Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.126
Filter
1.
Clin Chim Acta ; 564: 119929, 2025 Jan 01.
Article in English | MEDLINE | ID: mdl-39154700

ABSTRACT

Irisin, a hormone-like adipo-myokine, has garnered considerable attention in recent years for its potential impact in metabolic diseases. Its physiological effects are similar to those of thyroid hormones, prompting numerous investigations into potential correlations and interactions between irisin and thyroid function through various in vitro and animal experiments. However, existing studies suggest that the relationship between irisin and thyroid diseases is highly complex and multifaceted. In this paper, we have summarized the research results on serum irisin and thyroid function, providing an overview of advancements and constraints in current research on irisin and thyroid hormones. The aim is to offer insights and directions for future clinical trials in this field.


Subject(s)
Fibronectins , Thyroid Diseases , Humans , Fibronectins/blood , Fibronectins/metabolism , Thyroid Diseases/blood , Thyroid Diseases/metabolism , Animals , Thyroid Hormones/blood , Thyroid Hormones/metabolism
2.
Cell Metab ; 36(10): 2170-2172, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39357508

ABSTRACT

Classically, the central actions of thyroid hormones (THs) on metabolism occur within the hypothalamus. A recent article published in Cell by Sabatini and colleagues demonstrates that TH modulates cerebral cortical circuits of male mice, which might integrate exploratory behavior and whole-body metabolism.


Subject(s)
Cerebral Cortex , Thyroid Hormones , Animals , Thyroid Hormones/metabolism , Mice , Male , Cerebral Cortex/metabolism , Hypothalamus/metabolism , Exploratory Behavior , Humans
3.
Cell Death Dis ; 15(10): 728, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39368995

ABSTRACT

Patients with lung adenocarcinoma (LUAD) generally have poor prognosis. Abnormal cellular energy metabolism is a hallmark of LUAD. Glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) is a member of the γ-glutamylcyclotransferase family and an unfolded protein response pathway regulatory gene. Its biological function and molecular regulatory mechanism, especially regarding energy metabolism underlying LUAD, remain unclear. By utilizing tissue microarray and data from The Cancer Genome Atlas and Gene Expression Omnibus, we found that CHAC1 expression was markedly higher in LUAD tissues than in non-tumor tissues, and was positively correlated with poor prognosis. Phenotypically, CHAC1 overexpression enhanced the proliferation, migration, invasion, tumor sphere formation, and glycolysis ability of LUAD cells, resulting in tumor growth both in vitro and in vivo. Mechanistically, through a shotgun mass spectrometry-based proteomic approach and high-throughput RNA sequencing, we found that CHAC1 acted as a bridge connecting UBA2 and PKM2, enhancing the SUMOylation of PKM2. The SUMOylated PKM2 then transferred from the cytoplasm to the nucleus, activating the expression of glycolysis-related genes and enhancing the Warburg effect. Lastly, E2F Transcription Factor 1 potently activated CHAC1 transcription by directly binding to the CHAC1 promoter in LUAD cells. The results of this study implied that CHAC1 regulates energy metabolism and promotes glycolysis in LUAD progression.


Subject(s)
Adenocarcinoma of Lung , Carrier Proteins , Glucose , Lung Neoplasms , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Thyroid Hormones/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Glucose/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Animals , Disease Progression , gamma-Glutamylcyclotransferase/metabolism , gamma-Glutamylcyclotransferase/genetics , Mice , Cell Line, Tumor , Cell Proliferation , Mice, Nude , Cell Nucleus/metabolism , Male , Gene Expression Regulation, Neoplastic , Glycolysis , Female , Cell Movement , Mice, Inbred BALB C
4.
Sci Rep ; 14(1): 23664, 2024 10 10.
Article in English | MEDLINE | ID: mdl-39390134

