bims-endanx Biomed News
on Endocrine Anxiety
Issue of 2025–01–26
seventeen papers selected by
Logan K. Townsend, McMaster University



  1. Curr Mol Med. 2025 Jan 20.
       BACKGROUND: Growth Differentiation Factor 15 (GDF15) has been described as influencing skeletal physiology. Nevertheless, no systematic appraisal of the effect of GDF15 on skeletal muscle tissues has been developed to the present day.
    OBJECTIVE: The aim of the present work was to review the evidence on the topic.
    METHODS: In this preregistered systematic review (https://osf.io/wa8xr), articles were retrieved from MEDLINE/PubMed, EMBASE, and WebOfScience. Inclusion criteria comprised studies on humans or animal models, assessment of peripheral or local tissue GDF15 concentrations, as well as the direct expression of GDF15 in skeletal muscle, and direct or indirect correlates of GDF15 with physical activity/ sarcopenia/trophism/ function.
    RESULTS: A total of 646 studies were retrieved, and 144 finally included. Molecular inducers or inhibitors of GDF15 in skeletal muscle tissues were described. GDF15 was reported to promote skeletal muscle health, metabolic homeostasis, and overall physical conditioning. In pathology, GDF15 seems to be correlated to the degree of muscle impairment and mitochondrial stress. GDF15 has also been described as having the potential to stratify patients based on clinical prognosis and functional outcome.
    CONCLUSION: A hormetic hypothesis for GDF15 on skeletal muscle was proposed. In fact, GDF15 exhibited beneficial effects when expressed at high levels facing acute stressors (i.e., "myoprotection"). Conversely, GDF15 exhibited maladaptive effects, such as chronic low-grade inflammation, when chronically expressed in pathological processes (e.g., obesity, aging). GDF15 may be a potential molecular target for disease-modifying interventions. The current review underscores the need for further research on GDF15 to elucidate its therapeutic potential across different pathological states. The study protocol, registered before data collection and analysis, can be retrieved at https://osf.io/wa8xr. It should be noted that the study deviated from the protocol after peer review, including other electronic databases beyond MEDLINE/PubMed alone.
    Keywords:  Cytokines; Growth Factors; Mitokines; Muscle Fatigue; Muscle Strength; Myokines; Psychomotor Performance; TGF-β Superfamily.
    DOI:  https://doi.org/10.2174/0115665240327723241018073535
  2. Diabetol Metab Syndr. 2025 Jan 23. 17(1): 31
       BACKGROUND: Growth differentiation factor 15 (GDF15) is significantly correlated with glycolipid metabolic disorders. Increased GDF15 levels are associated with obesity, insulin resistance, and diabetes as well as a poorer diabetes progression and prognosis. This is a prospective cohort study investigated the association between circulating GDF15 and diabetic peripheral artery disease.
    METHODS: A total of 174 diabetic patients aged 20-80 were enrolled. Plasma GDF15 levels were measured using ELISA. Peripheral Artery Disease (PAD) was evaluated with the Ankle brachial index (ABI) and the Cardio-ankle vascular index (CAVI).
    RESULTS: We found that diabetic patients with higher serum GDF15 levels (mean: 2521.5 pg/mL) had a higher incidence of peripheral artery disease. Multivariate logistic regression analysis indicated that patients with high serum GDF15 levels were at an increased risk of developing peripheral artery disease. High GDF15 levels were associated with ABI < 0.9 (right and left mean 19.5% p = 0.80, OR:1.13; 95%CI: [0.44-2.90]). Increased age (p = 0.025 OR:1.02; 95% CI [0.13-0.87]), family history (p = 0.001 OR:1.37; 95%CI: [0.37-5.05]), heart failure (p = 0.002 OR:4.96; 95%CI: [1.76-13.97]), sodium-glucose linked transporter 2 (SGLT 2) inhibitor use (p = 0.026), estimated glomerular filtration rate (eGFR) (p = < 0.001), and uric acid (p = < 0.001) was also positively associated with high GDF15 levels. Urine albumin-to-creatinine ratio (UACR) (p = < 0.010) was associated with higher GDF15 levels after one year of follow up.
    CONCLUSIONS: Elevated GDF15 was significantly associated with worsening metabolic parameters and an increased risk of peripheral artery disease. Thus, it may be a stronger predictor of these outcomes in people with diabetes.
