Bayne's Biochemistry Chapter 39 - Neurotransmitters

Choose a study mode

Play Quiz
Study Flashcards
Spaced Repetition
Chat to Lesson

Podcast

Play an AI-generated podcast conversation about this lesson
Download our mobile app to listen on the go
Get App

Questions and Answers

The blood-brain barrier's (BBB) selectivity is most accurately described by which statement?

  • The BBB is absolute, preventing any molecule synthesized outside the brain from entering the CSF.
  • The BBB restricts all molecules larger than 500 Da, ensuring complete protection of the brain.
  • The BBB allows passage based solely on charge, favoring positively charged molecules.
  • The BBB's permeability is relative and depends on the size of the molecule in question. (correct)

In the context of neurology, the differing myelin composition in the CNS and PNS is most significantly relevant to:

  • Differential diagnosis of demyelinating diseases. (correct)
  • Regulating the synthesis of CSF protein pool within the brain.
  • Determining the structural integrity of the dura mater.
  • Facilitating faster nerve conduction velocities in the PNS compared to the CNS.

The presence of asialotransferrin in CSF, but its rapid removal from systemic circulation, is due to what?

  • The brain lacks reticuloendothelial cells along the path of CSF flow. (correct)
  • The blood-brain barrier actively degrades asialotransferrin.
  • Asialotransferrin is exclusively produced in the liver and cannot cross the BBB.
  • Systemic circulation contains higher concentrations of sialic acid.

Which statement best describes the role of GLUT1 and GLUT3 transporters in brain glucose metabolism?

<p>GLUT1 is insulin-independent and found on endothelial cells and astroglial endfeet, while GLUT3, also insulin-independent, is expressed in neurons. (C)</p>
Signup and view all the answers

Which of the following is the primary mechanism by which neurons ensure the efficient transport of materials over their long axonal lengths?

<p>Anterograde and retrograde transport using kinesin and dynein motor proteins, respectively. (B)</p>
Signup and view all the answers

What is the significance of N-acetyl-L-aspartate (NAA) in the context of neuronal metabolism and brain imaging?

<p>NAA is a marker of the metabolic competency of brain neurons and can be quantified by MRI. (B)</p>
Signup and view all the answers

How do astrocytes facilitate neuronal energy requirements and neurotransmitter function?

<p>By providing lactate as a complementary energy source and neurotransmitter precursors like glutamine to neurons. (C)</p>
Signup and view all the answers

What is the role of clathrin in synaptic transmission?

<p>Clathrin facilitates the invagination of the nerve terminal membrane to recycle membrane after vesicle fusion. (D)</p>
Signup and view all the answers

How does caffeine affect neurotransmission in the brain?

<p>It inhibits phosphodiesterase, mimicking the effects of adrenergic neurotransmission by preventing cAMP breakdown. (A)</p>
Signup and view all the answers

What is the primary mechanism by which atropine affects cholinergic neurotransmission?

<p>It blocks muscarinic receptors. (B)</p>
Signup and view all the answers

Which of the following best describes the role and mechanism of action of botulinum toxin in therapeutic applications?

<p>It hydrolyzes presynaptic proteins involved in neurotransmitter release, reducing muscle contraction. (B)</p>
Signup and view all the answers

What is a key distinction between Lambert-Eaton syndrome and myasthenia gravis related to their effects on synaptic transmission?

<p>Lambert-Eaton syndrome involves a presynaptic block, while myasthenia gravis involves a postsynaptic block of neurotransmitter action. (A)</p>
Signup and view all the answers

What is the significance of the presence of oligoclonal bands in CSF but not in serum when diagnosing neurological disorders?

<p>It suggests local synthesis of immunoglobulins within the brain, indicative of intrathecal immune activity. (B)</p>
Signup and view all the answers

In the context of long-term potentiation (LTP), what is the role of glutamate?

<p>Glutamate mediates excitatory transmission and plays a role in synaptic plasticity by activating different classes of receptors. (A)</p>
Signup and view all the answers

In Alzheimer's disease, how do amyloid-ẞ (Aβ) oligomers affect cholinergic neurons and cognitive function?

<p>Aβ oligomers preferentially impair cholinergic neurons in the brain septum, causing progressive loss of cognitive function. (A)</p>
Signup and view all the answers

What is the role of the serotonin transporter (SERT) in neuronal function, and how do SSRIs impact this process?

<p>SERT transports serotonin from the synaptic cleft back into the presynaptic neuron, and SSRIs block this process, increasing serotonin levels in the synaptic cleft. (A)</p>
Signup and view all the answers

How do neurons respond to increased fatty acid concentrations during food deprivation?

<p>Neurons sense increased fatty acid concentrations as a signal to initiate eating behavior. (D)</p>
Signup and view all the answers

Why is distinguishing CSF rhinorrhea from local nasal secretions important and how is it typically done?

<p>Because CSF rhinorrhea suggests a dural tear and risk of meningitis, often confirmed by testing for asialotransferrin. (A)</p>
Signup and view all the answers

What is the primary mechanism by which GABA exerts its inhibitory effect on postsynaptic neurons?

<p>By binding to specific GABA receptors, causing an influx of chloride ions and hyperpolarization. (B)</p>
Signup and view all the answers

In vision, what is the order of events after a photon is detected by rhodopsin in rod cells?

<p>Activation of rhodopsin → decrease in cGMP levels → sodium channels close → cell hyperpolarizes. (D)</p>
Signup and view all the answers

The dura mater serves as a significant dividing line between which two nervous system components?

<p>The central and peripheral nervous systems. (B)</p>
Signup and view all the answers

Which cells produce myelin in the CNS and PNS respectively?

<p>Oligodendrocytes in the CNS, Schwann cells in the PNS (D)</p>
Signup and view all the answers

Approximately what percentage of the cerebrospinal fluid (CSF) protein pool is synthesized within the brain itself?

<p>15% (D)</p>
Signup and view all the answers

Which of the following describes the role of the brain parenchyma in the context of CSF production?

<p>It contributes brain-specific proteins, such as transthyretin, to the CSF. (D)</p>
Signup and view all the answers

What is the consequence of a thiamine deficiency, and how does it impact neuronal function?

<p>Inhibition of pyruvate dehydrogenase, leading to decreased acetyl-CoA synthesis and impaired energy production. (C)</p>
Signup and view all the answers

In Alzheimer's Disease (AD), what role do biomarkers such as Aβ42 and tau protein play in the central nervous system (CNS)?

<p>Aβ42 levels reflect amyloid deposition, while tau levels correlate with neurodegeneration, assisting in disease diagnosis even before severe cognitive impairment. (D)</p>
Signup and view all the answers

How do the locations of neuronal cell bodies and synapses relate to the need for an efficient axonal transport system?

<p>Distances which can be up to a meter require a transport system. (D)</p>
Signup and view all the answers

How is the action of catecholamines terminated?