ABSTRACT

The widespread presence of microplastics in the environment has raised significant concerns regarding their potential impact on human and animal health. Among various microplastic types, polyethylene microplastics (PE-MPs) are particularly prevalent due to the extensive use in packaging and consumer products. Exploring the uncharted therapeutic potentials of naringin, this study delves into its mitigating effects on disruptions in kallikrein-3 levels, steroidal-thyroidal hormone balance, and antioxidant defense triggered by PE-MPs exposure, paving the way for novel interventions in environmental toxin-induced endocrine and oxidative stress disorders. Male Wistar rats (n = 24) were randomly grouped into four: Control, PE-MPs (1.5 mg/kg), PE-MPs + NAR (1.5 mg/kg PE-MPs + 100 mg/kg NAR), and NAR (100 mg/kg). Hormonal and antioxidant parameters were assessed after 28 days of exposure. PE-MPs exposure caused a significant increase(p < 0.005) in the level of kallikrein-3 (KLK-3) while it significantly reduces the levels of testosterone (TST), luteinizing hormone, thyroid stimulating hormone (TSH) and Free-triiodothyronine (fT3) and Total cholesterol (TChol) concentration. PE-MPs exposure also disrupted significantly (p < 0.005) antioxidant profile by down-regulating the activities of glutathione-S-transferase, catalase (CAT), superoxide dismutase (SOD) and reducing levels of glutathione (GSH) and ascorbic acid (AA) while concentration of malondialdehyde (MDA) levels were increased relative to control. However, the mitigating potentials of naringin on disruptions in hormonal and antioxidant profiles caused by PE-MPs exposure were demonstrated, as NAR normalized KLK-3, steroid, and thyroid hormone levels, cholesterol concentration, and enhanced antioxidant defense. This suggests that NAR is a promising protective agent against endocrine and oxidative damage induced by environmental contaminants such as microplastics.


Subject(s)
Antioxidants , Flavanones , Microplastics , Polyethylene , Rats, Wistar , Animals , Male , Antioxidants/metabolism , Flavanones/pharmacology , Rats , Microplastics/toxicity , Oxidative Stress/drug effects , Thyroid Hormones/metabolism , Kallikreins/metabolism , Thyrotropin/blood , Thyrotropin/metabolism
5.
J Transl Med ; 22(1): 914, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380010

ABSTRACT

The heterogeneous nuclear ribonucleoprotein C (HNRNPC) plays a crucial role in tumorigenesis, yet its role in papillary thyroid carcinoma (PTC) remains elusive. Herein, we elucidated the function and molecular mechanism of HNRNPC in PTC tumorigenesis and progression. Our study unveiled a significant upregulation of HNRNPC in PTC, and knockdown of HNRNPC markedly inhibited the proliferation, invasion, and metastasis of BCPAP cells. Furthermore, HNRNPC modulated PKM alternative splicing in BCPAP cells primarily through m6A modification. Additionally, by upregulating PKM2 expression, HNRNPC promoted aerobic glycolysis in BCPAP cells, thereby facilitating malignant progression in PTC. In summary, our findings demonstrate that HNRNPC regulates PKM alternative splicing through m6A methylation modification and promotes the proliferation, invasion and metastasis of PTC through glucose metabolism pathways mediated by PKM2. These discoveries provide new biomarkers for screening and diagnosing PTC patients and offer novel therapeutic targets for personalized treatment strategies.


Subject(s)
Alternative Splicing , Carrier Proteins , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Glycolysis , Heterogeneous-Nuclear Ribonucleoprotein Group C , Membrane Proteins , Thyroid Cancer, Papillary , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Thyroid Neoplasms , Up-Regulation , Humans , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Up-Regulation/genetics , Cell Line, Tumor , Membrane Proteins/metabolism , Membrane Proteins/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Alternative Splicing/genetics , Thyroid Hormones/metabolism , Glycolysis/genetics , Methylation , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Animals , Neoplasm Invasiveness , Neoplasm Metastasis , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Mice, Nude , Adenosine/analogs & derivatives , Adenosine/metabolism
6.
Mol Cell ; 84(19): 3574-3575, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39366344

ABSTRACT

In this issue of Molecular Cell, Anastasakis et al. describe a novel function of the metabolic enzyme PKM2 as an RNA G-quadruplex binding protein, which could contribute to cancer biology.


