Preclinical WebsiteClinical Website
Last Updated Date: August 06, 2024
Authors: Laurent Potvin-Trottier, Ph.D. and Barry J. Bedell, M.D., Ph.D.

Why quantify microglia morphology?

Microglia play a crucial role in many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson’s disease (PD) (Salter, 2017; Hickman, 2018). Many potential therapeutics for neurodegenerative diseases target microglia, for instance by modulating their phenotype or by promoting the clearance of Aβ plaques (Gao, 2023). Microglia are highly dynamic cells, capable of assuming various phenotypes, as demonstrated by single cell ‘omics technologies. Recently, about one hundred scientists co-authored a publication (Paolicelli, 2022) aiming to clarify and build a consensus around the terminology of these phenotypes, using terms such as homeostatic signature, white matter-associated microglia, and disease-associated microglia (DAM).

Microglia also show drastic morphological changes in reactive conditions. In this resource, we highlight commonly quantified morphological phenotypes (Savage, 2020). In homeostatic conditions, microglia typically exhibit the ramified morphology characterized by long, thin, extended processes. Hypertrophic microglia, typically associated with reactive conditions, have enlarged soma with shorter, thicker, more ramified processes. Amoeboid microglia have enlarged soma and are almost devoid of processes, making them morphologically similar to macrophages. Dystrophic microglia feature beaded, spherical swellings of their processes, which can appear fragmented. Finally, rod cells are highly elongated with a single orientation and minimal radial processes. While functional phenotypes and distinct morphologies are two complementary characterizations of microglia, a mechanistic link between morphological changes (ramification) and function (cytokine release) has been identified (Madry, 2018). In addition, morphological metrics have been found to be correlated to functional markers in individual microglia (Kozlowski, 2012; Fernández-Arjona, 2019).

Microscopy images and schematic representations of four selected morphologies of microglia.

Example microscopy images and schematic representations of four selected morphologies of microglia. Figure reproduced and adapted from (Reddaway, 2023), based on morphology definitions from (Savage, 2019), under the Creative Commons Attribution License.

At Biospective, we hypothesized that high-throughput quantification of microglial morphology could enhance the understanding of their state in preclinical studies. In mouse models of Alzheimer’s disease, Parkinson’s disease, and ALS, we found that analyzing microglial morphology provided a more sensitive metric of disease state than simply measuring the microgliosis by Iba-1 stain density.

How can we quantify microglia morphology in tissue sections?

Experimental Conditions: Staining, Section Thickness, and Microscopy

To quantify microglial morphology, microglia need to be labeled with a stain that is specific and sensitive to both their processes and soma, and is expressed in all phenotypes (typically Iba-1). Thick tissue sections up to 150 µm can be imaged as a z-stack using confocal microscopy for a full 3D characterization of microglia morphology. For high-throughput quantification in 2D, thinner sections (5-20 µm) can be imaged with a digital slice scanner (Franco-Bocanegra, 2021; Leyh, 2021). The trade-off between throughput and 3D imaging depends on the study’s objective. For example, high throughput 2D imaging is suitable for preclinical therapeutic efficacy studies and can scale to millions of cells. A simple metric, such as soma size, which consistently changes in reactive conditions (Kozlowski, 2012; Verdonk, 2016; Davis, 2017; Fletcher, 2020; Silburt, 2022), can be accurately quantified in 2D. For instance, one study focused solely on the soma morphology and found a shift from ramified to hypertrophic microglia in the retina of aged mice (Choi, 2022).

Steps for Morphological Quantification

The necessary steps for automated quantification common to most approaches are depicted in the figure below. Various approaches have been reviewed by Reddaway et al. (Reddaway, 2023). First, microglia need to be individually detected and separated - the object detection task in computer vision. The cell and soma of each cell are segmented from the background. The skeleton of the processes is algorithmically obtained. Finally, different morphological metrics (i.e. morphometrics), such as the cell area, perimeter, soma area, number of branches in the skeleton, etc. are measured in each cell.

