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
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (117)

Search Parameters:
Keywords = vascular injury biomarkers

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 587 KiB  
Review
Uric Acid and Preeclampsia: Pathophysiological Interactions and the Emerging Role of Inflammasome Activation
by Celia Arias-Sánchez, Antonio Pérez-Olmos, Virginia Reverte, Isabel Hernández, Santiago Cuevas and María Teresa Llinás
Antioxidants 2025, 14(8), 928; https://doi.org/10.3390/antiox14080928 - 29 Jul 2025
Viewed by 477
Abstract
Preeclampsia (PE) is a multifactorial hypertensive disorder unique to pregnancy and a leading cause of maternal and fetal morbidity and mortality worldwide. Its pathogenesis involves placental dysfunction and an exaggerated maternal inflammatory response. Uric acid (UA), traditionally regarded as a marker of renal [...] Read more.
Preeclampsia (PE) is a multifactorial hypertensive disorder unique to pregnancy and a leading cause of maternal and fetal morbidity and mortality worldwide. Its pathogenesis involves placental dysfunction and an exaggerated maternal inflammatory response. Uric acid (UA), traditionally regarded as a marker of renal impairment, is increasingly recognized as an active contributor to the development of PE. Elevated UA levels are associated with oxidative stress, endothelial dysfunction, immune activation, and reduced renal clearance. Clinically, UA is measured in the second and third trimesters to assess disease severity and guide obstetric management, with higher levels correlating with early-onset PE and adverse perinatal outcomes. Its predictive accuracy improves when combined with other clinical and biochemical markers, particularly in low-resource settings. Mechanistically, UA and its monosodium urate crystals can activate the NLRP3 inflammasome, a cytosolic multiprotein complex of the innate immune system. This activation promotes the release of IL-1β and IL-18, exacerbating placental, vascular, and renal inflammation. NLRP3 inflammasome activation has been documented in placental tissues, immune cells, and kidneys of women with PE and is associated with hypertension, proteinuria, and endothelial injury. Experimental studies indicate that targeting UA metabolism or inhibiting NLRP3 activation, using agents such as allopurinol, metformin, or MCC950, can mitigate the clinical and histopathological features of PE. These findings support the dual role of UA as both a biomarker and a potential therapeutic target in the management of the disease. Full article
Show Figures

Graphical abstract

10 pages, 377 KiB  
Article
Functional MPO Polymorphisms and Haplotypes Affect Both Myeloperoxidase Levels and Association with Hypertensive Disorders of Pregnancy
by Daniela Alves Pereira, Marcelo Rizzatti Luizon, Ricardo Carvalho Cavalli, Jose Eduardo Tanus-Santos and Valéria Cristina Sandrim
Int. J. Mol. Sci. 2025, 26(15), 7071; https://doi.org/10.3390/ijms26157071 - 23 Jul 2025
Viewed by 222
Abstract
Preeclampsia (PE) shares common pathophysiological mechanisms with cardiovascular diseases, including endothelial dysfunction and exacerbated inflammatory response. Myeloperoxidase (MPO) has been suggested as a biomarker for cardiovascular risk, and its circulating levels are contradictory in PE. Elevated levels of MPO can promote host tissue [...] Read more.
Preeclampsia (PE) shares common pathophysiological mechanisms with cardiovascular diseases, including endothelial dysfunction and exacerbated inflammatory response. Myeloperoxidase (MPO) has been suggested as a biomarker for cardiovascular risk, and its circulating levels are contradictory in PE. Elevated levels of MPO can promote host tissue damage and trigger vascular injury. MPO gene polymorphisms affect circulating MPO levels under different conditions. To date, no studies have investigated whether MPO polymorphisms influence MPO levels in hypertensive disorders of pregnancy. In this study, we examined the impact of two specific MPO polymorphisms—rs2243828 and rs2071409—and their associated haplotypes on MPO levels. We also explored their potential association with gestational hypertension (GH) and preeclampsia (PE). Our study included 136 healthy pregnant women (HP), including 118 with GH and 140 with PE. Genotyping was performed using TaqMan allele discrimination assays, and MPO levels were quantified using an ELISA assay. The TT genotype of the rs2243828 polymorphism was associated with lower MPO concentration in GH, and the CC genotype presented a higher frequency in the GH group than the HP group. The AC+CC rs2071409 polymorphism was associated with lower MPO concentration in GH. We also found that the ‘C, C’ haplotype was less frequent and was associated with lower MPO concentration in PE. Our findings suggest that both rs2243828 and rs2071409 polymorphisms might contribute to MPO levels in GH and that the haplotype ‘C, C’ formed by them may protect against PE. Full article
Show Figures

