardiomyocyte ferroptosis and cardiac I/R injury. Collectively, our study revealed that p53 and NAT10 interdependently cooperate to form a positive feedback loop that promotes cardiomyocyte ferroptosis to exacerbate cardiac I/R injury, suggesting that targeting the NAT10/Mybbp1a/p53 axis may be a novel approach for treating cardiac I/R.


Ferroptosis , Myocardial Reperfusion Injury , Myocytes, Cardiac , Tumor Suppressor Protein p53 , Ferroptosis/genetics , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/genetics , Acetyltransferases/metabolism , Acetyltransferases/genetics , Feedback, Physiological , Signal Transduction , Disease Models, Animal , Male , Humans , Apoptosis
8.
J Physiol ; 602(9): 1953-1966, 2024 May.
Article En | MEDLINE | ID: mdl-38630963

Dynamic cerebral autoregulation (dCA) is the mechanism that describes how the brain maintains cerebral blood flow approximately constant in response to short-term changes in arterial blood pressure. This is known to be impaired in many different pathological conditions, including ischaemic and haemorrhagic stroke, dementia and traumatic brain injury. Many different approaches have thus been used both to analyse and to quantify this mechanism in a range of healthy and diseased subjects, including data-driven models (in both the time and the frequency domain) and biophysical models. However, despite the substantial body of work on both biophysical models and data-driven models of dCA, there remains little work that links the two together. One of the reasons for this is proposed to be the discrepancies between the time constants that govern dCA in models and in experimental data. In this study, the processes that govern dCA are examined and it is proposed that the application of biophysical models remains limited due to a lack of understanding about the physical processes that are being modelled, partly due to the specific model formulation that has been most widely used (the equivalent electrical circuit). Based on the analysis presented here, it is proposed that the two most important time constants are arterial transit time and feedback time constant. It is therefore time to revisit equivalent electrical circuit models of dCA and to develop a more physiologically realistic alternative, one that can more easily be related to experimental data. KEY POINTS: Dynamic cerebral autoregulation is governed by two time constants. The first time constant is the arterial transit time, rather than the traditional 'RC' time constant widely used in previous models. This arterial transit time is approximately 1 s in the brain. The second time constant is the feedback time constant, which is less accurately known, although it is somewhat larger than the arterial transit time. The equivalent electrical circuit model of dynamic cerebral autoregulation should be replaced with a more physiologically representative model.


Cerebrovascular Circulation , Homeostasis , Homeostasis/physiology , Cerebrovascular Circulation/physiology , Humans , Feedback, Physiological , Models, Cardiovascular , Brain/physiology , Brain/blood supply , Animals
9.
New Phytol ; 242(5): 2026-2042, 2024 Jun.
Article En | MEDLINE | ID: mdl-38494681

Seed dormancy governs germination timing, with both evolutionary and applied consequences. Despite extensive studies on the hormonal and genetic control of these processes, molecular mechanisms directly linking dormancy and germination remain poorly understood. By screening a collection of lines overexpressing Arabidopsis transcription factors, we identified ERF50 as a key gene to control dormancy and germination. To study its regulation, we measured seed-related physiological parameters in loss-of-function mutants and carried out transactivation, protein interaction and ChIP-PCR analyses. We found direct ERF50-mediated repression of DOG1 and activation of EXPA2 transcription, which results in enhanced seed germination. Although ERF50 expression is increased by DOG1 in dormant seeds, ERF50 germination-promoting activity is blocked by RGL2. The physiological, genetic and molecular evidence gathered here supports that ERF50 controls germination timing by regulating DOG1 levels to leverage its role as enhancer of seed germination, via RGL2 antagonism on EXPA2 expression. Our results highlight the central role of ERF50 as a feedback regulator to couple and fine-tune seed dormancy and germination.


Arabidopsis Proteins , Arabidopsis , Feedback, Physiological , Gene Expression Regulation, Plant , Germination , Plant Dormancy , Seeds , Transcription Factors , Germination/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis/growth & development , Transcription Factors/metabolism , Transcription Factors/genetics , Seeds/growth & development , Seeds/physiology , Seeds/genetics , Plant Dormancy/genetics , Time Factors , Protein Binding
10.
Apoptosis ; 29(5-6): 882-897, 2024 Jun.
Article En | MEDLINE | ID: mdl-38491252

