Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 439
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1325286, 2024.
Article in English | MEDLINE | ID: mdl-39381436

ABSTRACT

Introduction: Reducing Optic Atrophy 1 (OPA1) expression in skeletal muscle in male mice induces Activation Transcription Factor 4 (ATF4) and the integrated stress response (ISR). Additionally, skeletal muscle secretion of Fibroblast Growth Factor 21 (FGF21) is increased, which mediates metabolic adaptations including resistance to diet-induced obesity (DIO) and glucose intolerance in these mice. Although FGF21 induction in this model can be reversed with pharmacological attenuation of ER stress, it remains to be determined if ATF4 is responsible for FGF21 induction and its metabolic benefits in this model. Methods: We generated mice with homozygous floxed Opa1 and Atf4 alleles and a tamoxifen-inducible Cre transgene controlled by the human skeletal actin promoter to enable simultaneous depletion of OPA1 and ATF4 in skeletal muscle (mAO DKO). Mice were fed high fat (HFD) or control diet and evaluated for ISR activation, body mass, fat mass, glucose tolerance, insulin tolerance and circulating concentrations of FGF21 and growth differentiation factor 15 (GDF15). Results: In mAO DKO mice, ATF4 induction is absent. Other indices of ISR activation, including XBP1s, ATF6, and CHOP were induced in mAO DKO males, but not in mOPA1 or mAO DKO females. Resistance to diet-induced obesity was not reversed in mAO DKO mice of both sexes. Circulating FGF21 and GDF15 illustrated sexually dimorphic patterns. Loss of OPA1 in skeletal muscle increases circulating FGF21 in mOPA1 males, but not in mOPA1 females. Additional loss of ATF4 decreased circulating FGF21 in mAO DKO male mice, but increased circulating FGF21 in female mAO DKO mice. Conversely, circulating GDF15 was increased in mAO DKO males and mOPA1 females, but not in mAO DKO females. Conclusion: Sex differences exist in the transcriptional outputs of the ISR following OPA deletion in skeletal muscle. Deletion of ATF4 in male and female OPA1 KO mice does not reverse the resistance to DIO. Induction of circulating FGF21 is ATF4 dependent in males, whereas induction of circulating GDF15 is ATF4 dependent in females. Elevated GDF15 in males and FGF21 in females could reflect activation by other transcriptional outputs of the ISR, that maintain mitokine-dependent metabolic protection in an ATF4-independent manner.


Subject(s)
Activating Transcription Factor 4 , Fibroblast Growth Factors , GTP Phosphohydrolases , Mice, Knockout , Muscle, Skeletal , Sex Characteristics , Animals , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Mice , Male , Muscle, Skeletal/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Female , GTP Phosphohydrolases/metabolism , GTP Phosphohydrolases/genetics , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Obesity/metabolism , Obesity/genetics , Diet, High-Fat , Mice, Inbred C57BL
2.
Cell Biol Toxicol ; 40(1): 79, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289208

ABSTRACT

Noise-induced hidden hearing loss (HHL) is a newly uncovered form of hearing impairment that causes hidden damage to the cochlea. Patients with HHL do not have significant abnormalities in their hearing thresholds, but they experience impaired speech recognition in noisy environments. However, the mechanisms underlying HHL remain unclear. In this study, we developed single-cell transcriptome profiles of the cochlea of mice with HHL, detailing changes in individual cell types. Our study revealed a transient threshold shift, reduced auditory brainstem response wave I amplitude, and decreased number of ribbon synapses in HHL mice. Our findings suggest elevated oxidative stress and GDF15 expression in cochlear hair cells of HHL mice. Notably, the upregulation of GDF15 attenuated oxidative stress and auditory impairment in the cochlea of HHL mice. This suggests that a therapeutic strategy targeting GDF15 may be efficacious against HHL.


Subject(s)
Growth Differentiation Factor 15 , Hearing Loss, Noise-Induced , Oxidative Stress , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Hearing Loss, Noise-Induced/metabolism , Mice , Cochlea/metabolism , Cochlea/pathology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Male , Mice, Inbred C57BL , Evoked Potentials, Auditory, Brain Stem , Noise/adverse effects , Transcriptome/genetics , Disease Models, Animal , Hearing Loss, Hidden
3.
J Toxicol Sci ; 49(9): 399-408, 2024.
Article in English | MEDLINE | ID: mdl-39231684

