Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (120)

Search Parameters:
Keywords = immune-mediated neurological disorders

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 2511 KiB  
Article
Depression, Anxiety, and MSQOL-54 Outcomes in RRMS Patients Receiving Fingolimod or Cladribine: A Cross-Sectional Comparative Study
by Müttalip Özbek, Adalet Arıkanoğlu and Mehmet Ufuk Aluçlu
Medicina 2025, 61(8), 1409; https://doi.org/10.3390/medicina61081409 - 3 Aug 2025
Viewed by 190
Abstract
Background and Objectives: Multiple sclerosis (MS) is a chronic immune-mediated neurological disorder that primarily affects young adults and is frequently accompanied by psychiatric comorbidities such as depression and anxiety, both of which significantly diminish patients’ quality of life (QoL). This study investigated [...] Read more.
Background and Objectives: Multiple sclerosis (MS) is a chronic immune-mediated neurological disorder that primarily affects young adults and is frequently accompanied by psychiatric comorbidities such as depression and anxiety, both of which significantly diminish patients’ quality of life (QoL). This study investigated the effect of two oral disease-modifying therapies (DMTs), fingolimod and cladribine, on mental health and QoL in patients with relapsing-remitting MS (RRMS). The aim of the study was to compare levels of depression, anxiety, and health-related quality of life (HRQoL) in RRMS patients treated with fingolimod or cladribine, and to evaluate their associations with clinical and radiological parameters. Materials and Methods: Eighty RRMS patients aged 18 to 50 years with Expanded Disability Status Scale (EDSS) scores of 3.0 or less, no recent disease relapse, and no history of antidepressant use were enrolled. Forty patients were treated with fingolimod and forty with cladribine. Depression and anxiety were assessed using the Hamilton Depression Rating Scale (HDRS) and the Hamilton Anxiety Rating Scale (HARS). QoL was evaluated using the Multiple Sclerosis QoL-54 (MSQOL-54) instrument. Additional clinical data, including MRI-based lesion burden, EDSS scores, age, disease duration, and occupational status, were collected. Results: No statistically significant differences were observed between the two groups regarding HDRS and HARS scores (p > 0.05). However, patients treated with fingolimod had significantly higher scores in the Energy/Fatigue subdomain (7.55 ± 2.02 vs. 6.56 ± 2.57, p = 0.046) and Composite Mental Health (CMH) score (64.73 ± 15.01 vs. 56.00 ± 18.93, p = 0.029) compared to those treated with cladribine. No significant differences were found in the independent items of the MSQOL-54. A negative correlation was identified between total lesion load and QoL scores. Conclusions: Although fingolimod and cladribine exert comparable effects on depression and anxiety levels, fingolimod may be associated with better mental health outcomes and reduced fatigue in RRMS patients. Furthermore, lesion burden and clinical parameters such as age and EDSS score may independently influence QoL, regardless of the DMT used. Full article
Show Figures

Figure 1

16 pages, 956 KiB  
Review
The Potential Therapeutic Role of Bruton Tyrosine Kinase Inhibition in Neurodegenerative Diseases
by Francesco D’Egidio, Housem Kacem, Giorgia Lombardozzi, Michele d’Angelo, Annamaria Cimini and Vanessa Castelli
Appl. Sci. 2025, 15(15), 8239; https://doi.org/10.3390/app15158239 - 24 Jul 2025
Viewed by 266
Abstract
Bruton Tyrosine Kinase (BTK) has emerged as a critical mediator in the pathophysiology of neuroinflammation associated with neurodegenerative diseases. BTK, a non-receptor tyrosine kinase predominantly expressed in cells of the hematopoietic lineage, modulates B-cell receptor signaling and innate immune responses, including microglial activation. [...] Read more.
Bruton Tyrosine Kinase (BTK) has emerged as a critical mediator in the pathophysiology of neuroinflammation associated with neurodegenerative diseases. BTK, a non-receptor tyrosine kinase predominantly expressed in cells of the hematopoietic lineage, modulates B-cell receptor signaling and innate immune responses, including microglial activation. Recent evidence implicates aberrant BTK signaling in the exacerbation of neuroinflammatory cascades contributing to neuronal damage in disorders such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, ischemic stroke, and Huntington’s disease. Pharmacological inhibition of BTK has shown promise in attenuating microglial-mediated neurotoxicity, reducing pro-inflammatory cytokine release, and promoting neuroprotection in preclinical models. BTK inhibitors, originally developed for hematological malignancies, demonstrate favorable blood–brain barrier penetration and immunomodulatory effects relevant to central nervous system pathology. This therapeutic approach may counteract detrimental neuroimmune interactions without broadly suppressing systemic immunity, thus preserving host defense. Ongoing clinical trials are evaluating the safety and efficacy of BTK inhibitors in patients with neurodegenerative conditions, with preliminary results indicating potential benefits in slowing disease progression and improving neurological outcomes. This review consolidates current knowledge on BTK signaling in neurodegeneration and highlights the rationale for BTK inhibition as a novel, targeted therapeutic strategy to modulate neuroinflammation and mitigate neurodegenerative processes. Full article
(This article belongs to the Section Applied Biosciences and Bioengineering)
Show Figures

