Engineered Microglia: A Paradigm Shift in Blood-Brain Barrier Navigation
- Bryan White
- Dec 31, 2025
- 14 min read

Abstract
The central nervous system (CNS) remains the most fortified and inaccessible organ in the human body, sequestered behind the Blood-Brain Barrier (BBB). This physiological rampart, while essential for maintaining neuronal homeostasis, has historically thwarted the delivery of therapeutic biologics, rendering the vast majority of neurodegenerative and metabolic brain disorders untreatable. In 2025, a paradigm-shifting therapeutic platform emerged from the University of California, Irvine (UCI), specifically from the laboratory of Professor Mathew Blurton-Jones. This approach, colloquially termed the "Trojan Horse" therapy, bypasses the BBB not by breaching its integrity, but by replacing the brain's resident immune sentinels—the microglia—with genetically engineered, inhibitor-resistant human induced pluripotent stem cell-derived microglia (iMG).
This report provides an exhaustive, multi-dimensional analysis of this breakthrough. We detail the molecular engineering of the CSF1R-G795A receptor variant, a critical innovation described in Molecular Therapy (November 2025) that permits the chemical ablation of endogenous microglia while sparing engineered donor cells, thereby enabling near-total engraftment (>99%) of the microglial niche. We subsequently explore the application of this platform in two landmark studies: the pathology-responsive delivery of neprilysin to resolve amyloidosis in Alzheimer's Disease models (Cell Stem Cell, April 2025), and the restoration of β-hexosaminidase activity to reverse lethal neurodegeneration in Sandhoff Disease (Nature Communications, August 2025). By synthesizing data on molecular mechanisms, preclinical efficacy, and translational feasibility, this report posits that the "Trojan Horse" microglial platform represents a foundational shift in neurotherapeutics—moving from passive drug delivery to active, cellular biological remediation.
1. Introduction: The Fortress and the Failure
1.1 The Physiological Imperative of the Blood-Brain Barrier
To understand the magnitude of the innovation developed at UCI, one must first appreciate the obstacle it overcomes. The human brain consumes approximately 20% of the body's total energy budget while constituting only 2% of its mass. This extreme metabolic demand supports a complex electrochemical signaling network that is exquisitely sensitive to fluctuations in ionic composition and the presence of neuroactive toxins. Evolutionary pressure has thus selected for a rigorous exclusionary interface: the Blood-Brain Barrier (BBB).
First conceptualized by Paul Ehrlich in the late 19th century and defined by Edwin Goldmann’s dye experiments in 1913, the BBB is not a static wall but a dynamic, metabolically active interface. It is composed of continuous non-fenestrated endothelial cells linked by tight junctions (claudins, occludins, and junctional adhesion molecules).1 These endothelial cells are ensheathed by pericytes and the end-feet of astrocytes, forming the "neurovascular unit." This unit effectively segregates the CNS interstitial fluid from the systemic circulation, preventing the entry of pathogens, immune cells, and hydrophilic molecules.1
1.2 The Pharmacological Graveyard
While the BBB is vital for survival, it is the primary reason for the stagnation in neuropharmacology. The "Lipinski Rule of 5," which guides oral drug design, fails at the BBB. It is estimated that 98% of all small-molecule drugs and nearly 100% of large-molecule biologics (recombinant proteins, monoclonal antibodies, gene therapy vectors) cannot cross the BBB in therapeutically relevant concentrations via passive diffusion.1
Historical attempts to circumvent the barrier have generally fallen into three categories, each with critical limitations:
Invasive Delivery: Intracerebroventricular (ICV) or intraparenchymal injections are highly invasive, carry risks of infection, and due to the brain's tortuous extracellular space, achieve very poor tissue distribution (often limited to a few millimeters from the needle track).