    Keywords:  Growth differentiation factor 15; Peripheral artery disease; Type 2 DM
    DOI:  https://doi.org/10.1186/s13098-025-01588-w
  3. Physiol Rev. 2025 Jan 17.
      Metabolic energy stored mainly as adipose tissue is homeostatically regulated. There is strong evidence that human body weight (BW) is physiologically regulated, i.e. maintained within a relatively narrow range in most mammals, including humans. Nevertheless, the prevalence of obesity has increased markedly in recent decades and now constitutes major medical and socioeconomic problems worldwide. This review focuses on understanding this paradox and the clinical issues that it has spawned: how and why do individuals become obese and how can we help those with obesity lose excess BW and body fat and maintain whatever loss they achieve. Excess BW gain occurs when physiological responses that usually resist short-term weight gain fail to compensate for excess caloric intake occurring over extended periods of time, often over many years or even decades. On the other hand, the difficulties of achieving BW loss and maintenance of reduced BW in obese subjects are due, in part, to the operation of the same physiological regulatory system that helps maintain a healthy BW in individuals without obesity. But, given obesity's association with many pathological conditions, we maintain that the physiological processes that resist BW loss and persistently drive regain are examples of dysregulation. Here we review research in humans and animals addressing these and other unresolved issues in the physiology of obesity. We bring important unresolved problems into focus, and, in some cases, propose hypotheses that can further elucidate their mechanisms to provide research opportunities into modalities that might lead to more effective treatments of obesity.
    Keywords:  Central nervous system; energy homeostasis; leptin; obesity; weight regain
    DOI:  https://doi.org/10.1152/physrev.00002.2024
  4. Hepatol Int. 2025 Jan 21.
       BACKGROUND: Our previous research demonstrated that growth differentiation factor 15 (GDF15) exhibited superior predictive capability for metabolic dysfunction-associated steatohepatitis (MASH) development with an AUC of 0.86 at 10 years before disease diagnosis. However, the specific pathways and molecular mechanisms associated with GDF15 expression during MASH development remain to be fully investigated in humans.
    METHODS: A nested case-control study comprising a MASH group of 78 individuals and three age- and sex-matched control groups (156 metabolic dysfunction-associated steatosis, 78 viral hepatitis, and 156 normal liver controls) was conducted. The baseline levels of GDF15-related transcription factors and upstream signaling pathways associated with the identified transcription factors were analysed prospectively.
    RESULTS: The significantly higher level of nuclear factor of activated T cells 3 (NFATC3), a transcription factor for GDF15, was identified in the circulation in MASH patients compared to controls. Expression of the non-canonical Wnt signaling pathway that is upstream of NFATC3, and its related proteins CTHRC1, FRZB, SFRP1, and SFRP4, were highest in the MASH group, suggesting a non-canonical Wnt signaling/NFATC3/GDF-15 cascade in MASH disease pathogenesis. A predictive model for MASH development based on four biomarkers (CTHRC1, FRZB, NFATC3, and GDF15) showed an AUC of 0.90 at 10 years. A protein-clinical model that included these four circulating proteins and BMI yielded an AUC of 0.93 at 10 years.
    CONCLUSIONS: Non-canonical Wnt signaling pathway may activate NFATC3 to promote GDF15 expression in MASH disease pathogenesis. These molecular mechanisms provide novel insights for developing targeted therapies that could modulate the non-canonical Wnt/NFATC3/GDF15 cascade to prevent/treat MASH.
    Keywords:  Metabolic dysfunction-associated steatohepatitis; Metabolic dysfunction-associated steatotic liver disease; Non-canonical Wnt signaling; Nuclear factor of activated T cells 3; Predictive biomarkers
    DOI:  https://doi.org/10.1007/s12072-024-10775-2
  5. Annu Rev Pathol. 2025 Jan;20(1): 483-509
      The immune system plays fundamental roles in maintaining physiological homeostasis. With the increasing prevalence of obesity-a state characterized by chronic inflammation and systemic dyshomeostasis-there is growing scientific and clinical interest in understanding how obesity reshapes immune function. In this review, we propose that obesity is not merely an altered metabolic state but also a fundamentally altered immunological state. We summarize key seminal and recent findings that elucidate how obesity influences immune function, spanning its classical role in microbial defense, its contribution to maladaptive inflammatory diseases such as asthma, and its impact on antitumor immunity. We also explore how obesity modulates immune function within tissue parenchyma, with a particular focus on the role of T cells in adipose tissue. Finally, we consider areas for future research, including investigation of the durable aspects of obesity on immunological function even after weight loss, such as those observed with glucagon-like peptide-1 (GLP-1) receptor agonist treatment. Altogether, this review emphasizes the critical role of systemic metabolism in shaping immune cell functions, with profound implications for tissue homeostasis across various physiological contexts.