Figure 1

23 pages, 2202 KiB  
Article
Afucosylated IgG Promote Thrombosis in Mouse Injected with SARS-CoV-2 Spike Expressing Megakaryocytes
by Meryem Mabrouk, Farah Atifi, Hicham Wahnou, Afaf Allaoui, Nabil Zaid, Abdallah Naya, Ejaife O. Agbani, Loubna Khalki, Meriem Khyatti, Youssef Tijani, Khadija Akarid, Damien Arnoult, Haissam Abou-Saleh, Othman El Faqer, Salma Labied, Mounia Ammara, Fadila Guessous, Farid Jalali and Younes Zaid
Int. J. Mol. Sci. 2025, 26(14), 7002; https://doi.org/10.3390/ijms26147002 - 21 Jul 2025
Viewed by 531
Abstract
Despite the prevalence of fucosylated IgG in plasma, specific IgGs with low core fucosylation sporadically emerge in response to virus infections and blood cell alloantigens. This low fucosylation of IgG is implicated in the pathogenesis of SARS-CoV-2 and dengue infections. In COVID-19, the [...] Read more.
Despite the prevalence of fucosylated IgG in plasma, specific IgGs with low core fucosylation sporadically emerge in response to virus infections and blood cell alloantigens. This low fucosylation of IgG is implicated in the pathogenesis of SARS-CoV-2 and dengue infections. In COVID-19, the presence of IgGs with low core fucosylation (afucosylated IgGs) targeting spike protein predicts disease progression to a severe form and actively mediates this progression. This study reveals that SARS-CoV-2 infection of megakaryocytes promotes the generation of pathogenic afucosylated anti-spike IgGs, leading to outcomes, such as pulmonary vascular thrombosis, acute lung injury, and mortality in FcγRIIa-transgenic mice. Platelets from mice injected with virus-infected human megakaryocytes express significant activation biomarkers, indicating a direct link between the immune response and platelet activation. Mice injected with virus-infected human megakaryocytes demonstrate an elevated rate of thrombus formation induced by FeCl3 (4%) and a reduction in bleeding time, emphasizing the intricate interplay of viral infection, immune response, and hemostatic complications. Treatment with inhibitors targeting FcγRIIa, serotonin, or complement anaphylatoxins of mice injected with spike-expressing MKs successfully prevents observed platelet activation, thrombus formation, and bleeding abnormalities, offering potential therapeutic strategies for managing severe outcomes associated with afucosylated IgGs in COVID-19 and related disorders. Full article
(This article belongs to the Special Issue The Molecular Role of Platelets in Human Diseases)
Show Figures

Figure 1

20 pages, 695 KiB  
Review
The Pathogenic Role of C-Reactive Protein in Diabetes-Linked Unstable Atherosclerosis
by Melania Sibianu and Mark Slevin
Int. J. Mol. Sci. 2025, 26(14), 6855; https://doi.org/10.3390/ijms26146855 - 17 Jul 2025
Viewed by 368
Abstract
C-reactive protein (CRP) has long been recognized as a biomarker of systemic inflammation and cardiovascular disease (CVD) risk. However, emerging evidence highlights the distinct and potent pro-inflammatory role of its monomeric form (mCRP), which is predominantly tissue-bound and directly implicated in vascular injury [...] Read more.
C-reactive protein (CRP) has long been recognized as a biomarker of systemic inflammation and cardiovascular disease (CVD) risk. However, emerging evidence highlights the distinct and potent pro-inflammatory role of its monomeric form (mCRP), which is predominantly tissue-bound and directly implicated in vascular injury and plaque destabilization. This narrative review explores the interactions and overlapping pathways that converge within and modulate CRP, mCRP, the associated pathophysiology of diabetes mellitus, and cardiovascular disease. We examine how mCRP promotes endothelial dysfunction, leukocyte recruitment, platelet activation, and macrophage polarization, thereby contributing to the formation of unstable atherosclerotic plaques. Furthermore, we discuss the critical influence of diabetes in amplifying mCRP’s pathogenic effects through metabolic dysregulation, chronic hyperglycemia, and enhanced formation of advanced glycation end products (AGEs). The synergistic interaction of mCRP with the AGE-receptor for AGE (RAGE) axis exacerbates oxidative stress and vascular inflammation, accelerating atherosclerosis progression and increasing cardiovascular risk in diabetic patients. Understanding these mechanistic pathways implicates mCRP as both a biomarker and therapeutic target, particularly in the context of diabetes-associated CVD. This review highlights the need for further research into targeted interventions that disrupt the mCRP-[AGE-RAGE] inflammatory cycle to reduce plaque instability and improve cardiovascular outcomes in high-risk populations. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

