• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/598

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

598 Cards in this Set

  • Front
  • Back
  • 3rd side (hint)

What are the 3 layers of cell development in the embryonic disc

Ectoderm


Mesoderm


Endoderm

What is the formulation of our nervous system called?

Gastrulation

The 6 basic processes of neurodevelopment

Cell birth/proliferation


Cell migration


Cell differentiation and maturation


Synaptogenesis


Cell death and synaptic pruning


Myelination (myelogenesis

2 ways cells migrate

Chemical signals


Physical support provided by radial glia

2 types of dendritic developments

Dendritic arborisation (branching)


Growth of dendritic spines (little swellings of the dendrite on the neuron)

Whats the growing end of the axon called

A growth cone

What are the extentions called that a growth cone makes in order to make contact with another cell

Filopodia

What 2 molecules are secreted from target sites to attract growth cones

Cell adhesion molecules (CAM)


Topic Molecules (attract or repel growth cones)

What is "successful neural connections survive and others die" called

Neural Darwinism

2 Neurotrophic Factors

Nerve Growth Factor


Brain Derived Neurotrophic Factor

Proteins


NGF


BDNF

Word for programmed cell death

Apoptosis

Other type of cell death to apoptosis

Necrosis

(damaging, from brain damage/too much excitability)

For immature neurons to avoid apoptosis and survive it needs:

Neurotrophins


Communications with other neurons (strengthen synapses - Hebb)

2 things

What is dendritic branch changes related to

Learning or experience

What produces myelin

Glia

How quick is the process of myelination

Slow - gradual for decades

What glia produces myelination in the CNS?

Oligodendroglia

What glia produces myelination in PNS

Schwann cells

What stage of development does the brain begin to develop

Embryonic

What is spina bifida

Failure of the closure of the neural fold at the level of the spinal cord

Type of neural tube deficit that entails...

What prevents spina bifida

Folic acid

What is anencephaly

Brain fails to develop

Alcohol enhances which process of cell neurodevelopment

Apoptosis

What is potential

Where male and female anatomy is the same during prenatal development

What are the female ducts called

Müllerian

What are the male ducts called?

Wolffian

What causes the development of the Wolffian ducts to develop into seminal vesicles and vas deferens?

Testosterone

What causes degeneration of the Müllarian ducts?

Müllerian Inhibiting hormone

What do males have more of instead of estrogen (opposite to females)

Androgen

3 brain sex differences

Males have more dendriric branching in the visual cortex.


Females have more dendritic branching in the motor cortex.


Part of corpus callosum bit thicker in females than males.

3 stages of prenatal development

Germinal stage


Embryonic stage


Fetal stage

When is neurogenesis mostly completed?

7 months of gestation

Where was new neuronotenesis postnatally first discoverer

In songbirds

What 2 parts of the adult brain shows neurogenesis

Olfactory bulb


Hippocampus

Does the cerebral cortex show neurogenesis in adults?

Not generally


Exceptions following injury or stroke

The recovery of damaged axons involves what

Collateral sprouting

What is collateral sprouting

Non-damaged axons attaching to vacant spots left from damaged axons

Evidence of "use it or lose it" principle

Hebb - rats kept in home (richer enviroment) faster to solve maze than those in lab

How does enriched environments affect rats brains?

Increases dendritic branching (correlated with improved ability to learn)

MRI scans reveal what 3 things in musicians?

Temporal lobe in right hemisphere is 30% larger.


Thicker grey matter responsible for hand control and vision.


Larger than normal area of the postcentral gyrus in the right hemisphere for movement of left hand.

3 broad waves for critical period in order (younger - older)

Senses


Language


Higher cognition

Richard Tees - train ride

Blakemore and coopers critical period example

Kitten verticle lines no horizontal

What are the 3 parts of Truine Brain - Mc Leans

Neomammalian


Paleomammalian (limbic system)


Reptilian

How does the Truine Brain explain the romanian orphans developmental delays and attachment problems

In neglect, neomammalian brain underdeveloped, less able to control reptillian brain.


In stress/trauma, overdeveloped midbrain/reptillian brain.


= more impulsivity, substance abuse, emotional problems, violence

Explanation for why adolescents are more impulsive

Prefrontal cortex is immature (decision making)


Nucleus accumbens (midbrain) well-developed (pleasure and pain)

What test shoes adolescent impulsivity?

Anti-saccade task (look away from powerful attention getter)

How does the grey matter in childhood change gradually

Thickens in childhood, thins out gradually after

Why does the grey matter change in adolescents?

Synaptic pruning


Increase in white matter (myelination)


Use it or lose it

What are the 4 nucleotide bases?

Adenine (A)


Thymine (T)


Cytosine (C)


Gyanine (G)

What does DNA stand for

Deoxyribonucleic acid

What are the 2 base pairs

Adenine - Thymine


Guyanine - Cytosine

What turns on genes?

Transcription factors

What is transcription

In the nucleus, the gene's dna sequence is copied into messenger RNA (mRNA)

For gene expression

What is gene translation?

A ribosome attaches to the mRNA and moves along it, reading each triplet codon (3 bases) and using transfer RNAs (tRNA) to put together the amino acid chain to make a protein.

In gene expression

What does mitosis make

Somatic cells (daughter cells identical to parents)

What does meiosis make

Gametes (daughter cells contain half number of chromosomes)

What is the 'crossing over' between nonsister chromatids called?

Homologous recombination

What inheritance is Mendel's law

Mendalian

What is Mendelain inheritance

Inheritance through 'transmissible units'


Law of segregation of genes


Law of independent assortments (copies)


Law of dominance (over recessive)

What did peas show for Mendel's law

Tall peas = dominant


Dwarf peas = recessive



TT + dd = 4 × Td


Td + Td = TT, Td, Td, dd

What is it called if genes are identical?

Homozygous

TT or dd

What is it called if genes are not identical?

Heterozygous

Td

What is the name an example of human dominant inheritance

Huntington's Chorea

HC

What does Huntington's Chorea lead to

Progressive deterioration of movement, temperament and cognition

If a parent has huntingtons, what % of the offspring will develop Huntington

50%

What is the cause of Huntington's

Single gene disorder


on chromosome 4


Due to excessive repeat of CAG biases

Name of an example of recessive inheritance

Phenylketonuria (PKU)

What is phenylketonuria?

Build up of phenylalanine toxic to developing brain:


Learning difficulties


Behavioural difficulties


Epilepsy

If both parents are carriers of phenylketonuria, what % of offspring will have the disease

25%

If both parents have phenylketonuria, what % of offspring will be carriers?

50%

What causes phenylketonuria?

Mutation in the PAH gene - enzyme that breaks down dietary phenylalanine

How can phenylketonuria (PKU) be prevented

Diet control

What is monosomy

Single copy of a chromosome

What is trisomy?

Three copies of a chromosome

In downs syndrome, there is a trisomy in which chromosome?

21

What are the characteristics of Downs syndrome

Small brain size


Facial features


Poor muscle tone


Heart condition


Early onset Alzheimer's disease

How do our bodies deal with ensuring cells function normally with either one or 2 X chromosomes

Random X inactivation early in development

The process of X-inactivation can be shown where

Cats

What is an X linked disorder almost exclusively affecting females

Rett syndrome

What does Rett syndrome lead to

Profound mental impairment

What does Rett syndrome do

Mutation in the gene MeCP2 - a transcription repressor turning off expression of unwanted genes

Why is Rett syndrome specific to females

Not all cells express mutated MePC2 gene so milder symptoms

What is an X-linked disorder mostly associated with males

Fragile X

Why is fragile X symptoms predominantly in males

Milder penetrance in females due to X-inactivation so not always recognised

What are alleles

Variants of a gene

What are epigenetics?

