Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.

Page created by Ida Bowen
 
CONTINUE READING
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Models applicable in studying
  Parkinson disease: from
 vertebrates to invertebrates

       Olasunmbo Afolayan Ph.D.
            Neuroscience Unit
          Department of Anatomy
            College of Medicine
            University of Lagos
                  Nigeria
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Outline
• Overview of Parkinson disease (PD)
• Pathogenesis of PD
• Why modeling PD?
• Criteria for selecting models for PD
• Parkinson disease models
• conclusion
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Overview
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
PARKINSON DISEASE

     • Is a progressive neurodegenerative movement disorder

     • Affects about ~1% of population of >65 years

     • Is the second most common after AD (Cabreira and Massano, 2019)

     • Is the most common neurological NCD in the aging African
       population (Derek et al., 2020)

     • Sporadic (15% among patients) and familial (5% among patients)
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Pathological hallmarks of PD
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Neuropathological features

                             Powel et al., 2017, Nat Rev. Disease Primer
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Molecular mechanism in the pathogenesis of PD

                              Powel et al., 2017, Nat Rev. Disease Primer
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Clinical symptoms associated with Parkinson disease

                                  Powel et al., 2017, Nat Rev. Disease Primer
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Models of Parkinson Disease
Models applicable in studying Parkinson disease: from vertebrates to invertebrates - Olasunmbo Afolayan Ph.D.
Why modelling PD?

• To understand the heterogeneity in the pathogenesis
  of PD

• To design and provide test beds for novel disease-
  modifying therapies
Criteria for
                                Constructive   Predictive
Animal model    Face validity
                                  validity      validity
 validation.
Models of Parkinson Disease
 Chemically induced models

 Genetic models

 Combined (Chemical and Genetic) models
Chemically induced models
Cont’d
 It involves the use of chemicals
 such as:
 •   1-methyl-4-phenyl-1,2,3,6-
     tetrahydropyridine (MPTP)
 •   6-hydroxydopamine (6-OHDA)
 •   Paraquat and Maneb
 •   Rotenone
 A variety of vertebrates and non-
 vertebrates can be used in these
 models (Blesa et al., 2012).
Features of chemical models

                              Blesa et al., 2012
Genetic models
• Involves the use of genetically modified animals following the
  understanding of the genetic interplay in PD.

• Genes that have been implicated with familial form of PD include:

   • alpha synuclein which increases the formation of Lewy bodies

   • Parkin (causes early onset of PD)

   • PTEN-induced putative kinase 1 (PINK1) (shows Lewy-related pathology)

   • DJ-1 (autosomal recessive PD with mild motor deficits without dopaminergic
     neuron loss).

• The major disadvantage of this model is that it is unclear if the results
  can be directly applied to humans.
Chemical and Genetic models

This involves the use of a chemical (neurotoxin) in a genetically modified
animal

   • MPTP induces more neuronal loss in DJ-1 knockout mice

Advantages:
   1. captures the multifaceted nature of PD
   2. might be the most ideal and most promising type of model
   3. High sensitivity to neurotoxin
Animals used in modelling PD
Broadly categorized into vertebrates and invertebrates models

• Vertebrates
             non-human primates- Marmosets
             Rodents – Rats and mice
             fishes – zebra fish
• Invertebrates
             Drosophila
             C. elegans
Vertebrates
Animals used in modelling PD Cont’d
Non-human primates
Such as the new world monkeys and old world monkeys

Advantages:
• Have anatomic, genetic, physiologic and behavioral proximity
  to humans
• Functional organization of the striatal regions are similar to
  humans
• Manifest many hallmarks of PD such as motor and non-motor                   Disadvantages:
  skills
                                                                              1. Ethical and
• Monkeys have better developed fine motor skills than rodents                   economical hurdles
    • a similar hand-foot use for measuring complex motor skill such as the   2. Not suitable for
      hand-eye coordination                                                      conducting pilot
    • vertical body position, which allows better analysis of postural           basic science
      imbalances                                                                 research
                                                                              3. No high throughput
                                                                                 screening
MURINE MODEL
                                                                                                        Advantages:

                                                                                                        1. Biological processes between
                                                                                                           rodents and humans are highly
                                                                                                           conserved
                                                                                                        2. Ability to mimic and
                                                                                                           recapitulate some complex
                                                                                                           pathways and features of PD

