Preclinical WebsiteClinical Website

Biospective is a global preclinical CRO specializing in Parkinson’s disease mouse models, with deep expertise in alpha-synuclein pathology. We support biotech and pharmaceutical drug development programs using validated Parkinson’s disease models for efficacy, biodistribution, mechanism-of-action, target engagement, and PK/PD studies across small molecules, antisense oligonucleotides, gene therapy, antibodies, and other biologics. Biospective’s alpha-synuclein PFF and AAV-A53T mouse models recapitulate key features of human Parkinson’s disease, including protein aggregation, neuroinflammation, dopaminergic neuron loss, and motor dysfunction. Studies include translational biomarkers such as MRI and PET imaging, fluid biomarkers including neurofilament light chain (NfL), and quantitative IHC. With fully integrated, end-to-end preclinical services, and over a decade of continuous experience executing Parkinson’s disease contract research studies in animal models, Biospective enables translational Parkinson’s disease research from study design through data interpretation.

Why Choose Biospective as Your Parkinson’s Disease CRO?

  • Specialized Parkinson's Disease CRO: Focused exclusively on Parkinson’s and neurodegenerative disease models, not a generalist animal provider.

  • Multiple Validated Models: Transgenic models, viral vector induced, and seeding-based rodent models of Parkinson’s disease are readily available for studies.

  • Alpha-Synuclein Expertise: Deep scientific expertise in α-synuclein biology and pathology, a central misfolded protein in PD.

  • Integrated Services: Fully integrated preclinical services from study design to data interpretation, ensuring seamless execution.

  • Proven Efficacy Data: Industry-standard α-synuclein efficacy datasets and extensive historical controls for robust benchmarking.

  • In-House Transgenic Colony: Internal colony of A53T α-synuclein transgenic mice (M83 line), enabling rapid study start with well-characterized animals.

  • Accelerated Timelines: Rapid study initiation and efficient workflows to compress timelines without sacrificing quality.

  • Translational Biomarkers: Advanced biomarkers (MRI, PET imaging, CSF/blood assays) that bridge preclinical findings to clinical outcomes.

  • Flexible Study Designs: Our scientists work with your team to customize the study design to best fit your goals.
  • Global Support: Experience supporting biotech and pharmaceutical industry clients worldwide, with responsive project management and communication.

Our scientists work as an extension of your internal team, collaborating closely to ensure scientific rigor, reproducibility, and translational relevance at every stage of your Parkinson’s disease research program.

Alpha-Synuclein Mouse Models – Our Core Expertise

Alpha-synuclein protein aggregation and propagation are central to Parkinson’s disease pathophysiology. Biospective has built specialized capabilities around α-synuclein–based PD animal models, making this a core differentiator of our CRO services.

These rodent models enable direct evaluation of target engagement and downstream neurodegenerative processes under pathological conditions. Notably, our α-synuclein models recapitulate key features of human Parkinson’s disease – including α-synuclein aggregation, dopaminergic neuron loss, neuroinflammation, motor impairments, and even non-motor symptoms such as sleep disturbances. Our animal model portfolio emphasizes reproducibility, well-defined phenotypes, and the integration of behavioral, imaging, biochemical, molecular, and histopathological endpoints to enable comprehensive in vivo Parkinson's disease efficacy studies and exploration of mechanism-of-action.

Alpha-Synuclein Preformed Fibril (PFF) Mouse Model

The pathologic spread of misfolded α-synuclein that characterizes human Parkinson’s disease can be recapitulated in rodent brains by stereotaxic injection of α-synuclein preformed fibrils (PFFs). In this PFF seeding and spreading model, exogenous recombinant α-synuclein fibrils seed the aggregation of endogenous α-synuclein and propagate pathology across the brain. This model can be induced in transgenic mice overexpressing human α-syn (e.g. M83 mice with the A53T mutation) as well as in wild-type mice or rats. This model is considered one of the best PD animal models for drug testing given its strong translational relevance.