Molecular Trojan Horses (Antibody-Fusion): This strategy involves fusing a drug payload to an antibody targeting a receptor capable of transcytosis, such as the Transferrin Receptor (TfR) or Insulin Receptor (IR). While theoretically sound, this approach faces "lysosomal trapping," where the endothelial cell endocytoses the complex but degrades it in the lysosome rather than releasing it into the brain. Furthermore, high affinity for TfR can trap the drug in the vasculature, preventing deep parenchymal penetration.4
Viral Vectors (AAV): Adeno-associated viruses (AAVs), particularly serotypes like AAV9 and AAV-PHP.B, have shown the ability to cross the BBB. However, their clinical utility is hamstrung by immunogenicity (pre-existing neutralizing antibodies), limited cargo capacity (<4.7 kilobases), and the risk of hepatotoxicity due to high systemic accumulation.6
1.3 The Cellular Trojan Horse: A Biological Precedent
The concept of a "Trojan Horse" entering the brain is not novel to medicine; nature perfected it eons ago. Pathogens such as Cryptococcus neoformans, HIV, and Zika virus utilize a "Trojan Horse" mechanism to infect the CNS. They infect peripheral immune cells (monocytes or macrophages) and ride inside them as they migrate across the BBB during inflammation.8
The Blurton-Jones Lab at UCI asked a provocative question: If pathogens can use immune cells to smuggle destruction into the brain, can we use the same cells to smuggle a cure?
2. The Microglial Renaissance: Ontogeny as Opportunity
To engineer a cellular vehicle for the brain, one must choose the right cell. Peripheral macrophages are transient visitors to the CNS, usually restricted to perivascular spaces or entering only during severe trauma. The true residents of the brain are the microglia.
2.1 Distinct Ontogeny and Function in Microglia
Microglia are the resident macrophages of the CNS, comprising 10-15% of all brain cells. Unlike other neural cells (neurons, astrocytes, oligodendrocytes) which are derived from the neuroectoderm, and unlike peripheral macrophages which are derived from bone marrow hematopoietic stem cells (HSCs), microglia have a unique origin. They originate from erythro-myeloid progenitors (EMPs) in the yolk sac during early embryogenesis (around embryonic day 8.5 in mice). They migrate into the developing brain before the BBB closes and establish a self-renewing population that persists for the lifespan of the organism.11
This distinction is crucial for therapy. Bone marrow-derived cells (monocytes) can enter the brain after irradiation and bone marrow transplant, but they do not become true microglia. They retain a distinct transcriptomic signature and often fail to integrate into the neuronal network in a homeostatic manner. True microglia possess unique functions—synaptic pruning, surveillance, and neurotrophic support—that make them the ideal "gardeners" of the CNS.11
2.2 The Challenge of the "Full House"
The therapeutic use of microglia faces a physical problem: the microglial niche. The brain has a specific carrying capacity for microglia. These cells exhibit "tiling" behavior, maintaining distinct, non-overlapping territories. Under normal conditions, the niche is fully occupied. If one were to inject healthy donor microglia into a patient, the endogenous (dysfunctional) cells would outcompete them, or the donors would die from lack of survival signals.13
Therefore, to install a new microglial system, one must first delete the old one.
3. The Technical Breakthrough: The CSF1R-G795A Platform
The foundational technology underpinning the UCI approach is not the therapeutic payload, but the engraftment platform. This platform was detailed in a seminal paper published in Molecular Therapy in November 2025, titled "CSF1R inhibitor-resistant model for CNS-wide microglia replacement strategies".15
3.1 The Achilles' Heel: CSF1R Dependency
Microglia are strictly dependent on signaling through the Colony Stimulating Factor 1 Receptor (CSF1R) for survival. The binding of ligands CSF1 or IL-34 to this receptor triggers tyrosine kinase activity, phosphorylation cascades (PI3K/Akt, MAPK), and subsequent survival gene expression.
Small-molecule inhibitors of CSF1R, such as PLX3397 (pexidartinib) and PLX5622, have revolutionized microglial research. These drugs can cross the BBB and, by blocking the ATP-binding pocket of the receptor, cause rapid apoptosis of >99% of microglia in the brain.13 However, these drugs have a limitation as a conditioning agent: they kill all microglia, including any potential donor cells one might try to transplant. Furthermore, once the drug is stopped, the few remaining endogenous survivors proliferate explosively to repopulate the brain.