Changing how DNA are expressed based on things binding to or changing how the DNA sequence is transcribed, blocking access to genes or making genes more accessible

In epigenetics, does the phenotype or genotype change?

Phenotype

What is the main method for epigenetics

DNA methylation

How does DNA methylation work as an epigenetic mechanism

Methylation repressed gene transcription, difficult to break DNA to get a strain so blocks it

Other type of epigenetic mechanism

Histone modification

What is genomic imprinting

Development form of epigenetics where some genes are myelinated based on if their inherited maternally or paternally (switched off)

What chromosome shows an example of genomic imprinting

Chromosome 15

If a gene is maternally imprinted, what does this mean?

Only express/activate the gene from father

What 2 syndromes can chromosomal 15 depletion create

Prader-Willi syndrome


Angelman syndrome

How is Prader-willi syndrome caused?

Deletion on paternal chromosome, only maternal genes present

How is Angelmann syndrome caused

Deletion on maternal chromosome


Only paternal gene present

What is Prader-Willi syndrome characterised by

Intellectual disability, decreased muscle tone, short stature, emotional lability and insatiable appetite (can = obesity)

What is angelman syndrome characterised by

Delayed development, severe lang impairment, movement and balance problems, small head, sociable behaviour, frequent smiling

Evidence of rats in epigenetics

Maternal care (pup licking) switches on serotonin, activate transcription factor, switch on gene expressing glucocorticoid receptor (GR).


Absense of licking: no transcription factor, promoter methylated - gene becomes methylated


= low levels of GR


loss of GR feedback in HPA axis = ^ stress hormones, ^ anxiety/depression

How do transformational epigenetics work?

Disrupted histones in sperm cells (e.g. smoking/drinking) showed alterned RNA profile in offspring and grand-offspring

What % of our DNA is shared

99.9%

What are natural variations in our DNA known as

Single Nucleotide Polymorphisms (SNPs)

What has looked for what SNPs soetbwith disease state

Genome wide association studies (GWAS)

What is a functional SNP

A SNP that has an effect on expression (by itself)

What's a genetic tagging SNP

A SNP that is associated with a functional change if next to a mutation or variant that is important

What disease can be explained via polygenic, different variants

Alzheimer's

What SNP differences are shown through Alzheimer's disease

APOE3 and APOE4

What is the cell body of a neuron called?

Soma

What is found at the end of the axon, location of the synapse, communication point with other neuron

Terminal button

What are the two sides of the phospholipid molecule in neuronal membranes?

Hydrophillic (likes water)


Hydrophobic

What acts as a framework, holding up the shape of the cell in the neuronal membrane?

Cytoskeleton proteins

What is the resting mebrabe potential?

-65 to -70 milli Volts

Inside the cell

What have they used to observe the resting membrane potential

Giant squid axon in sea water

What are the two forces that cause a membrane potention

Diffusion


Electrostatic force

What is equilibrium potential?

Diffusion force = electrostatic force

What ions is there more of outside of a neuron?

Sodium and chloride

What ions are there more of inside a neuron?

Potassium


Organic anions

During resting state, what forces are acting upon sodium?

Diffusion and electrostatic forces pulling sodium into the neuron

During resting state, what forces are acting upon potassium?

Diffusion pushing potassium out of the cell


Electrostatic keeping in the cell

During resting state, what forces are acting upon chloride

Diffusion pulling Chloride into the cell


Electrostatic pushing out

3 names for the things that allow ions to move into/out of the cell

Ion channel


Leak channel


Resting channel

Why is the overall resting membrane potential negative

Way more potassium channels open than sodium channels


More possibility for potassium to leave than sodium to enter


Resting membrane potential more towards potassium membrane potential

What is used to calculate the equilibrium potential?

The Nernst equation

What does the Nernst equation take into account

Body temperature

What maintains the sodium and potassium gradients across the membrane?

Sodium-Potassium pump

What is broken down to release energy which forces the ions to move against their concentration gradient

ATP

Where is an AP generated

Axon hillock

How is an AP generated

Either by summation of converging inputs from the dendrites or by electrical stimulation (experimentally)

Whats it called when membrane potential becomes more negative than RMP

Hyperpolarisation

Whats it called when membrane potential becomes more positive than RMP

Depolarisation

What is it called when following a small stimulation of a neuron there is a small degree of depolarisation that decays along the length of the neuron?

Decremental conductance

Decay

What opens when the membrane becomes depolarised

Voltage gated channels

What has been used to look at propeties of voltage gated channels

Voltage clamp experiments

Positive membrane potentials (very high degrees of depolarisation) result in what for the channels

Inactivation of the Na+ channels

Depolarising a neuron by +23mV leads to what (as shown by voltage clamp experiments)

Transient inward current followed by a slow outward current


Sodium current: fast (in)


Potassium current: slow (out)

What is conductance

The rate of ion travel through channel

What are the 3 states of the voltage gated channels

Closed


Open


Refractory

What causes refractory periods in voltage gated channels

Inactivation gate closes after activation gate opens

When does sodium voltage gated channels not allow for activation (in an AP)

If +50/65 membrane potential/lots of depolarisation, goes straight into refractory

How are potassium gated channels different to sodium gated channels?

A lot of depolarisation opens the channels

In an AP, what is the first movement of potassium and why

Moves out of the neuron by diffusion

What is it called when the sodium channels hit refractory and potassium still leaves the neuron?

Repolarisation

What causes the hyperpolarisation?

Few remaining open K+ channels and high concentration of K+ outside of the neurone

After hyperpolarisation, what happens?

Na+/K+ pumps work like mad to restore RMP

What happens to the potassium left outside the cell?

Diffuses away

What is propogation for unmyelinated axons?

Unidirectional

Direction

What is the bit inbetween myelin called

Nodes of Ranvier

When an axon is myelinated, where does an AP occur

Only at nodes of Ranvier

Whar happens to the depolarisation caused by AP in myelinated sections

Decays

What are the advantages of myelinated axons?

Less APs are needed to send a nerve impulse


APs require energy (e.g. sodium-potassium pump activations) and time

What is multiple sclerosis?

Disease where neurons have damaged myelin sheath


Leads to:


Loss of sensitivity, muscle weakness, difficulty with coordination and balance

Where is the toxin tetrodoxin (TTX) found?

Puffer fish

What does the toxin tetrodotoxin (TTX) do?

Blocks voltage gated Na+ channels, leads to paralysis then death

Where is the toxin alpha- dendrotoxin found

Green mamba

What does the toxin alpha-dendrotoxin do

Blocks voltage gated K+ channels leading to convulsions

What are the 2 types if synapses

Electrical synapse


Chemical synapse

What are the 3 types of synapses

Axodendric


Axosomatic


Axoaxonic

What is the most common type of synapse

Axodendritic

Lots of inhibitory connection are which synapse

Axosomatic

Why does synapse location matter

EPSP (excitatory post synaptic potentials) decay along dendrite (decremental decay)

Where are EPSPs and IPSPs summated?

Soma (cell body)

When APs get to a synapse, depolarisation opens what?

Voltage-gated calcium channels

What does an influx of calcium im the presynaptic terminal lead to

Neurotransmitter release

Neurotransmitters binding to and activating receptors on the dendrite of the postsynaptic cell leads to what

Depolarisation (excitation) or hyperpolarisation (inhibition) of the postsynaptic cells

If summation shows there is enough depolarisation, what happens

AP is generated at the axon hillock

What is Dales law

If a particular neurotransmitter is released by one of a neurons synaptic endings, the same chemical is released at all synaptic endings of that neuron

What are neurotransmitters stored in presynaptically

Vesicles

4 steps of neurotransmitter release

Depolarisation


Calcium influx


Vesicle infusion


Detatch/move away from dock

2 types of receptors on the postsynaptic membrane

Ionotropic receptor


Metabotropic receptor

Structure of an ionotropic receptor

Protein subunits around a central pore

How quick is the transmission in ionotropic receptors

Fast- immediate change

How can ionotropic receptors have inhibitory/excitatory effects?

