lecture 18 - Using animal models to investigate genetic effects on behaviour Flashcards

(31 cards)

1
Q

What is an animal model?

A

‘A living, non-human being used to understand the biological basis of healthy and pathological human phenotypes, and how to alleviate the latter, without the risk of harming
an actual human being during the process’
Criteria for a good animal model
* ‘Face validity’ (i.e. does the model resemble the human phenotype?)
* ‘Construct validity’ (i.e. do the model and the human phenotype share common
biological underpinnings?)
* ‘Predictive validity’ i.e. do therapeutic drugs have same effect in humans and model? Can model be used to screen for new treatments?

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Types of animal model - surgical

A
  • Occlusion of middle cerebral artery (stroke)
  • Brain lesions
  • Gonadectomy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Types of animal model - Administration of chemical or biological agents or radiation

A
  • Metrazol (pentylenetetrazol) administration (epilepsy)
  • Immunisation with auto-antigen (autoimmune disorders)
  • Administration of pathogenic and non-pathogenic micro-organisms (infectious diseases) (effects of gut
    microbiota on brain function)
  • Neurotransmitter agonists/antagonists or enzyme inhibitors (healthy/pathological behaviours)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Types of animal model - Genetic

A
  • Manipulation of genomic DNA
  • Administration of genetic material (to affect transcription/translation, as an experimental tool)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Advantages of using animal models to understand gene (dys)function

A
  • Examine in vivo effects of manipulation on brain function/behaviour (emergent property
    of integrated physiological systems) cf. cellular models; similarity of physiology to humans
  • Accessibility of neural tissue and amenability to procedures that would not be ethical in
    humans e.g. interactions between drug administration and genetic lesion
  • Can be maintained in large colonies
  • Good breeders with short generation times
  • Experimental control (regulated genetic background, environment)
  • Genomes amenable to genetic manipulation; similarity with human genome
  • Wide repertoire of sophisticated behaviours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Disadvantages of (genetic) animal models

A
  • Genetic and physiological divergence from humans
  • Different evolutionary histories (e.g. sensory modalities, social groupings)
  • Limited range of genetic modifications possible - now there are more technologies
  • Relevance to complex human behaviours influenced by combined effects of many genes –
    endophenotypes! Hard to look at age-related diseases e.g. AD or HD
  • Ethical issues regarding possible adverse effects (e.g. ‘psychiatric’ phenotypes)
  • Inability to accurately model human-specific phenotypes e.g. language, psychosis
  • Models rarely have true face, construct and predictive validity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Commonly used genetic animal models

A
  • GM non-human primates are very rarely used for research,
    but do exist (rhesus monkey ANDi, GFP transgene)
    Chan et al. (2001) Science 291:309-12
  • Caenorhabditis elegans (nematode worm)
  • Drosophila melanogaster (fruit fly)
  • Danio rerio (zebrafish)
  • Prairie (monogamous) and meadow (promiscuous) voles
  • Mus musculus (and other mouse sub-species)
  • Rattus norvegicus (rat)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

C. elegans

A
  • Molecular and developmental characterisation by Brenner (early 1970s); first multicellular organism to have its genome sequenced
  • Well-defined developmental fate for every cell (1031 in adult male); transparent
  • Simplest organism with a nervous system (302 neurons); ‘connectome’ characterised
  • Many strains with defined genetic mutations; can be frozen and thawed for storage and transport
  • Can be exposed to double-stranded RNAi (infusion, injection or through bacterial ingestion) to disable individual genes
  • Can be administered drugs readily
  • Exhibits chemotaxis, thermotaxis, learning and memory, mating behaviours
  • Can be used to study complex processes e.g. nicotine dependence (acute response, tolerance, withdrawal and sensitisation)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