19 pages, 1543 KiB  
Article
Peripheral Leukocyte Syndecan-3 Is Elevated in Alzheimer’s Disease: Evidence from a Human Study
by Anett Hudák, Annamária Letoha and Tamás Letoha
Int. J. Mol. Sci. 2025, 26(14), 6587; https://doi.org/10.3390/ijms26146587 - 9 Jul 2025
Viewed by 702
Abstract
Syndecan-3 (SDC3), a transmembrane heparan sulfate proteoglycan involved in cell signaling and endocytosis, has recently been implicated in the pathogenesis of neurodegenerative disorders. While preclinical studies have demonstrated its role in Alzheimer’s disease (AD), its diagnostic relevance in peripheral blood remains unexplored. In [...] Read more.
Syndecan-3 (SDC3), a transmembrane heparan sulfate proteoglycan involved in cell signaling and endocytosis, has recently been implicated in the pathogenesis of neurodegenerative disorders. While preclinical studies have demonstrated its role in Alzheimer’s disease (AD), its diagnostic relevance in peripheral blood remains unexplored. In this human cohort study, we measured SDC3 expression in peripheral blood mononuclear cells (PBMCs) from 22 clinically diagnosed AD patients and 20 cognitively unimpaired non-AD controls using a custom ELISA. The findings were compared with plasma p-tau217 levels and a panel of systemic laboratory markers. PBMC-expressed SDC3 was significantly elevated in AD patients and moderately correlated with AD status (r = 0.309, p = 0.0465) independent of age. Notably, SDC3 levels were inversely correlated with systemic inflammatory markers, including C-reactive protein (CRP; r = −0.421, p = 0.0055) and D-dimer (r = −0.343, p = 0.038), suggesting an AD-associated immune phenotype distinct from acute-phase or vascular inflammation. Conversely, plasma p-tau217 levels did not significantly differ between groups but correlated with markers of tissue injury and inflammation (LDH, GOT, and ferritin), potentially reflecting systemic influences in non-AD controls. A multivariable logistic regression model incorporating SDC3, p-tau217, and age demonstrated high diagnostic accuracy (AUC = 0.85). These findings identify PBMC-expressed SDC3 as a promising blood-based biomarker candidate for AD, warranting further validation in larger, biomarker-confirmed cohorts. Full article
Show Figures

Figure 1

20 pages, 5004 KiB  
Article
Maresin1 Alleviates Ischemia Reperfusion Injury After Lung Transplantation by Inhibiting Ferroptosis via the PKA-Hippo-YAP Signaling Pathway
by Peng Deng, You Wu, Li Wan, Xiangfu Sun and Quanchao Sun
Biomedicines 2025, 13(7), 1594; https://doi.org/10.3390/biomedicines13071594 - 30 Jun 2025
Viewed by 426
Abstract
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of [...] Read more.
Background: Lung ischemia reperfusion injury (LIRI) is a severe complication after lung transplantation (LT). Ferroptosis contributes to the pathogenesis of LIRI. Maresin1 (MaR1) is an endogenous pro-resolving lipid mediator that exerts protective effects against multiorgan diseases. However, the role and mechanism of MaR1 in the ferroptosis of LIRI after LT need to be further investigated. Methods: A mouse LT model and a pulmonary vascular endothelial cell line after hypoxia reoxygenation (H/R) culture were established in our study. Histological morphology and inflammatory cytokine levels predicted the severity of LIRI. Cell viability and cell injury were determined by CCK-8 and LDH assays. Ferroptosis biomarkers, including Fe2+, MDA, 4-HNE, and GSH, were assessed by relevant assay kits. Transferrin receptor (TFRC) and Acyl-CoA Synthetase Long Chain Family Member 4 (ACSL4) protein levels were examined by western blotting. In vitro, lipid peroxide levels were detected by DCFH-DA staining and flow cytometry analysis. The ultrastructure of mitochondria was imaged using transmission electron microscopy. Furthermore, the potential mechanism by which MaR1 regulates ferroptosis was explored and verified with signaling pathway inhibitors using Western blotting. Results: MaR1 protected mice from LIRI after LTx, which was reversed by the ferroptosis agonist Sorafenib in vivo. MaR1 administration decreased Fe2+, MDA, 4-HNE, TFRC, and ACSL4 contents, increased GSH levels, and ameliorated mitochondrial ultrastructural injury after LTx. In vitro, Sorafenib resulted in lower cell viability and worsened cell injury and enhanced the hallmarks of ferroptosis after H/R culture, which was rescued by MaR1 treatment. Mechanistically, the protein kinase A and YAP inhibitors partly blocked the effects of MaR1 on ferroptosis inhibition and LIRI protection. Conclusions: This study revealed that MaR1 alleviates LIRI and represses ischemia reperfusion-induced ferroptosis via the PKA-Hippo-YAP signaling pathway, which may offer a promising theoretical basis for the clinical application of organ protection after LTx. Full article
(This article belongs to the Special Issue The Role of Iron in Human Diseases)
Show Figures

