bims-endanx Biomed News
on Endocrine Anxiety
Issue of 2025–02–09
twenty papers selected by
Logan K. Townsend, McMaster University



  1. ChemMedChem. 2025 Feb 05. e202400961
      Growth differentiation factor 15 (GDF15) is a TGF-β superfamily member involved in diverse physiological and pathological processes. It is expressed in various tissues and its circulating levels rise during exercise, aging, pregnancy, and conditions such as cancer, cardiovascular disease, and infections. The biological activities of GDF15, including anorexia and cachexia, are primarily mediated through the GFRAL receptor, localized in the brainstem and functioning via RET co-receptor recruitment. This signaling is crucial for energy homeostasis and nausea induction. Recent studies suggest a broader GFRAL distribution, potentially explaining GDF15's distinct roles. These findings sparked interest in leveraging GDF15-GFRAL pathways for therapeutic development. Two primary strategies include GDF15 analogues as GFRAL agonists for obesity treatment and GDF15-derived peptides as antagonists to counteract cancer-induced cachexia and related disorders. This review highlights advancements in understanding GDF15-GFRAL signaling and its implications, summarizing bioactive GDF15-derived molecules, their pharmacological applications, and offering insights into novel treatment avenues for GDF15-associated conditions.
    Keywords:  CYTOKINES; Growth factors; Peptides; Proteins; Receptors
    DOI:  https://doi.org/10.1002/cmdc.202400961
  2. Diabetes Obes Metab. 2025 Feb 05.
       AIMS: This study aims to determine the predictive capability for metabolic dysfunction-associated steatohepatitis (MASH) long before its diagnosis by using six previously identified diagnostic biomarkers for metabolic dysfunction-associated steatotic liver disease (MASLD) with proteomic data from the UK Biobank.
    MATERIALS AND METHODS: A nested case-control study comprising a MASH group and three age- and sex-matched control groups (metabolic dysfunction-associated steatosis, viral hepatitis and normal liver controls) was conducted. Olink proteomics, anthropometric and biochemical data at baseline levels were obtained from the UK Biobank. The baseline levels of CDCP1, FABP4, FGF21, GDF15, IL-6 and THBS2 were analysed prospectively to determine their predictive accuracy for subsequent diagnosis with a mean lag time of over 10 years.
    RESULTS: At baseline, GDF15 demonstrated the best performance for predicting MASH occurrence at 5 and 10 years later, with AUCs of 0.90 at 5 years and 0.86 at 10 years. A predictive model based on four biomarkers (GDF15, FGF21, IL-6 and THBS2) showed AUCs of 0.88 at both 5 and 10 years. Furthermore, a protein-clinical model that included these four circulating protein biomarkers along with three clinical factors (BMI, ALT and TC) yielded AUCs of 0.92 at 5 years and 0.89 at 10 years.
    CONCLUSIONS: GDF15 at baseline levels outperformed other individual circulating protein biomarkers for the early prediction of MASH. Our data suggest that GDF15 and the GDF15-based model may be used as easy-to-implement tools to identify patients with high risks of developing MASH at a mean lag time of over 10 years.