    Keywords:  GLP-1 receptor agonists; PPARγ; adipose tissue immunology; high-fat diet; immune system; inflammatory disease; microbiome; obesity; tumor immunity
    DOI:  https://doi.org/10.1146/annurev-pathmechdis-051222-015350
  6. Elife. 2025 Jan 20. pii: RP96724. [Epub ahead of print]13
      Stress is a potent modulator of pain. Specifically, acute stress due to physical restraint induces stress-induced analgesia (SIA). However, where and how acute stress and pain pathways interface in the brain are poorly understood. Here, we describe how the dorsal lateral septum (dLS), a forebrain limbic nucleus, facilitates SIA through its downstream targets in the lateral hypothalamic area (LHA) of mice. Taking advantage of transsynaptic viral-genetic, optogenetic, and chemogenetic techniques, we show that the dLS→LHA circuitry is sufficient to drive analgesia and is required for SIA. Furthermore, our results reveal that the dLS→LHA pathway is opioid-dependent and modulates pain through the pro-nociceptive neurons in the rostral ventromedial medulla (RVM). Remarkably, we found that the inhibitory dLS neurons are recruited specifically when the mice struggle to escape under restraint and, in turn, inhibit excitatory LHA neurons. As a result, the RVM neurons downstream of LHA are disengaged, thus suppressing nociception. Together, we delineate a poly-synaptic pathway that can transform escape behavior in mice under restraint to acute stress into analgesia.
    Keywords:  analgesia; chronic pain; mouse; neuroscience; septum; stress
    DOI:  https://doi.org/10.7554/eLife.96724
  7. Mol Metab. 2025 Jan 18. pii: S2212-8778(25)00006-7. [Epub ahead of print] 102099
       OBJECTIVE: Obesity-associated metabolic dysfunction is a major public health concern worldwide. Endothelial dysfunction is a hallmark of metabolic dysfunction, and endothelial cells affect metabolic functions. Because autophagy-related gene 7 (ATG7) is involved in various cellular physiology, we investigated the roles of endothelial cell-ATG7 (EC-ATG7) on high-fat diet-induced obesity and its related metabolic dysfunction.
    METHODS: We generated an endothelial-specific Atg7 knock-out mouse by breeding Atg7flox/flox mouse with the Chd5-Cre mouse, and investigated the metabolic phenotypes associated with high-fat diet (HFD)-induced obesity. Body weight, food intake, glucose tolerance, insulin sensitivity, and liver fat accumulation were measured in endothelial Atg7 deficient (Atg7ΔEnd) and control mice (Atg7f/f). Adipose tissue inflammation was assessed by measuring the expression of pro-inflammatory genes. Furthermore, we performed indirect calorimetry and examined the insulin signaling pathway molecules.
    RESULTS: We found that deletion of EC-Atg7 ameliorated HFD-induced weight gain, fatty liver, and adipocyte hypertrophy and inflammatory response in adipose tissue, and improved insulin sensitivity without changing glucose tolerance. These metabolic effects seem to be due to the reduced food intake because there were no differences in energy expenditure, energy excretion to feces, and physical activity. Interestingly, the deletion of EC-Atg7 protected from HFD-induced vascular rarefaction, and the knock-down of Atg7 in endothelial cells protected from fatty acid-induced cell death.
    CONCLUSIONS: Our results suggest that EC-Atg7 deletion ameliorates HFD-induced obesity and its related metabolic dysfunction, such as insulin resistance and fatty liver by attenuating appetite and vascular rarefaction. The EC-Atg7 deletion may protect the endothelial cells from lipotoxicity and impaired angiogenesis, which preserves the endothelial function in metabolic tissues. These findings may have implications for developing new therapeutic strategies for preventing and treating obesity and its associated health risks.