Subject(s)
Carrier Proteins , G-Quadruplexes , Membrane Proteins , Neoplasms , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Transcriptome , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/enzymology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Gene Expression Regulation, Neoplastic
7.
Endocrinology ; 165(11)2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39279445

ABSTRACT

Thyroid hormone regulates the rate of testis maturation in mammals. Manipulations of thyroid hormone levels in neonatal animals affect various aspects of testis biology. However, there have been no studies examining the effects of thyroid hormone on the rete testis (RT). Here, we used animal models of neonatal hyperthyroidism (injections of triiodothyronine, or T3) and hypothyroidism (goitrogen 6-propyl-2-thiouracil [PTU] treatment) and found that higher levels of thyroid hormone accelerate RT development, while lower levels of thyroid hormone delay it. T3 and PTU treatments influence RT size, proliferation of RT cells, and expression of DMRT1 and androgen receptor in the RT. T3 supplementation accelerates RT development in an organ testicular culture, which indicates the local action of thyroid hormone. Additionally, it was found that follicle-stimulating hormone could be involved in the regulation both of RT proliferation and RT size. The fact that RT cells in a cell culture do not respond to T3 suggests indirect action of thyroid hormone on the RT in vivo or the loss of the responsiveness to the hormone in vitro.


Subject(s)
Animals, Newborn , Testis , Thyroid Hormones , Triiodothyronine , Animals , Male , Mice , Testis/drug effects , Testis/metabolism , Testis/growth & development , Triiodothyronine/pharmacology , Thyroid Hormones/metabolism , Thyroid Hormones/pharmacology , Propylthiouracil/pharmacology , Cell Proliferation/drug effects , Hyperthyroidism/chemically induced , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Hypothyroidism/chemically induced , Follicle Stimulating Hormone/pharmacology , Receptors, Androgen/metabolism
8.
Int J Mol Sci ; 25(18)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39337448

ABSTRACT

Myxedema is a potentially life-threatening condition typically observed in severe hypothyroidism. However, localized or diffuse myxedema is also observed in hyperthyroidism. The exact cause and mechanism of this paradoxical situation is not clear. We report here the analysis of body fluid distribution by bioelectrical impedance analysis (BIA) in 103 thyroid patients, subdivided according to their functional status. All BIA parameters measured in subclinical thyroid dysfunctions did not significantly differ from those observed in euthyroid controls. On the contrary, they were clearly altered in the two extreme, opposite conditions of thyroid dysfunctions, namely overt hyperthyroidism and severe hypothyroidism, indicating the occurrence of a typical hormetic condition. Surprisingly, differences in BIA parameters related to fluid body composition were even more evident in hyperthyroidism than in hypothyroidism. A hormetic response to thyroid hormone (TH)s was previously reported to explain the paradoxical, biphasic, time- and dose-dependent effects on other conditions. Our results indicate that myxedema, observed in both hypothyroid and hyperthyroid conditions, represents another example of a hormetic-type response to THs. BIA offers no additional valuable information in evaluating fluid body composition in subclinical thyroid dysfunctions, but it represents a valuable method to analyze and monitor body fluid composition and distribution in overt and severe thyroid dysfunctions.


Subject(s)
Hyperthyroidism , Hypothyroidism , Myxedema , Humans , Hypothyroidism/complications , Hypothyroidism/metabolism , Hyperthyroidism/complications , Hyperthyroidism/metabolism , Hyperthyroidism/physiopathology , Female , Male , Middle Aged , Adult , Electric Impedance , Hormesis , Aged , Thyroid Hormones/metabolism , Thyroid Hormones/blood , Body Composition
10.
Elife ; 132024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264698