    Keywords:  growth differentiation factor 15; metabolic dysfunction‐associated steatohepatitis; metabolic dysfunction‐associated steatotic liver disease; predictive biomarkers; proteomics
    DOI:  https://doi.org/10.1111/dom.16233
  3. bioRxiv. 2025 Jan 22. pii: 2025.01.17.633599. [Epub ahead of print]
      People frequently experience cycles of weight gain and loss. This weight cycling has been demonstrated, in humans and animal models, to increase cardiometabolic disease and disrupt glucose homeostasis. Obesity itself - and to an even greater extent weight regain - causes adipose tissue inflammation, resulting in metabolic dysfunction. Studies show that even after weight loss, increased numbers of lipid associated macrophages and memory T cells persist in adipose tissue and become more inflammatory upon weight regain. These findings suggest that the immune system retains a "memory" of obesity, which may contribute to the elevated inflammation and metabolic dysfunction associated with weight cycling. Here, we show that blocking the CD70-CD27 axis, critical for formation of immunological memory, decreases the number of memory T cells and reduces T cell clonality within adipose tissue after weight loss and weight cycling. Furthermore, while mice with impaired ability to create obesogenic immune memory have similar metabolic responses as wildtype mice to stable obesity, they are protected from the worsened glucose tolerance associated with weight cycling. Our data are the first to target metabolic consequences of weight cycling through an immunomodulatory mechanism. Thus, we propose a new avenue of therapeutic intervention by which targeting memory T cells can be leveraged to minimize the adverse consequences of weight cycling. These findings are particularly timely given the increasing use of efficacious weight loss drugs, which will likely lead to more instances of human weight cycling.
    DOI:  https://doi.org/10.1101/2025.01.17.633599
  4. Front Endocrinol (Lausanne). 2024 ;15 1456948
      Obesity is a major modifiable risk factor leading to neuroinflammation and neurodegeneration. Excessive fat storage in obesity promotes the progressive infiltration of immune cells into adipose tissue, resulting in the release of pro-inflammatory factors such as cytokines and adipokines. These inflammatory mediators circulate through the bloodstream, propagating inflammation both in the periphery and in the central nervous system. Gut dysbiosis, which results in a leaky intestinal barrier, exacerbates inflammation and plays a significant role in linking obesity to the pathogenesis of neuroinflammation and neurodegeneration through the gut-brain/gut-brain-liver axis. Inflammatory states within the brain can lead to insulin resistance, mitochondrial dysfunction, autolysosomal dysfunction, and increased oxidative stress. These disruptions impair normal neuronal function and subsequently lead to cognitive decline and motor deficits, similar to the pathologies observed in major neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Understanding the underlying disease mechanisms is crucial for developing therapeutic strategies to address defects in these inflammatory and metabolic pathways. In this review, we summarize and provide insights into different therapeutic strategies, including methods to alter gut dysbiosis, lifestyle changes, dietary supplementation, as well as pharmacological agents derived from natural sources, that target obesity-induced neuroinflammation and neurodegeneration.
    Keywords:  body-brain interactions; metabolic dysfunction; neurodegeneration; neuroinflammation; obesity; therapeutic targeting
    DOI:  https://doi.org/10.3389/fendo.2024.1456948
  5. Cell Metab. 2025 Jan 29. pii: S1550-4131(24)00526-6. [Epub ahead of print]
      Systemic metabolism ensures energy homeostasis through inter-organ crosstalk regulating thermogenic adipose tissue. Unlike the well-described inductive role of the sympathetic system, the inhibitory signal ensuring energy preservation remains poorly understood. Here, we show that, via the mechanosensor Piezo2, sensory neurons regulate morphological and physiological properties of brown and beige fat and prevent systemic hypermetabolism. Targeting runt-related transcription factor 3 (Runx3)/parvalbumin (PV) sensory neurons in independent genetic mouse models resulted in a systemic metabolic phenotype characterized by reduced body fat and increased insulin sensitivity and glucose tolerance. Deletion of Piezo2 in PV sensory neurons reproduced the phenotype, protected against high-fat-diet-induced obesity, and caused adipose tissue browning and beiging, likely driven by elevated norepinephrine levels. Finding that brown and beige fat are innervated by Runx3/PV sensory neurons expressing Piezo2 suggests a model in which mechanical signals, sensed by Piezo2 in sensory neurons, protect energy storage and prevent a systemic hypermetabolic phenotype.
