We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
EditorialFree Access

Lipid rafts, G proteins and the etiology of and treatment for depression: progress toward a depression biomarker

    Robert J Donati

    University of Illinois at Chicago College of Medicine, Department of Physiology & Biophysics, Chicago, IL 60612-7342, USA and, Basic & Health Science Department, Illinois College of Optometry, Chicago, IL 60616, USA.

    &
    Mark M Rasenick

    † Author for correspondence

    University of Illinois at Chicago College of Medicine, Departments of Physiology & Biophysics & Psychiatry, 835 S Wolcott Ave, M/C 901, Rm E202, Chicago, IL 60612-7342, USA.

    Published Online:https://doi.org/10.2217/14796708.3.5.511

    Despite decades of research, the cellular target(s) of chronic antidepressant treatment remain unknown. For the most part, antidepressant treatments, whether drug or direct stimulation, require repeated administration for approximately a month before achieving clinical benefit, while cognitive therapy typically requires 12–16 weeks not including follow-up visits. Many antidepressant drugs are suggested to increase synaptic monoamine content as they prevent the reuptake or breakdown of monoamines by presynaptic nerve terminals. These actions are contemporaneous with acute drug exposure and it is difficult to correlate these actions with the required chronic treatment. We hypothesize that chronic antidepressant drug treatment reorganizes the synaptic membrane and modifies neurotransmitter signaling and that these effects are relevant to the biology and treatment of depression.

    Receptors for monoamine neurotransmitters are often G protein-coupled, and those that are coupled to G proteins, activate adenylyl cyclase and generate cAMP. Extensive studies on the role of antidepressant treatment and the G–adenylyl cyclase signaling cascade have been reviewed previously [1]. Both in vivo and in vitro studies have demonstrated that chronic, but not acute, antidepressant treatment with a variety of antidepressant compounds leads to an increase in cellular cAMP as well as increased physical coupling between G and adenylyl cyclase. Several mechanisms proposed for chronic antidepressant action are consistent with a long-term increase in cAMP production [1]. Closely related to this is the regulation of cAMP response element binding protein (CREB) levels in depression and by antidepressant treatment. A comprehensive review by Blendy indicates that there is evidence for both increased and decreased levels of CREB and pCREB following chronic treatment with certain antidepressants [2], and a later study discovered that CREB-deficient mice display a ‘depression-resistant’ phenotype [3].

    A concurrent area of study has been on the role of antidepressant treatment on hippocampal neurogenesis. This remains controversial since there are studies suggesting both the importance of neurogenesis in antidepressant action and those stating that antidepressants work independently of neurogenesis. A review by Sahay and Hen covers both the neurogenesis-dependent and the neurogenesis-independent mechanisms leading to the behavioral effects seen by chronic treatment with various antidepressants [4]. It should be noted that while several brain regions are involved in both depression and antidepressant responsiveness, neurogenesis is restricted to the hippocampus. This is one of many details that remains to be explained about neurogenesis and the relationship of that phenomenon to antidepressant action.

    The localization of G proteins to specific membrane domains such as caveolae and lipid rafts has generated interest as to how these cholesterol and sphingolipid-rich, detergent-resistant membrane domains modulate G-protein targeting and function [5–9]. A recent study by Allen et al. suggests that G is targeted to lipid rafts during the process of desensitization and, as such, lipid rafts represent a membrane domain of diminished G–adenylyl cyclase signaling. Conversely, intact caveolae membrane domains appear essential for signaling via certain G-coupled receptors [10]. Thus, lipid raft/caveolae membrane domains appear to be important regulatory domains for neurotransmitter signaling (see [11] for a review) and this has direct clinical relevance in that lipid rafts/caveolae have been implicated in a number of diseases of the nervous system [12].

    Using a fluorescent version of G, we demonstrated that G is internalized through lipid rafts, and the Gthat is sequestered in lipid rafts is not in the pool of G that activates adenylyl cyclase [7]. Rybin et al. have demonstrated that cholesterol depletion increases adenylyl cyclase signaling in cardiac myocytes [13], and we have recently seen the same phenomenon in C6 glioma cells where cholesterol was modified or where lipid rafts/caveolae were disrupted genetically [Allen et al., Unpublished Data]. This suggests that, rather than a wholesale disruption of caveolae or lipid rafts, some specific lipid raft anchor of G is modified in depression or by antidepressant treatment.

