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

Journals

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
remove_circle_outline

Search Results (448)

Search Parameters:
Keywords = dysregulated macrophage

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
32 pages, 1605 KB  
Review
Nanomedicine-Driven Therapeutic Strategies for Rheumatoid Arthritis-Associated Depression: Mechanisms and Pharmacological Progress
by Jiaxiang Hu, Mingqin Shi, Miao Tian, Baiqing Xie, Yi Tan, Dongxu Zhou, Tengfei Qian and Dongdong Qin
Pharmaceuticals 2026, 19(1), 94; https://doi.org/10.3390/ph19010094 - 4 Jan 2026
Viewed by 240
Abstract
Rheumatoid arthritis (RA) is frequently accompanied by depression, a comorbidity arising from the interplay of chronic systemic inflammation, neuroimmune activation, oxidative stress, and dysregulation of the gut–brain axis. Increasing evidence suggests that nanomedicine offers unique opportunities for the integrated management of RA-associated depression [...] Read more.
Rheumatoid arthritis (RA) is frequently accompanied by depression, a comorbidity arising from the interplay of chronic systemic inflammation, neuroimmune activation, oxidative stress, and dysregulation of the gut–brain axis. Increasing evidence suggests that nanomedicine offers unique opportunities for the integrated management of RA-associated depression by enabling precise modulation of both peripheral inflammation and central nervous system (CNS) pathology. This review outlines the biological mechanisms linking RA and depression—including cytokine cascades, mitochondrial dysfunction, reactive oxygen species (ROS) accumulation, and microbial metabolite imbalance—and highlights recent progress in nanocarrier platforms capable of dual-site intervention. Liposomes, polymeric nanoparticles (NPs), exosomes, inorganic nanozymes, and emerging carbon-based nanomaterials have demonstrated the ability to target inflamed synovium, reprogram macrophage phenotypes, traverse the blood–brain barrier (BBB), suppress microglial overactivation, enhance neuroplasticity, and restore gut microbial homeostasis. Furthermore, stimulus-responsive nanoplatforms activated by ROS, pH, enzymes, or hypoxia provide spatiotemporally controlled drug release, thereby improving therapeutic precision. Finally, we discuss integrative designs such as dual-targeting nanomedicines, co-delivery systems, and microbiota-modulating nano-interventions, which offer promising strategies for the comprehensive treatment of RA-associated depression. This review aims to provide mechanistic insights and design principles to guide the development of next-generation nanomedicine for coordinated systemic-central modulation in RA comorbidity. Full article
(This article belongs to the Special Issue Application of Nanotechnology in Drug Delivery)
Show Figures

Graphical abstract

24 pages, 1094 KB  
Review
The Involvement of MicroRNAs in Innate Immunity and Cystic Fibrosis Lung Disease: A Narrative Review
by Annalucia Carbone, Namra Sajid, Piera Soccio, Pasquale Tondo, Donato Lacedonia, Sante Di Gioia and Massimo Conese
Curr. Issues Mol. Biol. 2026, 48(1), 58; https://doi.org/10.3390/cimb48010058 - 2 Jan 2026
Viewed by 198
Abstract
Pulmonary involvement in cystic fibrosis (CF) is characterised by respiratory infections caused by bacteria, viruses, and fungi, as well as by dysregulated inflammatory and immune responses. Although essential for the host’s initial defence against these microorganisms, the innate immune response is altered in [...] Read more.
Pulmonary involvement in cystic fibrosis (CF) is characterised by respiratory infections caused by bacteria, viruses, and fungi, as well as by dysregulated inflammatory and immune responses. Although essential for the host’s initial defence against these microorganisms, the innate immune response is altered in its main cellular (airway epithelial cells (AECs), monocytes, macrophages, and neutrophils) and molecular (cytokines, chemokines, signal transduction pathways, and transcription factors) components. MicroRNAs (miRNAs) form a regulatory network at the level of inflammatory and immune responses, and their dysregulation has been observed in immortalised and primary CF AECs as well as in monocytes, macrophages, and neutrophils from CF patients. Although the study of individual miRNAs is helping to dissect the specific altered events in CF lung disease (CFLD), large-scale genomic and transcriptomic studies are more likely to capture its full complexity. The studies we identified suggest that miRNAs are involved in various processes related to CFLD, including impaired pathogen response, compensation for hyperinflammation, altered antigen presentation, and wound healing in AECs and macrophages. However, clinical studies involving large cohorts of patients are needed to obtain meaningful results and identify new therapeutic targets. Equally important will be the study of the miRNome as circulating biomarkers for the purposes of diagnostic and prognostic precision medicine. Full article
(This article belongs to the Special Issue Complex Molecular Mechanism of Monogenic Diseases: 3rd Edition)
Show Figures