    Keywords:  PIEZO2; Runx3/PV sensory neurons; body composition; brown and beige adipose tissues; glucose tolerance; insulin sensitivity; mechanosensing; metabolic diseases; norepinephrine; systemic metabolism
    DOI:  https://doi.org/10.1016/j.cmet.2024.12.016
  6. Obesity (Silver Spring). 2025 Feb 04.
       OBJECTIVE: Both alternate-day fasting (ADF) and calorie restriction (CR) are effective weight loss strategies. However, most individuals find it difficult to adhere to CR. Furthermore, CR can induce an excessive loss of not only fat but also muscle mass. This study aimed to compare the effects of ADF and pair-feeding (PF) CR on metabolic pathways underlying obesity in mice with high-fat diet (HFD)-induced obesity.
    METHODS: Male C57BL/6N Tac mice (n = 10 per group) were fed an HFD for 8 weeks to establish a diet-induced obesity model. Mice were then continued on the HFD with either alternate-day access to food or PF for the next 8 weeks. We measured body weight, adiposity, plasma biomarkers, and molecular mechanisms involving lipolysis and autophagy.
    RESULTS: Both ADF and PF resulted in comparable weight and fat loss. Compared with PF, ADF showed a significant reduction in liver weight and hepatic triglyceride levels. ADF significantly increased plasma ketone body levels and white adipose tissue lipolysis. Compared with PF, ADF tended to activate autophagy elongation and autophagosome formation, which were insignificant.
    CONCLUSIONS: These findings indicated that ADF is a promising intervention for metabolic diseases, potentially due to its superior efficacy in promoting ketogenesis and lipolysis compared with PF.
    DOI:  https://doi.org/10.1002/oby.24211
  7. Prog Neuropsychopharmacol Biol Psychiatry. 2025 Feb 03. pii: S0278-5846(25)00034-X. [Epub ahead of print]137 111280
      Major depressive disorder (MDD), also known as depression, is a prevalent mental disorder that leads to severe disease burden worldwide. Over the past two decades, significant progress has been made in understanding the pathogenesis and developing novel treatments for MDD. Among the complicated etiologies of MDD, chronic stress is a major risk factor. Exploring the underlying brain circuit mechanisms of chronic stress regulation has been an area of active research for recent years. A growing body of preclinical and clinical research has revealed that abnormalities in the brain circuits are closely associated with failures in coping with stress in depressed individuals. Nevertheless, neural circuit mechanisms underlying chronic stress processing and the onset of depression remain a major puzzle. Here, we review recent literature focusing on circuit- and cell-type-specific dissection of depression-like behaviors in chronic stress-related animal models of MDD and outline the key questions.
    Keywords:  Chemogenetics; Chronic stress; Major depressive disorder; Neural circuit tracing; Optogenetics
    DOI:  https://doi.org/10.1016/j.pnpbp.2025.111280
  8. Cell Rep Med. 2025 Jan 31. pii: S2666-3791(25)00009-6. [Epub ahead of print] 101936
      Amphetamine (AMPH) exerts metabolic and cardiovascular effects. The central melanocortin system is a key regulator of both metabolic and cardiovascular functions. Here, we show that the melanocortin system partially mediates AMPH-induced anorexia, energy expenditure, tachycardia, and hypertension. AMPH increased α-melanocyte stimulating hormone (αMSH) secretion from the hypothalamus, elevated blood pressure and heart rate (HR), increased brown adipose tissue (BAT) thermogenesis, and reduced both food intake (FI) and body weight (BW). In melanocortin 4 receptor-deficient (MC4R knockout [KO]) mice, metabolic and cardiovascular effects of AMPH were significantly attenuated. Antagonism of serotonergic and noradrenergic neurotransmitter systems attenuated AMPH-induced αMSH secretion as well as AMPH-induced metabolic and cardiovascular effects. We propose that AMPH increases serotonergic activation of proopiomelanocortin (POMC) neurons and reduces the noradrenergic inhibition of POMC neurons, thereby disinhibiting them. Together, these presynaptic mechanisms result in increased POMC activity, increased αMSH secretion, and increased activation of MC4R pathways that regulate both the metabolic and cardiovascular systems.