Excitatory = +ve ions move into the neuron e.g. glutamate, depolarising


Inhibitory = -ve ions move into the neuron e.g. GABA-A receptors, hyperpolarising

How do Metabotropic receptors work?

Neurotransmitter binds to receptor and activates the G-protein couples receptor


GTP replaces GDP


The G-protein splits and activates other enzymes


Breakdown of GTP turns off G protein activity


Series of chemical reactions leads to amplification of the signal - second messenger system

How quick is the transmission in metabotropic receptors

Slow but bigger effects

What stops neurotransmitters

Deactivating enzymes

What are the 5 different classes of neurotransmitters

Amino acids


Monoamines


Soluble gases


Acetylcholine


Neuropeptide

Endorphins are part of what class of neurotransmitter?

Neuropeptides

Serotonin, dopamine, norepinephrine and epinephrine are part of what class?

Monoamines

Glutamate and GABA are part of what class of neurotransmitters

Amino acids

What are 2 neural integrations boosting EPSPs (name and explanation)

Spatial (multiple excitatory inputs at the same time on dendrite from dif axons)


Temporal (multiple excitatory inputs at different time on dendrite from same axon)

Where are autoreceptors located

Presynaptic terminal

What is the role of autoreceptors

Regulate internal process controlling the synthesis and release of neurotransmitter


Negative feedback mechanism

What are autoreceptors not the same as

Reuptake sites

Are autoreceptors ionotropic or Metabotropic?

Metabotropic (G-protein coupled)

What is neuromodulation?

Alter the presynaptic cell's ability to release more transmitter or the postsynaptic cells ability to respond

Where are amino acids, monoamines and acetylcholine synthesized?

Locally in presynaptic terminal

Where are neuropeptides synthesized?

Cell soma (transported to the terminal)

What does neuropeptides need

Way more calcium globally, not just locally

4 criteria for neurotransmitters

Created presynaptically


Electrical stimulation leads to release


Physiological effect


Termination

CEPT

What does too much glutamate/too little GABA cause

Hyperexcitability - epilepsy


Excitoxicity (cell death response in the brain)

What else causes excitoxicity?

Cerebral ischemia (insufficient blood flow)

Terminology for what transmitter binds to the receptor and how drugs interact

Pharmacology

Receptors vary in what 4 things

Pharmacology


Kinetics


Selectivity


Conductance

What is kinetics

Rate of transmission binding and channel gating (determine the duration of effects)

What are receptors selectivity

What ions are fluxed

Whats a receptor's conductance

The rate of flux

What does glutamate ionotropic receptors flux

Na+

What does GABA ionotropic receptors flux?

Cl-

3 types of ionotropic glutamate receptors

NMDA receptor


AMPA receptor


Kainate receptor

Differences between AMPA and NMDA receptors

Binding of glutamate alone leads to opening for AMPA


NMDA requires a co-agonist: glutamate and glycine, as it is blocked at rest by Mg2+


Voltage-dependent blockage (depolarised e.g. -30mV pushes Mg2+ out, REQUIRED)


NMDA also permeable to Na2+, K+, Ca2+ (AMPA = Na2+, slightly K+)


4 main things

Difference in kinetics of AMPA and NMDA receptors

Slower kinetics of NMDA meaning channels stays open longer


AMPA = fast

Conductance of AMPA and NMDA

Ampa- fast (rate of flux)


NMDA - slow (rate of flux)

Blocking NMDA receptors by drugs leads to what

Symptoms that resemble the hallucinations associated with schizophrenia

What else does dysregulation of NMDA receptors cause (not schizo)

Glutamate excitoxicity (excessive Ca2+ influx)

What is GABA synthesized from?

Glutamate

Whats the effects of too much GABA?

Sedation/coma

At the right dose, what can drugs that increase GABA transmission be used to treat?

Epilepsy

2 GABA agonists

Alcohol


Barbituates

What are the 2 main families of GABA receptors

GABA-A ionotropic receptors


GABA-B metabotropic receptors

What is the structure of GABA-A receptors

Heteropentameric structure - 2 alpha + 3 more subunits (beta, gamma or delta)


6 possible subunits



(Can have 1 alpha i think)

Function of GABA-A receptors

Increases chloride permeability and hyperpolarises the neuron


Decreasing the depolarising effects of an excitatory input

Name of an agonist binding at GABA binding sites that makes people "drunk worse than vodka"

Muscimol

What is an indirect GABA agonist

Benzodiazepines

What do drugs increasing GABA activity reduce?

Anxiety

Name of a GABA antagonist that increases anxiety

Flumazenil

Patients with panic disorder have less what?

Benzodiazepine binding sites

Barbiturates how what effect on the RMP with GABA

More hyperpolarisation

Problems with barbiturates

General depression of neuronal activity includes vital functions like breathing


Poor therapeutic ratio, OD easily, high suicide risk.


LT treatment = dependence and withdrawal

3 things

What is barbiturates used for now

Insomnia and seizures

Benzodiazepines use (5)

Anxiolytic


Anticonvulsant


Sedative


Muscle relaxant


Amnestic

Examples of neuromodulating neurotransmitters

Dopamine


Serotonin


Noradrenaline


Acetylcholine


Histamine


Neuropeptides

What does FM and AM stand for

Frequency modulation


Amplitude modulation

2 exceptions to the rule that the effect of a neurotransmitter is ultimately determined by receptor type

Some neurotransmitters act as neuromodulators (GABA at GABA-B receptors)


Some neuromodulators act as neurotransmitters (ACh at ACh receptors)

4 diffuse modulatory systems

Dopamine


Serotonin


Noradrenaline


Acetylcholine

Where does dopamine originate

Ventral Tegmental Area (VTA) and substantia nigra in midbrain

2 places

3 dopamine systems/projections names

Nigrostriatal system


Mesolimbic system


Mesocortical system

Role of dopamine in the nigrostriatal system

Movement

Dysfunction in the nigrostrial system could lead to what

Parkingsons disease


Huntingtons disease

2 things

Role of dopamine in the Mesolimbic system

Reinforcement (reward)

Dysfunction in Mesolimbic system could lead to what

Addiction (enhanced DA release in NAcc)

Role of dopamine in mesocortical system

Working memory and planning

Dysfunction in the mesocortical system could lead to what

Schizophrenia/addiction

What esssential amino acid is dopamine from

Tyrosine

What is tyrosine catalysed by

Tyrosine hydroxylase

What does tyrosine catalysed lead to

L-Dopa

3 ways for controlling the amount of dopamine

Feedback inhition (too much dopamine lowers tyrosine hydroxylase (TH)) = down regulation of synthesis


DA release leads to phosphorylation of TH increasing activity (feedback still)= up regulation of synthesis (replenish pool)


Prolonged activity in the presynaptic neuron leads to increase in transcription of the TH gene = up regulate (increase size of pool)

What effect does Reserpine have

Impairs storage of monoamines in synaptic vesicles


Causes catalepsy (look soulless, trouble moving)

Rabbits

What is L-DOPA used to treat

Parkinsons

What drug inactives TH

AMPT

How do cocaine, amphetamine and methylphenidate work

Block the reuptake of monoamines into terminals

Where does the noradrenergic system project from?

Locus coerulus

Role of the noradrenergic system

Arousal and attention

What 3 things is dysfunction in the noradrenergic system linked to?

