Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.035
Filter
1.
Mol Biol Rep ; 51(1): 624, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38710963

ABSTRACT

BACKGROUND: Thyroid hormones are primarily responsible for the brain development in perinatal mammals. However, this process can be inhibited by external factors such as environmental chemicals. Perinatal mammals are viviparous, which makes direct fetal examination difficult. METHODS: We used metamorphic amphibians, which exhibit many similarities to perinatal mammals, as an experimental system. Therefore, using metamorphic amphibians, we characterized the gene expression of matrix metalloproteinases, which play an important role in brain development. RESULTS: The expression of many matrix metalloproteinases (mmps) was characteristically induced during metamorphosis. We also found that the expression of many mmps was induced by T3 and markedly inhibited by hydroxylated polychlorinated biphenyls (PCBs). CONCLUSION: Overall, our findings suggest that hydroxylated PCBs disrupt normal brain development by disturbing the gene expression of mmps.


Subject(s)
Brain , Matrix Metalloproteinases , Metamorphosis, Biological , Polychlorinated Biphenyls , Thyroid Hormones , Xenopus laevis , Animals , Brain/metabolism , Brain/drug effects , Brain/growth & development , Xenopus laevis/metabolism , Xenopus laevis/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Polychlorinated Biphenyls/toxicity , Metamorphosis, Biological/drug effects , Metamorphosis, Biological/genetics , Thyroid Hormones/metabolism , Gene Expression Regulation, Developmental/drug effects , Hydroxylation
2.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 636-643, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708495

ABSTRACT

OBJECTIVE: To investigate the effect of Jisuikang formula-medicated serum for promoting spinal cord injury (SCI) repair in rats and explore the possible mechanism. METHODS: Thirty adult SD rats were randomized into sham-operated group, SCI (induced using a modified Allen method) model group, and Jisuikang formula-medicated serum treatment group. After the operations, the rats were treated with normal saline or Jisuikang by gavage on a daily basis for 14 days, and the changes in hindlimb motor function of the rats was assessed with Basso-Beattie-Bresnahan (BBB) scores and inclined-plate test. The injured spinal cord tissues were sampled from the SCI rat models for single-cell RNA sequencing, and bioinformatics analysis was performed to identify the target genes of Jisuikang, spinal cord injury and glycolysis. In the cell experiment, cultured astrocytes from neonatal SD rat cortex were treated with SOX2 alone or in combination with Jisuikang-medicated serum for 21 days, and the protein expressions of PKM2, p-PKM2 and YAP and colocalization of PKM2 and YAP in the cells were analyzed with Western blotting and immunofluorescence staining, respectively. RESULTS: The SCI rats with Jisuikang treatment showed significantly improved BBB scores and performance in inclined-plate test. At the injury site, high PKM2 expression was detected in various cell types. Bioinformatic analysis identified the HIPPO-YAP signaling pathway as the target pathway of Jisuikang. In cultured astrocytes, SOX2 combined with the mediated serum, as compared with SOX2 alone, significantly increased PKM2, p-PKM2 and YAP expressions and entry of phosphorylated PKM2 into the nucleus, and promoted PKM2 and YAP co-localization in the cells. CONCLUSION: Jisuikang formula accelerates SCI repair in rats possibly by promoting aerobic glycolysis of the astrocytes via activating the PKM2/YAP axis to induce reprogramming of the astrocytes into neurons.


Subject(s)
Astrocytes , Pyruvate Kinase , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord Injuries , YAP-Signaling Proteins , Animals , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Rats , Astrocytes/metabolism , Astrocytes/drug effects , Signal Transduction/drug effects , Thyroid Hormone-Binding Proteins , Thyroid Hormones/metabolism , Carrier Proteins/metabolism , Drugs, Chinese Herbal/pharmacology , Disease Models, Animal , Membrane Proteins/metabolism
3.
Cells ; 13(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38786060

