echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Study of Nervous System > Mol Psychiatry: A must-see for depression research [Review]: Summarize the six molecular mechanisms of depression from the perspective of synapses

    Mol Psychiatry: A must-see for depression research [Review]: Summarize the six molecular mechanisms of depression from the perspective of synapses

    • Last Update: 2022-10-25
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com

    Major depressive disorder (MDD) is a psychiatric disorder
    whose molecular etiology is unknown.
    Studies at different molecular levels have shown that the pathogenesis of depression involves a variety of highly complex and interrelated metabolic pathways, including monoamines, HPA axes, neurotrophic factors and neurogenesis, excitatory and inhibitory neurotransmission, mitochondrial dysfunction, epigenetics, inflammation, opioid system, myelination, and gut-brain axis
    .

    An article published in the journal Mol Psychiatry on October 6, 2022 by Theo Rein of the Max Planck Institute of Psychiatry in Germany comprehensively summarizes the six major pathogenesis hypotheses of depression, elucidating how signaling pathways and molecular systems interact in depression, and how each pathway or system relates
    to synaptic transmission.

    Figure 1: Molecular mechanisms of depression

    1

    The monoamine hypothesis

    The initial evidence supporting the "monoamine hypothesis for depression" was that monoamine oxidase inhibitors and tricyclic antidepressants could improve depressive symptoms
    by enhancing the activity of serotonin and norepinephrine.
    Although there have been many studies to support this hypothesis, the downside is that antidepressants usually take weeks to clinically act, while drugs can raise monoamine levels
    almost instantaneously.

    In addition, approximately one-third of patients with depression do not respond to antidepressants that act only by inhibiting monoamine reabsorption, and limiting the availability of tryptophan, a serotonin precursor, does not induce a depressive episode
    in all patients.
    Therefore, the monoamine deficiency hypothesis may not be widespread in depressed patients, suggesting that other pathways and neurotransmitters are associated with
    depression.

    Depression is associated with other neurotransmitter disorders in the brain, cerebrospinal fluid, and peripheral tissues, including GABA and glutamatergic systems
    .
    Depressed patients have reduced
    glutamate levels in specific brain regions.
    Therefore, newly developed antidepressant treatments focus on reversing glutamate and GABA deficiencies
    by targeting AMPA receptors or type 2 metabolic glutamate receptors.
    This has also led to the discovery
    of rapid antidepressants such as ketamine.

    In basic and clinical studies, ketamine rapidly increases glutamate signaling, producing rapid, sustained antidepressant effects
    .
    Ketamine increases the overall activity
    of the prefrontal cortex by blocking NMDA receptors and thus excitatory glutamate signaling in GABAergic neurons.

    Monoamines not only directly affect synaptic nerve transmission, but also indirectly affect intracellular pathways
    through their G protein-coupled receptors.
    Among them, opioid receptors interact functionally with
    5-HT and dopamine receptors through heterodimeration.
    A large body of clinical and preclinical evidence suggests the involvement of opioid receptors in MDD pathology
    .
    Opioid receptors negatively regulate neurotransmitter release and excitability of neurons by activating G protein-mediated mechanisms, resulting in enhanced potassium channel function, cellular depolarization, inhibition of voltage-gated calcium channel function, and negative regulation of neurotransmitter release, further affecting neuronal activity and plasticity
    .

    Figure 2: Receptor-related signaling pathways of serotonin, opioids, and BDNF regulate neuronal activity and synaptic function

    2

    The neurotrophic factor hypothesis

    The "neurotrophic hypothesis for depression" suggests that disruption of neurotrophic support is a key mechanism
    of MDD-related synaptic and brain-related functional changes.
    Neurotrophic factors are responsible for neuronal network formation, support, and plasticity
    .
    Among them, BDNF is an important member of the neurotrophic factor family, which can activate promyosin-associated kinase (Trk) and p75 receptors
    .
    Numerous studies have shown a decrease
    in blood levels of neurotrophic factors in patients with persistent depression and relapse, in animal models of depression.
    Notably, antidepressant therapy and electroconvulsive therapy increased BDNF levels
    .

    Traditional and rapid antidepressants not only require BDNF expression and its downstream signaling to take effect, but also antidepressants can directly bind to the transmembrane domain of TrkB dimer to form stable conformations of multi-protein complexes and promote the binding
    of TrkB and BDNF.
    Through the Trk receptor, neurotrophic factors can activate cell signaling pathways that regulate cell fate, axon growth, dendritic growth and pruning, and overall normal neuronal function
    .

    One of the most notable roles of BDNF is to promote hippocampal adult neurogenesis, which may play a role
    through most of the signaling described above.
    Hippocampal neurogenesis defects in MDD are associated
    with a decrease in hippocampal size and volume, a decrease in the number of neurons and glial cells, and a decrease in cell size found at autopsy.
    Studies have found a significant correlation between neurogenesis and synaptic activity, including long-term enhancement (LTP).

    Adult neonatal neurons can regulate dendritic spike density and excitatory synaptic transmission
    by reassigning existing synapses.
    Importantly, antidepressants induce neurogenesis, increase plasticity, and reverse hippocampal atrophy
    .

    3

    HPA axis hypothesis

    Stress exposure, especially early in life, is the most well-studied and identified risk factor
    for depression.
    The HPA axis is key
    to coordinating the body's stress response.
    The body terminates the stress response
    by activating the negative feedback mechanism of glucocorticoid receptors (GRs) by stress-secreted glucocorticoids.
    Elevated cortisol levels, HPA hyperactivity, and dysfunction
    of negative HPA axis feedback have been found in some patients with depression.
    Therefore, multiple drugs targeting the HPA axis have been developed to treat depression, including corticosteroid synthesis inhibitors, GR antagonists, adrenocorticotropin-releasing hormone receptor antagonists
    .