Figure 1

22 pages, 640 KB  
Review
Unraveling Jawbone Susceptibility: Distinctive Features Underlying Medication-Related Osteonecrosis
by Balázs Paczona, József Piffkó and Ágnes Janovszky
Dent. J. 2026, 14(1), 18; https://doi.org/10.3390/dj14010018 - 1 Jan 2026
Viewed by 196
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a devastating complication arising primarily after invasive dentoalveolar procedures in patients treated with antiresorptive, antiangiogenic, or targeted therapies. Although recognized risk factors are established, the distinctive vulnerability of jawbones compared to long bones is not fully [...] Read more.
Medication-related osteonecrosis of the jaw (MRONJ) is a devastating complication arising primarily after invasive dentoalveolar procedures in patients treated with antiresorptive, antiangiogenic, or targeted therapies. Although recognized risk factors are established, the distinctive vulnerability of jawbones compared to long bones is not fully understood. This review comprehensively synthesizes recent advances regarding the embryological, anatomical, and physiological disparities that contribute to region-specific susceptibility to MRONJ. Recent evidence suggests that jawbones diverge significantly from long bones in embryonic origin, ossification pathways, vascular architecture, innervation patterns, and regenerative capacities. These differences affect bone metabolism, healing dynamics, response to pharmacologic agents, and local cellular activities, such as enhanced bisphosphonate uptake and specialized microcirculation. Experimental and clinical evidence reveals that mandibular periosteal cells exhibit superior osteogenic and angiogenic potentials, and the jaws respond differently to metabolic challenges, trauma, and medication-induced insults. Furthermore, site-specific pharmacologic and inflammatory interactions, including altered periosteal microcirculation and leukocyte–endothelial interactions, may explain the development of MRONJ, although rare cases of medication-related osteonecrosis have also been reported in long bones. Emerging research demonstrates that immune dysregulation, particularly M1 macrophage polarization with overexpression of matrix metalloproteinase-13 (MMP-13), plays a crucial role in early MRONJ development. Understanding these mechanisms highlights the critical need for region-specific preventive measures and therapeutic strategies targeting the unique biology of jawbones. This comparative perspective offers new translational insights for designing targeted interventions, developing tissue engineering solutions, and improving patient outcomes. Future research should focus on gene expression profiling and cellular responses across skeletal regions to further delineate MRONJ pathogenesis and advance personalized therapies for affected patients. Full article
(This article belongs to the Special Issue Dental Oncology)
Show Figures

Figure 1

22 pages, 1385 KB  
Review
miRNA in the Progression of Diabetic Kidney Disease: New Insight
by Zhiyue Zou, Ning Zhou and Chun Zhang
Int. J. Mol. Sci. 2026, 27(1), 420; https://doi.org/10.3390/ijms27010420 - 31 Dec 2025
Viewed by 203
Abstract
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes and a leading cause of end-stage renal disease worldwide. Despite advances in metabolic and blood pressure control, the prevalence of DKD continues to rise, creating a significant clinical and socioeconomic burden. Recent [...] Read more.
Diabetic kidney disease (DKD) is a major microvascular complication of diabetes and a leading cause of end-stage renal disease worldwide. Despite advances in metabolic and blood pressure control, the prevalence of DKD continues to rise, creating a significant clinical and socioeconomic burden. Recent studies have revealed that non-coding RNAs, particularly microRNAs (miRNAs), play an important role in the development and progression of DKD. Distinct patterns of miRNA dysregulation have been identified in specific renal cell types, including podocytes, mesangial cells, tubular epithelial cells, endothelial cells, fibroblasts, and macrophages. These alterations drive characteristic cellular injuries such as podocyte loss, mesangial matrix expansion, tubular epithelial–mesenchymal transition, endothelial dysfunction, and interstitial fibrosis. Certain miRNAs, such as miR-21, miR-192, and miR-214, reinforce profibrotic TGF-β/Smad signaling, whereas protective groups, including the miR-29 and miR-30 families, maintain epithelial stability and restrict matrix deposition. Beyond their regulatory roles, circulating and urinary miRNAs have emerged as stable, non-invasive biomarkers that reflect renal injury and disease progression. This review summarizes recent progress in elucidating cell-specific miRNA networks in DKD and highlights their potential as diagnostic indicators and therapeutic targets for precision management of diabetic kidney disease. Full article
(This article belongs to the Special Issue mRNA/miRNAs Network in Diabetes and Its Complications)
Show Figures

