E-mail:BD@ebraincase.com
Tel:+8618971215294
English 中文版
Virus Vector - Viral Vector Production - BrainCaseVirus Vector - Viral Vector Production - BrainCase
  • Home
  • Virus product library
    CRISPRRNAiHSV-helperRV-helperNeurophilic virusCalcium SensorsOptogenetics activationOptogenetics inhibitionChemical geneticsSparse labelingFluorescent proteinBiosensorsRecombinaseApoptosis & AutophagyDisease ModelNeurotoxicityOther
  • Products & Service

    Product Center

    Virus

    VSV-vaccine and gene therapy research
    Retrovirus-RCAS-TVA
    Lentivirus Vector-Lentivirus Production
    Rabies Virus Vector-RBV Vector
    Herpes simplex virus-Oncolytic and anterograde tracing
    PRV-retrograd multisynaptic-Peripheral
    AAV-gene therapy vectors-neuroscience

    Animal Model

    Neurological Disease Models
    Tumor animal models-anti-tumor
    Digestive System Disease Animal Model
    Cardiovascular System Disease Animal Models

    Plasmid Construction

    Library Construction
    Plasmid design and construction

    Popular Applications

    Gene Regulation

    Gene Overexpression-Brain Case
    RNA interference(RNAi)-siRNA-Brain Case
    Gene Editing - CRSIPR cloning - BrainCase

    Neural Circuit Function Research

    Optogenetics - BrainCase
    Chemical genetics-DREADDs-Brain Case
    Calcium signal recording-Gels- Brain Case
    GRAB Neurotransmitter Fluorescent Sensor- Brain Case
    Functional Magnetic Resonance Imaging Technology- Brain Case

    Research on the structure of neural circuits

    Direct Input and Output-viral vectors- Brain Case
    Anterograde Mono-synaptic Tracing -HSV- Brain Case
    Antrograde Muti-synaptic Tracing-HSV & VSV-Brain Case
    Retrograde Mono-synaptic Tracing-Rabies Virus-Brain Case
    Retrograde Muti-synaptic Tracing-PRV-Brain Case

    Featured Services

    Nervous System Disease Drug Effect

    Alzheimer's disease-AD
    Depression-mental disorders
    Parkinson's disease--PD
    Epilepsy-an ancient neurological disorder

    AAV Serotype Screening

    AAV Serotypes screening-gene therapy

    Collaboration Products

    Kiryl Piatkevich lab
    Zhifei Fu lab
    Yulong Li lab

    Efficacy of Oncolytic Virus

    Oncolytic virus-for cancer therapy-Brain Case
    Herpes Virus Vector-anti-tumor- BrainCase
    Vesicular stomatitis virus-killing tumor cells-Brain Case
  • News
    Corporate News New Product Launch Team Investor News
  • Support
    Literature interpretation Customer article Video Zone FAQs
  • About Us
    Virus product library Products & Service News Support About Us Contact
  • Contact
    Contact Us Join us
  • 中文
    English 中文版
  • Home
  • News
  • New Product Launch
  • News
  • Corporate News
  • New Product Launch
  • Team
  • Investor News

Brain Case New Release|Rapid, Precise Construction of PD/AD/AS Animal Models

Time:2025-09-30 15:37:13
Animal modeling is a core tool in life science and medical research. By simulating human physiology and diseases, it avoids the ethical and practical limitations of human experimentation, providing a controllable platform for mechanism exploration and drug development. It serves as a critical bridge between basic research and clinical translation.

Traditional animal models—such as transgenic animals, chemical induction, surgical methods, and spontaneous disease models (natural mutation/breeding)—often suffer from long timelines, high costs, and complex operations. Recombinant adeno-associated virus (AAV), widely used as a delivery vector in gene therapy, offers significant advantages in animal model construction.
 

Key Advantages of AAV:

Fast & Easy: Enables efficient gene delivery through direct in vivo injection (e.g., brain or intravenous), eliminating complex embryonic manipulations and shortening the modeling cycle.

Stable, Long-Term Expression: Viral genomes remain episomal for months to years, supporting sustained gene expression ideal for chronic disease research.

Precise Targeting: Different serotypes exhibit distinct tissue tropisms, and further refinement via specific promoters or capsid engineering allows cell-level targeting.

Flexible Strategies: Supports diverse modeling approaches—gene-of-function (overexpression), loss-of-function (RNAi), and gene editing (CRISPR)—with spatial and temporal control of gene regulation.

Broad Applicability: Suitable for mice, rats, and non-human primates, facilitating studies in models with physiological structures closer to humans.

Mature & Cost-Effective: Established production processes and protocols enable stable, scalable, and economical implementation in laboratories.

With its speed, simplicity, high efficiency, and versatile strategies, AAV has become a preferred tool for constructing disease animal models, particularly for neurological and metabolic disorders.


 

Applications of AAV in Animal Modeling

Parkinson’s Disease (PD)
PD is a progressive neurodegenerative disorder of the central nervous system that primarily affects middle-aged and elderly individuals, leading to a range of motor and non-motor symptoms. Its key pathological features include progressive degeneration and loss of dopaminergic neurons in the substantia nigra pars compacta, along with the formation of Lewy bodies and Lewy neurites—intracellular inclusions mainly composed of abnormally aggregated α-synuclein (α-syn).

In disease model construction, AAV vectors carrying the α-syn gene (expressing either wild-type or mutant forms such as A53T) are commonly used. By overexpressing α-syn in specific brain regions, researchers can mimic its abnormal aggregation and pathogenic process, thereby establishing Parkinson’s disease–related animal models.

 

Case Study

1. mGluR5 Protects Against α-Synuclein–Induced Microglial Inflammation and Neurotoxicity in a Parkinson’s Disease Model
💠Experimental Animals: Sprague–Dawley rats
💠Viral Vector: AAV/9-α-syn
💠Injection Protocol: Stereotaxic injection into the substantia nigra (SN), 3 μL; 8.17 × 10¹³ vg/mL; 8-week expression period
💠Experimental Findings: To investigate the in vivo role of metabotropic glutamate receptor 5 (mGluR5) in α-synuclein–induced microglial activation and inflammatory responses in a PD model, AAV-α-syn or control AAV-GFP was injected into the right substantia nigra of rats. Behavioral assessments and pathological analyses were performed at multiple time points. Results showed that rats overexpressing α-syn developed significant motor impairments beginning 8 weeks after viral injection, persisting through 16 weeks, accompanied by progressive loss of dopaminergic neurons in the substantia nigra. α-syn levels increased while mGluR5 expression decreased. Inflammatory factors in microglia (iNOS, IL-1β, TNF-α) rose by day 10, peaked at 4 weeks, and subsided by 8 weeks. These findings indicate early reactive microglial inflammation during PD pathology, consistent with previous studies.

Figure 1. Inflammatory response associated with progressive neurodegeneration in a rat Parkinson’s disease model induced by AAV-α-syn.

 
2. Overexpression of A53T-α-Synuclein in the Substantia Nigra via AAV1/2 Induces Nigrostriatal Degeneration with Lewy Body–Like Pathology and Motor Dysfunction: A Novel Parkinson’s Disease Mouse Model
🔹Experimental Animals: Male wild-type mice
🔹Viral Vector: AAV1/2-A53T-α-syn
🔹Injection Protocol: Stereotaxic injection into the substantia nigra (SN), 1.5 μL; 5.16 × 10¹² vg/mL; 10-week expression period
🔹Experimental Findings: Using mice injected with AAV1/2-EV as controls, quantitative analysis was performed 10 weeks after injection. Results showed that in the AAV1/2-A53T-α-syn group, the number of TH⁺ dopaminergic neurons and total neurons in the ipsilateral substantia nigra was significantly reduced compared with controls. In addition, the ipsilateral striatal TH⁺ fiber optical density and presynaptic DAT levels decreased by 20% and 29%, respectively. These findings indicate that AAV1/2-A53T-α-syn injection induces neurodegeneration of the nigrostriatal pathway.

Figure 2. Injection of AAV1/2-A53T-α-syn into the substantia nigra leads to neurodegeneration of the nigrostriatal pathway.
 

Atherosclerosis (AS)

AS is a systemic disease related to lipid metabolism disorders. It is characterized by the deposition of lipid-rich plaques within arterial walls, leading to arterial narrowing and hardening.

In disease model construction, AAV vectors carrying the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene (wild-type or mutant) are commonly used. Through intravenous or local vascular injection, PCSK9 is overexpressed in the liver or vascular tissues, promoting abnormal low-density lipoprotein cholesterol (LDL-C) metabolism and accelerating lipid deposition in the arterial wall. This approach enables the establishment of atherosclerosis animal models and is often combined with a high-fat diet to enhance modeling efficiency.