Bone marrow mesenchymal stem cell (BMSC) transplantation is a promising regenerative therapy; however, the survival rate of BMSCs after transplantation is low. Oxidative stress is one of the main reasons for the high apoptosis rate of BMSCs after transplantation, so there is an urgent need to explore the mechanism of oxidative stress-induced apoptosis of BMSCs. Our previous transcriptome sequencing results suggested that the expression of P53-induced nuclear protein 1 (TP53INP1) and the tumor suppressor P53 (P53) was significantly upregulated during the process of oxidative stress-induced apoptosis of BMSCs. The present study further revealed the role and mechanism of TP53INP1 and P53 in oxidative stress-induced apoptosis in BMSCs. Overexpression of TP53INP1 induced apoptosis of BMSCs, knockdown of TP53INP1 alleviated oxidative stress apoptosis of BMSCs. Under oxidative stress conditions, P53 is regulated by TP53INP1, while P53 can positively regulate the expression of TP53INP1, so the two form a positive feedback loop. To clarify the mechanism of feedback loop formation. We found that TP53INP1 inhibited the ubiquitination and degradation of P53 by increasing the phosphorylation level of P53, leading to the accumulation of P53 protein. P53 can act on the promoter of the TP53INP1 gene and increase the expression of TP53INP1 through transcriptional activation. This is the first report on a positive feedback loop formed by TP53INP1 and P53 under oxidative stress. The present study clarified the formation mechanism of the positive feedback loop. The TP53INP1-P53 positive feedback loop may serve as a potential target for inhibiting oxidative stress-induced apoptosis in BMSCs.


Apoptosis , Mesenchymal Stem Cells , Oxidative Stress , Tumor Suppressor Protein p53 , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Apoptosis/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Humans , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Ubiquitination , Carrier Proteins/genetics , Carrier Proteins/metabolism , Phosphorylation , Cells, Cultured , Feedback, Physiological , Mice
11.
Cancer Sci ; 115(5): 1587-1601, 2024 May.
Article En | MEDLINE | ID: mdl-38438251

Both lysine and arginine methyltransferases are thought to be promising therapeutic targets for malignant tumors, yet how these methyltransferases function in malignant tumors, especially hepatocellular carcinoma (HCC), has not been fully elucidated. Here, we reported that SMYD4, a lysine methyltransferase, acts as an oncogene in HCC. SMYD4 was highly upregulated in HCC and promoted HCC cell proliferation and metastasis. Mechanistically, PRMT5, a well-known arginine methyltransferase, was identified as a SMYD4-binding protein. SMYD4 monomethylated PRMT5 and enhanced the interaction between PRMT5 and MEP50, thereby promoting the symmetrical dimethylation of H3R2 and H4R3 on the PRMT5 target gene promoter and subsequently activating DVL3 expression and inhibiting expression of E-cadherin, RBL2, and miR-29b-1-5p. Moreover, miR-29b-1-5p was found to inversely regulate SMYD4 expression in HCC cells, thus forming a positive feedback loop. Furthermore, we found that the oncogenic effect of SMYD4 could be effectively suppressed by PRMT5 inhibitor in vitro and in vivo. Clinically, high coexpression of SMYD4 and PRMT5 was associated with poor prognosis of HCC patients. In summary, our study provides a model of crosstalk between lysine and arginine methyltransferases in HCC and highlights the SMYD4-PRMT5 axis as a potential therapeutic target for the treatment of HCC.


Carcinoma, Hepatocellular , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Liver Neoplasms , MicroRNAs , Protein-Arginine N-Methyltransferases , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Animals , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Mice , Methylation , Male , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Feedback, Physiological , Female , Mice, Nude
12.
Nature ; 628(8008): 596-603, 2024 Apr.
Article En | MEDLINE | ID: mdl-38509371

Motor neurons are the final common pathway1 through which the brain controls movement of the body, forming the basic elements from which all movement is composed. Yet how a single motor neuron contributes to control during natural movement remains unclear. Here we anatomically and functionally characterize the individual roles of the motor neurons that control head movement in the fly, Drosophila melanogaster. Counterintuitively, we find that activity in a single motor neuron rotates the head in different directions, depending on the starting posture of the head, such that the head converges towards a pose determined by the identity of the stimulated motor neuron. A feedback model predicts that this convergent behaviour results from motor neuron drive interacting with proprioceptive feedback. We identify and genetically2 suppress a single class of proprioceptive neuron3 that changes the motor neuron-induced convergence as predicted by the feedback model. These data suggest a framework for how the brain controls movements: instead of directly generating movement in a given direction by activating a fixed set of motor neurons, the brain controls movements by adding bias to a continuing proprioceptive-motor loop.