α-Synuclein PFF Model Induction:

  • Injection of recombinant α-synuclein preformed fibrils into the CNS 
  • Applicable to A53T α-synuclein transgenic mice (M83 line) or wild-type mice

Validated PFF Injection Sites:

  • Anterior Olfactory Nucleus (AON) - an early-affected region in PD (Braak stage 1)
  • Medial Forebrain Bundle (MFB)
  • Striatum (± overlying cortex)

Disease Features Modeled (PFF):

  • Progressive spreading of α-synuclein pathology in a well-defined spatiotemporal pattern
  • Neuroinflammation (microglial and astroglial activation)
  • Neurodegeneration (loss of vulnerable neuronal populations)
  • Measurable behavioral impairments: motor deficits and non-motor symptoms (e.g. sleep disturbances)

Our α-synuclein PFF-induced PD models are highly reproducible and widely regarded as a gold standard for testing disease-modifying therapeutics. The PFF mouse model is commonly used to evaluate therapies targeting α-synuclein aggregation, propagation, and neurodegeneration in Parkinson’s disease. Biospective has 10+ years of experience executing preclinical studies with α-syn PFF models to evaluate therapeutic biodistribution, target engagement, mechanism of action, and efficacy

3 validated injection sites of the Anterior Olfactory Nucleus (AON)

Our validated injection sites: Anterior Olfactory Nucelus (AON). Striatum +/- Overlying Cerebral Cortex, and Medial Forebrain Bundle (MFB).

IHC images showing phosphorylated α-synuclein of piriform cortex

Phosphorylated α-synuclein (pSyn129) IHC of ipsilateral (left) and contralateral (right) piriform cortex 12 weeks after unilateral α-synuclein PFF injection into the AON of an M83+/- mouse.

Graphs showing elevated NF-L levels in plasma

Highly elevated levels of neurofilament light (NF-L) are observed in the plasma from the α-synuclein fibril seeding mice.

AAV-A53T Alpha-Synuclein Mouse Model

Generation of α-synuclein pathology in the adult rodent brain can also be achieved via injection of adeno-associated virus (AAV) vectors encoding mutant α-synuclein. In this PD mouse model, adult wild-type (C57BL/6) mice (or suitable transgenic backgrounds) receive a unilateral stereotaxic injection of an AAV vector overexpressing human α-synuclein with the A53T mutation directly into the substantia nigra pars compacta (SNc). Our skilled surgical team uses high-precision digital stereotaxic guidance with automated microinjectors for accurate viral delivery to the target region. This model replicates dopaminergic neuron loss and other hallmark phenotypes of Parkinson's disease, measurable via translational biomarkers, making it among the best PD animal models for drug evaluation.

AAV-A53T α-Syn Model Induction:

  • Unilateral stereotaxic injection of AAV expressing human A53T α-synuclein into the SNc

Validated Injection Site:

  • Substantia Nigra pars compacta (SNc) – to induce nigrostriatal degeneration

Disease Features Modeled (AAV):

In addition to standard endpoints, Biospective can perform non-invasive imaging studies on AAV α-synuclein mice – such as MRI volumetric analysis and PET imaging (e.g. [18F]FDG PET for glucose metabolism, [18F]DOPA PET for dopaminergic function) – to generate clinically translational imaging biomarkers (e.g. regional brain atrophy, cerebral hypometabolism, striatal dopamine terminal loss). This AAV-A53T model is also excellent for high-throughput studies (including screening studies) of disease-modifying therapeutics, thanks to its rapid start-up (no breeding needed), use of wild-type animals, cost-effective induction, and suitability for large cohort studies.

Image showing dopaminergic neuron loss and denervation in caudate-putamen

Severe dopaminergic neuron loss and dopaminergic denervation in the ipsilateral (left hemisphere) caudate-putamen following unilateral AAV-hA53Tα-Syn injection into the SNc of a C57BL/6 mouse.

Graphs showing effects of loss of dopaminergic innervation on behavioral tests

Loss of dopaminergic innervation corresponds with unilateral motor deficits, including increased ipsilateral paw use (Cylinder Test), reduced latency to fall (Rotarod), and increased contralateral swings (Tail Suspension Swing Test).