<p>Reuptake and degradation to aldehydes by mitochondrial monoamine oxidases (A)</p>
Signup and view all the answers

What are the functional outcomes of light striking rhodopsin in a rod cell?

<p>Decreased level of cGMP and cell hyperpolarization (C)</p>
Signup and view all the answers

Upon the detection of a single photon of light by the human eye, transducing it into an electrical signal to be interpreted by the nervous system culminates in:

<p>An action potential being generated within a retinal ganglion neuron, subsequently propagating along the optic nerve. (A)</p>
Signup and view all the answers

Given the role of gut microbiome modulation via probiotic therapy in the context of irritable bowel syndrome (IBS) and considering the gut-brain axis, how does the modulation impact neural pathways?

<p>Influencing CNS pathways and cognitive symptoms, besides improving gut-related symptoms and psychological stress, via the vagus nerve. (D)</p>
Signup and view all the answers

The clinical presentation of an elderly patient exhibiting significant memory loss combined with visuospatial deficits and language impairments led to a preliminary diagnosis of neurodegenerative disease. To differentiate between various forms of dementia, including Alzheimer's disease and vascular dementia, which set of CSF biomarkers should be ordered for diagnostic clarification?

<p>Amyloid β42 (Aβ42), total tau, and phosphorylated tau levels for Alzheimer's (D)</p>
Signup and view all the answers

What role do G-protein coupled receptors of glutamate play in synaptic neurotransmission?

<p>They activate downstream signaling cascades (B)</p>
Signup and view all the answers

Which of the following statements is most accurate concerning the role of myelin in the nervous system?

<p>Myelin insulates axons, facilitating nerve transmission, and is generated by oligodendrocytes in the CNS and Schwann cells in the PNS. (D)</p>
Signup and view all the answers

A researcher is studying the long-term potentiation (LTP) in a neuronal circuit. Which signaling molecule would be most crucial to investigate due to its effects in inducing LTP?

<p>Glutamate (A)</p>
Signup and view all the answers

What distinguishes the myelin produced by oligodendrocytes in the CNS from that produced by Schwann cells in the PNS?

<p>The lipid composition and protein components differ due to their synthesis by different cell types. (B)</p>
Signup and view all the answers

Considering the role of the blood-brain barrier (BBB) in maintaining central nervous system (CNS) homeostasis, what is the most critical implication of its selective permeability?

<p>It necessitates specific transport mechanisms for essential nutrients and signaling molecules, regulating the CNS microenvironment. (A)</p>
Signup and view all the answers

Under conditions of prolonged hypoglycemia, how does the brain maintain its energy supply, and what are the limitations of this adaptation?

<p>Ketone bodies, specifically β-hydroxybutyrate, are utilized as an alternative fuel source, although they cannot entirely replace the brain's glucose requirements. (A)</p>
Signup and view all the answers

How does the unique structural arrangement of brain capillaries and the presence of tight junctions contribute to the function of the blood-brain barrier (BBB)?

<p>It restricts paracellular transport, necessitating transcellular transport mechanisms that selectively control substance entry into the CNS. (C)</p>
Signup and view all the answers

In the context of neurotransmission, what is the primary role of axonal transport, and how do disruptions in this process lead to neurodegenerative diseases?

<p>It is essential for the bidirectional movement of organelles, proteins, and signaling molecules necessary for neuronal function and survival; disruptions can cause accumulation of toxic proteins and energy deficits. (A)</p>
Signup and view all the answers

Considering the role of astrocytes in neuronal metabolism, how does the astrocyte-neuron lactate shuttle contribute to brain energy metabolism, and what are the implications of its dysfunction?

<p>Astrocytes take up glucose and convert it to lactate, which is then released to neurons as an alternative energy substrate, particularly during high neuronal activity. (B)</p>
Signup and view all the answers

What is the significance of N-acetyl-L-aspartate (NAA) as a biomarker in brain imaging, and how does its concentration reflect neuronal health and metabolic competency?

<p>NAA is predominantly found in neurons and is synthesized from mitochondrial acetyl-CoA; its level reflects the metabolic competency and integrity of neurons. (C)</p>
Signup and view all the answers

How does the process of long-term potentiation (LTP) contribute to synaptic plasticity, and what are the critical molecular mechanisms involved?

<p>It is characterized by an increase in synaptic strength following high-frequency stimulation, mediated by changes in glutamate receptor expression and postsynaptic signaling pathways. (C)</p>
Signup and view all the answers

What are the key mechanisms by which microglia contribute to both neuroprotection and neurotoxicity in the central nervous system (CNS)?

<p>Microglia can exert neuroprotective effects through phagocytosis of debris and secretion of growth factors, but also induce neurotoxicity by releasing inflammatory cytokines and reactive oxygen species. (C)</p>
Signup and view all the answers

How do the distinct types of glutamate receptors (ionotropic and metabotropic) contribute to the diverse roles of glutamate in synaptic transmission and neural plasticity?

<p>Ionotropic receptors produce fast excitatory postsynaptic potentials (EPSPs) through direct ion channel activation, while metabotropic receptors modulate synaptic transmission through G-protein-coupled signaling pathways. (D)</p>
Signup and view all the answers

What are the primary mechanisms by which neurons maintain the ionic gradients necessary for resting membrane potential, and how do disturbances in these gradients lead to neurological disorders?

<p>Active transport via ion channels continuously pumps sodium out and potassium in, and mutations in sodium channels can cause disorders like hyperkalemic periodic paralysis. (A)</p>
Signup and view all the answers

How does the mechanism by which botulinum toxin inhibits neurotransmitter release differ from that of Lambert-Eaton syndrome, and what implications does this have for their clinical presentations?

<p>Botulinum toxin hydrolyzes presynaptic proteins, inhibiting neurotransmitter release, while Lambert-Eaton syndrome involves antibodies blocking calcium channels necessary for neurotransmitter release. (D)</p>
Signup and view all the answers

How do Wernicke-Korsakoff encephalopathy and Alzheimer's disease affect cholinergic neurotransmission, and what are the specific mechanisms underlying these effects?

<p>Alzheimer's disease causes damage through lesions and amyloid plaques/tangles, ultimately impairing acetylcholine production. (B)</p>
Signup and view all the answers

Considering the relationship between the gut microbiome and the brain, how might alterations in gut microbiota influence neurotransmission and behavior, and what is the role of the vagus nerve in this process?

<p>The vagus nerve carries neural signals, treatment via probiotic bacteria can affect neurotransmission in the brain. (C)</p>
Signup and view all the answers

How do the cellular mechanisms underlying the detection of a single photon of light by rhodopsin in rod cells ultimately result in an action potential in the optic nerve?

<p>Rhodopsin ultimately catalyzes hydrolysis of cGMP, sodium closes and causes hyperpolarization of the rod cell and ultimately an action potential in the optic nerve. (C)</p>
Signup and view all the answers

How does the drug edrophonium improve muscle strength in myasthenia gravis, and what is the underlying mechanism of its action on cholinergic neurotransmission?