ABSTRACT

Cells defend against oxidative stress by enhancing antioxidant capacity, including stress-activated metabolic alterations, but the underlying intracellular signaling mechanisms remain unclear. This paper reports that immunoglobulin superfamily containing leucine-rich repeat (ISLR) functions as a redox sensor that responds to reactive oxygen species (ROS) stimulation and modulates the antioxidant capacity by suppressing pyruvate kinase isozyme M2 (PKM2) activity. Following oxidative stress, ISLR perceives ROS stimulation through its cysteine residue 19, and rapidly degrades in the autophagy-lysosome pathway. The downregulated ISLR enhances the antioxidant capacity by promoting the tetramerization of PKM2, and then enhancing the pyruvate kinase activity, PKM2-mediated glycolysis is crucial to the ISLR-mediated antioxidant capacity. In addition, our results demonstrated that, in triple-negative breast cancer, cisplatin treatment reduced the level of ISLR, and PKM2 inhibition sensitizes tumors to cisplatin by enhancing ROS production; and argued that PKM2 inhibition can synergize with cisplatin to limit tumor growth. Our results demonstrate a molecular mechanism by which cells respond to oxidative stress and modulate the redox balance.


Subject(s)
Antioxidants , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species , Humans , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Antioxidants/metabolism , Antioxidants/pharmacology , Oxidative Stress/drug effects , Animals , Cisplatin/pharmacology , Female , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins , Mice , Pyruvate Kinase/metabolism , Glycolysis/drug effects , Autophagy/drug effects , Carrier Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/enzymology
4.
Mol Cancer ; 23(1): 94, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720298

ABSTRACT

BACKGROUND: The hypoxic tumor microenvironment is a key factor that promotes metabolic reprogramming and vascular mimicry (VM) in ovarian cancer (OC) patients. ESM1, a secreted protein, plays an important role in promoting proliferation and angiogenesis in OC. However, the role of ESM1 in metabolic reprogramming and VM in the hypoxic microenvironment in OC patients has not been determined. METHODS: Liquid chromatography coupled with tandem MS was used to analyze CAOV3 and OV90 cells. Interactions between ESM1, PKM2, UBA2, and SUMO1 were detected by GST pull-down, Co-IP, and molecular docking. The effects of the ESM1-PKM2 axis on cell glucose metabolism were analyzed based on an ECAR experiment. The biological effects of the signaling axis on OC cells were detected by tubule formation, transwell assay, RT‒PCR, Western blot, immunofluorescence, and in vivo xenograft tumor experiments. RESULTS: Our findings demonstrated that hypoxia induces the upregulation of ESM1 expression through the transcription of HIF-1α. ESM1 serves as a crucial mediator of the interaction between PKM2 and UBA2, facilitating the SUMOylation of PKM2 and the subsequent formation of PKM2 dimers. This process promotes the Warburg effect and facilitates the nuclear translocation of PKM2, ultimately leading to the phosphorylation of STAT3. These molecular events contribute to the promotion of ovarian cancer glycolysis and vasculogenic mimicry. Furthermore, our study revealed that Shikonin effectively inhibits the molecular interaction between ESM1 and PKM2, consequently preventing the formation of PKM2 dimers and thereby inhibiting ovarian cancer glycolysis, fatty acid synthesis and vasculogenic mimicry. CONCLUSION: Our findings demonstrated that hypoxia increases ESM1 expression through the transcriptional regulation of HIF-1α to induce dimerization via PKM2 SUMOylation, which promotes the OC Warburg effect and VM.


Subject(s)
Carrier Proteins , Fatty Acids , Membrane Proteins , Neoplasm Proteins , Ovarian Neoplasms , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Tumor Microenvironment , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Animals , Thyroid Hormones/metabolism , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Fatty Acids/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Warburg Effect, Oncologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays , Cell Proliferation , Proteoglycans
5.
Expert Rev Endocrinol Metab ; 19(3): 207-215, 2024 May.
Article in English | MEDLINE | ID: mdl-38712738