Anxiety


Heroin withdrawal


Depression

5 functions of serotonin systems

Mood


Sleep


Pain


Emotion


Appetite

What amino acid is serotonin made from

Tryptophan

What is trytophan broken into

5-hydroxytryptophan (5-HTP)

What is the rate limiting stage in dopamine synthesis

Tyrosine hydroxylase catalyzing

What is the rate limiting step in serotonin synthesis

Tryprophan obtaining

What complex is involved in the cholinergic systems projections to hippocampus and cortex?

Basal forebrain complex

What neurons project locally in the striatum and cortex

Cholinergic interneurons

What is the rate limiting step in acetylcholine synthesis

Amount of choline

What is acetylcholine rapidly degraded in synaptic cleft by

Acetylcholinesterase (AChE)

AChE inhibitors have helped what

Treatments for Alzheimers disease


Myasthenia gravis (autoimmune disorder)

Botox acts where

Directly at synapse in Neuro-Musclular Junction

Where is ACh released

Neuro-Musclular Junction (NMJ)

Where does tetanus toxin work

Inhibitory (glycine) synapses on chlinergic motor neurons of spinal cord

What does tetanus toxin do

Disinhibits the cholinergic neurons so they continuously fire resulting in permanent muscle contraction

What is the mechanism underlying synaptic strengthening

Long term potentiation

What part of the brain was LTP first observed/studied

Hippocampus

3 properties of LTP

Temporal = summation of inputs reaches a stimulus threshold that leads to the induction of LTP e.g. repetitive stimulation (HFS).


Associative = simultaneously stimulation of a strong and weak pathway will induce LTP at both pathways (spatial summation).


Specific: LTP at one synapse is not propagated to adjacent synapses (input specific).

What's happening at the synapse before LTP? (Small activation)

Glutamate release onto inactive cell (membrane resting)


AMPA receptor activated


NMDA blocked by Mg2+


Depolarisation (from AMPA) not enough to expel Mg2+

Whats happening at the synapse during high activation (freq activation or neighbouring cell) during LTP?

Glutamate release onto an active cell (membrane depolarised)


AMPA receptor activated


Mg2+ block relieved


Na+ through AMPA and NMDA


Ca2+ through NMDA

What does the Ca2+ that has entered through the NMDA receptor in LTP do?

Activate CaMKII

What does CaMKII do?

Phosphorylate existing AMPA receptors increasing their effectiveness


Stimulates the insertion of new AMPA receptors into the membrane

2 things

How does CaMKII have sustained activity after repolarisation

Phosphorylates itself


Molecular switch maintaining increased excitability of neuron for minutes to hours

What are the presynaptic events in LTP

Postsynaptic neuron feeds back to presynaptic neuron by retrograde neurotransmitter - nitric oxide


Leads to increased glutamate release

What is required for long-lasting LTP

Protein synthesis

What is a transcription factor involved in late phase LTP

CREB

What is CREB phosphorylated by

Kinases e.g. PKA or CaMKII

What does late phase LTP do

Can involve morphological changes and the establishment of new synapses

How long does early phase LTP last for

Minutes to an hour

How long does late phase LTP last for

Hours, days or months

What are silent synapses?

Synapses with only NMDA receptors

What is the process of recruiting new synapses in LTP?

EPSPs lead to unused AMPA receptors being recruited, spreading to neighbouring synapses, turning on silent synapses.


Synapses also may split into 2, increasing the number of them

What is the opposite of Long Term Potentiation

Long Term Depression

How is Long Term Depression caused

Low frequency stimulation

What happens to AMPA receptors in Long Term Depression

Dephosphorylated and removed from the membrane

What causes the AMPA receptors to be dephosphorylated in LTD

Same NMDA process, however lower level rises in Ca2+ activate phophatase rather than kinase

What did the Morris Water Maze show

Hippocampal lesion rat does not learn where platform is


NMDA antagonist swims like it has no hippocampus

What did Bear show when test LTP on human inferotemporal cortex removed during surgery

High Frequency Stimulations = LTP


LFS = LTD

What is the issue with titanic stimulation

Artificially high stimulation

Is there a physiological equivalent to tetanic stimulation?

Yes - hippocampus theta rhythms

When do theta waves become active

When animal is active - running, swimming, head movements and spatially orientated response


Arousal/alertness, fire during exploration etc

Depolarising stimulation of a neuron at a peak of a theta waves leads to what?

LTP

Depolarising stimulation of a neuron at a through of a theta waves leads to what?

LTD

Genetically increasing the amount of NMDA shows what in Morris Water Maze

Boosts LTP and significantly increased rate in learning


Spend more time in quadrant where platform is in probe trial

Aging shows what in the Morris Water Maze

Decreased acquisition in the Morris Water Maze (takes longer to learn)


Decreased LTP


Decreased expression of the NMDA receptor

How does enrichment effect the Morris Water Maze?

Enhanced acquisition


Increased LTP

One suggestion to how to reverse aging effect on LTP

Enrichment

Evidence of enrichment reversing effect on aging

Spatial maze task:


Old mice in impoverished environment show greater deficits than those in standard or enriched conditions. Can change via environment change.

Associative LTP entails what summation?

Spatial summation

What has been used to prove LTP underlies fear memories (not just correlational)

Optogenetics (shining lights on neurons that have been modified to respond to light)

How can you engineer a memory with LTD and LTP

Instead of using tone, use optogenetics, with shock and can elicit same response of fear/freezing.



Fire lots of light (high freq stim) = LTP, fire little light (LFS) = LTD

Increasing NMDA function in the forebrain leads to what?

Improved spatial memory

What do patients of panic disorder lack in the brain?

Sufficient inhibitory control in cortical and limbic regions to suppress inappropriate fear responses

GABA-A antagonist

Flumazenil

How do benzodiazepines work on GABA

Makes it more effective at opening the Cl- channel


Greater influx of chloride


Greater hyperpolarisation


More inhibition

What does benzodiazepines do at GABAa receptors with no GABA

Nothing (no opening of channel)

What does anxiolytic mean

Reduce anxiety

Are GABA agonist anxiolytic or anxiogenic

Anxiolytic

What is the problem with barbiturates

Activitate all GABA-A = shut down effect

Partial agonists have a better what

Therapeutic ratio

Where is there a high density of alpha 2 GABAa receptors

Limbic system


Hippocampus


Amygdala

2 animal model to study anxiety

Elevated + maze


Conditioned Emotional Response test

What is the effect of diazepam on the elevated + maze

Increases amount of time spent in the aversive open arms of the maze

What did using an agonist at alpha 2/3 subunit of GABAA receptors show for the elevated + maze?

Spent equivalent/possibly greater time in open arms of maze than general diazepam showing they are the ones influencing anxiety

What is the process of the conditioned emotional response

Train mice to press level for food


Learn 2 things=


Bell paired with mild footshock


Just a light (no consequence)


Change in level pressing ratio: how much pressing when bell played/pressing when light + pressing when bell

What is the effect of agonist at a2/a3 subunit containing GABAa receptors on the conditioned emotional response task

More agonist = less suppression (less change in pressing)


More anxiolytic

What is the effect of removing alpha 2 subunits (knockout) in mice for the conditioned emotional response task?

No difference in acquisition or lever pressing for food



Knockouts show greater supression (more anxiety?) than wildtypes

What is the effect of benzodiazepines on GABAa a2 subunit knockout mice in the conditioned emotional response task?