Translational Pathology and Biomarkers in Parkinson’s Disease Models

As a Preclinical Neuroscience CRO, we design our PD models with translational relevance to mirror key aspects of the human disease. A major differentiator of Biospective is our focus on translational biomarkers that align preclinical findings with clinical outcomes – including advanced neuroimaging and fluid biomarkers. We incorporate:

  • Alpha-synuclein–related biomarkers (pathology and spread)

  • Neuroinflammation markers (microglial/astrocyte activation)

  • Neurodegeneration endpoints (neuron loss, atrophy)

  • Mechanism-of-action confirmation (target/pathway engagement)

Our modeling and biomarker strategies ensure that preclinical successes meaningfully predict clinical potential, de-risking the transition from animal studies to human trials.

IF image of synuclein in low magnitude

Immunofluorescence of phosphorylated synuclein (pSyn129) in Parkinson's disease animal models reveals pronounced accumulation in neuronal soma and processes. 

Alpha-Synuclein Aggregates

Misfolded α-synuclein aggregates are a pathological hallmark of Parkinson’s disease. In patients, Lewy bodies and Lewy neurites (α-syn–rich inclusion bodies) are observed in the dopaminergic neurons of the SNc and in other brain regions, following a characteristic spatiotemporal progression (Braak, 2003). In our PFF- and AAV-induced PD models, we likewise observe robust α-synuclein pathology – including high levels of phosphorylated α-syn (pSyn129) accumulation in neuronal cell bodies and processes, and, in the PFF models, extensive seeded spread of α-syn aggregates throughout connected brain regions.

Images showing activated Microglia & Reactive Astrocytes

Activated microglia (red boxes) in the hippocampus of mice injected into the AON with PBS (top) or α-synuclein PFFs (bottom).

Activated Microglia & Reactive Astrocytes

Neuroinflammation is a key pathologic feature of Parkinson’s disease, with chronic activation of microglia and astrocytes contributing to neurodegeneration (Kam, 2020Chen, 2023). In our AAV and PFF PD models, we see strong neuroinflammatory responses with distinct spatiotemporal patterns. We apply cutting-edge image analysis – including proprietary computer vision and deep learning algorithms – to detect and quantify changes in microglial and astrocytic morphology in brain tissue. (See our Initiative: Microglia, Astrocytes, and Neurodegenerative Diseases and our Innovation: Microglial Activation in an α-Synuclein Mouse Model of Parkinson's Disease for more details.) These approaches allow sensitive tracking of neuroinflammation in relation to α-syn pathology.

Graph showing AAV - EBST Test Results (Box and Whiskers)

Tail Suspension Swing Test showing that AAV-A53T α-syn mice exhibit increased contralateral swings due to a unilateral dopaminergic deficit, compared to AAV-null control mice.
**** p<0.0001.

Dopaminergic Neuron Loss & Motor Deficits

Extrapyramidal motor symptoms in Parkinson’s disease are primarily driven by the degeneration of dopaminergic neurons in the substantia nigra pars compacta and the resultant loss of dopaminergic projections to the striatum. In our PD models, we induce α-synuclein pathology targeting the SNc via either AAV-mediated gene delivery or PFF injection . Both interventions produce significant dopaminergic neuron loss in the SNc and striatal dopamine terminal loss, leading to motor impairments that we quantify through behavioral assays (e.g. rotarod, tail suspension swing, cylinder, and hindlimb clasping tests). (See our Resource: Preformed Fibrils – A Guide to Cell and Animal Models for additional background on these approaches.) These motor phenotypes in the models parallel the movement disorders seen in human PD, enabling evaluation of therapeutic effects on motor function.

Graph showing M83 Sleep Bout Length % per bin

Sleep analysis showing that mice injected with α-syn PFFs into the AON exhibit disrupted sleep architecture, including shorter sleep bout lengths and reduced overall sleep duration.

Sleep Alterations

Sleep disturbances are among the most prevalent non-motor symptoms of Parkinson’s disease, affecting up to ~85% of patients (Stefani, 2020; Asadpoordezaki, 2025). Using a non-invasive sleep monitoring system, we have shown that PFF-seeded α-syn pathology can disrupt sleep-wake architecture in transgenic mice. For example, injecting α-syn PFFs into the AON of A53T (M83) mice leads to altered sleep architecture, including changes in total sleep time and significantly shorter sleep bout lengths (more fragmented sleep). Such findings mirror the insomnia and REM sleep behavior disorders observed in PD patients. (Sleep phenotypes have not yet been evaluated in our AAV model.)