D. melanogaster

A
  • Used as a genetic model from early 1900s onwards; genome sequenced and published in 2000
    • Have lots of mitochondria for flight - neurodegenerative diseases such as alchzeimers are caused by changes in mitochondria function - can use flies to see how to treat
  • Only four pairs of chromosomes (3 autosome pairs, and one sex chromosome pair); used to
    study fundamental mechanisms of transcription and translation
  • Genome can be readily manipulated (since 1987)
  • Morphology (including ‘nervous system’) easily identifiable
  • Used as a genetic model for neurodegenerative
    disorders (PD, AD, HD) and effects of oxidative
    stress/ageing
  • Also used to examine genetics of circadian rhythm,
    sensory function, locomotor activity, courtship, pain,
    and learning and memory
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Zebrafish

A
  • Used as a lab model from 1960s onwards; reference genome sequence published in 2013
  • Genome can be readily manipulated
  • Expression of specific genes can be acutely altered through use of ‘morpholino antisense oligonucleotides’
    (bind to mRNA sequences and prevent translation to protein)
  • Embryos large, robust, transparent and able to develop outside of the mother
  • Well-characterised, easily observable and testable range of (developmental) behaviours
  • Diurnal sleep cycle
  • Anxiety-related and exploratory behaviours
  • Chemosensory behaviours
  • Response choice and inhibition
  • Social behaviours
  • Cognitive and executive functions
  • Similar response to mammals in toxicity testing – utility for high-throughput screening of novel therapeutics?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

three- choice serial reaction time task for zebrafish - Parker et al

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Rodents (mice and rats)

A
  • Mice used as lab models since 16th Century by, inter alia, Harvey, Hooke, Priestley and Mendel; rats used as
    models from early 1800s – first animal domesticated for purely scientific reasons
  • Mouse (C57BL/6 strain) genome sequenced and published in 2002 (second mammalian genome after
    human); rat genome sequenced and published in 2004
  • Mouse genome readily manipulated; rat genome less so, until recently (see later)
  • Mammals, therefore high degree of genetic and physiological homology with humans
  • Range of sophisticated behavioural phenotypes; can examine genetic effects on:
  • Courtship and mating behaviours
  • Dam-pup interactions
  • Social behaviours
  • Circadian rhythms
  • Motor function
  • Anxiety-related and exploratory behaviours
  • Cognition and executive function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Genetic rodent models

A

The rodent genome may be modified in a number of ways to assess effects on brain and behaviour:
* Selective breeding (inbreeding/outbreeding)
* Gene ‘knockout’
* Transgenesis and ‘knock-in’
* Mutagenesis using chemicals or radioactivity
* Chromosomal mutations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

selective breeding

A

Inbreeding (mainly mice): Selected members of a founder strain are repeatedly inbred over many generations to
(theoretically) ensure genetic homogeneity (→ less phenotypic variability, can dissociate genetic vs. environment
influences)
Commonly used inbred strains include C57BL/6, BALB/c, 129 and BTBR (autism), and Spontaneously Hypertensive
Rat (ADHD); inbred strains can differ significantly in appearance and behaviour (polymorphisms)
Outbreeding: Members of a founder strain are bred to unrelated individuals to ensure genetic heterogeneity
(→ more phenotypic variability (more like humans?), ‘hybrid vigour’)
Commonly used outbred strains include CD-1, MF1, Swiss-Webster (mice) and Lister Hooded, Long-Evans,
Sprague Dawley and Wistar (rats)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

recombinant inbred strains

A

crosses between phenotypically-distinct inbred strains for several generations can
help identify regions of the genome affecting behaviour
Highly active Moderately active
* Examine activity phenotype for
hundreds of individual animals
* Examine origin of genetic material
at multiple genomic sites for all
animals
* Correlate the two to see which
genetic sites seem to influence the
behaviour most

less black = less active

genetic diagram in notes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Gene knockout: removal of gene function
Prof Sir Martin Evans (Cardiff University), Nobel Prize 2007

A

Genetic insert can be designed so that ‘knockout’ only occurs at certain developmental stages
or in certain tissues → ‘conditional knockout’
put genetically modified cell from C57BL6 mouse into blastocyst of another mouse - white fur
In some mice genetic change is passed into sperm or egg and then offspring so then offspring have genetic change in one of each pair of chromosome = hetereozygous then breed to together so have genetic change in all chromosome