Figure 1

19 pages, 2085 KB  
Review
T-Cell-Driven Immunopathology and Fibrotic Remodeling in Hypertrophic Cardiomyopathy: A Translational Scoping Review
by Antonio da Silva Menezes Junior, Henrique Lima de Oliveira, Khissya Beatryz Alves de Lima, Silvia Marçal Botelho and Isabela Jubé Wastowski
Cells 2026, 15(1), 61; https://doi.org/10.3390/cells15010061 - 29 Dec 2025
Viewed by 271
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is increasingly recognized as a disorder shaped not only by sarcomeric mutations but also by complex immunogenetic and metabolic interactions. Emerging transcriptomic and single-cell analyses implicate immune dysregulation, RNA methylation, and necroptosis as critical modulators of myocardial remodeling. [...] Read more.
Background: Hypertrophic cardiomyopathy (HCM) is increasingly recognized as a disorder shaped not only by sarcomeric mutations but also by complex immunogenetic and metabolic interactions. Emerging transcriptomic and single-cell analyses implicate immune dysregulation, RNA methylation, and necroptosis as critical modulators of myocardial remodeling. Objectives: This scoping review synthesizes bioinformatic, transcriptomic, and experimental data to delineate the immunogenetic architecture of HCM and identify candidate molecular targets for immune–metabolic modulation. Methods: Following Joanna Briggs Institute and PRISMA-ScR guidelines, we systematically searched PubMed, Embase, Web of Science, and GEO through September 2025 for studies evaluating immune infiltration, RNA regulation, and necroptosis in human HCM. Data were narratively synthesized across histologic, clinical, and multi-omics domains. Results: Among 8191 screened records, 25 studies met the inclusion criteria. Key immune–epigenetic regulators included the lncRNA–mRNA pair MIR210HG–BPIFC, m6A readers IGFBP3 and YTHDC1, and necroptosis gene JAK2. The HCM myocardium exhibited the depletion of reparative M2 macrophages and Tregs; enrichment of cytotoxic CD8+ T cells; and activation of the TNFα–NFκB, IL-6–JAK–STAT3, and PI3K–Akt pathways. Machine learning biomarkers (RASD1, FCN3, and PIK3R1) exhibited diagnostic accuracy (AUC > 0.85). Drug target predictions identified ruxolitinib and celecoxib as potential immunometabolic modulators (agents predicted to modulate both immune and metabolic pathways based on gene expression signatures). Conclusions: These findings support a hypothesis that HCM may involve immunogenetic mechanisms, rather than being exclusively sarcomeric in nature, although this remains to be validated. The integration of molecular and imaging biomarkers may enable precision immunotherapy, redefining HCM from a structural cardiomyopathy to a biologically stratified condition. Full article
Show Figures

Graphical abstract

25 pages, 1727 KB  
Review
Macrophage Plasticity and Regulatory Networks During the Transition from Inflammation to Fibrosis in the Kidney
by Yehun Moon, Jintaek Hong, Jinwoo Chung and Jea-Hyun Baek
Life 2026, 16(1), 52; https://doi.org/10.3390/life16010052 - 29 Dec 2025
Viewed by 399
Abstract
Kidney fibrosis represents the final common pathway of nearly all progressive renal diseases, linking acute kidney injury (AKI) and chronic kidney disease (CKD) through a maladaptive repair process. Regardless of etiology, persistent inflammation and excessive extracellular matrix (ECM) deposition drive irreversible structural distortion [...] Read more.
Kidney fibrosis represents the final common pathway of nearly all progressive renal diseases, linking acute kidney injury (AKI) and chronic kidney disease (CKD) through a maladaptive repair process. Regardless of etiology, persistent inflammation and excessive extracellular matrix (ECM) deposition drive irreversible structural distortion and functional decline in the kidney. Among cellular mediators, macrophages occupy a central role across the continuum from acute injury to fibrosis, orchestrating both tissue injury and repair through dynamic transitions between pro-inflammatory (M1) and pro-fibrotic (M2) states in response to local cues. Here, we synthesize macrophage-driven mechanisms of renal fibrosis, emphasizing recruitment, infiltration, and local proliferation mediated by chemokine–receptor networks and mechanosensitive ion channels. In addition, in this review paper, we provide an overview on the dual roles of macrophages in acute inflammation and chronic remodeling through key cytokine signaling pathways (TLR4/NF-κB, IL-4/STAT6, TGF-β/Smad, IL-10/STAT3), highlighting how metabolic reprogramming, mechanochemical feedback via Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) signaling, and epigenetic modulators collectively stabilize the fibrotic macrophage phenotype. Also, emerging insights into mitochondrial dysfunction, succinate–succinate receptor 1 (SUCNR1) signaling, and autophagy dysregulation reveal the metabolic basis of macrophage persistence in fibrotic kidneys. Understanding these multilayered regulatory circuits offers a framework for therapeutic strategies that selectively target macrophage-dependent fibrogenesis to halt the transition from acute injury to chronic renal failure. Full article
(This article belongs to the Special Issue New Insights into Cellular Inflammation and Regeneration)
Show Figures