Schematic representation of the process for the analysis of microglia morphology. Microglia are identified on an immunohistochemistry (shown here stained with Iba-1) or immunofluorescence tissue section. For each identified cell, the soma and processes are segmented, and the skeleton of the processes is obtained. Many morphological metrics, such as the cell and soma size, are then obtained. The cell can then optionally be classified as a specific morphology using a machine learning model. In the example shown, the blue boxes represent ramified morphology, and the red boxes represent non-ramified morphology. Different statistics about the cell can then be aggregated by region-of-interest (ROI), subject (e.g. animal), and group. Morphology metric schematic reproduced and adapted from (Leyh, 2021) under the Creative Commons Attribution License.

Quantitative Analysis Between Groups

The cell morphometrics are aggregated by region-of-interest (ROI), by animal, and by group. To compare the groups, there are three main approaches:

1. Compare group morphometric distribution. The simplest approach is simply to compare the distribution of a particular metric, such as the soma size, across groups. Examples: (Kozlowski, 2012; Heindl, 2018).

 

2. Compare the distribution of cells across distinct clusters. Machine learning methods can identify clusters of cells with similar morphologies. The distribution of cell across these clusters can then be compared across groups. Examples: (Fernández-Arjona, 2017; Heindl, 2018; Salamanca, 2019).

 

3. Compare the distribution of cells across classified morphologies. Machine learning models can be trained to recognize user-specified morphologies (e.g. ramified, hypertrophic, amoeboid, etc.). The distribution and the number of cells with these morphologies can then be compared across groups. Examples: (Leyh, 2021; Choi, 2022).

What changes in microglia morphology have been observed in neurodegenerative diseases?

In this section, we highlight common findings in three neurodegenerative diseases, namely ALS, AD, and PD.

Amyotrophic Lateral Sclerosis (ALS)

Morphological changes in microglia have been observed in three different mouse model of ALS. In a SOD1-G93A model, a progressive shift to a morphology akin to hypertrophy (larger cell body, shorter processes) was observed during the disease progression (Ohgomori, 2016). In the inducible hTDP43ΔNLS model, a similar shift was observed during the disease “recovery” after expression of pathological TDP-43 was stopped (Spiller, 2018). An increase in the number of microglia and a shift toward hypertrophy was observed in a C9orf72 model (GA-repeats). This effect was largely eliminated when the mice were immunized to the GA-repeats (Zhou, 2020). At Biospective, using the TDP-43 ΔNLS mouse model, we have found morphological changes in both “Off Dox” and “Low Dox” models. The density of non-ramified microglia was highly correlated with the clinical composite motor scores.

Microglia morphological changes are highly correlated with clinical motor score in a mouse TDP-43 ΔNLS model.

Microglia morphological changes are highly correlated with clinical motor score in a mouse TDP-43 ΔNLS model. The composite motor score is shown as a function of the Iba-1 stain density (fraction of segmented pixels, left) and the density of microglia with non-ramified (“reactive”) morphology (right) in the caudate putamen (CP) across groups with different levels of disease severity. A linear fit to the log of the x-axis is shown, and the Pearson correlation coefficient increases from 0.38 with the Iba-1 stain density to 0.83 for the density of non-ramified microglia. Different disease severity groups are shown in the linear fit, such as “On Dox (control)", “4 weeks Low Dox”, “6 weeks Low Dox”, and “3 weeks Off Dox”. A linear fit combining the log-density of non-ramified microglia in four ROIs (caudate putamen, motor cortex, non-motor cortex, and corticospinal tract) results in a correlation coefficient of 0.93 +/- 0.06 (range obtained from standard deviation of a 5-fold cross-validation).