    Keywords:  ATG7; Autophagy; Endothelial cell; Insulin resistance; Obesity
    DOI:  https://doi.org/10.1016/j.molmet.2025.102099
  8. Bioessays. 2025 Jan 19. e202400203
      Adipose tissue (AT) inflammation, a hallmark of the metabolic syndrome, is triggered by overburdened adipocytes sending out immune cell recruitment signals during obesity development. An AT immune landscape persistent throughout weight loss and regain constitutes an immune-obesogenic memory that hinders long-term weight loss management. Lipid-associated macrophages (LAMs) are emerging as major players in diseased, inflamed metabolic tissues and may be key contributors to an obesogenic memory in AT. Our previous study found that LAM abundance increases with weight loss via intermittent fasting (IF) in obese mice, which is driven by adipocyte p53 signalling. However, the specific signals causing LAM accumulation in AT under IF remain unknown. In this piece, we hypothesise on a range of adipocyte-secreted signals that can harbor immune-attractive features upon fasting/refeeding cycles. We highlight possible mechanisms including cell death signalling, matrikines, and other damage-associated molecular patterns (DAMPs), as well as adipo(-cyto)kines, lipid mediators, metabolites, extracellular vesicles, and epigenetic rewiring. Finally, we consider how advances in mechanisms of AT LAM recruitment gleaned from preclinical models might be translatable to long-term weight management in humans. Thus, we provide vantage points to study signals driving monocyte recruitment, polarisation towards LAMs, and LAM retention, to harness the therapeutic potential of modulating AT LAM levels by impacting the immune-obesogenic memory in metabolic disease.
    Keywords:  adipose; lipid‐associated macrophages; metabolic syndrome; metaflammation; obesity
    DOI:  https://doi.org/10.1002/bies.202400203
  9. J Neuroinflammation. 2025 Jan 19. 22(1): 10
      Major depressive disorder is a prevalent mental disorder, yet its pathogenesis remains poorly understood. Accumulating evidence implicates dysregulated immune mechanisms as key contributors to depressive disorders. This review elucidates the complex interplay between peripheral and central immune components underlying depressive disorder pathology. Peripherally, systemic inflammation, gut immune dysregulation, and immune dysfunction in organs including gut, liver, spleen and adipose tissue influence brain function through neural and molecular pathways. Within the central nervous system, aberrant microglial and astrocytes activation, cytokine imbalances, and compromised blood-brain barrier integrity propagate neuroinflammation, disrupting neurotransmission, impairing neuroplasticity, and promoting neuronal injury. The crosstalk between peripheral and central immunity creates a vicious cycle exacerbating depressive neuropathology. Unraveling these multifaceted immune-mediated mechanisms provides insights into major depressive disorder's pathogenic basis and potential biomarkers and targets. Modulating both peripheral and central immune responses represent a promising multidimensional therapeutic strategy.
    Keywords:  Major depressive disorder; Neuroimmune interactions; Neuroinflammation; Peripheral-central crosstalk
    DOI:  https://doi.org/10.1186/s12974-024-03312-3
  10. Mol Syst Biol. 2025 Jan 23.
      Elevated cortisol in chronic stress and mood disorders causes morbidity including metabolic and cardiovascular diseases. There is therefore interest in developing drugs that lower cortisol by targeting its endocrine pathway, the hypothalamic-pituitary-adrenal (HPA) axis. However, several promising HPA-modulating drugs have failed to reduce long-term cortisol in mood disorders, despite effectiveness in other hypercortisolism conditions such as Cushing's syndrome. The reasons for these failures remain unclear. Here, we use a mathematical model of the HPA axis to demonstrate that the pituitary and adrenal glands compensate for drug effects by adjusting their functional mass, a feedback mechanism absent in Cushing tumors. Our systematic in silico analysis identifies two interventions targeting corticotropin-releasing hormone (CRH) as effective for lowering long-term cortisol. Other targets either fail due to gland mass compensation or harm other aspects of the HPA axis. We propose CRH-neutralizing antibodies and CRH-synthesis inhibitors as potential targets for reducing long-term cortisol in mood disorders and chronic stress. More generally, this study indicates that understanding the slow compensatory mechanisms in endocrine axes can be crucial to prioritize drug targets.