ABSTRACT

Nasopharyngeal carcinoma (NPC) originates from the nasopharynx epithelium, and luteolin is recognized as an important anti-cancer agent. This study investigated the effects of luteolin on ferroptosis in NPC cells. NPC cells were cultured and exposed to varying concentrations of luteolin. Cell viability, malondialdehyde (MDA) levels, superoxide dismutase (SOD) activity, glutathione (GSH) levels, Fe2+ concentration, and glutathione peroxidase 4 (GPX4) protein level were assessed. Additionally, SRY-related high-mobility-group box 4 (SOX4) expression was measured. Subsequently, the binding of SOX4 to the growth differentiation factor-15 (GDF15) promoter and GDF15 mRNA levels were evaluated. The impact of the SOX4/GDF15 axis on luteolin-induced ferroptosis in NPC cells was assayed. Luteolin treatment induced cell ferroptosis, evidenced by decreased cell viability, increased MDA and Fe2+ levels, and reduced SOD, GSH, and GPX4 levels. Furthermore, luteolin downregulated SOX4 expression, while overexpression of SOX4 reversed luteolin's pro-ferroptotic effects in NPC cells. SOX4 was found to up-regulate GDF15 transcription by directly binding to its promoter. Conversely, overexpression of GDF15 mitigated the ferroptotic effects induced by luteolin in NPC cells. Therefore, luteolin induces ferroptosis in NPC cells via modulation of the SOX4/GDF15 axis. In conclusion, luteolin reduces the binding of SOX4 to the GDF15 promoter by suppressing SOX4 expression, thereby down-regulating GDF15 transcription levels and inducing ferroptosis in NPC cells.


Subject(s)
Cell Survival , Ferroptosis , Growth Differentiation Factor 15 , Luteolin , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Ferroptosis/drug effects , Ferroptosis/genetics , Luteolin/pharmacology , Humans , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Cell Survival/drug effects , Cell Line, Tumor , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Malondialdehyde/metabolism , Antineoplastic Agents/pharmacology , Superoxide Dismutase/metabolism , Superoxide Dismutase/genetics , Promoter Regions, Genetic/genetics
4.
Proc Natl Acad Sci U S A ; 121(39): e2316161121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39298490

ABSTRACT

Uveitis is a vision-threatening disease primarily driven by a dysregulated immune response, with retinal microglia playing a pivotal role in its progression. Although the transcription factor EGR2 is known to be closely associated with uveitis, including Vogt-Koyanagi-Harada disease and Behcet's disease, and is essential for maintaining the dynamic homeostasis of autoimmunity, its exact role in uveitis remains unclear. In this study, diminished EGR2 expression was observed in both retinal microglia from experimental autoimmune uveitis (EAU) mice and inflammation-induced human microglia cell line (HMC3). We constructed a mice model with conditional knockout of EGR2 in microglia and found that EGR2 deficiency resulted in increased intraocular inflammation. Meanwhile, EGR2 overexpression downregulated the expression of inflammatory cytokines as well as cell migration and proliferation in HMC3 cells. Next, RNA sequencing and ChIP-PCR results indicated that EGR2 directly bound to its downstream target growth differentiation factor 15 (GDF15) and further regulated GDF15 transcription. Furthermore, intravitreal injection of GDF15 recombinant protein was shown to ameliorate EAU progression in vivo. Meanwhile, knockdown of GDF15 reversed the phenotype of EGR2 overexpression-induced microglial inflammation in vitro. In summary, this study highlighted the protective role of the transcription factor EGR2 in AU by modulating the microglial phenotype. GFD15 was identified as a downstream target of EGR2, providing a unique target for uveitis treatment.


Subject(s)
Autoimmune Diseases , Early Growth Response Protein 2 , Growth Differentiation Factor 15 , Microglia , Uveitis , Animals , Humans , Mice , Autoimmune Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Cell Line , Disease Models, Animal , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/genetics , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Phenotype , Retina/metabolism , Retina/pathology , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology , Uveitis/genetics
5.
Clin Epigenetics ; 16(1): 124, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256775

ABSTRACT

BACKGROUND: Plasma growth differentiation factor 15 (GDF15) and N-terminal proB-type natriuretic peptide (NT-proBNP) are cardiovascular biomarkers that associate with a range of diseases. Epigenetic scores (EpiScores) for GDF15 and NT-proBNP may provide new routes for risk stratification. RESULTS: In the Generation Scotland cohort (N ≥ 16,963), GDF15 levels were associated with incident dementia, ischaemic stroke and type 2 diabetes, whereas NT-proBNP levels were associated with incident ischaemic heart disease, ischaemic stroke and type 2 diabetes (all PFDR < 0.05). Bayesian epigenome-wide association studies (EWAS) identified 12 and 4 DNA methylation (DNAm) CpG sites associated (Posterior Inclusion Probability [PIP] > 95%) with levels of GDF15 and NT-proBNP, respectively. EpiScores for GDF15 and NT-proBNP were trained in a subset of the population. The GDF15 EpiScore replicated protein associations with incident dementia, type 2 diabetes and ischaemic stroke in the Generation Scotland test set (hazard ratios (HR) range 1.36-1.41, PFDR < 0.05). The EpiScore for NT-proBNP replicated the protein association with type 2 diabetes, but failed to replicate an association with ischaemic stroke. EpiScores explained comparable variance in protein levels across both the Generation Scotland test set and the external LBC1936 test cohort (R2 range of 5.7-12.2%). In LBC1936, both EpiScores were associated with indicators of poorer brain health. Neither EpiScore was associated with incident dementia in the LBC1936 population. CONCLUSIONS: EpiScores for serum levels of GDF15 and Nt-proBNP associate with body and brain health traits. These EpiScores are provided as potential tools for disease risk stratification.