                                                                                                          Disadvantages:

                                                                                                          1. Ethical restrictions
                                                                                                          2. Not suitable for high
                                                                                                             throughput screening
                                                                                                          3. Distinct sensitivity of
                                                                                                             strains to neurotoxins

Photomicrograph of a 6-OHDA lesioned rat striatum immunostained for tyrosine
hydroxylase (TH). Densities of TH immunoreactivity striatal fibers are clearly reduced
after the 6- OHDA injection (right side) as compared to the densities of striatal TH-
immunoreactivity fibers in control rat (left side).                                      Blesia et al.,2012 Journal of Biomedicine and Biotechnology
Paraquat injection induced significant decrease in falling time, number of crosses and percentage
alternation behaviour with a concomitant increase in the duration of cataleptic behaviour in the rotarod,
open field, Y‐maze and bar tests, respectively, which was ameliorated by GPD treatment. PQT also
increased lipid peroxidation, peroxynitrite and TNF‐α generations as well as deficit in superoxide
dismutase and GSH activities in the midbrain. PQT‐induced oxidative stress and neuroinflammation was
attenuated by GPD treatment. Findings from this study showed that GPD prevents PQT‐induced motor
dysfunction, memory impairment, oxidative stress and neuroinflammation through enhancement of
antioxidant defense system and inhibition of pro‐inflammatory cytokine release. Thus, GPD could be a
potential adjunct in the management of Parkinsonism.
Zebrafish, Danio rerio (vertebrate)
• Are small and transparent in nature

• Neuronal circuitries involved in movement are
  well characterized

• Molecular   mechanism,    biological   and
  neurobehavioural phenomenology in humans The   approximate anatomical location of the dopaminergic (orange), histaminergic
                                             (purple), noradrenergic (blue), and serotonergic (green) regions are represented in
                                                                        zebrafish brain.
                                                                                                        Vaz et al. 2018 Frontiers in Neurology

• Swimming is used in accessing dopaminergic
  activity

• Reduced swimming indicates bradykinesia
• Several transgenic, knockdown, and mutant zebrafish lines have been generated and characterized

• Knockdown of the β- or γ1-synuclein subunits induces motor impairments

• Knockout of the Parkin gene, produces a moderate (20%) loss of dopaminergic neurons, reduced
  mitochondrial complex I activity and increased susceptibility to toxins

• However, there is no loss of dopaminergic cells when PINK1 is knocked out. however, it alters
  dopaminergic projections and induces locomotor deficits              Saxena, 2013 material and methods.

• Down regulation of PINK1 results in mitochondrial dysfunction that leads to augmented levels of ROS
  and activation of the apoptotic signaling pathway in zebrafish
Invertebrates
Drosophila melanogaster in PD
• Has a short life span

• About 77% of human disease genes are conserved in Drosophila

• gene sequence and function are highly conserved between flies
  and humans

• Suitable for studying mechanisms of PD-related
  neurodegeneration

• The brain and spinal cord are extraordinarily complex

• many PD-related genes are found to have homologues in
  Drosophila

• most of DA neurons are generated at embryogenesis, matured
  and gathered into clusters during first larval stage
                                                                             (a) Illustration of six DA neuron clusters in Drosophila larval central brain. (b)
                                                                             DA neuron clusters in Drosophila adult central brain.

                                          Dung and Thao,2018. Advances in Experimental Medicine and Biology
Drosophila melanogaster as a model
• Expression of α-synuclein in flies confers
  age-dependent motor difficulties and a
  compromise of dopaminergic neurons
  (Feany and Bender, 2000)

• α -synuclein transgenic Drosophila models
  premature loss of climbing ability indicates
  degeneration of dopaminergic neurons
  and α -synuclein formation.