    Keywords:  Dynamic Compensation; HPA Axis; Mood Disorders; Systems Endocrinology; Systems Pharmacology
    DOI:  https://doi.org/10.1038/s44320-024-00083-0
  11. J Biol Chem. 2025 Jan 17. pii: S0021-9258(25)00053-5. [Epub ahead of print] 108206
      It has been well established that adenosine plays a key role in the control of inflammation through G protein coupled receptors and recently shown that it can regulate thermogenesis. Here we investigated the specific requirements of the adenosine A2A receptor (A2AR) in mature adipocytes for thermogenic functionality and metabolic homeostasis. We generated fat tissue specific adenosine A2A receptor knock-out mice to assess the influence of signaling through this receptor on brown and beige fat functionality, obesity, insulin sensitivity, inflammation and liver function. Fat specific A2AR knock-out and wild type littermate mice were compared for potential differences in cold tolerance and energy metabolism. In addition, we measured glucose metabolism, AT inflammation and liver phenotypes in mice of the two genotypes after exposure to a diet rich in fat. Our results provide novel evidence indicating that loss of the adenosine A2A receptor specifically in adipocytes is associated with cold intolerance and decreased oxygen consumption. Furthermore, mice with fat specific ablation of the A2AR exposed to a diet rich in fat showed increased propensity to obesity, decreased insulin sensitivity, elevated adipose tissue inflammation and hepato-steatosis and -steatitis. Overall, our data provide novel evidence that A2AR in mature adipocytes safeguards metabolic homeostasis, suggesting the possibility of targeting this receptor selectively in fat for the treatment of metabolic disease.
    Keywords:  UCP1; adenosine A2A receptor; brown and beige adipose tissue; insulin resistance; obesity
    DOI:  https://doi.org/10.1016/j.jbc.2025.108206
  12. Front Physiol. 2024 ;15 1522471
      Lifestyle-related diseases, such as atherosclerosis and diabetes, are now considered to be a series of diseases caused by chronic inflammation. Adipose tissue is considered to be an endocrine organ that not only plays a role in lipid storage, heat production, and buffering, but also produces physiologically active substances and is involved in chronic inflammation. Perivascular adipose tissue (PVAT) surrounding blood vessels similarly produces inflammatory and anti-inflammatory physiologically active substances that act on blood vessels either directly or via the bloodstream. Epicardial adipose tissue (EAT), which is in direct contact with the coronary arteries inside the pericardium, is thought to have a direct effect on the coronary arteries as well. The presence and inflammatory status of these adipose tissues can be evaluated by imaging tests, and has been shown to be associated with the presence of current cardiovascular disease (CVD) and to be a prognostic factor. It is also expected to become a new diagnostic and therapeutic target for CVD.
    Keywords:  atherosclerosis; cardiovascular disease (CVD); chronic inflammation; epicardial adipose tissue (EAT); perivascular adipose tissue (PVAT); vasa vasorum (VV)
    DOI:  https://doi.org/10.3389/fphys.2024.1522471
  13. J Physiol Sci. 2024 ;pii: S1880-6546(24)00098-2. [Epub ahead of print]74(1): 11
      Many hormones act on the hypothalamus to control hunger and satiety through various pathways closely associated with several factors. When food is present in the gastro intestinal (GI) tract, enteroendocrine cells (EECs) emit satiety signals such as cholecystokinin (CCK), glucagon like peptide-1 (GLP-1) and peptide YY (PYY), which can then communicate with the vagus nerve to control food intake. More specifically, satiety has been shown to be particularly affected by the GLP-1 hormone and its receptor agonists that have lately been acknowledged as a promising way to reduce weight. In addition, there is increasing evidence that normal flora is also involved in the peripheral, central, and reward system that impact satiety. Moreover, neurologic pathways control satiety through neurotransmitters. In this review, we discuss the different roles of each of the GLP-1 hormone and its agonist, gut microbiomes, as well as neurotransmitters and their interconnected relation in the regulation of body's satiety homeostasis.
