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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,532)

Search Parameters:
Keywords = muscle phenotypes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
8 pages, 901 KB  
Case Report
Beyond Neurodevelopmental Delay: BICRA-Related Coffin–Siris Syndrome 12 with Severe Intestinal Dysmotility and Recurrent Pneumothorax
by Hua Wang
Genes 2026, 17(1), 81; https://doi.org/10.3390/genes17010081 - 11 Jan 2026
Abstract
Background: Coffin–Siris syndrome 12 (CSS12) is a recently described neurodevelopmental disorder caused by heterozygous pathogenic variants in BICRA, a gene encoding a core subunit of the non-canonical BAF (ncBAF) chromatin-remodeling complex. The condition is characterized by developmental delay, hypotonia, hypertrichosis, and joint [...] Read more.
Background: Coffin–Siris syndrome 12 (CSS12) is a recently described neurodevelopmental disorder caused by heterozygous pathogenic variants in BICRA, a gene encoding a core subunit of the non-canonical BAF (ncBAF) chromatin-remodeling complex. The condition is characterized by developmental delay, hypotonia, hypertrichosis, and joint laxity. However, long-term data remain limited, and systemic manifestations are incompletely defined. Case Description: We report a 22-year-old male with a de novo BICRA frameshift variant, c.2479_2480delinsA (p.Ala827Thrfs*15), previously included in the original cohort reported by Barish et al. Longitudinal follow-up revealed an expanded phenotype extending beyond neurodevelopmental features. Early findings included global developmental delay, growth hormone deficiency, short stature, and joint hypermobility. In adolescence and adulthood, he developed severe intestinal dysmotility requiring total colectomy, recurrent spontaneous pneumothoraces from bilateral apical bullous disease, and portal-vein thrombosis, representing visceral and vascular complications not previously emphasized in BICRA-related disorders. The identified BICRA variant truncates the coiled-coil domain critical for BRD9/BRD4 interaction, consistent with a loss-of-function mechanism. The patient’s systemic features suggest that BICRA haploinsufficiency affects not only neurodevelopmental pathways but also smooth-muscle and connective-tissue integrity. Conclusions: This case expands the phenotypic spectrum of BICRA-related CSS12, demonstrating that visceral and vascular involvement can occur alongside neurodevelopmental and connective-tissue features. Recognition of these broader manifestations underscores the need for lifelong multidisciplinary surveillance and contributes to understanding the diverse biological roles of the ncBAF complex in human development. Full article
(This article belongs to the Section Genetic Diagnosis)
Show Figures

Figure 1

36 pages, 1741 KB  
Review
Extracellular Vesicles as Biological Templates for Next-Generation Drug-Coated Cardiovascular Devices: Cellular Mechanisms of Vascular Healing, Inflammation, and Restenosis
by Rasit Dinc and Nurittin Ardic
Cells 2026, 15(2), 121; https://doi.org/10.3390/cells15020121 - 9 Jan 2026
Viewed by 70
Abstract
While drug-eluting cardiovascular devices, including drug-eluting stents and drug-coated balloons, have significantly reduced restenosis rates, they remain limited by delayed vascular healing, chronic inflammation, and late adverse events. These limitations reflect a fundamental mismatch between current device pharmacology, which relies on nonselective antiproliferative [...] Read more.
While drug-eluting cardiovascular devices, including drug-eluting stents and drug-coated balloons, have significantly reduced restenosis rates, they remain limited by delayed vascular healing, chronic inflammation, and late adverse events. These limitations reflect a fundamental mismatch between current device pharmacology, which relies on nonselective antiproliferative drugs, and the highly coordinated, cell-specific programs that orchestrate vascular repair. Extracellular vesicles (EVs), nanometer-scale membrane-bound particles secreted by virtually all cell types, provide a biologically evolved platform for intercellular communication and cargo delivery. In the cardiovascular system, EVs regulate endothelial regeneration, smooth muscle cell phenotype, extracellular matrix remodeling, and macrophage polarization through precisely orchestrated combinations of miRNA, proteins, and lipids. Here, we synthesize mechanistic insights into EV biogenesis, cargo selection, recruitment, and functional effects in vascular healing and inflammation and translate these into a formal framework for EV-inspired device engineering. We discuss how EV-based or EV-mimetic coatings can be designed to sense the local microenvironment, deliver encoded biological “instruction sets,” and function within ECM-mimetic scaffolds to couple local stent healing with systemic tissue repair. Finally, we outline the manufacturing, regulatory, and clinical trial issues that must be addressed for EV-inspired cardiovascular devices to transition from proof of concept to clinical reality. By shifting the focus from pharmacological suppression to biological regulation of healing, EV-based strategies offer a path to resolve the long-standing tradeoff between restenosis prevention and durable vascular healing. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Cardiac Repair and Regeneration)
23 pages, 1108 KB  
Review
Senescence as a Driver of Smooth Muscle Cell Plasticity and Atherosclerosis: Mechanisms and Therapeutic Opportunities
by Lisa Steegen and Mandy O. J. Grootaert
Cells 2026, 15(2), 114; https://doi.org/10.3390/cells15020114 - 8 Jan 2026
Viewed by 231
Abstract
Cell senescence is increasingly recognized as a key driver of atherosclerosis progression. Senescent smooth muscle cells (SMCs) lose their proliferative capacity and adopt a pro-inflammatory profile, contributing to impaired vessel repair and weakening of the fibrous cap. Moreover, senescence promotes SMC dedifferentiation and [...] Read more.
Cell senescence is increasingly recognized as a key driver of atherosclerosis progression. Senescent smooth muscle cells (SMCs) lose their proliferative capacity and adopt a pro-inflammatory profile, contributing to impaired vessel repair and weakening of the fibrous cap. Moreover, senescence promotes SMC dedifferentiation and phenotypic modulation into unfavorable phenotypes associated with plaque destabilization. In this review, we will discuss how cell senescence is induced in atherosclerotic plaques, how this influences SMC plasticity, and how this impacts plaque stability. We will also evaluate the potential of current and experimental anti-atherosclerotic drugs to target SMC senescence and/or SMC phenotypic modulation. Full article
Show Figures

Figure 1

22 pages, 7811 KB  
Article
BDH1 Mediates Aerobic Exercise-Induced Improvement in Skeletal Muscle Metabolic Remodeling in Type 2 Diabetes Mellitus
by Mingyu Wu, Xiaotong Ma, Wei Dai, Ke Li, Haoyang Gao, Yifan Guo and Weihua Xiao
Biomolecules 2026, 16(1), 115; https://doi.org/10.3390/biom16010115 - 8 Jan 2026
Viewed by 164
Abstract
Background: Type 2 diabetes mellitus (T2DM) is typically characterized by the dysregulation of metabolic remodeling. As a systemic metabolic disease, T2DM can affect the mass and function of skeletal muscle by inducing impaired energy metabolism, mitochondrial dysfunction, and chronic low-grade inflammation. β-Hydroxybutyrate dehydrogenase [...] Read more.
Background: Type 2 diabetes mellitus (T2DM) is typically characterized by the dysregulation of metabolic remodeling. As a systemic metabolic disease, T2DM can affect the mass and function of skeletal muscle by inducing impaired energy metabolism, mitochondrial dysfunction, and chronic low-grade inflammation. β-Hydroxybutyrate dehydrogenase 1 (BDH1) is a rate-limiting enzyme involved in ketone body metabolism, and its activity is down-regulated in various models of diabetic complications. Aerobic exercise (AE) is recognized as an effective intervention to promote energy homeostasis and alleviate metabolic stress. Whether its protective effect on skeletal muscle in T2DM involves the regulatory control of BDH1 expression remains unclear. Methods: Wild-type (WT) and systemic BDH1 knockout (BDH1−/−) male C57BL/6J mice were used to establish the sedentary control (SED) and AE models of T2DM by providing a high-fat diet combined with streptozotocin injection. The indicators related to metabolic remodeling were detected by hematoxylin and eosin staining, immunofluorescence staining, quantitative real-time PCR, and Western blot assays. Results: After 8 weeks of AE, we found that AE improved glycolipid metabolic disorders and mitochondrial quality control in the gastrocnemius muscle of T2DM mice by up-regulating BDH1, thereby alleviating oxidative stress, inflammation, and fibrosis. Compared with the WT mice, the BDH1−/− T2DM mice in the SED group exhibited more severe phenotypic impairment. The metabolic improvement effect of AE was attenuated in the BDH1−/− mice. Conclusions: BDH1 is a key effector enzyme that may mediate the AE-induced improvement in metabolic remodeling in the gastrocnemius muscle of mice with T2DM. Full article
Show Figures

Figure 1

24 pages, 3255 KB  
Review
Molecular Mechanisms Underlying Atherosclerosis and Current Advances in Targeted Therapeutics
by Bo Zhu
Int. J. Mol. Sci. 2026, 27(2), 634; https://doi.org/10.3390/ijms27020634 - 8 Jan 2026
Viewed by 169
Abstract
Atherosclerosis is a chronic, multifactorial vascular disease and the leading global cause of cardiovascular morbidity. Its development reflects interconnected disturbances in lipid metabolism, endothelial function, inflammation, smooth muscle cell (SMC) phenotypic switching, and extracellular matrix remodeling. Genetic predisposition, including monogenic disorders such as [...] Read more.
Atherosclerosis is a chronic, multifactorial vascular disease and the leading global cause of cardiovascular morbidity. Its development reflects interconnected disturbances in lipid metabolism, endothelial function, inflammation, smooth muscle cell (SMC) phenotypic switching, and extracellular matrix remodeling. Genetic predisposition, including monogenic disorders such as familial hypercholesterolemia and polygenic risk variants, modulates disease susceptibility by altering lipid homeostasis as well as inflammatory and thrombotic pathways. Epigenetic regulators and noncoding RNAs, such as histone modifications, microRNAs, and long noncoding RNAs, further shape gene expression and link environmental cues to vascular pathology. Endothelial injury promotes lipoprotein retention and oxidation, triggering monocyte recruitment and macrophage-driven foam cell formation, cytokine secretion, and necrotic core development. Persistent inflammation, macrophage heterogeneity, and SMC plasticity collectively drive plaque growth and destabilization. Emerging insights into immune cell metabolism, intracellular signaling networks, and novel regulatory RNAs are expanding therapeutic possibilities beyond lipid-lowering. Current and evolving treatments include statins, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, anti-inflammatory agents targeting interleukin-1 beta (IL-1β) or NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3), and advanced approaches such as gene editing, siRNA, and nanoparticle-based delivery. Integrating multi-omics, biomarker-guided therapy, and precision medicine promises improved risk stratification and next-generation targeted interventions. This review summarizes recent molecular advances and highlights translational opportunities for enhancing atherosclerosis prevention and treatment. Full article
(This article belongs to the Special Issue Molecular Insights and Therapeutic Advances in Atherosclerosis)
Show Figures

Figure 1

17 pages, 11668 KB  
Article
Can the Spatial Heterogeneity in the Epiligament Explain the Differential Healing Capacities of the ACL and MCL?
by Lyubomir Gaydarski, Boycho Landzhov, Richard Shane Tubbs and Georgi P. Georgiev
J. Clin. Med. 2026, 15(2), 510; https://doi.org/10.3390/jcm15020510 - 8 Jan 2026
Viewed by 154
Abstract
Background: The anterior cruciate ligament (ACL) and medial collateral ligament (MCL) display strikingly different healing behaviors, despite their similar structural roles within the knee. The epiligament (EL)—a vascular and cellular envelope surrounding each ligament—has emerged as a critical determinant of repair capacity. The [...] Read more.
Background: The anterior cruciate ligament (ACL) and medial collateral ligament (MCL) display strikingly different healing behaviors, despite their similar structural roles within the knee. The epiligament (EL)—a vascular and cellular envelope surrounding each ligament—has emerged as a critical determinant of repair capacity. The aim of this study was to perform a region-specific, comparative analysis of EL molecular profiles in the ACL and MCL to elucidate the mechanisms underlying their contrasting reparative outcomes. Methods: Human ACL and MCL specimens were obtained from 12 fresh knee joints. Immunohistochemical labeling for CD34, α-smooth muscle actin (α-SMA), and vascular endothelial growth factor (VEGF) was performed across proximal, mid-substance, and distal EL regions. Quantitative image analysis using IHC Profiler for ImageJ generated semiquantitative (negative, low-positive, positive) distributions, and inter-ligament comparisons were quantified using t-tests (p  <  0.05). Results: Distinct, region-specific EL signatures were identified. The ACL EL exhibited strong proximal α-SMA expression (0% neg/66.8% low+/33.2%+) and notable distal CD34 positivity (0% neg/83.3% low+/16.7%+), while VEGF expression was confined to the mid-substance (≈55% low+/26%+). In contrast, the MCL EL was largely negative for CD34 and VEGF across all regions, showing a homogeneous but functionally oriented α-SMA profile: proximally negative, sparse mid positivity, and high distal low-positive staining (93.4% low+). Differences in proximal and distal CD34 and α-SMA expression between the ACL and MCL were highly significant (p  <  0.0001–0.001), confirming a mechanistic divergence in EL organization. Conclusions: The ACL EL is regionally heterogeneous, vascularly biased, and enriched in contractile α-SMA+ cells, suggesting localized but poorly coordinated reparative potential. In contrast, the MCL EL is structurally uniform, with distributed α-SMA activity supporting stable wound contraction and tissue continuity, despite limited angiogenic signaling. These findings indicate that the ACL’s failure to heal is not attributable to the absence of progenitor or angiogenic factors, but rather to its fragmented spatial organization and dominant contractile phenotype. Therapeutically, preserving and modulating the EL, particularly its CD34+ and α-SMA+ compartments, could be key to enhancing intrinsic ACL repair and improving outcomes in ligament reconstruction and regeneration. Full article
(This article belongs to the Special Issue Acute Trauma and Trauma Care in Orthopedics: 2nd Edition)
Show Figures

Figure 1

25 pages, 2932 KB  
Article
Pain Hypersensitivity in a Mouse Model of Marfan Syndrome
by Rebecca Kordikowski, Joana Coutinho, Ignacio Martínez-Martel, Clara Penas, Beatriz Martín-Mur, Belén Pérez, Francesc Jiménez-Altayó and Olga Pol
Antioxidants 2026, 15(1), 80; https://doi.org/10.3390/antiox15010080 (registering DOI) - 8 Jan 2026
Viewed by 177
Abstract
Marfan syndrome (MFS) is a genetic disorder caused by mutations in the fibrillin-1 (Fbn1) gene, leading to structurally abnormal elastic fibers and diverse clinical manifestations. Aortic root dilation represents the most serious threat, often requiring prophylactic surgical repair. Emerging evidence suggests that MFS [...] Read more.
Marfan syndrome (MFS) is a genetic disorder caused by mutations in the fibrillin-1 (Fbn1) gene, leading to structurally abnormal elastic fibers and diverse clinical manifestations. Aortic root dilation represents the most serious threat, often requiring prophylactic surgical repair. Emerging evidence suggests that MFS patients experience increased pain sensitivity, contributing to functional impairment and reduced quality of life. Here, we used C57BL/6 wild-type and Fbn1C1041G/+ (MFS) mice to examine brain transcriptomics, aortic histology, nociceptive behaviors, grip strength, and spinal cord gene expression in both sexes at 2, 4, 6, 8, and 16 months of age. Transcriptomic analysis revealed reduced activation of pain-related pathways in young males and aged females, with a reversal in aged males, suggesting age- and sex-dependent differences in pain modulation. Behavioral testing showed progressive mechanical and thermal hypersensitivity in MFS mice, with cold allodynia as the earliest manifestation with late-onset muscle weakness. In the spinal cord of 16-month-old MFS mice, increased expression of key excitatory and nociceptive markers was observed, consistent with the pain hypersensitivity phenotype. In addition, aged female MFS mice exhibited elevated spinal expression of pro-inflammatory cytokines, inducible nitric oxide synthase, and Nox4, whereas males showed increased transforming growth factor-β1 and Nox1, reflecting distinct inflammatory and oxidative stress profiles. These findings demonstrate that Fbn1C1041G/+ mice reproduce pain hypersensitivity and muscle deficits observed in MFS patients, supporting their use as a preclinical model. Our results suggest that enhanced spinal excitatory/nociceptive signaling, together with neuroinflammation and oxidative stress, contributes to sex- and age-specific pain mechanisms in MFS. Full article
(This article belongs to the Special Issue Chronic Pain and Oxidative Stress)
Show Figures

Figure 1

26 pages, 1058 KB  
Review
The Adipokine Axis in Heart Failure: Linking Obesity, Sarcopenia and Cardiac Dysfunction in HFpEF
by Luka Komić, Jelena Komić, Nikola Pavlović, Marko Kumrić, Josipa Bukić, Iris Jerončić Tomić and Joško Božić
Int. J. Mol. Sci. 2026, 27(2), 612; https://doi.org/10.3390/ijms27020612 - 7 Jan 2026
Viewed by 223
Abstract
Obesity, sarcopenia, and heart failure with preserved ejection fraction (HFpEF) constitute an interconnected clinical triad driven by multisystem mechanisms centered on the adipokine axis. Adipose tissue, now recognized as a dynamic endocrine organ, undergoes pathological remodeling in obesity, characterized by hypoxia, chronic low-grade [...] Read more.
Obesity, sarcopenia, and heart failure with preserved ejection fraction (HFpEF) constitute an interconnected clinical triad driven by multisystem mechanisms centered on the adipokine axis. Adipose tissue, now recognized as a dynamic endocrine organ, undergoes pathological remodeling in obesity, characterized by hypoxia, chronic low-grade inflammation, and dysregulated adipokine secretion. These changes impair endothelial function, promote myocardial fibrosis, and disrupt skeletal muscle metabolism, thereby linking cardiometabolic and musculoskeletal dysfunction. This review integrates current evidence on homeostatic adipokines, such as adiponectin, apelin, and omentin, that preserve vascular and muscular resilience, as well as stress-inducible adipokines, such as leptin, resistin, and GDF15, that reflect or amplify metabolic and inflammatory injury. A maladaptive adipokine milieu associates with a self-reinforcing cycle of endothelial dysfunction, myocardial stiffening, and muscle atrophy that characterizes s HFpEF in the context of obesity and sarcopenia. We further discuss emerging translational applications, including diagnostic and prognostic adipokine signatures, targeted modulation of adipokine pathways, and the therapeutic impact of GLP-1 receptor agonists on adipose–cardiovascular–muscle crosstalk. Remaining challenges, including the adiponectin paradox and pleiotropic adipokine effects, highlight the need for precision-medicine approaches integrating multimodal biomarker profiling with cardiometabolic and musculoskeletal phenotyping. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

20 pages, 1354 KB  
Review
Central and Peripheral Sensitization in Temporomandibular Disorders: Proposed Mechanisms of Botulinum Toxin Therapy
by Basit Ali Chaudhry, Christopher L. Robinson, Edoardo Caronna, Freda Dodd-Glover, Amrittej Singh Virk, Mario Fernando Prieto Peres, Hope L. O’Brien, Marcela Romero-Reyes and Sait Ashina
Toxins 2026, 18(1), 28; https://doi.org/10.3390/toxins18010028 - 6 Jan 2026
Viewed by 362
Abstract
Temporomandibular disorders (TMDs) are common musculoskeletal chronic orofacial pain conditions involving peripheral and central sensitization within trigeminal nociceptive pathways, manifesting as mechanical allodynia and functional impairment. Botulinum toxin type A (BoNT-A) has been explored as a treatment targeting both muscle hyperactivity and nociceptive [...] Read more.
Temporomandibular disorders (TMDs) are common musculoskeletal chronic orofacial pain conditions involving peripheral and central sensitization within trigeminal nociceptive pathways, manifesting as mechanical allodynia and functional impairment. Botulinum toxin type A (BoNT-A) has been explored as a treatment targeting both muscle hyperactivity and nociceptive modulation. Preclinical and clinical evidence demonstrate that BoNT-A reduces peripheral neurotransmitter release, neurogenic inflammation, and central neuronal excitability, leading to attenuation of mechanical allodynia in TMD models and patients. Clinical trials show modest and variable analgesic effects, with patients displaying sensory sensitization appearing to respond more favorably, though methodological heterogeneity limits definitive conclusions. Safety concerns related to muscle weakening, changes in bone density, and structural changes underscore the need for standardized protocols optimizing dosing and monitoring, in addition to prospective studies. These findings suggest that BoNT-A may serve as an adjunctive, mechanism-based therapy within multimodal TMD management. Future research should focus on standardized sensory phenotyping and trial design to clarify BoNT-A’s role in modulating central sensitization and improving patient outcomes. Full article
Show Figures

Figure 1

20 pages, 2378 KB  
Article
Phosphomimetic Thrombospondin-1 Modulates Integrin β1-FAK Signaling and Vascular Cell Functions
by Assala Raya, Bálint Bécsi and Anita Boratkó
Biomolecules 2026, 16(1), 84; https://doi.org/10.3390/biom16010084 - 4 Jan 2026
Viewed by 295
Abstract
Thrombospondin-1 (TSP1) is a multifunctional glycoprotein that plays a crucial role in angiogenesis and vascular remodeling. Ser93 of TSP1 has recently been identified as a novel phosphorylation site, influencing angiogenic properties; however, the underlying signaling mechanism remains unclear. Here, we investigated the functional [...] Read more.
Thrombospondin-1 (TSP1) is a multifunctional glycoprotein that plays a crucial role in angiogenesis and vascular remodeling. Ser93 of TSP1 has recently been identified as a novel phosphorylation site, influencing angiogenic properties; however, the underlying signaling mechanism remains unclear. Here, we investigated the functional impact of Ser93 phosphorylation using phosphomimetic (TSP1S93D) and phosphonull (TSP1S93A) mutants. Endothelial cell (EC) migration was analyzed using scratch assay and electric cell-substrate impedance sensing. Activation of key pathways (Akt, p38, ERK, and FAK) was analyzed by immunoblotting. TSP1 secretion was quantified by ELISA. Downstream effects on smooth muscle cells were examined by Western blot using conditioned media of endothelial cells. Expression of TSP1S93D significantly impaired endothelial migration and wound closure, associated with reduced phosphorylation of FAK and paxillin. Upstream of FAK signaling, TSP1S93D showed enhanced binding to integrin β1 and promoted its clustering. In contrast, TSP1S93D stimulated smooth muscle cell proliferation, migration, cytoskeletal remodeling, and phenotypic switching toward a synthetic, pro-inflammatory state characterized by elevated marker protein expression. Together, these findings demonstrate that the impaired angiogenic properties induced by TSP1S93D result from the modulation of integrin β1-FAK pathways in ECs, suppressing endothelial motility while promoting smooth muscle activation, suggesting a role in early vascular remodeling and inflammation. Full article
(This article belongs to the Section Cellular Biochemistry)
Show Figures

Figure 1

19 pages, 1035 KB  
Article
The Transforming Growth Factor β Genes and Susceptibility to Musculoskeletal Injuries in a Physically Active Caucasian Cohort
by Agata Rzeszutko-Bełzowska and Agata Leońska-Duniec
J. Clin. Med. 2026, 15(1), 358; https://doi.org/10.3390/jcm15010358 - 3 Jan 2026
Viewed by 182
Abstract
Background/Objectives: Changes in the physiological activity of transforming growth factor-beta (TGF-β) family caused by genetic variability may significantly affect the phenotype of the musculoskeletal system and, consequently, the risk of sports injuries. This study aimed to investigate whether the TGFBI (rs1442), TGFBR3 [...] Read more.
Background/Objectives: Changes in the physiological activity of transforming growth factor-beta (TGF-β) family caused by genetic variability may significantly affect the phenotype of the musculoskeletal system and, consequently, the risk of sports injuries. This study aimed to investigate whether the TGFBI (rs1442), TGFBR3 (rs1805113 and rs1805117), and MSTN (rs11333758) polymorphisms, either individually or in combination, were associated with susceptibility to muscle injury, anterior cruciate ligament (ACL) rupture, and other injuries. Methods: The study group included 202 physically active Caucasians with reported sport injuries and 133 healthy controls. All the samples were genotyped using real-time polymerase chain reaction (real-time PCR). Results: The results revealed that (1) the TGFBR3 rs1805117 TC genotype was nominally associated with increased ACL injury risk; (2) the MSTN rs11333758 heterozygotes was more frequent in the one injury group (vs controls) and in the ACL group, whereas in the multiple vs. one comparison the over-dominant model suggested lower odds for heterozygotes; and (3) the TGFBI rs1442 CG genotype was nominally associated with lower odds of fractures, dislocations or sprains. In addition, simultaneous analysis of chosen SNPs revealed interactions between TGFBR3 rs1805117 and rs1805113, with a nominal association of the rs1805113 G allele with increased injury risk, as did rs11333758 and rs1805113, with a potential effect of rs11333758 on injury status. However, haplotype analysis of the TGFBR3 SNPs revealed no significant associations. After Bonferroni correction, none of the associations remained statistically significant. Conclusions: The results suggested that carrying specific TGFBI, TGFBR3, and MSTN genotypes may be potentially associated with susceptibility to musculoskeletal injuries in a physically active Caucasians. Full article
(This article belongs to the Section Sports Medicine)
Show Figures

Figure 1

16 pages, 2046 KB  
Article
Distinct Role of γ-Synuclein in the Regulation of Motor Performance and Behavioral Responses in Mice
by Iuliia S. Sukhanova, Kirill D. Chaprov, Olga A. Morozova, Ruslan K. Ovchinnikov, Olga A. Kukharskaya, Valeria N. Zalevskaya, Nadezhda M. Yusupova, Anastasia A. Lugovskaya, Natalia N. Ninkina and Michail S. Kukharsky
Biomedicines 2026, 14(1), 92; https://doi.org/10.3390/biomedicines14010092 - 2 Jan 2026
Viewed by 321
Abstract
Background: The three Synuclein family members (α-, β-, and γ-synuclein) are presynaptic proteins that regulate synaptic vesicle trafficking and thereby influence neurotransmitter release. Synucleins belong to a class of intrinsically disordered proteins and are prone to aggregation into pathological deposits, which may impair [...] Read more.
Background: The three Synuclein family members (α-, β-, and γ-synuclein) are presynaptic proteins that regulate synaptic vesicle trafficking and thereby influence neurotransmitter release. Synucleins belong to a class of intrinsically disordered proteins and are prone to aggregation into pathological deposits, which may impair their physiological synaptic functions. Knockout (KO) mouse lines, commonly used to model synuclein depletion in the nervous system, reveal a range of phenotypes with different motor and behavioral deficits. However, given the high sequence homology and functional interplay among the three synucleins, the specific contribution of each family member to these phenotypes remains poorly understood. Objective: In this study, we conducted a comparative phenotypic analysis of γ-synuclein KO, α- and β-synuclein KO, and αβγ-synuclein KO mice. Methods: Mice were subjected to a battery of behavioral tests assessing motor activity and coordination, anxiety-like behavior, and spatial learning and memory. Synaptic vesicle proteins were analyzed in brain tissues using Western blotting. Results: We observed that knocking out γ-synuclein but not α- and β-synucleins reduces mouse lifespan and leads to sustained reduction in muscle strength implicating that γ-synuclein is essential for longevity and motor system function. Another consequence of γ-synuclein deficiency is altered anxiety-like behavior manifested as a diminished aversive response, while exploratory behavior and memory remain intact. The triple KO mice mirror γ-synuclein KO mice in some behavioral changes, including shortened lifespan, reduced muscle strength, and decreased anxiety-like behavior. However, the triple KO mice additionally exhibit hyperactivity, which is not present in the other groups. No changes in synaptic vesicle marker levels were detected, indicating that the observed motor and behavioral abnormalities are not attributable to impaired synaptic connectivity. Conclusions: Taken together, these findings demonstrate nonredundant functions of individual synuclein family members and highlight a distinct role of γ-synuclein in regulating motor performance and behavioral responses. Full article
(This article belongs to the Section Neurobiology and Clinical Neuroscience)
Show Figures

Figure 1

9 pages, 1016 KB  
Article
Risk Factors Associated with the Development of Thrombotic Microangiopathy in Patients with Dermatomyositis
by Fabiola Cassiano-Quezada, Daniel Alberto Carrillo-Vázquez, Jiram Torres-Ruiz, Nancy Raquel Mejía-Domínguez, Karina Santana-de Anda, Ericka Abigail Guevara-Rojas and Diana Gómez-Martín
Int. J. Mol. Sci. 2026, 27(1), 315; https://doi.org/10.3390/ijms27010315 - 27 Dec 2025
Viewed by 234
Abstract
Thrombotic microangiopathy (TMA) is an infrequent and poorly understood manifestation in dermatomyositis (DM) associated with poor outcomes and refractoriness to treatment. The aim of this study is to describe the clinical characteristics and risk factors for its development. We conducted a nested case–control [...] Read more.
Thrombotic microangiopathy (TMA) is an infrequent and poorly understood manifestation in dermatomyositis (DM) associated with poor outcomes and refractoriness to treatment. The aim of this study is to describe the clinical characteristics and risk factors for its development. We conducted a nested case–control study comparing patients with DM who developed TMA to those with DM without this complication. Disease activity was evaluated using the Myositis Disease Activity Assessment Tool (MDAAT), the Manual Muscle Test of eight muscle groups (MMT8), and muscle enzyme levels. A binomial logistic regression analysis was performed to identify risk factors for the development of TMA among patients with DM. All patients with TMA had DM. Patients with DM/TMA had a shorter time since disease onset (p = 0.033), lower levels of C3 (p = 0.07) and C4 (p = 0.046), as well as higher leukocyte (p = 0.044), neutrophil (p = 0.033), and creatine phosphokinase (CK) levels (p = 0.005). They also exhibited higher constitutional (p = 0.0008), pulmonary (p = 0.008), and muscle disease activity (p = 0.027). In the univariate analysis, a shorter time since disease onset (OR 0.42, p = 0.0042) indicated an increased risk for TMA, as did low complement levels (C3: OR 1.11, p = 0.01; C4: OR 1.18, p = 0.02) and higher constitutional (OR 2.27, p = 0.0014), pulmonary (OR 5.50, p = 0.0004), and muscle disease activity (OR 2.1, p = 0.003). Although elevated CK levels (OR 1.001, p = 0.0008) reached statistical significance, the effect size was minimal and should not be interpreted as a clinically relevant increase in risk. Confocal microscopy of muscle biopsy specimens demonstrated neutrophil extracellular traps (NETs) infiltrating muscle tissue. Patients with DM who develop TMA appear to exhibit a distinct clinical phenotype characterized by leukocytosis, neutrophilia, hypocomplementemia, shorter disease duration, and greater constitutional, pulmonary, and muscular disease activity. Although limited by the small sample size, these findings suggest a potential role of NETs in microvascular and tissue injury associated with DM-related TMA. Larger studies are warranted to validate these observations and further elucidate the underlying pathogenic mechanisms. Full article
(This article belongs to the Special Issue Advances in the Genetics, Epidemiology and Management of Myopathies)
Show Figures

Figure 1

11 pages, 669 KB  
Article
Associations Between the Severity of Sarcopenia and Health-Related Quality of Life in Older Adults
by Wei-Syun Hung, Ying-Jen Chen, Tz-Shiu Tsai, Chern-Horng Lee, Ji-Tseng Fang, Ming-Shien Wen, Chun-Yen Lin, Kuo-Chen Liao and Chieh-Li Yen
J. Clin. Med. 2026, 15(1), 161; https://doi.org/10.3390/jcm15010161 - 25 Dec 2025
Viewed by 272
Abstract
Background: Sarcopenia is a progressive skeletal muscle disorder associated with adverse outcomes. Although the association between sarcopenia and quality of life (QoL) has been discussed, the specific relationship between different stages of sarcopenia severity—particularly distinguishing between muscle mass loss and functional impairment—and health-related [...] Read more.
Background: Sarcopenia is a progressive skeletal muscle disorder associated with adverse outcomes. Although the association between sarcopenia and quality of life (QoL) has been discussed, the specific relationship between different stages of sarcopenia severity—particularly distinguishing between muscle mass loss and functional impairment—and health-related quality of life (HRQoL) remains unclear. Method: This cross-sectional study enrolled 100 elderly participants from a geriatric outpatient clinic. Participants were categorized into four groups (normal, possible sarcopenia, sarcopenia and severe sarcopenia) based on the 2019 Asian Working Group for Sarcopenia (AWGS) criteria. HRQoL was assessed using the Short-Form 36-Item (SF-36) questionnaire. Result: The severe sarcopenia group was significantly older and had lower calf circumference compared to the normal group. Notably, the possible sarcopenia group presented with the highest body mass index and body fat percentage, resembling a “dynapenic obesity” phenotype. In terms of QoL, participants with confirmed sarcopenia did not exhibit significant differences compared to the normal group. However, the severe sarcopenia group demonstrated significantly lower scores across almost all SF-36 domains compared to the normal group. Multivariate linear regression analysis revealed that severe sarcopenia was independently and significantly negatively associated with multiple QoL domains, including physical functioning, general health and vitality. Additionally, age, social activity and body fat were identified as independent correlates of specific QoL domains. Conclusions: Our findings suggest a non-linear relationship between sarcopenia and HRQoL. A comprehensive decline in HRQoL is strongly linked to the severity of sarcopenia (functional impairment) rather than the diagnosis of muscle mass loss alone. These results highlight the clinical importance of preserving physical performance and suggest that categorizing different severities of sarcopenia and stage-specific management strategies are necessary to improve quality of life in older adults. Full article
(This article belongs to the Section Geriatric Medicine)
Show Figures

Figure 1

14 pages, 3925 KB  
Article
CDKN2B Inhibits Vascular Smooth Muscle Phenotypic Switching in Corpus Spongiosum Surrounding the Urethral Plate in Hypospadias
by Jiayao Huang, Zihan Xu, Jiacheng Huang, Xiaoqin Yin, Yichen Huang and Fang Chen
Biomedicines 2026, 14(1), 32; https://doi.org/10.3390/biomedicines14010032 - 23 Dec 2025
Viewed by 317
Abstract
Objective: Phenotypic switching of vascular smooth muscle cells (VSMCs) in the corpus spongiosum may contribute to abnormal urethral development in hypospadias, but the underlying molecular regulators remain unclear. This study aimed to identify hub genes associated with VSMCs phenotypic switching in the corpus [...] Read more.
Objective: Phenotypic switching of vascular smooth muscle cells (VSMCs) in the corpus spongiosum may contribute to abnormal urethral development in hypospadias, but the underlying molecular regulators remain unclear. This study aimed to identify hub genes associated with VSMCs phenotypic switching in the corpus spongiosum using RNA sequencing and Weighted Gene Co-expression Network Analysis (WGCNA), and to functionally characterize the top candidate gene CDKN2B. Methods: Corpus spongiosum tissue samples were collected from seven patients with proximal hypospadias and five patients with urethral stricture (control group). The expression of the VSMCs contractile markers Calponin 1 and α-SMA, and the secretory marker OPN, was evaluated by qRT-PCR and Western blotting to assess VSMCs phenotypic state. RNA sequencing and Weighted Gene Co-expression Network Analysis (WGCNA) were performed to identify hub genes, which were then validated by qRT-PCR. Primary VSMCs were isolated from corpus spongiosum tissue and transduced with lentiviral vectors to either suppress or overexpress CDKN2B. Changes in VSMC marker expression and in key signaling pathways associated with phenotypic switching—specifically TGF/Smad and SRF/MYOCD—were analyzed using qRT-PCR and Western blotting. Results: In hypospadias tissue, the decreased expression of α-SMA and Calponin 1, together with increased OPN, indicated a shift in VSMCs from a contractile to a secretory phenotype. RNA-seq and WGCNA identified 11 differentially expressed genes, among which CDKN2B showed a marked downregulation in hypospadias samples. In control VSMCs, CDKN2B inhibition led to reduced α-SMA and Calponin 1, elevated OPN, and suppressed activity of TGF/Smad and SRF/MYOCD signaling. Conversely, CDKN2B overexpression in VSMCs from hypospadias samples restored α-SMA and Calponin 1 expression, decreased OPN, and enhanced TGF/Smad and SRF/MYOCD pathway activation. Conclusions: VSMCs in the corpus spongiosum surrounding the urethral plate in hypospadias undergo a transition from a contractile to a secretory phenotype. CDKN2B emerges from unbiased transcriptomic screening as a key hub gene and functions as a critical regulator of this process, maintaining the contractile phenotype by modulating canonical TGF/Smad and SRF/MYOCD signaling. The CDKN2B–TGF/Smad axis may represent a central pathway linking VSMC phenotypic switching to abnormal vascular remodeling in hypospadias. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

Back to TopTop