    Keywords:  BAT; MC4R; amphetamine; blood pressure; body weight; cardiovascular; energy expenditure; food intake; hypertension; melanocortin system; obesity; thermogenesis; weight loss
    DOI:  https://doi.org/10.1016/j.xcrm.2025.101936
  9. bioRxiv. 2025 Jan 24. pii: 2025.01.21.634167. [Epub ahead of print]
      GLP-1 receptor agonists (GLP1RAs) effectively reduce feeding to treat obesity, although nausea and other aversive side effects of these drugs can limit their use. Brainstem circuits that promote satiation and that mediate the physiologic control of body weight can be distinguished from those that cause aversion. It remains unclear whether brainstem Glp1r neurons contribute to the normal regulation of energy balance and whether GLP1RAs control appetite via circuits distinct from those that mediate aversive responses, however. Hence, we defined roles for AP and NTS Glp1r -expressing neurons (AP Glp1r and NTS Glp1r neurons, respectively) in the physiologic control of body weight, the GLP1RA-dependent suppression of food intake, and the GLP1RA-mediated stimulation of aversive responses. While silencing non-aversive NTS Glp1r neurons interfered with the physiologic restraint of feeding and body weight, restoring NTS Glp1r neuron Glp1r expression on an otherwise Glp1r -null background failed to enable long-term body weight suppression by GLP1RAs. In contrast, selective Glp1r expression in AP Glp1r neurons restored both aversive responses and long-term body weight suppression by GLP1RAs. Thus, while non-aversive NTS Glp1r neurons control physiologic feeding, aversive AP Glp1r neurons mediate both the anorectic and weight loss effects of GLP1RAs, dictating the functional inseparability of these pharmacologic GLP1RA responses at a circuit level.
    DOI:  https://doi.org/10.1101/2025.01.21.634167
  10. Diabetes Metab J. 2025 Feb 05.
       Background: Serotonin (5-hydroxytryptamine [5-HT]) is a monoamine neurotransmitter that has various functions in central and peripheral tissues. While 5-HT is known to regulate various biological processes in liver, direct role of 5-HT and its receptors, especially 5-HT receptor 2A (HTR2A) and HTR2B, in development and progression of alcoholic liver disease (ALD) in vivo is not well understood.
    Methods: Blood 5-HT level was measured from both human ALD patients and ethanol (EtOH) diet-fed mouse models. Gut-specific tryptophan hydroxylase 1 (Tph1) knockout mice, liver-specific Htr2a knockout mice, and liver-specific Htr2b knockout mice were fed with EtOH diet. Then we evaluated liver damage, hepatic steatosis, endoplasmic reticulum (ER) stress, and inflammation.
    Results: Blood 5-HT concentrations are increased in both humans and mice with ALD. Both gut-specific Tph1 knockout and liver- specific Htr2a knockout mice are resistant to steatosis by down-regulating lipogenic pathways in liver of chronic EtOH diet-fed mice. Moreover, genetic inhibition of both gut-derived serotonin (GDS) synthesis and hepatic HTR2A signaling prevents ER stress in liver of chronic EtOH diet-fed mice. Additionally, we found that ablation of HTR2A signaling protects against disease progression by attenuating liver injury and inflammation in chronic plus binge EtOH diet-fed mice. Also, inhibiting HTR2A signaling ameliorates alcohol-induced liver injury and ER stress in an acute EtOH diet-fed mice model.
    Conclusion: GDS directly regulates lipogenesis and ER stress via signaling through hepatic HTR2A in the context of ALD. Inhibiting HTR2A signaling protects against alcohol-induced steatosis, liver injury and disease progression in various ALD mouse models and may also provide a novel therapeutic strategy for ALD.