Drosophila melanogaster , Motor Neurons , Movement , Posture , Proprioception , Animals , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Feedback, Physiological/physiology , Head/physiology , Models, Neurological , Motor Neurons/physiology , Movement/physiology , Posture/physiology , Proprioception/genetics , Proprioception/physiology , Male
13.
Phytomedicine ; 128: 155417, 2024 Jun.
Article En | MEDLINE | ID: mdl-38518642

BACKGROUND: The role of the glioblastoma (GBM) microenvironment is pivotal in the development of gliomas. Discovering drugs that can traverse the blood-brain barrier and modulate the tumor microenvironment is crucial for the treatment of GBM. Dioscin, a steroidal saponin derived from various kinds of plants and herbs known to penetrate the blood-brain barrier, has shown its powerful anti-tumor activity. However, little is known about its effects on GBM microenvironment. METHODS: Bioinformatics analysis was conducted to assess the link between GBM patients and their prognosis. Multiple techniques, including RNA sequencing, immunofluorescence staining, Western blot analysis, RNA-immunoprecipitation (RIP) assays, and Chromatin immunoprecipitation (CHIP) analysis were employed to elucidate the mechanism through which Dioscin modulates the immune microenvironment. RESULTS: Dioscin significantly impaired the polarization of macrophages into the M2 phenotype and enhanced the phagocytic ability of macrophages in vitro and in vivo. A strong correlation between high expression of RBM47 in GBM and a detrimental prognosis for patients was demonstrated. RNA-sequencing analysis revealed an association between RBM47 and the immune response. The inhibition of RBM47 significantly impaired the recruitment and polarization of macrophages into the M2 phenotype and enhanced the phagocytic ability of macrophages. Moreover, RBM47 could stabilize the mRNA of inflammatory genes and enhance the expression of these genes by activating the NF-κB pathway. In addition, NF-κB acts as a transcription factor that enhances the transcriptional activity of RBM47. Notably, we found that Dioscin could significantly inhibit the activation of NF-κB and then downregulate the expression of RBM47 and inflammatory genes protein. CONCLUSION: Our study reveals that the positive feedback loop between RBM47 and NF-κB could promote immunosuppressive microenvironment in GBM. Dioscin effectively inhibits M2 polarization in GBM by disrupting the positive feedback loop between RBM47 and NF-κB, indicating its potential therapeutic effects in GBM treatment.


Diosgenin , Diosgenin/analogs & derivatives , NF-kappa B , Tumor Microenvironment , Diosgenin/pharmacology , Humans , NF-kappa B/metabolism , Tumor Microenvironment/drug effects , Animals , Macrophages/drug effects , Macrophages/metabolism , Glioma/drug therapy , Glioma/metabolism , Mice , Cell Line, Tumor , RNA-Binding Proteins/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Feedback, Physiological/drug effects
14.
JCI Insight ; 9(8)2024 Mar 12.
Article En | MEDLINE | ID: mdl-38470486

IL-17C is an epithelial cell-derived proinflammatory cytokine whose transcriptional regulation remains unclear. Analysis of the IL17C promoter region identified TCF4 as putative regulator, and siRNA knockdown of TCF4 in human keratinocytes (KCs) increased IL17C. IL-17C stimulation of KCs (along with IL-17A and TNF-α stimulation) decreased TCF4 and increased NFKBIZ and ZC3H12A expression in an IL-17RA/RE-dependent manner, thus creating a feedback loop. ZC3H12A (MCPIP1/Regnase-1), a transcriptional immune-response regulator, also increased following TCF4 siRNA knockdown, and siRNA knockdown of ZC3H12A decreased NFKBIZ, IL1B, IL36G, CCL20, and CXCL1, revealing a proinflammatory role for ZC3H12A. Examination of lesional skin from the KC-Tie2 inflammatory dermatitis mouse model identified decreases in TCF4 protein concomitant with increases in IL-17C and Zc3h12a that reversed following the genetic elimination of Il17c, Il17ra, and Il17re and improvement in the skin phenotype. Conversely, interference with Tcf4 in KC-Tie2 mouse skin increased Il17c and exacerbated the inflammatory skin phenotype. Together, these findings identify a role for TCF4 in the negative regulation of IL-17C, which, alone and with TNF-α and IL-17A, feed back to decrease TCF4 in an IL-17RA/RE-dependent manner. This loop is further amplified by IL-17C-TCF4 autocrine regulation of ZC3H12A and IL-17C regulation of NFKBIZ to promote self-sustaining skin inflammation.