17
Q

Transgenesis

A

Function is to either: a) insert exogenous sequence into genome (e.g. gene from one species into another) or b)
to have more copies (i.e. higher expression) of a particular gene than normal
* Pronuclear injection of DNA sequence of interest; lack of specificity re target and copy number
Lots of control manipulations needed to work out where gene is inserted and what it is effecting

18
Q

‘Knock-in’

A
  • Delete target gene
  • Insert altered version of the gene
    containing mutation of interest
    ‘Knock-in’

These are genotype driven approaches

19
Q

Mutagenesis using chemicals (‘phenotype-driven approach’)

A

Wildtype male mice treated with mutagen
e.g. N-ethyl-N-nitrosourea (ENU) →
~1000 random mutations in genome;
breed to wildtype female mice
* Identify any progeny mice with interesting phenotypes and select for breeding
* Over several generations, selectively breed mice showing phenotype of interest
* Eventually, all phenotypically interesting descendants will theoretically possess just one point mutation
which is associated with the phenotype of interest; this can then be identified by genome sequencing

Where you look at behaviour and work out what caused it

20
Q

New technologies

A

Custom-made genetically-altered animals can be readily easily and cheaply generated using new technologies
which selectively edit chosen parts of the genome
Zinc finger nucleases (ZFNs): ‘molecular scissors’ (Guerts et al. (2009) Science 325:433)
CRISPR (Clustered Regularly-Interspaced Short Palindromic Repeats):
RNA-guided genome-editing tool based around the components of the prokaryotic immune system (Guan et al.
(2014) Methods Enzymology 546:297-317)
Potential ethical
issues re humans?

21
Q

fragile X syndrome

A

caused by a genetic change in a gene on the X chromosome called FMR1 and the genetic change is the repeat of this cgg sequence so you get lots of CGGs repeating which causes the gene to become dysfunctional as this gene sequence is extensively methylated

animal models have should you can use crispr technology to get rid of the extensive silencing and can restore gene function that is typically lost

that might have clinical benefits in terms of treatment of fragile X

22
Q

Chromosomal mutations

A
  • Occasionally, spontaneous chromosomal mutations may occur in rodents (e.g. due to impaired segregation of
    the chromosomes at meiosis); rates may be increased by mutagens e.g. radioactivity
  • Some of these animals may be fertile and can be bred from e.g. 39,XY*O mouse - mutant mice where the x chromosome and the y chromosome have become stuck together which causes a deletion of the STS gene - this may predispose to ADHD - to test this looked at activity across 24 hours in these mutant mice compared to normal male mice - found mutant mice tend to be more active during the night which is mirroring one aspect of ADHD some degree of face validity
    Trent et al. (2012) Psychoneuroendocrinology 37:221-9

constraint validity in the deletion of STS in humans is associated with the conditions called X-linked theosis associated with an increased risk or likelihood of being diagnosed with ADHD

then went on to test mice in the mouse version of the zebrafish behaviour test - test of attention so in mice its the five time serial reaction time task. a light flashes on briefly and mice have to poke the light when it appears, if they respond correctly they a condensed milk award. you can alter the attentional demands of the task by varying the duration for the light stimulus is presented - it was varied from 0.7 seconds to 0.1 seconds. you look at the number of time that the mice missed the stimulus. the chromosomal mutant mice tended to miss the stimulus more under the most attentionally demanding conditions when the light flashed on for 0.1 seconds.
these mice seem to be hyperactive, inattentive so may be a viable model for understanding the neurobiology of ADHD. chromosomal mutant mice can slo be used to study human chromosomal conditions like down syndrome

23
Q

mouse model for Down syndrome (trisomy 21) (O’Doherty et al. (2005) Science 309:2033-2037)

A

In humans down syndrome is caused by having 3 copies of chromosome 21 rather than the usual 2
in this Tc1 (‘transchromosomic 1’) model, mice have a human chromosome 21 (inserted at ES cell stage) in
addition to their own set of chromosomes
* These mice exhibit phenotypes relevant to DS including altered:
* behaviour
* synaptic plasticity
* cerebellar morphology
* heart development
* mandible size