Figure 1

18 pages, 570 KB  
Review
The Central Role of Macrophages in Long COVID Pathophysiology
by Philip Mcmillan, Anthony J. Turner and Bruce D. Uhal
Int. J. Mol. Sci. 2026, 27(1), 313; https://doi.org/10.3390/ijms27010313 - 27 Dec 2025
Viewed by 1126
Abstract
This review article attempts to provide a unifying hypothesis to explain the myriad of symptoms and predispositions underlying the development of PASC (Postacute Sequelae of COVID), often referred to as Long COVID. The hypothesis described here proposes that Long COVID is best understood [...] Read more.
This review article attempts to provide a unifying hypothesis to explain the myriad of symptoms and predispositions underlying the development of PASC (Postacute Sequelae of COVID), often referred to as Long COVID. The hypothesis described here proposes that Long COVID is best understood as a disorder of persistent immune dysregulation, with chronic macrophage activation representing the fundamental underlying pathophysiology. Unlike transient post-viral syndromes, Long COVID involves a sustained innate immune response, particularly within monocyte-derived macrophages, driven by persistent spike protein (peripherally in MAIT cells and centrally in Microglial cells), epigenetic imprinting, and gut-related viral reservoirs. These macrophages are not merely activated temporarily but also become epigenetically “trained” into a prolonged inflammatory state, as demonstrated by enduring histone acetylation markers such as H3K27acDNA Reprogramming. It is proposed that recognizing macrophage activation as the central axis of Long COVID pathology offers a framework for personalized risk assessment, targeted intervention, and therapeutic recalibration. Full article
(This article belongs to the Special Issue Host Responses to Virus Infection)
Show Figures

Figure 1

32 pages, 4225 KB  
Article
Dysregulated Efferocytosis in CAD: TNF-α and TGF-β Silencing Reveals Functional Divergence in M1 and M2 Macrophages
by Fatin Najiah Mohd Idrus, Hayat Asaad Hameed Al-Ali, Zahidah Nasuha Mohd Yasin, Maryam Azlan, Rapeah Suppian, See Too Wei Cun, Hoe Chee Hock, Nurul Shuhadah Ahmad, Zurkurnai Yusof, Wan Yus Haniff Wan Isa, Akbar Ali Mohamed Ali and Yvonne-Tee Get Bee
Immuno 2026, 6(1), 6; https://doi.org/10.3390/immuno6010006 - 26 Dec 2025
Viewed by 257
Abstract
Efferocytosis, the process by which macrophages clear apoptotic cells, plays a vital role in maintaining immune homeostasis. This study explores the influence of inflammatory cytokines—tumor necrosis factor-alpha (TNF-α) and transforming growth factor-beta (TGF-β)—on efferocytosis dysregulation in coronary artery disease (CAD). Peripheral blood samples [...] Read more.
Efferocytosis, the process by which macrophages clear apoptotic cells, plays a vital role in maintaining immune homeostasis. This study explores the influence of inflammatory cytokines—tumor necrosis factor-alpha (TNF-α) and transforming growth factor-beta (TGF-β)—on efferocytosis dysregulation in coronary artery disease (CAD). Peripheral blood samples were collected from 27 non-obstructive and 29 obstructive CAD patients to isolate monocytes, which were then differentiated into M1 and M2 macrophages using specific cytokine stimuli. These macrophages were transfected with TNF-α and TGF-β siRNA to assess cytokine impact on efferocytosis. Expression levels of the efferocytosis receptor MERTK and its regulatory protease ADAM17 were quantified via qPCR. Statistical analysis revealed significantly higher MERTK expression in M2 macrophages compared to M1 (p = 0.002). Notably, TNF-α silencing enhanced efferocytosis in M2 macrophages, with increased clearance of early apoptotic bodies in non-obstructive CAD and late apoptotic bodies in obstructive CAD (both p < 0.001). These findings suggest that macrophage phenotype, apoptotic stage, and cytokine environment influence efferocytosis efficiency and may involve pathways beyond MERTK-ADAM17. They offer preliminary mechanistic insights into cytokine-mediated modulation of efferocytosis in CAD. Further in vivo studies are needed to confirm these observations and evaluate their relevance for future therapeutic strategies. Full article
(This article belongs to the Section Innate Immunity and Inflammation)
Show Figures