3.2 Engineering the "Shield": The G795A Mutation
To create a donor cell that could survive this chemical assault, the Blurton-Jones team looked to the structure of the receptor itself. Through molecular modeling and mutagenesis screens, they identified a single point mutation in the human CSF1R gene: a substitution of Glycine with Alanine at position 795 (G795A).13
This mutation is located within the kinase domain of the receptor. Its impact is precise:
Inhibitor Resistance: The additional methyl group of the Alanine residue creates steric hindrance in the ATP-binding pocket. This prevents bulky inhibitor molecules like PLX3397 and PLX5622 from binding effectively.
Functional Integrity: Crucially, the mutation does not interfere with the binding of the natural ligands (CSF1/IL-34) or the conformational changes required for kinase activation. The receptor remains functional; it simply becomes "invisible" to the drug.13
3.3 The Engraftment Protocol: A "Condition-and-Replace" Strategy
This genetic innovation allowed for a novel transplantation protocol, described by lead author Jean Paul Chadarevian:
Genome Editing: Human iPSCs are edited using CRISPR-Cas9 to introduce the G795A mutation into the CSF1R locus.19
Differentiation: These iPSCs are differentiated into hematopoietic progenitor cells (HPCs) and then into microglia (iMG) using a specialized cytokine cocktail (IL-34, TGF-β1, M-CSF).20
Transplantation & Selection: The G795A-iMG are injected into the recipient brain. Simultaneously, the recipient is treated with PLX3397.
Niche Takeover: The inhibitor wipes out the endogenous microglia. The G795A-iMG, impervious to the drug, sense the "empty niche" and the abundance of survival ligands. They proliferate rapidly, migrating away from the injection site to colonize the entire CNS parenchyma.13
The result is a brain where the immune system has been swapped out—>99% of the microglia are now human, engineered, and permanent.13
4. Therapeutic Application I: Targeted Proteolysis in Alzheimer's Disease
With the delivery platform established, the UCI team turned to their primary target: Alzheimer's Disease (AD). This work culminated in a publication in Cell Stem Cell in April 2025.1
4.1 The Amyloid Hypothesis and Clearance Failure
A neuropathological hallmark of AD is the accumulation of extracellular plaques composed of amyloid-beta (Aβ) peptides. In the healthy brain, Aβ is cleared by enzymes such as Neprilysin (NEP) and Insulin-Degrading Enzyme (IDE). In AD, these clearance mechanisms fail, leading to oligomerization, synaptic toxicity, and plaque formation.1
Previous attempts to restore NEP levels via viral gene therapy failed because constitutive overexpression of NEP is deleterious—it degrades other essential neuropeptides (e.g., enkephalins, substance P). The therapy needed to be "smart."
4.2 The "Smart" Cargo: Pathology-Responsive Neprilysin
The Blurton-Jones lab engineered their G795A-iMG to carry a payload: a gene for secreted neprilysin (sNEP). However, they placed this gene under the control of a plaque-responsive promoter.
The Sensor: Microglia naturally transition into a "Disease-Associated Microglia" (DAM) state when they encounter amyloid plaques. This state is defined by the upregulation of specific genes (e.g., CD9, APOE, CLEC7A).
The Circuit: By driving sNEP expression with a DAM-specific promoter, the researchers ensured that the enzyme was only produced when the microglia were physically interacting with a plaque. In healthy regions of the brain, the cells remained quiescent.