MRI cortical thickness maps

MRI cortical thickness maps showing PBS-injected mice (top) and AON α-syn PFF-injected mice (bottom), with PFF-treated animals displaying cortical thinning.

Regional Brain Atrophy

Neuroimaging biomarkers are widely used in clinical trials of Parkinson’s disease to assess neurodegeneration. Magnetic resonance imaging (MRI) can detect regional brain atrophy and cortical thinning, which are sensitive indicators of neuron loss in PD (Tremblay, 2021Abdelgawad, 2023). We apply high-resolution MRI with fully automated volumetric analysis to our PD models. Using these techniques, we have demonstrated reproducible brain atrophy in both AAV and PFF mouse models of PD, including significant cortical thinning in PFF-injected mice relative to controls. (See our Innovation: Brain Atrophy Analysis in Mouse Models of Neurodegeneration to explore this capability.) These MRI endpoints provide quantitative, translational biomarkers that can be directly compared to human PD imaging data.

Graph showing M83 plasma CSF NF-L

α-Syn PFF-injected mice in the AON and MFB show elevated CSF NF-L levels compared to control mice.

Elevated Neurofilament Light in CSF & Plasma

Neurofilament light chain (NfL) is a well-established fluid biomarker of neurodegeneration. NfL levels are elevated in the CSF and plasma of Parkinson’s disease patients, and NfL is frequently measured in PD clinical trials (Bäckström, 2020Urso, 2023Pedersen, 2024). Elevated NfL has also been reported in several preclinical PD models. Consistent with clinical findings, our Parkinson’s mouse models exhibit significant increases in NfL in both plasma and CSF. In particular, PFF-injected transgenic mice show marked NfL elevation in biofluids (plasma, CSF) after α-syn fibril injection into regions like the AON or MFB. (See our Resource: Neurofilament Light Chain in Parkinson’s Disease Models for additional data.) This biomarker provides a quantifiable readout of neurodegeneration in our studies, strengthening the clinical relevance of the model outcomes.

Pathological and Phenotypical Profiles of PFF versus AAV α-Synuclein Parkinson's Disease Models?

The tables below summarize how each animal model of Parkinson's disease reproduces hallmark features of human Parkinson’s disease, enabling a quick comparison of their characteristics and use-cases.

Pathology – Key PD-associated pathological features (α-syn aggregates, neuroinflammation, dopaminergic neuron loss) in each mouse model:

Feature/DomainPFF ModelsAAV Models

α-Syn Aggregates

✔️
Present (robust seeding & widespread spreading)

✔️
Present (localized aggregates in SNc & striatum)

Neuroinflammation

✔️
Strong microglial & astroglial activation

✔️
Strong microglial & astroglial activation

Dopaminergic Neuron Loss

✔️
Moderate loss (widespread, include some non-dopaminergic regions)

✔️
Severe loss (selective SNc neurons, plus striatal denervation)

 

Functional Features – Behavioral and physiological impairments linked to the pathology:

Feature/Domain

PFF Models

AAV Models

Motor Deficits

✔️
Yes (moderate motor phenotype; onset over longer timeframe ~2-4 months)

✔️
Yes (severe unilateral motor deficits; earlier onset in weeks)

Sleep Disturbances

✔️
Yes (sleep/wake alterations observed)

N/A – Not yet evaluated in AAV model

 

Translational Biomarkers – Non-invasive imaging and fluid biomarkers reflecting neurodegeneration:

Feature/Domain

PFF Models

AAV Models

MRI Brain Atrophy

✔️
Observed

✔️
Observed

Neurofilament Light (NF-L)

✔️
Observed

✔️
Observed

 

Summary: Both the PFF and AAV α-synuclein mouse models exhibit robust α-synuclein pathology and significant neuroinflammation, closely mirroring the core features of human Parkinson’s disease. The PFF model uniquely recapitulates the prion-like spread of α-synuclein aggregates through the brain (including involvement of non-dopaminergic regions), whereas the AAV model produces more pronounced, regionally selective dopaminergic neuron loss in the SNc along with earlier-onset, severe motor deficits. Sleep disturbances have been demonstrated in the PFF model (due to its longer timeline and limbic involvement) but have not yet been assessed in the AAV model. Notably, both models support the use of MRI-based brain atrophy measures and elevated NfL in biofluids as translational biomarkers of neurodegeneration, facilitating direct comparison to clinical trial endpoints. Together, the PFF and AAV models provide complementary platforms that allow a comprehensive evaluation of Parkinson’s disease mechanisms and therapeutic interventions across diverse experimental needs.