ABSTRACT

INTRODUCTION: Intracranial aneurysms (IAs) occur in 3-5% of the general population and are characterized by localized structural deterioration of the arterial wall with loss of internal elastic lamina and disruption of the media. The risk of incidence and rupture of aneurysms depends on age, sex, ethnicity, and other different factors, indicating the influence of genetic and environmental factors. When an aneurysm ruptures, there is an estimated 20% mortality rate, along with an added 30-40% morbidity in survivors. The alterations in hormonal levels can influence IAs, while the rupture of an aneurysm can have various impacts on endocrine pathways and affect their outcome. AREA COVERED: This review explores the reciprocal relationship between endocrinological changes (estrogen, growth hormone, and thyroid hormones) and IAs, as well as the effects of aneurysm ruptures on endocrine fluctuations. EXPERT OPINION: Based on the data presented in this paper, we recommend further exploration into the influence of hormones on aneurysm formation and rupture. Additionally, we propose conducting endocrine assessments for patients who have experienced a rupture of IAs. Monitoring hormonal changes in patients with IAs could serve as a potential risk factor for rupture, leading to interventions in the approach to managing IAs.


Subject(s)
Intracranial Aneurysm , Humans , Aneurysm, Ruptured , Risk Factors , Thyroid Hormones/metabolism , Estrogens , Growth Hormone/metabolism , Hormones/metabolism
7.
Article in English | MEDLINE | ID: mdl-38663833

ABSTRACT

Disruption of the thyroid hormone system by synthetic chemicals is gaining attention owing to its potential negative effects on organisms. In this study, the effects of the dio-inhibitor iopanoic acid (IOP) on the levels of thyroid hormone and related gene expression, swim bladder inflation, and swimming performance were investigated in Japanese medaka. Iopanoic acid exposure suppressed thyroid-stimulating hormone ß (tshß), tshß-like, iodotyronin deiodinase 1 (dio1), and dio2 expression, and increased T4 and T3 levels. In addition, IOP exposure inhibited swim bladder inflation, reducing swimming performance. Although adverse outcome pathways of thyroid hormone disruption have been developed using zebrafish, no adverse outcome pathways have been developed using Japanese medaka. This study confirmed that IOP inhibits dio expression (a molecular initiating event), affects T3 and T4 levels (a key event), and reduces swim bladder inflation (a key event) and swimming performance (an adverse outcome) in Japanese medaka.


Subject(s)
Air Sacs , Iopanoic Acid , Oryzias , Swimming , Thyroid Hormones , Animals , Oryzias/physiology , Air Sacs/drug effects , Air Sacs/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/blood , Iopanoic Acid/toxicity , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation/drug effects , Thyroxine/blood , Triiodothyronine/blood , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism
8.
Chemosphere ; 358: 142105, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657690

ABSTRACT

Di(2-ethylhexyl) terephthalate (DEHTP) is an alternative plasticizer widely used in numerous consumer products, replacing di(2-ethylhexyl) phthalate (DEHP). Hence, DEHTP has been frequently detected in the environment and humans. As a structural isomer and functional analog of DEHP, DEHTP is a suspected endocrine disruptor. Here, we evaluated thyroid-disrupting effects of DEHTP using embryo-larval and adult male zebrafish. We also investigated its sex hormone disruption potential in the adult zebrafish. After 5- and 7-days of exposure to DEHTP, significant increases in whole-body thyroid hormonal levels were observed in the larval fish. Down-regulation of several thyroid-regulating genes, including trh, tshß, nis, and dio2, was observed, but only after 5-day exposure. Following a 21-day exposure, the adult male zebrafish exhibited a significant decrease in total triiodothyronine and an increase in thyroid-stimulating hormones. Potential changes in the deiodination of thyroid hormones, supported by the up-regulation of two deiodinases, dio1 and dio3a, along with the down-regulation of dio2, could explain the thyroid hormone changes in the adult zebrafish. Moreover, significant trends of decrease in estradiol and 11-ketotestosterone, along with increase of testosterone (T), were observed in the adult zebrafish. Up-regulation of several steroidogenic genes may explain elevated T, while exact mechanisms of action warrant further investigation. Our results demonstrate that DEHTP can cause disruptions of thyroid and sex hormones at different life stages in zebrafish.