4.3 Preclinical Efficacy in 5xFAD Mice
The engineered cells were transplanted into 5xFAD mice, an aggressive model of amyloidosis. The results were visually and statistically profound:
Global Plaque Reduction: Immunohistochemical analysis revealed that the G795A-sNEP microglia clustered around plaques and significantly reduced the total amyloid burden across the hippocampus and cortex.1
Synaptic Preservation: The ultimate goal of AD therapy is not just to clean plaques, but to save synapses. Using synaptophysin staining as a proxy for synaptic density, the study found that treated mice retained synaptic levels comparable to wild-type controls, whereas untreated 5xFAD mice showed severe synaptic loss.1
Mechanism of Action: The sNEP degraded the local high concentrations of Aβ oligomers—the species most toxic to synapses—creating a "halo of protection" around the neurons.3
Jean Paul Chadarevian summarized the findings: "Remarkably, we found that placing the microglia in specific brain areas could reduce toxic amyloid levels and other AD-associated neuropathologies throughout the brain".22
5. Therapeutic Application II: Metabolic Cross-Correction in Sandhoff Disease
The versatility of the platform was further demonstrated in August 2025 with a publication in Nature Communications focusing on Sandhoff Disease, a prototypical Lysosomal Storage Disorder (LSD).23
5.1 The Metabolic Defect: Hexosaminidase Deficiency
Sandhoff disease is a fatal genetic disorder caused by mutations in the HEXB gene. This leads to a deficiency in both β-hexosaminidase A and B enzymes. Without these enzymes, GM2 gangliosides cannot be degraded and accumulate to toxic levels within the lysosomes of neurons, leading to cellular ballooning, dysfunction, and rapid neurodegeneration.24
5.2 The Concept of Cross-Correction
Unlike AD, where the problem is extracellular junk (plaques), LSDs involve intracellular storage. How can a microglia cell fix a neuron's lysosome? The answer lies in Mannose-6-Phosphate (M6P) trafficking.
Secretory Pathway: A fraction of lysosomal enzymes are normally secreted into the extracellular space. These enzymes carry M6P tags.
Uptake: Neighboring cells (neurons) express M6P receptors on their surface, which bind the secreted enzymes and traffic them to their own lysosomes.This phenomenon, known as cross-correction, means that a small number of healthy cells can serve as "enzyme factories" for the entire tissue.4
5.3 Restoration of the Hex-GM2-MGL2 Axis
The Blurton-Jones team transplanted G795A-iMG expressing functional HEXB into Hexb-/- knockout mice (a model of Sandhoff disease).
Biochemical Rescue: The transplanted microglia successfully secreted functional β-hexosaminidase. Lipidomic analysis confirmed a massive reduction in GM2 ganglioside levels in the brain parenchyma.24
Neuroprotection: The treatment normalized the lysosomal phenotype of neurons (reducing the characteristic "storage bodies") and reversed apoptotic gene signatures.
Behavioral Improvement: Treated mice showed significantly improved performance on motor tasks (e.g., rotarod) and extended survival compared to untreated controls.24
This study established the critical role of the "microglia-neuron Hex-GM2-MGL2 axis" in maintaining brain homeostasis, proving that myeloid-derived enzymes are sufficient to prevent neurodegeneration.26
6. Emerging Frontiers and Broader Implications
The success in AD and Sandhoff disease suggests that the G795A-iMG platform could serve as a "universal chassis" for CNS drug delivery.
6.1 Sanfilippo Syndrome (MPS IIIA)
Parallel work by Alina Chadarevian and the UCI team is targeting Mucopolysaccharidosis Type IIIA (Sanfilippo A), caused by SGSH deficiency. Preliminary data presented at the 2025 UCI Stem Cell Symposium suggests that transplantation of gene-corrected iMG can reduce heparan sulfate accumulation and improve cognitive function in MPS IIIA mice.29
6.2 Neuro-Oncology: Glioblastoma
The lab has also begun exploring the use of these cells in neuro-oncology. Glioblastomas are notorious for recruiting microglia and converting them into immunosuppressive "Tumor-Associated Macrophages" (TAMs).