Subject(s)
Biomarkers , DNA Methylation , Diabetes Mellitus, Type 2 , Growth Differentiation Factor 15 , Natriuretic Peptide, Brain , Peptide Fragments , Humans , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/genetics , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Peptide Fragments/blood , Peptide Fragments/genetics , Male , Female , Aged , Middle Aged , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , DNA Methylation/genetics , Biomarkers/blood , Scotland , Dementia/blood , Dementia/genetics , Epigenesis, Genetic , Ischemic Stroke/blood , Ischemic Stroke/genetics , Bayes Theorem , Cohort Studies
6.
Front Biosci (Landmark Ed) ; 29(8): 298, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39206905

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disease that remains a serious global health issue. Ferroptosis has been recognized as a vital driver of pathological progression of AD. However, the detailed regulatory mechanisms of ferroptosis during AD progression remain unclear. This study aimed to explore the regulatory role and mechanism of methyltransferase like 14 (METTL14) in ferroptosis in AD models. METHODS: Serum samples were collected from 18 AD patients and 18 healthy volunteers to evaluate clinical correlation. Scopolamine-treated mice and Aß1-42-stimulated SH-SY5Y cells were served as the in vivo and in vitro models of AD. Ferroptosis was detected by reactive oxygen species (ROS), Fe2+, total iron levels, and ferroptosis-related proteins glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11). Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide (MTT) assay. The N6-methyladenosine (m6A) modification was detected by RNA methylation quantification kit and methylated RNA immunoprecipitation sequencing-quantitative real-time polymerase chain reaction (MeRIP-qPCR). Molecular mechanisms were investigated by RNA pull-down, RNA immunoprecipitation (RIP), and co-immunoprecipitation (Co-IP) assays. Cognitive disorder of AD mice was measured by Morris water maze test. RESULTS: METTL14 was down-regulated, while lncRNA taurine upregulated gene 1 (TUG1) was up-regulated in clinical patients and experimental models of AD. Functional experiments demonstrated that METTL14 overexpression or TUG1 silencing effectively attenuated Aß1-42-induced ferroptosis and neurotoxicity in SH-SY5Y cells. Mechanistically, METTL14-mediated m6A modification reduced the stability of TUG1. Moreover, TUG1 promoted the ubiquitination and degradation of growth differentiation factor 15 (GDF15) by directly interacted with Smad ubiquitin regulatory factor 1 (SMURF1), which consequently inactivated nuclear factor erythroid 2-related factor 2 (NRF2). Rescue experiments indicated that GDF15 depletion reversed sh-TUG1-mediated protection against ferroptosis and neurotoxicity. Finally, Mettl14 overexpression repressed ferroptosis to ameliorate the cognitive disorder via modulating Tug1/Gdf15/Nrf2 pathway in vivo. CONCLUSION: METTL14 inhibited ferroptosis to ameliorate AD pathological development by m6A modification of TUG1 to activate GDF15/NRF2 axis, providing a novel therapeutic target for AD.


Subject(s)
Alzheimer Disease , Ferroptosis , Growth Differentiation Factor 15 , Methyltransferases , RNA, Long Noncoding , Ubiquitination , Ferroptosis/drug effects , Ferroptosis/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Humans , Animals , Methyltransferases/metabolism , Methyltransferases/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Male , Mice , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Female , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Line, Tumor , Aged , Disease Models, Animal , Amyloid beta-Peptides/metabolism , Mice, Inbred C57BL
7.
Am J Physiol Endocrinol Metab ; 327(4): E552-E562, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39196800

