Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (358)

Search Parameters:
Keywords = glucocorticoid signalling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 1257 KB  
Review
Connective Tissue Disease-Associated Pulmonary Arterial Hypertension: Current Therapeutic Strategies and Future Prospects
by Yukina Mizuno Yokoyama, Ryu Watanabe, Tomohiro Yamaguchi, Ryuhei Ishihara, Mayu Shiomi, Yuya Fujita, Masao Katsushima, Kazuo Fukumoto, Yoichiro Haji, Shinsuke Yamada and Motomu Hashimoto
Biomolecules 2026, 16(1), 140; https://doi.org/10.3390/biom16010140 - 13 Jan 2026
Viewed by 169
Abstract
Connective tissue disease-associated pulmonary arterial hypertension (CTD-PAH) is a severe form of pulmonary hypertension with poor prognosis. It most commonly arises in systemic sclerosis (SSc), followed by systemic lupus erythematosus (SLE) and mixed connective tissue disease (MCTD). Its pathogenesis involves a complex interplay [...] Read more.
Connective tissue disease-associated pulmonary arterial hypertension (CTD-PAH) is a severe form of pulmonary hypertension with poor prognosis. It most commonly arises in systemic sclerosis (SSc), followed by systemic lupus erythematosus (SLE) and mixed connective tissue disease (MCTD). Its pathogenesis involves a complex interplay of immune dysregulation, chronic inflammation, endothelial injury, vascular remodeling, and fibrosis. Although vasodilators targeting the endothelin, nitric oxide, and prostacyclin pathways remain the therapeutic backbone, newer agents—including the activin signal inhibitor sotatercept and inhaled treprostinil—have expanded treatment options. Immune-targeted therapies such as glucocorticoids, cyclophosphamide, mycophenolate mofetil, rituximab, and IL-6 receptor inhibitors may benefit inflammation-dominant PAH phenotypes, while fibrotic phenotypes continue to demonstrate limited responsiveness. In addition to brain natriuretic peptide (BNP), N-terminal (NT)-proBNP and disease-specific autoantibodies, emerging biomarkers show promise for early detection, risk stratification, and personalized treatment, though validation in CTD-PAH is lacking. Advances in animal models replicating immune-mediated vascular injury and fibrosis have further improved mechanistic understanding. Despite these developments, substantial unmet needs remain, including the absence of disease-specific therapeutic strategies, limited biomarker integration into clinical practice, and a scarcity of large, well-designed trials targeting individual CTD subtypes. Addressing these gaps will be essential for improving prognosis in patients with CTD-PAH. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

24 pages, 2061 KB  
Review
The Impact of Chronic Stress on Treatment Outcomes of Cancer Patients with Divergent Survival Rates: A Systematic Review
by Katarzyna Herbetko, Justyna Kaczor, Adam Sołtyk, Monika Kisielewska, Marcel Opęchowski, Aleksandra Sztuder and Julita Kulbacka
Int. J. Mol. Sci. 2026, 27(2), 686; https://doi.org/10.3390/ijms27020686 - 9 Jan 2026
Viewed by 184
Abstract
This systematic review investigates the impact of chronic stress on treatment outcomes among cancer patients with divergent survival rates, focusing on breast, prostate, pancreatic, and ovarian cancers. The analysis explores how chronic stress influences molecular pathways and tumor progression while comparing cancers with [...] Read more.
This systematic review investigates the impact of chronic stress on treatment outcomes among cancer patients with divergent survival rates, focusing on breast, prostate, pancreatic, and ovarian cancers. The analysis explores how chronic stress influences molecular pathways and tumor progression while comparing cancers with five-year survival rates above and below 50%. A comprehensive literature search was conducted in PubMed and Scopus for studies published between 2014 and 2025 using combinations of keywords related to “chronic stress,” “psychological stress,” “psychotherapy,” and selected cancer types. All studies met the inclusion criteria according to the PRISMA 2020 guidelines. Evidence suggests that chronic stress is associated with the activation of neuroendocrine and immune mechanisms, including β-adrenergic and glucocorticoid signaling. These multifactorial processes are associated with disease progression and survival, particularly in pancreatic and ovarian cancers; however, these links remain primarily associative rather than causative. Conversely, psychotherapeutic interventions alleviate stress-related biological responses, improve quality of life, and may indirectly enhance therapeutic efficacy. By structuring the evidence around cancers with higher versus lower five-year survival, our review provides a survival informed synthesis of cancer type specific stress biology and stress-mitigating interventions, highlighting potentially targetable pathways and clear evidence gaps for future trials. The findings underscore the need to integrate psychological care into oncological practice to improve overall outcomes. Full article
Show Figures

Figure 1

37 pages, 2753 KB  
Review
Osteoporosis After Menopause and After Drug Therapy: The Molecular Mechanism of Bone Loss and Its Treatment
by Kelly I-Rong Lee, Jie-Hong Chen and Kuo-Hu Chen
Int. J. Mol. Sci. 2026, 27(2), 641; https://doi.org/10.3390/ijms27020641 - 8 Jan 2026
Viewed by 432
Abstract
Osteoporosis is a prevalent skeletal disorder characterized by reduced bone mass and microarchitectural deterioration, leading to increased fracture risk, particularly in aging populations. Postmenopausal osteoporosis (PMOP) remains the most common primary form and results from abrupt estrogen deficiency after menopause, which disrupts bone [...] Read more.
Osteoporosis is a prevalent skeletal disorder characterized by reduced bone mass and microarchitectural deterioration, leading to increased fracture risk, particularly in aging populations. Postmenopausal osteoporosis (PMOP) remains the most common primary form and results from abrupt estrogen deficiency after menopause, which disrupts bone remodeling by accelerating the receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis, suppressing Wnt/β-catenin signaling, and promoting inflammatory cytokine production. In contrast, drug-induced osteoporosis (DIOP) encompasses a heterogeneous group of secondary bone disorders arising from pharmacologic exposures. Glucocorticoids suppress osteoblastogenesis, enhance osteoclast activity, and increase reactive oxygen species; long-term bisphosphonate therapy may oversuppress bone turnover, resulting in microdamage accumulation; denosumab withdrawal triggers a unique rebound surge in RANKL activity, often leading to rapid bone loss and multiple vertebral fractures. Medications including aromatase inhibitors, SSRIs, proton pump inhibitors, heparin, and antiepileptic drugs impair bone quality through diverse mechanisms. Standard antiresorptive agents remain first-line therapies, while anabolic agents such as teriparatide, abaloparatide, and romosozumab provide enhanced benefits in high-risk or drug-suppressed bone states. Transitional bisphosphonate therapy is essential when discontinuing denosumab, and individualized treatment plans—including drug holidays, lifestyle interventions, and monitoring vulnerable patients—are critical for optimizing outcomes. Emerging approaches such as small interfering RNA (siRNA)-based therapeutics, anti-sclerostin agents, digital monitoring technologies, and regenerative strategies show promise for future precision medicine management. Understanding the distinct and overlapping molecular mechanisms of osteoporosis is essential for improving fracture prevention and long-term skeletal health. Full article
(This article belongs to the Special Issue Osteoporosis: From Molecular Research to Novel Therapies)
Show Figures

Figure 1

25 pages, 1317 KB  
Review
Hormonal and Behavioral Consequences of Social Isolation and Loneliness: Neuroendocrine Mechanisms and Clinical Implications
by Volodymyr Mavrych, Ghaith K. Mansour, Ahmad W. Hajjar and Olena Bolgova
Int. J. Mol. Sci. 2026, 27(1), 84; https://doi.org/10.3390/ijms27010084 - 21 Dec 2025
Cited by 1 | Viewed by 679
Abstract
Social isolation and loneliness represent critical psychosocial stressors associated with profound hormonal dysregulation and adverse behavioral outcomes. This review synthesizes current evidence on neuroendocrine mechanisms linking perceived and objective social disconnection to health consequences, emphasizing hypothalamic–pituitary–adrenal axis dysfunction, altered glucocorticoid signaling, and inflammatory [...] Read more.
Social isolation and loneliness represent critical psychosocial stressors associated with profound hormonal dysregulation and adverse behavioral outcomes. This review synthesizes current evidence on neuroendocrine mechanisms linking perceived and objective social disconnection to health consequences, emphasizing hypothalamic–pituitary–adrenal axis dysfunction, altered glucocorticoid signaling, and inflammatory pathways. Loneliness activates conserved transcriptional responses with upregulated proinflammatory gene expression and downregulated antiviral responses, mediated through sustained cortisol elevation and glucocorticoid resistance. Neural circuit alterations in reward processing, particularly the ventral tegmental area-nucleus accumbens pathway, contribute to anhedonia, social withdrawal, and cognitive decline. Sex differences in neuroendocrine responses reveal distinct hormonal profiles and circuit-specific adaptations. Emerging interventions targeting oxytocin and arginine vasopressin systems, alongside behavioral approaches addressing loneliness-induced cognitive biases, show promise. Critical research gaps include a mechanistic understanding of epigenetic modifications, sex-specific therapeutic responses, and translational applications across diverse populations. Understanding the endocrine–behavior interface in social disconnection offers opportunities for targeted interventions addressing this growing public health challenge. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

21 pages, 2483 KB  
Article
Glucocorticoid Signaling-Associated Gene Expression in the Hippocampus and Frontal Cortex of Chronically Isolated Normotensive and Hypertensive Rats and the Responsiveness to Acute Restraint Stress
by Alexey Kvichansky, Liya Tretyakova, Yulia Moiseeva, Viktoriia Ovchinnikova, Diana Mamedova, Olga Nedogreeva, Natalia Lazareva, Natalia Gulyaeva and Mikhail Stepanichev
Int. J. Mol. Sci. 2025, 26(24), 12050; https://doi.org/10.3390/ijms262412050 - 15 Dec 2025
Viewed by 457
Abstract
Genotypic characteristics may determine the body’s response to stressful conditions as well as its susceptibility to cardiovascular diseases and stroke. Old age worsens the course of these diseases, and often concomitant hypertension can negatively affect brain function, especially in cases of social isolation. [...] Read more.
Genotypic characteristics may determine the body’s response to stressful conditions as well as its susceptibility to cardiovascular diseases and stroke. Old age worsens the course of these diseases, and often concomitant hypertension can negatively affect brain function, especially in cases of social isolation. In this work, we studied how social isolation and hypertension affect the transcription activity of genes associated with glucocorticoid signaling in the rat brain. The study was performed on 10-month-old rats of the outbred Wistar stock (n = 48) and the inbred spontaneously hypertensive (SHR) strain (n = 28). The animals of each genotype were divided into groups, one of which was kept in home cages in groups of 3–4 individuals, and the other in single cages for 3 months. Physiological parameters and plasma corticosterone were controlled before the start and after 3 months of isolation. Each group was additionally divided into two subgroups: one subjected to 1 h of restraint stress, and changes in blood glucose and corticosterone levels were assessed. At the end, the levels of Nr3c1, Nr3c2, Hsd11b1, and Fkbp5 mRNAs were measured in the hippocampus and frontal cortex using the Q-PCR technique. After isolation, weight gain stopped in SHRs, although blood pressure did not change, and heart rate increased in rats of both genotypes. In response to restraint, there was practically no increase in corticosterone in isolated Wistar rats, whereas in SHRs, there were significant glucose and corticosterone responses. Significant disruptions in the system responsible for corticosterone-activated signaling cascades were found in the brains of SHR rats. The transcriptional activity of genes encoding corticosterone receptors and proteins regulating their action was reduced in the hippocampus and frontal cortex in SHRs compared to Wistar rats. However, neither isolation nor acute stress significantly affected the contents of transcripts studied. Meanwhile, after isolation, the relationships between the expression of these genes changed significantly, in different directions, in rats of the studied genotypes, both within and between brain structures. Thus, the SHR genotype is associated with persistent changes in the brain that affect the expression of glucocorticoid-associated genes. This indicates a more complex regulation of the stress response, not limited only by the feedback system within the hypothalamic–pituitary–adrenocortical or sympatho-adrenomedullary systems, but operated at the level of the limbic system and the cerebral cortex. Full article
(This article belongs to the Special Issue Molecular Crosstalk in Stress-Induced Psychopathology)
Show Figures

Figure 1

17 pages, 1124 KB  
Review
St. John’s Wort for Depression: From Neurotransmitters to Membrane Plasticity
by Verena M. Merk, Georg Boonen and Veronika Butterweck
Int. J. Mol. Sci. 2025, 26(24), 11925; https://doi.org/10.3390/ijms262411925 - 10 Dec 2025
Viewed by 978
Abstract
Depression is a multifactorial disorder shaped by genetic, psychosocial, and biological influences, with hypotheses ranging from monoamine deficiency and neuroplasticity deficits to inflammation and stress-induced dysregulation. St. John’s wort (Hypericum perforatum L.) has long been used as an herbal antidepressant and is [...] Read more.
Depression is a multifactorial disorder shaped by genetic, psychosocial, and biological influences, with hypotheses ranging from monoamine deficiency and neuroplasticity deficits to inflammation and stress-induced dysregulation. St. John’s wort (Hypericum perforatum L.) has long been used as an herbal antidepressant and is supported by clinical evidence for efficacy and safety in mild-to-moderate depression. While its multimodal mechanisms have been linked to neurotransmitter reuptake inhibition, neuroendocrine regulation, and modulation of neuroplasticity, recent findings suggest an additional role at the membrane level. Emerging lipidomic studies highlight that Ze 117, a low-hyperforin H. perforatum extract, counteracts stress- and glucocorticoid-induced increases in membrane fluidity by modulating lipid composition and cholesterol metabolism. These effects normalize receptor mobility and signal transduction, particularly of β1-adrenoceptors, and modulate glycerophospholipid metabolism in both cellular and animal models. Such membrane-stabilizing properties may represent a novel mechanistic pathway complementing classical neurochemical actions. This review revisits the mechanisms of St. John’s wort with a special focus on its impact on membrane lipids, positioning lipidomics as a promising tool for elucidating antidepressant activity. These insights may open avenues toward personalized therapeutic strategies in depression. Full article
(This article belongs to the Special Issue The Role of Lipids in Health and Diseases)
Show Figures

Figure 1

18 pages, 1986 KB  
Article
Myoprotective Role of Quercus acuta Thunb. Fruit Extract Through IGF-1–Akt–FOXO Axis Modulation in Dexamethasone-Induced Sarcopenia
by Da-In Choi, HuiJun Lee, Seokhoon Heo, Ji-Ae Hong, Donghyuk Bae and Chul-Yung Choi
Appl. Sci. 2025, 15(24), 12978; https://doi.org/10.3390/app152412978 - 9 Dec 2025
Viewed by 340
Abstract
Sarcopenia, characterized by the progressive loss of skeletal muscle mass and function, is exacerbated by glucocorticoid exposure. Although there is growing interest in natural therapies for muscle atrophy, the effects of Quercus acuta Thunb. fruit extract (QA) on sarcopenia or glucocorticoid-induced muscle loss [...] Read more.
Sarcopenia, characterized by the progressive loss of skeletal muscle mass and function, is exacerbated by glucocorticoid exposure. Although there is growing interest in natural therapies for muscle atrophy, the effects of Quercus acuta Thunb. fruit extract (QA) on sarcopenia or glucocorticoid-induced muscle loss had not been previously investigated. QA is an evergreen oak known for its antioxidant and anti-inflammatory properties, with polyphenolic components reported to enhance oxidative and metabolic homeostasis in various tissues. Based on these properties, we hypothesized that QA could counteract muscle atrophy by modulating anabolic and catabolic signaling pathways. The research utilized both in vitro (C2C12 myotubes) and in vivo (ICR mice) models to assess QA’s effects. Daily oral administration of QA (100–200 mg/kg) was given to mice with dexamethasone (Dex)-induced muscle atrophy. Techniques included H&E staining to assess muscle mass and fiber cross-sectional area (CSA), Western blot, and ELISA analyses to investigate signaling pathways. Confocal imaging was also used to confirm cellular changes. In vitro QA treatment improved myotube integrity by increasing myogenic differentiation markers (MyoD, MyoG) and suppressing atrophy-related E3 ligases, specifically MuRF-1 and FBX32/Atrogin-1. Confocal imaging showed that QA inhibited the nuclear localization of FOXO1 and reduced FBX32 expression. In vivo, daily oral administration of QA significantly preserved gastrocnemius muscle mass and fiber cross-sectional area in Dex-treated mice. QA restored the IGF-1/PI3K/Akt signaling pathway and attenuated FOXO1-dependent proteolytic activation. Collectively, these findings demonstrate that QA possesses potent anti-atrophic and myoprotective effects mediated through the modulation of the IGF-1/Akt-FOXO axis. QA has potential as a novel natural therapeutic for preventing glucocorticoid-induced sarcopenia. Full article
Show Figures

Figure 1

30 pages, 1519 KB  
Review
Chronic Stress and Astrocyte Dysfunction in Depression: Molecular Mechanisms and Gene Expression Changes
by Natalia Bochenska, Julia Tomczak and Malwina Lisek
Antioxidants 2025, 14(12), 1464; https://doi.org/10.3390/antiox14121464 - 6 Dec 2025
Viewed by 1084
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous psychiatric condition with high global prevalence and significant personal and societal burdens. While traditionally focused on neuronal dysfunction, emerging research highlights a critical role for astrocytes—glial cells essential for maintaining brain homeostasis in the [...] Read more.
Major depressive disorder (MDD) is a complex and heterogeneous psychiatric condition with high global prevalence and significant personal and societal burdens. While traditionally focused on neuronal dysfunction, emerging research highlights a critical role for astrocytes—glial cells essential for maintaining brain homeostasis in the pathogenesis of depression. This review explores how chronic stress, a major risk factor for MDD, disrupts astrocyte function through multiple converging mechanisms. We detail the normal physiological roles of astrocytes in synaptic regulation, neurotransmitter cycling, metabolic support, and neurovascular integrity, and examine how these functions are compromised under chronic stress. Key molecular pathways implicated include glucocorticoid receptor (GR) signaling dysregulation, neuroinflammatory responses, glutamate excitotoxicity, oxidative stress, and epigenetic alterations. Evidence from histological and transcriptomic studies in both human postmortem tissue and rodent models reveals consistent changes in astrocyte-specific genes, such as GFAP, SLC1A2, SLC1A3, BDNF, and AQP4, supporting their involvement in depressive pathology. Finally, we discuss therapeutic strategies targeting astrocyte dysfunction—including EAAT2 upregulation, neuromodulation, anti-inflammatory approaches, GR modulation, and glial-focused epigenetic therapies. Understanding astrocyte pathology in the context of chronic stress not only refines our understanding of MDD but also opens novel avenues for treatment development. Full article
Show Figures

Figure 1

13 pages, 1973 KB  
Article
Non-Genomic Cortisol Signaling Regulates Early Myogenic Gene Expression in Rainbow Trout Skeletal Muscle
by Consuelo Figueroa, Rodrigo Zuloaga, Giorgia Daniela Ugarte, Phillip Dettleff, Jorge Eduardo Aedo, Alfredo Molina and Juan Antonio Valdés
Fishes 2025, 10(12), 621; https://doi.org/10.3390/fishes10120621 - 4 Dec 2025
Viewed by 369
Abstract
Glucocorticoids are key regulators of vertebrate physiology, orchestrating metabolic, immune, and developmental processes that enable adaptation to stress. In teleosts, cortisol is the primary glucocorticoid, acting through classical genomic pathways and rapid non-genomic mechanisms. Although genomic signaling has been widely characterized, non-genomic actions [...] Read more.
Glucocorticoids are key regulators of vertebrate physiology, orchestrating metabolic, immune, and developmental processes that enable adaptation to stress. In teleosts, cortisol is the primary glucocorticoid, acting through classical genomic pathways and rapid non-genomic mechanisms. Although genomic signaling has been widely characterized, non-genomic actions remain poorly understood in skeletal muscle, a tissue of both biological and economic importance. In this study, we examined the effects of cortisol and its membrane-impermeable analog, cortisol-BSA, on rainbow trout (Oncorhynchus mykiss) skeletal muscle under in vivo and in vitro conditions. Transcript analysis demonstrated that cortisol and cortisol-BSA rapidly induced pax3 (2.28 ± 0.22- and 2.48 ± 0.45-fold change, respectively) and myf5 expression (3.03 ± 0.47- and 2.31 ± 0.29-fold change, respectively) at 1 h, whereas prolonged cortisol and cortisol-BSA exposure resulted in their downregulation (0.34 ± 0.07- and 0.38 ± 0.14-fold change, respectively). In cultured myotubes, cortisol-BSA activated protein kinase A (PKA) (2.24 ± 0.25-fold change) and enhanced phosphorylation of its downstream target CREB (3.2 ± 0.21-fold change) in a time-dependent manner; these effects were abolished by the PKA inhibitor H89. Moreover, inhibition of PKA signaling suppressed cortisol-BSA–induced pax3 and myf5 expression (1.31 ± 0.28-fold change and 1.89 ± 0.28-fold change, respectively). Together, these findings provide the first mechanistic evidence that non-genomic cortisol signaling regulates the PKA–CREB axis in fish skeletal muscle, promoting the early transcriptional activation of promyogenic factors. This work underscores the complementary role of rapid cortisol actions in fine-tuning myogenic responses under acute stress, offering new perspectives on muscle plasticity in teleosts. Full article
(This article belongs to the Special Issue Genomics Applied to Fish Health)
Show Figures

Graphical abstract

23 pages, 2577 KB  
Review
Molecular Pathogenesis and Targeted Therapies in Eosinophilic Granulomatosis with Polyangiitis: An Updated Review
by María López Paraja, Grisell Starita Fajardo, Ignacio Donate Velasco, David Lucena López, María Pilar Iranzo Alcolea, Francisco José Lirola Sánchez, Mercedes Peña Rodriguez, Andrés González García and Luis Manzano Espinosa
Int. J. Mol. Sci. 2025, 26(22), 11141; https://doi.org/10.3390/ijms262211141 - 18 Nov 2025
Viewed by 1137
Abstract
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare systemic vasculitis characterized by asthma, eosinophilia, and necrotizing inflammation of small- to medium-sized vessels. Accumulating evidence indicates that EGPA is a polygenic and heterogeneous disorder comprising distinct antineutrophil cytoplasmic antibody (ANCA)–defined endotypes with divergent genetic [...] Read more.
Eosinophilic granulomatosis with polyangiitis (EGPA) is a rare systemic vasculitis characterized by asthma, eosinophilia, and necrotizing inflammation of small- to medium-sized vessels. Accumulating evidence indicates that EGPA is a polygenic and heterogeneous disorder comprising distinct antineutrophil cytoplasmic antibody (ANCA)–defined endotypes with divergent genetic backgrounds, immune pathways, and clinical phenotypes. Its pathogenesis reflects the convergence of epithelial–alarmin signaling, type 2 inflammation, eosinophil effector mechanisms, and B-cell/autoantibody responses, with myeloperoxidase (MPO)-ANCA serving as a hallmark of the vasculitic subset. Recent advances in genomics, immunology, and multi-omics profiling have uncovered biomarkers and molecular circuits sustaining disease activity and guiding therapeutic stratification. The identification of the interleukin (IL)-5–eosinophil axis, epithelial-derived alarmins, and B-cell/IgG4 networks as central pathogenic nodes has enabled the development of targeted biologic therapies that are redefining treatment paradigms. Benralizumab (anti-IL-5Rα) has recently been approved for EGPA following the phase 3 head-to-head MANDARA trial, which demonstrated non-inferiority to mepolizumab in achieving remission (BVAS = 0 with ≤4 mg/day prednisone equivalent) at weeks 36 and 48. These results, together with the established efficacy of mepolizumab, inform practical selection between IL-5 and IL-5Rα blockade and support glucocorticoid-sparing approaches. A structured literature search (2015–2025) was conducted in PubMed, Scopus, and Web of Science to identify recent advances in epidemiology, genetics, biomarkers, and targeted therapies for EGPA. This updated review integrates molecular insights, clinical endotypes, and therapeutic innovations to outline current evidence and future precision-medicine strategies aimed at improving long-term patient outcomes. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutic Targets in Systemic Vasculitis)
Show Figures

Figure 1

15 pages, 936 KB  
Review
Anti-Cytokine Drugs in the Treatment of Canine Atopic Dermatitis
by Agnieszka Wichtowska and Małgorzata Olejnik
Int. J. Mol. Sci. 2025, 26(22), 10990; https://doi.org/10.3390/ijms262210990 - 13 Nov 2025
Viewed by 3702
Abstract
Canine atopic dermatitis (cAD) is a chronic, pruritic, inflammatory skin disease with complex immunopathogenesis involving dysregulated cytokine networks. In recent years, targeted therapies have transformed the management of cAD by directly or indirectly modulating cytokine activity. Lokivetmab, a monoclonal antibody neutralizing interleukin-31, represents [...] Read more.
Canine atopic dermatitis (cAD) is a chronic, pruritic, inflammatory skin disease with complex immunopathogenesis involving dysregulated cytokine networks. In recent years, targeted therapies have transformed the management of cAD by directly or indirectly modulating cytokine activity. Lokivetmab, a monoclonal antibody neutralizing interleukin-31, represents a breakthrough in veterinary dermatology, providing rapid and sustained reduction in pruritus with a favorable safety profile. Janus kinase inhibitors, including oclacitinib and the newer ilunocitinib, act downstream by blocking cytokine signal transduction, offering effective control of both acute and chronic phases of disease. Ciclosporin, a calcineurin inhibitor, remains a valuable immunosuppressant for long-term cAD management, while topical tacrolimus provides localized benefits. Together, these therapies mark a paradigm shift from non-specific immunosuppressants to precision medicine. In this context, precision medicine refers to therapeutic strategies that selectively target key cytokines or intracellular signaling pathways central to the pathogenesis of cAD, such as IL-31 or the JAK/STAT axis. Unlike traditional immunosuppressants such as glucocorticoids, which exert broad and non-selective immune suppression, these agents modulate defined molecular mechanisms, thereby improving efficacy and minimizing adverse effects. Consequently, they enable improved quality of life for affected dogs and their owners. Future strategies will likely focus on patient stratification and personalized approaches based on immunological endotypes. Full article
(This article belongs to the Special Issue Cytokines and Other Biomarkers of Health Status)
Show Figures

Figure 1

19 pages, 6662 KB  
Article
Stigmasterol Protects Against Dexamethasone-Induced Muscle Atrophy by Modulating the FoxO3–MuRF1/MAFbx Signaling Pathway in C2C12 Myotubes and Mouse Skeletal Muscle
by Young-Sool Hah, Seung-Jun Lee, Yeung-Ho Ji, Jeongyun Hwang, Han-Gil Kim, Young-Tae Ju, Jun-Il Yoo and Seung-Jin Kwag
Biomolecules 2025, 15(11), 1551; https://doi.org/10.3390/biom15111551 - 5 Nov 2025
Viewed by 929
Abstract
Glucocorticoid therapy, using agents like dexamethasone (Dexa), often leads to muscle atrophy by increasing protein degradation via the ubiquitin–proteasome system while suppressing protein synthesis. Stigmasterol, a phytosterol with known bioactivities, has an unexplored role in muscle atrophy. This study investigated stigmasterol’s protective effects [...] Read more.
Glucocorticoid therapy, using agents like dexamethasone (Dexa), often leads to muscle atrophy by increasing protein degradation via the ubiquitin–proteasome system while suppressing protein synthesis. Stigmasterol, a phytosterol with known bioactivities, has an unexplored role in muscle atrophy. This study investigated stigmasterol’s protective effects against Dexa-induced muscle atrophy and its impact on the FoxO3 and mTORC1 signaling pathways. Differentiated C2C12 myotubes were treated with Dexa (50 µM) ± stigmasterol (10 µM), and the morphology, viability, and protein levels in the FoxO3/MuRF1/MAFbx catabolic and mTOR/p70S6K/4E-BP1 anabolic signaling pathways were assessed. C57BL/6 mice received Dexa (20 mg/kg/day i.p.) ± stigmasterol (3 mg/kg/day oral) for 21 days, and the body/muscle mass, bone mineral density (BMD), fiber cross-sectional area (CSA), and muscle protein expression were measured. Stigmasterol (10 µM) was non-toxic and attenuated Dexa-induced reductions in myotube diameter and fusion in vitro, concurrent with suppressing Dexa-induced upregulation of FoxO3/MuRF1/MAFbx proteins and preventing the Dexa-induced dephosphorylation of mTOR/p70S6K/4E-BP1 proteins. In vivo, stigmasterol mitigated Dexa-induced losses in body weight, muscle mass, BMD, and fiber CSA. This protection was associated with attenuated upregulation of FoxO3 and MAFbx proteins in muscle tissue. Stigmasterol protected against Dexa-induced muscle atrophy in vitro and in vivo via modulation of the FoxO3–MAFbx catabolic pathway. These findings suggest stigmasterol inhibits excessive glucocorticoid-induced muscle protein breakdown. It therefore warrants further investigation as a potential therapeutic agent for glucocorticoid myopathy. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

16 pages, 1426 KB  
Systematic Review
Long Non-Coding RNAs in Multiple Sclerosis—Differential Expression and Functional Implications
by Kaalindi Misra, Aishwary Nerkar, Ferdinando Clarelli, Melissa Sorosina and Federica Esposito
Genes 2025, 16(11), 1327; https://doi.org/10.3390/genes16111327 - 3 Nov 2025
Viewed by 709
Abstract
Background/Objectives: Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators of immune pathways and may hold diagnostic and therapeutic relevance in autoimmune diseases such as Multiple Sclerosis (MS). However, research on lncRNAs in MS remains fragmented and geographically clustered. This systematic review [...] Read more.
Background/Objectives: Long non-coding RNAs (lncRNAs) are increasingly recognized as key regulators of immune pathways and may hold diagnostic and therapeutic relevance in autoimmune diseases such as Multiple Sclerosis (MS). However, research on lncRNAs in MS remains fragmented and geographically clustered. This systematic review aimed to collate and critically evaluate studies of lncRNA expression in MS, assess consistency of findings across studies, and synthesize proposed functional implications of the most frequently studied lncRNAs. Methods: This PROSPERO-registered review (CRD420250575938), conducted in accordance with PRISMA, searched PubMed, Scopus, Embase, and Web of Science (2010–2024) for studies evaluating lncRNA expression in adult MS (≥18 years of age). Eligible studies included ≥20 participants and assessed lncRNAs in blood, PBMCs, serum, plasma, or CSF using qRT-PCR, RNA-seq, or microarrays. Pediatric, review, animal, and in vitro studies were excluded. Two reviewers independently screened and extracted data, with risk of bias evaluated using QUADAS-2. Results: Narrative synthesis of 51 studies identified 77 unique lncRNAs. A limited set (MALAT1, GAS5, MEG3, H19) demonstrated consistent dysregulation in MS, whereas others (THRIL, IFNG-AS1, HOTAIR, TUG1) exhibited context-dependent expression influenced by treatment, relapse status, or demographics. Functional annotations converged on immune pathways, including NF-κB, STAT3, IFN-γ/Th1, and glucocorticoid signaling. Conclusions: This review identifies reproducible and context-specific lncRNA dysregulation in MS, emphasizing the need for transcriptome-wide approaches, standardized methods, and multi-center validation. Current evidence is constrained by geographic clustering, preselection bias, and methodological heterogeneity. Full article
Show Figures

Figure 1

16 pages, 1167 KB  
Systematic Review
Bone Quality Beyond DXA in People Living with HIV: A Systematic Review of HR-pQCT, TBS, Microindentation, and Vertebral Fractures
by David Vladut Razvan, Ovidiu Rosca, Felix Bratosin, Vlad Predescu, Silviu Valentin Vlad and Adrian Vlad
J. Clin. Med. 2025, 14(21), 7669; https://doi.org/10.3390/jcm14217669 - 29 Oct 2025
Cited by 1 | Viewed by 697
Abstract
Background and Objectives: People living with HIV (PLWH) have excess fragility fractures not fully explained by areal DXA. We reviewed bone “quality” in PLWH—microarchitecture, estimated strength, tissue-level properties—and vertebral fractures (VFs). Methods: PRISMA-conform systematic review (2000–2025) of randomized, cohort, and cross-sectional [...] Read more.
Background and Objectives: People living with HIV (PLWH) have excess fragility fractures not fully explained by areal DXA. We reviewed bone “quality” in PLWH—microarchitecture, estimated strength, tissue-level properties—and vertebral fractures (VFs). Methods: PRISMA-conform systematic review (2000–2025) of randomized, cohort, and cross-sectional studies assessing HR-pQCT (±finite-element analysis), trabecular bone score (TBS), impact microindentation (BMSi), femoral QCT/MRI, and VF imaging (DXA-VFA or radiography). Risk of bias used ROBINS-I (non-randomized) and RoB 2 (randomized/switch). No meta-analysis was performed due to clinical/methodological heterogeneity; evidence was synthesized narratively per SWiM. Results: Fourteen studies met criteria. HR-pQCT showed cortical/trabecular deficits with lower finite-element–estimated strength in PLWH. BMSi was 3–4 units lower; it declined after ART initiation but improved after TDF→TAF switch. TBS was modestly lower and reclassified risk when BMD was non-osteoporotic. VF prevalence was 12–25% and frequently occurred at non-osteoporotic BMD. Signals aligned with modifiable risks (smoking, glucocorticoids) and specific ART exposures. Conclusions: Beyond DXA, PLWH exhibit quantifiable decrements in microarchitecture, estimated strength, and tissue-level properties alongside a meaningful VF burden. TBS and VFA are pragmatic, scalable adjuncts to refine risk; HR-pQCT/BMSi add mechanistic value in research/tertiary settings. Prospective studies linking these metrics to incident fractures are warranted. Full article
(This article belongs to the Section Orthopedics)
Show Figures

Figure 1

21 pages, 28334 KB  
Article
Irisin Increases Sirtuin 1 to Improve Glucocorticoid-Induced Sarcopenia and Mitochondrial Dysfunction
by Hongwei Shi, Wen Sun, Xiaoyuan Cao, Xuepeng Fan, Wenjuan Xie, Xiaojing Hao, Simiao Wang, Jiayin Lu, Yi Yan, Xiaomao Luo, Yanjun Dong, Haidong Wang and Juan Wang
Cells 2025, 14(21), 1675; https://doi.org/10.3390/cells14211675 - 27 Oct 2025
Cited by 1 | Viewed by 1004
Abstract
Sarcopenia, characterized by progressive skeletal muscle mass, strength, and functional loss, imposes a substantial global health burden. Irisin, a myokine derived from fibronectin type III domain-containing protein 5 (FNDC5), is critical for muscle health. Here, we investigate its role in mitigating glucocorticoid-induced sarcopenia [...] Read more.
Sarcopenia, characterized by progressive skeletal muscle mass, strength, and functional loss, imposes a substantial global health burden. Irisin, a myokine derived from fibronectin type III domain-containing protein 5 (FNDC5), is critical for muscle health. Here, we investigate its role in mitigating glucocorticoid-induced sarcopenia using a mouse and C2C12 myotubes model. We quantified FNDC5/irisin levels in skeletal muscle and plasma and assessed muscle function (body weight, grip strength, wire-hanging, and locomotor activity), histology, and mitochondrial features following irisin administration to dexamethasone-treated mice. Western blot analyzed synthesis/hydrolysis regulators, apoptosis markers, and mitochondrial regulators in mouse muscle tissues and C2C12 myotubes. The results show that FNDC5/irisin was significantly downregulated in sarcopenic mice and atrophic C2C12 myotubes; exogenous irisin rescued muscle mass loss and functional impairment, improving body weight, muscle mass, grip strength, and mobility. Mechanistically, irisin bound SIRT1 with −12.7 kcal/mol affinity, activating a deacetylation cascade that suppressed FoxO3a transcriptional activity (attenuating proteasomal degradation) and enhanced mTORC1-mediated protein synthesis in C2C12 myotubes. Additionally, irisin potentiated PGC-1α signaling in mouse myocytes, promoting mitochondrial biogenesis and restoring contractile function in dystrophic fibers. Collectively, these findings demonstrate irisin alleviates glucocorticoid-induced muscle atrophy via SIRT1-dependent pathways, rebalancing muscle physiology and systemic energy homeostasis. This highlights irisin-based therapeutics as a promising exercise surrogate for sarcopenia management, offering novel clinical avenues. Full article
Show Figures

Figure 1

Back to TopTop