ABSTRACT

Reactive astrocytes play critical roles in the occurrence of various neurological diseases such as multiple sclerosis. Activation of astrocytes is often accompanied by a glycolysis-dominant metabolic switch. However, the role and molecular mechanism of metabolic reprogramming in activation of astrocytes have not been clarified. Here, we found that PKM2, a rate-limiting enzyme of glycolysis, displayed nuclear translocation in astrocytes of EAE (experimental autoimmune encephalomyelitis) mice, an animal model of multiple sclerosis. Prevention of PKM2 nuclear import by DASA-58 significantly reduced the activation of mice primary astrocytes, which was observed by decreased proliferation, glycolysis and secretion of inflammatory cytokines. Most importantly, we identified the ubiquitination-mediated regulation of PKM2 nuclear import by ubiquitin ligase TRIM21. TRIM21 interacted with PKM2, promoted its nuclear translocation and stimulated its nuclear activity to phosphorylate STAT3, NF-κB and interact with c-myc. Further single-cell RNA sequencing and immunofluorescence staining demonstrated that TRIM21 expression was upregulated in astrocytes of EAE. TRIM21 overexpressing in mice primary astrocytes enhanced PKM2-dependent glycolysis and proliferation, which could be reversed by DASA-58. Moreover, intracerebroventricular injection of a lentiviral vector to knockdown TRIM21 in astrocytes or intraperitoneal injection of TEPP-46, which inhibit the nuclear translocation of PKM2, effectively decreased disease severity, CNS inflammation and demyelination in EAE. Collectively, our study provides novel insights into the pathological function of nuclear glycolytic enzyme PKM2 and ubiquitination-mediated regulatory mechanism that are involved in astrocyte activation. Targeting this axis may be a potential therapeutic strategy for the treatment of astrocyte-involved neurological disease.


Subject(s)
Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Ribonucleoproteins , Up-Regulation , Animals , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Mice , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mice, Inbred C57BL , Pyruvate Kinase/metabolism , Pyruvate Kinase/genetics , Active Transport, Cell Nucleus , Female , Glycolysis , Ubiquitination , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Nucleus/metabolism
11.
Front Endocrinol (Lausanne) ; 15: 1443051, 2024.
Article in English | MEDLINE | ID: mdl-39253586

ABSTRACT

The hypometabolism induced by fasting has great potential in maintaining health and improving survival in extreme environments, among which thyroid hormone (TH) plays an important role in the adaptation and the formation of new energy metabolism homeostasis during long-term fasting. In the present review, we emphasize the potential of long-term fasting to improve physical health and emergency rescue in extreme environments, introduce the concept and pattern of fasting and its impact on the body's energy metabolism consumption. Prolonged fasting has more application potential in emergency rescue in special environments. The changes of THs caused by fasting, including serum biochemical characteristics, responsiveness of the peripheral and central hypothalamus-pituitary-thyroid (HPT) axis, and differential changes of TH metabolism, are emphasized in particular. It was proposed that the variability between brain and liver tissues in THs uptake, deiodination activation and inactivation is the key regulatory mechanism for the cause of peripheral THs decline and central homeostasis. While hypothalamic tanycytes play a pivotal role in the fine regulation of the HPT negative feedback regulation during long-term fasting. The study progress of tanycytes on thyrotropin-releasing hormone (TRH) release and deiodination is described in detail. In conclusion, the combination of the decrease of TH metabolism in peripheral tissues and stability in the central HPT axis maintains the basal physiological requirement and new energy metabolism homeostasis to adapt to long-term food scarcity. The molecular mechanisms of this localized and differential regulation will be a key research direction for developing measures for hypometabolic applications in extreme environment.


Subject(s)
Energy Metabolism , Fasting , Thyroid Hormones , Humans , Fasting/metabolism , Fasting/physiology , Thyroid Hormones/metabolism , Animals , Energy Metabolism/physiology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Thyroid Gland/metabolism , Thyroid Gland/physiology , Homeostasis
12.
Mol Biol Rep ; 51(1): 969, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249564