Adaptor Proteins, Signal Transducing , Interleukin-17 , Keratinocytes , Receptors, Interleukin-17 , Ribonucleases , Signal Transduction , Transcription Factor 4 , Animals , Transcription Factor 4/metabolism , Transcription Factor 4/genetics , Humans , Interleukin-17/metabolism , Interleukin-17/genetics , Mice , Keratinocytes/metabolism , Ribonucleases/metabolism , Ribonucleases/genetics , Receptors, Interleukin-17/metabolism , Receptors, Interleukin-17/genetics , Inflammation/metabolism , Inflammation/genetics , Disease Models, Animal , Epidermis/metabolism , Dermatitis/metabolism , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Feedback, Physiological , Gene Expression Regulation
15.
J Plant Res ; 137(3): 445-453, 2024 May.
Article En | MEDLINE | ID: mdl-38367196

Thioredoxin (Trx) is a small redox mediator protein involved in the regulation of various chloroplast functions by modulating the redox state of Trx target proteins in ever-changing light environments. Using reducing equivalents produced by the photosynthetic electron transport chain, Trx reduces the disulfide bonds on target proteins and generally turns on their activities. While the details of the protein-reduction mechanism by Trx have been well investigated, the oxidation mechanism that counteracts it has long been unclear. We have recently demonstrated that Trx-like proteins such as Trx-like2 and atypical Cys His-rich Trx (ACHT) can function as protein oxidation factors in chloroplasts. Our latest study on transgenic Arabidopsis plants indicated that the ACHT isoform ACHT2 is involved in regulating the thermal dissipation of light energy. To understand the role of ACHT2 in vivo, we characterized phenotypic changes specifically caused by ACHT2 overexpression in Arabidopsis. ACHT2-overexpressing plants showed growth defects, especially under high light conditions. This growth phenotype was accompanied with the impaired reductive activation of Calvin-Benson cycle enzymes, enhanced thermal dissipation of light energy, and decreased photosystem II activity. Overall, ACHT2 overexpression promoted protein oxidation that led to the inadequate activation of Calvin-Benson cycle enzymes in light and consequently induced negative feedback control of the photosynthetic electron transport chain. This study highlights the importance of the balance between protein reduction and oxidation in chloroplasts for optimal photosynthetic performance and plant growth.


Arabidopsis Proteins , Arabidopsis , Photosynthesis , Plants, Genetically Modified , Thioredoxins , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Thioredoxins/metabolism , Thioredoxins/genetics , Photosystem II Protein Complex/metabolism , Photosystem II Protein Complex/genetics , Chloroplasts/metabolism , Oxidation-Reduction , Feedback, Physiological , Light , Gene Expression Regulation, Plant , Electron Transport
16.
Plant Commun ; 5(5): 100833, 2024 May 13.
Article En | MEDLINE | ID: mdl-38327058

Plants can sense temperature changes and adjust their growth accordingly. In Arabidopsis, high ambient temperatures stimulate stem elongation by activating a key thermoresponsive regulator, PHYTOCHROME INTERACTING FACTOR 4 (PIF4). Here, we show that warmth promotes the nighttime transcription of GI, which is necessary for the high temperature-induced transcription of TOC1. Genetic analyses suggest that GI prevents excessive thermoresponsive growth by inhibiting PIF4, with this regulatory mechanism being partially reliant on TOC1. GI transcription is repressed by ELF3 and HY5, which concurrently inhibit PIF4 expression and activity. Temperature elevation causes the deactivation or degradation of ELF3 and HY5, leading to PIF4 activation and relief of GI transcriptional repression at high temperatures. This allows PIF4 to further activate GI transcription in response to elevated temperatures. GI, in turn, inhibits PIF4, establishing a negative feedback loop that fine-tunes PIF4 activity. In addition, we demonstrate that ELF3, HY5, and PIF4 regulate GI transcription by modulating the enrichment of histone variant H2A.Z at the GI locus. Together, our findings suggest that thermal release of a negative feedback loop finely adjusts plant thermomorphogenesis.