Subject(s)
Endocrine Disruptors , Thyroid Gland , Thyroid Hormones , Zebrafish , Animals , Male , Endocrine Disruptors/toxicity , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Thyroid Hormones/metabolism , Gonadal Steroid Hormones/metabolism , Plasticizers/toxicity , Larva/drug effects , Water Pollutants, Chemical/toxicity , Phthalic Acids/toxicity , Triiodothyronine , Diethylhexyl Phthalate/toxicity , Diethylhexyl Phthalate/analogs & derivatives
9.
Ecotoxicol Environ Saf ; 277: 116348, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38669872

ABSTRACT

Alkylphenols, such as nonylphenol and 4-tert-octylphenol (OP), are byproducts of the biodegradation of alkylphenol ethoxylates and present substantial ecological and health risks in aquatic environments and higher life forms. In this context, our study aimed to explore the effect of OP on reproductive endocrine function in both female and male zebrafish. Over a period of 21 days, the zebrafish were subjected to varying concentrations of OP (0, 0.02, 0.1, and 0.5 µg/L), based on the lowest effective concentration (EC10 = 0.48 µg/L) identified for zebrafish embryos. OP exposure led to a pronounced increase in hepatic vitellogenin (vtg) mRNA expression and 17ß-estradiol biosynthesis in both sexes. Conversely, OP exhibits anti-androgenic properties, significantly diminishes gonadal androgen receptor (ar) mRNA expression, and reduces endogenous androgen (testosterone and 11-ketotestosterone) levels in male zebrafish. Notably, cortisol and thyroid hormone (TH) levels demonstrated concentration-dependent elevations in zebrafish, influencing the regulation of gonadal steroid hormones (GSHs). These findings suggest that prolonged OP exposure may result in sustained reproductive dysfunction in adult zebrafish, which is largely attributable to the intricate reciprocal relationship between hormone levels and the associated gene expression. Our comprehensive biological response analysis of adult zebrafish offers vital insights into the reproductive toxicological effects of OP, thereby enriching future ecological studies on aquatic systems.


Subject(s)
Endocrine Disruptors , Estrogens , Phenols , Receptors, Androgen , Thyroid Hormones , Vitellogenins , Water Pollutants, Chemical , Zebrafish , Animals , Phenols/toxicity , Male , Water Pollutants, Chemical/toxicity , Female , Vitellogenins/metabolism , Endocrine Disruptors/toxicity , Thyroid Hormones/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Estrogens/toxicity , Estradiol/toxicity , Androgen Antagonists/toxicity , Testosterone/metabolism , Testosterone/analogs & derivatives , Hydrocortisone
10.
Nat Metab ; 6(4): 639-650, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38671149

ABSTRACT

Thyroid hormones (THs) are key hormones that regulate development and metabolism in mammals. In man, the major target tissues for TH action are the brain, liver, muscle, heart, and adipose tissue. Defects in TH synthesis, transport, metabolism, and nuclear action have been associated with genetic and endocrine diseases in man. Over the past few years, there has been renewed interest in TH action and the therapeutic potential of THs and thyromimetics to treat several metabolic disorders such as hypercholesterolemia, dyslipidaemia, non-alcoholic fatty liver disease (NAFLD), and TH transporter defects. Recent advances in the development of tissue and TH receptor isoform-targeted thyromimetics have kindled new hope for translating our fundamental understanding of TH action into an effective therapy. This review provides a concise overview of the historical development of our understanding of TH action, its physiological and pathophysiological effects on metabolism, and future therapeutic applications to treat metabolic dysfunction.


Subject(s)
Thyroid Hormones , Humans , Thyroid Hormones/metabolism , Animals , Metabolic Diseases/metabolism , Receptors, Thyroid Hormone/metabolism
11.
Eur J Pharmacol ; 972: 176551, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38570082