 

Case Study

1. Induction of Atherosclerosis in Mice and Rats Without Germline Genetic Engineering
🔸Experimental Animals: C57 mice
🔸Viral Vector: rAAV8-D377Y-mPCSK9
🔸Injection Protocol: Tail vein injection of 2.0 × 10¹⁰, 1.0 × 10¹¹, or 5.0 × 10¹¹ vg; 12-week expression period
🔸Diet Regimens: Western diet – 21% fat and 0.21% cholesterol. Paigen diet – 16% fat, 1.25% cholesterol, and 0.5% sodium cholate
🔸Experimental Findings: Mice were sacrificed 12 weeks after rAAV8-D377Y-mPCSK9 injection for quantitative assessment of atherosclerosis. Control mice, regardless of diet, showed no or only minimal lesions. In contrast, all rAAV8-injected mice developed atherosclerosis in a dose-dependent manner. Histological analysis of aortic root lesions revealed advanced atherosclerotic plaques containing foam cells, smooth muscle cells, and fibrous tissue. Except for one high-dose rAAV8-D377Y-mPCSK9 mouse and one low-density lipoprotein receptor knockout (Ldlr−/−) mouse, all others exhibited necrotic core formation.

Figure 3. Atherosclerosis induced by rAAV8-D377Y-mPCSK9.

 
2. Ceramide Aggravates Atherosclerosis via CYSLTR2 and P2RY6 Sensing
🔺Experimental Animals: C57BL/6J mice
🔺Viral Vector: rAAV8-D377Y-hPCSK9
🔺Injection Protocol: Tail vein injection, 2.0 × 10¹¹ vg; 12-week expression period
🔺Diet Regimen: HFD — standard casein-based formula containing 1.25% cholesterol and 0.5% sodium cholate
🔺Experimental Findings: To explore whether CYSLTR2 and P2RY6 mediate ceramide-induced exacerbation of atherosclerosis, 8-week-old WT, Cysltr2⁻/⁻, P2ry6⁻/⁻, and double-knockout mice were injected with AAV8-hPCSK9-D377Y and fed a high-fat diet for 12 weeks. WT mice injected with AAV-Control and fed a normal diet served as blank controls. Starting at 10 weeks of age, “AAV-PCSK9 + high-fat” mice received intravenous C16:0 ceramide (5 mg/kg every 48 h). Results showed that C16:0 ceramide did not affect blood lipid levels but doubled plasma ceramide concentrations and significantly promoted plaque formation in WT mice. This effect was partially reduced in single knockouts and further attenuated in double knockouts, which exhibited less plaque formation, lipid accumulation, and macrophage infiltration. These findings demonstrate that CYSLTR2 and P2RY6 receptors synergistically mediate ceramide-induced aggravation of atherosclerosis.

Figure 4. Deletion of CYSLTR2/P2RY6 mitigates ceramide-aggravated atherosclerosis.
 
 

Alzheimer’s Disease (AD)

AD is a common age-related neurodegenerative disorder characterized by progressive cognitive decline and neuronal damage, with symptoms worsening over time. Neuropathologically, AD brains exhibit amyloid-β (Aβ) deposition forming senile plaques, hyperphosphorylated Tau protein aggregation forming neurofibrillary tangles (NFTs), as well as neuronal loss and synaptic damage.

In AD research, adeno-associated virus (AAV) vectors are frequently used to deliver AD-related pathogenic genes—such as mutant APP or mutant Tau—into specific brain regions (e.g., hippocampus, cortex) of experimental animals. This induces Aβ deposition, Tau aggregation, and cognitive impairment, creating animal models that closely replicate AD pathology and phenotype for mechanistic studies and drug screening.

 

Case Study

1. AAV-Mediated Tau Expression Induces Pyramidal Neuron Degeneration via Cell-Cycle Reentry Without Neurofibrillary Tangle Formation in Wild-Type Mice
💠Experimental Animals: Wild-type FVB/N mice
💠Viral Vector: rAAV1/2-hsyn-hTau (P301L)
💠Injection Protocol: Stereotaxic injection into the hippocampus, 1.0 × 10⁸ vg; 12-week expression period
💠Experimental Findings: Injection of AAV-Tau (P301L) into the mouse brain induced dose-dependent neurodegeneration. Low doses caused mild neuronal loss and thinning of the hippocampal CA1/2 region, with hTau levels decreasing as neuronal loss progressed due to reduced protein synthesis in degenerating neurons. Pathology first appeared in the CA2 region at 1.5 weeks post-injection, extended to CA1 by 3 weeks, and by 6–12 weeks nearly all CA pyramidal cells were lost, with degeneration spreading to the cortex. Fluoro-Jade B (FJB) staining confirmed degenerating neurons. Microglial activation emerged transiently and subsided by 12 weeks, while astrocyte activation persisted, indicating ongoing inflammation. Control mice injected with AAV-EGFP showed no neuronal loss or microglial activation. These results demonstrate that AAV-Tau (P301L) drives degeneration of hippocampal CA and cortical pyramidal neurons, with microglial activation specifically linked to Tau-mediated neurodegeneration rather than amyloid pathology or viral components.