Figure 1

42 pages, 6458 KB  
Review
Clonal Hematopoiesis of Indeterminate Potential and Cardiometabolic Disease: Challenges, Controversies and Future Perspectives
by Ioanna A. Anastasiou, Dimitris Kounatidis, Natalia G. Vallianou, Eleni Rebelos, Irene Karampela and Maria Dalamaga
Int. J. Mol. Sci. 2026, 27(1), 233; https://doi.org/10.3390/ijms27010233 - 25 Dec 2025
Viewed by 517
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is defined by the expansion of hematopoietic stem cells harboring leukemogenic mutations in the absence of overt malignancy. Strongly associated with advancing age, CHIP is detected by next-generation sequencing of peripheral blood in more than 20% of [...] Read more.
Clonal hematopoiesis of indeterminate potential (CHIP) is defined by the expansion of hematopoietic stem cells harboring leukemogenic mutations in the absence of overt malignancy. Strongly associated with advancing age, CHIP is detected by next-generation sequencing of peripheral blood in more than 20% of individuals over 80, most commonly through mutations in DNMT3A, TET2, ASXL1, and PPM1D. While CHIP confers over a four-fold increased risk of hematologic malignancy, it has recently emerged as a key determinant of cardiometabolic health. Epidemiological data indicated a 40% higher cardiovascular disease (CVD) risk events and a 34% increase in all-cause mortality among CHIP carriers, with specific mutations and larger clone sizes conferring greater cardiovascular burden. Preclinical studies have shown that macrophages deficient in TET2 or DNMT3A drive interleukin (IL)-1β/IL-6 inflammasome activation, thereby promoting atherosclerosis and metabolic dysfunction, whereas the JAK2V617F mutation accelerates thrombosis. CHIP integrates into a broader network of dysregulation encompassing adiposity and inflammaging, which underlies its association with diverse comorbidities, including type 2 diabetes (T2D), chronic kidney disease (CKD), and chronic obstructive pulmonary disease (COPD). Multi-omics approaches have identified epigenetic and proteomic signatures correlated with CHIP expansion, providing potential biomarkers for risk stratification. Despite growing evidence of its systemic impact, CHIP screening remains limited to research settings. Emerging therapeutic strategies, including inflammasome inhibition, STING modulation, and epigenetic restoration, highlight its potential as a modifiable risk factor. This narrative review synthesizes current epidemiological, mechanistic, and translational insights, framing CHIP as an emerging causal factor in cardiometabolic disease and as a promising target for precision medicine in aging populations. Full article
Show Figures