Illustrative Example of Our Alpha-Synuclein Mouse Models

(Interactive Data Presentation – AAV A53T α-Synuclein Mouse Model)

The interactive viewer below highlights some of the key motor, imaging, and pathological findings from Biospective’s AAV-A53T α-synuclein mouse model of Parkinson’s disease. In this example study, 12-week-old C57BL/6 mice were injected unilaterally in the left SNc with either AAV-hA53T-α-synuclein or a control AAV-null vector (2 µL, infused at 0.4 µL/min). A digital stereotaxic system with automated microinjection was used to ensure precise targeting of the SNc (see the coronal atlas view of the injection site).

Multiplex immunofluorescence (mIF) imaging was performed on brain sections, staining for phospho-α-syn (pSyn129), tyrosine hydroxylase (TH), GFAP (astrocytes), Iba1 (microglia), and nuclei (DAPI). Whole-slide high-resolution scans were analyzed using Biospective’s quantitative image analysis software. The results show a substantial loss of TH-positive dopaminergic neurons in the injected (ipsilateral) SNc compared to the contralateral hemisphere, accompanied by dense pSyn129-positive α-syn aggregates and robust glial activation in the affected regions. Quantitative analysis confirmed a highly significant reduction of TH immunoreactivity in the ipsilateral SNc, reflecting the targeted neurodegeneration.

Using the Interactive Viewer: Navigate through the “Image Story” via the left-hand panel or the on-screen arrows. You can pan around high-resolution microscopy images with your mouse, and zoom in/out using the scroll wheel or the +/- controls. The Control Panel (top-right) allows toggling of image channels and segmentation overlays. For the best experience, we recommend switching to full-screen mode. This Interactive Presentation enables you to explore the model’s neuropathology and associated functional deficits in detail, as if looking directly down the microscope.

Neurodegeneration & Neuroinflammation in the AAV-Synuclein Mouse Model

1/12

Biospective Preclinical Logo

This Interactive Presentation illustrates some of the interesting motor function, brain imaging, and pathologic features of Biospective's AAV A53T α-synuclein mouse model of Parkinson’s disease (PD).

This model was generated by injecting 12 week-old C57BL/6 mice with AAV-human-A53T-synuclein or AAV-null (control) vectors unilaterally into the left substantia nigra pars compacta (SNc). 2 µL of vector was infused at a rate of 0.4 µL/min using a digital stereotaxic device with an automated microinjector.

Coronal Image of Mouse Brain with AAV Injection Site in the SNc

Coronal Atlas View of SNc Injection Site

Multiplex immunofluorescence (mIF) images were generated by immunostaining for phospho-Syn129, GFAP, Iba-1, Tyrosine Hydroxylase, Dopaminergic Nuclei, and counterstained with the DAPI nuclear stain. Tissue sections were digitized using a high-throughput slide scanner and were processed using Biospective's PERMITSTM software platform.

To navigate though this Image Story, you can use the arrows and/or the Table of Contents icon in the upper right corner of this panel.

Navigation Panel with Tooltips

You can also interact with the microscopy image in the viewer on the right at any time to further explore this high-resolution data.

Neurodegeneration in the Substantia Nigra

As can be seen in this microscopy image, there is substantial loss of TH-positive dopaminergic neurons in the ipsilateral SNc compared to the contralateral hemisphere. For reference, an illustration with atlas labels for this brain level is provided below.

Coronal Mouse Brain Section (Bregma -3.2) with Neuroanatomy Labels

Using our PERMITSTM quantitative analysis software, we have quantified the TH staining in the SNc. The plots below show a highly significant reduction in the ipsilateral hemisphere.