  • Model may now be used to investigate molecular
    pathways resulting in DS→ potential therapeutic
    approaches e.g. ‘inactivation’ of additional
    chromosome (Jiang et al. (2013) Nature 500:296-
    300)
  • Overlap between DS and AD → common
    underlying mechanisms?

can use it to explore new clinical approaches

individuals with DS have massively increased likelihood of getting early onset dementia - in chromosome 21 gene called APP and if DS you overexpress the APP gene which might make it more available for abnormal processing so increases risk of alzheimers

using the animal model we can look at the relationship between DS and vulnerability to early onset alzheimers disease

24
Q

Administration of molecules affecting gene/protein expression

A

‘Transient gene knockdown’ to characterise function of poorly-annotated gene
* Molecules (e.g. antisense oligonucleotides, short interfering RNAs (siRNAs) or morpholinos) introduced into
the brain (directly or via viruses) to inhibit transcription/translation of a specific gene
* Issues with poor permeability/diffusivity in adult brain but technique is routinely done in utero

Zhang et al. (2013) The X-linked intellectual
disability protein PHF6 associates with the PAF1 complex and
regulates neuronal migration in the mammalian brain Neuron
78(6):986-93
* Drugs altering epigenome e.g. sodium butyrate

you can take a pregnant mouse and give it a C-section and full out its womb and the lumps are the pups typically get 6 or 7 and you induce a little electrical charge across the head of each these mice, inject a particle genetic construct which inhibits the functions of a particular protein in the brain of that animal. once you have done that it inhibits the function of this protein transiently you then put the womb back into the mum. let her give birth normally let the pups grow up and behaviourally test the pups when their adults and you can look to see whether this transient disruption of expression to proteins which you think might be important in brain development in these mice has impacted upon their later behaviour and later brain structure. these techniques are used to study genes that are associated with neurodevelopmental conditions such as ADHD, autism, intellectual disability. we can see whether those particular genes, those particular proteins are important in early prenatal Brain development and whether they are associated with similar types of phenotypes when these pups group up and become adults.