<p>Edrophonium also causes the death of the autoantibodies. (C)</p>
Signup and view all the answers

How does caffeine affect neurotransmission in the brain, and what is its primary mechanism of action at the molecular level?

<p>An impulse can also be regulated at the level of the second messenger, such as cAMP, which is broken down by the enzyme phosphodiesterase. Phosphodiesterase is inhibited by caffeine and other methylxanthines, thereby mimicking many of the effects of adrenergic neurotransmission. (A)</p>
Signup and view all the answers

How does serotonin transporter (SERT) dysfunction contribute to the pathophysiology of autism spectrum disorder (ASD), and what are the specific mechanisms involved?

<p>Mutations of the serotonin transporter are implicated in autism, attention deficit hyperactivity disorder, and parkinsonism. (B)</p>
Signup and view all the answers

Given that neurons do not divide postpartum, what implications does this have for neuronal recovery following injury, and how does it influence the development of neurodegenerative diseases?

<p>Adult neurons cannot divide following injury, contributing to neurodegeneration. (B)</p>
Signup and view all the answers

How does the differential diagnosis of CSF rhinorrhea from common nasal secretions impact treatment strategies, and what are the key diagnostic markers used to distinguish between these fluids?

<p>ENT surgeons do not have to test for this. (A)</p>
Signup and view all the answers

Following the detection of a single photon of light by the human eye, the process of transducing it into an electrical signal involves:

<p>Direct conversion of the emitted light to an electrical form. (A)</p>
Signup and view all the answers

If you wanted to test for a CSF leak via Rhinorrhea, which clinical marker is most useful?

<p>Asialotransferrin (D)</p>
Signup and view all the answers

Which factor has the MOST influence on the permeability of the blood-brain barrier?

<p>Molecular Size (D)</p>
Signup and view all the answers

Guillain-Barre syndrome typically affects myelin in which part of the nervous system?

<p>PNS (A)</p>
Signup and view all the answers

What is the glucose transporter found in neurons?

<p>GLUT3 (A)</p>
Signup and view all the answers

Where does anterograde and retrograde axonal transport take place?

<p>Neurofilaments (B)</p>
Signup and view all the answers

Which two molecular motors are involved in axonal transport?

<p>Kinesin and Dynein (D)</p>
Signup and view all the answers

Neurons typically use what molecule for a main source of energy?

<p>Glucose (C)</p>
Signup and view all the answers

Astroglial cells release what molecule as a complementary energy source for the neurons?

<p>Lactate (C)</p>
Signup and view all the answers

Astroglial cells will produce what in the CNS when there is injury to the cells?

<p>GFAP (A)</p>
Signup and view all the answers

What protein moves the synaptic vesicle?

<p>Clathrin (C)</p>
Signup and view all the answers

What is the rate limiting enzyme in the synthesis of Acetylcholine?

<p>Choline Acetyltransferase (A)</p>
Signup and view all the answers

What enzyme breaks down catecholamines?

<p>Monoamine Oxidase (D)</p>
Signup and view all the answers

Which of the following proteins is associated with Alzheimer's Disease?

<p>All of the above (D)</p>
Signup and view all the answers

What neurotransmitters are thought to primarily mediate inhibitory signals in the brain?

<p>GABA (D)</p>
Signup and view all the answers

What accounts for the distinct chemical composition observed between CNS and PNS myelin?

<p>The production of myelin by oligodendrocytes in the CNS versus Schwann cells in the PNS. (B)</p>
Signup and view all the answers

What role do the meninges play in CSF dynamics under pathological conditions?

<p>They become a source of CSF leading to increased protein concentrations. (B)</p>
Signup and view all the answers

In the visual transduction pathway, how does the activation of rhodopsin lead to the hyperpolarization of rod cells?

<p>Rhodopsin activates transducin, which activates cGMP phosphodiesterase, reducing cGMP levels and closing sodium channels. (B)</p>
Signup and view all the answers

How does the brain's reliance on glucose as its primary energy substrate influence neuronal responses to food deprivation?

<p>Neurons initiate eating behavior through sensing increased fatty acid concentrations. (B)</p>
Signup and view all the answers

What are the implications of the absence of cell division in postpartum neurons for recovery from CNS injuries and neurodegenerative diseases?

<p>It necessitates reliance on axonal transport for neuronal maintenance and repair. (D)</p>
Signup and view all the answers

How does the strategic allocation of GLUT1 and GLUT3 transporters facilitate brain glucose metabolism?

<p>GLUT1 on endothelial cells ensures glucose entry into the brain, while GLUT3 in neurons supports neuronal activity. (C)</p>
Signup and view all the answers

What is the primary mechanism by which caffeine affects neurotransmission, and how does this lead to its stimulant effects?

<p>By inhibiting phosphodiesterase, which increases cAMP levels, mimicking adrenergic neurotransmission. (C)</p>
Signup and view all the answers

Which of the following mechanisms contributes most significantly to the unique ionic environment of cerebrospinal fluid (CSF) compared to plasma?

<p>The active transport of ions across the blood-CSF barrier. (B)</p>
Signup and view all the answers

How does the mechanism of botulinum toxin's action on synaptic transmission differ from that observed in Lambert-Eaton syndrome?

<p>Botulinum toxin causes a presynaptic blockage of neurotransmitter release by hydrolyzing proteins, whereas Lambert-Eaton syndrome involves autoimmune antibodies against calcium channels. (D)</p>
Signup and view all the answers

How does the interdependent relationship between neurons and astrocytes in the brain manifest in energy metabolism and neurotransmitter recycling?

<p>Neurons shuttle glutamate to astrocytes, which convert it to glutamine and return it to neurons for glutamate synthesis. (D)</p>
Signup and view all the answers

What role does the enteric vagal nerve play in the bidirectional communication between the gut microbiome and the CNS?

<p>It transmits signals about the composition of the gut microbiome to the brain, influencing neurotransmission and behavior. (A)</p>
Signup and view all the answers

What are the key cellular mechanisms that contribute to both the neuroprotective and neurotoxic effects of microglia in the central nervous system?

<p>Microglia mediate synaptic pruning to refine neural circuits while also releasing pro-inflammatory cytokines that can damage neurons. (A)</p>
Signup and view all the answers

How does the differential diagnosis of CSF rhinorrhea from common nasal secretions guide clinical management and prevent potential complications?

<p>Accurate identification of CSF rhinorrhea prompts surgical repair to prevent meningitis, while misdiagnosis can lead to inappropriate treatments. (A)</p>
Signup and view all the answers

How does the synthesis and metabolism of acetylcholine (ACh) influence cognitive function, and what implications does this have for neurodegenerative diseases such as Alzheimer's?