The Strategy: By transplanting engineered microglia that are resistant to this conversion and programmed to secrete anti-tumor cytokines (e.g., IL-12, IFN-Γ) or oncolytic viruses, the team aims to turn the tumor's support system into its executioner.22
6.3 Multiple Sclerosis (MS)
In demyelinating disorders, microglia are essential for clearing myelin debris to allow oligodendrocyte precursor cells (OPCs) to remyelinate axons. The G795A platform could be used to deliver trophic factors (IGF-1, BDNF) to stimulate this repair process.22
7. Comparative Analysis: The "Trojan Horse" vs. The Field
To contextualize the significance of the UCI platform, it is instructive to compare it with existing therapeutic modalities.
Feature | AAV Gene Therapy | Antibody "Trojan Horses" | HSC Transplant (BMT) | UCI Microglia Platform |
Delivery Vehicle | Viral Capsid (e.g., AAV9) | Fusion Protein (mAb-Drug) | Monocyte / Macrophage | Resident Microglia (iMG) |
Barrier Penetration | Variable / inefficient | Receptor-mediated (saturable) | Requires toxic conditioning | Direct Replacement |
Duration of Effect | Transient to Long-term | Short (requires weekly doses) | Permanent | Permanent (Self-renewing) |
Cargo Capacity | Limited (<4.7 kb) | Limited by protein stability | Full Genome | Unlimited (Genome Editing) |
Spatial Control | Non-specific / Global | Global / Vascular trap | Perivascular | Pathology-Targeted (Chemotactic) |
Toxicity Profile | Immunogenic / Hepatotoxic | Systemic exposure | GvHD / Chemotherapy risks | Low (CNS-restricted) |
Table 1: Comparative analysis of CNS delivery modalities. The UCI Microglia Platform offers distinct advantages in duration, capacity, and spatial precision.
The data clearly indicates that while AAVs and antibodies struggle with "getting in," the microglial platform solves the problem by "moving in."
8. Translational Roadmap and Challenges
8.1 Manufacturing and Scalability (GMP)
The transition from preclinical mouse models to human patients requires a massive scale-up in cell production.
Differentiation: The current protocol to generate iMG takes 30-40 days. Automating this process under Good Manufacturing Practice (GMP) standards is a major logistical hurdle.20
Allogeneic Banking: To make the therapy economically viable, the field must move away from patient-specific (autologous) cells. The solution lies in hypoimmunogenic iPSC lines (e.g., HLA-class I/II knockout + CD47 overexpression). This would allow for the creation of a "universal donor" bank of G795A-iMG, drastically reducing costs and lead times.32
8.2 Safety Considerations: The "Kill Switch"
The permanence of the graft is its greatest strength but also its primary safety risk. If the engineered cells were to undergo malignant transformation or cause unchecked inflammation, they would be difficult to remove.
Mitigation: The G795A cells are resistant to specific inhibitors (PLX3397). They likely remain susceptible to other tyrosine kinase inhibitors (TKIs) targeting different residues, or to high-dose chemotherapy.
Suicide Genes: Clinical-grade cell lines will almost certainly incorporate an inducible suicide gene, such as iCaspase9 (activated by a distinct small molecule) or HSV-TK (activated by ganciclovir), providing a fail-safe mechanism to ablate the graft if adverse events occur.1
8.3 Ethical Considerations
The therapy involves the permanent replacement of a substantial portion of the patient's brain cells (the immune system). While mouse data suggests the human cells integrate homeostatically 13, the long-term cognitive and psychological implications of hosting "alien" microglia—even if human—remain a topic for future bioethical discourse.
9. Conclusion
The "Trojan Horse" microglia therapy developed by the Blurton-Jones Lab at UC Irvine represents a seminal moment in the history of neurotherapeutics. It marks the transition from the era of passive delivery—where we struggled to push molecules across the unyielding wall of the BBB—to the era of active remediation, where we install a living, programmable repair crew inside the fortress itself.
By combining the unparalleled plasticity of induced pluripotent stem cells, the precision of CRISPR gene editing, and the unique biology of the microglial niche, this platform offers a solution that is as elegant as it is powerful. Whether degrading the plaques of Alzheimer's or replenishing the enzymes of Sandhoff disease, these "living couriers" provide a level of spatial precision and temporal control that no synthetic drug can match. As the technology moves toward clinical trials, it carries with it the promise of not just treating, but potentially curing, some of the most devastating diseases of the human experience.