    Keywords:  Alcoholics; Endoplasmic reticulum stress; Fatty liver; Liver diseases; Receptors, serotonin; Serotonin; Tryptophan hydroxylase
    DOI:  https://doi.org/10.4093/dmj.2024.0215
  11. J Clin Endocrinol Metab. 2025 Feb 07. pii: dgaf073. [Epub ahead of print]
       OBJECTIVE: Fetuin B is a steatosis-responsive hepatokine that induces glucose intolerance in mice. Recently, we found that fetuin B in white adipose tissue was positively associated with peripheral insulin resistance in mice and a small study population, possibly through a fetuin B-induced inflammatory response in adipocytes. This translational study aimed to investigate the link between plasma fetuin B and the adipose tissue transcriptome and plasma proteome in a large cohort of humans.
    METHODS: Continuous linear regression analysis in R was applied to investigate the link between plasma fetuin B and the adipose tissue transcriptome (n=207) and plasma proteome (n=558) in humans, after adjustment for sex, age and study centre (model 1), model 1 + BMI (model 2) and model 2 + insulin sensitivity (MATSUDA-index) (model 3).
    RESULTS: Plasma fetuin B was associated with >100 genes in white adipose tissue, belonging to pathways related to cytokine/chemokine signaling (models 1 and 2) and insulin signaling (all models), and with >146 plasma proteins, involved in pathways related to metabolic processes and insulin signaling (all models).
    CONCLUSION: Plasma fetuin B is related to adipose tissue genes and plasma proteins involved in metabolic processes and insulin signaling. Our findings provide evidence for the involvement of white adipose tissue in fetuin B-induced insulin resistance.
    Keywords:  Fetuin B; glucose homeostasis; inflammation; insulin resistance; inter-organ crosstalk
    DOI:  https://doi.org/10.1210/clinem/dgaf073
  12. Biopsychosoc Sci Med. 2025 Feb-Mar 01;87(2):87(2): 153-159
       OBJECTIVE: Several studies suggest a bidirectional association between inflammation, and anxiety and depression. Elevated inflammatory cytokines generate and aggravate neuroinflammation, which may play a part in developing psychological symptoms. Growth differentiation factor 15 (GDF-15) is a novel biomarker possibly reflecting fibrosis and inflammation. The aim of the current study was to investigate the associations between levels of GDF-15 and symptoms of anxiety and depression in the general population.
    METHODS: We measured GDF-15 in middle-aged persons participating in the Akershus Cardiac Examination 1950 Study. Symptoms of anxiety and depression were assessed using the Hospital Anxiety and Depression Scale (HADS), with HADS ≥8 denoting significant symptoms. We used multivariable regression analysis to assess the associations between GDF-15 and HADS, adjusting for levels of C-reactive protein (CRP), demographics, and comorbidities.
    RESULTS: A total of 3638 participants had valid assessment of HADS and measurements of GDF-15 and CRP. The mean age was 63.9 (SD 0.65) years, and 48.8% were women. In adjusted models, levels of GDF-15 were associated with the continuous HADS-D score (β = 0.27, 95% confidence interval [CI] = 0.12 to 0.43) and HADS-D score ≥8 (odds ratio = 1.41, 95% CI = 1.12 to 1.78), but not with the continuous HADS-A score (β = 0.06, 95% CI = -0.12 to 0.24) or HADS-A score ≥8 (odds ratio = 1.06, 95% CI = 0.88 to 1.27).
    CONCLUSIONS: Levels of GDF-15 are independently associated with symptoms of depression in the general population. Our results reinforce the notion that inflammation may be a contributing factor for the development of clinical depression.
    REGISTRATION: ClinicalTrials.gov identifier NCT01555411 (Akershus Cardiac Examination [ACE] 1950 Study), https://clinicaltrials.gov/study/NCT01555411.