<p>ACh synthesis depends on acetyl-CoA generation by pyruvate dehydrogenase, and its impairment can lead to cognitive deficits seen in Alzheimer's. (D)</p>
Signup and view all the answers

What is the role of the blood-brain barrier (BBB) and the choroid plexus in maintaining the unique composition of the cerebrospinal fluid (CSF), and how do these structures respond to inflammation or injury?

<p>The BBB selectively filters blood components to produce CSF, while the choroid plexus regulates ion and protein concentrations. (B)</p>
Signup and view all the answers

How does the role of anterograde and retrograde axonal transport differ in maintaining neuronal health, and what are the implications of their disruption in neurodegenerative diseases?

<p>Anterograde transport supplies essential proteins and mitochondria to the nerve terminal, while retrograde transport returns signaling molecules and damaged components for degradation. (A)</p>
Signup and view all the answers

How do the distinct types of glutamate receptors, namely ionotropic and metabotropic, contribute to synaptic transmission and neuronal plasticity?

<p>Ionotropic receptors directly gate ion channels for fast synaptic transmission, while metabotropic receptors activate intracellular signaling cascades for neuronal plasticity. (B)</p>
Signup and view all the answers

How does the interplay between glutamate and GABA contribute to neuronal function, and what diseases occur due to the imbalances between the two?

<p>Glutamate mediates fast excitatory neurotransmission while GABA facilitates inhibitory neurotransmission, thus imbalances in either results in seizures. (D)</p>
Signup and view all the answers

How do inflammatory conditions of the CNS or PNS affect the production of oligoclonal bands and what test is used to detect them?

<p>Locally synthesized antibodies give a unique pattern in CSF, detected via electrophoresis. (D)</p>
Signup and view all the answers

How is the action of catecholamines terminated in the synapse and why?

<p>Catecholamines are terminated by their reuptake and degradation to aldehydes by mitochondrial monoamine oxidases in the presynaptic terminal. (A)</p>
Signup and view all the answers

What role do the glutamate receptors play in LTP?

<p>They play a role in synaptic plasticity, termed long-term potentiation (LTP). (C)</p>
Signup and view all the answers

How do oligomers of amyloid-β (Aβ) affect cholinergic neurons and cognitive processes in the brain following the diagnosis of Alzheimer's Disease?

<p>The Aβ42 oligomers, preferentially impair cholinergic neurons in the brain septum, causing progressive loss of cognitive function. (A)</p>
Signup and view all the answers

How do neurons maintain the ionic gradients necessary for resting membrane potential, and how do disturbances in these gradients lead to neurological disorders?

<p>At rest, the neuron pumps ions, maintaining the ionic gradients causing mutations of sodium channels to occur at different sites and give rise to hyperkalemic periodic paralysis. (D)</p>
Signup and view all the answers

What protein moves materials through the axon?

<p>Kinase (B)</p>
Signup and view all the answers

What is the function of the protein, clathrin?

<p>An inward puckering movement of the membrane in nerve terminals. (D)</p>
Signup and view all the answers

What pathology/disease can result from the deficiency of thiamine in a patient and what treatment is used to resolve this?

<p>Wernicke-Korsakoff encephalopathy, because the activities of complexes of pyruvate dehydrogenase and ketoglutarate dehydrogenase are inhibited due to the deficit of their cofactor thiamine pyrophosphate. (C)</p>
Signup and view all the answers

What is the sequence of reactions that catecholamines, epinephrine and norepinephrine, are synthesized from?

<p>Synthesized from L-tyrosine in the sequence of reactions catalyzed by tyrosine hydroxylase/L-aromatic amino acid decarboxylase (AADC), then dopamine AADC and phentolamine-N-methyltransferase, yielding dopamine, norepinephrine, and epinephrine. (D)</p>
Signup and view all the answers

What are some drugs used to treat the brain using the serotonin symporter?

<p>Serotonin reuptake inhibitors (SSRIs) and serotonin-noradrenaline reuptake inhibitors (SNRIs). (C)</p>
Signup and view all the answers

In the brain, what happens due to the increase in glutamate at the synapse?

<p>Prolonged depolarization of the postsynaptic cells yielding a rise in intracellular Ca2+, free oxygen, nitrosyl, and synthesis of fatty acid radicals which results in structural damage take place. (D)</p>
Signup and view all the answers

In the visual system, what is the series of events that occurs following the moment that a photon interacts with the rhodopsin?

<p>Rhodopsin becomes activated, the level of cGMP decreases, Sodium entry into the cell is blocked, The rod cell hyperpolarizes, release of glutamate, action potential depolarizes the adjacent bipolar cell. (C)</p>
Signup and view all the answers

What distinguishes the configuration of oligoclonal bands in CSF and serum in neurosarcoidosis, compared to other intrathecal immune responses?

<p>Oligoclonal bands exhibit a 'mirror pattern,' being the same in both CSF and serum, reflecting passive transfer of immunoglobulins. (B)</p>
Signup and view all the answers

How does the brain ensure a stable rate of glucose provision to neurons, and what specific mechanisms are involved?

<p>Through the action of GLUT3 transporters, which are expressed exclusively in neurons, providing a high-affinity, insulin-independent mechanism for glucose uptake. (B)</p>
Signup and view all the answers

What role does the relative impermeability of the blood-brain barrier (BBB) play in maintaining central nervous system (CNS) homeostasis, and how does this impact drug delivery to the brain?

<p>Its selective permeability restricts the entry of large molecules and many drugs, necessitating specific transport mechanisms or drug modifications for effective CNS targeting. (A)</p>
Signup and view all the answers

In the context of axonal transport, how do kinesin and dynein facilitate the bidirectional movement of materials, and what implications do disruptions in their function have on neuronal health?

<p>Kinesin facilitates anterograde transport, moving materials from the cell body to the synapse, while dynein mediates retrograde transport, essential for signaling and waste removal, and their dysfunction contributes to neurodegenerative diseases. (D)</p>
Signup and view all the answers

Given the interdependent metabolic relationship between neurons and astrocytes, how does the astrocyte-neuron lactate shuttle (ANLS) contribute to brain energy metabolism and what are the consequences of its disruption?

<p>Astrocytes metabolize glucose to lactate, which neurons then use as an energy substrate, especially during high activity. Disruption of the ANLS leads to energy deficits, impaired neurotransmission, and neurodegeneration. (C)</p>
Signup and view all the answers

Considering that neurons do not undergo cell division postnatally, what are the implications for neuronal recovery after injury in the central nervous system (CNS), and how does this influence the progression of neurodegenerative diseases?

<p>The inability of neurons to divide postnatally limits their ability to regenerate or replace damaged cells, leading to reliance on plasticity and compensatory mechanisms for functional recovery, and contributing to the irreversible nature of neurodegenerative diseases. (D)</p>
Signup and view all the answers

What is the role of the meninges in CSF dynamics, and how are these dynamics altered under pathological conditions such as meningitis?