Figure 1

22 pages, 1501 KB  
Review
The Convergent Immunopathogenesis of Cigarette Smoke Exposure: From Oxidative Stress to Epigenetic Reprogramming in Chronic Disease
by Aysen Kutan Fenercioglu, Hafize Uzun and Durisehvar Ozer Unal
Int. J. Mol. Sci. 2026, 27(1), 187; https://doi.org/10.3390/ijms27010187 - 24 Dec 2025
Viewed by 289
Abstract
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid [...] Read more.
Cigarette smoking is the leading preventable cause of chronic diseases (e.g., COPD, cardiovascular disease, cancer), largely driven by persistent immune-inflammatory mechanisms. This review synthesizes the molecular and cellular cascades linking cigarette smoke (CS) exposure to chronic pathology. CS constituents, particularly ROS/RNS, induce rapid oxidative stress that overwhelms antioxidant defenses and generates damage-associated molecular patterns (DAMPs). These DAMPs activate pattern recognition receptors (PRRs) and the NLRP3 inflammasome, initiating NF-κB signaling and the release of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). CS exposure causes profound innate immune dysregulation, including airway epithelial barrier disruption, hyperactivated neutrophils, and dysfunctional alveolar macrophages (AMs) that release destructive proteases (e.g., MMP-12) and acquire foam-cell–like characteristics. Furthermore, CS drives adaptive immunity toward a Th1/Th17-dominant phenotype while suppressing regulatory T-cell (Treg) function, thereby promoting autoimmunity and chronic tissue injury. Critically, CS induces epigenetic reprogramming (e.g., DNA methylation, miRNA dysregulation), locking immune cells into a persistent pro-inflammatory state. This convergence of oxidative stress, innate and adaptive immune dysregulation, and epigenetic alterations underlies the systemic low-grade inflammation that fuels smoking-related chronic diseases, highlighting key targets for novel therapeutic interventions. Full article
Show Figures

Graphical abstract

51 pages, 2000 KB  
Review
Leptin as a Potential Modifier of Neuroinflammation: Contrasting Roles in Alzheimer’s Disease and Multiple Sclerosis
by Naghmeh Abbasi Kasbi, Barbara Elena Stopschinski, Alanna Gabrielle Polyak, Agastya Reddy Malladi, Navid Manouchehri, Philipp E. Scherer and Olaf Stuve
Int. J. Mol. Sci. 2026, 27(1), 168; https://doi.org/10.3390/ijms27010168 - 23 Dec 2025
Viewed by 397
Abstract
The neuroendocrine and immune systems interact bidirectionally through shared ligands and receptors during inflammation, thereby regulating immune responses. Leptin, primarily known for its role in energy metabolism and appetite regulation, also modulates neuroinflammatory pathways. Its receptors are widely expressed on immune cells and [...] Read more.
The neuroendocrine and immune systems interact bidirectionally through shared ligands and receptors during inflammation, thereby regulating immune responses. Leptin, primarily known for its role in energy metabolism and appetite regulation, also modulates neuroinflammatory pathways. Its receptors are widely expressed on immune cells and contribute to immune mechanisms implicated in the pathogenesis of neuroinflammatory disorders such as multiple sclerosis (MS) and Alzheimer’s disease (AD). This review highlights recent advances in understanding leptin’s role in immune regulation, with a focus on its impact on MS and AD. A comprehensive literature review was conducted until October 2025, using PubMed, Google Scholar, and Scopus to identify studies investigating leptin in neuroinflammatory conditions, particularly MS and AD. Leptin exerts broad immunomodulatory effects by activating T cells, dendritic cells, and microglia, and promoting their proliferation and phagocytosis. Its elevation enhances Th1 and Th17 responses, drives pro-inflammatory macrophage phenotype polarization, and suppresses regulatory T cell and Th2 responses, immune pathways involved in MS. Peripheral leptin levels are increased in MS, especially during disease exacerbations. In contrast, in AD, they are typically reduced, particularly in patients with normal body mass index (BMI), where their decline contributes to amyloid-β and tau pathology. These divergent patterns position leptin as a bidirectional regulator at the intersection of immunity and neurodegeneration. Additionally, its protective or detrimental effects likely depend on whether it acts under physiological conditions or in the context of obesity-induced leptin resistance. Elevated leptin levels in obesity exacerbate inflammation and diminish its neuroprotective effects. In conclusion, leptin is elevated in MS patients but downregulated in AD, reflecting its bidirectional effects. In leptin resistance, peripheral proinflammatory signaling is maintained while central leptin signaling is restricted, thereby potentially promoting autoimmunity in MS and limiting neuroprotection in AD. Further mechanistic and longitudinal studies are needed to clarify the relationship between leptin dysregulation, leptin resistance, neuroinflammatory and neurodegenerative diseases. Full article
(This article belongs to the Special Issue Molecular Research and Treatment in Multiple Sclerosis)
Show Figures