    One focus of our research has been on the effects of chronic antidepressant treatment on the localization of G to both Triton X-100 (TX-100)-resistant raft-enriched and TX-100-soluble nonraft membrane domains in cultured cells and in the human brain. These studies have demonstrated that G is localized to TX-100-resistant membrane domains and the tips of elongated processes of C6 glioma cells, and this localization is altered after chronic antidepressant treatment [14–16]. Similarly, we have observed an altered distribution of G in the TX-100-resistant and the TX-100-soluble membrane domains in the prefrontal cortex and cerebellum of depressed suicide subjects compared with control subjects [17]. The distribution of other G proteins, as well as a number of other raft-associated proteins, is not altered in depression or by antidepressant treatment [15,17]. The TX-100-soluble G is more likely to interact with adenylyl cyclase because the TX-100-resistant membrane regions serve as inhibitory domains for G signaling [7,11] and they are much more rigid cholesterol and sphingolipid-rich membrane structures [18]. It should be noted that antidepressant drugs appear to concentrate in lipid rafts [19].

    Dysfunctional cAMP signaling represents a consistent finding in human cerebral cortex tissue from suicide subjects [20–25]. Some of these studies demonstrate a consistent decrease in forskolin-stimulated adenylyl cyclase activity without a change in expression or concentration of G protein [20,25], while other studies have demonstrated a small increase in G protein and mRNA levels in subjects with major depression [21,23]. Downstream in the cAMP signaling pathway, decreased PKA activity was observed in these same suicide subjects [22,24].

    Many studies have employed cells from human blood to model the biochemistry of the brain. Recent studies using human platelets suggest that adenylyl cyclase may be attenuated in depression [26–30]. The most recent of these studies demonstrated markedly lower levels of basal, forskolin-, cesium fluoride- and Gpp(NH)p-stimulated platelet adenylyl cyclase activity in subjects with a history of major depression compared with control subjects [30]. This suggests diminished coupling between G and adenylyl cyclase, consistent with our postmortem results. Furthermore, in both depression and response to antidepressants, we observe a redistribution of Gwithout any major change in the content of that protein. This suggests that a reordering of membrane domains likely accompanies both depression and chronic antidepressant therapy and is consistent with the extended time of treatment required for therapeutic effectiveness.

    Curiously, our findings demonstrated increased lipid raft localization of G in the cerebellum in depression. However, recent behavioral, imaging and biochemical evidence suggests that the cerebellum may be involved in the etiology of major depression. The cerebellum has neuronal connections to brainstem nuclei that supply the limbic system and prefrontal cortex with various monoamines, including serotonin and norepinephrine [31]. In fact, subjects with cerebellar lesions have displayed characteristics such as passivity, blunting of emotion and disinhibition of restraint similar to the depressed and manic states characteristic of mood disorders [32]. Furthermore, the cerebellum is rich in the G-coupled receptors, CRF-1 and -2. The CRF system has been implicated in the relationship between early-life stress and depression [33,34]. Finally, caveolin-1 (the major structural protein component of caveolae) has been identified in the cerebellum of mice, and caveolin-1-knockout mice exhibit neurological abnormalities associated with cortico-striatal, hippocampal and cerebellar regions [35].

    In summary, previous studies in both rats and C6 glioma cells demonstrated that chronic antidepressant treatment liberates G, but not G or G, from the inhibitory TX-100-resistant membrane raft domains [14–16], increasing its association with adenylyl cyclase [16]. Coupled with postmortem human brain data, these observations demonstrate that the increased localization of G in the TX-100-resistant membrane domains of depressed individuals may prevent G from associating with and activating adenylyl cyclase, thus preventing the propagation of the cAMP signal. These findings are supported by recent evidence suggesting that antidepressant drugs concentrate in raft-like plasma membrane domains [19]. The intercalation of these drugs may physically inhibit the localization of G to the raft domains. Thus, it appears that antidepressants may exert their observed effects on cAMP signaling by liberating G from TX-100-resistant membrane domains, where it accumulates during the course of depression. Furthermore, the ratio of TX-100-soluble to TX-100-resistant G may prove to be a useful biomarker for human depression and a rapid (3–4 days) harbinger of effective response to antidepressant therapy. Finally, revelation of new mechanisms for depression and new targets for antidepressant drugs may lead to the development of novel classes of therapeutic agents.

    Financial & competing interests disclosure

    The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

    No writing assistance was utilized in the production of this manuscript.

    Bibliography

    • Donati RJ, Rasenick MM: G protein signaling and the molecular basis of antidepressant action. Life Sci.73,1–17 (2003).
    • Blendy JA: The role of CREB in depression and antidepressant treatment. Biol. Psych.59,1144–1150 (2006).
    • Gur TL, Conti AC, Holden J et al.: cAMP response element-binding protein deficiency allows for increased neurogenesis and a rapid onset of antidepressant response. J. Neurosci.27,7860–7868 (2007).
    • Sahay A, Hen R: Adult hippocampal neurogenesis in depression. Nat. Neurosci.10,1110–1115 (2007).
    • Bayewitch ML, Nevo I, Avidor-Reiss T et al.: Alterations in detergent solubility of heterotrimeric G proteins after chronic activation of Gi/o-coupled receptors: changes in detergent solubility are in correlation with onset of adenylyl cyclase superactivation. Mol. Pharmacol.57,820–825 (2000).
    • Brown DA: Lipid rafts, detergent-resistant membranes, and raft targeting signals. Physiology (Bethesda)21,430–439 (2006).
    • Allen JA, Yu JZ, Donati RJ, Rasenick MM: β-adrenergic receptor stimulation promotes Gαs internalization through lipid rafts: a study in living cells. Mol. Pharmacol.67,1493–1504 (2005).
    • Moffett S, Brown DA, Linder ME: Lipid-dependent targeting of G proteins into rafts. J. Biol. Chem.275,2191–2198 (2000).
    • Ostrom RS, Insel PA: The evolving role of lipid rafts and caveolae in G protein-coupled receptor signaling: implications for molecular pharmacology. Br. J. Pharmacol.143,235–245 (2004).
    • 10  Bhatnagar A, Sheffler DJ, Kroeze WK, Compton-Toth B, Roth BL: Caveolin-1 interacts with 5-HT2A serotonin receptors and profoundly modulates the signaling of selected Gαq-coupled protein receptors. J. Biol. Chem.279,34614–34623 (2004).
    • 11  Allen JA, Halverson-Tamboli RA, Rasenick MM: Lipid raft microdomains and neurotransmitter signalling. Nat. Rev. Neurosci.8,128–140 (2007).
    • 12  Benarroch EE: Lipid rafts, protein scaffolds, and neurologic disease. Neurology69,1635–1639 (2007).
    • 13  Rybin VO, Xu X, Lisanti MP, Steinberg SF: Differential targeting of β-adrenergic receptor subtypes and adenylyl cyclase to cardiomyocyte caveolae. A mechanism to functionally regulate the cAMP signaling pathway. J. Biol. Chem.275,41447–41457 (2000).
    • 14  Donati RJ, Thukral C, Rasenick MM: Chronic treatment of C6 glioma cells with antidepressant drugs results in a redistribution of G. Mol. Pharmacol.59,1426–1432 (2001).
    • 15  Donati RJ, Rasenick MM: Chronic antidepressant treatment prevents accumulation of gsα in cholesterol-rich, cytoskeletal-associated, plasma membrane domains (lipid rafts). Neuropsychopharmacology30,1238–1245 (2005).
    • 16  Toki S, Donati RJ, Rasenick MM: Treatment of C6 glioma cells and rats with antidepressant drugs increases the detergent extraction of G from plasma membrane. J. Neurochem.73,1114–1120 (1999).
    • 17  Donati RJ, Dwivedi Y, Roberts RC et al.: Postmortem brain tissue of depressed suicides reveals increased G localization in lipid raft domains where it is less likely to activate adenylyl cyclase. J. Neurosci.28,3042–3050 (2008).
    • 18  Brown DA, London E: Structure and function of sphingolipid- and cholesterol-rich membrane rafts. J. Biol. Chem.275,17221–17224 (2000).
    • 19  Eisensamer B, Uhr M, Meyr S et al.: Antidepressants and antipsychotic drugs colocalize with 5-HT3 receptors in raft-like domains. J. Neurosci.25,10198–10206 (2005).
    • 20  Dowlatshahi D, MacQueen GM, Wang JF, Reiach JS, Young LT: G protein-coupled cyclic AMP signaling in postmortem brain of subjects with mood disorders: effects of diagnosis, suicide, and treatment at the time of death. J. Neurochem.73,1121–1126 (1999).
    • 21  Dwivedi Y, Rizavi HS, Conley RR et al.: mRNA and protein expression of selective α subunits of G proteins are abnormal in prefrontal cortex of suicide victims. Neuropsychopharmacology27,499–517 (2002).
    • 22  Dwivedi Y, Rizavi HS, Shukla PK et al.: Protein kinase A in postmortem brain of depressed suicide victims: altered expression of specific regulatory and catalytic subunits. Biol. Psych.55,234–243 (2004).
    • 23  Pacheco MA, Stockmeier C, Meltzer HY et al.: Alterations in phosphoinositide signaling and G-protein levels in depressed suicide brain. Brain Res723,37–45 (1996).
    • 24  Pandey GN, Dwivedi Y, Ren X et al.: Brain region specific alterations in the protein and mRNA levels of protein kinase A subunits in the post-mortem brain of teenage suicide victims. Neuropsychopharmacology30,1548–1556 (2005).
    • 25  Stewart RJ, Chen B, Dowlatshahi D, MacQueen GM, Young LT: Abnormalities in the cAMP signaling pathway in post-mortem brain tissue from the Stanley Neuropathology Consortium. Brain Res. Bull.55,625–629 (2001).
    • 26  Menninger JA, Tabakoff B: Forskolin-stimulated platelet adenylyl cyclase activity is lower in persons with major depression. Biol. Psych.42,30–38 (1997).
    • 27  Menninger JA, Baron AE, Conigrave KM et al.: Platelet adenylyl cyclase activity as a trait marker of alcohol dependence. WHO/ISBRA Collaborative Study Investigators. International Society for Biomedical Research on Alcoholism. Alcohol Clin. Exp. Res.24,810–821 (2000).
    • 28  Mooney JJ, Samson JA, McHale NL et al.: Signal transduction by platelet adenylate cyclase: alterations in depressed patients may reflect impairment in the coordinated integration of cellular signals (coincidence detection). Biol. Psych.43,574–583 (1998).
    • 29  Hoffman PL, Glanz J, Tabakoff B: Platelet adenylyl cyclase activity as a state or trait marker in alcohol dependence: results of the WHO/ISBRA Study on State and Trait Markers of Alcohol Use and Dependence. Alcohol Clin. Exp. Res.26,1078–1087 (2002).
    • 30  Hines LM, Tabakoff B: Platelet adenylyl cyclase activity: a biological marker for major depression and recent drug use. Biol. Psych.58,955–962 (2005).
    • 31  Schmahmann JD: Disorders of the cerebellum: ataxia, dysmetria of thought, and the cerebellar cognitive affective syndrome. J. Neuropsychiatry Clin. Neurosci.16,367–378 (2004).
    • 32  Schmahmann JD, Sherman JC: The cerebellar cognitive affective syndrome. Brain121(Pt 4),561–579 (1998).
    • 33  Arborelius L, Owens MJ, Plotsky PM, Nemeroff CB: The role of corticotropin-releasing factor in depression and anxiety disorders. J. Endocrinol160,1–12 (1999).
    • 34  Owens MJ, Nemeroff CB: Corticotropin-releasing factor antagonists in affective disorders. Expert Opin. Investig. Drugs8,1849–1858 (1999).
    • 35  Trushina E, Du Charme J, Parisi J, McMurray CT: Neurological abnormalities in caveolin-1 knock out mice. Behav. Brain Res.172,24–32 (2006).