    DOI:  https://doi.org/10.1097/PSY.0000000000001365
  13. Cell. 2025 Jan 29. pii: S0092-8674(25)00047-9. [Epub ahead of print]
      Hunger is evolutionarily hardwired to ensure that an animal has sufficient energy to survive and reproduce. Just as important as knowing when to start eating is knowing when to stop eating. Here, using spatially resolved single-cell phenotyping, we characterize a population of neuropeptidergic neurons in the brainstem's dorsal raphe nucleus (DRN) and describe how they regulate satiation. These neurons track food from sensory presentation through ingestion, integrate these signals with slower-acting humoral cues, and express cholecystokinin (CCK). These CCK neurons bidirectionally regulate meal size, driving a sustained meal termination signal with a built-in delay. They are also well positioned to sense and respond to ingestion: they express a host of metabolic signaling factors and are integrated into an extended network known to regulate feeding. Together, this work demonstrates how DRN CCK neurons regulate satiation and identifies a likely conserved cellular mechanism that transforms diverse neurohumoral signals into a key behavioral output.
    Keywords:  cholecystokinin; dorsal raphe nucleus; feeding; molecular profiling; satiation
    DOI:  https://doi.org/10.1016/j.cell.2025.01.018
  14. bioRxiv. 2025 Jan 20. pii: 2025.01.17.633623. [Epub ahead of print]
      Circadian clocks present throughout the brain and body coordinate diverse physiological processes to support daily homeostasis and respond to changing environmental conditions. The local dependencies within the mammalian clock network are not well defined. We previously demonstrated that the skeletal muscle clock controls transcript oscillations of genes involved in fatty acid metabolism in the liver, yet whether the liver clock also regulates the muscle was unknown. Here, we use hepatocyte-specific Bmal1 KO mice (Bmal1 hep-/- ) and reveal that approximately one third of transcriptional rhythms in skeletal muscle are regulated by the liver clock vivo. Treatment of myotubes with serum harvested from Bmal1 hep-/- mice inhibited expression of genes involved in metabolic pathways, including oxidative phosphorylation. Overall, the transcriptional changes induced by liver clock-driven endocrine-communication revealed from our in vitro system were small in magnitude, leading us to surmise that the liver clock acts to fine-tune metabolic gene expression in muscle. Strikingly, treatment of myotubes with serum from Bmal1 hep-/- mice inhibited mitochondrial ATP production compared to WT and this effect was only observed with serum harvested during the active phase. Overall, our results reveal communication between the liver clock and skeletal muscle-uncovering a bidirectional endocrine communication pathway dependent on clocks in these two key metabolic tissues. Targeting liver and muscle circadian clocks may represent a potential avenue for exploration for diseases associated with dysregulation of metabolism in these tissues.
    DOI:  https://doi.org/10.1101/2025.01.17.633623
  15. Nat Commun. 2025 Feb 04. 16(1): 1330
      Physiological adaptations to fasting enable humans to survive for prolonged periods without food and involve molecular pathways that may drive life-prolonging effects of dietary restriction in model organisms. Mobilization of fatty acids and glycerol from adipocyte lipid stores by canonical neutral lipases, including the rate limiting adipose triglyceride lipase (Pnpla2/ATGL), is critical to the adaptive fasting response. Here we discovered an alternative mechanism of lipolysis in adipocytes involving a lysosomal program. We functionally tested lysosomal lipolysis with pharmacological and genetic approaches in mice and in murine and human adipocyte and adipose tissue explant culture, establishing dependency on lysosomal acid lipase (LIPA/LAL) and the microphthalmia/transcription factor E (MiT/TFE) family. Our study establishes a model whereby the canonical pathway is critical for rapid lipolytic responses to adrenergic stimuli operative in the acute stage of fasting, while the alternative lysosomal pathway dominates with prolonged fasting.
    DOI:  https://doi.org/10.1038/s41467-025-56613-3
  16. Neurosci Biobehav Rev. 2025 Feb 03. pii: S0149-7634(25)00040-5. [Epub ahead of print] 106040
      The experience of stress and the need to regulate emotions are pervasive in everyday life. Emotion regulation (ER) is particularly required under stress to facilitate successful adaptation and recovery. Importantly, a growing body of work has identified stress and ER deficits as transdiagnostic risk factors for psychopathology. This highlights the relevance of understanding how stress impacts ER to elucidate individual vulnerability to mental disorders. Stress alters cognitive and emotional functioning via stress hormones secreted by the two major stress systems: sympathetic nervous system and hypothalamus-pituitary adrenocortical axis. This review aims to compile and synthesize empirical studies in humans investigating the effects of acute stress and stress hormones on ER. A systematic literature search yielded 14 relevant studies, 11 investigating acute stress effects and 3 examining the influence of pharmacological cortisol elevations on ER. The results of the stress studies are mixed revealing either impairing, beneficial or no effects at all. Cortisol administration mostly facilitated ER attempts. Notably, we detected timing differences in measuring ER performance relative to stress exposure that potentially reconcile divergent findings. Here, we propose the PRESSURE-model (Predominant Stress System Underpins Regulation of Emotions) postulating that the direction and magnitude of stress effects on ER depends on the relative predominance of one stress system over the other. Additionally, sex-stress hormone interactions, stimulus intensity and ER strategy are discussed as possible moderators. Finally, we highlight limitations in current research and provide recommendations for future studies that will further advance our understanding of the intricate relationship between stress and ER.
    Keywords:  catecholamines; cognitive reappraisal; distraction; emotion control; glucocorticoids; stress; timing
    DOI:  https://doi.org/10.1016/j.neubiorev.2025.106040
  17. Cell Rep. 2025 Jan 30. pii: S2211-1247(25)00024-5. [Epub ahead of print]44(2): 115253
      Acute-stress-induced anxiety helps animals avoid danger, but the neural and molecular mechanisms controlling this behavior remain largely elusive. Here, we find that acute physical stress activates many neurons in the primary somatosensory cortex, trunk region (S1Tr). Single-cell sequencing reveals that the S1Tr c-fos-positive neurons activated by acute stress are largely GABAergic somatostatin (Sst) neurons. These S1TrSst neurons desensitize during subsequent anxiety-like behavior tests. Inhibiting or inducing apoptosis of S1TrSst neurons mimics acute-stress effects and induces anxiety, while activating these neurons reduces acute-stress-induced anxiety. S1TrSst cells receive inputs from secondary auditory cortex, dorsal area (AUD) GABAergic neurons to modulate this anxiety. Spatial transcriptome sequencing and targeted Pde4b protein knockdown show that acute stress reduces Pde4b-regulated cAMP signaling in AUDGABA-S1TrSst projections, leading to decreased S1TrSst neuron activity in subsequent behavioral tests. Our study reports a neural and molecular mechanism for acute-stress-induced anxiety, providing a basis for treating anxiety disorders.
    Keywords:  CP: Cell biology; CP: Neuroscience; acute stress; anxiety; cAMP signaling pathway; primary somatosensory cortex; somatostatin
    DOI:  https://doi.org/10.1016/j.celrep.2025.115253
  18. J Cachexia Sarcopenia Muscle. 2025 Feb;16(1): e13714
       BACKGROUND: Fat mass (FM) and fat-free mass (FFM) are body composition estimates commonly reported in research studies and clinical settings. Recently, fat-free mass indexed to height (fat-free mass index; FFMI) has been shown to be positively associated with impaired insulin sensitivity or insulin resistance. Consequently, hypertrophic resistance training which can increase FFM was also questioned. This paper sets out to evaluate these propositions.
    METHODS: In this narrative review, we discuss possible reasons that link FFMI to adverse metabolic health outcomes including the limitations of the body composition model that utilizes FFM. The safety of resistance training is also briefly discussed.