    Keywords:  GLP-1; Microbiota; Neuroscience; Neurotransmitters; Satiety
    DOI:  https://doi.org/10.1186/s12576-024-00904-9
  14. Neuroscience. 2025 Jan 21. pii: S0306-4522(25)00033-8. [Epub ahead of print]
      There is mounting evidence for the involvement of the immune system, neuroinflammation and disturbed gut microbiota, or dysbiosis, in attentive-deficit/hyperactivity disorder (ADHD). Gut dysbiosis is strongly implicated in many physical, autoimmune, neurological, and neuropsychiatric conditions, however knowledge of its particular pathogenic role in ADHD is sparse. As such, this narrative review examines and synthesizes the available evidence related to inflammation, dysbiosis, and neural processes in ADHD. Minimal differences in microbiota diversity measures between cases and controls were found, however many relative abundance differences were observed at all classification levels (phylum to strain). Compositional differences of taxa important to key gut-brain axis pathways, in particular Bacteroides species and Faecalibacterium, may contribute to inflammation, brain functioning differences, and symptoms, in ADHD. We have identified one possible model of ADHD etiopathogenesis involving systemic inflammation, an impaired blood-brain barrier, and neural disturbances as downstream consequences of gut dysbiosis. Nevertheless, studies conducted to date have varied degrees of methodological rigour and involve diverse participant characteristics and analytical techniques, highlighting a need for additional research.
    Keywords:  Attention-deficit/hyperactivity disorder; Dysbiosis; Gut Microbiome; Gut-Brain Axis; Inflammation; Neuroinflammation; Oxidative Stress
    DOI:  https://doi.org/10.1016/j.neuroscience.2025.01.031
  15. J Hepatol. 2025 Jan 15. pii: S0168-8278(25)00002-9. [Epub ahead of print]
       BACKGROUND & AIMS: Lanifibranor is a pan-PPAR agonist that improves glucose/lipid metabolism and reverses steatohepatitis and fibrosis in adults with MASH. We tested its effect on insulin resistance at the level of different target tissues in relationship to change in intrahepatic triglyceride (IHTG) content.
    METHODS: This phase 2, single center, study randomized (1:1) 38 patients with T2D and MASLD to receive lanifibranor 800 mg or placebo for 24 weeks. The primary endpoint was the change in IHTG (1H-MRS). The main prespecified secondary endpoint was the change in hepatic, muscle and adipose tissue insulin sensitivity using the gold-standard euglycemic hyperinsulinemic clamp technique measuring glucose turnover. Other secondary endpoints included changes in cardiometabolic parameters (i.e., HbA1c, lipid profile, adiponectin).
    RESULTS: Lanifibranor compared to placebo significantly lowered IHTG (full analysis set [FAS] -44% vs. -12%, respectively; least squares mean difference -31%, 95% CI -51 to -12%; in completers -50% vs. -16%; both p<0.01). More patients reached ≥30% IHTG reduction with lanifibranor compared to placebo (FAS 65% vs. 22%; completers 79% vs. 29%; both p<0.01) and steatosis resolution (FAS 25% vs. 0%; p<0.05). Lanifibranor significantly improved hepatic and peripheral insulin resistance (i.e., fasting endogenous [primarily hepatic] glucose production, hepatic IR, and insulin-stimulated muscle glucose disposal or Rd). Secondary metabolic endpoints also improved (fasting glucose, insulin, HOMA-IR, HbA1c; HDL-C), and adiponectin increased 2.4-fold (all p<0.001). Lanifibranor caused modest weight gain (+2.7%). Adverse events were mild (gastrointestinal side effects, hemoglobin decrease) and drug-related TEAE leading to study discontinuation were balanced between groups.
    CONCLUSIONS: Lanifibranor significantly improves hepatic, muscle and adipose tissue insulin resistance. Lanifibranor treatment was safe and effective in reducing hepatic steatosis and cardiometabolic risk factors associated with metabolic dysfunction.
    IMPACT AND IMPLICATIONS: No prior studies have evaluated the effect of lanifibranor on insulin sensitivity at the level of muscle, liver and adipose tissue and its relationship to changes in intrahepatic triglyceride (IHTG) content in insulin resistant subjects with MASLD and T2D. We observed a significant decrease in IHTG after 24 weeks of treatment (by ∼50%, p < 0.001 versus placebo) that was associated with a major improvement in hepatic and peripheral (Rd) insulin sensitivity, restoration of adipose tissue function with more than two-fold increase in plasma adiponectin concentration and improvement in cardiometabolic risk factors. This is the first in-depth study on how a pan-PPAR approach reverses steatosis and metabolic dysfunction in patients with T2D and MASLD. It has important clinical implications because it offers proof-of-concept that by targeting the key underlying metabolic defects in MASLD (i.e., insulin resistance, lipotoxicity and hyperglycemia) one can restore cardiometabolic health and offers a compelling rationale for treating with lanifibranor individuals with MASLD, either alone or in combination with weight loss and other treatment strategies.