ABSTRACT

BACKGROUND: Mitochondria are known to be involved in mediating the calorigenic effects of thyroid hormones. With an abundance of these hormones, alterations in energy metabolism and cellular respiration take place, leading to the development of cardiac hypertrophy. Vitamin D has recently gained attention due to its involvement in the regulation of mitochondrial function, demonstrating promising potential in preserving the integrity and functionality of the mitochondrial network. The present study aimed to investigate the therapeutic potential of Vitamin D on cardiac hypertrophy induced by hyperthyroidism, with a focus on the contributions of mitophagy and apoptosis as possible underlying molecular mechanisms. METHODS AND RESULTS: The rats were divided into three groups: control; hyperthyroid; hyperthyroid + Vitamin D. Hyperthyroidism was induced by Levothyroxine administration for four weeks. Serum thyroid hormones levels, myocardial damage markers, cardiac hypertrophy indices, and histological examination were assessed. The assessment of Malondialdehyde (MDA) levels and the expression of the related genes were conducted using heart tissue samples. Vitamin D pretreatment exhibited a significant improvement in the hyperthyroidism-induced decline in markers indicative of myocardial damage, oxidative stress, and indices of cardiac hypertrophy. Vitamin D pretreatment also improved the downregulation observed in myocardial expression levels of genes involved in the regulation of mitophagy and apoptosis, including PTEN putative kinase 1 (PINK1), Mitofusin-2 (MFN2), Dynamin-related Protein 1 (DRP1), and B cell lymphoma-2 (Bcl-2), induced by hyperthyroidism. CONCLUSIONS: These results suggest that supplementation with Vitamin D could be advantageous in preventing the progression of cardiac hypertrophy and myocardial damage.


Subject(s)
Apoptosis , Cardiomegaly , Cardiotonic Agents , Disease Models, Animal , Hyperthyroidism , Mitophagy , Thyroxine , Vitamin D , Animals , Hyperthyroidism/complications , Hyperthyroidism/metabolism , Hyperthyroidism/drug therapy , Mitophagy/drug effects , Apoptosis/drug effects , Rats , Thyroxine/pharmacology , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Vitamin D/pharmacology , Male , Cardiotonic Agents/pharmacology , Oxidative Stress/drug effects , Rats, Wistar , Myocardium/metabolism , Myocardium/pathology , Protein Kinases/metabolism , Protein Kinases/genetics , Malondialdehyde/metabolism , Thyroid Hormones/metabolism
13.
Med Sci Monit ; 30: e945444, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39318065

ABSTRACT

BACKGROUND Technetium (99mTc)-labelled Methoxy-2-Isobutylisonitrile (MIBI) is a diagnostic lipophilic cationic radiotracer used to evaluate the cardiac, breast, thyroid, and parathyroid pathology. This study aimed to evaluate the role of MIBI combined with Tc-99m pertechnetate thyroid scintigraphy, thyroid ultrasonography, and measurement of thyrotropin, thyroid hormones, and autoantibodies to subtype amiodarone-induced thyrotoxicosis (AIT) and the contribution of semi-quantitative analysis of MIBI uptake. MATERIAL AND METHODS This cross-sectional study included 36 patients with AIT who underwent thyrotropin, thyroid hormone, and autoantibody analysis using chemiluminescent method, ultrasonography, pertechnetate, and MIBI thyroid scintigraphy with semi-quantitative uptake, including calculation of the target-to-background ratio (TBR) with 2 different background regions. The MIBI washout rate (WR) was analyzed in all groups. Statistical analysis was performed using descriptive statistics, correlations, and the receiver operating characteristic curve - area under the curve (ROC-AUC). The results were compared with the control group. RESULTS Based on visual and semi-quantitative analyses, patients were successfully categorized into AIT groups (AIT-1, AIT-2 and AIT-3) but the latter method enabled better differentiation of MIBI uptake between all groups. Additionally, ROC-AUC analysis determined cutoff values which enabled discerning between AIT-1 and AIT-2 groups, and AIT-1 and AIT-3 groups. WR showed no significant difference between all AIT groups and controls (P>0.05). CONCLUSIONS Visual MIBI analysis enabled differentiation between AIT-1 and 2 groups, but the method was substantially improved with semi-quantitative analysis, especially in defining AIT-3 group. However, multicenter collaboration with larger studies is needed to standardize the method and obtain more accurate and consistent results.