Figure 1

15 pages, 455 KiB  
Article
Dead or Alive? Identification of Postmortem Blood Through Detection of D-Dimer
by Amy N. Brodeur, Tai-Hua Tsai, Gulnaz T. Javan, Dakota Bell, Christian Stadler, Gabriela Roca and Sara C. Zapico
Biology 2025, 14(7), 784; https://doi.org/10.3390/biology14070784 - 28 Jun 2025
Viewed by 370
Abstract
At crime scenes, apart from the detection of blood, it may be important to determine whether a person was alive at the time of blood deposition. Based on the rapid onset of fibrinolysis after death, this pathway could be considered to identify potential [...] Read more.
At crime scenes, apart from the detection of blood, it may be important to determine whether a person was alive at the time of blood deposition. Based on the rapid onset of fibrinolysis after death, this pathway could be considered to identify potential biomarkers for postmortem blood. Fibrinolysis is the natural process that breaks down blood clots after healing a vascular injury. One of its products, D-dimer, could be a potential biomarker for postmortem blood. SERATEC® (SERATEC® GmbH, Göttingen, Germany) has developed the PMB immunochromatographic assay to simultaneously detect human hemoglobin and D-dimer. The main goals of this study were to assess the possibility of using this test to detect postmortem blood, evaluate D-dimer levels in antemortem, menstrual, and postmortem blood, and assess the ability to obtain STR profiles from postmortem blood. Except for one degraded sample, all postmortem blood samples reacted positively for the presence of D-dimer using the SERATEC® PMB test. All antemortem blood samples from living individuals showed negative results for D-dimer detection, except for one liquid sample with a weak positive result, probably due to pre-existing health conditions. Menstrual blood samples gave variable results for D-dimer. The DIMERTEST® Latex assay was used for semi-quantitative measurement of D-dimer concentrations, with postmortem and menstrual blood yielding higher D-dimer concentrations compared to antemortem blood. Full STR profiles were developed for all postmortem samples tested except for one degraded sample, pointing to the possibility of not only detecting postmortem blood at the crime scene but also the potential identification of the victim. Full article
Show Figures

Figure 1

20 pages, 751 KiB  
Article
Crosstalk Between Metabolic Biomarkers and Pulse Wave Analysis in Hypertensive Patients
by Mirela Baba, Mihaela Ioana Maris, Adina Bucur, Daniela Jianu, Simina Mariana Moroz, Dana Stoian, Constantin Tudor Luca and Ioana Mozos
Biomedicines 2025, 13(7), 1514; https://doi.org/10.3390/biomedicines13071514 - 20 Jun 2025
Viewed by 522
Abstract
Background/Objectives: Hypertension is strongly linked to changes in vascular function and lipid metabolism. This study aimed to examine the relationship between lipid profiles, various metabolic biomarkers, and pulse wave analysis in patients with hypertension. Methods: A group of 66 hypertensive patients, aged 64 [...] Read more.
Background/Objectives: Hypertension is strongly linked to changes in vascular function and lipid metabolism. This study aimed to examine the relationship between lipid profiles, various metabolic biomarkers, and pulse wave analysis in patients with hypertension. Methods: A group of 66 hypertensive patients, aged 64 ± 10 years, participated in pulse wave analysis utilizing an oscillometric device. Multiple lipid serum biomarkers were assessed, such as total cholesterol (TC), triglycerides (TG), and non-HDL cholesterol (non-HDL). Lipid balance index (LBI) was determined by considering TG, LDL, HDL levels, and lipid-lowering medications. Results: Notable correlations were observed for SBP, DBP, and early vascular aging (EVA) with lipid biomarkers. In addition to serum lipids, metabolic syndrome, insulin resistance, and non-alcoholic fatty liver disease (NAFLD) were significantly linked to pulse wave analysis variables. Multiple regression analysis showed that only TC continued to have a significant association with DBP. Conclusions: Total cholesterol, triglycerides, non-HDL cholesterol, and lipid balance index provide information about systolic and diastolic blood pressure, as well as early vascular aging in hypertensive patients. LBI offers valuable vascular insights in hypertensive individuals with cardiovascular risk factors, early vascular aging, insulin resistance, and NAFLD. The connection between metabolic biomarkers and pulse wave measurements in individuals with hypertension offers a comprehensive method for the early identification of vascular injury and could enhance the prediction of major cardiovascular events. Full article
(This article belongs to the Special Issue Lipids and Lipid Metabolism in Cardiovascular Health and Disease)
Show Figures

Figure 1

25 pages, 1580 KiB  
Review
Inflammasomes and Signaling Pathways: Key Mechanisms in the Pathophysiology of Sepsis
by Jhan S. Saavedra-Torres, María Virginia Pinzón-Fernández, Martin Ocampo-Posada, H. A. Nati-Castillo, Laura Alejandra Jiménez Hincapie, Eder J. Cadrazo-Gil, Marlon Arias-Intriago, Marlon Rojas-Cadena, Andrea Tello-De-la-Torre, Walter Osejos and Juan S. Izquierdo-Condoy
Cells 2025, 14(12), 930; https://doi.org/10.3390/cells14120930 - 19 Jun 2025
Cited by 1 | Viewed by 1892
Abstract
Sepsis is a life-threatening syndrome characterized by a dysregulated immune response to infection, frequently leading to multiorgan failure and high mortality. Inflammasomes—cytosolic multiprotein complexes of the innate immune system—serve as critical platforms for sensing pathogen- and damage-associated molecular patterns (PAMPs and DAMPs). Key [...] Read more.
Sepsis is a life-threatening syndrome characterized by a dysregulated immune response to infection, frequently leading to multiorgan failure and high mortality. Inflammasomes—cytosolic multiprotein complexes of the innate immune system—serve as critical platforms for sensing pathogen- and damage-associated molecular patterns (PAMPs and DAMPs). Key sensors such as NLRP3, AIM2, and IFI16 initiate caspase-1 activation, IL-1β and IL-18 maturation, and gasdermin D–mediated pyroptosis. In sepsis, excessive inflammasome activation drives oxidative stress, endothelial dysfunction, immunothrombosis, and immune exhaustion. This maladaptive cascade is further aggravated by the release of DAMPs and procoagulant factors, compromising vascular integrity and immune homeostasis. Prolonged activation contributes to immunoparalysis, lymphopenia, and increased susceptibility to secondary infections. Inflammasome signaling also intersects with necroptosis and ferroptosis, amplifying systemic inflammation and tissue injury. Additionally, various pathogens exploit immune evasion strategies to modulate inflammasome responses and enhance virulence. Therapeutic interventions under investigation include selective NLRP3 inhibitors, IL-1 blockers, gasdermin D antagonists, and extracorporeal cytokine hemoadsorption. Emerging approaches emphasize biomarker-guided immunomodulation to achieve personalized therapy. While preclinical studies have shown promising results, clinical translation remains limited. Targeting inflammasomes may offer a path toward precision immunotherapy in sepsis, with potential to reduce organ dysfunction and improve survival. Full article
Show Figures

Figure 1

9 pages, 557 KiB  
Article
Diagnostic Value of Sirtuin-1 in Predicting Contrast-Induced Nephropathy After Percutaneous Coronary Intervention
by Melis Ardic and Cuma Bulent Gul
J. Clin. Med. 2025, 14(11), 3953; https://doi.org/10.3390/jcm14113953 - 3 Jun 2025
Viewed by 507
Abstract
Objectives: Contrast-induced acute kidney injury (CI-AKI) remains a frequent and serious complication after cardiac catheterization. Sirtuin-1 (SIRT1), a NAD+-dependent deacetylase, plays a central role in renal protection against ischemia-reperfusion injury, inflammation, and vascular dysfunction. We aimed to investigate whether serum SIRT1 levels could [...] Read more.
Objectives: Contrast-induced acute kidney injury (CI-AKI) remains a frequent and serious complication after cardiac catheterization. Sirtuin-1 (SIRT1), a NAD+-dependent deacetylase, plays a central role in renal protection against ischemia-reperfusion injury, inflammation, and vascular dysfunction. We aimed to investigate whether serum SIRT1 levels could serve as an early diagnostic biomarker for CI-AKI. Methods: This prospective case-control study included 50 patients undergoing elective percutaneous coronary intervention (PCI) for stable angina. Serum SIRT1 levels were measured at baseline, 24 h, and 72 h post-PCI. The occurrence of CI-AKI was defined by a standard rise in serum creatinine, and patients were stratified accordingly. Results: Although SIRT1 levels tended to be lower in patients who developed CI-AKI (n = 17) compared to those without (n = 33), the differences were not statistically significant at any time point (p > 0.05). However, a significant between-group difference was observed in the 72-h change in SIRT1 levels (Δ0–72 h, p = 0.037), with a greater decline in the CI-AKI group. Multivariable logistic regression also revealed a trend-level inverse association between 72-h SIRT1 levels and CI-AKI (β = −0.536, p = 0.099). Conclusions: While SIRT1 is biologically plausible as a renal protective factor, our findings suggest that serial SIRT1 measurement may offer added value as a dynamic biomarker rather than a static diagnostic tool. Confirmatory trials incorporating serial SIRT1 measurements may help translate this molecular signal into clinically actionable tools for early detection of CI-AKI. Full article
(This article belongs to the Special Issue Advances in the Diagnosis and Treatment of Acute Kidney Injury)
Show Figures

Figure 1

16 pages, 1934 KiB  
Review
Pathophysiology in Systemic Sclerosis: Current Insights and Future Perspectives
by Suzan Al-Gburi, Pia Moinzadeh and Thomas Krieg
Sclerosis 2025, 3(2), 17; https://doi.org/10.3390/sclerosis3020017 - 27 May 2025
Viewed by 1215
Abstract
Background: Systemic sclerosis (SSc) is a rare connective tissue disease characterized by vasculopathy, autoimmunity, and fibrosis. Due to its low prevalence and heterogeneous clinical presentation, early diagnosis remains challenging, often delaying appropriate treatment. The disease progresses from microvascular dysfunction, manifesting as Raynaud’s phenomenon, [...] Read more.
Background: Systemic sclerosis (SSc) is a rare connective tissue disease characterized by vasculopathy, autoimmunity, and fibrosis. Due to its low prevalence and heterogeneous clinical presentation, early diagnosis remains challenging, often delaying appropriate treatment. The disease progresses from microvascular dysfunction, manifesting as Raynaud’s phenomenon, to systemic fibrosis affecting multiple organs, including the lungs, gastrointestinal tract, heart, and kidneys. There have been considerable advancements in understanding the pathophysiology of the disease during the last few years and this has already resulted in the improvement of the therapeutic approaches used to control organ-specific manifestations. However, the underlying cause of the disease still remains incompletely elucidated. Methods: Here, we summarize the current knowledge on the SSc pathogenesis. Results: The pathophysiology involves an interplay of chronic inflammation, impaired vascular function, and excessive extracellular matrix deposition, leading to progressive organ damage. Endothelial dysfunction in SSc is driven by immune-mediated injury, oxidative stress, and the imbalance of vasoconstrictors and vasodilators, leading to capillary loss and chronic hypoxia. Autoantibodies against endothelial cells or other toxic factors induce apoptosis and impair angiogenesis, further exacerbating vascular damage. Despite increased angiogenic factor levels, capillary repair mechanisms are defective, resulting in progressive ischemic damage. Dysregulated immune responses involving Th2 cytokines, B cells, and macrophages contribute to fibroblast activation and excessive collagen deposition. Transforming growth factor-beta (TGF-β) plays a central role in fibrotic progression, while fibroblasts resist apoptosis, perpetuating tissue scarring. The extracellular matrix in SSc is abnormally stiff, reinforcing fibroblast activation and creating a self-perpetuating fibrotic cycle. Conclusions: Advances in molecular and cellular understanding have facilitated targeted therapies, yet effective disease-modifying treatments remain limited. Future research should focus on precision medicine approaches, integrating biomarkers and novel therapeutics to improve patient outcomes. Full article
(This article belongs to the Special Issue Recent Advances in Understanding Systemic Sclerosis)
Show Figures

Figure 1

25 pages, 1595 KiB  
Review
The Molecular Mechanisms of Cognitive Dysfunction in Long COVID: A Narrative Review
by Elena Popa, Andrei Emilian Popa, Mihaela Poroch, Vladimir Poroch, Monica Iuliana Ungureanu, Ana Maria Slanina, Agnes Bacusca and Elena Adorata Coman
Int. J. Mol. Sci. 2025, 26(11), 5102; https://doi.org/10.3390/ijms26115102 - 26 May 2025
Viewed by 3258
Abstract
Cognitive dysfunction represents one of the most persistent and disabling features of Long COVID, yet its molecular underpinnings remain incompletely understood. This narrative review synthesizes current evidence on the pathophysiological mechanisms linking SARS-CoV-2 infection to long-term neurocognitive sequelae. Key processes include persistent neuroinflammation, [...] Read more.
Cognitive dysfunction represents one of the most persistent and disabling features of Long COVID, yet its molecular underpinnings remain incompletely understood. This narrative review synthesizes current evidence on the pathophysiological mechanisms linking SARS-CoV-2 infection to long-term neurocognitive sequelae. Key processes include persistent neuroinflammation, blood–brain barrier (BBB) disruption, endothelial dysfunction, immune dysregulation, and neuroendocrine imbalance. Microglial activation and cytokine release (e.g., IL-6, TNF-α) promote synaptic dysfunction and neuronal injury, while activation of inflammasomes such as NLRP3 amplifies CNS inflammation. Vascular abnormalities, including microthrombosis and BBB leakage, facilitate the infiltration of peripheral immune cells and neurotoxic mediators. Hypothalamic–pituitary–adrenal axis dysfunction and reduced vagal tone further exacerbate systemic inflammation and autonomic imbalance. Biomarkers such as GFAP, NFL, IL-6, and S100B have been associated with both neuroinflammation and cognitive symptoms. Notably, transcriptomic signatures in Long COVID overlap with those observed in Alzheimer’s disease, highlighting shared pathways involving tau dysregulation, oxidative stress, and glial reactivity. Understanding these mechanisms is critical for identifying at-risk individuals and developing targeted therapeutic strategies. This review underscores the need for longitudinal research and integrative biomarker analysis to elucidate the molecular trajectory of cognitive impairment in Long COVID. Full article
Show Figures

Figure 1

15 pages, 263 KiB  
Article
Biomarkers of Calcification, Endothelial Injury, and Platelet-Endothelial Interaction in Patients with Aortic Valve Stenosis
by Paweł Bańka, Klaudia Męcka, Adrianna Berger-Kucza, Karolina Wrona-Kolasa, Anna Rybicka-Musialik, Beata Nowak, Marek Elżbieciak, Magdalena Mizia-Szubryt, Wojciech Wróbel, Tomasz Francuz, Michał Lelek, Agnieszka Kosowska, Wojciech Garczorz, Tomasz Bochenek, Andrzej Swinarew, Jarosław Paluch, Maciej Wybraniec and Katarzyna Mizia-Stec
Int. J. Mol. Sci. 2025, 26(10), 4873; https://doi.org/10.3390/ijms26104873 - 19 May 2025
Viewed by 578
Abstract
Aortic stenosis (AS) is a progressive valvular heart disease characterized by fibrocalcific remodeling, inflammation, and hemodynamic disturbances. Serum biomarkers may indirectly reflect these processes. Autotaxin (ATX) and lysophosphatidic acid (LPA) have been implicated in osteogenic differentiation of valvular interstitial cells, while growth differentiation [...] Read more.
Aortic stenosis (AS) is a progressive valvular heart disease characterized by fibrocalcific remodeling, inflammation, and hemodynamic disturbances. Serum biomarkers may indirectly reflect these processes. Autotaxin (ATX) and lysophosphatidic acid (LPA) have been implicated in osteogenic differentiation of valvular interstitial cells, while growth differentiation factor-15 (GDF-15) reflects cellular stress and vascular changes. Thrombomodulin (TM) indicates endothelial injury and interacts with thrombin. This study aimed to evaluate biomarkers focusing on serum ATX, LPA, GDF-15, and TM levels and flow-mediated dilatation (FMD) in patients with AS. Overall, 149 patients were included in the study: 86 consecutive patients with AS hospitalized due to qualification for invasive treatment of AS and 63 controls. The clinical characteristics, echocardiographic data, FMD, and the following biomarkers—ATX, LPA, GDF-15, and TM—were included in the analysis. AS patients presented increased serum levels of ATX, GDF-15, and TM as compared to the controls. Differences in LPA levels were not statistically significant. FMD values were significantly lower in AS patients. The biomarkers mentioned above and FMD correlated with AS severity. There were no differences in both biomarkers’ serum levels and FMD regarding the hemodynamic AS phenotype. GDF-15 serum level was a risk factor for all-cause mortality and MACCE in the 12-month follow-up. Full article
(This article belongs to the Special Issue Cardiovascular Diseases: From Pathology to Therapeutics)
26 pages, 2831 KiB  
Article
Catalpol Protects Against Retinal Ischemia Through Antioxidation, Anti-Ischemia, Downregulation of β-Catenin, VEGF, and Angiopoietin-2: In Vitro and In Vivo Studies
by Howard Wen-Haur Chao, Windsor Wen-Jin Chao and Hsiao-Ming Chao
Int. J. Mol. Sci. 2025, 26(9), 4019; https://doi.org/10.3390/ijms26094019 - 24 Apr 2025
Viewed by 592
Abstract
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen–glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study [...] Read more.
Retinal ischemic disorders present significant threats to vision, characterized by inadequate blood supply oxygen–glucose deprivation (OGD), oxidative stress, and cellular injury, often resulting in irreversible injury. Catalpol, an iridoid glycoside derived from Rehmannia glutinosa, has demonstrated antioxidative and neuroprotective effects. This study aimed at investigating the protective effects and mechanisms of catalpol against oxidative stress or OGD in vitro and retinal ischemia in vivo, focusing on the modulation of key biomarkers of retinal ischemia, including HIF-1α, vascular endothelial growth factor (VEGF), angiopoietin-2, MCP-1, and the Wnt/β-catenin pathway. Cellular viability was assessed using retinal ganglion cell-5 (RGC-5) cells cultured in DMEM; a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed. H2O2 (1 mM)/OGD was utilized. Vehicle or different catalpol concentrations were administered 15 min before the ischemic-like insults. The Wistar rat eyes’ intraocular pressure was increased to 120 mmHg for 60 min to induce retinal ischemia. Intravitreous injections of catalpol (0.5 or 0.25 mM), Wnt inhibitor DKK1 (1 μg/4 μL), anti-VEGF Lucentis (40 μg/4 μL), or anti-VEGF Eylea (160 μg/4 μL) were administered to the rats’ eyes 15 min before or after retinal ischemia. Electroretinogram (ERG), fluorogold retrograde labeling RGC, Western blotting, ELISA, RT-PCR, and TUNEL were utilized. In vitro, both H2O2 and OGD models significantly (p < 0.001/p < 0.001; H2O2 and OGD) induced oxidative stress/ischemic-like insults, decreasing RGC-5 cell viability (from 100% to 55.14 ± 2.19%/60.84 ± 4.57%). These injuries were insignificantly (53.85 ± 1.28% at 0.25 mM)/(63.46 ± 3.30% at 0.25 mM) and significantly (p = 0.003/p = 0.012; 64.15 ± 2.41%/77.63 ± 8.59% at 0.5 mM) altered by the pre-administration of catalpol, indicating a possible antioxidative and anti-ischemic effect of 0.5 mM catalpol. In vivo, catalpol had less effect at 0.25 mM for ERG amplitude ratio (median [Q1, Q3] 14.75% [12.64%, 20.48%]) and RGC viability (mean ± SE 63.74 ± 5.13%), whereas (p < 0.05 and p < 0.05) at 0.5 mM ERG’s ratio (35.43% [24.35%, 43.08%]) and RGC’s density (74.34 ± 5.10%) blunted the ischemia-associated significant (p < 0.05 and p < 0.01) reduction in ERG b-wave amplitude (6.89% [4.24%, 10.40%]) and RGC cell viability (45.64 ± 3.02%). Catalpol 0.5 mM also significantly protected against retinal ischemia supported by the increased amplitude ratio of ERG a-wave and oscillatory potential, along with recovering a delayed a-/b-wave response time ratio. When contrasted with DKK1 or Lucentis, catalpol exhibited similar protective effects against retinal ischemia via significantly (p < 0.05) blunting the ischemia-induced overexpression of β-catenin, VEGF, or angiopoietin-2. Moreover, ischemia-associated significant increases in apoptotic cells in the inner retina, inflammatory biomarker MCP-1, and ischemic indicator HIF-1α were significantly nullified by catalpol. Catalpol demonstrated antiapoptotic, anti-inflammatory, anti-ischemic (in vivo retinal ischemia or in vitro OGD), and antioxidative (in vitro) properties, counteracting retinal ischemia via suppressing upstream Wnt/β-catenin and inhibiting downstream HIF-1α, VEGF, and angiopoietin-2, together with its decreasing TUNEL apoptotic cell number and inflammatory MCP-1 concentration. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