Figure 1

35 pages, 2933 KiB  
Review
NEU1-Mediated Extracellular Vesicle Glycosylation in Alzheimer’s Disease: Mechanistic Insights into Intercellular Communication and Therapeutic Targeting
by Mohd Adnan, Arif Jamal Siddiqui, Fevzi Bardakci, Malvi Surti, Riadh Badraoui and Mitesh Patel
Pharmaceuticals 2025, 18(6), 921; https://doi.org/10.3390/ph18060921 - 19 Jun 2025
Viewed by 683
Abstract
Alzheimer’s disease (AD), a progressive neurodegenerative disorder, is marked by the pathological accumulation of amyloid-β plaques and tau neurofibrillary tangles, both of which disrupt neuronal communication and function. Emerging evidence highlights the role of extracellular vesicles (EVs) as key mediators of intercellular communication, [...] Read more.
Alzheimer’s disease (AD), a progressive neurodegenerative disorder, is marked by the pathological accumulation of amyloid-β plaques and tau neurofibrillary tangles, both of which disrupt neuronal communication and function. Emerging evidence highlights the role of extracellular vesicles (EVs) as key mediators of intercellular communication, particularly in the propagation of pathological proteins in AD. Among the regulatory factors influencing EV composition and function, neuraminidase 1 (NEU1), a lysosomal sialidase responsible for desialylating glycoproteins has gained attention for its involvement in EV glycosylation. This review explores the role of NEU1 in modulating EV glycosylation, with particular emphasis on its influence on immune modulation and intracellular trafficking pathways and the subsequent impact on intercellular signaling and neurodegenerative progression. Altered NEU1 activity has been associated with abnormal glycan profiles on EVs, which may facilitate the enhanced spread of amyloid-β and tau proteins across neural networks. By regulating glycosylation, NEU1 influences EV stability, targeting and uptake by recipient cells, primarily through the desialylation of surface glycoproteins and glycolipids, which alters the EV charge, recognition and receptor-mediated interactions. Targeting NEU1 offers a promising therapeutic avenue to restore EV homeostasis and reduces pathological protein dissemination. However, challenges persist in developing selective NEU1 inhibitors and effective delivery methods to the brain. Furthermore, altered EV glycosylation patterns may serve as potential biomarkers for early AD diagnosis and monitoring. Overall, this review highlights the importance of NEU1 in AD pathogenesis and advocates for deeper investigation into its regulatory functions, with the aim of advancing therapeutic strategies and biomarker development for AD and related neurological disabilities. Full article
(This article belongs to the Special Issue Pharmacotherapy for Alzheimer’s Disease)
Show Figures

Graphical abstract

28 pages, 2140 KiB  
Review
Mammalian PI-Phospholipase C Isozymes: Structural and Functional Insights and Roles in Health and Disease
by May Hamdi, Mohammed Al-Matwi, Nour Elghoul, Hissa Al-Kuwari, Tahseen S. Sayed, Emna Riguene and Michail Nomikos
Medicina 2025, 61(6), 1054; https://doi.org/10.3390/medicina61061054 - 7 Jun 2025
Viewed by 1061
Abstract
The Phosphoinositide Specific-Phospholipase C (PI-PLC) family of enzymes plays a crucial role in various cellular processes by catalyzing the hydrolysis of phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which are essential messengers mediating critical intracellular signaling [...] Read more.
The Phosphoinositide Specific-Phospholipase C (PI-PLC) family of enzymes plays a crucial role in various cellular processes by catalyzing the hydrolysis of phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which are essential messengers mediating critical intracellular signaling pathways. Herein, we carry out a comprehensive analysis of the structure, function, regulation, and implications of the PI-PLC family enzymes in both physiological and pathological contexts. More specifically, we discuss the structural features of PI-PLCs, elucidating their conserved domains and catalytic mechanisms. Furthermore, we explore the multifaceted roles of PI-PLCs in signal transduction, cellular homeostasis, and membrane dynamics, whilst highlighting the intricate regulatory mechanisms governing their activity such as protein–protein interactions, post-translational modifications, and lipid modulation. Lastly, we assess the involvement of PI-PLCs in various diseases, such as cancer, neurological disorders, immune dysregulation, and male infertility, emphasizing their potential as therapeutic targets. Full article
(This article belongs to the Section Genetics and Molecular Medicine)
Show Figures

Figure 1

24 pages, 393 KiB  
Review
Faecal Microbiota Transplantation as an Adjuvant Treatment for Extraintestinal Disorders: Translating Insights from Human Medicine to Veterinary Practice
by Alice Nishigaki, Julian R. Marchesi and Renato L. Previdelli
Vet. Sci. 2025, 12(6), 541; https://doi.org/10.3390/vetsci12060541 - 3 Jun 2025
Viewed by 2626
Abstract
Faecal microbiota transplantation (FMT) has emerged as a transformative therapy in human medicine, particularly for managing recurrent Clostridioides difficile infections and other gastrointestinal (GI) disorders. Beyond the GI tract, FMT has shown potential in addressing extraintestinal conditions in people, including metabolic, immune-mediated, dermatological, [...] Read more.
Faecal microbiota transplantation (FMT) has emerged as a transformative therapy in human medicine, particularly for managing recurrent Clostridioides difficile infections and other gastrointestinal (GI) disorders. Beyond the GI tract, FMT has shown potential in addressing extraintestinal conditions in people, including metabolic, immune-mediated, dermatological, neurological, and infectious diseases. Research in people has highlighted its efficacy in decolonising multidrug-resistant organisms in infection, mitigating autoimmune diseases, and improving outcomes in metabolic disorders such as obesity and diabetes. Furthermore, FMT has also been linked to enhanced responses to immunotherapy in cancer and improved management of hepatic and renal conditions. These findings underscore the intricate connections between the gut microbiome and systemic health, opening novel therapeutic avenues. In veterinary medicine, while FMT has demonstrated benefits for GI disorders, its application in extraintestinal diseases remains largely unexplored. Emerging evidence suggests that conditions such as atopic dermatitis, chronic kidney disease, immune-mediated diseases, and behavioural disorders in companion animals could benefit from microbiome-targeted therapies. However, significant gaps in knowledge persist, particularly regarding the long-term safety and efficacy for veterinary applications. This review synthesises findings from human medicine to assess their relevance for veterinary applications and future research. Full article
(This article belongs to the Section Veterinary Biomedical Sciences)
Show Figures

Figure 1

54 pages, 904 KiB  
Review
Gut Feelings: How Microbes, Diet, and Host Immunity Shape Disease
by Benjamin Frederick Theis, Jun Sung Park, Jong Sung Anthony Kim, Sareh Zeydabadinejad, Matam Vijay-Kumar, Beng San Yeoh and Piu Saha
Biomedicines 2025, 13(6), 1357; https://doi.org/10.3390/biomedicines13061357 - 31 May 2025
Viewed by 1529
Abstract
The human gut microbiome is intricately linked to systemic and organ-specific immune responses and is highly responsive to dietary modulation. As metagenomic techniques enable in-depth study of an ever-growing number of gut microbial species, it has become increasingly feasible to decipher the specific [...] Read more.
The human gut microbiome is intricately linked to systemic and organ-specific immune responses and is highly responsive to dietary modulation. As metagenomic techniques enable in-depth study of an ever-growing number of gut microbial species, it has become increasingly feasible to decipher the specific functions of the gut microbiome and how they may be modulated by diet. Diet exerts both supportive and selective pressures on the gut microbiome by regulating a multitude of factors, including energy density, macronutrient and micronutrient content, and circadian rhythm. The microbiome, in turn, contributes to local and systemic immune responses by providing colonization resistance against pathogens, shaping immune cell activity and differentiation, and facilitating the production of bioactive metabolites. Emerging research has strengthened the connections between the gut microbiome and cardiometabolic disorders (e.g., cardiovascular disease, obesity, type-2 diabetes), autoimmune conditions (e.g., type-1 diabetes, rheumatoid arthritis, celiac disease), respiratory disease, chronic kidney and liver disease, inflammatory bowel disease, and neurological disorders (e.g., Alzheimer’s, Parkinson’s disease, depressive disorders). Here, we outline ways in which dietary factors impact host response in diseases through alterations of gut microbiome functionality and composition. Consideration of diet-mediated microbial effects within the context of the diseases discussed highlights the potential of microbiome-targeted treatment strategies as alternative or adjunct therapies to improve patient outcomes. Full article
Show Figures

Figure 1

41 pages, 11116 KiB  
Article
In Silico Identification and Characterization of Spiro[1,2,4]triazolo[1,5-c]quinazolines as Diacylglycerol Kinase α Modulators
by Lyudmyla Antypenko, Kostiantyn Shabelnyk, Oleksii Antypenko, Mieko Arisawa, Oleksandr Kamyshnyi, Valentyn Oksenych and Serhii Kovalenko
Molecules 2025, 30(11), 2324; https://doi.org/10.3390/molecules30112324 - 26 May 2025
Viewed by 769
Abstract
A new class of spiro[1,2,4]triazolo[1,5-c]quinazoline derivatives is presented as promising modulators of diacylglycerol kinase α (DGK-α), a target implicated in cancer, neurological disorders, and immune dysfunction. Through structure-based computational design using the CB-Dock2 platform with human DGK-α (PDB ID: 6IIE), 40 [...] Read more.
A new class of spiro[1,2,4]triazolo[1,5-c]quinazoline derivatives is presented as promising modulators of diacylglycerol kinase α (DGK-α), a target implicated in cancer, neurological disorders, and immune dysfunction. Through structure-based computational design using the CB-Dock2 platform with human DGK-α (PDB ID: 6IIE), 40 novel compounds were systematically evaluated along with established inhibitors (ritanserin, R59022, R59949, BMS502, and (5Z,2E)-CU-3) across five distinct binding pockets. Several compounds demonstrated binding profiles at the level of or surpassing the reference compounds. The physicochemical analysis revealed balanced drug-like properties with favorable molecular weights (252–412 g/mol) and appropriate three-dimensionality. The toxicological assessment indicated reassuring safety profiles with predicted LD50 values of 1000–2000 mg/kg and minimal hepatotoxicity, carcinogenicity, and mutagenicity potential. Notably, compound 33 (adamantyl-substituted) emerged as exceptionally promising, exhibiting strong binding affinity, moderate solubility, and selective CYP inhibition patterns that minimize drug–drug interaction risks. Detailed molecular interaction mapping identified critical binding determinants, including strategic hydrogen bonding with TRP151, GLU166, and ARG126. The multidimensional evaluation identified compounds 13, 18, 33, and 40 as particularly promising candidates that balance potent target engagement with favorable pharmaceutical profiles, establishing this scaffold as a valuable platform for developing next-generation therapeutics targeting DGK-α -mediated signaling pathways. Full article
Show Figures

Graphical abstract

21 pages, 1089 KiB  
Article
Discrepancy Between Vaccination Willingness and Actual SARS-CoV-2 Vaccination Status in People with Multiple Sclerosis: A Longitudinal Study
by Felicita Heidler, Michael Hecker, Niklas Frahm, Julia Baldt, Silvan Elias Langhorst, Pegah Mashhadiakbar, Barbara Streckenbach, Katja Burian, Jörg Richter and Uwe Klaus Zettl
J. Clin. Med. 2025, 14(11), 3689; https://doi.org/10.3390/jcm14113689 - 24 May 2025
Viewed by 509
Abstract
Background/Objectives: Infection with severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) poses a significant health risk, especially for individuals with chronic medical conditions. Multiple sclerosis (MS) is the most prevalent chronic, immune-mediated neurological disorder, and vaccinations are essential to its management. This study [...] Read more.
Background/Objectives: Infection with severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) poses a significant health risk, especially for individuals with chronic medical conditions. Multiple sclerosis (MS) is the most prevalent chronic, immune-mediated neurological disorder, and vaccinations are essential to its management. This study aimed to compare the reported willingness to be vaccinated against SARS-CoV-2 with the actual vaccination status among people with MS (pwMS) and identify factors explaining the discrepancy. Methods: In a longitudinal, two-center study, we analyzed 149 patients aged 18 or older with a diagnosis of clinically isolated syndrome or MS. The participants completed three surveys: a baseline survey (from June 2019 to June 2020), a pre-vaccine follow-up (from May to July 2020), and a post-vaccine follow-up (from October 2021 to January 2022). The data included sociodemographic, clinical, and psychological information. Results: Among the 149 participants, 122 (81.9%) received a SARS-CoV-2 vaccination, while 27 (18.1%) did not. The pwMS who were unwilling to become vaccinated and remained unvaccinated were less likely to live with a partner, had higher smoking rates, took more medications, had a higher number of previously discontinued disease-modifying therapies, and found pandemic policies inappropriate. No significant associations were found between vaccination willingness/status and factors like age, sex, depression, or anxiety. Conclusions: This study highlights the gap between vaccination willingness and actual status in pwMS, revealing factors associated with vaccine hesitancy. The findings of this study offer insights into addressing vaccine uptake. Full article
(This article belongs to the Section Clinical Neurology)
Show Figures

Figure 1

11 pages, 224 KiB  
Case Report
Use of Cyclosporine and Itraconazole as Palliative Treatment for Proventricular Dilatation Disease in Psittacine Birds
by Laura M. Kleinschmidt, Sharman M. Hoppes, Jeffrey M. B. Musser, Ian Tizard and J. Jill Heatley
Vet. Sci. 2025, 12(5), 459; https://doi.org/10.3390/vetsci12050459 - 12 May 2025
Viewed by 691
Abstract
Proventricular dilatation disease (PDD) is a neurologic syndrome of birds caused by the infectious agent Psittacine Bornavirus (PaBV). Clinical disease may be based on the T-cell-mediated immune response to PaBV within the central and peripheral nervous system, similar to Borna disease virus, a [...] Read more.
Proventricular dilatation disease (PDD) is a neurologic syndrome of birds caused by the infectious agent Psittacine Bornavirus (PaBV). Clinical disease may be based on the T-cell-mediated immune response to PaBV within the central and peripheral nervous system, similar to Borna disease virus, a closely related mammalian virus. Lymphoplasmacytic infiltrations may occur in ganglia, nerve plexuses, peripheral nerves and the central nervous system of the infected bird. Clinical disease may result in multiple neurologic disorders and life-threatening morbidity. Treatment of PDD with antivirals and non-steroidal anti-inflammatories has thus far been non-curative and unsuccessful long-term. Cyclosporine is an immunosuppressant drug that decreases cell-mediated immune responses by inhibiting T-cell proliferation and decreasing cytokine production. In avian species, cyclosporine is a potent immunosuppressant with T-cell-specific action. A pilot study performed in PaBV-infected cockatiels showed increased weight gain and a lack of morbidity or mortality following experimental PaBV infection and cyclosporine treatment at 10 mg/kg orally every 12 h. In this case series of six psittacine birds affected by PDD, cyclosporine at this dose alleviated or reduced clinical signs in multiple birds without severe sequelae. Itraconazole was used concurrently in these cases to prevent secondary fungal infections during immunosuppression but may have had a synergetic effect when used in combination with cyclosporine. Further prospective research is indicated to better evaluate cyclosporine use in birds with PDD. However, these preliminary clinical findings suggest that cyclosporine and itraconazole administration is a treatment option for palliation of PDD in psittacine patients, especially those refractory to other treatments. Full article
(This article belongs to the Section Veterinary Biomedical Sciences)
41 pages, 1230 KiB  
Review
Human T-Lymphotropic Virus (HTLV): Epidemiology, Genetic, Pathogenesis, and Future Challenges
by Francesco Branda, Chiara Romano, Grazia Pavia, Viola Bilotta, Chiara Locci, Ilenia Azzena, Ilaria Deplano, Noemi Pascale, Maria Perra, Marta Giovanetti, Alessandra Ciccozzi, Andrea De Vito, Angela Quirino, Nadia Marascio, Giovanni Matera, Giordano Madeddu, Marco Casu, Daria Sanna, Giancarlo Ceccarelli, Massimo Ciccozzi and Fabio Scarpaadd Show full author list remove Hide full author list
Viruses 2025, 17(5), 664; https://doi.org/10.3390/v17050664 - 1 May 2025
Cited by 1 | Viewed by 1748
Abstract
Human T-lymphotropic viruses (HTLVs) are deltaretroviruses infecting millions of individuals worldwide, with HTLV-1 and HTLV-2 being the most widespread and clinically relevant types. HTLV-1 is associated with severe diseases such as adult T-cell leukemia/lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), while HTLV-2 [...] Read more.
Human T-lymphotropic viruses (HTLVs) are deltaretroviruses infecting millions of individuals worldwide, with HTLV-1 and HTLV-2 being the most widespread and clinically relevant types. HTLV-1 is associated with severe diseases such as adult T-cell leukemia/lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), while HTLV-2 shows a lower pathogenic potential, with occasional links to neurological disorders. HTLV-3 and HTLV-4, identified in Central Africa, remain poorly characterized but are genetically close to their simian counterparts, indicating recent zoonotic transmission events. HTLVs replicate through a complex cycle involving cell-to-cell transmission and clonal expansion of infected lymphocytes. Viral persistence is mediated by regulatory and accessory proteins, notably Tax and HBZ in HTLV-1, which alter host cell signaling, immune responses, and genomic stability. Integration of proviral DNA into transcriptionally active regions of the host genome may contribute to oncogenesis and long-term viral latency. Differences in viral protein function and intracellular localization contribute to the distinct pathogenesis observed between HTLV-1 and HTLV-2. Geographically, HTLV-1 shows endemic clusters in southwestern Japan, sub-Saharan Africa, the Caribbean, South America, and parts of the Middle East and Oceania. HTLV-2 is concentrated among Indigenous populations in the Americas and people who inject drugs in Europe and North America. Transmission occurs primarily via breastfeeding, sexual contact, contaminated blood products, and, in some regions, zoonotic spillover. Diagnostic approaches include serological screening (ELISA, Western blot, LIA) and molecular assays (PCR, qPCR), with novel biosensor and AI-based methods under development. Despite advances in understanding viral biology, therapeutic options remain limited, and preventive strategies focus on transmission control. The long latency period, lack of effective treatments, and global neglect complicate public health responses, underscoring the need for increased awareness, research investment, and targeted interventions. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

40 pages, 5811 KiB  
Review
Metabolic Dysfunction and Dietary Interventions in Migraine Management: The Role of Insulin Resistance and Neuroinflammation—A Narrative and Scoping Review
by Cinzia Cavestro
Brain Sci. 2025, 15(5), 474; https://doi.org/10.3390/brainsci15050474 - 29 Apr 2025
Cited by 2 | Viewed by 1769
Abstract
Introduction: Migraine is a prevalent neurological disorder characterized by recurrent headaches with autonomic and neurological symptoms, significantly impacting quality of life globally. Its pathogenesis involves genetic, neurological, inflammatory, and metabolic factors, with insulin resistance and metabolic dysfunction increasingly recognized as important contributors. Historically, [...] Read more.
Introduction: Migraine is a prevalent neurological disorder characterized by recurrent headaches with autonomic and neurological symptoms, significantly impacting quality of life globally. Its pathogenesis involves genetic, neurological, inflammatory, and metabolic factors, with insulin resistance and metabolic dysfunction increasingly recognized as important contributors. Historically, it has been known that certain foods can trigger migraine attacks, which led for many years to the recommendation of elimination diets—now understood to primarily target histamine-rich foods. Over the past two decades, attention has shifted toward underlying metabolic disturbances, leading to the development of dietary approaches specifically aimed at addressing these dysfunctions. Methods: A scoping literature review was conducted using PubMed and Embase to evaluate the relationships among migraine, insulin-related mechanisms, neurogenic inflammation, and dietary interventions. Initial searches focused on “MIGRAINE AND (neurogenic inflammation)” (2019–15 April 2025), followed by expanded searches from 1950 onward using terms such as “MIGRAINE AND (insulin, insulin resistance, hyperinsulinism)”, and “MIGRAINE AND (diet, dietary, nutrition, nutritional)”. A specific search also targeted “(INSULIN OR insulin resistance OR hyperinsulinism) AND (neurogenic inflammation)”. Abstracts were screened, full texts were retrieved, and duplicates or irrelevant publications were excluded. No filters were applied by article type or language. Systematic reviews and meta-analyses were prioritized when available. Results: Migraine pathogenesis involves trigeminovascular system activation, neurogenic inflammation mediated by CGRP and PACAP, immune dysregulation, mast cell activation, and cortical spreading depression (CSD). Emerging evidence highlights significant associations between migraine, insulin resistance, and hyperinsulinism. Hyperinsulinism contributes to migraine through TRPV1 sensitization, increased CGRP release, oxidative stress, mitochondrial dysfunction, and systemic inflammation. Metabolic dysfunction, including obesity and insulin resistance, exacerbates migraine severity and frequency. Dietary interventions, particularly anti-inflammatory, Mediterranean, and ketogenic diets, show promise in reducing migraine frequency and severity through mechanisms involving reduced inflammation, oxidative stress, improved mitochondrial function, and glucose metabolism stabilization. Conclusions: The interplay between insulin resistance, metabolic dysfunction, and neuroinflammation is crucial in migraine pathophysiology. Targeted dietary interventions, including ketogenic and Mediterranean diets, demonstrate significant potential in managing migraines, emphasizing the need for personalized nutritional strategies to optimize therapeutic outcomes. Full article
(This article belongs to the Special Issue Advances in Neuroinflammation and Pain Medicine)
Show Figures

Graphical abstract

29 pages, 2290 KiB  
Review
Beyond the Gut: Unveiling Butyrate’s Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review
by Arda Erkan Kalkan, Mona N. BinMowyna, António Raposo, Md Faruque Ahmad, Faiyaz Ahmed, Abdullah Y. Otayf, Conrado Carrascosa, Ariana Saraiva and Sercan Karav
Nutrients 2025, 17(8), 1305; https://doi.org/10.3390/nu17081305 - 9 Apr 2025
Cited by 6 | Viewed by 5341
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within [...] Read more.
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate’s role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate’s growing recognition as a modulator of neurological health via its interaction with the gut–brain axis. Additionally, butyrate’s neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer’s, Parkinson’s, autism spectrum disorder, and Huntington’s disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected. Full article
Show Figures

Figure 1

24 pages, 1610 KiB  
Review
Targeting the Endocannabinoidome: A Novel Approach to Managing Extraintestinal Complications in Inflammatory Bowel Disease
by Dinesh Thapa, Anjali Ghimire, Leon N. Warne and Rodrigo Carlessi
Pharmaceuticals 2025, 18(4), 478; https://doi.org/10.3390/ph18040478 - 27 Mar 2025
Viewed by 1210
Abstract
Background: Inflammatory bowel disease (IBD) is a chronic inflammatory disorder marked by persistent gastrointestinal inflammation and a spectrum of systemic effects, including extraintestinal manifestations (EIMs) that impact the joints, skin, liver, and eyes. Conventional therapies primarily target intestinal inflammation, yet they frequently [...] Read more.
Background: Inflammatory bowel disease (IBD) is a chronic inflammatory disorder marked by persistent gastrointestinal inflammation and a spectrum of systemic effects, including extraintestinal manifestations (EIMs) that impact the joints, skin, liver, and eyes. Conventional therapies primarily target intestinal inflammation, yet they frequently fail to ameliorate these systemic complications. Recent investigations have highlighted the complex interplay among the immune system, gut, and nervous system in IBD pathogenesis, thereby underscoring the need for innovative therapeutic approaches. Methods: We conducted a comprehensive literature search using databases such as PubMed, Scopus, Web of Science, Science Direct, and Google Scholar. Keywords including “cannabinoids”, “endocannabinoid system”, “endocannabinoidome”, “inflammatory bowel disease”, and “extraintestinal manifestations” were used to identify peer-reviewed original research and review articles that explore the role of the endocannabinoidome (eCBome) in IBD. Results: Emerging evidence suggests that eCBome—a network comprising lipid mediators, receptors (e.g., CB1, CB2, GPR55, GPR35, PPARα, TRPV1), and metabolic enzymes—plays a critical role in modulating immune responses, maintaining gut barrier integrity, and regulating systemic inflammation. Targeting eCBome not only improves intestinal inflammation but also appears to mitigate metabolic, neurological, and extraintestinal complications such as arthritis, liver dysfunction, and dermatological disorders. Conclusions: Modulation of eCBome represents a promising strategy for comprehensive IBD management by addressing both local and systemic disease components. These findings advocate for further mechanistic studies to develop targeted interventions that leverage eCBome as a novel therapeutic avenue in IBD. Full article
Show Figures

Graphical abstract

22 pages, 2256 KiB  
Article
Mild Zika Virus Infection in Mice Without Motor Impairments Induces Working Memory Deficits, Anxiety-like Behaviors, and Dysregulation of Immunity and Synaptic Vesicle Pathways
by Jaime Alexander Chivatá-Ávila, Paola Rojas-Estevez, Alejandra M. Muñoz-Suarez, Esthefanny Caro-Morales, Aura Caterine Rengifo, Orlando Torres-Fernández, Jose Manuel Lozano and Diego A. Álvarez-Díaz
Viruses 2025, 17(3), 405; https://doi.org/10.3390/v17030405 - 12 Mar 2025
Viewed by 1087
Abstract
Background: The Zika virus (ZIKV) is an arbovirus linked to “Congenital Zika Syndrome” and a range of neurodevelopmental disorders (NDDs), with microcephaly as the most severe manifestation. Milder NDDs, such as autism spectrum disorders and delays in neuropsychomotor and language development, often go [...] Read more.
Background: The Zika virus (ZIKV) is an arbovirus linked to “Congenital Zika Syndrome” and a range of neurodevelopmental disorders (NDDs), with microcephaly as the most severe manifestation. Milder NDDs, such as autism spectrum disorders and delays in neuropsychomotor and language development, often go unnoticed in neonates, resulting in long-term social and academic difficulties. Murine models of ZIKV infection can be used to mimic part of the spectrum of motor and cognitive deficits observed in humans. These can be evaluated through behavioral tests, enabling comparison with gene expression profiles and aiding in the characterization of ZIKV-induced NDDs. Objectives: This study aimed to identify genes associated with behavioral changes following a subtle ZIKV infection in juvenile BALB/c mice. Methods: Neonatal mice were subcutaneously inoculated with ZIKV (MH544701.2) on postnatal day 1 (DPN) at a dose of 6.8 × 103 PFU. Viral presence in the cerebellum and cortex was quantified at 10- and 30-days post-infection (DPI) using RT-qPCR. Neurobehavioral deficits were assessed at 30 DPI through T-maze, rotarod, and open field tests. Next-Generation Sequencing (NGS) was performed to identify differentially expressed genes (DEGs), which were analyzed through Gene Ontology (GO) and KEGG enrichment. Gene interaction networks were then constructed to explore gene interactions in the most enriched biological categories. Results: A ZIKV infection model was successfully established, enabling brain infection while allowing survival beyond 30 DPI. The infection induced mild cognitive behavioral changes, though motor and motivational functions remained unaffected. These cognitive changes were linked to the functional repression of synaptic vesicles and alterations in neuronal structure, suggesting potential disruptions in neuronal plasticity. Conclusions: Moderate ZIKV infection with circulating strains from the 2016 epidemic may cause dysregulation of genes related to immune response, alterations in cytoskeletal organization, and modifications in cellular transport mediated by vesicles. Despite viral control, neurocognitive effects persisted, including memory deficits and anxiety-like behaviors, highlighting the long-term neurological consequences of ZIKV infection in models that show no apparent malformations. Full article
(This article belongs to the Special Issue Arboviral Lifecycle 2025)
Show Figures

Figure 1

24 pages, 1204 KiB  
Review
Modelling Peroxisomal Disorders in Zebrafish
by Chenxing S. Jiang and Michael Schrader
Cells 2025, 14(2), 147; https://doi.org/10.3390/cells14020147 - 20 Jan 2025
Viewed by 2349
Abstract
Peroxisomes are ubiquitous, dynamic, oxidative organelles with key functions in cellular lipid metabolism and redox homeostasis. They have been linked to healthy ageing, neurodegeneration, cancer, the combat of pathogens and viruses, and infection and immune responses. Their biogenesis relies on several peroxins (encoded [...] Read more.
Peroxisomes are ubiquitous, dynamic, oxidative organelles with key functions in cellular lipid metabolism and redox homeostasis. They have been linked to healthy ageing, neurodegeneration, cancer, the combat of pathogens and viruses, and infection and immune responses. Their biogenesis relies on several peroxins (encoded by PEX genes), which mediate matrix protein import, membrane assembly, and peroxisome multiplication. Defects in peroxins or peroxisomal enzymes can result in severe disorders, including developmental and neurological abnormalities. The drive to understand the role of peroxisomes in human health and disease, as well as their functions in tissues and organs or during development, has led to the establishment of vertebrate models. The zebrafish (Danio rerio) has become an attractive vertebrate model organism to investigate peroxisomal functions. Here, we provide an overview of the visualisation of peroxisomes in zebrafish, as well as the peroxisomal metabolic functions and peroxisomal protein inventory in comparison to human peroxisomes. We then present zebrafish models which have been established to investigate peroxisomal disorders. These include model zebrafish for peroxisome biogenesis disorders/Zellweger Spectrum disorders, and single enzyme deficiencies, particularly adrenoleukodystrophy and fatty acid beta-oxidation abnormalities. Finally, we highlight zebrafish models for deficiencies of dually targeted peroxisomal/mitochondrial proteins. Advantages for the investigation of peroxisomes during development and approaches to the application of zebrafish models for drug screening are discussed. Full article
(This article belongs to the Special Issue Modeling Developmental Processes and Disorders in Zebrafish)
Show Figures

Figure 1

Back to TopTop