No effect (doesn't reduce supression)

Benzodiazepines are dependent on what part of the structure of an alpha subunit

Histidine

What is it called when you change the DNA sequence so that GABA-A a2 subunits have an argenine rather than a histidine

Knock in

What difference did a knock in mice for GABA-A alpha 1 show as a response to diazepam

Lost sedation effects and reduced Anticonvulsant effects

What difference did a knock in mice for GABA-A alpha 1 show as a response to diazepam

Lost sedation effects and reduced Anticonvulsant effects

What difference did a knock in mice for GABA-A alpha 2 show as a response to diazepam

No anxiolytic effect

What is the anxiety effect of a barbiturate in GABA-A subunit alpha 2 knock in mouse and what does this show

Still works - have the same anxiolytic effect


Show alpha 2 containing receptors still there just not effected by BZD

What difference did a knock in mice for GABA-A alpha 3 show as a response to diazepam

No difference

Want to find selective BZD agonist that acts specifically where?

A2 receptors

What is a syndrome that leads to little/no fear when both right and left medial temporal lobes of brain malfunction

Kluver-Bucy syndrome

What did SM case study have

Bilateral amygdala lesions

What did SM case study show

Fear response significantly reduced compared to normal

Where is the amygdala

Deep in brain of temporal lobes

What subjects show increased amygdala activation during public speaking vs control

Subjects with social anxiety disorder

What is there a high density of in the amygdala

Benzodiazepine binding sites

Injection of soluble BZD into amygdala induces what

Anxiolytic effect

After amygdala destruction, does BZD have an effect and what does this show

Yes, some, anxiety controlled by other brain areas as well

What 2 things does the amygdala excite?

Locus coeruleus + hypothalamus

What does the H stand for in HPA axis?

Hypothalamus releases CRH

What it does

What does the P stand for in HPA axis?

Pituitary releases ACTH

What it does

What does the P stand for in HPA axis?

Pituitary releases ACTH

What it does

What does the A stand for in HPA axis?

Adrenal cortex release cortisol (stress hormone) and adrenaline

In fear response, what does the locus coerulus release

Noradrenaline

What part of the brain provides info about contextual stimuli that are important for fear condition

Hippocampus

Lesions here have anxiolytic effdcg

Increasing GABA inhibitions in hippocampus leads to what

Decrease in contextual fear response (freezing less in context learnt)

What effect does noradrenaline have

Increases arousal and attention

What effect does benzodiazepines have on Locus coeruleus?

Decrease its release of Noradrenaline (increase GABAergic inhibition)

What balances the adrenaline (arousal)

Raphe nuclei (serotonergic projections) = behaviour inhibition, freezing

Freezing

What do benzodiazepines do to the serotonin activity

Decrease it in the raphe nuclei

Benzodiazepine effect on serotonin evidence

Increase in social interaction in rats following midazolan administered directly into the raphe nuclei

What disorders do benzodiazepines work well on

GAD


Panic D

What disorders do benzodiazepines not work well in

OCD


PTSD

What is a common treatment for all 4 anxiety disorders

SSRIs

What is the pattern of effect for SSRIs

Can be anxiogenic in short term (first few days)


Anxiolytic effect may not become apparent for weeks

What is another common treatment for GAD that takes 4-6 weeks to exert therapeutic potential

Buspirone (5-HT 1A receptor partial agonist)

Why do SSRIs take a few days to work?

Presynaptic receptor feedback to presynaptic cell to stop making serotonin as already some in synapse

Why does buspirone take a few weeks to have an effect?

Autoreceptors decrease 5-HT cell firing, changes in brain LT

Main role of acetylcholine

Attention/learning

What type of receptors do neuromodulators activate?

Metabotropic

What gated ion channels are ionotropic receptors

Ligand

Which type of acetylcholine receptors are ionotropic

Nicotinic

Which GABA receptor is Metabotropic?

GABA B

Term for transfering signals into cell

Transduce

Stages of activation of Metabotropic receptors in signal tranduction cascade

G protein


⬇️


Effector protein


⬇️


Second messenger


⬇️


Kinase


↙️ ↘️


Channelactivation Genetranscription

5/6

How was ionotropic receptors discovered/named

Pharmacology (effects of drugs)

How were dif G protein coupled receptors discovered?

Cloned by homology (screening dif DNA), work out which ligands were binding or which neurotransmitter were specific to activate each of those dif receptor subtypes

Neurotransmitter binding to the extracellular domain of a Metabotropic receptor causes what

Uncoupling of a heteromeric G protein on the intracellular surface (allowing them to move freely)


Signal is transduced across the cell membrane

What are the components of a G protein

Subunits alpha, beta and gamma

When a G protein is not active, what does it have attached to it?

GDP molecule

What replaces the GDP molecule when a ligand is bound to the receptor

GTP

What does the G protein split into

Alpha subunit + GTP


Beta and gamma subunits

What happens to the GTP attached just to the alpha subunit

Converts to GDP after some time, switching off the activity

3 types of alpha subunits

Gs stimulates adenylyl cyclase


Gi inhibits adenylyl cylase


Gq stimulates phosphate C

And what they do

What do beta gamma complexes activate

K+ channels directly

What does stimulating the Gs protein stimulate the synthesis of?

cAMP

(Second messenger)

What does cAMP activate

Protein Kinase A (PKA)

What is the opposite of protein kinase

Protein phosphatase (takes off phosphorylate group)

What does phosphorylating do?

Increases the effectiveness of the channel (allowing it to open more/easier)

Does one transmitter bound receptor uncouple one G protein

No, can uncouple multiple

2 names of types of presynaptic receptors

Autoreceptors


Heteroreceptors

How does the presynaptic dopamine receptor activation work

Phosphorylates tyrosine hydroxylase (increase dopamine)


Phosphorylates K+ channels (channels open, hyperpolarisation, further release of vesicles inhibited - promotes phasic release (big bursts))

Difference between autoreceptors and heteroreceptors

Auto- regulate own neurotransmitter release


Hetero - modulate release of another transmitter

Example of a heteroreceptor

Nicotinic receptors - ligand gated ion channel flux Ca2+ activation lead to release of dif transmitter (e.g. dopamine)

What 2 types of longterm synaptic changes can signal transduction/second messenger cascades lead to?

Structural and biochemical

What 2 ways do neuropeptides differ from other kibds of neurotransmitters

Methods of synthesis (cell body)


And release (global Ca2+)

What is the structure of neuropeptides?

Short polypeptide chains (6 to 36 amino acids)

Neuropeptides are produced by cleavage of what?

Propeptides synthesized directly from mRNA

The hippocampal Theta waves are controlled by what

Neuromodulatory acetylcholine release in the hippocampus

What is an antagonist of muscarinic acetylcholine receptors (metabotropic)

Scopolamine

What does scopolamine do?

Disrupt hippocampal waves


Disrupt learning and memory

What system is the first to degenerate in alzheimers disease?

Acetylcholine system

Opioid/opiate receptors agonists

Opiates e.g. morphine, heroin

Opioid/opiate receptors antagonist

Naloxone, naltrexone

Are opioid/opiate receptors ionotropic or Metabotropic?

Metabotropic

2 roles of the opioidergic system

Analgesia (inability to feel pain)


Relaxation (regulating noradrenaline release)

Dif between morphine and heroin

Heroin administered and get to brain in seconds


Morphine in minutes

How does opiates create the relaxing effect

Inhibiting the firing of noradrenergic neurons in the Locus Coeruleus

Opioid receptors are couple to what G?

Gi

You can work it out

Withdrawal leads to increased or decreased firing in LC?

Increase d

What is the balance of Gs and Gi coupled receptors during opiate withdrawal

Lack Gi, hypersensitivity of Gs

What is upregulated in tolerance to opiates

Gs pathway component (to match Gi)

Alongside the Gi pathways decreasing PKA and inhibiting the neuron, what does the G(beta gamma) subunit do?