<p>The meninges represent a key site for CSF reabsorption into the venous sinuses, and inflammation, such as in meningitis, can significantly impede this process, leading to hydrocephalus. (B)</p>
Signup and view all the answers

In the context of differential diagnosis, what is the significance of distinguishing CSF rhinorrhea from common nasal secretions, and what specific diagnostic markers are used to differentiate between these fluids?

<p>CSF rhinorrhea requires surgical repair to prevent meningitis, and is differentiated from nasal secretions by the presence of asialotransferrin and beta-2 transferrin, which are specific to CSF. (D)</p>
Signup and view all the answers

How do the cellular mechanisms underlying detection of a single photon of light by rhodopsin ultimately initiate an action potential in the optic nerve?

<p>Photoisomerization of retinal leads to a cascade that closes sodium channels, hyperpolarizing the rod cell and reducing glutamate release, which disinhibits bipolar cells and leads to an action potential in ganglion cells. (D)</p>
Signup and view all the answers

How does the chronic deficiency of thiamine, commonly seen in Wernicke-Korsakoff encephalopathy, impair cholinergic neurotransmission, and what are the specific mechanisms underlying these effects?

<p>Thiamine deficiency impairs the activity of pyruvate dehydrogenase, reducing the production of acetyl-CoA, a necessary precursor for acetylcholine synthesis, and disrupting energy metabolism which leads to neuronal damage. (D)</p>
Signup and view all the answers

How does the administration of edrophonium improve muscle strength in myasthenia gravis, and what is the underlying mechanism of its action on cholinergic neurotransmission?

<p>Edrophonium inhibits acetylcholinesterase, increasing the concentration of acetylcholine in the synaptic cleft and prolonging its action at remaining functional receptors, compensating for the reduced number of receptors due to autoantibodies. (A)</p>
Signup and view all the answers

In vision, what are the specific steps that result in the hyperpolarization of rod cells after a photon is detected by rhodopsin?

<p>Activation of rhodopsin leads to a cascade where transducin activates phosphodiesterase, which hydrolyzes cGMP, closing sodium channels and causing hyperpolarization. (D)</p>
Signup and view all the answers

Given that synapses in the human brain are incredibly dense, what mechanisms ensure the efficient removal of neurotransmitters from the synaptic cleft to facilitate subsequent signal transmission?

<p>Enzymatic degradation, reuptake by presynaptic neurons and astrocytes, and diffusion collectively regulate neurotransmitter concentrations in the synaptic cleft, allowing for termination of synaptic signaling and subsequent transmission. (C)</p>
Signup and view all the answers

How does serotonin transporter (SERT) dysfunction contribute to the pathophysiology of autism spectrum disorder (ASD), and what are the specific mechanisms potentially involved?

<p>SERT dysfunction results in reduced serotonin levels in the synaptic cleft, leading to decreased serotonergic neurotransmission and contributing to dysregulation of mood, social behavior, and repetitive behaviors seen in ASD. (C)</p>
Signup and view all the answers

How do the locations of neuronal cell bodies and synapses, distributed throughout the nervous system, relate to the necessity for an efficient axonal transport system?

<p>The long distances between neuronal cell bodies (containing the nucleus) and synapses necessitate efficient axonal transport to deliver newly synthesized proteins and organelles, as well as recycle materials, essential for neuronal function and survival. (D)</p>
Signup and view all the answers

In the context of the adverse effects of amyloid-ẞ (Aβ) oligomers in Alzheimer's disease, how do these oligomers specifically disrupt cholinergic neurotransmission and contribute to cognitive decline?

<p>Aβ oligomers preferentially impair cholinergic neurons in the brain septum, disrupt long-term potentiation, stimulate tau hyperphosphorylation, and trigger synaptic and neuronal loss, thus leading to cognitive decline. (C)</p>
Signup and view all the answers

How is the action of catecholamines terminated in the synapse, and why is this termination mechanism crucial for neuronal function?

<p>Catecholamine action is terminated primarily by their reuptake into the presynaptic terminal and degradation by mitochondrial monoamine oxidases or methylation, which is essential to regulate signaling, prevent overstimulation, and maintain synaptic efficiency. (A)</p>
Signup and view all the answers

In neurons, upon an increase in glutamate at the synapse, what is the most immediate response and how do these responses play a role in neuronal signaling?

<p>Increased glutamate binding to ionotropic receptors triggers an influx of Na⁺ and Ca²⁺, leading to depolarization and activation with subsequent action potential. (A)</p>
Signup and view all the answers

Guillain-Barré syndrome typically affects myelin in which part of the nervous system, and what is the underlying immunological mechanism?

<p>Guillain-Barré syndrome affects myelin in the peripheral nervous system via molecular mimicry, where antibodies against gangliosides on peripheral nerves cause demyelination. (C)</p>
Signup and view all the answers

How is the synthesis of acetylcholine (ACh) regulated and what is the rate-limiting step?

<p>ACh synthesis is regulated by the availability of acetyl-CoA and choline, with choline acetyltransferase (ChAT) catalyzing the rate-limiting step. (A)</p>
Signup and view all the answers

What is the primary function of the protein clathrin in synaptic transmission, and how does it contribute to the recycling of synaptic vesicles?

<p>Clathrin mediates the endocytosis of synaptic vesicle membrane components, driving the formation of coated vesicles from the plasma membrane for recycling. (B)</p>
Signup and view all the answers

In the context of neurotransmission, how does the mechanism of action of botulinum toxin differ from that of Lambert-Eaton syndrome, and what implications does this have for their clinical presentations?

<p>Botulinum toxin blocks the presynaptic release of acetylcholine by hydrolyzing proteins, while Lambert-Eaton syndrome is caused by antibodies against voltage-gated calcium channels, reducing acetylcholine release and thus also leading to muscle weakness. However, they present with different progression and antibody presence. (A)</p>
Signup and view all the answers

What are the distinct characteristics of myelin produced by oligodendrocytes in the CNS compared to Schwann cells in the PNS, and what implications do these differences have for nerve regeneration and neurological disorders?

<p>CNS myelin is produced by oligodendrocytes, which can myelinate multiple axons, while PNS myelin is produced by Schwann cells, which myelinate a single axon, influencing remyelination efficiency and the pathology of demyelinating diseases. (B)</p>
Signup and view all the answers

Why is the brain considered to be an 'energy-demanding' organ, and what proportion of the body's total glucose consumption does it account for under resting conditions?

<p>The brain is energy-demanding because it needs to maintain neurotransmission, ionic gradients and resting membrane; therefore it accounts for approximately 2% of the body mass, but accounts for 20% of the glucose consumption under resting conditions. (C)</p>
Signup and view all the answers

How does long-term potentiation (LTP) contribute to synaptic plasticity, and what are the critical molecular mechanisms involved in this process?