Subject(s)
Amiodarone , Technetium Tc 99m Sestamibi , Thyroid Gland , Thyrotoxicosis , Humans , Amiodarone/adverse effects , Thyrotoxicosis/chemically induced , Thyrotoxicosis/diagnostic imaging , Female , Pilot Projects , Male , Middle Aged , Cross-Sectional Studies , Aged , Thyroid Gland/diagnostic imaging , Thyroid Gland/metabolism , Radionuclide Imaging/methods , Adult , Thyrotropin/blood , Thyrotropin/metabolism , Ultrasonography/methods , Radiopharmaceuticals , ROC Curve , Thyroid Hormones/metabolism , Autoantibodies/blood
14.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39276028

ABSTRACT

The identification of critical factors that can worsen the mechanisms contributing to the pathophysiology of Alzheimer disease is of paramount importance. Thyroid hormones (TH) fit this criterion. Epidemiological studies have identified an association between altered circulating TH levels and Alzheimer disease. The study of human and animal models indicates that TH can affect all the main cellular, molecular, and genetic mechanisms known as hallmarks of Alzheimer disease. This is true not only for the excessive production in the brain of protein aggregates leading to amyloid plaques and neurofibrillary tangles but also for the clearance of these molecules from the brain parenchyma via the blood-brain barrier and for the escalated process of neuroinflammation-and even for the effects of carrying Alzheimer-associated genetic variants. Suboptimal TH levels result in a greater accumulation of protein aggregates in the brain. The direct TH regulation of critical genes involved in amyloid beta production and clearance is remarkable, affecting the expression of multiple genes, including APP (related to amyloid beta production), APOE, LRP1, TREM2, AQP4, and ABCB1 (related to amyloid beta clearance). TH also affects microglia by increasing their migration and function and directly regulating the immunosuppressor gene CD73, impacting the immune response of these cells. Studies aiming to understand the mechanisms that could explain how changes in TH levels can contribute to the brain alterations seen in patients with Alzheimer disease are ongoing. These studies have potential implications for the management of patients with Alzheimer disease and ultimately can contribute to devising new interventions for these conditions.


Subject(s)
Alzheimer Disease , Thyroid Hormones , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/epidemiology , Alzheimer Disease/etiology , Thyroid Hormones/metabolism , Animals , Brain/metabolism , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism
15.
Expert Opin Ther Pat ; 34(10): 953-962, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39279560

ABSTRACT

INTRODUCTION: Cancer cells adopt a glycolytic phenotype to fulfill their energy needs in unfavorable conditions. In metabolic rewiring, cancer cells upregulate the expression of glycolytic pathway regulators including glucose transporter 1, hexokinase 2, and PKM2 (pyruvate kinase) into its M2 splice form. Among these regulators, PKM2 plays a major role in metabolic reprogramming and is overexpressed in various diseases, including cancer. Dimerization of PKM2 causes the generation of synthetic precursors from glycolytic intermediates, which are essential for cellular growth and cancer cell proliferation. COVERED AREAS: This article is focused on examining recent patents (2018-2023) on PKM2 activators, inhibitors and their biological and synthesis properties by using the advanced search service of the European Patent Office (EPO). Moreover, other databases including PubMed, Google Scholar and Elsevier were also examined for scientific data. On basis of their chemical structures, PKM2 activators and inhibitors are classified into pyrazole, pyrolidine-pyrazole, phenol, benzoxazine, isoselenazolo-pyridinium, phthalazine, and propiolylamide derivatives. EXPERT OPINION: Activating PKM2 reduces proliferation and development of cells by reducing the quantity of biomolecules needed for cell formation. PKM2 activators and inhibitors are highly effective in treating many cancer pathogens. It is important to find new, more potent and selective molecules for PKM2 activation and inhibition.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Neoplasms , Patents as Topic , Pyruvate Kinase , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Pyruvate Kinase/metabolism , Pyruvate Kinase/antagonists & inhibitors , Thyroid Hormone-Binding Proteins , Membrane Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Thyroid Hormones/metabolism , Carrier Proteins/metabolism , Carrier Proteins/antagonists & inhibitors , Drug Development , Glycolysis/drug effects
16.
Ecotoxicol Environ Saf ; 284: 116995, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39236656