Activate K+ channels = more inhibition

Cascade involved in opioid agonists (morphine)

Gi coupled opens K+ channel (hyperpolarises) and inhibits the adenylyl cylase = no cAMP = no PKA = no Na+ channel phosphorylation = LC cell inhibited


What are the compensatory changes in tolerance of morphine

Increased expression of adenylyl cyclase


Increased expression of PKA


(Decrease degradation of PKA)

4 components of withdraw process from morphine or Naloxone (mu antagonist)

Compensatory changes still active


No inhibition through mu receptors


cAMP pathways overactive


LC firing increases above normal levels

What are the negative symptoms of schizophrenia

Flattened emotional response, poverty of speech, lack of initiative and persistence, anhedonia and social withdrawal

5

What are the cognitive symptoms of schizophrenia

Low psychomotor speed


Problems in sustained attention


Deficits in learning and memory


Poor abstract thinking


Poor problem solving

A rare mutation in what gene has been linked to schizophrenia

DISC1

DISC1 increases the chance of schizophrenia by a factor of what

50

How does parental age relate to schizophrenia

Children of older fathers more likely to develop schizophrenia

Why are older fathers more likely to have schiz children

Most likely due to mutations in the spermatocytes (cells that produce sperm) as they divide much more than a woman's oocytes - more change for mutation

What % of DZ twins have schizophrenia

17%

What % of DZ twins have schizophrenia

48%

What are the 2 types of MZ twins

Monochronionic (same placentas)


Dichrionic (dif placentas)

What is the difference in concordance rates for schizophrenia in dichorionic and monochorionic twins?

11%-M


60%-D

3 neurodevelopmental theories to schizophrenia

The early neurodevelopmental model


The late neurodevelopmental model


Two-hit model

3 early evidence for schizophrenia suggesting abnormal brain development

Home movies from families with a schizophrenic child (more -ve affect, more abnormal movements)


Danish children videod while eating lunch, blind raters found schiz displayed less sociability and deficient psychomotor functioning


Minor physical abnormalities in ppl with schizophrenia

What is the late neurodevelopmental model of schizophrenia

Schizophrenia may result from abnormality in adolescence when synaptic pruning takes place

What is the pattern of developmental pruning in schizophrenic people

Much greater in adolescence

Do men or women exhibit schizophrenia later?

Women

3 structural changes in brain due to schizophrenia

Ventricular enlargement


Reduced brain volume (less grey matter) in temporal and frontal lobes and hippocampus


Faulty cellular arrangement in the cortex and hippocampus

How much bigger is the ventricles in schizo patients

More than 2x

7 neurocognitive deficits in schiz patients

Lower IQ


Planning and info processing deficits


Attentional deficits


Working memory deficits


Sensory-motor gating deficits


Antisaccade task (look away from attracting stimulus)


Occolomotor function (eye tracking)

Weinberger suggested the negative symptoms of schizophrenia are caused primarily by...

Hypofrontality, decreased activity of the frontal lobes, the dIPFC in particular

What task shows attentional deficits in schizophrenic patients

Stroop test

What task shows working memory deficits in schizophrenic people

Wisconsin card sort test

What does the Wisconsin card sort test show neurobiologically in schizophrenic people

Lack of increase in regional blood flow to the dIPFC (the normal response) = hypofrontality

In sensory motor gating deficits, what signal does not diminish when 2 stimuli (clicks) is presented

P50

In sensory motor gating deficits not learning to inhibit a startle response shows what

Pre-pulse inhibition (deficit)

What is the oculomotor function in schizophrenic people

Eye movement not smooth, catchup saccades

4 neurochemical hypotheses of schizophrenia

The dopamine hypothesis


The glutamate hypothesis


Neuroinflammatory hypothesis


The estrogen hypothesis

What is the prevalent theory of schizophrenia

Dopamine

Where is there overactivity of dopamine in the dopamine hypothesis of schizophrenia

Mesolimbic system

What does overactivity of dopamine in the mesolimbic system result in

+ve symptoms

Where is there underactivity of dopamine in the dopamine hypothesis of schizophrenia

Mesocortical system

What does interactivity of dopamine in the mesolimbic system lead to?

Negative and cognitive symptoms of schizophrenia

What do DA agonists do

Produce symptoms that resemble the +ve symptoms of schizophrenia

4 types of DA agonists

Amphetamine


Cocaine


Methylphenidate


L-DOPA

What was the first DA antagonist

Chlorpromazine (CPZ)

For schizo (typical)

What do DA antagonists do

Block D2 receptors

(Chlorpromazine)

What does affinity mean (drug talk)

How well it binds to receptor

What did a radioactive tracer of IBZM show for schizophrenic people

More displacement of IBZM, showing more DA activity in striatum correlated with positive symptoms

What are the consequences of LT drug treatment of schizophrenia

Symptoms relating to Parkinson's: slowness in movement, lack of facial expression and general weakness.


In around 1/3rd patients = tardive dyskinesia (unable to stop moving, usually around face: smacking of lips or protusion of the tongue)

How do newer, atypical antipsychotic drugs work

Have a lower affinity for the D2 receptor (so no extra-pyramidal side effects)

What do atypical antipsychotic drugs help with (for schizophrenia)

Improve positive and negative symptoms of schizophrenia.


Improve the performance in neuropsychological tests.

What was the first atypical antipsychotics (the king)

Clozapine

3 issues with the dopamine system

It explains only a part of schizophrenia (+ve symptoms)


Atypical drugs work better


Under activity of dopamine in mesocortical pathway, not over, causes -ve symtpoms

Which of the receptors are linked to schizophrenia in the glutamate hypothesis of schizophrenia?

NMDA

Mutations in what two gene codes for NMDA receptors could cause schizophrenia

GRIN1, GRIN2A

Is it the glutamate hyperfrontality or hypofrontality hypothesis?

Hypofrontality

What could the NMDA receptor hypofrontality explain?

Why there are so many treatment resistant negative symptoms


Why the onset is in early adulthood and,


Why the disorder is associated with structural changes and cognitive deficits

3 things

What 2 drugs can cause all symptoms of schizophrenia

PCP and Ketamine

What antagonists and PCP and Ket

NMDA

What did a monkey administered with repeated PCP show

Decrease in activity in PFC, showed deficit in task where had to reach around a barrier to get a piece of food (same as those with lesion to PFC)

What is the explanation of positive symptoms from the glutamate hypothesis?

Glutamate stimulates GABA


GABA hypofrontality so does not inhibit Glutamate


Disinhibited glutamate stimulates overproduction of dopamine

What is the explanation of the negative and cognitive symptoms of schizophrenia from the glutamate hypothesis

Glutamate hypofrontality NMDA synapse


GABA not able to inhibit


Disinhibted glutamate


GABA over excited in VTA


Overinhibiting (silence) dopamine signal


Reducing the dopamine production in PFC = -ve and cog symptoms

What is the Neuroinflammatory hypothesis of schizophrenia

Hyperactivation of microglia (brain's immune system)

Evidence for the Neuroinflammatory hypothesis of schizophrenia

PET imaging shows increase in microglial activity as severity of illness increases


Symptoms reversed with antipsychs/antibiotics that reduce microglial activation


Chromosome linked with schizophrenia includes gene region involved in acquiring immunity

There are gender differences in schizophrenia in regards to:

Rate and onset of disease (later in women)


Severity and progression (less severe)


Response to antipsych treatment (better)

3 things

What % of the population have depression

5%

Different in rates of depression in males and females

3x more likely in women than men

What % heritability is depression?

37%

2 hypotheses to the cause of depression

The monoamine hypothesis


The stress-diathesis Hypothesis (Neurogenic Hypothesis)

What is the prevalent theory for depression cause

The monoamine hypothesis

4 types of monoamines

Dopamine


Serotonin


Norepinephrine (noradrenaline)


Epinephrine (adrenaline)

What breaks down monoamine in the synapse

Monoamine Oxidase (MAO)


Catechol-O-methyltransferase (COMT)

Early evidence pointing towards chemical inbalance

Lower levels of the 5-HT, dopamine or norepinephrine metabolite (bit left over when monoamine broken down) in the CSF of depressed individuals

Evidence from reserpine for the monoamine hypothesis for depression

Used to treat blood pressure


Caused dep as a side effect


Blocks packaging of monoamines into the vesicles, so when the neurons are activated no neurotransmitter is released


Rabbits

All effective antidepressants affect what system

5HT/NA

What drugs created linked to monoamine hypothesis for depression

Monoamine oxidase inhibitors (MAOIs)

What side effect does MAOIs lead to

Increased symathetic system stimulation and the cheese effect - cheese type products contain tyramine broken down by MAO in liver, not broken down = stimulation of sympathetic system

3 other antidepressants besides monoamine oxidase inhibitors

Tricyclic antidepressants


SSRIs (prozac)


SNRIs serotonin- norepinephrine reuptake inhibitors

What do tricyclic antidepressants do

Inhibit the reuptake or serotonin and norepinephrine

What do monoamine oxidase inhibitors do

Inhibit the breakdown of monoamines in the presynaptic terminal so they increase the level of monoamines taken up into the vesicles

2 issues with the monoamine hypothesis of depression

Decrease in serotonergic activity in healthy people doesn't induce depression, only those w/ history. (Cocktail depleted of tryptophan)



Time-course, SSRIs and SNRIs increase levels of 5HT or NE rapidly, but effect takes several weeks

What shrinks with the duration of untreated depression

Hippocampal volume

What is the negative feedback look in HPA

Amydgala encourages HPA to create cortisol


Cortisol detected in hippocampus, inhibiting the HPA to create cortisol

What does chronic stress lead to

Chronic activation of glucocorticoid receptors = increased Ca2+ into neurons, too much = excitotoxic = cell die


Hippocampus can't feedback to limit cortisol production

Hippocampus

Evidence for the dysfunctional stress system in depression

Artificial cortisol in normal controls activates GR and the negative feedback system, leading to decreased ACTH and cortisol


In depressed, they are less sensitive to it (poor -ve feeback), more sensitive to ACTH

What is the stress-diathesis explanation of why antidepressants take 2-4 weeks to have an effect

Takes a while for "newborn" cells to grow

Injections of what in the rat brain reduces neuronal death and protects neurons that have been treated with neural toxins

BDNF

Chronic stress does what to the levels of BDNF

Decreases them

What has been used to explain susceptibility to depression

Serotonin transport

Which serotonin transporter has a big interaction with stressful life events and depression

Short/short (s/s) alleles from parents

2 other benefits of having long alleles for serotonin transporters

More likely to respond to treatment


Much better LT outcomes


Tryptophan deletion less likely to produce symptoms

What is a novel antidepressant?

Ketamine

What antagonists is Ketamine

NMDA

2 alternative therapies to antidepressants

Transcranial Magnetic Stimulation (TMS)


Electroconvulsive Therapy (ECT)

Difference between pharmacokinetics and pharmacodynamics

K = drug movement, entering our body, what our body does to the drug


D = drug change, what the drug does to us, when it binds, how it effects us

4 categories of criteria of substance use disorders

Impaired control


Social impairment


Risky use


Pharmacological - tolerance and withdrawal

What is the severity cut offs for substance use disorders

Mild = 2-3 symptoms


Moderate = 4-5


Severe = 6+

Do you need tolerance and/or withdrawal for addiction

No

Which brain system is involed in addiction (structures)

The mesocorticolimbic dopamine system

What is the projections involved in the mesocortolimbic dopamine system

Dopamine neurons projecting from the VTA to nucleus accumbens and prefrontal cortex

What is the role of the mesocorticolimbic dopamine system

Critical pathway for rewards and reinforcement

Natural reinforcers (e.g. food and sex) increase what in where?

Dopamine in the nucleus accumbens

What is the changed set point hypothesis for addiction

Dopamine in nucleus accumbens have been used for little pleasure, but suddenly used to much more

Direct agonists increasing dopamine availability/release in Nucleus accumbens

Cocaine


Amphetamine


Nicotine

How do cocaine/amphetamine work

Increase availability of dopamine at synapse by blocking or reversing dopamine transporters

How does nicotine work

Direct excitation of VTA neurons by action at nicotinic receptors (leading to dopamine increase)

Indirect agonists increasing dopamine availability/release in NAc

Opioids


Cannabinoids

How do opioids/cannabinoids work

Action at opioid or cannabinoid receptors indirectly leads to modulation of VTA activity


E.g. inhibition of GABAergic projections onto VTA neurons allows them to fire resulting in increases of DA release

What explains wanting a cigarette when you go into a pub

Associative learning


Coincident firing in NAc induces LTP, strengthening sensory inputs synaptic connections

What does the dopamine cascade lead to

Synaptic re-modelling - increased spines and dendritic branches - enhaced LTP

Hypoactivity in what brain areas may underlie inability to control in addiction

Frontal systems

What did the self administration model show for rats

Damage to nucleus accumbens decreases self-administration of heroin

Does cocain or amphetamine reverse the transporter of DA

Amphetamine

What else does cocaine and amphetamine effect (altho dopamine is primary)

5HT NA

How do opiates cascades work

Act at endogenous opioid receptors (Gi/Go coupled)


Inhibitory


Decrease adenylyl cyclase activity


Lead to open K+ channels and close Na+ channels

How does GABA inhibition have an effect/on dopamine (2 ways)

Disinhibits the DA neurons in VTA


Action at opiate receptors in the NAc - independent of DA release

What is alcohol an agonist of

GABAa (inhibitory)

What is alcohol an antagonist of

NMDA

How does alcohol have an effect on dopamine in NAc

Suppression of cortical output


No GABA activation in PFC


Disinhibited DA neurons in VTA


Rewarding n reinforcing in NAc

How is alcohol involved in the opiate system

Naltrexone reduces EtOH (alc) self administration in animals


Used as a treatmend to reduce EtOH consumption, relapse and craving

Explain the nicotine receptors

Nicotininc acetylcholine receptors


Ligand gated ion channels pre and post synaptically


Excitatory

How does nicotine effect dopamine

Activation of nACh receptors on cell body in the VTA (increasing firing)


Facilitation of DA release by pre-synaptic receptors in NAc

What can block nicotine induced behaviour and self administration

Opiate and DA antagonists

What is the prevalent model for addiction

Impaired response inbibition and salience attribution model of addiction

What is the impaired response inhibition and salience attribution model of addiction

Weakened top down inhibitory control functions and strengthened bottom up functions are features of the addicted brain

What is the heredity of addiction

40-60%

What is the heredity of addiction

40-60%

When does addiction usually begin

Adolescence

3 key subtypes of behavioural impulsivity in addiction

Inability to inhibit actions


Inability to delay gratification


Inability to reflect before making a decision

2 measurements of impulsivity

Self-report measures (questionnaires)


The Matching Familiar Figures Test

The matching familiar figures test shows there are 2 types of impulsivity levels in kids called:

Impulsives (<10 secs)


Reflectives (30-40 secs)

What does impulsivity lead to in treatment

Quicker and more drop out

Evidence for impulsivity Predisposing later substance abuse

Field et al impulsivity in adolescents predicted alc involvement 6m later


Converse relationship wasn't observed (alc =/ impulse)