Tyrosine Hydroxylase and Cell Density in the Substantia Nigra

TH stain density and cell density for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

We have found significant brain atrophy in the SNc by generating regional volume data from in vivo anatomical MRI scans, which corresponds well with the loss of TH-positive neurons. MR images were acquired from mice injected with different doses of AAV-Synuclein at 4 weeks post-inoculation using a 7T animal MRI scanner.

Anatomical MRI with segmented SNc, as well as a plot of relative difference between ipsilateral and contralateral SNc. Injected AAV-Syn doses (GC) were 1×109 (yellow), 5×109 (blue), and 1×1010 (aqua). *p<0.05, **p<0.01.

Dopaminergic Neurons in the Contralateral SNc

This microscopy image shows the contralateral (right hemisphere) SNc which shows unaffected TH-positive cell bodies and processes in red. The DAPI-counterstained nuclei are shown in blue.

Loss of Dopaminergic Neurons in the Ipsilateral SNc

This microscopy image shows the ipsilateral (left hemisphere) SNc, which demonstrates a substantial reduction of TH-positive cell bodies and processes (in red) compared to the contralateral hemisphere. The DAPI-counterstained nuclei are shown in blue.

Neurodegeneration in the Caudate-Putamen & Dopaminergic Motor Deficits

This microscopy image show severe dopaminergic denervation of the ipsilateral (left hemisphere) caudate-putamen (loss of TH-positive terminals). For reference, an illustration with atlas labels for this approximate brain level is provided below.

Coronal Image of Mouse Brain at the Level of the Striatum

Coronal Mouse Brain Section (Bregma +0.86) with Neuroanatomy Labels

Using our PERMITSTM quantitative analysis software, we have quantified the TH staining in the Caudate-Putamen. The plot below shows a highly significant reduction in the ipsilateral hemisphere.

Tyrosine Hydroxylase Staining in the Caudate-Putamen

TH stain density for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

This loss of dopaminergic innervation corresponds well with unilateral motor deficits in these mice, including a highly significant increase in use of the ipsilateral paw during the Cylinder Test, decreased latency to fall in the Rotarod Test, increased swings to the contralateral side in the Tail Suspension Swing Test (TSST), and progressively increased hindlimb clasping.

https://opt003stagmediafiles.blob.core.windows.net/image/22081b3660b5456da7546684dbe757ef

Cylinder Test data for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

https://opt003stagmediafiles.blob.core.windows.net/image/aec8fe14b2444c5ab9874463a60d2227

Rotarod Test data for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

https://opt003stagmediafiles.blob.core.windows.net/image/e7d5e05ba3324a08b78eecfc643cb480

Tail Swing Suspension Test (TSST) data for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

Weekly Hindlimb Clasping Test for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, **p<0.01, **** p<0.0001.

Loss of Dopaminergic Terminals in the Ipsilateral Caudate-Putamen

This high magnification view shows the severe extent of loss of dopaminergic (TH-positive) terminals in the ipsilateral striatum. There are some remaining (albeit dystrophic) axons present.

Similar to our findings in the SNc, we have observed brain atrophy in the caudate-putamen by quantitative analysis of high-resolution anatomical MRI scans, which establishes an in vivo-ex vivo relationship between neuroimaging and IF measures.

MRI Atlas and Volume Data at the Level of the Striatum

Anatomical MRI with segmented caudate-putamen, as well as a plot of the relative difference between ipsilateral and contralateral caudate-putamen. Injected AAV-Syn doses (GC) were 1×109 (yellow), 5×109 (blue), and 1×1010 (aqua). *p<0.05, **p<0.01.

Cerebrospinal Fluid (CSF) Neurofilament Light Chain (NF-L) in the AAV-Synuclein Mouse Model

NF-L is a neuron-specific cytoskeletal protein released into the extracellular fluid following axonal damage and/or neurodegeneration. Elevated NF-L levels serve as a highly sensitive biomarker for neuronal injury and damage.

In PD patients, NF-L concentrations are increased relative to healthy controls, and have been reported to correlate with clinical measures of disease severity and progression (Pilotto, 2021; Ou, 2024).

https://opt003stagmediafiles.blob.core.windows.net/image/1075128c3f6045a890167f0d44347615

CSF NF-L concentrations for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

CSF NF-L quantification in the AAV-synuclein mouse model provides a sensitive and quantifiable readout of neurodegeneration.