Arabidopsis Proteins , Arabidopsis , Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/growth & development , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Feedback, Physiological , Temperature , Hot Temperature , Vernalization
17.
J Virol ; 98(2): e0188823, 2024 Feb 20.
Article En | MEDLINE | ID: mdl-38289104

Human cytomegalovirus (HCMV) utilizes peripheral blood monocytes as a means to systemically disseminate throughout the host. Following viral entry, HCMV stimulates non-canonical Akt signaling leading to the activation of mTORC1 and the subsequent translation of select antiapoptotic proteins within infected monocytes. However, the full extent to which the HCMV-initiated Akt/mTORC1 signaling axis reshapes the monocyte translatome is unclear. We found HCMV entry alone was able to stimulate widescale changes to mRNA translation levels and that inhibition of mTOR, a component of mTORC1, dramatically attenuated HCMV-induced protein synthesis. Although monocytes treated with normal myeloid growth factors also exhibited increased levels of translation, mTOR inhibition had no effect, suggesting HCMV activation of mTOR stimulates the acquisition of a unique translatome within infected monocytes. Indeed, polyribosomal profiling of HCMV-infected monocytes identified distinct prosurvival transcripts that were preferentially loaded with ribosomes when compared to growth factor-treated cells. Sirtuin 1 (SIRT1), a deacetylase that exerts prosurvival effects through regulation of the PI3K/Akt pathway, was found to be highly enriched following HCMV infection in an mTOR-dependent manner. Importantly, SIRT1 inhibition led to the death of HCMV-infected monocytes while having minimal effect on uninfected cells. SIRT1 also supported a positive feedback loop to sustain Akt/mTORC1 signaling following viral entry. Taken together, HCMV profoundly reshapes mRNA translation in an mTOR-dependent manner to enhance the synthesis of select factors necessary for the survival of infected monocytes.IMPORTANCEHuman cytomegalovirus (HCMV) infection is a significant cause of morbidity and mortality among the immunonaïve and immunocompromised. Peripheral blood monocytes are a major cell type responsible for disseminating the virus from the initial site of infection. In order for monocytes to mediate viral spread within the host, HCMV must subvert the naturally short lifespan of these cells. In this study, we performed polysomal profiling analysis, which demonstrated HCMV to globally redirect mRNA translation toward the synthesis of cellular prosurvival factors within infected monocytes. Specifically, HCMV entry into monocytes induced the translation of cellular SIRT1 to generate an antiapoptotic state. Defining the precise mechanisms through which HCMV stimulates survival will provide insight into novel anti-HCMV drugs able to target infected monocytes.


Cytomegalovirus , Host Microbial Interactions , Mechanistic Target of Rapamycin Complex 1 , Monocytes , Protein Biosynthesis , RNA, Messenger , Humans , Apoptosis , Cell Survival/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/transmission , Cytomegalovirus Infections/virology , Feedback, Physiological , Mechanistic Target of Rapamycin Complex 1/metabolism , Monocytes/cytology , Monocytes/metabolism , Monocytes/virology , Phosphatidylinositol 3-Kinases/metabolism , Polyribosomes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Sirtuin 1/biosynthesis , Sirtuin 1/genetics , Sirtuin 1/metabolism , Virus Internalization
18.
Sportis (A Coruña) ; 10(1): 212-245, 2024. ilus, tab
Article Es | IBECS | ID: ibc-229142

El balonmano, un deporte altamente dinámico y competitivo, involucra una interacción compleja de variables que influyen en el rendimiento de jugadores y equipos, entre ellas está la comunicación. Este artículo se centra en examinar la relación entre la comunicación y el entrenamiento en el balonmano durante el período 2013-2023. El objetivo de esta revisión sistemática es revelar el estado actual de la investigación en este ámbito. 12 estudios fueron exhaustivamente revisados, abordando la comunicación, el rendimiento y la formación en el contexto del balonmano a partir de una revisión sistemática siguiendo las directrices del PRISMA, recopilando evidencia de una exhaustiva búsqueda de literatura en la Web of Science, abarcando artículos publicados entre 2013 y 2023. Los resultados resaltan un interés creciente en la comunicación entre entrenadores y jugadores, especialmente enfocado en mejorar el rendimiento de los jugadores. La literatura subraya la importancia de una comunicación efectiva en la formación y el desempeño, pero se identifica una carencia en el enfoque integral de la comunicación como variable crítica. A pesar de la evidencia abundante sobre su importancia, la exploración de la comunicación en el contexto del balonmano aún está en sus etapas iniciales en comparación con otros deportes. Se concluye que existe un creciente interés en la relación entre la comunicación del entrenador y el rendimiento de los jugadores en balonmano, pero se necesita un enfoque más integral. Los entrenadores pueden utilizar esta información para mejorar la formación y el rendimiento (AU)