• PINK1 Knock out in Drosophila indicates a
  reduction in the number of Dopaminergic
  neurons & impaired climbing ability.
Caenorhabditis elegans as a model
•   C. elegans has an extremely rapid life cycle (4 days)
•   1/3 of its cells are made up of neurons
•   8 dopaminergic neurons are involved in motor activity
•   Prolific and very amenable to genetic manipulation (Teschendorf and Link, 2009)
•   The transparent nature of C. elegans throughout its life cycle facilitates the use of GFP
    fusion proteins to visualize; neurons, synaptic connections and neuronal death in living
    worms by the morphological appearance of vacuolated neurons (Teschendorf and Link,
    2009)
Caenorhabditis elegans as a model

• over-expressing wild-type or mutant human α-synuclein (A53T or A30P)
  display degeneration of dopaminergic neurons alongside loss of the
  basal slowing response.

• Mutations within the gene leads to the typical decrease in locomotion
  (basal slowing)

• over-expression of wild-type LRRK2 increased survival in response to
  paraquat and rotenone indicating LRRK2 mutations may enhance
  vulnerability in PD
Conclusion
  An ideal model of PD should display pathophysiologic features
  and symptoms of PD.

  However, the current models are not able to recapitulate all PD
  features.

  Each model has both advantages and disadvantages, and the
  selection of a suitable model depends on particular purposes of
  the research.
Thank you for your attention.
References
  Blesa, J., Phani, S., Jackson-Lewis, V., and Przedborski, S. (2012). Classic and New Animal Models of Parkinson’s Disease. Journal of Biomedicine and Biotechnology
  Bose, A., and Beal, M. F. (2016). Mitochondrial dysfunction in Parkinson's disease. Journal of neurochemistry, 139 Suppl 1, 216–231.
  Buhidma, Y., Rukavina, K., Chaudhuri, K.R. et al (2020). Potential of animal models for advancing the understanding and treatment of pain in Parkinson’s disease. npj Parkinsons Dis. 6, 1.
  Cabreira, V., and Massano, J. (2019). Doença de Parkinson: Revisão Clínica e Atualização [Parkinson's Disease: Clinical Review and Update]. Acta medica portuguesa, 32(10), 661–670.
  Cuenca, L., Gil-Martinez, A. L., Cano-Fernandez, L., Sanchez-Rodrigo, C., Estrada, C., Fernandez-Villalba, E., and Herrero, M. T. (2019). Parkinson's disease: a short story of 200 years. Histology
  and histopathology, 34(6), 573–591.
  Dekker, M., Coulibaly, T., Bardien, S., Ross, O. A., Carr, J., and Komolafe, M. (2020). Parkinson's Disease Research on the African Continent: Obstacles and Opportunities. Frontiers in
  neurology, 11, 512.
  Dung V.M., Thao D.T.P. (2018) Parkinson’s Disease Model. In: Yamaguchi M. (eds) Drosophila Models for Human Diseases. Advances in Experimental Medicine and Biology, vol 1076. Springer,
  Singapore.
  Duty, S., and Jenner, P. (2011). Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. British journal of pharmacology, 164(4), 1357–1391.
  Feany MB and Bender WW (2000). A Drosophila model of Parkinson's disease. Nature 404(6776):394-8.
  Jackson-Lewis, V., Przedborski, S (2007). Protocol for the MPTP mouse model of Parkinson's disease. Nat Protoc 2, 141–151.
  Konnova EA, Swanberg M. Animal Models of Parkinson’s Disease. In: Stoker TB, Greenland JC, editors. Parkinson’s Disease: Pathogenesis and Clinical Aspects [Internet]. Brisbane (AU): Codon
  Publications; 2018 Dec 21. Chapter 5.
  Porras, G., Li, Q., & Bezard, E. (2012). Modeling Parkinson's disease in primates: The MPTP model. Cold Spring Harbor perspectives in medicine, 2(3)
  Reich, S. G., and Savitt, J. M. (2019). Parkinson's Disease. The Medical clinics of North America, 103(2), 337–350.
  Sveinbjornsdottir S. (2016). The clinical symptoms of Parkinson's disease. Journal of neurochemistry, 139 Suppl 1, 318–324.
  Teschendorf, D and Link, C.D (2009). What have worm models told us about the mechanisms of neuronal dysfunction in human neurodegenerative diseases?. Mol Neurodegeneration 4, 38.
  Vaz, Rita L., Outeiro, Tiago F., and Ferreira, Joaquim J. (2018). Zebrafish as an Animal Model for Drug Discovery in Parkinson’s Disease and Other Movement Disorders: A Systematic Review.
  Frontiers in Neurology, 9(347).
You can also read