    CLINICALTRIALS:
    GOV IDENTIFIER: NCT03459079.
    Keywords:  Metabolic dysfunction-associated steatotic liver disease (MASLD); insulin resistance; lanifibranor; peroxisome proliferator-activated receptor (PPAR); type 2 diabetes
    DOI:  https://doi.org/10.1016/j.jhep.2024.12.045
  16. Front Pharmacol. 2024 ;15 1451895
       Introduction: Stress-evoked dysfunctions of the frontal cortex (FC) are correlated with changes in the functioning of the glutamatergic system, and evidence demonstrates that noradrenergic transmission is an important regulator of this process. In the current study, we adopted a restraint stress (RS) model in male Wistar rats to investigate whether the blockade of β1 adrenergic receptors (β1AR) with betaxolol (BET) in stressed animals influences the body's stress response and the expression of selected signaling proteins in the medial prefrontal cortex (mPFC).
    Methods: The study was divided into two parts. In the first part, rats were exposed to RS for 3, 7, or 14 days, and the expression of glutamate signaling proteins (p(S845)/t GluA1, p(Y1472)/t GluN2B, VGLUT1, and VGLUT2) in the FC was analyzed to determine the optimal RS duration for studying the mechanisms of hypofrontality. In the second part, rats were exposed to RS for 14 days, and BET (5 mg/kg, p. o.) was administered during the last 8 days immediately after RS. The body's stress reaction was assessed by analyzing body weight and blood levels of adrenocorticotropic hormone (ACTH) and corticosterone (CORT). Behavioral responses were evaluated using the novel object recognition (NOR) and elevated plus maze (EPM) tests. The impact of RS and BET on the expression of p(Y530)/t Fyn and p (S133)/t CREB in the mPFC was measured via Western blotting.
    Results and Discussion: The first part of the study demonstrated a decreased level of glutamate receptors in rats exposed to 14 days of RS, following an initial increase observed after 7 days of RS. Results from the second part revealed that chronic RS reduced body weight, impaired recognition memory in the NOR test, augmented blood levels of ACTH, and increased the expression of p(Y530) Fyn in the mPFC. However, β1AR blockade did not alter the effects of RS on weight gain, cognitive function, or the expression of p(Y530) Fyn. β1AR blockade normalized only the blood concentration of ACTH. These results suggest that decreased Fyn kinase activity, indicated by phosphorylation at Y530, underlies the stress-evoked downregulation of GluN2B in the FC in a manner independent of β1AR activity.
    Keywords:  Fyn kinase; adrenocorticotropic hormone; betaxolol; elevated plus maze; frontal cortex; glutamate receptors; novel object recognition; restraint stress
    DOI:  https://doi.org/10.3389/fphar.2024.1451895
  17. Mol Med. 2025 Jan 21. 31(1): 16
      Severe sepsis is cognate with life threatening multi-organ dysfunction. There is a disturbance in endocrine functions with alterations in several hormonal pathways. It has frequently been linked with dysfunction in the hypothalamic pituitary-adrenal axis (HPA). Increased cortisol or cortisolemia is evident throughout the acute phase, along with changes in the hypothalamic pituitary thyroid (HPT) axis, growth hormone-IGF-1 axis, insulin-glucose axis, leptin, catecholamines, renin angiotensin aldosterone axis, ghrelin, glucagon, hypothalamic pituitary gonadal (HGA) axis, and fibroblast growth factor-21. These changes and metabolic alterations constitute the overall response to infection in sepsis. Further research is essential to look into the hormonal changes that occur during sepsis, not only to understand their potential relevance in therapy but also because they may serve as prognostic indicators.
    Keywords:  Endocrine; Hormones; Inflammation; Metabolism; Sepsis; Septic shock
    DOI:  https://doi.org/10.1186/s10020-025-01074-z