ABSTRACT

Fibroblast-like synoviocytes (FLS) play an important role in rheumatoid arthritis (RA)-related swelling and bone damage. Therefore, novel targets for RA therapy in FLS are urgently discovered for improving pathologic phenomenon, especially joint damage and dyskinesia. Here, we suggested that pyruvate kinase M2 (PKM2) in FLS represented a pharmacological target for RA treatment by antimalarial drug artemisinin (ART). We demonstrated that ART selectively inhibited human RA-FLS and rat collagen-induced arthritis (CIA)-FLS proliferation and migration without observed toxic effects. In particular, the identification of targets revealed that PKM2 played a crucial role as a primary regulator of the cell cycle, leading to the heightened proliferation of RA-FLS. ART exhibited a direct interaction with PKM2, resulting in an allosteric modulation that enhances the lactylation modification of PKM2. This interaction further promoted the binding of p300, ultimately preventing the nuclear translocation of PKM2 and inducing cell cycle arrest at the S phase. In vivo, ART obviously suppressed RA-mediated synovial hyperplasia, bone damage and inflammatory response to further improve motor behavior in CIA-rats. Taken together, these findings indicate that directing interventions towards PKM2 in FLS could offer a hopeful avenue for pharmaceutical treatments of RA through the regulation of cell cycle via PKM2 lactylation.


Subject(s)
Arthritis, Rheumatoid , Cell Proliferation , Synoviocytes , Synoviocytes/drug effects , Synoviocytes/metabolism , Synoviocytes/pathology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Animals , Cell Proliferation/drug effects , Humans , Rats , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Pyruvate Kinase/metabolism , Thyroid Hormone-Binding Proteins , Male , Thyroid Hormones/metabolism , Arthritis, Experimental/pathology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Cell Movement/drug effects , Molecular Targeted Therapy , Membrane Proteins/metabolism , Carrier Proteins/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry
12.
Sci Rep ; 14(1): 9355, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38654093

ABSTRACT

Thyroid hormones (TH) play critical roles during nervous system development and patients carrying coding variants of MCT8 (monocarboxylate transporter 8) or THRA (thyroid hormone receptor alpha) present a spectrum of neurological phenotypes resulting from perturbed local TH action during early brain development. Recently, human cerebral organoids (hCOs) emerged as powerful in vitro tools for disease modelling recapitulating key aspects of early human cortex development. To begin exploring prospects of this model for thyroid research, we performed a detailed characterization of the spatiotemporal expression of MCT8 and THRA in developing hCOs. Immunostaining showed MCT8 membrane expression in neuronal progenitor cell types including early neuroepithelial cells, radial glia cells (RGCs), intermediate progenitors and outer RGCs. In addition, we detected robust MCT8 protein expression in deep layer and upper layer neurons. Spatiotemporal SLC16A2 mRNA expression, detected by fluorescent in situ hybridization (FISH), was highly concordant with MCT8 protein expression across cortical cell layers. FISH detected THRA mRNA expression already in neuroepithelium before the onset of neurogenesis. THRA mRNA expression remained low in the ventricular zone, increased in the subventricular zone whereas strong THRA expression was observed in excitatory neurons. In combination with a robust up-regulation of known T3 response genes following T3 treatment, these observations show that hCOs provide a promising and experimentally tractable model to probe local TH action during human cortical neurogenesis and eventually to model the consequences of impaired TH function for early cortex development.


Subject(s)
Cerebral Cortex , Monocarboxylic Acid Transporters , Neurogenesis , Organoids , RNA, Messenger , Symporters , Thyroid Hormone Receptors alpha , Female , Humans , Pregnancy , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/metabolism , Organoids/metabolism , Pregnancy Trimester, First/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Symporters/genetics , Symporters/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/genetics
13.
J Autoimmun ; 145: 103198, 2024 May.
Article in English | MEDLINE | ID: mdl-38428341

ABSTRACT

OBJECTIVES: Expansion of follicular helper T (Tfh) cells and abnormal glucose metabolism are present in patients with systemic lupus erythematosus (SLE). Pyruvate kinase M2 (PKM2) is one of the key glycolytic enzymes, and the underlying mechanism of PKM2-mediated Tfh cell glycolysis in SLE pathogenesis remains elusive. METHODS: We analyzed the percentage of Tfh cells and glycolysis in CD4+ T cells from SLE patients and healthy donors and performed RNA sequencing analysis of peripheral blood CD4+ T cells and differentiated Tfh cells from SLE patients. Following Tfh cell development in vitro and following treatment with PKM2 activator TEPP-46, PKM2 expression, glycolysis, and signaling pathway proteins were analyzed. Finally, diseased MRL/lpr mice were treated with TEPP-46 and assessed for treatment effects. RESULTS: We found that Tfh cell percentage and glycolysis levels were increased in SLE patients and MRL/lpr mice. TEPP-46 induced PKM2 tetramerization, thereby inhibiting Tfh cell glycolysis levels. On the one hand, TEPP-46 reduced the dimeric PKM2 entering the nucleus and reduced binding to the transcription factor BCL6. On the other hand, TEPP-46 inhibited the AKT/GSK-3ß pathway and glycolysis during Tfh cell differentiation. Finally, we confirmed that TEPP-46 effectively alleviated inflammatory damage in lupus-prone mice and reduced the expansion of Tfh cells in vivo. CONCLUSIONS: Our results demonstrate the involvement of PKM2-mediated glycolysis in Tfh cell differentiation and SLE pathogenesis, and PKM2 could be a key therapeutic target for the treatment of SLE.


Subject(s)
Cell Differentiation , Disease Models, Animal , Glycolysis , Lupus Erythematosus, Systemic , Mice, Inbred MRL lpr , T Follicular Helper Cells , Animals , Mice , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Humans , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism , Female , Pyruvate Kinase/metabolism , Inflammation/immunology , Inflammation/metabolism , Thyroid Hormones/metabolism , Signal Transduction , Thyroid Hormone-Binding Proteins , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics
14.
J Biol Chem ; 300(4): 107139, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447792

ABSTRACT

Androgen receptor (AR) is one of the key targets for the treatment of castration-resistant prostate cancer (CRPC). Current endocrine therapy can greatly improve patients with CRPC. However, with the change of pathogenic mechanism, acquired resistance often leads to the failure of treatment. Studies have shown that tanshinone IIA (TS-IIA) and its derivatives have significant antitumor activity, and have certain AR-targeting effects, but the mechanism is unknown. In this study, the TS-IIA analog TB3 was found to significantly inhibit the growth of CRPC in vitro and in vivo. Molecular docking, cellular thermal shift assay, and cycloheximide experiments confirmed that AR was the target of TB3 and promoted the degradation of AR. Furthermore, TB3 can significantly inhibit glycolysis metabolism by targeting the AR/PKM2 axis. The addition of pyruvic acid could significantly alleviate the inhibitory effect of TB3 on CRPC cells. Besides, the knockdown of AR or PKM2 also could reverse the effect of TB3 on CRPC cells. Taken together, our study suggests that TS-IIA derivative TB3 inhibits glycolysis to prevent the CRPC process by targeting the AR/PKM2 axis.


Subject(s)
Abietanes , Glycolysis , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen , Thyroid Hormone-Binding Proteins , Animals , Humans , Male , Mice , Abietanes/pharmacology , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Glycolysis/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Nude , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Thyroid Hormones/metabolism
15.
Biochim Biophys Acta Rev Cancer ; 1879(3): 189089, 2024 May.
Article in English | MEDLINE | ID: mdl-38458358

ABSTRACT

Pyruvate Kinase M2, a key enzyme in glycolysis, has garnered significant attention in cancer research due to its pivotal role in the metabolic reprogramming of cancer cells. Originally identified for its association with the Warburg effect, PKM2 has emerged as a multifaceted player in cancer biology. The functioning of PKM2 is intricately regulated at multiple levels, including controlling the gene expression via various transcription factors and non-coding RNAs, as well as adding post-translational modifications that confer distinct functions to the protein. Here, we explore the diverse functions of PKM2, encompassing newly emerging roles in non-glycolytic metabolic regulation, immunomodulation, inflammation, DNA repair and mRNA processing, beyond its canonical role in glycolysis. The ever-expanding list of its functions has recently grown to include roles in subcellular compartments such as the mitochondria and extracellular milieu as well, all of which make PKM2 an attractive drug target in the pursuit of therapeutics for cancer.


Subject(s)
Glycolysis , Neoplasms , Warburg Effect, Oncologic , Humans , Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/genetics , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Membrane Proteins/metabolism , Animals , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , DNA Repair
16.
FEBS Open Bio ; 14(5): 843-854, 2024 May.
Article in English | MEDLINE | ID: mdl-38514913

ABSTRACT

Glioblastoma (GBM) is a primary tumor in the central nervous system with poor prognosis. It exhibits elevated glucose uptake and lactate production. This metabolic state of aerobic glycolysis is known as the Warburg effect. N6-isopentenyladenosine (iPA), a natural cytokine modified with an isopentenyl moiety derived from the mevalonate pathway, has well-established anti-tumor activity. It inhibits cell proliferation in glioma cells, inducing cell death by apoptosis and/or necroptosis. In the present study, we found that iPA inhibits aerobic glycolysis in unmodified U87MG cells and in the same cell line engineered to over-express wild-type epidermal growth factor receptor (EGFR) or EGFR variant III (vIII), as well as in a primary GBM4 patient-derived cell line. The detection of glycolysis showed that iPA treatment suppressed ATP and lactate production. We also evaluated the response of iPA treatment in normal human astrocyte primary cells, healthy counterpart cells of the brain. Aerobic glycolysis in treated normal human astrocyte cells did not show significant changes compared to GBM cells. To determine the mechanism of iPA action on aerobic glycolysis, we investigated the expression of certain enzymes involved in this metabolic pathway. We observed that iPA reduced the expression of pyruvate kinase M2 (PKM2), which plays a key role in the regulation of aerobic glycolysis, promoting tumor cell proliferation. The reduction of PKM2 expression is a result of the inhibition of the inhibitor of nuclear factor kappa-B kinase subunit, beta/nuclear factor-kappa B pathway upon iPA treatment. In conclusion, these experimental results show that iPA may inhibit aerobic glycolysis of GBM in stabilized cell lines and primary GBM cells by targeting the expression and activity of PKM2.


Subject(s)
Carrier Proteins , Cell Proliferation , Glioblastoma , Glycolysis , Isopentenyladenosine , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Thyroid Hormones/metabolism , Glycolysis/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Isopentenyladenosine/pharmacology , Isopentenyladenosine/metabolism , Cell Proliferation/drug effects , Cell Line, Tumor , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects
17.
BMC Mol Cell Biol ; 25(1): 11, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553695

ABSTRACT

Disruption of circadian rhythms is associated with neurological, endocrine and metabolic pathologies. We have recently shown that mice lacking functional type 3 deiodinase (DIO3), the enzyme that clears thyroid hormones, exhibit a phase shift in locomotor activity, suggesting altered circadian rhythm. To better understand the physiological and molecular basis of this phenotype, we used Dio3+/+ and Dio3-/- mice of both sexes at different zeitgeber times (ZTs) and analyzed corticosterone and thyroxine (T4) levels, hypothalamic, hepatic, and adipose tissue expression of clock genes, as well as genes involved in the thyroid hormone action or physiology of liver and adipose tissues. Wild type mice exhibited sexually dimorphic circadian patterns of genes controlling thyroid hormone action, including Dio3. Dio3-/- mice exhibited altered hypothalamic expression of several clock genes at ZT12, but did not disrupt the overall circadian profile. Expression of clock genes in peripheral tissues was not disrupted by Dio3 deficiency. However, Dio3 loss in liver and adipose tissues disrupted circadian profiles of genes that determine tissue thyroid hormone action and physiology. We also observed circadian-specific changes in serum T4 and corticosterone as a result of DIO3 deficiency. The circadian alterations manifested sexual dimorphism. Most notable, the time curve of serum corticosterone was flattened in Dio3-/- females. We conclude that Dio3 exhibits circadian variations, influencing the circadian rhythmicity of thyroid hormone action and physiology in liver and adipose tissues in a sex-specific manner. Circadian disruptions in tissue physiology may then contribute to the metabolic phenotypes of DIO3-deficient mice.