ABSTRACT

Environmental pollutants capable of interfering with the thyroid hormone (TH) system increasingly raise concern for both human and environmental health. Recently, resorcinol has received attention as a compound of concern due to its endocrine disrupting properties. It is a known inhibitor of thyroperoxidase (TPO), an enzyme required in TH synthesis, and therapeutic use of resorcinol exposure has led to hypothyroidism in humans. There is limited evidence concerning ecotoxicologically relevant effects of resorcinol in fish. A set of adverse outcome pathways (AOPs) has recently been developed linking thyroid hormone system disruption (THSD) to impaired swim bladder inflation and eye development in fish. In the present study, these AOPs were used to provide the background for testing potential THSD effects of resorcinol in zebrafish eleutheroembryos. We exposed zebrafish eleutheroembryos to resorcinol and assessed TH levels, swim bladder inflation and eye morphology. As a TPO inhibitor, resorcinol is expected to affect TH levels and eye morphology but not swim bladder inflation during embryonic development. Indeed, thyroxine (T4) levels were significantly decreased following resorcinol exposure. In contrast to our hypothesis, swim bladder inflation was impaired at 5 days post fertilization (dpf) and no effects on eye morphology were detected. Therefore, in vitro assays were performed to identify potential additional thyroid hormone system disruption-related mechanisms through which resorcinol may act. Two new mechanisms were identified: TH receptor (TR) antagonism and transthyretin (TTR) binding inhibition. Both of these mechanisms can plausibly be linked to impaired swim bladder inflation and could, therefore, explain the observed effect. Overall, our study contributes to the knowledge of the THSD potential of resorcinol both in vivo in the zebrafish model as well as in vitro.


Subject(s)
Endocrine Disruptors , Resorcinols , Thyroid Hormones , Zebrafish , Animals , Resorcinols/toxicity , Endocrine Disruptors/toxicity , Thyroid Hormones/metabolism , Water Pollutants, Chemical/toxicity , Air Sacs/drug effects , Eye/drug effects , Embryo, Nonmammalian/drug effects , Thyroxine
17.
Nat Immunol ; 25(10): 1884-1899, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39327500

ABSTRACT

TCF1high progenitor CD8+ T cells mediate the efficacy of immunotherapy; however, the mechanisms that govern their generation and maintenance are poorly understood. Here, we show that targeting glycolysis through deletion of pyruvate kinase muscle 2 (PKM2) results in elevated pentose phosphate pathway (PPP) activity, leading to enrichment of a TCF1high progenitor-exhausted-like phenotype and increased responsiveness to PD-1 blockade in vivo. PKM2KO CD8+ T cells showed reduced glycolytic flux, accumulation of glycolytic intermediates and PPP metabolites and increased PPP cycling as determined by 1,2-13C glucose carbon tracing. Small molecule agonism of the PPP without acute glycolytic impairment skewed CD8+ T cells toward a TCF1high population, generated a unique transcriptional landscape and adoptive transfer of agonist-treated CD8+ T cells enhanced tumor control in mice in combination with PD-1 blockade and promoted tumor killing in patient-derived tumor organoids. Our study demonstrates a new metabolic reprogramming that contributes to a progenitor-like T cell state promoting immunotherapy efficacy.


Subject(s)
CD8-Positive T-Lymphocytes , Hepatocyte Nuclear Factor 1-alpha , Immunotherapy , Pentose Phosphate Pathway , Thyroid Hormone-Binding Proteins , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Immunotherapy/methods , Glycolysis , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Knockout , Thyroid Hormones/metabolism , Programmed Cell Death 1 Receptor/metabolism , Immune Checkpoint Inhibitors/pharmacology , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/metabolism , Pyruvate Kinase
18.
Cell Biochem Funct ; 42(7): e4125, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39327771

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the deadly malignant tumors that directly leads to the death of nearly one million people worldwide every year, causing a serious burden on society. In the presence of sufficient oxygen, HCC cells rapidly generate energy through aerobic glycolysis, which promotes tumor cell proliferation, immune evasion, metastasis, angiogenesis, and drug resistance. Pyruvate kinase M2 (PKM2) is a key rate-limiting enzyme in glycolysis. In recent years, studies have found that PKM2 not only exerts pyruvate kinase activity in the process of glucose metabolism, but also exerts protein kinase activity in non-metabolic pathways to affect tumor cell processes, and its activity is flexibly regulated by various posttranslational modifications such as acetylation, phosphorylation, lactylation, ubiquitination, SUMOylation, and so forth. This review summarizes the role of posttranslational modifications of PKM2-related sites in the development of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Protein Processing, Post-Translational , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins , Pyruvate Kinase/metabolism , Membrane Proteins/metabolism , Animals , Carrier Proteins/metabolism
19.
Eur Thyroid J ; 13(5)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39312733