Alzheimer’s Disease

Studies in human tissue have generally shown no significant changes in the total number of microglia in AD versus aged-matched control (Davies, 2017; Heindl, 2018; Paasila, 2019; Martini, 2020; Franco-Bocanegra, 2021). However, a shift away from the homeostatic ramified morphology has been observed. An increase in the number of microglia with dystrophic morphology has been reported (Bachstetter, 2015; Davies, 2017; Martini, 2020). Multiple morphological metrics, such as the total process length, have been found to be correlated to the Aβ and/or tau pathological load (Heindl, 2018; Paasila, 2019; Franco-Bocanegra, 2021). Interestingly, Franco-Bocanegra et al. (Franco-Bocanegra, 2021) observed a treatment effect - patients immunized against Aβ42 had increased fraction of microglia with ramified morphology as compared to non-treated AD patients and healthy controls. Our studies in an APP/PS1 mouse model of AD have found morphological changes in microglia that were highly localized in proximity to the Aβ plaques, and were more sensitive than the Iba-1 stain density at assessing disease progression.

Parkinson’s Disease & Synucleinopathies

In rat and marmoset models of PD, a shift from the ramified morphology to hypertrophic and amoeboid form has been observed (Sanchez-Guajardo, 2010; Barkholt, 2012). Similarly, in a mouse model of multiple system atrophy (MSA; PLP-alpha-synuclein), a shift from the ramified morphology to the hypertrophic morphology was observed without an increase in overall microglia number (Refolo, 2018). A treatment effect was observed in a rat model of PD - changes typical of a transition to the hypertrophic morphology (increase in soma size, density, etc.) were observed in the striatum of levodopa-treated rats, but not in the saline-treated rats (Fletcher, 2020). Using Biospective’s mouse model of PD (injection of human preformed fibrils into the anterior olfactory nucleus of hemizygous M83 mice), we found an increase in total microglia and a shift towards hypertrophic morphology, correlating with disease progression. The density of non-ramified microglia was a more sensitive metric of the disease state than the Iba-1 stain density.

Our team would be happy to answer any questions about Microglia Morphology in ALS, Alzheimer's Disease, and Parkinson's Disease or provide specific information about the models that we use for therapeutic efficacy studies.

Discover more about our Neurodegenerative Diseases Models

FAQs

What are the advantages and disadvantages of the different quantitative approaches for comparing groups?

Using the morphometric distribution can be simple and efficient, but the metrics need to be chosen in advance to avoid losing statistical power due to multiple hypothesis testing. In addition, care must be taken to represent the intra-group biological variability, for example by bootstrapping on the animals (to avoid overrepresenting differences due to the high number of cells per animal). Using morphological classification (e.g. ramified, hypertrophic) can be powerful and precise, but requires defining these morphologies, and thus can be less suited to discovering new morphologies. While clustering approaches do not require defining morphologies a priori, the number of clusters, the model, and the morphometrics used to define the clusters require user input and can change the clusters obtained.


What are the different methods to classify the morphologies using machine learning?

There are two main approaches to classify the morphology of microglia. One approach is based on using the morphometrics (e.g. soma area, number of branches in skeleton, etc.) – in other words, a set of numbers for each cell – to classify cells in each morphology (sometimes called a classical machine learning approach). The other method is to directly use the image of the cell as the input to the model and let the model learn the necessary features to classify the morphologies (i.e. deep learning approach). Deep-learning approaches can be precise and do not require a choice of features, but are less directly explainable and require more data to train. Machine learning models based on morphometrics can be more directly explainable; for example, a cell has been classified in this particular morphology because its soma size was larger. In addition, many morphometrics were found to be linearly correlated with expression of functional markers (e.g. IL-1β; Fernández-Arjona, 2019), which motivates the use of models based on linear combination of these metrics (e.g. logistic regression) for characterizing microglia.


Have treatment effects been observed using microglia morphology analysis?

Yes. Microglia morphology analysis revealed changes in the microglia morphology in putative therapeutic treatments for ALS (Zhou, 2020), AD (Franco-Bocanegra, 2021), and PD (Fletcher, 2020).


In what other conditions & diseases has microglial morphology been observed to change?