<p>LTP is the long-lasting strengthening of synapses through increased glutamate production and the modulation of postsynaptic receptor expression and sensitivity via processes like NMDA receptor activation and downstream signaling cascades. (C)</p>
Signup and view all the answers

What are the primary mechanisms by which microglia contribute to both neuroprotection and neurotoxicity in the central nervous system (CNS)?

<p>Microglia contribute by phagocytosing pathogens and cellular debris (neuroprotection) as well as releasing neurotoxic substances and creating inflammatory cascades (neurotoxicity), with their role highly dependent on the context and activation state. (C)</p>
Signup and view all the answers

How might dysfunctions of the enteric vagal nerve affect gut motility, and what role does the gut microbiome play in this context?

<p>Dysfunctions of the vagal nerve induce problems with intestinal motility, as the vagal nerve serves as means of communication between the gut and the brain, with the gut microbiome acting upon the vagal nerve. (C)</p>
Signup and view all the answers

The peripheral nervous system (PNS) is located entirely within the dura.

<p>False (B)</p>
Signup and view all the answers

The blood-brain barrier's permeability is constant and does not depend on the size of the molecule.

<p>False (B)</p>
Signup and view all the answers

Approximately 75% of the CSF protein pool is synthesized within the brain.

<p>False (B)</p>
Signup and view all the answers

Increased chloride ion concentration in the CSF, compared to plasma, helps maintain anion balance.

<p>True (A)</p>
Signup and view all the answers

The blood-CSF barrier provides the primary source of CSF, accounting for about two-thirds of its volume.

<p>True (A)</p>
Signup and view all the answers

Asialotransferrin is a useful marker protein in the CSF due to its abundance in systemic circulation.

<p>False (B)</p>
Signup and view all the answers

Neurons typically constitute approximately 75% of the cells in the human nervous system.

<p>False (B)</p>
Signup and view all the answers

GLUT1 insulin-dependent transporters facilitate glucose transport into the brain.

<p>False (B)</p>
Signup and view all the answers

The process of axonal transport, essential for neuronal function, relies solely on retrograde transport to move materials.

<p>False (B)</p>
Signup and view all the answers

Neurons are incapable of dividing postnatally.

<p>True (A)</p>
Signup and view all the answers

Astrocytes help filter materials from the blood for neurons in the white matter.

<p>False (B)</p>
Signup and view all the answers

During an action potential, potassium influxes into the cell and sodium effluxes out of the cell, leading to repolarization.

<p>False (B)</p>
Signup and view all the answers

Lambert-Eaton syndrome primarily affects the M/N subtype of calcium channels due to molecular mimicry.

<p>False (B)</p>
Signup and view all the answers

Neurotransmitters are removed from the synaptic cleft solely through hydrolysis by enzymes.

<p>False (B)</p>
Signup and view all the answers

Synaptic transmission involves primarily the unidirectional flow of information from the presynaptic to the postsynaptic neuron without any recycling of components.

<p>False (B)</p>
Signup and view all the answers

Acetylcholine (ACh) is synthesized from acetyl-CoA and choline by pyruvate dehydrogenase.

<p>False (B)</p>
Signup and view all the answers

Nicotine receptors are characterized by effects originally demonstrated with muscarine.

<p>False (B)</p>
Signup and view all the answers

The most common neurotransmitter receptor is the metabotropic receptor.

<p>True (A)</p>
Signup and view all the answers

Decreased levels of A$\beta$42 and tau in the CSF indicate neurodegeneration in Alzheimer's Disease.

<p>False (B)</p>
Signup and view all the answers

Dopamine is synthesized from L-tyrosine in a single-step reaction catalyzed by tyrosine hydroxylase.

<p>False (B)</p>
Signup and view all the answers

Match the glial cell type with its primary function in the central nervous system:

<p>Astrocytes = Provide metabolic support and contribute to the blood-brain barrier Oligodendrocytes = Generate myelin sheaths to insulate axons Microglia = Act as resident macrophages, scavenging for debris and pathogens Ependymal cells = Secrete cerebrospinal fluid</p>
Signup and view all the answers

Match the neurotransmitter with its primary role in the brain:

<p>Glutamate = Primary excitatory neurotransmitter involved in learning and memory GABA = Primary inhibitory neurotransmitter, reducing neuronal excitability Dopamine = Involved in reward, motivation, and motor control Acetylcholine = Plays a role in muscle movement and cognitive functions</p>
Signup and view all the answers

Match the component of the neuron with its role in axonal transport:

<p>Anterograde transport = Moves materials from the cell body to the synapse Retrograde transport = Moves materials from the synapse to the cell body Kinesin = Motor protein involved in anterograde transport Dynein = Motor protein involved in retrograde transport</p>
Signup and view all the answers

Match the process with its description related to synaptic transmission:

<p>Neurotransmitter release = Action potential triggers exocytosis of neurotransmitters into the synaptic cleft Receptor binding = Neurotransmitters bind to receptors on the postsynaptic neuron Signal termination = Neurotransmitters are cleared from the synaptic cleft via reuptake or degradation Vesicle recycling = Synaptic vesicles are reformed and refilled with neurotransmitters for subsequent release</p>
Signup and view all the answers

Match each condition with its corresponding description related to myelin:

<p>Multiple sclerosis = Demyelination in the CNS, affecting oligodendrocytes Guillain-Barré syndrome = Demyelination in the PNS, often triggered by an infection Myelin = Insulates nerve axons and is made of oligodendrocytes in the CNS and Schwann cells in the PNS Nodes of Ranvier = Gaps in the myelin sheath that are parallel to sites of depolarization within the axon</p>
Signup and view all the answers

Match the condition with its effect on the nervous system:

<p>Alzheimer's disease = Characterized by amyloid plaques and neurofibrillary tangles, leading to cognitive decline Parkinson's disease = Involves loss of dopaminergic neurons in the substantia nigra, affecting motor control Myasthenia gravis = Autoantibodies block nicotinic acetylcholine receptors, causing muscle weakness Wernicke-Korsakoff encephalopathy = Thiamine deficiency inhibits pyruvate dehydrogenase</p>
Signup and view all the answers

Match cell type with the marker proteins:

<p>Neurons = Neuron-specific enolase Astrocytes = GFAP (glial fibrillary acidic protein) Oligodendrocytes = Myelin basic protein Microglia = Ferritin</p>
Signup and view all the answers

Match the receptor to its description:

<p>Ionotropic receptors = Fast-acting; ligand-gated ion channels; dimeric structures Metabotropic receptors = Slower, long-term effect; G-protein coupled receptors NMDA receptors = Ionotropic glutamate receptors; fast excitatory receptors Muscarinic receptors = Metabotropic glutamate receptors; activates G-proteins</p>
Signup and view all the answers

Match the term related to vision with its appropriate description:

<p>Rod cell = Hyperpolarizes when light strikes Rhodopsin = G-protein coupled receptor involved in vision Transretinal = Form that cis-retinal is converted to to initiate vision Glutamate = Neurotransmitter released in the human eye</p>
Signup and view all the answers

Match each term to its role within catecholamine signaling:

<p>Epinephrine = Synthesized from L-tyrosine Dopamine = Transmitter in the dopaminergic neurons of several brain areas α-adrenergic receptor = Blocked by phentolamine β-adrenergic receptor = Blocked by propranolol</p>
Signup and view all the answers

Flashcards

What does the central nervous system (CNS) include?