25
new genetic approaches
‘Optogenetics’ (Gero Miesenbock, University of Oxford, Karl Deisseroth, Stanford University) its a technique where you can selectively activate a bit of the brain that you're interested in and look at how that impacts upon behaviour. to do this you take a virus and stick a gene in it that encodes a receptor that responds to light - a gene from a particular algae. you then insert that virus and that gene into the brain regions and the neurons that you are particularly interested in. you then implant a cannula into the brain of your animal switch on a light which is then detected by the receptors in the particular brain region of interest and that then activates that specific brain circuity. you could then turn the light off and the activity on that Brains circitury goes back to normal. by using this technique by switching the light on and off you can activate and deactivate particular circuits within the brain and you can look at what activation of those particular circuits does to the animals behaviour
26
genetic mouse model relevant to OCD
these mice tended to groom themselves compulsively. stroked their faces till they wiped all the fur of their faces and the researchers identified that this behavioural phenotype was due to hypo activity under activity of cortical striatal and circuitry using ontogenetic techniques they inserted this virus and algae gene into the skull of straital circuity of these mice, they implanted the light they switched the lights on activated the cortical striatal circuitry and show that by activating the circuity they restored function to normal levels and these mice could reduce levels of compulsive grooming optogenetics might be a therapeutic strategy for use in humans potentially and people with OCD who have hypoactive cortical striatal regions but theres issues with it in humans in terms of the human brain being much bigger than that of a mouse so its difficult for light to permeate through it so there lots of work now being done to try to develop a technique which gets around these problems.
27
‘DREADDs’ (‘Designer Receptor Exclusively Activated by Designer Drugs’)
* DNA encoding designer G-protein coupled receptors that do not respond to endogenous chemicals inserted into specific neurons/circuits * Drugs binding to these receptors introduced into the animal to modulate (turn up, turn down) the activity of the circuit → assess resultant effects on behaviour Ferguson et al. (2013) Direct-pathway striatal neurons regulate the retention of decision-making strategies J Neurosci 33(28): 11668-76
28
Summary
* Animal models offer greater experimental control and amenability than human studies for examining genetic effects on behaviour * There are a number of commonly used genetic animal models, each with its own set of advantages and disadvantages * Alterations to the genomic DNA sequence can lead to absence of gene expression (‘knockout’), altered gene function (‘knockin’) or increased gene expression (‘transgenesis’); these changes can be made to occur at selected timepoints/in selected tissues (‘conditional’ approaches) * Brain gene expression can also be altered in a specific manner by introducing small genetic sequences that impair transcription and/or translation * New genetic techniques (e.g. CRISPR, optogenetics and DREADDs) are revolutionising our knowledge about the genetic substrates and neural circuitry underlying particular behaviours
29
Practice essay question
‘Discuss how research in animals might be used to develop new genetic therapies’
30
* Yizhar (2012) Optogenetic insights into social behaviour function Biol Psychiatry 71(12):1075-80
1. Introduction * Stress is a major risk factor for mental health disorders. * Chronic stress affects brain function, particularly in areas linked to emotion and cognition. * Understanding stress mechanisms can help develop better treatments. 2. Stress and the Brain * Chronic stress impacts key brain regions: ○ Hippocampus: Involved in memory; stress reduces its size and function. ○ Amygdala: Controls emotions; stress increases its activity, leading to heightened fear and anxiety. ○ Prefrontal Cortex: Regulates emotions and decision-making; stress impairs its function. * Stress alters neurotransmitters like serotonin, dopamine, and glutamate, which are critical for mood regulation. 3. The Role of Cortisol and the HPA Axis * The Hypothalamic-Pituitary-Adrenal (HPA) Axis controls stress responses by releasing cortisol. * Chronic stress leads to dysregulation of cortisol, which can contribute to anxiety and depression. * High cortisol levels damage brain structures and disrupt neural communication. 4. Early-Life Stress and Long-Term Effects * Childhood trauma and stress increase the risk of mental disorders in adulthood. * Early stress exposure alters brain development, making individuals more vulnerable to stress later in life. * Epigenetics plays a role in how stress effects can be passed down through generations. 5. Genetic and Environmental Interactions * Individual genetic differences affect stress responses. * Environmental factors like social support can buffer stress effects. * Gene-environment interactions determine resilience or susceptibility to stress-related disorders. 