ABSTRACT

Type 1 diabetes (T1D) is a chronic metabolic disease resulting from an autoimmune destruction of pancreatic beta cells. Beta cells activate various stress responses during the development of T1D, including senescence, which involves cell cycle arrest, prosurvival signaling, and a proinflammatory secretome termed the senescence-associated secretory phenotype (SASP). We previously identified growth and differentiation factor 15 (GDF15) as a major SASP factor in human islets and human EndoC-ßH5 beta cells in a model of DNA damage-mediated senescence that recapitulates features of senescent beta cells in T1D. Soluble GDF15 has been shown to exert protective effects on human and mouse beta cells during various forms of stress relevant to T1D; therefore, we hypothesized that secreted GDF15 may play a prosurvival role during DNA damage-mediated senescence in human beta cells. We found that elevated GDF15 secretion was associated with endogenous senescent beta cells in an islet preparation from a T1D donor, supporting the validity of our DNA damage model. Using antibody-based neutralization, we found that secreted endogenous GDF15 was not required for senescent human islet or EndoC cell viability. Rather, neutralization of GDF15 led to reduced expression of specific senescence-associated genes, including GDF15 itself and the prosurvival gene BCL2-like protein 1 (BCL2L1). Taken together, these data suggest that SASP factor GDF15 is not required to sustain senescent human islet viability, but it is required to maintain senescence-associated transcriptional responses.NEW & NOTEWORTHY Beta cell senescence is an emerging contributor to the pathogenesis of type 1 diabetes, but candidate therapeutic targets have not been identified in human beta cells. In this study, we examined the role of a secreted factor, GDF15, and found that although it is not required to maintain viability during senescence, it is required to fine-tune gene expression programs involved in the senescence response during DNA damage in human beta cells.


Subject(s)
Cellular Senescence , DNA Damage , Diabetes Mellitus, Type 1 , Growth Differentiation Factor 15 , Insulin-Secreting Cells , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Humans , Insulin-Secreting Cells/metabolism , Cellular Senescence/genetics , Cellular Senescence/physiology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/genetics , Senescence-Associated Secretory Phenotype , Cells, Cultured , Cell Survival , Transcription, Genetic
8.
Cell Rep ; 43(8): 114577, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39096490

ABSTRACT

Growth and differentiation factor 15 (GDF15) has recently emerged as a weight loss and insulin-sensitizing factor. Growing evidence also supports a role for GDF15 as a physiological, exercise-induced stress signal. Here, we tested whether GDF15 is required for the insulin-sensitizing effects of exercise in mice and humans. At baseline, both under a standard nutritional state and high-fat feeding, GDF15 knockout (KO) mice display normal glucose tolerance, systemic insulin sensitivity, maximal speed, and endurance running capacity when compared to wild-type littermates independent of sex. When submitted to a 4-week exercise training program, both lean and obese wild-type and GDF15 KO mice similarly improve their endurance running capacity, glucose tolerance, systemic insulin sensitivity, and peripheral glucose uptake. Insulin-sensitizing effects of exercise training were also unrelated to changes in plasma GDF15 in humans. In summary, we here show that GDF15 is dispensable for the insulin-sensitizing effects of chronic exercise.


Subject(s)
Growth Differentiation Factor 15 , Insulin Resistance , Insulin , Mice, Knockout , Physical Conditioning, Animal , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Humans , Male , Insulin/metabolism , Insulin/blood , Female , Mice , Mice, Inbred C57BL , Adult
9.
Nat Commun ; 15(1): 7173, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169003

ABSTRACT

Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.


Subject(s)
Adipose Tissue , Diabetes Mellitus, Type 2 , Fatty Liver , Growth Differentiation Factor 15 , Hepatocytes , Macrophages , Obesity , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Obesity/metabolism , Obesity/pathology , Hepatocytes/metabolism , Male , Macrophages/metabolism , Mice , Humans , Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Mice, Inbred C57BL , Liver/metabolism , Liver/pathology , Disease Models, Animal , Signal Transduction
10.
Eur J Pharmacol ; 982: 176894, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39147013

ABSTRACT

Sepsis is a systemic inflammatory response syndrome triggered by infection, presenting with symptoms such as fever, increased heart rate, and low blood pressure. In severe cases, it can lead to multiple organ dysfunction, posing a life-threatening risk. Sepsis-induced cardiomyopathy (SIC) is a critical factor in the poor prognosis of septic patients, leading to myocardial dysfunction characterized by cell death, inflammation, and diminished cardiac function. Ferroptosis, an iron-dependent form of programmed cell death, is a key mechanism causing cardiomyocyte damage in SIC. Growth differentiation factor 15 (GDF15), a member of the TGF-ß superfamily, is associated with various cardiovascular diseases and can inhibit oxidative stress, reduce reactive oxygen species (ROS), and suppress ferroptosis. Elevated serum GDF15 levels in sepsis are correlated with organ injuries, suggesting its potential as a therapeutic target. However, its role and mechanisms in SIC remain unclear. Glutathione peroxidase 4 (GPX4), the only enzyme capable of reducing lipid peroxides within cells, protects cells by reducing lipid peroxidation levels and inhibiting ferroptosis. Investigating the regulatory factors of GPX4 may provide a theoretical basis for SIC treatment. In this study, a mouse SIC model revealed that elevated GDF15 exerts a protective effect. Antagonizing GDF15 exacerbates myocardial damage. Through transcriptomic analysis and other methods, we confirmed that GDF15 inhibits the expression of SOCS1 by activating the ALK5-SMAD2/3 pathway, thereby activates the JAK2/STAT3 pathway, promotes the transcription of GPX4, inhibits ferroptosis in cardiomyocytes, and plays a myocardial protective role in SIC.