Microglial morphology has been observed to change in many contexts, such as in aging (Castro, 2024; Shaerzadeh, 2020; Shahidehpour, 2021), dementia with Lewy bodies (Bachstetter, 2015), demyelination (cuprizone model) (Olah, 2012), hippocampal sclerosis of aging (Bachstetter, 2015), Huntington’s disease (Sapp, 2001; Franciosi, 2012; Savage, 2020), inflammation through lipopolysaccharide (LPS) treatment (Kozlowski, 2012; Verdonk, 2016), and many others (reviewed in Reddaway, 2023).


References

Bachstetter, A. D., Van Eldik, L. J., Schmitt, F. A., Neltner, J. H., Ighodaro, E. T., Webster, S. J., Patel, E., Abner, E. L., Kryscio, R. J., Nelson, P. T. Disease-related microglia heterogeneity in the hippocampus of Alzheimer’s disease, dementia with Lewy bodies, and hippocampal sclerosis of aging. Acta Neuropathol. Commun., 3: 32, 2015; doi: 10.1186/s40478-015-0209-z

Barkholt, P., Sanchez-Guajardo, V., Kirik, D., Romero-Ramos, M. Long-term polarization of microglia upon α-synuclein overexpression in nonhuman primates. Neuroscience, 208: 85–96, 2012; doi: 10.1016/j.neuroscience.2012.02.004

Castro, R. W., Lopes, M. C., De Biase, L. M., Valdez, G. Aging spinal cord microglia become phenotypically heterogeneous and preferentially target motor neurons and their synapses. Glia, 72: 206–221, 2024; doi: 10.1002/glia.24470

Choi, S., Hill, D., Guo, L., Nicholas, R., Papadopoulos, D., Cordeiro, M. F. Automated characterisation of microglia in ageing mice using image processing and supervised machine learning algorithms. Sci. Rep., 12: 2022; doi: 10.1038/s41598-022-05815-6 

Davies, D. S., Ma, J., Jegathees, T., Goldsbury, C. Microglia show altered morphology and reduced arborization in human brain during aging and Alzheimer’s disease. Brain Pathology, 27: 795–808, 2017; doi: 10.1111/bpa.12456

Davis, B. M., Salinas-Navarro, M., Cordeiro, M. F., Moons, L., Groef, L. De. Characterizing microglia activation: A spatial statistics approach to maximize information extraction. Sci. Rep., 7: 2017; doi: 10.1038/s41598-017-01747-8

Deczkowska, A., Keren-Shaul, H., Weiner, A., Colonna, M., Schwartz, M., Amit, I. Disease-Associated Microglia: a universal immune sensor of neurodegeneration. Cell, 173: 1073–1081, 2018; doi: 10.1016/j.cell.2018.05.003

Fernández-Arjona, M. del M., Grondona, J. M., Fernández-Llebrez, P., López-Ávalos, M. D. Microglial morphometric parameters correlate with the expression level of IL-1β, and allow identifying different activated morphotypes. Front. Cell. Neurosci., 13: 2019; doi: 10.3389/fncel.2019.00472

Fernández-Arjona, M. del M., Grondona, J. M., Granados-Durán, P., Fernández-Llebrez, P., López-Ávalos, M. D. Microglia morphological categorization in a rat model of neuroinflammation by hierarchical cluster and principal components analysis. Front. Cell. Neurosci., 11: 2017; doi: 10.3389/fncel.2017.00235

Fletcher, E. J. R., Finlay, C. J., Amor Lopez, A., Crum, W. R., Vernon, A. C., Duty, S. Neuroanatomical and microglial alterations in the striatum of levodopa-treated, dyskinetic hemi-Parkinsonian rats. Front. Neurosci., 14: 2020; doi: 10.3389/fnins.2020.567222

Franciosi, S., Ryu, J. K., Shim, Y., Hill, A., Connolly, C., Hayden, M. R., McLarnon, J. G., Leavitt, B. R. Age-dependent neurovascular abnormalities and altered microglial morphology in the YAC128 mouse model of Huntington disease. Neurobiol. Dis., 45: 438–449, 2012; doi: 10.1016/j.nbd.2011.09.003