Brain and spinal cord

What does the peripheral nervous system (PNS) include?

Nerves outside the dura.

What is the dura?

A thick, fibrous covering containing cerebrospinal fluid (CSF) that divides the CNS and PNS.

What is myelin?

Insulates axons and have distinct chemical compositions in the CNS vs. PNS.

Signup and view all the flashcards

What cells produce myelin?

Oligodendrocytes (CNS) and Schwann cells (PNS)

Signup and view all the flashcards

Is the blood-brain barrier (BBB) absolute?

Not absolute; its permeability depends on the size of the molecule.

Signup and view all the flashcards

What are the sources of CSF proteins?

Proteins pass from external cells/tissues into the CSF under normal and pathologic conditions.

Signup and view all the flashcards

What are the six sources of CSF?

Blood-brain barrier, blood-CSF barrier, dorsal root ganglia, brain parenchyma, circulating CSF cells, and meninges.

Signup and view all the flashcards

What are ependymal cells?

Ciliated cells secreting brain-specific proteins

Signup and view all the flashcards

What are the major types of glial cells?

Astrocytes, oligodendrocytes, and microglia

Signup and view all the flashcards

What are the significant features of neurons?

Length, prolific interconnections, and inability to divide postpartum

Signup and view all the flashcards

Why do neurons depend on axonal transport?

To transport materials between the nucleus and synapse

Signup and view all the flashcards

What 'molecular motors' mediate axonal transport?

Kinesin (anterograde) and dynein (retrograde)

Signup and view all the flashcards

Which motor proteins facilitate anterograde and retrograde transport?

Kinesin in anterograde and dynein for retrograde transport.

Signup and view all the flashcards

What is the primary energy substrate for the brain?

Glucose

Signup and view all the flashcards

What cells utilize glucose and release lactate for neurons?

Astrocytes

Signup and view all the flashcards

What transporter secures glucose provision in neurons?

GLUT3

Signup and view all the flashcards

What molecule is a marker of neuron metabolic competency?

N-acetyl-L-aspartate (NAA)

Signup and view all the flashcards

What process requires large amounts of energy in neurons?

Synaptic transmission

Signup and view all the flashcards

What protein do astrocytes synthesize in response to CNS injury?

Glial fibrillary acidic protein (GFAP)

Signup and view all the flashcards

What is the function of oligodendrocytes?

Insulate neuronal processes and stop cross-talk.

Signup and view all the flashcards

What is a unique chemical characteristic of the brain?

The massively high density of synapses between different neurons.

Signup and view all the flashcards

How is Synaptic Transmission achieved?

Presynaptic neuron releases neurotransmitter, which binds to receptors on the postsynaptic neuron

Signup and view all the flashcards

What are the two groups of neurotransmitter receptors?

Ionotropic and metabotropic

Signup and view all the flashcards

What is the best-studied neurotransmitter?

Acetylcholine (ACh)

Signup and view all the flashcards

What enzyme synthesizes acetylcholine?

Choline acetyltransferase (ChAT)

Signup and view all the flashcards

What are the two types of acetylcholine receptors?

Nicotinic and muscarinic receptors

Signup and view all the flashcards

What is the best-studied blocker for the nicotinic receptor?

a-bungarotoxin

Signup and view all the flashcards

What is a precursor of norepinephrine and epinephrine?

Dopamine

Signup and view all the flashcards

Which diseases are associated with dopamine metabolism disturbance?

Parkinson's disease, schizophrenia, and restless legs syndrome.

Signup and view all the flashcards

What precursor to dopamine crosses the blood-brain barrier?

L-DOPA

Signup and view all the flashcards

What are the two separate catecholamine receptors?

α-adrenergic and β-adrenergic

Signup and view all the flashcards

What returns serotonin from the synaptic cleft?

Serotonin transporter

Signup and view all the flashcards

To what family of proteins does serotonin transporter belong?

Sodium symporters

Signup and view all the flashcards

What conditions implicate mutations of the serotonin transporter?

Autism, ADHD, and parkinsonism

Signup and view all the flashcards

What does glutamate mediate?

Learning, memory formation, and cognition

Signup and view all the flashcards

To what two classes of receptors does glutamate bind?

ionotropic and metabotropic receptors

Signup and view all the flashcards

What enzyme converts glutamate to glutamine in astroglial cells?

Glutamine synthetase

Signup and view all the flashcards

What causes glutamate toxicity?

Excessive glutamate release/impaired uptake

Signup and view all the flashcards

What is the chief inhibitory neurotransmitter in the brain?

GABA

Signup and view all the flashcards

What drugs act as GABA receptor agonists?

Barbiturates, benzodiazepines, chloral hydrate, and valproate

Signup and view all the flashcards

Multiple Sclerosis

Demyelination of CNS, leading to scar tissue plaques.

Signup and view all the flashcards

Guillain-Barré syndrome

Demyelination of PNS, leading to motor weakness.

Signup and view all the flashcards

Asialotransferrin Marker

Loss of smell, CSF leak (rhinorrhea).

Signup and view all the flashcards

Circulating CSF cells

Mainly lymphocytes within the CNS, synthesize local antibodies.

Signup and view all the flashcards

Meningitis Treatment

In brain infections such as meningitis, steroids are given in addition to antibiotics to suppress the potentially devastating effects, within this confined space, of inflammation associated with the intrathecal immune response.

Signup and view all the flashcards

Brain Parenchyma

The part of the brain that produces brain-specific proteins.

Signup and view all the flashcards

Protoplasmic Astrocytes

They help to 'filter' materials from the blood, and a separate set of processes surrounding the neurons.

Signup and view all the flashcards

Ion Channel Mutations

Mutations of sodium channels causes hyperkalmeic periodic paralysis. The chloride anion moves through separate channels, which are implicated in specific pathologic states, such as myotonia.

Signup and view all the flashcards

Lambert-Eaton syndrome

Autoantibodies block presynaptic neurotransmitter release via molecular mimicry.

Signup and view all the flashcards

Botulinum Toxin

A protein derived from anaerobic bacteria which contains enzymes to hydrolyze the presynaptic proteins involved in the release of neurotransmitters.

Signup and view all the flashcards

Drugs causing addiction

Excessive stimulation of the release and the increase of dopamine level in the synaptic cleft. They also stimulate serotoninergic and norepinephrinergic transmission in the brain.