Subject(s)
Ferroptosis , Growth Differentiation Factor 15 , Myocytes, Cardiac , Phospholipid Hydroperoxide Glutathione Peroxidase , Sepsis , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Animals , Male , Mice , Cardiomyopathies/metabolism , Cardiomyopathies/etiology , Disease Models, Animal , Ferroptosis/drug effects , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Sepsis/complications , Sepsis/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Suppressor of Cytokine Signaling 1 Protein/genetics
11.
EMBO Mol Med ; 16(9): 2080-2108, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103698

ABSTRACT

Chemotherapy, the standard of care treatment for cancer patients with advanced disease, has been increasingly recognized to activate host immune responses to produce durable outcomes. Here, in colorectal adenocarcinoma (CRC) we identify oxaliplatin-induced Thioredoxin-Interacting Protein (TXNIP), a MondoA-dependent tumor suppressor gene, as a negative regulator of Growth/Differentiation Factor 15 (GDF15). GDF15 is a negative prognostic factor in CRC and promotes the differentiation of regulatory T cells (Tregs), which inhibit CD8 T-cell activation. Intriguingly, multiple models including patient-derived tumor organoids demonstrate that the loss of TXNIP and GDF15 responsiveness to oxaliplatin is associated with advanced disease or chemotherapeutic resistance, with transcriptomic or proteomic GDF15/TXNIP ratios showing potential as a prognostic biomarker. These findings illustrate a potentially common pathway where chemotherapy-induced epithelial oxidative stress drives local immune remodeling for patient benefit, with disruption of this pathway seen in refractory or advanced cases.


Subject(s)
Adenocarcinoma , Carrier Proteins , Colorectal Neoplasms , Growth Differentiation Factor 15 , Oxaliplatin , Humans , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Carrier Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
12.
Mol Metab ; 88: 102002, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111389

ABSTRACT

OBJECTIVE: Growth differentiation factor 15 (GDF15), a stress related cytokine, was recently identified as a novel satiety signal acting via the GFRAL receptor located in the hindbrain. Bitter compounds are known to induce satiety via the release of glucagon-like peptide 1 (GLP-1) through activation of bitter taste receptors (TAS2Rs, 25 subtypes) on enteroendocrine cells in the gut. This study aimed to investigate whether and how bitter compounds induce a stress response in intestinal epithelial cells to affect GDF15 expression in patients with obesity, thereby facilitating satiety signaling from the gut. METHODS: The acute effect of oral intake of the bitter-containing medication Plaquenil (hydroxychloroquine sulfate) on plasma GDF15 levels was evaluated in a placebo-controlled, double-blind, randomized, two-visit crossover study in healthy volunteers. Primary crypts isolated from the jejunal mucosa from patients with obesity were stimulated with vehicle or bitter compounds, and the effect on GDF15 expression was evaluated using RT-qPCR or ELISA. Immunofluorescence colocalization studies were performed between GDF15, epithelial cell type markers and TAS2Rs. The role of TAS2Rs was tested by 1) pretreatment with a TAS2R antagonist, GIV3727; 2) determining TAS2R4/43 polymorphisms that affect taste sensitivity to TAS2R4/43 agonists. RESULTS: Acute intake of hydroxychloroquine sulfate increased GDF15 plasma levels, which correlated with reduced hunger scores and plasma ghrelin levels in healthy volunteers. This effect was mimicked in primary jejunal cultures from patients with obesity. GDF15 was expressed in enteroendocrine and goblet cells with higher expression levels in patients with obesity. Various bitter-tasting compounds (medicinal, plant extracts, bacterial) either increased or decreased GDF15 expression, with some also affecting GLP-1. The effect was mediated by specific intestinal TAS2R subtypes and the unfolded protein response pathway. The bitter-induced effect on GDF15/GLP-1 expression was influenced by the existence of TAS2R4 amino acid polymorphisms and TAS2R43 deletion polymorphisms that may predict patient's therapeutic responsiveness. However, the effect of the bitter-tasting antibiotic azithromycin on GDF15 release was mediated via the motilin receptor, possibly explaining some of its aversive side effects. CONCLUSIONS: Bitter chemosensory and pharmacological receptors regulate the release of GDF15 from human gut epithelial cells and represent potential targets for modulating metabolic disorders or cachexia.