Franco-Bocanegra, D. K., Gourari, Y., McAuley, C., Chatelet, D. S., Johnston, D. A., Nicoll, J. A. R., Boche, D. Microglial morphology in Alzheimer’s disease and after Aβ immunotherapy. Sci. Rep., 11: 2021; doi: 10.1038/s41598-021-95535-0

Gao, C., Jiang, J., Tan, Y., Chen, S. Microglia in neurodegenerative diseases: mechanism and potential therapeutic targets. Signal Transduct. Target Ther., 8: 2023; doi: 10.1038/s41392-023-01588-0

Heindl, S., Gesierich, B., Benakis, C., Llovera, G., Duering, M., Liesz, A. Automated morphological analysis of microglia after stroke. Front. Cell. Neurosci., 12: 2018; doi: 10.3389/fncel.2018.00106

Hickman, S., Izzy, S., Sen, P., Morsett, L., El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci., 21: 2018; doi: 10.1038/s41593-018-0242-x

Kozlowski, C., Weimer, R. M. An automated method to quantify microglia morphology and application to monitor activation state longitudinally in vivo. PLoS One 7: 2012; doi: 10.1371/journal.pone.0031814

Leyh, J., Paeschke, S., Mages, B., Michalski, D., Nowicki, M., Bechmann, I., Winter, K. Classification of microglial morphological phenotypes using machine learning. Front. Cell. Neurosci., 15: 2021; doi: 10.3389/fncel.2021.701673

Madry, C., Kyrargyri, V., Arancibia-Cárcamo, I. L., Jolivet, R., Kohsaka, S., Bryan, R. M., Attwell, D. Microglial tamification, surveillance, and Interleukin-1β release are regulated by the two-pore domain K+ channel THIK-1. Neuron, 97: 299-312.e6, 2018; doi: 10.1016/j.neuron.2017.12.002

Martini, A. C., Helman, A. M., McCarty, K. L., Lott, I. T., Doran, E., Schmitt, F. A., Head, E. Distribution of microglial phenotypes as a function of age and Alzheimer’s disease neuropathology in the brains of people with Down syndrome. Alzheimer’s and Dementia: Diagnosis, Assessment and Disease Monitoring, 12: 2020; doi: 10.1002/dad2.12113

Ohgomori, T., Yamada, J., Takeuchi, H., Kadomatsu, K., Jinno, S. Comparative morphometric analysis of microglia in the spinal cord of SOD1G93A transgenic mouse model of amyotrophic lateral sclerosis. European J. Neurosci., 43: 1340–1351, 2016; doi: 10.1111/ejn.13227

Olah, M., Amor, S., Brouwer, N., Vinet, J., Eggen, B., Biber, K., Boddeke, H. W. G. M. Identification of a microglia phenotype supportive of remyelination. Glia, 60: 306–321, 2012; doi: 10.1002/glia.21266

Paasila, P. J., Davies, D. S., Kril, J. J., Goldsbury, C., Sutherland, G. T. The relationship between the morphological subtypes of microglia and Alzheimer’s disease neuropathology. Brain Pathol., 29: 726–740, 2019; doi: 10.1111/bpa.12717

Paolicelli, R. C., Sierra, A., Stevens, B., Tremblay, M. E., Aguzzi, A., Ajami, B., Amit, I., Audinat, E., Bechmann, I., Bennett, M., Bennett, F., Bessis, A., Biber, K., Bilbo, S., Blurton-Jones, M., Boddeke, E., Brites, D., Brône, B., … Wyss-Coray, T. Microglia states and nomenclature: a field at its crossroads. Neuron, 110: 3458–3483, 2022; doi: 10.1016/j.neuron.2022.10.020

Reddaway, J., Richardson, P. E., Bevan, R. J., Stoneman, J., Palombo, M. Microglial morphometric analysis: so many options, so little consistency. Front. Neuroinform., 17: 2023; doi: 10.3389/fninf.2023.1211188

Refolo, V., Bez, F., Polissidis, A., Kuzdas-Wood, D., Sturm, E., Kamaratou, M., Poewe, W., Stefanis, L., Angela Cenci, M., Romero-Ramos, M., Wenning, G. K., Stefanova, N. Progressive striatonigral degeneration in a transgenic mouse model of multiple system atrophy: translational implications for interventional therapies. Acta Neuropathol. Commun., 6: 2, 2018; doi: 10.1186/s40478-017-0504-y

Salamanca, L., Mechawar, N., Murai, K. K., Balling, R., Bouvier, D. S., Skupin, A. MIC-MAC: An automated pipeline for high-throughput characterization and classification of three-dimensional microglia morphologies in mouse and human postmortem brain samples. Glia, 67: 1496–1509, 2019; doi: 10.1002/glia.23623

Salter, M. W., Stevens, B. Microglia emerge as central players in brain disease. Nat., Med., 23: 1018–1027, 2017; doi: 10.1038/nm.4397

Sanchez-Guajardo, V., Febbraro, F., Kirik, D., Romero-Ramos, M. Microglia acquire distinct activation profiles depending on the degree of α-synuclein neuropathology in a rAAV based model of Parkinson’s disease. PLoS One, 5: 2010; doi: 10.1371/journal.pone.0008784

Sapp, E., Kegel, K. B., Hashikawa, T., Uchiyama, Y., Tohyama, K., Bhide, P. G., Vonsattel, J. P., Difiglia, M. Early and progressive accumulation of reactive microglia in the Huntington disease brain. J. Neuropathol. Exp. Neurol., 60: 2001; doi: 10.1093/jnen/60.2.161

Savage, J. C., Carrier, M., Tremblay, M. È. Morphology of microglia across contexts of health and disease. in Methods in Molecular Biology vol. 2034 13–26, Humana Press Inc., 2019.; doi: 10.1007/978-1-4939-9658-2_2

Savage, J. C., St-Pierre, M. K., Carrier, M., El Hajj, H., Novak, S. W., Sanchez, M. G., Cicchetti, F., Tremblay, M. È. Microglial physiological properties and interactions with synapses are altered at presymptomatic stages in a mouse model of Huntington’s disease pathology. J. Neuroinflammation, 17: 2020; doi: 10.1186/s12974-020-01782-9

Shaerzadeh, F., Phan, L., Miller, D., Dacquel, M., Hachmeister, W., Hansen, C., Bechtle, A., Tu, D., Martcheva, M., Foster, T. C., Kumar, A., Streit, W. J., Khoshbouei, H. Microglia senescence occurs in both substantia nigra and ventral tegmental area. Glia, 68: 2228–2245, 2020; doi: 10.1002/glia.23834

Shahidehpour, R. K., Higdon, R. E., Crawford, N. G., Neltner, J. H., Ighodaro, E. T., Patel, E., Price, D., Nelson, P. T., Bachstetter, A. D. Dystrophic microglia are associated with neurodegenerative disease and not healthy aging in the human brain. Neurobiol. Aging., 99: 19–27, 2021; doi: 10.1016/j.neurobiolaging.2020.12.003

Silburt, J., Aubert, I. MORPHIOUS: an unsupervised machine learning workflow to detect the activation of microglia and astrocytes. J. Neuroinflammation, 19: 2022; doi: 10.1186/s12974-021-02376-9

Spiller, K. J., Restrepo, C. R., Khan, T., Dominique, M. A., Fang, T. C., Canter, R. G., Roberts, C. J., Miller, K. R., Ransohoff, R. M., Trojanowski, J. Q., Lee, V. M. Y. Microglia-mediated recovery from ALS-relevant motor neuron degeneration in a mouse model of TDP-43 proteinopathy. Nat. Neurosci., 21: 329–340, 2018; doi: 10.1038/s41593-018-0083-7