What system is involved in movement

Somatosensory

What are the 2 types of skin humans have

Hairy skin


Glabrous skin

What test reveals differences in skin sensitivity across the body

2 point discrimination test

What parts of our body are the most sensitive

Fingers (most)


Lips


Toes

3 main types of somatosensory perception (names)

Nocioception


Hapsis


Proprioception

What is nocioception perceiving

Pain and temp

What is hapsis perceiving

Pressure and fine touch

What is proprioception perceiving

Movement and location of the body

What is the structure of nocioceptors

Free nerve endings


Damage to dendrite or surrounding cells release chemical that stimulate dendrite and produces an AP

What is the similarity between the different haptic receptors

Mechanical stimulation produces AP

What are the 4 layers of haptic receptors in order from surface to deep

Merkel


Meissner


Ruffini


Pacinian

3 key proprioceptors

Muscle spindles


Gogli tendon organs


Joint receptors

What is a rapidly adapting receptor (meaning)

Receptor that responds briefly to the beginning and end of a stimulus

What is a slowly adapting receptor (meaning)

Body sensory receptor that responds as long as a sensory stimulus is on the body

Does meissner's corpuscle have slow or fast adaption

Fast

Does merkel's disc have slow or fast adaptation

Slow

Is pacinian corpuscle rapid or slow adaptation

Rapid

Does Ruffini have rapid or slow adaptation

Slow

What neurons do the somatosensory info go through

Dorsal-root ganglion neurons

How is the structure of a dorsal-root ganglion cell different

Dendrite and axon are continuous

Which somatosensory neuron/s is/are myelinated

Proprioceptive and Haptic Neurons

What are the 2 key somatosensory pathways to the brain

Dorsal spinothalamic tract


Ventral spinothalamic tract

What info does the dorsal spinothalamic tract carry

Haptic and proprioceptive info

Does the dorsal or ventral spinothalamic tract cross immediately in the spinal cord

Ventral

Damage in left side of spinal cord would cause what in each leg

Left leg- loss of sensation of touch and movement


Right leg - loss of pain and temp

What shows a break in the spinothalamic pathways

Monosynaptic reflex e.g. knee jerk

What system in involved in balance

Vestibular

What does the vestibular organ contain

3 semicircular canals


Otolith organs (utricle and saccule)

The semicircular canals in the ear respond to what movement?

Angular acceleration (spinning on a chair)

Uccicle and saccule respond to what movement

Linear acceleration (moving head back and forth)

What does the primary somatosensory cortex do

Receive projections from thalamus


Begin constriction of perception

What does the secondary somatosensory cortex do

Continue the construction of perceptions


Project to the frontal cortex

Who was the first to discover and map out the somatosensory homunculus

Penfield

Who refined the somatosensory homunculus into 4 seperate parts

Kaas

What were the 4 separate somatosensory homunculi Kaas found

Area 1: skin (fast)


Area 2: joints, pressure


Area 3a: muscles


Area 3b: skin (slow)

What was the hierarchical organisation of Kaas' somatosensory homunculus

3a


3b


1


2

What is the components of the hierarchical control of movement

Neocortex (conscious)


Brain stem and spinal cord (automatic)

What did Karl Lashley add to the theory of initiation of movement

Preprogrammed motor sequences

Role of prefrontal cortex in movement

Planning and specifying the goal

Role of premotor cortex

Organises motor sequences (select movements appropriate to the context)

Evidence of the premotor cortex involvement

Monkey study, damage to this cortex shows cannot put motor sequences together

Primary motor cortex role

Produce specific skilled movements

Evidence for the frontal lobe involvement in movement

fMRI shows cerebral fluid blood flow in each area for specific tasks


Push a lever = motor


Perform sequence of movements = motor and premotor


Find route through maze = motor, premotor and prefrontal

Who was the first to map out the motor cortex

Fitsch and Hitzig

Dogs

What is the map called that Penfield created

Homunculus

Whag did Graziano show for the motor coetex

Used precise stimulation and found recognizable actions


Motor cortex represents 3 types of organisation:


The part of the body that is to be moved, the spatial location to which the movement is directed, and the movements function

Who found the area of motor cortex retains size with rehabilitation/ decreases without if damaged

Nudo and colleagues 1996

How does info from the motor cortex get to parts of body

Via corticospinal/pyramidal tracts

What main tract?

What are the 2 corticospinal tracts called

Lateral


Ventral

What does the lateral corticospinal tract do

Moves the limbs and digits on the opposite side of body, crosses at brain stem

What does the ventral corticospinal tract do

Moves the muscles of the midline (trunk) on the same side of the body

What are the 2 neurons inbthe spinal columns ventral horm

Interneurons


Motor neurons

How are the motor neurons arranged

Laterally located motor neurons = fingers and hand muscles


Intermediate = arms and shoulder muscles


Medially = trunk

What are the pairs that limb muscles are arranged in?

Extensor and flexor

Does flexor muscles move towards or away from the trunk

Towards

What a good extensor example

Triceps

Whats it called if you have paralysis of the legs and arms

Quadriplegia

What did a monkey flexing with weights show

Flexor motor neurons fire more

What did live imagine of motor cortex using calcium imaging show

A subset of neurons or a "neuronal ensemble" is activated during behaviour (e.g. running)

What is the part of the brain responsible for allowing us to adjust the force of our movements

Basal ganglia

What is the basal ganglia

Collection of subcortical nuclei within the forebrain


Receives input from all areas of the neocortex and limbic cortex


Projects back to the motor cortex

What is the hypothesis called about the basal ganglia

Volume hypothesis

What acts like a volume dial and projectsbto the motor cortex in the volume hypothesis

The internal globus pallidus

What are the 2 pathways within the basal ganglia

Direct and indirect

Names

Which neurons do the direct pathway express

D1R (domapine 1 receptor)

What does the direct pathway within the basal ganglia do

Inhibitory effect of the globus pallidus internal (disinhibition): too much activity leads to overactivity in thalamus and amplified force of movement

What does the indirect pathway within the basal ganglia do

Excitatory effect on globus pallidus internal leading to underactivity in the thalamus and reduced force of movement

Damage to the basal ganglia cab produce what 2 main types of symptoms

Hyperkinetic and hypokinetic symptoms

What does hyperkinetic symptoms show and where is it seen

Excessive involuntary movement


Huntington's chorea


Hemiballism

What does hypokinetic symptoms of basal ganglia damage show and where is it seen

Paucity of movement


Parkinson's disease

How does Huntingtons affect the volume hypothesis

DR2 neurons degenerate


Disinhibitted indirect pathway


Thalamus overactive

What does hemiballism show in the volume hypothesis

Stroke in the subthalamic nucleus


Indirect pathway not working properly so doesn't excite the globus pallidus internal to inhibit the thalamus


Involuntary fast movements

What is used to relieve the symptoms of huntingtons and hemiballism

Antipsycha clozapine dopamine blocker

What is used as a treatment to parkinsons

L Dopa

How does parkinsons affect the volume hypothesis

Underactivity of in the direct pathway and overactivity in the indirect pathway


More inhinition


Turning down of motor activity

What did a study using parkinsonian mice show

Optogenetic activation if D1R expressing striatal neurons improved symptoms

How does the cerebellum affect movement

Acquiring a movememnt skill

What is the flocculus involved in in movement

Eye movement and balance

What is the lateral parts of the hemispheres of the cerebellum involved in in movement

Controls movement of limbs hands feet and digits

What are the medial parts of the hemispheres of the cerebellum involved in in movement

Face and midline of mody

3 main motor functions of the cerebellum

Timing of movements


Maintaining movement accuracy


Motor associative learning

Damage to the cerebellum leads to problems in what 3 tasks

Tapping along to a metronome


Throwing darts using prison goggles


Eyeblinks conditioning response after airpuffs