Figure 5. Dose-dependent and temporal progression of neurodegeneration induced by AAV-Tau (P301L).
 
2. Hippocampal Overexpression of Human Tau to Establish a Non-Human Primate Model with Alzheimer’s-Like Pathology
🔸Animal Model: Adult rhesus monkeys (7–15 years old)
🔸Viral Vector: rAAV9-hTau (WT)
🔸Injection Protocol: Bilateral stereotaxic injections into the hippocampus, 10¹⁰–10¹¹ GC; expression for 6–12 weeks / up to 50 weeks
🔸Experimental Findings: To develop a non-human primate (NHP) model of Tauopathy with AD-like pathology, adult rhesus monkeys were injected bilaterally in the hippocampus with an AAV vector overexpressing human Tau. Immunostaining, PET, and MRI analyses revealed an AAV-mediated gene transduction efficiency of approximately 75%, with stable Tau expression maintained from 6 to 50 weeks. These results demonstrate that AAV enables efficient, widespread, and long-lasting Tau expression in the primate hippocampus, providing a robust NHP model for Alzheimer’s disease research.

Figure 6. Bilateral stereotaxic injection of AAV into the hippocampus achieves sustained high-level Tau expression in a non-human primate model.


 
Relevant AAV Modeling Tool Viruses Are All Available from Brain Case
Product Categories Product Number Product Name
PD Modeling BC-0997 rAAV-hSyn-mSNCA-P2A-EGFP
BC-1212 rAAV-hSyn-hSNCA-3xFLAG
BC-2789 rAVV-CMV-hSNCA(A30P A53T)
BC-1113 rAAV-CMV-α-synuclein(A30P A53T)-P2A-mCherry
BC-0237 rAAV-hSyn-hm-α-synuclein(A30P A53T)
AS Modeling BC-0463 rAAV-hAAT-mPCSK9(D377Y)
BC-1568 rAAV-hAAT-hPCSK9(D374Y)
AD Modeling BC-2097 rAAV-CMV-EGFP-Tau(human)
BC-2098 rAAV-CAG-Tau(human P301L)
BC-0136 rAAV-EF1a-DIO-Tau(human)-mCherry
BC-0131 rAAV-CaMKIlα-Tau(Bos mutus)-tdTomato
BC-0133 rAAV-hSyn-Tau(Bos mutus)-tdTomato

Service Type :

Select the service you'd like to purchase.

Order Information(Premade-AAVs)

Please provide us some information about the service you'd like to order.

How did you hear about BrainCase Biotech or our products?

Detailed requirements:

scroll

Order Information(Custom AAV/Lentivirus)

Please provide us some information about the service you'd like to order.

Gene ID or gene information:

Selection of the reporting gene:

Special Instructions:

scroll

Order Information(Others)

Please provide us some information about the service you'd like to order.

Virus name/Detailed requirements:

scroll

Related products

Brain Case New Release|Rapid, Precise Construction of PD/AD/AS Animal Models

Brain Case New Release|Rapid, Precise Construction of PD/AD/AS Animal Models

Nature Methods|mYongHong: A New Generation of Ultra-Stable Monomeric Red Fluorescent Protein

Nature Methods|mYongHong: A New Generation of Ultra-Stable Monomeric Red Fluorescent Protein

[Research Tool Highlight] Featured in Cell! Brain Case launches new product — Enhancer AAV Vectors, a breakthrough for  research Tool !

[Research Tool Highlight] Featured in Cell! Brain Case launches new product — Enhancer AAV Vectors, a breakthrough for research Tool !

Brain Case New Arrival | Novel High-Brightness Red PRV Virus – A Powerful Tool for Peripheral Nervous System Research

Brain Case New Arrival | Novel High-Brightness Red PRV Virus – A Powerful Tool for Peripheral Nervous System Research

map
{dede:global.cfg_webname/}

Virus product library

CRISPR
RNAi
Neurophilic virus
Optogenetics activation
Biosensors

News

Corporate News
New Product Launch
Team
Investor News

Support

Literature interpretation
Customer article
Video Zone
FAQs
微信

WhatsApp Business Account

Tel: +8618971215294
E-mail: BD@ebraincase.com

Address:-

Address:-

  • Copyright © 2024 Brain Case All Rights Reserved.