Verdonk, F., Roux, P., Flamant, P., Fiette, L., Bozza, F. A., Simard, S., Lemaire, M., Plaud, B., Shorte, S. L., Sharshar, T., Chrétien, F., Danckaert, A. Phenotypic clustering: A novel method for microglial morphology analysis. J. Neuroinflammation, 13: 2016; doi: 10.1186/s12974-016-0614-7

Zhou, Q., Mareljic, N., Michaelsen, M., Parhizkar, S., Heindl, S., Nuscher, B., Farny, D., Czuppa, M., Schludi, C., Graf, A., Krebs, S., Blum, H., Feederle, R., Roth, S., Haass, C., Arzberger, T., Liesz, A., Edbauer, D. Active poly‐GA vaccination prevents microglia activation and motor deficits in a C9orf72 mouse model. EMBO Mol. Med., 12: 2020; doi: 10.15252/emmm.201910919


Keywords

Alpha-Synuclein: a presynaptic neuronal protein that is genetically and neuropathologically associated with Parkinson's disease (PD).  

Alzheimer's Disease: a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and behavioral changes. It is associated with the accumulation of amyloid-beta plaques and tau tangles in the brain, and loss of neurons and synapses in the cerebral cortex and subcortical regions.

Amyotrophic Lateral Sclerosis (ALS): also known as Lou Gehrig's disease, it is the most common form of motor neuron disease and affects the upper and lower motor neurons. This fatal neuromuscular disease is characterized by progressive weakness of the muscles required to move, speak, eat, and breathe.

Disease-Associated Microglia (DAM): a subset of microglia with a specific transcriptional signature – conserved in humans and mice – thought to play an important role in neurodegenerative disorders. Originally discovered in AD, but also appear to be present in other neurodegenerative diseases (Deczkowska, 2018). Whether they are more accurately described as one signature across diseases or distinct signatures in distinct diseases is still under investigation (Paolicelli, 2022).

Doxycycline (Dox): a tetracycline analog that is used to regulate gene expression using a Tet-On or Tet-Off system. 

Low Dox Model: a variation of the standard ΔNLS model using a protocol developed by Biospective to generate a less severe, slower progressing phenotype.  

Microglia Morphometrics: quantitative measures of microglia morphology, such as cell area, soma perimeter, number of branching points in processes’ skeleton, etc.

Neurodegeneration: a complex, multifactorial process resulting in the loss of neurons.

Nuclear Localization Signal (NLS): a short peptide which facilitates the transport of a protein from the cytoplasm into the nucleus of a cell.

Parkinson's Disease (PD): a neurodegenerative disease that is characterized by: (a) movement-related (motor) symptoms, including resting tremor, bradykinesia, rigidity, and postural instability, related to the loss of dopaminergic neurons in the substantia nigra; and (b) non-motor symptoms including anxiety, depression, apathy, hallucinations, constipation, orthostatic hypotension, sleep disorders, hyposmia/anosmia, and cognitive impairment.

Preformed Fibril (PFF): recombinant, monomeric proteins (e.g. alpha-synuclein) that are incubated under specific conditions to generate aggregated, misfolded fibrils. 

Reactive Microglia: microglia that are responding to or reacting to a particular condition. The name was proposed by Paolicelli et al. (Paolicelli, 2022) in lieu of the discouraged term “activated” microglia, emphasizing that microglia can have many different “reactive states” in health and disease.

Transactive Response DNA Binding Protein of 43 kDa (TDP-43): a highly conserved nuclear RNA/DNA-binding protein encoded by the TARDBP gene involved in the regulation of RNA processing.


Related Content

Up-to-date information on neurodegenerative diseases and best practices related to the evaluation of therapeutic agents in neurodegenerative disease models.

More Information

Let us know what you’re interested in. Our team will be happy to discuss with you!

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

Name*
Email*
Phone Number*
Affiliation (Company/University)*
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.