6. Stress and Mental Health Disorders * Chronic stress is a major contributor to: ○ Depression: Linked to reduced hippocampal volume and serotonin dysfunction. ○ Anxiety Disorders: Increased amygdala activation and heightened fear responses. ○ Post-Traumatic Stress Disorder (PTSD): Impaired memory processing and exaggerated fear responses. 7. Coping Strategies and Treatments * Pharmacological Approaches: ○ Antidepressants (SSRIs) help regulate serotonin and reduce stress effects. ○ Cortisol blockers are being researched as potential treatments. * Behavioral and Psychological Interventions: ○ Cognitive Behavioral Therapy (CBT) helps reframe negative thought patterns. ○ Mindfulness and meditation reduce stress and improve emotional regulation. ○ Exercise promotes neuroplasticity and reduces stress hormones. * Social Support and Lifestyle Changes: ○ Strong social connections help buffer stress. ○ Proper sleep, nutrition, and relaxation techniques are key for stress management. 8. Conclusion * Stress plays a central role in mental health disorders through its effects on the brain and body. * Understanding these mechanisms can improve prevention and treatment strategies. Future research should focus on personalized approaches based on genetics, environment, and lifestyle factors.
31
* Walters et al. (2016) Advanced in vivo use of CRISPR/Cas9 and antisense DNA inhibition for gene manipulation in the brain Front Genet 6:362
Gene editing tools are crucial for understanding brain function and neuro disorders. Recent advancements, particularly CRISPR/Cas9 and anti-sense nucleotide-based techniques, allow for efficient genome modifications in the adult nervous system. CRISPR/Cas9 is highly versatile, enabling gene knockouts, mutations, expression control, and epigenetic modifications, even targeting multiple genes simultaneously. Though its advanced applications in neuroscience are still developing, the technology is widely accessible. Anti-sense nucleotide-based methods offer a simpler, rapid way to knock down genes with minimal expertise. This article explores these tools and their potential impact on neuroscience research Genes play a crucial role in human behavior, and dysfunctions in gene expression contribute to various neurological and neuropsychiatric disorders. Understanding how genes influence behavior is essential for both basic neuroscience and developing new treatments. Since gene regulation varies across brain regions, gene editing must be precise in both timing and location. To address these challenges, advanced gene editing tools have been developed. This article highlights CRISPR/Cas9 for its flexibility and ease of use, as well as anti-sense oligonucleotides for their accessibility and simplicity in gene knockdown applications in neuroscience research THE EVOLUTION OF GENE EDITING TOOLS The evolution of gene editing tools has progressed from pharmacological approaches to advanced genetic manipulation techniques. Early methods used drugs to modulate protein activity, but they lacked specificity. The development of transgenic knockout mice allowed for targeted gene manipulation, though early methods lacked temporal control. The Cre-LoxP system improved tissue specificity, and later inducible Cre systems allowed for precise timing of gene deletion. Viral delivery methods further enhanced control but required additional technical steps. RNA interference (RNAi) techniques, such as shRNA, enabled gene knockdown in wild-type rodents without transgenic models, though they required careful design to avoid off-target effects. Despite these advancements, gene editing tools remained time-intensive and complex. Newer technologies like CRISPR/Cas9 offer a highly flexible, user-friendly approach, allowing multiple genes to be studied simultaneously. Additionally, anti-sense nucleotides (ASOs and Morpholinos) provide a simpler alternative by directly modulating gene expression in the brain without the need for viral delivery or transgenic mice. These innovations represent the next generation of gene editing tools in neuroscience. USING CRISPR/Cas9 FOR GENE MODULATION IN THE BRAIN CRISPR/Cas9 has revolutionized gene editing due to its simplicity, flexibility, and efficiency. Unlike RNA interference (RNAi), which is time-consuming and often requires multiple iterations, CRISPR/Cas9 reliably produces gene knockouts and allows for rapid switching between targets, even enabling multiple genes to be edited simultaneously. It is user-friendly, requiring minimal specialized knowledge, and free resources are available for designing targeting sequences. Originally discovered as a bacterial immune defense, CRISPR/Cas9 uses an RNA-guided Cas9 protein to cut DNA at specific sites. Scientists have adapted this system to target and modify genes, introducing double-stranded DNA breaks that facilitate gene knockouts or knock-ins. This makes CRISPR/Cas9 a powerful tool for gene modulation in neuroscience and other fields. Using CRISPR/Cas9 for Gene Knockout CRISPR/Cas9 enables efficient and straightforward gene knockout by creating targeted double-stranded DNA breaks using a single guide RNA (sgRNA) and the Cas9 enzyme. These breaks are repaired through non-homologous end joining (NHEJ), leading to mutations that disrupt gene function. This method is simpler and more efficient than previous techniques, allowing for easy gene targeting and the simultaneous knockout of multiple genes. Studies have demonstrated CRISPR/Cas9’s effectiveness in the brain. For example, Swiech et al. (2015) successfully knocked out the Mecp2 gene in mice, resulting in impaired memory, highlighting the system's potential for behavioral research. They also used CRISPR/Cas9 to target three DNA methyltransferase genes (Dnmt1, Dnmt3a, and Dnmt3b) simultaneously, showing that multi-gene knockouts can reveal complex genetic interactions affecting brain function. However, multi-gene editing efficiency decreased with additional sgRNAs, suggesting potential challenges in targeting multiple genes at once. Despite this, CRISPR/Cas9 remains a powerful tool for neuroscience, offering high flexibility and the potential for large-scale genetic studies in the brain. Use of Cas9 for Knock-in Mutations CRISPR/Cas9 can also introduce knock-in mutations, which are useful for studying disease-related genetic changes. This is typically achieved through the homology-dependent repair (HDR) pathway, which repairs double-stranded DNA (dsDNA) breaks by using a homologous DNA template. However, HDR is inefficient in neurons, making its application in neuroscience challenging. Alternative methods are being explored, such as using the non-homologous end joining (NHEJ) pathway, which is active in neurons, and Cpf1, a Cas9-like enzyme that produces “sticky ends” to facilitate NHEJ-based knock-ins. These methods could allow for the introduction of disease-specific mutations or regulatory sequences like loxP sites directly into the brain. Beyond direct gene editing, CRISPR/Cas9 can also modulate gene expression without cutting DNA. This is achieved using a modified, nuclease-inactive dCas9, which acts as a scaffold to recruit other enzymes that regulate gene activity. Two strategies exist: one modifies dCas9 itself, while the other modifies the single guide RNA (sgRNA), both enabling precise control over gene expression without altering the genome. Using sgRNA/Cas9 as a Scaffold to Inhibit or Activate Genes CRISPR/Cas9 can be used as a scaffold to modify gene expression without altering the genome. By modifying the single guide RNA (sgRNA), transcriptional activators or repressors can be recruited to specific gene regions, such as promoters or enhancers, to either activate or inhibit gene activity. This can be done by fusing transcriptional regulators like HSF1 to the MS2 coat protein (MCP) or by using other RNA-binding proteins. An advantage of this method is the ability to multiplex and regulate multiple genes simultaneously, turning some genes on and others off within the same model. This is done by engineering sgRNAs with different stem loops that recruit specific proteins for activation or repression. This strategy is especially useful for studying neural behaviors by modulating gene expression in the brain. Additionally, this approach allows for position-specific epigenetic modifications, such as histone acetylation, at targeted gene regions, providing new insights into gene regulation and its relationship to behavior. While the use of CRISPRi (repression) and CRISPRa (activation) in the brain has not been fully demonstrated, there is potential for these methods to be applied in the CNS, especially when combined with light-activation techniques for temporal control. However, the main challenge lies in delivering the necessary constructs into the brain using viral vectors. OFF-TARGET EFFECTS OF CRISPR/Cas9 CRISPR/Cas9 can sometimes cause off-target effects, where the Cas9–sgRNA complex binds to sites in the genome that are similar but not identical to the intended target. The frequency of off-target binding varies depending on the guide sequence. To reduce off-target effects, strategies include: 1. Optimizing sgRNAs to minimize homology with other genomic sites. 2. Shortening sgRNA sequences (e.g., from 20 to 17–19 base pairs) to reduce off-target activity. 3. Modifying Cas9 (e.g., using eCas9) to decrease off-target activity. 4. Verifying results by using multiple sgRNAs targeting the same gene to confirm the results and minimize errors. 5. Paired nickases or FokI: This method requires two sgRNAs targeting different DNA strands to induce a double-stranded break (DSB), reducing off-target effects. FokI fuses to dCas9 (a mutated form of Cas9 without nuclease activity), and only when both sgRNAs bind close together will FokI dimerize to cut the DNA. While these methods reduce off-target effects, they can complicate experiments, especially for in vivo brain editing, as they may require additional vectors or viral delivery systems. DELIVERING CRISPR–Cas9 INTO THE BRAIN The application of CRISPR/Cas9 in neuroscience has been slow, partly due to the challenge of delivering the large Cas9 protein into the brain. Cas9's size often exceeds the packaging limit of AAV vectors, which typically range from 4 to 5 kb. However, there are several strategies to overcome this: 1. Cas9 Transgenic Mice: A mouse model with Cas9 knocked into the ROSA26 locus allows viral vectors to deliver sgRNAs and other proteins. This method eliminates packaging issues but requires a dedicated mouse line, limiting flexibility. 