24 pages, 3732 KiB  
Article
Acute Neurovascular Inflammatory Profile in Patients with Aneurysmal Subarachnoid Hemorrhage
by Ruby R. Taylor, Robert W. Keane, Begoña Guardiola, Raul Martí, Daniel Alegre, W. Dalton Dietrich, Jon Perez-Barcena and Juan Pablo de Rivero Vaccari
Biomolecules 2025, 15(5), 613; https://doi.org/10.3390/biom15050613 - 23 Apr 2025
Viewed by 744
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a life-threatening condition that results from intracranial aneurysm rupture, leading to the accumulation of blood between the arachnoid and pia mater. The blood breakdown products and damage-associated molecule patterns (DAMPs), which are released as a result of vascular [...] Read more.
Aneurysmal subarachnoid hemorrhage (aSAH) is a life-threatening condition that results from intracranial aneurysm rupture, leading to the accumulation of blood between the arachnoid and pia mater. The blood breakdown products and damage-associated molecule patterns (DAMPs), which are released as a result of vascular and cellular compromise following aneurysm rupture, elicit local endothelial reactions leading to the narrowing of cerebral arteries and ischemia. In addition, vascular inflammation, characterized by activated endothelial cells, perpetuates disruption of the neurovascular unit and the blood–brain barrier. The uncertain prognosis of aSAH patients contributes to the necessity of a fluid biomarker that can serve as a valuable adjunct to radiological and clinical evaluation. Limited studies have investigated vascular inflammation and angiogenic protein expression following aSAH. Reliable markers of the vascular inflammatory and angiogenic response associated with aSAH may allow for the earlier detection of patients at risk for complications and aid in the identification of novel pharmacologic targets. We investigated whether vascular inflammatory and angiogenesis signaling proteins may serve as potential biomarkers of aSAH. Serum and cerebrospinal fluid (CSF) from fifteen aSAH subjects and healthy age-matched controls as well as hydrocephalus (CSF) no-aneurysm controls were evaluated for levels of vascular inflammatory and angiogenesis proteins. Protein measurement was carried out using electrochemiluminescence. The area under the curve (AUC) was calculated using receiver operating characteristics (ROC) to obtain information on biomarker reliability, specificity, sensitivity, cut-off points, and likelihood ratio. In addition, patients were grouped by Glasgow Outcome Score—Extended at 3 months post-injury to determine the correlation between vascular inflammatory protein levels and clinical outcome measures. aSAH subjects demonstrated elevated vascular inflammatory protein levels in serum and CSF when compared to controls. Certain vascular injury and angiogenic proteins were found to be promising biomarkers of inflammatory response in aSAH in the CSF and serum. In particular, elevated levels of serum amyloid-alpha (SAA) were found to be correlated with unfavorable outcomes following aSAH. Determination of these protein levels in CSF and serum in aSAH may be utilized as reliable biomarkers of inflammation in aSAH and used clinically to monitor patient outcomes. Full article
Show Figures

Figure 1

Back to TopTop