Figure 1

19 pages, 3250 KB  
Article
Integrative Multi-Omics and Machine Learning Reveal Shared Biomarkers in Type 2 Diabetes and Atherosclerosis
by Qingjie Wu, Zhaochu Wang, Mengzhen Fan, Linglun Hao, Jicheng Chen, Changwen Wu and Bizhen Gao
Int. J. Mol. Sci. 2026, 27(1), 136; https://doi.org/10.3390/ijms27010136 - 22 Dec 2025
Viewed by 338
Abstract
Atherosclerosis (AS) is a leading cause of death and disability in type 2 diabetes mellitus (T2DM). However, the shared molecular mechanisms linking T2DM and atherosclerosis have not been fully elucidated. We analyzed AS- and T2DM-related gene expression profiles from the Gene Expression Omnibus [...] Read more.
Atherosclerosis (AS) is a leading cause of death and disability in type 2 diabetes mellitus (T2DM). However, the shared molecular mechanisms linking T2DM and atherosclerosis have not been fully elucidated. We analyzed AS- and T2DM-related gene expression profiles from the Gene Expression Omnibus (GEO) database to identify overlapping differentially expressed genes and co-expression signatures. Functional enrichment (Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG)) and protein–protein interaction (PPI) network analyses were then used to describe the pathways and interaction modules associated with these shared signatures, We next applied the cytoHubba algorithm together with several machine learning methods to prioritize hub genes and evaluate their diagnostic potential and combined CIBERSORT-based immune cell infiltration analysis with single-cell RNA sequencing data to examine cell types and the expression patterns of the shared genes in specific cell populations. We identified 72 shared feature genes. Functional enrichment analysis of these genes revealed significant enrichment of inflammatory- and metabolism-related pathways. Three genes—IL1B, MMP9, and P2RY13—emerged as shared hub genes and yielded robust ANN-based predictive performance across datasets. Immune deconvolution and single-cell analyses consistently indicated inflammatory amplification and an imbalance of macrophage polarization in both conditions. Biology mapped to the hubs suggests IL1B drives inflammatory signaling, MMP9 reflects extracellular-matrix remodeling, and P2RY13 implicates cholesterol transport. Collectively, these findings indicate that T2DM and AS converge on immune and inflammatory processes with macrophage dysregulation as a central axis; IL1B, MMP9, and P2RY13 represent potential biomarkers and therapeutic targets and may influence disease progression by regulating macrophage states, supporting translational application to diagnosis and treatment of T2DM-related atherosclerosis. These findings are preliminary. Further experimental and clinical studies are needed to confirm their validity, given the limitations of the present study. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Figure 1

19 pages, 7444 KB  
Article
Integrating Bioinformatics and Experimental Validation Identifies SCD as a Ferroptosis-Related Immune Regulator and Therapeutic Target in Crohn’s Disease
by Jingtong Wu, Lixiang Zhou, Dongmei Qiu, Tao Wei, Chaohui Xie, Ermei Chen, Mengjie Lin and Yanyun Fan
Int. J. Mol. Sci. 2026, 27(1), 19; https://doi.org/10.3390/ijms27010019 - 19 Dec 2025
Viewed by 188
Abstract
This study investigates the role of ferroptosis-related genes (FRGs) in the intestinal inflammation of Crohn’s disease (CD). Through integrated bioinformatics and experimental validation, we identified differentially expressed genes from RNA-seq data and intersected them with known FRGs to obtain ferroptosis-related differentially expressed genes [...] Read more.
This study investigates the role of ferroptosis-related genes (FRGs) in the intestinal inflammation of Crohn’s disease (CD). Through integrated bioinformatics and experimental validation, we identified differentially expressed genes from RNA-seq data and intersected them with known FRGs to obtain ferroptosis-related differentially expressed genes (FEDGs). Functional enrichment and immune infiltration analyses were performed, and seven hub FEDGs were selected using machine learning. A diagnostic model based on these genes showed strong predictive ability. Immune analysis revealed significant associations with macrophages, neutrophils, dendritic cells, and CD4+ T cells. Protein expression of key hub genes was validated in clinical CD samples and a DSS-induced colitis model. Importantly, localized inhibition of SCD alleviated disease severity in experimental colitis. These findings highlight the involvement of ferroptosis in CD immune dysregulation and propose SCD as a potential therapeutic target. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Figure 1