2. Multiple AAVs: One AAV delivers Cas9, while another delivers sgRNA and other proteins. This approach has been effective, with high co-transduction rates, and allows for multiplexing gene knockouts. Larger packaging viruses like HSV can be used to overcome size limitations. 3. Smaller Cas9 Homologs: Cas9 from Streptococcus aureus is smaller than the commonly used Streptococcus pyogenes Cas9, allowing it to be packaged with sgRNA and GFP into a single AAV. These strategies, along with ongoing innovations, are making CRISPR/Cas9 more feasible for in vivo use in neuroscience. Although advanced methods like CRISPRi, CRISPRa, and epigenetic modulation have not been widely tested in vivo, they are poised to transform brain gene editing and neuroepigenetics research. Disrupting gene expression in the brain without using viral vectors or mouse models is possible through anti-sense oligonucleotides (ASOs), which are short DNA sequences that bind to target mRNA, leading to its degradation or translational inhibition. ASOs are advantageous because they can be easily administered in vivo without the need for viral vectors or transgenic animals. However, the use of ASOs in vivo faces challenges, primarily due to their susceptibility to degradation by nucleases. To address this, researchers have developed modified ASOs with more stable backbones, such as phosphorothioate bonds, which improve their stability but may require higher doses and can lead to cytotoxicity. Another modification involves using ribose in the sugar ring, improving binding affinity and protecting ASOs from degradation. Some ASOs also use exon skipping, which can produce functionally inert proteins or restore gene function, providing potential therapeutic benefits, such as in the treatment of spinal muscular atrophy (SMA) by restoring function to the Smn2 gene. While ASOs are primarily used for gene knockdown, their potential to restore gene activity in specific contexts is also being explored, offering new possibilities for gene therapy and neuroscience research. Using Locked Nucleotides for Gene Knockdown Locked nucleic acids (LNAs) are a modification of antisense oligonucleotides (ASOs) that enhance binding affinity and knockdown efficiency by "locking" the ribose backbone into a stable structure. This modification also protects the oligos from degradation and allows them to cross the blood-brain barrier. However, LNAs face limitations, including the inability to recruit RNase H for degradation and potential toxic effects and off-target actions, which hinder their widespread use in vivo. To address these issues, a new generation of ASOs, called "gapmers," was developed. Gapmers combine LNA-modified regions at the ends with unmodified DNA in the middle, allowing RNase H-mediated degradation of the mRNA while benefiting from the stability and improved affinity of the LNA modifications. Gapmers have shown promise in vivo, successfully reducing gene expression in diseases like Huntington's disease and showing long-lasting effects with minimal off-target impacts. Despite their potential, gapmers require caution due to the risk of toxic side effects, especially at high doses. Advances in ASO design are needed to minimize these risks and maximize their effectiveness in neuroscience research. Overall, LNA-based gapmers are a powerful tool for gene knockdown in the brain and offer great promise for future therapeutic applications. USE OF MORPHOLINOS TO REGULATE GENE EXPRESSION IN THE BRAIN Morpholinos (PMOs) are synthetic oligonucleotides with a unique backbone that makes them resistant to nuclease degradation, allowing them to remain active for extended periods. Unlike ASOs, PMOs do not recruit RNase H, but they can still achieve gene knockdown by interfering with splicing, translation, or miRNA maturation. PMOs are unable to cross cell membranes unassisted, limiting their general use. However, modifications like vivo-Morpholinos, which link PMOs to dendrimers or cell-penetrating peptides, improve their cell permeability and make them more effective in vivo, especially when directly injected into the brain. vivo-Morpholinos have shown significant promise in gene suppression in the brain with minimal cytotoxicity, making them a valuable tool for gene editing in brain research. These modifications, along with gapmer-based ASOs, offer direct application to target areas without the need for viral vectors or mouse models, providing a powerful approach to gene regulation. In conclusion, recent gene-editing approaches like CRISPR/Cas9 and anti-sense mRNA inhibition are transforming neuroscience research by offering flexible, effective, and accessible tools for studying gene function in the brain. These methods allow for targeted gene disruption and rapid knockdown, providing essential insights into gene-behavior relationships.