Subject(s)
Corticosterone , Iodide Peroxidase , Male , Female , Mice , Animals , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Thyroid Hormones/metabolism , Circadian Rhythm/genetics , Gene Expression
18.
Sci Rep ; 14(1): 7200, 2024 03 26.
Article in English | MEDLINE | ID: mdl-38531895

ABSTRACT

Unlike other thyroid hormone receptors (THRs), the beta 2 isoform (THRB2) has a restricted expression pattern and is uniquely and abundantly phosphorylated at a conserved serine residue S101 (S102 in humans). Using tagged and or phosphorylation-defective (S101A) THRB2 mutant mice, we show that THRB2 is present in a large subset of POMC neurons and mitigates ROS accumulation during ROS-triggering events, such as fasting/refeeding or high fat diet (HFD). Excessive ROS accumulation in mutant POMC neurons was accompanied by a skewed production of orexigenic/anorexigenic hormones, resulting in elevated food intake. The prolonged exposure to pathogenic hypothalamic ROS levels during HFD feeding lead to a significant loss of POMC neurons in mutant versus wild-type (WT) mice. In cultured cells, the presence of WT THRB2 isoform, but not other THRs, or THRB2S101A, reduced ROS accumulation upon exogenous induction of oxidative stress by tert-butyl hydroperoxide. The protective function of phospho-THRB2 (pTHRB2) did not require thyroid hormone (TH), suggesting a TH-independent role of the THRB2 isoform, and phospho-S101 in particular, in regulating oxidative stress. We propose that pTHRB2 has a fundamental role in neuronal protection against ROS cellular damage, and mitigates hypothalamic pathological changes found in diet-induced obesity.


Subject(s)
Hypothalamus , Pro-Opiomelanocortin , Humans , Mice , Animals , Reactive Oxygen Species/metabolism , Phosphorylation , Pro-Opiomelanocortin/metabolism , Hypothalamus/metabolism , Feeding Behavior , Thyroid Hormones/metabolism , Diet, High-Fat , Receptors, Thyroid Hormone/metabolism , Protein Isoforms/metabolism , Mice, Inbred C57BL
19.
Drug Discov Today ; 29(5): 103949, 2024 May.
Article in English | MEDLINE | ID: mdl-38492882

ABSTRACT

Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme that regulates proliferating cell metabolism. The role of PKM2 in common diseases has been well established, but its role in rare diseases (RDs) is less understood. Over the past few years, PKM2 has emerged as a crucial player in RDs, including, neoplastic, respiratory, metabolic, and neurological disorders. Herein, we summarize recent findings and developments highlighting PKM2 as an emerging key player in RDs. We also discuss the current status of PKM2 modulation in RDs with particular emphasis on preclinical and clinical studies in addition to current challenges in the field.


Subject(s)
Rare Diseases , Humans , Animals , Rare Diseases/drug therapy , Thyroid Hormone-Binding Proteins , Pyruvate Kinase/metabolism , Thyroid Hormones/metabolism , Membrane Proteins/metabolism , Carrier Proteins/metabolism
20.
Front Endocrinol (Lausanne) ; 15: 1360188, 2024.
Article in English | MEDLINE | ID: mdl-38529399

ABSTRACT

Thyroid hormones are involved in many biological processes such as neurogenesis, metabolism, and development. However, compounds called endocrine disruptors can alter thyroid hormone signaling and induce unwanted effects on human and ecosystems health. Regulatory tests have been developed to detect these compounds but need to be significantly improved by proposing novel endpoints and key events. The Xenopus Eleutheroembryonic Thyroid Assay (XETA, OECD test guideline no. 248) is one such test. It is based on Xenopus laevis tadpoles, a particularly sensitive model system for studying the physiology and disruption of thyroid hormone signaling: amphibian metamorphosis is a spectacular (thus easy to monitor) life cycle transition governed by thyroid hormones. With a long-term objective of providing novel molecular markers under XETA settings, we propose first to describe the differential effects of thyroid hormones on gene expression, which, surprisingly, are not known. After thyroid hormones exposure (T3 or T4), whole tadpole RNAs were subjected to transcriptomic analysis. By using standard approaches coupled to system biology, we found similar effects of the two thyroid hormones. They impact the cell cycle and promote the expression of genes involves in cell proliferation. At the level of the whole tadpole, the immune system is also a prime target of thyroid hormone action.


Subject(s)
Ecosystem , Thyroid Hormones , Animals , Humans , Xenopus laevis/metabolism , Thyroid Hormones/metabolism , Thyroid Gland/metabolism , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL
...