Subject(s)
Glucagon-Like Peptide 1 , Growth Differentiation Factor 15 , Obesity , Receptors, Gastrointestinal Hormone , Humans , Obesity/metabolism , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Male , Adult , Female , Double-Blind Method , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/blood , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics , Taste , Middle Aged , Intestinal Mucosa/metabolism , Cross-Over Studies , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Young Adult
13.
PLoS One ; 19(7): e0307105, 2024.
Article in English | MEDLINE | ID: mdl-39008451

ABSTRACT

BACKGROUND AND PURPOSE: Growth differentiation factor 15 (GDF15) has emerged as a promising biomarker in cerebro-cardiovascular disease, particularly in acute and chronic inflammatory stress situations. However, understanding the origins, targets and functions of GDF15 in clinical situations, such as ischemic stroke, remains a complex challenge. This study aims to assess the sources of GDF15 production following an experimental ischemic stroke. METHODS: Adult male Wistar rats underwent cerebral embolization through microspheres injection into the left or right internal carotid artery. Two hours post-surgery, GDF15 expression was analyzed in the brain, blood, lungs, liver and heart using quantitative RT-PCR and Western blotting. RESULTS: Stroke model induced large cerebral infarcts accompanied by severe neurological deficits. GDF15 gene expression exhibited a substantial increase in the ipsilateral cortex and cerebellum, with a lesser extent in the contralateral cortex. Regarding GDF15 protein expression, proGDF15 levels were elevated in the 3 aforementioned organs mentioned and the heart. However, the mature form of GDF15 was exclusively present and increased in the heart. Finally, the expression of GDF15 expression was correlated with the neurological deficit score. CONCLUSIONS: Our findings suggest that both the GDF15 gene and pro-protein are expressed in the ischemic brain after a stroke, while only its mature form is expressed remotely in in the heart. The impact of increased GDF15 in the heart following a stroke remains to be established. This is particularly relevant in understanding its relationships with poor neurological outcomes, determining whether it may contribute to stroke-induced cardiac dysfunction.


Subject(s)
Growth Differentiation Factor 15 , Rats, Wistar , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Animals , Male , Rats , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/complications , Disease Models, Animal , Brain/metabolism , Brain/pathology , Myocardium/metabolism , Myocardium/pathology , Stroke/metabolism , Stroke/genetics
14.
Anal Cell Pathol (Amst) ; 2024: 2165242, 2024.
Article in English | MEDLINE | ID: mdl-39045215

ABSTRACT

This study aimed to construct an eukaryotic expression vector, pEGFP-N1-MIC-1, for overexpressing the mouse macrophage inhibitory cytokine-1 (MIC-1) gene. Additionally, we transfected the MFC cell line to observe the upregulation of MIC-1 gene expression and assess its impact on macrophage phenotype conversion. Enzyme digestion and DNA sequencing confirmed the successful construction of the pEGFP-N1-MIC-1 vector. The transfected MFC cells exhibited a significant increase in MIC-1 protein expression levels. Furthermore, transfection with pEGFP-N1-MIC-1 increased the migration and colony formation capabilities of MFC cells. These results may contribute to future research and the development of therapeutic interventions targeting MIC-1 in macrophages, particularly in the context of gastric cancer.


Subject(s)
Genetic Vectors , Growth Differentiation Factor 15 , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Animals , Mice , Cell Line, Tumor , Genetic Vectors/genetics , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Cell Movement/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Transfection , Macrophages/metabolism , Humans
15.
Hum Cell ; 37(5): 1405-1420, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39007956

ABSTRACT

Abnormal functions of trophoblast cells are associated with the pathogenesis of preeclampsia (PE). Nuclear receptor subfamily 2 group F member 1 (NR2F1) acts as a transcriptionally regulator in many diseases, but its role in PE remains unknown. Hypoxia/reoxygenation (H/R)-stimulated HTR-8/SVneo cells were used to mimic PE injury in vitro. NR2F1 overexpression alleviated trophoblast apoptosis, as evidenced by the decreased number of TUNEL-positive cells and the downregulation of caspase 3 and caspase 9 expression in cells. NR2F1 overexpression increased the invasion and migration ability of HTR-8/SVneo cells, accompanied by increased protein levels of matrix metalloproteinase (MMP)-2 and MMP-9. mRNA-seq was applied to explore the underlying mechanism of NR2F1, identifying growth differentiation factor 15 (GDF15) as the possible downstream effector. Dual-luciferase reporter, ChIP-qPCR, and DNA pull-down assays confirmed that NR2F1 bound to the promoter of GDF15 and transcriptionally inhibited its expression. GDF15 overexpression increased apoptosis and decreased the ability of invasion and migration in HTR-8/SVneo cells expressing NR2F1. MAPK pathway was involved in the regulation of PE. Administration of p38 inhibitor, ERK inhibitor, and JNK inhibitor reversed the effect of simultaneous overexpression NR2F1 and GDF15 on trophoblast apoptosis, invasion, and migration. Our findings demonstrated that NR2F1 overexpression inhibited trophoblast apoptosis and promoted trophoblast invasion and migration. NR2F1 might negatively regulate GDF15 expression by binding to its promoter region, which further inhibited MAPK signaling pathway in PE. Our study highlights that NR2F1 might sever as a potential target in PE.