Microgliosis in Response to Human A53T a-Synuclein Expression

In this low magnification image, one can readily appreciate the higher density of Iba-1 staining microglia in the ipsilateral hemisphere (indicated by the box) relative to the contralateral hemisphere in an AAV-Syn injected mouse brain.

The plots below show the Iba-1 stain density in various brain regions, with highly significant increased staining in the AAV-Syn mice.

PERMITS Data on Iba-1 Stain Density

Iba-1 stain density for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

We have performed a morphological analysis of microglia using a novel computer vision & machine learning approach developed by our team. This fully-automated algorithm classifies non-activated (ramified) and activated (non-ramified) microglia.

Examples of Non-activated and Activated Microglial Morphology

The plots below show the microglial activation in various brain regions, with highly significant increased microglial activation in the AAV-Syn mice.

Plots of PERMITS Data Showing Activated Microglia

Microglial activation for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

Iba-1 Staining in Proximity to Phosphorylated α-Synuclein

This high magnification view shows the increased density of Iba-1-stained microglia in areas with phosphorylated α-synuclein aggregates.

Astrogliosis in Response to Human A53T α-Synuclein Expression

This low magnification microscopy image show a higher density of GFAP-positive astrocytes in the ipsilateral hemisphere (indicated by the box) of an AAV-Syn injected mouse brain. The plots below show the GFAP stain density in various brain regions.

Plots of PERMITS Data Showing GFAP Stain Density

GFAP stain density for AAV-Syn compared to AAV-null (control) injections; mean ± SEM, t-test, **** p<0.0001.

Astrogliosis and Microgliosis

This high magnification microscopy image shows a high level of Iba-1-positive microglia and GFAP-positive astrocytes in the ipsilateral hemisphere. Note the “activated” morphology of these neuroinflammatory cells.

Summary

The AAV A53T α-syn mouse model recapitulates many of the hallmark features of Parkinson’s disease. This model demonstrates progressive development of asymmetric motor dysfunction (due to unilateral injection), and associated loss of TH-positive SNc neurons and striatal TH expression.

AAV A53T α-synuclein locally increases brain atrophy, microglial density and activation levels, and astrocyte density and hypertrophy. Studies are planned to further interrogate the spatial relationships between microglial activation, astrocyte hypertrophy, and α-synuclein aggregation.

The AAV A53T α-synuclein mouse model is well-suited for drug development given the quantitative in-life and ex vivo readouts. It also has advantages over other models as a screening tool for novel disease-modifying therapeutics targeting α-synuclein related pathology, including the relatively short timeframe required to perform preclinical studies in this model.

Please feel free to further explore the microscopy image in the viewer.

We would be happy to discuss this model and our characterization if you would like to Contact Us.

Table of Contents
Control Panel
Section: SNc Section 1
Channels

Characterization of our AAV-A53T-Synuclein mouse model, including in vivo data and high-resolution images of entire Multiplex Immunofluorescence tissue sections.

How are Parkinson's Disease Mouse Models Used in Drug Development?

We work closely with our biotech and pharmaceutical sponsors to:

  • Evaluate therapeutic efficacy and dose-response in Parkinson’s models

  • Assess target engagement and disease-modifying effects

  • Support translational biomarker strategies, including imaging and fluid biomarkers for clinical readiness

Our Parkinson’s disease mouse models are optimized for in vivo testing of multiple therapeutic modalities, including both traditional and advanced approaches:

Small Molecules

  • Brain penetration and PK/PD profile

  • Behavioral efficacy on motor and non-motor symptoms

  • Reduction of pathological hallmarks (α-syn aggregates, neuron loss)

RNA-Targeted Therapies

  • Target knockdown verification (e.g. mRNA or protein level reduction)
  • CNS biodistribution of ASOs/siRNA

  • Translational biomarker readouts to confirm pathway engagement

Gene Therapy & Viral Vectors

  • Transgene expression levels in target regions

  • Regional biodistribution of viral vectors (e.g. AAV spread)

  • Functional rescue or disease modification outcomes (behavioral and pathological improvements)