ABSTRACT

Objective: Metabolic dysfunction-associated steatohepatitis (MASH) is characterized by inflammation, fibrosis, and accumulation of fatty acids in the liver. MASH disease progression has been associated with reduced thyroid hormone (TH) signaling in the liver, including reduced expression of deiodinase type I (DIO1) and TH receptor beta (THRB). However, the underlying mechanisms mediating these effects remain elusive. Here, we hypothesized that epigenetic mechanisms may be involved in modulating hepatic TH action. Methods: Liver samples from patients with and without MASH were analyzed by qRT-PCR and correlated with clinical parameters. Luciferase reporter assays and overexpression of miRNA in HepG2 cells were used to validate the functional binding of miRNA to predicted targets. DNA methylation was analyzed by bisulfite pyrosequencing. Results: miR-34a-5p was upregulated in MASH patients and correlated positively with the clinical parameters of MASH. Using in silico and in vitro analysis, we demonstrate that miR-34a-5p is capable of targeting several modulators of local hepatic TH action, as evidenced by the functional binding of miR-34a-5p to the seed sequence in the THRB and DIO1 genes. Consequently, overexpression of miR-34a-5p in HepG2 cells reduced the expression of THRA, THRB, DIO1, and SLC10A1, thus potentially mediating an acquired hepatic resistance to TH in MASH. As an additional regulatory mechanism, DNA methylation of THRB intron 1 was increased in MASH and negatively correlated with THRB expression. Conclusion: miR-34a-5p constitutes a possible epigenetic master regulator of hepatic TH action, which together with THRB-specific DNA methylation could explain a possible developing TH resistance in the liver during MASH progression on the molecular level.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Iodide Peroxidase , MicroRNAs , Thyroid Hormone Receptors beta , Thyroid Hormones , Humans , Hep G2 Cells , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Female , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Middle Aged , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Liver/metabolism , Liver/pathology , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Adult
20.
Int Immunopharmacol ; 142(Pt B): 113247, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39321706

ABSTRACT

Pterostilbene (PTS) is a dietary phytochemical that has shown antitumor activity in many types of cancer, but the molecular mechanism remains unclear. It has also not been adequately studied on PTS against esophageal squamous cell carcinoma (ESCC). Thus, this study investigated the effect of PTS on ESCC in vitro and in vivo and explored the underlying molecular mechanism. We found that PTS can inhibit the proliferation, colony formation, and migration of ESCC cells. According to the bioinformatics analysis of proteomics, PTS had a great influence on the metabolic process of ESCC cells. KEGG analysis showed that PTS down-regulated the pyruvate metabolism pathway. Moreover, PTS can inhibit the PK activity, glucose consumption, and lactate production in ESCC cells. By administration of PTS into xenograft mice, experiment results demonstrated that PTS can suppress tumor progress and the PKM2/STAT3/c-MYC signaling pathway. We found that PTS inhibited the PKM2/STAT3/c-MYC signaling pathway by targeting PKM2 in ESCC cells. Collectively, this study revealed that PTS inhibited ESCC growth by suppressing PKM2 mediated aerobic glycolysis and PKM2/STAT3/c-MYC signaling pathway, which enriching the anti-tumor molecular mechanism of PTS and providing a theoretical basis for its clinical application.


Subject(s)
Carrier Proteins , Cell Proliferation , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Glycolysis , Membrane Proteins , Mice, Nude , Proto-Oncogene Proteins c-myc , STAT3 Transcription Factor , Signal Transduction , Stilbenes , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Xenograft Model Antitumor Assays , Animals , Humans , STAT3 Transcription Factor/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Cell Line, Tumor , Signal Transduction/drug effects , Thyroid Hormones/metabolism , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Glycolysis/drug effects , Stilbenes/pharmacology , Stilbenes/therapeutic use , Membrane Proteins/metabolism , Cell Proliferation/drug effects , Carrier Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL