bims-blobar Biomed News
on Blood brain barrier repair
Issue of 2025–09–28
sixteen papers selected by
Nicolas Rebergue



  1. bioRxiv. 2025 Sep 15. pii: 2025.09.09.675258. [Epub ahead of print]
      Vascular normalization [stabilization of aberrant angiogenesis and restoration of blood-brain barrier (BBB)] is critical for reducing long-term secondary sequelae after ischemic stroke. How immune and developmental signaling pathways coordinate these processes is poorly understood. Here we identify a unique brain endothelial cell (BEC) type one interferon (IFN1) signature in human and mouse ischemic stroke tissue. By leveraging two clinically-relevant murine ischemic stroke models, single-cell transcriptomics, and BBB functional assays, we find that deletion of endothelial IFN1 receptor (Ifnar1) exacerbates post-stroke BBB disruption and expands a BEC population expressing angiogenic and immature BBB markers. Conversely, IFNβ administration immediately after stroke reduces acute BBB disruption. Activation of IFN1 signaling in mouse BECs in vitro promotes junctional stabilization and reduces vascular endothelial growth factor (VEGF) signaling to enhance barrier properties, but suppress angiogenic features. Thus, endogenous endothelial IFN1 signaling modulates BBB dysfunction and angiogenesis to promote vascular normalization after ischemic brain injury.
    DOI:  https://doi.org/10.1101/2025.09.09.675258
  2. Eur Arch Psychiatry Clin Neurosci. 2025 Sep 27.
      The blood-brain barrier (BBB) integrity is important for central nervous system homeostasis, and dysfunction of its components may contribute to the pathogenesis of various neuropsychiatric disorders, including schizophrenia. An additional factor supporting this hypothesis is the association of schizophrenia with DiGeorge syndrome, in which a deletion of a part of one copy of chromosome 22 (22q11DS) leads to haplonephrenia in the Claudin-5 gene (a tight junction protein that forms the BBB). Individuals with 22q11DS have a 25-fold greater risk of schizophrenia than the population average. The aim of our review was to establish the role of tight junction proteins in the pathogenesis of schizophrenia. For this purpose, a systematic literature search was performed. The first part describes genetic polymorphisms of tight junction proteins and studies of patients with DiGeorge syndrome. The second and third parts of the review focus on the expression of tight junction proteins in postmortem samples and serum. The fourth part of the review describes in vitro studies. The fifth part includes animal studies. Mice models with incomplete absence of the Claudin-5 gene have shown specific behavioral and memory impairments. The final part describes the effects of antipsychotics on tight junction proteins and BBB function. The discussed studies support the role of tight junction proteins in the pathogenesis of schizophrenia and the increased permeability of the BBB. However, the number of studies on this topic is extremely small. This review shows the prospects of studying tight junction proteins in psychiatric diseases.
    Keywords:  Blood brain barrier; Claudin; DiGeorge syndrome; Schizophrenia; Tight junction
    DOI:  https://doi.org/10.1007/s00406-025-02060-6
  3. Brain Commun. 2025 ;7(5): fcaf332
      Age-associated vascular alterations make the brain more vulnerable to neuropathologies. Research in humans and rodents has demonstrated structural, molecular, and functional alterations of the aged brain vasculature that suggest blood-brain barrier dysfunction. However, these studies focused on particular features of the blood-brain barrier and specific brain regions. Thus, it remains unclear if and which blood-brain barrier age-associated phenotypes are conserved across brain areas. Moreover, there is very limited information about how blood-brain barrier dysfunction and cell-specific phenotypes relate to each other. In this manuscript, we use immunofluorescence, transmission electron microscopy, and permeability assays to assess how age-associated blood-brain barrier molecular, structural, and functional phenotypes correlate between the blood-brain barrier cell types at three brain regions (prefrontal cortex, hippocampus, and corpus callosum) during mouse early ageing. We discovered that at 18-20 months of age, changes to the mouse blood-brain barrier are subtle. The prefrontal cortex blood-brain barrier is the most affected by age, with alterations in brain endothelial cell protein expression, blood-brain barrier permeability, basement membrane thickness, and astrocyte endfoot size when compared with young mice. Here, we deliver a detailed multicellular characterization of region-dependent blood-brain barrier changes at early stages of ageing. Our data paves the way for future studies to investigate how region-specific blood-brain barrier dysfunction may contribute to disease-associated regional vulnerability.
    Keywords:  ageing; astrocyte endfoot; blood-brain barrier; brain endothelial cell; transmission electron microscopy
    DOI:  https://doi.org/10.1093/braincomms/fcaf332
  4. Acta Neuropathol Commun. 2025 Sep 24. 13(1): 195
      Stereotactic radiosurgery (SRS) is a procedure that delivers high-dose single fraction, targeted radiation to treat brain pathologies. Brain radiation necrosis is a significant side effect of SRS, resulting in severe clinical sequelae such as seizure, hemorrhage, stroke, and neurological deficit. While focused radiation causes DNA damage and cell death, radiation necrosis is mostly mediated by vascular injury. Yet the effects of SRS on the neurovascular unit (NVU) cells-microglia, astrocytes, and endothelial cells-remain poorly understood. This study establishes a mouse SRS model using 15 to 60 Gy to characterize NVU stress, providing histological and transcriptomic profiles of radiation-induced damage. Our findings demonstrate blood-brain-barrier (BBB) disruption, inflammatory cell infiltration, and microvascular pathology. Spatial transcriptomics identified differentially expressed genes and cell-cell communication across NVU components, revealing a coordinated stress response involving immune modulation, barrier integrity, and tissue remodeling pathways. This model provides a mechanistic framework for developing strategies to mitigate BBB and NVU stress.
    Keywords:  Adverse radiation effects (AREs); Blood-brain barrier (BBB); Brain damage; Mouse radiation model; Neurovascular unit; Stereotactic radiosurgery (SRS)
    DOI:  https://doi.org/10.1186/s40478-025-02112-x
  5. Prog Neuropsychopharmacol Biol Psychiatry. 2025 Sep 24. pii: S0278-5846(25)00264-7. [Epub ahead of print] 111510
      The neurogliovascular unit (NGVU) reflects the complex interplay between neural tissue and blood flow. Dysfunction in this NGVU system is involved in neuropsychiatric disorders, however, whether the alterations are a cause or consequence of these conditions remains unclear. This study investigates the role of NGVU adaptations in motivational deficits associated with depressive episodes, focusing on blood vessel structural changes and blood-brain barrier (BBB) permeability. We used brain samples from adult male C57BL/6jRj mice that were chronically treated with corticosterone (CORT), and which presented severe motivational deficits in an operant progressive ratio task, along with altered neural activation in brain regions involved in motivational processing (anterior insular cortex, basolateral amygdala, bed nucleus of the stria terminalis and ventral tegmental area), as assessed by FosB expression. NGVU modifications were first evaluated through immunofluorescence staining for microglia (IBA-1), endothelial tight junctions (ZO-1), and astrocytes (GFAP). BBB permeability was assessed using intravenous perfusion of fluorescent 40 kDa Dextran. Principal component analysis revealed that NGVU alterations in the ventral tegmental area and basolateral amygdala predicted motivational deficits in CORT-treated mice. Specifically, ZO-1 expression was downregulated, and Dextran extravasation was increased in these regions. These findings suggest that NGVU adaptations induced by chronic CORT exposure impact BBB integrity and are integral to understanding behavioural performance. In conclusion, NGVU modifications may play a key role in the cognitive and behavioural dysfunction seen in neuropsychiatric disorders, highlighting their relevance in the biological substrate of these conditions.
    Keywords:  Blood-brain barrier; Chronic distress; Motivation; Neurogliovascular unit; ZO-1
    DOI:  https://doi.org/10.1016/j.pnpbp.2025.111510
  6. Trends Pharmacol Sci. 2025 Sep 19. pii: S0165-6147(25)00182-8. [Epub ahead of print]
      Transcytosis across the blood-brain barrier (BBB) enables systemically administered large therapeutics to reach the brain parenchyma, but their fate in the parenchyma ultimately governs their therapeutic effect. Recent studies show that brain parenchymal cell uptake, internalization kinetics, reuptake at the BBB, and diffusion in the brain parenchyma shape the distribution and retention of therapeutics. Target engagement further influences their behavior beyond the BBB. These insights have prompted new strategies to enhance their distribution, retention, and target engagement. These include the selection of transport targets with favorable trafficking properties, the use of anchoring proteins, and modeling-based optimization. This opinion highlights emerging understanding of the fate of therapeutics in the brain parenchyma and outlines strategies to optimize this fate.
    Keywords:  blood–brain barrier; brain parenchyma; distribution; drug delivery; retention; transcytosis
    DOI:  https://doi.org/10.1016/j.tips.2025.08.006
  7. Food Chem Toxicol. 2025 Sep 24. pii: S0278-6915(25)00530-7. [Epub ahead of print] 115762
      Microplastics (MP) and nanoplastics (NP), including low-density polyethylene (LDPE), as well as carbon-based particles such as carbon black (CBP) and black carbon (BC) from incomplete combustion, are widespread environmental pollutants. Their co-occurrence in airborne particulate matter raises concerns about potential impacts on sensitive barriers such as the blood-brain barrier (BBB). This study investigates the effects of CBP and LDPE particles on human cerebral microvascular endothelium hCMEC/D3 cells, an in vitro BBB model. Using a stable NRF2 reporter cell line, we assessed barrier function via transendothelial electrical resistance (TEER), particle uptake through confocal microscopy, and morphological changes using CellProfiler. Inflammatory and oxidative stress responses were also evaluated. CBP exposure increased TEER values, suggesting an occluding effect on barrier integrity. Additionally, CBP enhanced phagocytic activity and reduced IL-1β levels, indicating modulation of inflammation. In contrast, although internalized by cells, LDPE particles had minimal impact on morphology, phagocytosis, inflammation, or barrier function. These findings offer new insights into the cellular responses to airborne pollutants, highlighting the differential effects of CBP and LDPE particles on the BBB and underscoring the need for further studies on their combined impact.
    Keywords:  air pollution; blood-brain barrier; carbon black particles; inflammation; low-density polyethylene; microplastic; oxidative stress
    DOI:  https://doi.org/10.1016/j.fct.2025.115762
  8. Ageing Res Rev. 2025 Sep 23. pii: S1568-1637(25)00246-6. [Epub ahead of print] 102900
      The glymphatic system is an essential component in modulating brain health and clearing out toxins from the central nervous system (CNS). The glymphatic system, especially the perivascular space, undergoes significant alterations during CNS inflammation. Various cerebrovascular disorders have implicated glymphatic dysfunction as the major contributing factor for cerebral edema, neuroinflammation, as well as damage to the blood-brain barrier (BBB). Intracerebral hemorrhage (ICH) is a major life-threatening stroke subtype with limited therapies available. ICH results from the rupture of cerebral arteries, accumulation of the blood and hematoma formation in the brain parenchyma. The toxic degradation components from the hematoma further exacerbates the oxidative stress, inflammatory response and cerebral edema, worsening the BBB disruption as well as the neurological outcomes post-ICH. Thus, making it imperative to understand the underlying mechanisms of secondary brain injury (SBI) and create targeted therapeutic strategies to improve patient recovery. Owing to the involvement of the glymphatic system in clearing out debris, it may contribute in alleviating SBI following ICH. In this review we intend to gain a better understanding of the glymphatic system's role in mitigating SBI as well as potential therapeutic approaches that target this system to improve patient recovery and reduce damage post-ICH.
    Keywords:  Blood Brain Barrier disruption; Glymphatic system; Intracerebral Hemorrhage; Neuroinflammation; Secondary Brain Injury
    DOI:  https://doi.org/10.1016/j.arr.2025.102900
  9. J Nat Med. 2025 Sep 26.
      This study investigated the modification of blood-brain barrier (BBB) permeability of alkaloids from Phellodendri Chinensis Cortex (PC) by wine processing. Phytochemical analysis using UPLC-QqQ-MS compared raw PC (RPC) and wine-processed PC (WPC), identifying significant changes in four major alkaloids (phellodendrine, magnoflorine, berberrubine, and berberine). Subsequent in vivo experiments demonstrated enhanced brain distribution of these alkaloids in WPC-treated mice. An in vitro BBB model was established using bEnd.3 endothelial cells and astrocytes to evaluate the permeability of four major alkaloids from RPC and WPC using UPLC-QqQ-MS. Tight junction protein expression and transporter interactions were analyzed through Western blotting and ligand fishing assays. Results showed WPC treatment significantly enhanced alkaloid transport across the BBB model, with mechanistic studies demonstrating downregulation of tight junction proteins (ZO-1 and Occludin), reduced P-glycoprotein expression, and increased binding to the absorptive transporter TfRc. These findings reveal that wine processing enhances BBB penetration of PC alkaloids by coordinately modulating both paracellular and transcellular transport pathways, providing scientific validation for traditional processing methods of Chinese Materia Medica and insights for developing brain-targeted herbal medicines.
    Keywords:  Affinity ultrafiltration; Alkaloids; Blood–brain barrier; Phellodendri Chinensis Cortex; UPLC-QqQ-MS; Wine processing
    DOI:  https://doi.org/10.1007/s11418-025-01952-0
  10. FASEB J. 2025 Sep 30. 39(18): e70868
      Diabetes-related cerebral small-vessel disease (CSVD) is an important causative factor of cognitive impairment, but its molecular mechanisms have not been clarified. The aim of this study was to investigate the role of the necrotic apoptotic pathway (RIP1/RIP3/MLKL) and the inflammatory response in diabetic CSVD. Wild-type C57BL/6 mice and leptin receptor-deficient db/db mice were categorized into six groups according to age (8-, 12-, and 16-week time points) and genotype. Cognitive function was assessed by the water maze experiment (escape latency, percentage of time spent in the target quadrant, and number of times through the table); cerebral atrophy and ventricular dilatation were detected by cranial MRI; cerebral microvascular structure, cortical neuronal damage, and ultrapathological changes in hippocampal mitochondria were observed by HE staining and transmission electron microscopy, respectively; and blood-brain barrier-associated proteins were detected by western blot and RT-qPCR (occludin, ZO-1, VEGFA) and necroptotic apoptotic pathway molecules (RIP1, RIP3, MLKL). Western blot and RT-qPCR were used to detect the protein and mRNA expression of blood-brain barrier-associated proteins (occludin, ZO-1, VEGFA) and necroptotic apoptotic pathway molecules (RIP1, RIP3, MLKL). Immunohistochemistry was used to localize the distribution of RIP1/RIP3/MLKL in the brain tissues; and plasma levels of inflammatory factors (IL-6, IL-10, TNF-α, NF-κB) were quantified by ELISA. In db/db mice: (1) spatial learning and memory abilities were reduced compared to wild-type (WT) mice; (2) at 16 weeks of age, db/db mice showed signs of temporal lobe atrophy and an enlarged fourth cerebral ventricle; (3) capillary proliferation and cortical injury were observed in the frontal cortex, along with mitochondrial swelling, degeneration, and nuclear membrane rupture in hippocampal cells; (4) the occludin and ZO-1 protein expression in the db/db-16W group decreased to 0.48- and 0.68-fold of the WT-8W group, respectively, and the VEGFA was elevated by 2.87-fold; the mRNA expression of RIP1/RIP3/MLKL was up-regulated to 3.02-, 3.12-, and 4.02-fold of the WT group, the relative expression of western blot proteins increased synchronously; the increase in the number of immunohistochemically positive cells increased synchronously, and (5) plasma inflammatory factors were significantly elevated in db/db mice: IL-6 ↑3.81-fold, TNF-α ↑4.23-fold, NF-κB ↑3.56-fold. This study reveals for the first time the molecular mechanism by which diabetes drives cerebral small vessel disease (CSVD) through the spatiotemporal-dependent activation of the necrotic apoptotic pathway (RIP1/RIP3/MLKL), and targeting the necrotic apoptotic pathway may serve as a potential therapeutic strategy for diabetes-associated cognitive deficits by concurrently protecting the blood-brain barrier and suppressing neuroinflammation.
    Keywords:  cerebral small vessel disease (CSVD); cognitive impairment; diabetes mellitus; inflammation; necroptosis
    DOI:  https://doi.org/10.1096/fj.202500772R
  11. bioRxiv. 2025 Sep 19. pii: 2025.09.18.676925. [Epub ahead of print]
      Development and delivery of treatments for neurological diseases are limited by the tight and selective human blood-brain barrier (BBB). Although animal models have been important research and preclinical tools, the rodent BBB exhibits species differences and fails to capture the complexity of human genetics. Microphysiological systems incorporating human-derived cells hold great potential for modeling disease and therapeutic development, with advantages in screening throughput, real-time monitoring, and tunable genetic backgrounds when combined with induced pluripotent stem cell (iPSC) technology. Existing 3D BBB-on-chip systems have incorporated iPSC-derived endothelial cells but not the other major brain cell types from iPSCs, each of which contributes to brain physiology and disease. Here we developed a 3D Brain-Chip system incorporating endothelial cells, pericytes, astrocytes, neurons, microglia, and oligodendroglia from iPSCs. To enable this multicellular 3D co-culture in-chip, we designed a GelChip microfluidic platform using a 3D printing-based approach and dextran-based engineered hydrogel. Leveraging this platform, we co-cultured and characterized iPSC-derived brain-on-chips and modeled the brain microvasculature of APOE4 , the strongest known genetic risk factor for sporadic Alzheimer's disease. These 3D brain-on-chips provide a versatile system to assess BBB vascular morphology and function, investigate downstream neurological effects in disease, and screen therapeutics to optimize delivery to the brain.
    Significance Statement: The blood-brain barrier (BBB) is both a contributing factor to neurological disease and a major obstacle to its treatment, yet human-relevant models remain limited. Most existing brain-on-chip systems incorporate only subsets of BBB cell types and cannot capture the full cellular complexity of the human neurovascular unit. Here, we establish a vascular-perfusable 3D Brain-Chip using human induced pluripotent stem cell-derived brain cells including endothelial cells, pericytes, astrocytes, neurons, microglia, and oligodendroglia. This system enables systematic analysis of human genetic risk factors, such as APOE4 in Alzheimer's disease, and provides a powerful platform to investigate BBB function and dysfunction and accelerate the development of more effective neurological therapies.
    DOI:  https://doi.org/10.1101/2025.09.18.676925
  12. Cells. 2025 Sep 15. pii: 1440. [Epub ahead of print]14(18):
      The Complement C3a Receptor (C3aR) plays a multifaceted role along the varying temporal phases of brain injury following cerebral ischemia. C3aR is a G-protein-coupled receptor (GPCR) that binds to its ligand, C3a an anaphylatoxin generated during activation of the complement cascade. During ischemia, complement is activated as part of the initial inflammatory response, with C3aRs playing a time-dependent role in both brain injury and repair mechanisms. In the acute phase (minutes to hours post-ischemia), C3aR activation promotes the recruitment of immune cells and the release of chemokines and cytokines, driving blood-brain barrier (BBB) permeability and brain edema. During the subacute phase (hours to days post-ischemia), C3aR continues to modulate immune cell activity, worsening secondary brain injury, although emerging evidence suggests that C3aR activation in this phase may also aid in the clearance of cellular debris and cell survival. In the chronic phase (days to weeks post-ischemia), chronically elevated C3aR activity can prolong neuroinflammation and impair recovery, whereas controlled C3aR signaling in the subacute/chronic phase can activate reparative pathways (e.g., microglial phagocytosis, astrocyte trophic support). As a result, targeting the C3aR requires careful timing to optimize its benefits. Given the dual impact of C3aR activation, which serves to exacerbate injury in the acute phase but supports repair beginning in the subacute and chronic phases, a targeted therapeutic approach should focus on context- and time-dependent modulation of the C3a/C3aR axis. This strategy would involve blocking the C3aR during the acute phase to reduce inflammation and BBB breakdown while controlling C3a signaling in later phases to promote tissue repair.
    Keywords:  complement C3a receptor (C3aR); ischemic stroke; neuroinflammation; temporal phases of brain injury; therapeutic modulation
    DOI:  https://doi.org/10.3390/cells14181440
  13. Biomark Res. 2025 Sep 26. 13(1): 116
    Alzheimer’s Disease All Markers (ADAM) Research Group
      Alzheimer's disease (AD) is the most common cause of dementia and accounts for approximately 60-80% of total dementia patients. Currently, accurate diagnosis for AD relies on cerebrospinal fluid (CSF) sampling or a positron emission tomography (PET) scan, methods that cannot be done in primary care centers where most people go with cognitive complaints. This Limitation calls for the urgent need to develop blood-related diagnostic tests that could facilitate early detection and enable timely treatment. Recent CSF proteomic research categorized AD into five molecular subtypes with discrete Genetic risk profiles. Subtypes 1-3, namely neuronal hyperplasticity, innate immune activation, and RNA dysregulation, were characterized by more classical AD-related changes, like accumulation of amyloid/tau and synaptic and immune dysfunction, respectively. On the contrary, non-traditional AD mechanisms in subtypes 4-5 were choroid plexus (CP) dysfunction and blood-brain barrier (BBB) dysfunction, emphasizing clearance deficits in association with brain barrier dysfunction. The unchanged tau levels later may be explained by an alternate disease mechanism (clearance dysfunction). These subtypes included BBB and CP dysfunction. Biomarker identification based on the mechanism of disease progression would increase the precision of diagnoses, allowing for tailored interventions and aiding in the creation of novel therapies for subtypes that might not react favorably to conventional amyloid/tau-targeting strategies. Finding biomarkers specific to each subtype would aid in patient classification, resulting in more individualized therapy as opposed to a "one-size-fits-all" strategy. The present review emphasized the importance of identifying blood-based biomarkers (BBMs) related to brain barrier dysfunction from CSF studies and personalized treatment strategies to streamline the diagnostic workup, and may be utilized in standard clinical practice for the early detection of AD.
    Keywords:  Alzheimer’s disease; BBB dysfunction; Blood Biomarkers; Choroid Plexus dysfunction; Early detection; Personalized treatment
    DOI:  https://doi.org/10.1186/s40364-025-00829-4
  14. Brain Behav Immun. 2025 Sep 19. pii: S0889-1591(25)00357-5. [Epub ahead of print]130 106115
      Chronic alcohol consumption is a major risk factor for neuroinflammation and cognitive decline, yet the molecular underpinnings connecting peripheral alcohol-induced injury to central nervous system (CNS) dysfunction remain poorly defined. Emerging evidence implicates purinergic P2X7 receptor (P2X7R) signaling and extracellular vesicles (EVs) as key mediators in peripheral-central communication. Ethanol exposure promotes oxidative stress, mitochondrial dysfunction, and blood-brain barrier (BBB) disruption, leading to sustained microglial activation and neuronal injury. Concurrently, alcohol-induced damage in the gut, liver, and lung, triggers systemic inflammation and EV release. These EVs, enriched in proinflammatory cytokines, miRNAs, mitochondrial DNA, and other DAMPs, can cross the compromised BBB and engage innate immune receptors, such as TLR4 and P2X7R, on glial cells, amplifying neuroimmune responses. In this review, we integrated recent findings on EV biogenesis, P2X7R signaling, and neurovascular dysfunction in the context of alcohol use disorder. We proposed a mechanistic model in which ethanol-triggered P2X7R activation drives EV release, turning these vesicles into inflammatory couriers that carry peripheral injury signals to the brain. We emphasize EV cargo as promising biomarkers of alcohol-related neurodegeneration and explore emerging therapies that target EV pathways or P2X7R to curb alcohol-induced CNS damage.
    Keywords:  Alcohol use disorder; Biomarkers; Blood–brain barrier; Extracellular vesicles; Inflammation; Neuroinflammation; Oxidative stress; P2X7 receptor
    DOI:  https://doi.org/10.1016/j.bbi.2025.106115
  15. Metab Brain Dis. 2025 Sep 26. 40(7): 276
      Type 2 Diabetes (T2D) and Alzheimer's Disease (AD) share common risk factors that can be seen through T2D nearly doubling an individual's likelihood of developing AD. Some AD patients show signs of metabolic dysfunction as well. This review focuses on the potential mechanisms associated with these two diseases, like insulin resistance, inflammation, oxidative damage, mitochondrial injury, and cell death. One of the notable elements in this connection is the "brain insulin resistance," most frequently named as "type 3 diabetes," which impairs glucose metabolism and facilitates amyloid beta (Aβ) plaque synthesis while reducing the action of insulin-degrading enzyme (IDE). Moreover, the overactivity of glycogen synthase kinase-3 beta (GSK-3β) also triggers taurine protein pathology. Raised concentrations of glucose in blood can produce advanced glycation end products (AGEs), which further exacerbate neuroinflammation in tandem with the mitigation of neurotoxic Aβ oligomers. Inflammation and subsequent damage to mitochondria lead to the dissolution of synapses. Current vascular insults include the breakdown of the blood-brain barrier (BBB) and decreased brain perfusion, along with other contributory factors to conditions conducive to neurotoxicity. Recently, novel therapies are emerging, including GLP-1 agonists, intranasal insulin, and mitochondrial antioxidants, that show surprising results for treating both conditions, but on the contrary, bioavailability and the timing of interventions remain a big challenge in the management of these diseases. Eventually, further research should center on understanding the mechanisms of integration along with the development of molecular biology, neuroimaging, and outcome-driven treatment strategies. Comprehensive strategies that exist between T2D-AD for integration and preservation of brain and metabolic health are addressed in this review.
    Keywords:  Advanced glycation end products (AGEs); Alzheimer’s disease (AD); Amyloid-beta (Aβ) accumulation; Insulin resistance; Mitochondrial dysfunction; Neuroinflammation; Type 2 diabetes mellitus (T2D)
    DOI:  https://doi.org/10.1007/s11011-025-01700-z
  16. Neurol Res. 2025 Sep 23. 1-15
       AIM: This study aims to assess the involvement of CD3+ T cells in the early stage of cerebral hemorrhage and investigate how depleting CD3+ T cells affects brain injury.
    METHODS: We proposed that CD3+ T cell infiltration aggravates hemorrhagic brain injury and observed the changes in brain injury by consuming these cells through the administration of anti-CD3 antibodies. Mice received an intraperitoneal injection of 50 μg of purified anti-CD3 monoclonal antibody 24 hours prior to the induction of intracerebral hemorrhage (ICH), with IgG antibody serving as the control. Results: Compared to the sham group, the ICH+IgG group showed a significant infiltration of CD3+ T cells and CD4+ T cells into the brain. This group also displayed elevated mNSS scores, a reduction in Nissl bodies, brain tissue disorganization, increased AQP4 protein expression, worsened brain edema, impaired BBB integrity, heightened inflammation, and decreased cerebral perfusion. Unexpectedly, depleting CD3+ T cells led to a reduction of these cells in both the circulation and brain, with CD4+ T cells being decreased, whereas CD8+ T cells showed an increase.
    CONCLUSION: CD3+ T cell infiltration can aggravate brain injury related to cerebral hemorrhage, while extensive consumption of CD3+ T cells disrupts immune homeostasis and further exacerbates the injury. Precise regulation of T cell subsets rather than total depletion may be a key strategy for ICH immunotherapy.
    Keywords:  CD3+ T cell; Immune cell infiltration; brain injury; immune dysregulation; intracerebral hemorrhage
    DOI:  https://doi.org/10.1080/01616412.2025.2559307