    RESULTS: Approximately 50% of FFM is comprised of skeletal muscle (SM), with the other 50% being viscera, skin, and bone; FFM and SM cannot be conflated. FFM and fat mass (FM) can both rise with increasing body weight and adiposity, indicating a positive correlation between the two compartments. Risk assessment models not adequately adjusting for this correlation may cause erroneous conclusions, however which way FM and FFM are indexed. Adipose tissue accumulation with weight gain, measured by dual-energy X-ray absorptiometry or bioelectrical impedance, can inflate FFM estimates owing to increased connective tissue. Increased adiposity can also result in fat deposition within skeletal muscle disrupting metabolic health. Importantly, non-skeletal muscle components of the FFM, i.e., the liver and pancreas, both critical in metabolic health, can also be negatively affected by the same lifestyle factors that impact SM. The most frequently used body composition techniques used to estimate FM and FFM cannot detect muscle, liver or pancreas fat infiltration. Prospective evidence demonstrates that resistance training is a safe and effective exercise modality across all ages, especially in older adults experiencing age- or disease-related declines in muscle health.
    CONCLUSIONS: The association between FFM and insulin resistance is largely an artefact driven by inadequate assessment of skeletal muscle. If FM and FFM are used, at the minimum, they need to be evaluated in context with one another. Body composition methods, such as magnetic resonance imaging, which measures skeletal muscle rather than fat-free mass, and adipose tissue as well as muscle ectopic fat, are preferred methods. Resistance training is important in achieving and maintaining good health across the lifespan. While strength and power are critical components of resistance training, the reduction of skeletal mass through ageing or disease may require hypertrophic training to mitigate and slow down the progression of this often-inevitable process.
    Keywords:  ectopic fat; fat‐free mass index; metabolic health; resistance training; skeletal muscle
    DOI:  https://doi.org/10.1002/jcsm.13714
  19. Science. 2025 Feb 07. 387(6734): 674-682
      Insulin resistance is a hallmark of obesity-associated type 2 diabetes. Insulin's actions go beyond metabolic cells and also involve blood vessels, where insulin increases capillary blood flow and delivery of insulin and nutrients. We show that adrenomedullin, whose plasma levels are increased in obese humans and mice, inhibited insulin signaling in human endothelial cells through protein-tyrosine phosphatase 1B-mediated dephosphorylation of the insulin receptor. In obese mice lacking the endothelial adrenomedullin receptor, insulin-induced endothelial nitric oxide-synthase activation and skeletal muscle perfusion were increased. Treating mice with adrenomedullin mimicked the effect of obesity and induced endothelial and systemic insulin resistance. Endothelial loss or blockade of the adrenomedullin receptor improved obesity-induced insulin resistance. These findings identify a mechanism underlying obesity-induced systemic insulin resistance and suggest approaches to treat obesity-associated type 2 diabetes.
    DOI:  https://doi.org/10.1126/science.adr4731
  20. Diabetes. 2025 Feb 03. pii: db240360. [Epub ahead of print]
      Glucolipotoxicity, caused by combined hyperglycemia and hyperlipidemia, results in β-cell failure and type 2 diabetes via cellular stress-related mechanisms. Activating transcription factor 4 (Atf4) is an essential effector of stress response. We show here that Atf4 expression in β-cells is minimally required for glucose homeostasis in juvenile and adolescent mice but it is needed for β-cell function during aging and under obesity-related metabolic stress. Henceforth, Atf4-deficient β-cells older than 2 months after birth display compromised secretory function under acute hyperglycemia. In contrast, they are resistant to acute free fatty acid-induced dysfunction and reduced production of several factors essential for β-cell identity. Atf4-deficient β-cells down-regulate genes involved in protein translation. They also upregulate several lipid metabolism or signaling genes, likely contributing to their resistance to free fatty acid-induced dysfunction. These results suggest that Atf4 activation is required for β-cell identity and function under high glucose. But Atf4 activation paradoxically induces β-cell failure in high levels of free fatty acids. Different transcriptional targets of Atf4 could be manipulated to protect β-cells from metabolic stress-induced failure.
    DOI:  https://doi.org/10.2337/db24-0360