Signup and view all the flashcards

Termination of Catecholamines

Degradation to aldehydes by mitochondrial monoamine oxidases.

Signup and view all the flashcards

Long-term potentiation (LTP)

This phenomenon takes place in the hippocampus and in the different regions of brain cortex, and it is involved in learning, memory formation, and other cognitive functions.

Signup and view all the flashcards

peripheral cholinergic neurons

Located in parasympathetic ganglia and innervate all visceral tissues.

Signup and view all the flashcards

NMDA

Caused by the excitotoxic effects of excessive activation of glutamatergic neurons.

Signup and view all the flashcards

Eye vision mechanism

The process by which the human eye can detect a single photon of light.

Signup and view all the flashcards

CSF compartment

Watertight space within the confines of the dura, containing CSF.

Signup and view all the flashcards

Differential Diagnosis of Nasal Discharge (Rhinorrhea)

Technique distinguishing CSF rhinorrhea from nasal secretions.

Signup and view all the flashcards

Rhodopsin

Produced by retina, causes action potential by retinal ganglion neuron.

Signup and view all the flashcards

Synaptic Transmission

Impulse occurs when electrical action potential triggers neurotransmitter release from axon's nerve terminals.

Signup and view all the flashcards

Neurone plasticity

Describes the ability of the brain to reorganize synaptic connections.

Signup and view all the flashcards

Synaptic Transmission Involves Recycling

Recycling of membrane constituents also associated with this process.

Signup and view all the flashcards

Resting membrane potential

Describes when neurotransmitter will finally trigger an action potential at the axon hillock.

Signup and view all the flashcards

Pyruvate Dehydrogenase

States that ACh synthesis and the functional competence of cholinergic neurons strongly depend on the provision of acetyl-CoA.

Signup and view all the flashcards

Neurotoxic signals

Results in aberrant proteolysis of amyloid precursor protein (APP), yielding production of amyloid-ẞ peptide (1–42) (Aẞ42).

Signup and view all the flashcards

Agrp neurons

Serve as the central components in the brain's regulation of hunger and satiety.

Signup and view all the flashcards

Neuron Diversity

Neurons present diverse morphologic features concerning cell bodies, axons, and dendrites, as well as neurotransmitter and functional phenotypes.

Signup and view all the flashcards

Astrocyte Function

Astrocytes uptake glucose and provide energy substrates (lactate) and neurotransmitter precursors (glutamine) to neurons, also make up part of the blood-brain barrier.

Signup and view all the flashcards

Axon Length

Axons of motor and sensory neurons can be up to 1 m long.

Signup and view all the flashcards

Membrane invagination

An increase in total membrane mass is redressed by invagination of the lateral aspects of the nerve terminals

Signup and view all the flashcards

Fast axonal transport

In the axon, there is fast axonal transport along microtubules via the motile proteins, kinesin (anterograde transport) or dynein (retrograde transport).

Signup and view all the flashcards

GABAB Function

Potassium efflux from the cell, hyperpolarizing the membrane.

Signup and view all the flashcards

GABA’s mechanism

The inhibitory effect of GABA on postsynaptic neurons results from binding to specific GABAĄ receptors.

Signup and view all the flashcards

Epilepsy Cause

Excessive glutamate release by pathologically stimulated glutamatergic neurons and/or the deficiency of inhibitory GABAergic transmission

Signup and view all the flashcards

Active Zone

Synaptic vesicles fuse within the active zone, facilitated by scaffold proteins.

Signup and view all the flashcards

Role of Calcium Ions

The movement of calcium ions within cells often provides a trigger for the cells to synchronize an activity such as synaptic release of neurotransmitter

Signup and view all the flashcards

Botulinum Exotoxin

Hydrolyzes the presynaptic proteins involved in the release of neurotransmitter, similar to Lambert-Eaton myasthenic syndrome

Signup and view all the flashcards

Serotonin transporter

A membrane protein present on the presynaptic neurons.

Signup and view all the flashcards

Rhodopsin activation

The light photons activate rhodopsin via G protein-coupling in rod cells

Signup and view all the flashcards

Blood-brain barrier action

CSF is partially produced, about one-third of the total volume, through the action of this.

Signup and view all the flashcards

Asialotransferrin

A marker protein in CSF, transferrin lacking sialic acid and recycled systemically.

Signup and view all the flashcards

Glial Cells

These are three major types of cells in the nervous system.

Signup and view all the flashcards

Energy substrate

Glucose is almost exclusively this for the brain

Signup and view all the flashcards

Inhibition by Methylxanthines

Molecular process involving caffeine, mimicking adrenergic neurotransmission.

Signup and view all the flashcards

Choline acetyltransferase (ChAT)

ACh is synthesized from acetyl-CoA and choline by this enzyme.

Signup and view all the flashcards

Pyruvate Dehydrogenase Inhibition

Encephalopathy caused by ACh deficits linked to inhibition/inactivation of this enzyme.

Signup and view all the flashcards

Pheochromocytoma

Excess compounds in urine may hint at adrenal medullar tumor

Signup and view all the flashcards

Vagus Nerve

The intestine communicates with the CNS through this

Signup and view all the flashcards

Brain and Peripheral Nerve Distinction

The division between the CNS and PNS, delineated by the dura mater.

Signup and view all the flashcards

Neuron Energy Generation

Neurons generate 60-80% of brain energy, crucial for restoring plasma membrane potentials.

Signup and view all the flashcards

Energy for Neurotransmission

Neurotransmission relies heavily on this to sustain signaling.

Signup and view all the flashcards

Components of the Nervous System

The nervous system contains diverse, tightly interacting structural, metabolic, and functional components

Signup and view all the flashcards

Axonal Transport

Axonal transport ensures bidirectional movement of organelles and proteins, maintaining neuronal integrity.

Signup and view all the flashcards

Glucose and Energy

Glucose transport and glycolysis facilitate high energy production in the nervous system.

Signup and view all the flashcards

Neurons Support

Neurons use metabolic support from astroglial and microglial cells, and structural support from oligodendroglial cells

Signup and view all the flashcards

Study Notes

GABA-ergic transmission

  • Several GABAA receptor agonists and GABA uptake inhibitors are used as sedatives, tranquilizers, or anxiolytic drugs: barbiturates, benzodiazepines, chloral hydrate, and valproate, and ethanol is also a receptor agonist.
  • GABAB receptors are heterodimeric GPCRs that activate the inwardly rectifying potassium channels and determine membrane excitability by promoting potassium efflux and hyperpolarizing the membrane.
  • GABA inhibits calcium channels to reduce calcium influx and link to the cAMP-protein kinase A (PKA) signaling pathway.

Studying That Suits You

Use AI to generate personalized quizzes and flashcards to suit your learning preferences.

Quiz Team

Related Documents

More Like This

Use Quizgecko on...
Browser
Browser