Subject(s)
Apoptosis , COUP Transcription Factor I , Growth Differentiation Factor 15 , MAP Kinase Signaling System , Pre-Eclampsia , Trophoblasts , Humans , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , Pregnancy , Female , Apoptosis/genetics , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , COUP Transcription Factor I/genetics , COUP Transcription Factor I/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Gene Expression/genetics , Cell Movement/genetics , Cells, Cultured
16.
Transl Vis Sci Technol ; 13(7): 2, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949633

ABSTRACT

Purpose: We sought to evaluate the efficacy of growth differentiation factor (GDF)-15 treatment for suppressing epithelial-mesenchymal transition (EMT) and alleviating transforming growth factor ß2 (TGFß2)-induced lens opacity. Methods: To test whether GDF-15 is a molecule that prevents EMT, we pretreated the culture with GDF-15 in neural progenitor cells, retinal pigment epithelial cells, and lens epithelial cells and then treated with factors that promote EMT, GDF-11, and TGFß2, respectively. To further investigate the efficacy of GDF-15 on alleviating lens opacity, we used mouse lens explant culture to mimic secondary cataracts. We pretreated the lens culture with GDF-15 and then added TGFß2 to develop lens opacity (n = 3 for each group). Western blot and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to measure EMT protein and gene expression, respectively. Results: In cell culture, GDF-15 pretreatment significantly attenuated EMT marker expression in cultured cells induced by treatment with GDF-11 or TGFß2. In the lens explant culture, GDF-15 pretreatment also reduced mouse lens opacity induced by exposure to TGFß2. Conclusions: Our results indicate that GDF-15 could alleviate TGFß2-induced EMT and is a potential therapeutic agent to slow or prevent posterior capsular opacification (PCO) progression after cataract surgery. Translational Relevance: Cataracts are the leading cause of blindness worldwide, with the only current treatment involving surgical removal of the lens and replacement with an artificial lens. However, PCO, also known as secondary cataract, is a common complication after cataract surgery. The development of an adjuvant that slows the progression of PCO will be beneficial to the field of anterior complications.


Subject(s)
Cataract , Epithelial-Mesenchymal Transition , Growth Differentiation Factor 15 , Lens, Crystalline , Transforming Growth Factor beta2 , Animals , Epithelial-Mesenchymal Transition/drug effects , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Cataract/pathology , Cataract/metabolism , Cataract/prevention & control , Mice , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/drug effects , Mice, Inbred C57BL , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Blotting, Western , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/metabolism
17.
Hepatol Int ; 18(5): 1448-1458, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38900410

ABSTRACT

BACKGROUND AND AIMS: Healthy weight (lean) patients with metabolic dysfunction-associated fatty liver disease (MAFLD) have a more favorable metabolic and histological profile in cross-sectional studies compared with their non-lean counterparts. Paradoxically, they also have higher overall mortality. The underpinning pathophysiology of this paradox is not understood. Telomere attrition is associated with increased mortality in various diseases. METHODS: We investigated the role of telomere length in the pathogenesis of lean MAFLD in cohorts with biopsy-proven MAFLD (n = 303). We measured serum malondialdehyde (MDA) levels and hepatic 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxy-2-nonenal (4-HNE) expression (reactive oxygen species (ROS) markers), growth/differentiation factor-15 (GDF-15) and tested the effect of H2O2 on telomere length and activity in hepatocyte cell lines. The association between leukocyte telomere length and mortality was examined. RESULTS: Telomere length was significantly lower in patients with lean MAFLD (p < 0.001). They also demonstrated an increase in ROS levels and decreases in GDF-15. H2O2 induced telomere shortening and reducing telomere activity in hepatocyte cell lines. We subsequently confirmed that telomere length shortening at baseline is associated with increased hazards of all-cause mortality; the deleterious effect was more profound in lean people. CONCLUSION: Differences in telomere length in part explain the increased mortality of lean compared to non-lean patients with MAFLD. The effect is in part mediated through ROS activation and provide opportunities for therapy.


Subject(s)
Telomere Shortening , Humans , Male , Female , Middle Aged , Telomere/metabolism , Reactive Oxygen Species/metabolism , Adult , Oxidative Stress , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Fatty Liver/metabolism , Fatty Liver/genetics , Fatty Liver/mortality , Malondialdehyde/blood , Malondialdehyde/metabolism , Cross-Sectional Studies
18.
Breast Cancer ; 31(5): 926-934, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38896170