    Stress induces atrophy of the cerebral dendritic apex and postsynaptic dendritic spines, leading to significant synaptic remodeling
    .
    Mechanically, glucocorticoids increase the pool
    of easily released glutamate vesicles in the prefrontal cortex by activating membrane receptors.
    The synaptic and behavioral effects of stress are also mediated
    through the opioid system.
    Signal transduction of glucocorticoids interacts with most depression-related pathways such as BDNF, FKBP51, and autophagy pathways
    .

    Figure 3: Stress signals are intertwined with multiple depression-related pathways

    4

    The cytokine hypothesis

    The "cytokine hypothesis for depression" holds that the dysregulation of MDD and inflammatory processes is bidirectional
    .
    Persistent immune responses, such as infections, malignancies, or autoimmune diseases, can lead to depression
    .
    In fact, an enhanced inflammatory response is associated with
    MDD.
    Specific pro-inflammatory cytokines and their receptors associated with MDD include IL-6, TNF-α, IL-1β, IL-2, IL-2, IL-2 receptors, IL-4, IL-10, IL-1 receptor antagonists, transforming growth factor-β, and c-reactive protein (CRP).

    Pro-inflammatory cytokines are also associated
    with MDD severity.

    Many mechanisms have been proposed to explain the occurrence of inflammation in MDD, including inflammasome signaling pathways, oxidative stress, changes in BBB permeability, and the entry of peripheral immune cells into the brain
    .
    Mechanisms by which inflammatory pathways affect synaptic activity include pro-inflammatory cytokines regulating the expression of NMDA and AMPA receptor subunits, reducing AMPA receptor phosphorylation, and ultimately affecting glutamate synapses and LTP-related processes
    .

    The immune system is closely related to the neuroendocrine system, and glucocorticoids exert pro- or anti-inflammatory effects in
    different situations.
    In addition, increased inflammatory mediators in MDD can significantly interfere with mitochondrial oxidative phosphorylation and ATP production, ultimately leading to increased
    oxidative stress.

    5

    The mitochondrial hypothesis and oxidative stress hypothesis

    The evidence supporting the "mitochondrial hypothesis of depression" is that a large number of studies have found that some depressed patients have mitochondrial dysfunction, changes in mitochondrial structure and function, including decreased ATP production, and disturbances
    in mitochondrial dynamics (fusion, division, mitophagy).
    Mitochondrial dysfunction also produces free radicals and oxidative stress
    .
    Markers of oxidative stress are elevated in depression, while antioxidant capacity is reduced
    .
    In addition, as the disease progresses, mitochondrial dysfunction and oxidative damage develop
    progressively.

    Therefore, the "oxidative stress hypothesis of depression" proposes that oxidative stress is the cause of
    brain structural changes in patients with depression.
    Normal levels of reactive oxygen species (ROS) are important signaling molecules that play a key role
    in neuronal cell function.
    However, when at high levels and low in the presence of antioxidants, ROS can be harmful
    to neurons and LTP.
    Increased oxidative stress may lead to further mitochondrial damage, increased apoptosis, and ultimately inflammatory signaling
    .

    Mitochondria regulate synaptic function and plasticity in a variety of ways, including ATP production, Ca2+ buffering and signaling, neurotransmitter synthesis, establishment and maintenance of membrane excitability, and regulation of synaptic vesicle pools and neurotransmitter release
    .
    Mitochondria can produce oxygen and nitrogen needed for synaptic plasticity and activate caspase in dendrites, inducing the clearance
    of postsynaptic dendritic spines.

    Figure 4: The role of mitochondrial dysfunction in depression and its impact on synaptic function

    6

    The "microbial-gut-brain axis" hypothesis

    The evidence supporting the "gut-brain axis" hypothesis is a change
    in the gut microbiome in MDD.
    Conversely, supplementation with probiotics or the Mediterranean diet has an antidepressant effect
    on patients.
    A causal relationship between microbiome alterations and depression-like behavior can also be confirmed
    from fecal bacteria transplantation microbiota or specific bacterial experiments.
    Microorganisms affect brain activity
    by regulating synaptic function through specific molecules.
    A typical example is the kynurenine pathway, which is a metabolite
    of the essential amino acid tryptophan.
    Tryptophan is one of
    the first nutrients reported (more than 60-80 years ago) associated with depression.

    The conversion of tryptophan to the neurotransmitter serotonin is clearly associated
    with synaptic function and depression.
    However, tryptophan mainly produces neurotoxic (e.
    g.
    , quinolinic acid) and neuroprotective (e.
    g.
    , kynurinic acid) metabolites
    primarily through kynurenine metabolic pathways.
    By binding to glycine binding sites, kynuuric acid acts directly on synapses
    as a glutamate receptor antagonist.
    Quinolinic acid, on the other hand, is a glutamate receptor agonist, which enhances the release of glutamate and inhibits the reuptake
    of glutamate by astrocytes.

    Figure 5: Illustration of the role of gut microbes and their metabolites in depression - kynurenine pathway

    summary

    This review summarizes the molecular connections
    between the main pathways and systems that contribute to the development and progression of depression.
    The relative contribution of each pathway varies between individual patients, reflecting the high complexity
    of the disease.
    It is
    unrealistic to fully understand the association of multiple molecular pathways with MDD.

    Original source:

    Fries, G.
    R.
    , Saldana, V.
    A.
    , Finnstein, J.
    et al.
    Molecular pathways of major depressive disorder converge on the synapse.
    Mol Psychiatry (2022).
    https://doi.
    org/10.
    1038/s41380-022-01806-1.

    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.