Handball, a highly dynamic and competitive sport, involves a complex interplay of variables influencing player and team performance, with communication being one of them. This article focuses on examining the relationship between communication and coaching in handball during the period 2013-2023. The objective of this systematic review is to unveil the current state of research in this field. Twelve studies were thoroughly reviewed, addressing communication, performance, and coaching in the context of handball through a systematic review following PRISMA guidelines, gathering evidence from an extensive literature search in the Web of Science, covering articles published between 2013 and 2023. The results highlight a growing interest in coach-player communication, particularly aimed at enhancing player performance. The literature underscores the importance of effective communication in training and performance but identifies a lack of a comprehensive approach to communication as a critical variable. Despite ample evidence of its significance, the exploration of communication in the context of handball is still in its early stages compared to other sports. It is concluded that there is a rising interest in the relationship between coach communication and player performance in handball, but a more comprehensive approach is needed. Coaches can use this information to enhance training and performance (AU)


Humans , Feedback, Physiological/physiology , Communication , Affect
19.
Science ; 382(6672): 820-828, 2023 11 17.
Article En | MEDLINE | ID: mdl-37917749

Mitochondria must maintain adequate amounts of metabolites for protective and biosynthetic functions. However, how mitochondria sense the abundance of metabolites and regulate metabolic homeostasis is not well understood. In this work, we focused on glutathione (GSH), a critical redox metabolite in mitochondria, and identified a feedback mechanism that controls its abundance through the mitochondrial GSH transporter, SLC25A39. Under physiological conditions, SLC25A39 is rapidly degraded by mitochondrial protease AFG3L2. Depletion of GSH dissociates AFG3L2 from SLC25A39, causing a compensatory increase in mitochondrial GSH uptake. Genetic and proteomic analyses identified a putative iron-sulfur cluster in the matrix-facing loop of SLC25A39 as essential for this regulation, coupling mitochondrial iron homeostasis to GSH import. Altogether, our work revealed a paradigm for the autoregulatory control of metabolic homeostasis in organelles.


ATP-Dependent Proteases , ATPases Associated with Diverse Cellular Activities , Glutathione , Mitochondria , Mitochondrial Proteins , Phosphate Transport Proteins , Glutathione/metabolism , Homeostasis , Iron/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Proteomics , Feedback, Physiological , Mitochondrial Proteins/metabolism , Phosphate Transport Proteins/metabolism , Humans , Iron-Sulfur Proteins/metabolism , Proteolysis , HEK293 Cells , ATP-Dependent Proteases/genetics , ATP-Dependent Proteases/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism
20.
Nature ; 624(7990): 130-137, 2023 Dec.
Article En | MEDLINE | ID: mdl-37993711

The termination of a meal is controlled by dedicated neural circuits in the caudal brainstem. A key challenge is to understand how these circuits transform the sensory signals generated during feeding into dynamic control of behaviour. The caudal nucleus of the solitary tract (cNTS) is the first site in the brain where many meal-related signals are sensed and integrated1-4, but how the cNTS processes ingestive feedback during behaviour is unknown. Here we describe how prolactin-releasing hormone (PRLH) and GCG neurons, two principal cNTS cell types that promote non-aversive satiety, are regulated during ingestion. PRLH neurons showed sustained activation by visceral feedback when nutrients were infused into the stomach, but these sustained responses were substantially reduced during oral consumption. Instead, PRLH neurons shifted to a phasic activity pattern that was time-locked to ingestion and linked to the taste of food. Optogenetic manipulations revealed that PRLH neurons control the duration of seconds-timescale feeding bursts, revealing a mechanism by which orosensory signals feed back to restrain the pace of ingestion. By contrast, GCG neurons were activated by mechanical feedback from the gut, tracked the amount of food consumed and promoted satiety that lasted for tens of minutes. These findings reveal that sequential negative feedback signals from the mouth and gut engage distinct circuits in the caudal brainstem, which in turn control elements of feeding behaviour operating on short and long timescales.


Appetite Regulation , Brain Stem , Eating , Feedback, Physiological , Food , Satiation , Stomach , Appetite Regulation/physiology , Brain Stem/cytology , Brain Stem/physiology , Eating/physiology , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/metabolism , Prolactin-Releasing Hormone/metabolism , Satiation/physiology , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Stomach/physiology , Taste/physiology , Time Factors , Animals , Mice
...