Antibodies & Biologics

  • CNS exposure and penetration of biologics (e.g. BBB engagement)

  • α-Synuclein aggregation clearance or reduction

  • Mechanism-of-action validation (target binding, downstream signaling changes)

End-to-End Parkinson’s Disease Preclinical CRO Services

Biospective offers fully integrated preclinical Parkinson’s disease contract research services, including:

  • Study design & model selection – expert guidance on choosing the right PD model and designing robust studies

  • In vivo efficacy studies – execution of treatment studies with comprehensive monitoring of outcomes

  • Biodistribution & PK/PD – analysis of drug distribution and pharmacokinetics/pharmacodynamics in CNS and periphery

  • Target engagement assays – confirmation that the therapeutic hits its molecular target (e.g. α-syn reduction, pathway modulation)

  • Behavioral analysis – motor and non-motor behavioral testing (rotarod, gait, grip strength, sleep monitoring, etc.)

  • In vivo multi-modality imaging – MRI, PET, SPECT, fluorescence, and bioluminescence imaging to track disease and treatment effects

  • Immunoassays – biomarker quantification in CSF, blood, and tissue (e.g. NfL, cytokines, chemokines)

  • Immunohistochemistry (IHC) & multiplex immunofluorescence (mIF) – post-mortem tissue staining & quantitative image analysis to assess pathology and therapeutic impact

  • Data analysis & reporting – rigorous quantitative analysis, statistics, and comprehensive reporting by our scientists

This end-to-end approach minimizes handoffs, accelerates timelines, and reduces risk for our sponsors by keeping all aspects of the study under one expert team.

Parkinson's Disease Animal Models Summary: How do PFF and AAV α-Synuclein Models Compare?

Together, the PFF and AAV α-synuclein rodent models provide complementary platforms for investigating Parkinson’s disease mechanisms and evaluating novel therapeutics. Both models faithfully reproduce core features of PD – including robust α-synuclein aggregation and measurable neuroinflammation – while each has unique strengths in terms of phenotype expression and timing. The PFF model offers a powerful system for studying the prion-like seeding and spreading of α-synuclein pathology across the brain over time, including limbic and cortical regions. In contrast, the AAV model produces pronounced, selective dopaminergic neuron loss in the SNc and rapid-onset motor deficits, more directly modeling the nigrostriatal degeneration responsible for classic PD motor symptoms. Sleep and other non-motor phenotypes have been demonstrated in the PFF model (over longer durations), whereas these endpoints remain to be characterized in the faster AAV model. Importantly, both models support translational endpoints – such as MRI-detected brain atrophy and elevated NfL in biofluids – that bridge preclinical findings to clinical measures of neurodegeneration. By leveraging both models, researchers can comprehensively evaluate Parkinson’s disease pathways and therapeutic effects, from molecular mechanisms to functional outcomes.

Learn more about our in-depth characterization of these Parkinson’s disease mouse models, our validated outcome measures, and the full scope of our Parkinson's disease CRO services.

FAQs

What Parkinson's disease mouse models do you offer?


Can these models be used to test potential Parkinson's disease therapies?


Do you work with non-small-molecule therapies?


Do you work with international clients?


What is α-synuclein?


What are the key differences between the PFF and AAV models?


How well do these mice models translate to human Parkinson's disease?


What are the advantages of injecting PFFs into transgenic mice?


How can neurodegeneration be measured in α-synuclein animal models?


Can non-motor symptoms be observed in α-synuclein mouse models?


References


Keywords


Related Content

Explore up-to-date insights on Parkinson’s disease and best practices for evaluating therapeutics in PD animal models.

More Information

Let us know what you’re interested in – our team of experts will be happy to discuss your project needs!

Email us at i[email protected] or simply complete & submit the form below. 

Name*
Email*
Purpose of Inquiry*
Affiliation (Company/Institution)*
Message*

Your privacy is important to us. We will protect your data as outlined in our Privacy Notice.

I agree to the terms in the Privacy Notice*

We use necessary cookies to make our site work. We also use other cookies to help us make improvements by measuring how you use the site or for marketing purposes. You have the choice to accept or reject them all. For more detailed information about the cookies we use, see our Privacy Notice.