13 pages, 52330 KB  
Article
Obesity Promotes Renal Inflammation and Fibrosis Independent of Sex in SS Leptin Receptor Mutant (SSLepR) Rats
by Karim M. Saad, Mohamed S. Gad, Jocelyn Tang, Kim Capehart, Rafik Abdelsayed, Jan M. Williams and Ahmed A. Elmarakby
Biomedicines 2025, 13(12), 3105; https://doi.org/10.3390/biomedicines13123105 - 17 Dec 2025
Viewed by 344
Abstract
Background: Obesity is a major contributor to chronic kidney disease (CKD) through mechanisms involving inflammation and metabolic dysregulation. Premenopausal female rats are known to be protected from cardiovascular disorders vs. age matched male rats. The current study investigates if there are sex [...] Read more.
Background: Obesity is a major contributor to chronic kidney disease (CKD) through mechanisms involving inflammation and metabolic dysregulation. Premenopausal female rats are known to be protected from cardiovascular disorders vs. age matched male rats. The current study investigates if there are sex differences in obesity-induced renal inflammation in SS leptin receptor mutant (SSLepR mutant) rats as a model of metabolic syndrome. Method: Male and female lean and obese SSLepR mutant rats were used in the current study to assess changes in metabolic parameters and markers of renal inflammation. Results: Obese SSLepR rats showed significant increases in body weight, hemoglobin A1c (HbA1c), and cholesterol vs. lean control, although their blood glucose levels remained comparable to lean rats. Plasma leptin, insulin, and TNF-α converting enzyme (TACE) levels were significantly elevated in obese SSLepR rats vs. lean control rats, with no apparent sex differences. Obesity was associated with an elevation in renal injury since protein and albumin excretion levels were significantly elevated in obese SSLepR rats vs. lean control rats, with no apparent sex differences. The elevation in renal injury was associated with increased renal fibrosis as evidenced by increased collagen deposition and TGF-β expression in the kidney of obese SSLepR rats vs. lean control rats. Increased renal fibrosis also coincided with increased renal inflammation and apoptosis as evidenced by increased macrophage infiltration and IL-6 expression in the kidneys of obese SSLepR rats vs. lean control rats. Conclusion: These findings indicate that obesity triggers renal inflammation and fibrosis independent of hyperglycemia in SSLepR rats, and these changes may override sex-based protective effects seen in females in other experimental rodent models of cardiovascular diseases. Full article
Show Figures

Figure 1

20 pages, 3953 KB  
Article
Sequential Dengue Virus Infection in Marmosets: Histopathological and Immune Responses in the Liver
by Daniele Freitas Henriques, Livia M. N. Casseb, Milene S. Ferreira, Larissa S. Freitas, Hellen T. Fuzii, Carla Pagliari, Luciane Kanashiro, Paulo H. G. Castro, Gilmara A. Siva, Orlando Pereira Amador Neto, Valter M. Campos, Beatriz C. Belvis, Flavia B. dos Santos, Lilian R. M. de Sá and Pedro Fernando da Costa Vasconcelos
Viruses 2025, 17(12), 1619; https://doi.org/10.3390/v17121619 - 15 Dec 2025
Viewed by 314
Abstract
This study evaluated hepatic pathological and phenotypic alterations, along with the inflammatory response, following sequential dengue virus (DENV) infection in Callithrix penicillata, a relevant model for human endemic scenarios. Twenty-six animals were initially infected subcutaneously with DENV-3. Thirteen were euthanized between 1 and [...] Read more.
This study evaluated hepatic pathological and phenotypic alterations, along with the inflammatory response, following sequential dengue virus (DENV) infection in Callithrix penicillata, a relevant model for human endemic scenarios. Twenty-six animals were initially infected subcutaneously with DENV-3. Thirteen were euthanized between 1 and 7 days post-infection (dpi) to assess the acute phase, and up to 60 dpi for the convalescent phase. The remaining animals received a secondary DENV-2 infection two months later. Liver samples underwent histopathological and immunohistochemical analysis. Viral antigens were identified in hepatocytes, Kupffer cells, and Councilman bodies. Observed liver changes included apoptosis, lytic necrosis, midzonal inflammation, Kupffer cell hyperplasia and hypertrophy, sinusoidal dilation, and hemosiderin deposition. Both primary and secondary infections increased activated macrophages, NK cells, S-100 protein, and B lymphocytes. Primary infection was associated with elevated CD4+ T cells, IFN-γ, TGF-β, IL-10, and Fas expression, whereas secondary infection induced higher IFN-γ, TNF-α, IL-8, Fas, and VCAM levels. These findings mirror hepatic alterations in severe human dengue cases and underscore the role of direct viral effects and immune dysregulation in liver injury. The results support C. penicillata as a suitable non-human primate model for studying DENV pathogenesis. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

Back to TopTop