Citations
1: UCI News / Cell Stem Cell press release regarding Alzheimer's/Neprilysin study.
13: Chadarevian et al., dissertation/paper on G795A mechanism.
3: ScienceDaily summary of Cell Stem Cell paper.
8: Chadarevian et al., Molecular Therapy (Nov 2025) on resistance mechanism.
17: Spangenberg et al., on PLX3397 depletion in 5xFAD mice.
4: Pardridge, W.M., on BBB transport mechanisms and Trojan Horse antibodies.
24: Tsourmas et al., Nature Communications (Aug 2025) on Sandhoff disease.
22: News-Medical interview with Jean Paul Chadarevian.
13: Detailed mechanism of G795A mutation.
26: Abstract on the Hex-GM2-MGL2 axis.
20: Protocol for generating G795A iMG.
Works cited
Engineered microglia show promise for treating Alzheimer's and ..., accessed December 30, 2025, https://mind.uci.edu/jpc_cellstemcell/
Harnessing the brain's repair crew - UC Irvine News, accessed December 30, 2025, https://news.uci.edu/2025/10/16/harnessing-the-brains-repair-crew/
Engineered microglia show promise for treating Alzheimer's and other brain diseases, accessed December 30, 2025, https://www.sciencedaily.com/releases/2025/04/250421162825.htm
A multicenter open-label extension study of intrathecal heparan-N-sulfatase in patients with Sanfilippo syndrome type A - ResearchGate, accessed December 30, 2025, https://www.researchgate.net/publication/353099502_A_multicenter_open-label_extension_study_of_intrathecal_heparan-N-sulfatase_in_patients_with_Sanfilippo_syndrome_type_A
Tau-specific immunotherapeutic strategies for the treatment of tauopathies - UQ eSpace - The University of Queensland, accessed December 30, 2025, https://espace.library.uq.edu.au/view/UQ:89e5155/s4354085_phd_thesis.pdf
Gene Therapy Approach with an Emphasis on Growth Factors: Theoretical and Clinical Outcomes in Neurodegenerative Diseases - PubMed Central, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8518903/
A novel gene editing system to treat both Tay-Sachs and Sandhoff diseases - PMC, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7260097/
CSF1R inhibition by PLX5622 reduces pulmonary fungal infection by depleting MHCIIhi interstitial lung macrophages | Request PDF - ResearchGate, accessed December 30, 2025, https://www.researchgate.net/publication/383298179_CSF1R_inhibition_by_PLX5622_reduces_pulmonary_fungal_infection_by_depleting_MHCIIhi_interstitial_lung_macrophages
Microglia in antiviral immunity of the brain and spinal cord - PMC - PubMed Central, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9934594/
Stem cell stories that caught our eye: the tale of a tail that grows back and Zika's devious Trojan Horse, accessed December 30, 2025, https://blog.cirm.ca.gov/2017/11/03/stem-cell-stories-that-caught-our-eye-the-tale-of-a-tail-that-grows-back-and-zikas-devious-trojan-horse/
Therapeutic efficacy of intracerebral hematopoietic stem cell gene therapy in an Alzheimer's disease mouse model - PubMed Central, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC11399302/
Monocytes can efficiently replace all brain macrophages and fetal liver monocytes can generate bona fide SALL1+ microglia - PubMed Central, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12094688/
UC Irvine - eScholarship, accessed December 30, 2025, https://escholarship.org/content/qt8zb198pz/qt8zb198pz.pdf
Engineering an inhibitor-resistant human CSF1R variant for microglia replacement - PMC, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9814156/
Mathew Blurton-Jones - Cure Alzheimer's Fund, accessed December 30, 2025, https://curealz.org/researchers/mathew-blurton-jones/
CSF1R inhibitor-resistant model for CNS-wide microglia replacement strategies, accessed December 30, 2025, https://www.researchgate.net/publication/397541641_CSF1R_inhibitor-resistant_model_for_CNS-wide_microglia_replacement_strategies
Eliminating microglia in Alzheimer's mice prevents neuronal loss without modulating amyloid-β pathology | Brain | Oxford Academic, accessed December 30, 2025, https://academic.oup.com/brain/article/139/4/1265/2464332
genetic modification of mammalian cells to confer resistance to csf1r antagonists - Justia Patents, accessed December 30, 2025, https://patents.justia.com/patent/20230248775
Therapeutic potential of human microglial transplantation in a chimeric model of CSF1R-related leukoencephalopathy - PMC - PubMed Central, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12357648/
(PDF) CRISPR generation of CSF1R-G795A human microglia for robust microglia replacement in a chimeric mouse model - ResearchGate, accessed December 30, 2025, https://www.researchgate.net/publication/372767475_CRISPR_generation_of_CSF1R-G795A_human_microglia_for_robust_microglia_replacement_in_a_chimeric_mouse_model
Engineered microglia show promise for treating Alzheimer's, other brain diseases, accessed December 30, 2025, https://news.uci.edu/2025/04/21/engineered-microglia-show-promise-for-treating-alzheimers-other-brain-diseases/
Engineered brain cells offer new hope for treating Alzheimer's - News-Medical.Net, accessed December 30, 2025, https://www.news-medical.net/news/20250421/Engineered-brain-cells-offer-new-hope-for-treating-Alzheimers.aspx
The evolution of microglia replacement: A new paradigm for CNS disease therapy, accessed December 30, 2025, https://www.researchgate.net/publication/398341395_The_evolution_of_microglia_replacement_A_new_paradigm_for_CNS_disease_therapy
Microglial replacement in a Sandhoff disease mouse model reveals myeloid-derived β-hexosaminidase is necessary for neuronal health - PubMed, accessed December 30, 2025, https://pubmed.ncbi.nlm.nih.gov/40866328/
New Approaches to Tay-Sachs Disease Therapy - Frontiers, accessed December 30, 2025, https://www.frontiersin.org/journals/physiology/articles/10.3389/fphys.2018.01663/pdf
Microglia–neuron crosstalk through Hex–GM2–MGL2 maintains brain homeostasis - PMC, accessed December 30, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12545202/
Brain-wide microglia replacement using a nonconditioning strategy ameliorates pathology in mouse models of neurological disorders | Request PDF - ResearchGate, accessed December 30, 2025, https://www.researchgate.net/publication/391330173_Brain-wide_microglia_replacement_using_a_nonconditioning_strategy_ameliorates_pathology_in_mouse_models_of_neurological_disorders
(PDF) Microglia–neuron crosstalk through Hex–GM2–MGL2 maintains brain homeostasis, accessed December 30, 2025, https://www.researchgate.net/publication/394368285_Microglia-neuron_crosstalk_through_Hex-GM2-MGL2_maintains_brain_homeostasis
Harnessing human iPSC-microglia for CNS-wide delivery of disease, accessed December 30, 2025, https://ouci.dntb.gov.ua/en/works/4zXXzNqw/
Recent Updates – Page 3 – Blurton-Jones Lab - UC Irvine, accessed December 30, 2025, https://faculty.sites.uci.edu/blurtonjoneslab/recent-updates/page/3/
UCI Stem Cell Research Center Sue & Bill Gross Hall February 12th, 2025 9:00 AM - 7:00 PM, accessed December 30, 2025, https://stemcell.uci.edu/Flyers/2025/2025-Symposium-Program-SCRC.pdf
Engineering an inhibitor-resistant human CSF1R variant for microglia replacement, accessed December 30, 2025, https://rupress.org/jem/article-abstract/220/3/e20220857/213788/Engineering-an-inhibitor-resistant-human-CSF1R?redirectedFrom=fulltext
Replacing brain immune cells in mice slows neurodegeneration in Stanford Medicine study, accessed December 30, 2025, https://med.stanford.edu/news/all-news/2025/08/brain-immune-cells.html



Comments