ABSTRACT

BACKGROUND: A history of severe nausea and vomiting during pregnancy (SNVP) is a risk factor for postoperative nausea and vomiting (PONV). This study aimed to explore potentially effective treatment strategies and potential genetic factors underlying SNVP risk-related PONV. METHODS: A total of 140 female patients undergoing breast cancer surgery were assigned to either the study group (70 with SNVP) or the control group (70 with mild to moderate nausea and vomiting during pregnancy (MNVP)). Patients in each group were randomly assigned to two different treatment subgroups and received either ondansetron plus dexamethasone (OD) or OD + TEAS (ODT) (transcutaneous electrical acupoint stimulation, TEAS). Blood samples were collected from patients before induction (D0) and 24 h (D1) after surgery for growth differentiation factor 15 (GDF-15) evaluation. The primary outcome was the incidence of PONV within 36 h. The secondary outcome was the serum GDF-15 level. RESULTS: The incidence of PONV in the SNVP group was significantly higher than that in the MNVP group within 24 h (P < 0.005). In the SNVP group, ODT-treated patients had less PONV than those in the OD-treated group during the 6-12 h (P = 0.033) and 12-24 h (P = 0.008) intervals, while within 6 h, there were fewer vomiting cases in the ODT-treated group (SNVP-ODT vs. SNVP-OD, 7/33 vs. 19/35, P = 0.005). The preoperative GDF-15 serum levels in patients with SNVP were significantly higher (P = 0.004). Moreover, higher preoperative GDF-15 serum levels correlated with a higher incidence of PONV (P = 0.043). CONCLUSIONS: TEAS showed significant effect on PONV treatment in patients with SNVP. A higher serum GDF-15 level was associated with a history of SNVP, as well as a higher risk of PONV.


Subject(s)
Antiemetics , Breast Neoplasms , Dexamethasone , Growth Differentiation Factor 15 , Ondansetron , Postoperative Nausea and Vomiting , Humans , Female , Postoperative Nausea and Vomiting/etiology , Breast Neoplasms/surgery , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Middle Aged , Adult , Antiemetics/therapeutic use , Dexamethasone/therapeutic use , Ondansetron/therapeutic use , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/genetics , Pregnancy , Genetic Predisposition to Disease , Risk Factors , Incidence
19.
Growth Factors ; 42(2): 84-100, 2024 05.
Article in English | MEDLINE | ID: mdl-38889447

ABSTRACT

N-acetylgalactosaminyltransferases (GALNTs) are a polypeptide responsible for aberrant glycosylation in breast cancer (BC), but the mechanism is unclear. In this study, expression levels of GALNT6, GALNT14, and Gal-3 were assessed in BC, and their association with GDF-15, ß-catenin, stemness (SOX2 and OCT4), and drug resistance marker (ABCC5) was evaluated. Gene expression of GALNT6, GALNT14, Gal-3, GDF-15, OCT4, SOX2, ABCC5, and ß-catenin in tumor and adjacent non-tumor tissues (n = 30) was determined. The same was compared with GEO-microarray datasets. A significant increase in the expression of candidate genes was observed in BC tumor compared to adjacent non-tumor tissue; and in pre-therapeutic patients compared to post-therapeutic. GALNT6, GALNT14, Gal-3, and GDF-15 showed positive association with ß-catenin, SOX2, OCT4, and ABCC5 and were significantly associated with poor Overall Survival. Our findings were also validated via in silico analysis. Our study suggests that GALNT6, GALNT14, and Gal-3 in association with GDF-15 promote stemness and intrinsic drug resistance in BC, possibly by ß-catenin signaling pathway.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Growth Differentiation Factor 15 , N-Acetylgalactosaminyltransferases , Polypeptide N-acetylgalactosaminyltransferase , beta Catenin , Humans , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , beta Catenin/metabolism , beta Catenin/genetics , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Neoplastic Stem Cells/metabolism , Middle Aged , Gene Expression Regulation, Neoplastic , Cell Line, Tumor
20.
Nat Commun ; 15(1): 5190, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890300

ABSTRACT

Mitochondrial-secreted growth differentiation factor-15 (GDF-15) promotes weight loss in animals. Its effects in humans remain unclear, due to limited research and potential measurement interference from the H202D-variant. Our post-hoc analysis investigates total (irrespective of genetic variants) and H-specific GDF-15 (detected only in H202D-variant absence) in humans under acute and chronic energy deprivation, examining GDF-15 interaction with leptin (energy homeostasis regulator) and GDF-15 biologic activity modulation by the H202D-variant. Total and H-specific GDF-15 increased with acute starvation, and total GDF-15 increased with chronic energy deprivation, compared with healthy subjects and regardless of leptin repletion. Baseline GDF-15 positively correlated with triglyceride-rich particles and lipoproteins. During acute metabolic stress, GDF-15 associations with metabolites/lipids appeared to differ in subjects with the H202D-variant. Our findings suggest GDF-15 increases with energy deprivation in humans, questioning its proposed weight loss and suggesting its function as a mitokine, reflecting or mediating metabolic stress response.


Subject(s)
Growth Differentiation Factor 15 , Leptin , Humans , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/blood , Leptin/metabolism , Leptin/blood , Male , Adult , Female , Energy Metabolism , Starvation/metabolism , Young Adult , Middle Aged , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL