Next Article in Journal
Administration of Epidermal Growth Factor (EGF) and Basic Fibroblast Growth Factor (bFGF) to Induce Neural Differentiation of Dental Pulp Stem Cells (DPSC) Isolates
Next Article in Special Issue
GABA and Combined GABA with GAD65-Alum Treatment Alters Th1 Cytokine Responses of PBMCs from Children with Recent-Onset Type 1 Diabetes
Previous Article in Journal
The Relationship between the Expression of GATA4 and GATA6 with the Clinical Characteristics and Prognosis of Resectable Pancreatic Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19

Department of Molecular and Medical Pharmacology, UCLA School of Medicine, University of California, Los Angeles, CA 90095-1735, USA
*
Authors to whom correspondence should be addressed.
Biomedicines 2023, 11(2), 254; https://doi.org/10.3390/biomedicines11020254
Submission received: 16 December 2022 / Revised: 16 January 2023 / Accepted: 16 January 2023 / Published: 18 January 2023

Abstract

:
GABA and GABAA-receptors (GABAA-Rs) play major roles in neurodevelopment and neurotransmission in the central nervous system (CNS). There has been a growing appreciation that GABAA-Rs are also present on most immune cells. Studies in the fields of autoimmune disease, cancer, parasitology, and virology have observed that GABA-R ligands have anti-inflammatory actions on T cells and antigen-presenting cells (APCs), while also enhancing regulatory T cell (Treg) responses and shifting APCs toward anti-inflammatory phenotypes. These actions have enabled GABAA-R ligands to ameliorate autoimmune diseases, such as type 1 diabetes (T1D), multiple sclerosis (MS), and rheumatoid arthritis, as well as type 2 diabetes (T2D)-associated inflammation in preclinical models. Conversely, antagonism of GABAA-R activity promotes the pro-inflammatory responses of T cells and APCs, enhancing anti-tumor responses and reducing tumor burden in models of solid tumors. Lung epithelial cells also express GABA-Rs, whose activation helps maintain fluid homeostasis and promote recovery from injury. The ability of GABAA-R agonists to limit both excessive immune responses and lung epithelial cell injury may underlie recent findings that GABAA-R agonists reduce the severity of disease in mice infected with highly lethal coronaviruses (SARS-CoV-2 and MHV-1). These observations suggest that GABAA-R agonists may provide off-the-shelf therapies for COVID-19 caused by new SARS-CoV-2 variants, as well as novel beta-coronaviruses, which evade vaccine-induced immune responses and antiviral medications. We review these findings and further advance the notions that (1) immune cells possess GABAA-Rs to limit inflammation in the CNS, and (2) this natural “braking system” on inflammatory responses may be pharmacologically engaged to slow the progression of autoimmune diseases, reduce the severity of COVID-19, and perhaps limit neuroinflammation associated with long COVID.

1. Introduction

GABA and GABA-Rs play key roles in neurotransmission and neurodevelopment in the central nervous system (CNS) and have been the subject of intensive investigations for decades by neuroscientists. Only relatively recently have the presence and functions of GABA-Rs on immune cells become evident. There are a growing number of studies reporting that the activation of GABA-Rs on human and rodent immune cells limits their pro-inflammatory activities while also promoting Treg responses and shifting APC toward anti-inflammatory phenotypes. Thereby, GABA-Rs are a natural immune cell braking system that has been pharmacologically targeted to ameliorate inflammatory disorders in preclinical models. Here, we review GABA-R physiological functions in the CNS, GABA-R-based therapeutics for autoimmune diseases and inflammatory disorders along with its currently understood mechanisms of action, GABA’s role as an immunosuppressant within some solid tumors, and recent observations of GABA-R agonists’ potential as therapeutics for COVID-19. Finally, we further advance the hypotheses that T cells express GABAA-Rs to limit inflammation in the CNS and that GABAA-R agonists could aid in the treatment of CNS inflammatory disorders including MS and neuroinflammation associated with long-COVID.

2. GABA and GABA-Receptors and Their Physiological Functions in the CNS

In vertebrate neurons, GABA is synthesized through the decarboxylation of glutamate by two glutamic acid decarboxylases (GAD65 and GAD67). These two isoforms of GAD are highly homologous but have distinct kinetic properties, cofactor requirements, and subcellular locations, allowing the fine-tuning of GABA production with neuronal activity and cellular metabolism [1,2,3,4,5]. There are two types of GABA-Rs that are evolutionarily and functionally unrelated: GABAA-Rs and GABAB-Rs. GABAA-Rs are pentamers of 19 possible subunits that form fast-acting chloride channels, which when activated let chloride passively flow in or out of the cell. The direction of Cl flux depends on the equilibrium potential of Cl relative to the cell membrane potential, which in turn is modulated by the activity of the cell’s Cl transporters [6,7,8]. The influx/efflux of Cl through GABAA-Rs modulates the levels of intracellular ions and consequently many signaling pathways involved in neurodevelopment and neurotransmission [6,7,8]. Different combinations of GABAA-R subunits form GABAA-Rs with varying pharmacological sensitivities and kinetics. The GABAB-Rs are heterodimers that form a slow-acting G-protein-coupled receptor that modulates adenylyl cyclase activity as well as potassium and calcium channels [9,10].
GABAA-Rs and GABAB-Rs have different pharmacologically targetable sites which can modulate their sensitivity and function [6,10]. Orally consumed GABA has little or no ability to pass through the blood–brain barrier (BBB) [11,12,13,14]. Therefore, many BBB-permeable GABAA-Rs and GABAB-Rs agonists and positive allosteric modulators (PAMs), such as benzodiazepines and barbiturates, were developed and have provided first-line treatments for neurological and neuropsychiatric disorders including seizures, anxiety, schizophrenia, and insomnia [6,8]. The neurological activity of these BBB-permeable drugs is an inherent drawback to their use when seeking to target peripheral GABA-Rs. In contrast, oral GABA can modulate peripheral GABA-Rs without CNS side effects. Additionally, while the GABAA-R-specific agonist homotaurine is BBB-permeable, it had no adverse CNS effects when tested in a large phase III clinical trial for Alzheimer’s disease and may also be an attractive candidate for modulating GABAA-Rs in both the periphery and the CNS, as discussed below.

3. GABA-R-Based Therapies for T Cell-Mediated Autoimmune Diseases and Other Inflammatory Disorders

In 1999, GABA was reported to inhibit T cell proliferation in vitro and that this was mediated through GABAA-Rs, but not GABAB-Rs. In vivo, GABA administration modestly limited classical Th1-mediated delayed-type hypersensitivity (DTH) responses [15]. The anti-inflammatory effect of GABA was mediated at least in part through inhibiting TCR-mediated T cell cycling without affecting cell viability [16]. It was proposed that because of the relatively high concentration of intracellular Cl in T cells, the activation of T cell GABAA-R Cl channels leads to Cl efflux, depolarization, and limitation of Ca2+ entry into the cell to reduce intracellular calcium levels, thereby reducing T cell proliferation [15,16], a notion that has been supported by studies of Ca2+ flux [17,18,19,20].
The observations that GABA could limit T cell proliferation in vitro and DTH responses in vivo suggested that GABA administration could help abate autoimmune processes in which autoreactive Th1 and/or Th17 cells mediate tissue damage. Indeed, a growing number of studies have now shown that treatment with GABA or the GABAA-R agonist homotaurine can inhibit the progression of a diverse array of T cell-mediated autoimmune diseases that occur in rodents with different genetic backgrounds, including models of multiple sclerosis (MS), type 1 diabetes (T1D), rheumatoid arthritis, Sjogren’s syndrome, as well as inflammation in type 2 diabetes [16,20,21,22,23,24,25,26,27]. Mechanistic studies revealed that treatment with a GABAA-R agonist in these disease models inhibited the inflammatory activities of rodent and human Th17 and Th1 CD4+ T cells and cytotoxic CD8+ T cells [15,16,18,19,23,24,25,28,29] and also promoted CD4+ and CD8+ Treg responses [20,22,23,24,28] (Figure 1). These differential outcomes may be due in part to Th1 and Th17 cells having the greatest dependency on Ca2+ influx for their activation and effector functions, while Tregs display the least dependency on Ca2+ influx [30,31,32,33,34,35]. Long-term GABA treatment did not desensitize antigen-specific T cell responses to cognate antigen upon re-exposure [21], indicating that GABA treatment did not exhaust pre-existing antigen-specific memory T cells.
While early studies of GABA-Rs in the immune system focused on T cells and autoimmune diseases, it became increasingly apparent that cells of the innate immune system also expressed GABAA-Rs and GABAB-Rs that modulated their function. As observed in the adaptive immune system, the activation of GABAA-Rs on macrophages, dendritic cells (DC), and NK cells of the innate immune system reduces their inflammatory activities and shifts them toward anti-inflammatory phenotypes [19,21,25,27,36,37,38,39,40,41]. Moreover, GABAB-R agonists were found to (1) inhibit murine DC activation and immune cell chemotaxis [42,43], (2) inhibit DC proinflammatory functions [42], (3) suppress antigen presentation activity of DCs to alleviate collagen-induced arthritis and contact dermatitis in mouse models [42,43], (4) inhibit the destruction of insulin-producing ß-cells in T1D-prone mice [44,45], and (5) attenuate TLR4-induced inflammatory signaling in human peripheral blood mononuclear cells (PBMCs) [46]. Although classical neuronal GABAB-Rs are heterodimers of GABAB1 and GABAB2 subunits, human immune cells express relatively high levels of the GABAB1 gene but little or no GABAB2 transcripts [45,47]. There is growing evidence that GABAB1 subunits have functional activities independent of GABAB2 subunits [48,49,50,51], but its mechanisms have not been elucidated.
A major factor driving relapses of human MS and experimental autoimmune encephalomyelitis (EAE, a model of MS) is thought to be the spreading of T cell autoreactivity to new myelin T cell determinants within the CNS [52,53,54,55]. While oral GABA administration did not affect EAE disease progression, presumably because it has little or no ability to pass through the BBB, treatment with the BBB-permeable GABAA-R-specific agonist homotaurine effectively induced disease remission in mice with advanced relapsing-remitting EAE [24,25]. Mechanistically, homotaurine treatment limited the spreading of T cell autoreactivities to new myelin antigen determinants, decreased autoreactive Th17 and Th1 responses in the CNS, and also enhanced CD4+ and CD8+ regulatory T cell responses [24,25]. The remission of EAE may have also involved the attenuation of the inflammatory activities of microglia, astrocytes, and inflammatory infiltrates in the CNS, which also possess GABAA-Rs and contribute to EAE pathogenesis [56,57,58,59,60,61]. In other studies of rats with an autism spectrum-like disorder caused by prenatal exposure to valproic acid [62], homotaurine treatment reduced the levels of IL-1ß, IL-6, and TNF-α, but increased the levels of IL-10 in the CNS, decreased neuronal loss, and ameliorated behavioral deficits [63].
In large drug screening assays, homotaurine was found to physically interfere with amyloid aggregation in vitro, making it a candidate treatment for Alzheimer’s disease. In a large phase III clinical study with Alzheimer’s patients, homotaurine (also known as Tramiprosate) treatment over 1.5 years failed to meet primary endpoints, but the treatment appeared to be very safe [64,65,66]. A follow-up MRI study of a subgroup of these patients indicated that homotaurine treatment slowed hippocampal atrophy with some evidence of a beneficial effect on cognition [67]. A recent small clinical study of individuals with mild cognitive impairment found that those who were given homotaurine for 12 months had reduced circulating levels of the proinflammatory cytokine IL-18 but increased levels of IL-10 and IL-33, and improved short-term memory performance [68]. Hence, homotaurine appears to be a promising candidate for testing in clinical trials for MS as well as other disorders in which inflammation in the CNS contributes to disease pathogenesis.
Recent metabolomic studies of murine T cell development have found that GABA also modulates murine T cell differentiation [69]. Depending on the context, intracellularly synthesized GABA can enter the TCA cycle to enhance bioenergetic capacity and the differentiation of Th17 cells, or GABA may be secreted where it acts in an autocrine manner to promote inducible Treg (iTreg) differentiation [69].
GABA also inhibits the secretion of pro-inflammatory cytokines and chemokines by human immune cells in vitro [19,70]. Consistent with those findings, in a small clinical trial (NCT02002130), treatment of newly diagnosed T1D children with GABA reduced their PBMC responses to a ß-cell autoantigen compared to PBMC from placebo-treated children over the 12-month study (Dr. Hubert Tse, University of Alabama at Birmingham, personal communication). Therefore, GABA-R agonists may be promising candidate therapeutics for human T cell-mediated autoimmune diseases.

4. Are T Cell GABAA-Rs a Braking System against T-Cell-Mediated Inflammation in the CNS? Lessons from Multiple Sclerosis Models

The presence and anti-inflammatory activities of GABAA-Rs on T cells raise basic questions as to (1) why T cells evolved to express receptors for this neurotransmitter, (2) what purpose do GABAA-Rs on T cells serve in states of health, and (3) where are T cells modulated by endogenous GABA? Given that the CNS is by far the major site of GABA production and secretion, we hypothesized that GABAA-Rs on T cells may be a natural braking system against inflammation in the CNS [16,24,25].
The vesicular release of GABA from presynaptic neurons in the CNS leads briefly to levels of ≈1 mM GABA within the synaptic cleft [71,72,73]. The GABAA-R subtypes on the postsynaptic membrane are comprised of subunits that confer a relatively low affinity for GABA. This allows the GABA to rapidly dissociate from the GABAA-Rs and be rapidly taken into cells again by transporters so that the postsynaptic neuron can quickly respond to the next release of GABA from presynaptic terminals. Some GABA can spill over from the synaptic cleft and GABA is also released from (1) neurons in a non-vesicular action potential-independent fashion from extrasynaptic sites and (2) glia [60,61,74], such that the extracellular GABA levels in the CNS are thought to be in the nanomolar to low micromolar range [75,76,77]. Notably, the GABAA-R subtypes on cells in the periphery, such as T cells, are composed of subunits that confer a high affinity for GABA (in the nanomolar range) [78,79,80,81,82,83,84]. Accordingly, the levels of extracellular GABA in the CNS should be sufficient to activate the high-affinity GABAA-Rs on CNS-infiltrating T cells.
The notion that GABA in the CNS may limit the inflammatory activities of infiltrating activated T cells is supported by the observation that GABAA-R-deficient myelin-specific T cells mediated a more severe EAE than their GABAA-R-expressing counterparts [69]. This suggests that extracellular CNS GABA had some inhibitory effect on infiltrating GABAA-R-expressing encephalitogenic T cells; however, the degree of inhibition was insufficient to prevent the robust experimentally induced autoreactive T cell responses from mediating EAE, although it was less severe. The aforementioned findings that administering the BBB-permeable GABAA-R agonist homotaurine, but not GABA (BBB-impermeable), led to EAE remission [24,25] indicate that pharmacological augmentation of the endogenous GABAA-R braking system is needed to control this acute neuroinflammatory disease. EAE remission may have also involved homotaurine-mediated diminution of the inflammatory activities of microglia, astrocytes, and other inflammatory infiltrates in the CNS, all of which express GABAA-Rs and contribute to EAE pathogenesis [56,57,58,59,60,61]. Together, these observations suggest that endogenous GABA acts as a natural braking system on inflammation in the CNS and that this can be pharmacologically augmented.

5. GABA Inhibits Tumor-Infiltrating Immune Responses and Modulates Tumor Cell Proliferation

Recent studies have made the surprising finding that B cells (but not T cells) express GAD67 and secrete GABA [19]. The B cell-secreted GABA was found to inhibit antitumor responses by suppressing the infiltration and activity of cytotoxic T cells and macrophages. Tumors within B cell-deficient mice had more tumor-infiltrating CD8+ T cells with enhanced cytotoxic and inflammatory marker expression. Moreover, treatment of wildtype mice with a GABAA-R-specific antagonist significantly increased the cytotoxic activity of tumor-infiltrating CD8+ T cells and inflammatory marker expression in tumor-associated macrophages. Tumors implanted into mice with GAD67-deficient B cells had reduced growth, and the tumor-infiltrating CD8+ T cells had greater cytotoxicity and stronger pro-inflammatory properties than did CD8+ T cells isolated from tumors in wildtype animals [19]. These findings suggest that pharmacological inhibition of T cell GABAA-Rs, or the adoptive transfer of GABAA-R-deficient tumor-reactive T cells or APCs may enhance the efficacy of anti-cancer treatments [19,85,86]. These observations also lend support to the notion that extracellular GABA in the CNS may contribute to the immunosuppressive environment.
The activation of GABA-Rs and GABAA-R PAMs has also been found to modulate the metabolism and signaling pathways of some tumor cells, resulting in the inhibition or promotion of their replication and migration. The particular outcome of GABA-R activation on tumor cell proliferation and migration has varied depending on the type of tumor and the type of GABA-R and its subunit composition (recently reviewed in [87]). Benzodiazepine use has been epidemiologically associated with increased risk for some cancers [88,89]; however, care should be taken in interpreting those findings since some benzodiazepines, such as diazepam, have high affinity for the mitochondrial translocator protein (TSPO), previously known as the “peripheral benzodiazepine-binding receptor,” which is widely expressed and modulates many cellular processes [90].
A recent study found that some tumor cells express GAD67 and secrete GABA [85]. This tumor-secreted GABA can (1) activate GABAB-Rs on tumor cells, which facilitates GSK-3β inactivation and leads to enhanced β-catenin signaling and tumor growth, and (2) suppresses tumor-infiltrating CD8+ T cells [85]. The above studies demonstrate that the GABA/GABA-R system can help tumor cells escape from immune surveillance in different types of solid tumors. Accordingly, the targeting of GAD67/GAD65 or specific GABA-R subunits might be of therapeutic value for limiting the progression of these types of tumors. We also speculate that the ability of glioblastomas and neuroblastomas to produce and secrete GABA ([60,91,92,93,94,95,96,97], and our unpublished observations) could contribute to tumor cell proliferation and the immunosuppressive environment in these types of malignant tumors.

6. GABA-R Agonists Modulate Airway Epithelial Cells and Pulmonary Immune Cells

Both GABAA-R and GABAB-R expression has been observed in rodent and human lung epithelia [98,99,100]. Although there are some variations in observations between studies, it appears that rodent and human airway epithelial cells express GAD67 (primarily) and GAD65, with GAD67 found in alveolar epithelial type II (ATII) but not ATI cells [98,101,102]. GABAA-Rs have been demonstrated on rat ATII cells electrophysiologically, which acted to increase Cl efflux [102].
GABA and GABAA-R PAMs reduce inflammation and improve alveolar fluid clearance and recovery of lung function in different rodent models of acute lung injury [100,103,104,105,106,107,108,109]. Likewise, GABAA-R PAMs reduce pulmonary inflammation and improve clinical outcomes in ventilated human patients [110,111,112]. Of potential relevance to COVID-19 treatment, GABA decreases the secretion of inflammatory factors from cultured human bronchial epithelial cells [104] and GABAA-R PAMs can reduce the numbers of macrophages and the levels of inflammatory cytokines in bronchoalveolar lavage fluid, as well as rodent and human macrophage inflammatory responses [36,37,108,113,114,115,116]. GABAA-R ligands also inhibit (1) the activation of human neutrophils and neutrophil extracellular trap (NET) formation [117,118,119] and (2) platelet aggregation [120], both of which contribute to pulmonary thrombosis.

7. GABAA-Rs Agonists Are Effective Therapeutics in Preclinical Models of COVID-19

The ability of GABAA-R agonists and potentiators to limit inflammation and acute lung injury in vivo suggests that they have the potential to reduce some of the major contributors to the development of severe COVID-19. On the other hand, their anti-inflammatory activities may limit key antiviral responses and exacerbate the severity of the infection.
Murine Hepatitis Virus Strain 1 (MHV-1) studies. Like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), MHV-1 is a beta coronavirus and was used as a model of SARS-CoV infection [121,122,123,124]. Wildtype A/J mice infected with MHV-1 develop severe pneumonitis with high mortality [121,122,123,124]. We observed that oral GABA treatment just after MHV-1 inoculation, or 3 days later when clinical symptoms had begun to appear, protected 80–90% of virus-infected mice from developing severe disease [125]. Additionally, GABA treatment modestly reduced viral load in the lungs and greatly reduced lung inflammation in mice [125]. Follow-up studies using the GABAA-R-specific agonist homotaurine, or the GABAB-R-specific agonist baclofen, revealed that homotaurine, but not baclofen, was as effective as GABA in preventing severe disease and limiting pneumonitis, indicating that GABAA-Rs primarily mediated GABA’s therapeutic benefits in MHV-1 infected mice [125].
SARS-CoV-2 studies. Because MHV-1 and SARS-CoV-2 bind to different cellular receptors (CEACAM1 and ACE2, respectively) and the immune response to virus infections varies greatly depending on the virus strain [126], we sought to more stringently test the therapeutic potential of GABA as a therapy for COVID-19. We therefore studied transgenic K18-hACE2 mice which express human ACE2. Following infection with SARS-CoV-2, K18-hACE2 mice develop severe pneumonitis, providing a widely used acute and lethal model of COVID-19 [127,128,129,130,131,132]. We found that oral GABA administration beginning just after SARS-CoV-2 infection, or a few days later near the peak of lung viral load, reduced disease severity, pneumonitis, and death rates in K18-hACE2 mice [133]. Notably, GABA-treated mice had reduced viral loads in their lungs (23-fold lower) and displayed shifts in circulating cytokine and chemokine levels that are associated with better outcomes in COVID-19 patients [133]. Thus, administration of this GABA-R agonist had multiple beneficial effects that are also desirable for the treatment of COVID-19 (Figure 1).
The ability of GABA treatment to modestly reduce viral loads in both MHV-1 and SARS-CoV-2-infected mice was unexpected. The activation of GABAA-Rs leads to the influx or efflux of Cl depending on the equilibrium potential of Cl relative to the cell membrane potential. Consequently, activating GABAA-Rs on adult neurons or islet α-cells leads to Cl influx and hyperpolarization, while activation of GABAA-Rs on T cells, islet ß-cells, and lung ATII cells, causes Cl efflux and depolarization [17,18,19,102,115,134,135]. Coronaviruses promote Ca2+ influx into host cells to enhance their replication [136,137]. Conceivably, the activation of GABAA-Rs on lung epithelial cells leads to Cl efflux and membrane depolarization, which limits the influx of extracellular Ca2+ and reduces intracellular calcium levels, making the intracellular environment less conducive to viral replication. This scenario, as well as possible GABA-R-mediated changes in virus receptor levels, surfactant production/absorption, and/or inflammatory responses and autophagy, warrants further investigations (Table 1).
Figure 1. The effects of the GABA and GABA-R system on inflammatory responses in different disease states. GABAA-R agonists (1) inhibit pathogenic/effector Th1, Th17, and CD8+ T cell responses, reducing the production of IFNγ, IL-17, and other pro-inflammatory mediators, (2) limit APC inflammatory activities and shift them toward anti-inflammatory phenotypes, and (3) promote CD4+ and CD8+ Treg responses to enhance the production of anti-inflammatory IL-10 and other mediators. Moreover, GABAB-R agonists inhibit murine APC activation and proinflammatory functions. These actions limit the pathogenesis of T-cell-mediated autoimmune diseases, such as T1D, MS, rheumatoid arthritis, and Sjogren’s syndrome, as well as inflammation in type 2 diabetes, in preclinical models. Within certain types of tumors, these actions attenuate antitumor responses. Furthermore, GABAA-Rs agonists limit lung inflammation, virus replication, and inflammatory cytokine/chemokine responses to reduce the severity of disease and mortality in MHV-1 and SARS-CoV-2 mouse models of COVID-19. Therefore, besides the roles GABA and GABA-Rs play in neurotransmission in the CNS and in regulating pancreatic islet cell functions, they also act as brakes on inflammatory immune responses and may be a therapeutic target for the intervention of inflammatory diseases.
Figure 1. The effects of the GABA and GABA-R system on inflammatory responses in different disease states. GABAA-R agonists (1) inhibit pathogenic/effector Th1, Th17, and CD8+ T cell responses, reducing the production of IFNγ, IL-17, and other pro-inflammatory mediators, (2) limit APC inflammatory activities and shift them toward anti-inflammatory phenotypes, and (3) promote CD4+ and CD8+ Treg responses to enhance the production of anti-inflammatory IL-10 and other mediators. Moreover, GABAB-R agonists inhibit murine APC activation and proinflammatory functions. These actions limit the pathogenesis of T-cell-mediated autoimmune diseases, such as T1D, MS, rheumatoid arthritis, and Sjogren’s syndrome, as well as inflammation in type 2 diabetes, in preclinical models. Within certain types of tumors, these actions attenuate antitumor responses. Furthermore, GABAA-Rs agonists limit lung inflammation, virus replication, and inflammatory cytokine/chemokine responses to reduce the severity of disease and mortality in MHV-1 and SARS-CoV-2 mouse models of COVID-19. Therefore, besides the roles GABA and GABA-Rs play in neurotransmission in the CNS and in regulating pancreatic islet cell functions, they also act as brakes on inflammatory immune responses and may be a therapeutic target for the intervention of inflammatory diseases.
Biomedicines 11 00254 g001

8. Could Homotaurine’s Anti-Inflammatory Effects in the Periphery and in the CNS Help Limit the Neurological Sequelae of COVID-19?

Many COVID-19 patients suffer from long-term neurological symptoms. These post-acute sequelae of COVID-19 frequently involve cognitive impairments (“brain fog”), difficulty concentrating, anxiety, and depression. Histological studies of brains obtained from COVID-19 patients have noted immune cell infiltrates and increased frequencies of glial cells with inflammatory phenotypes, which are indicative of neuroinflammatory responses [138,139,140,141,142,143,144,145]. Currently, there are no specific treatments for this condition.
We speculate that homotaurine is a good candidate for clinical trials with individuals suffering from neurological sequelae of COVID-19 based on (1) the ability of homotaurine to inhibit autoreactive T cell responses and ameliorate advanced EAE [24,25], (2) microglia and astrocytes also express GABAA-Rs, which act to downregulate their inflammatory activities [60], (3) GABA-R agonists protected MHV-1and SARS-CoV-2-infected mice from severe illness and reduced the levels of circulating inflammatory cytokines that might enter the CNS [125], (4) homotaurine treatment decreased proinflammatory and increased anti-inflammatory cytokine profiles in the CNS, preserved neurons, and ameliorated behavioral deficits in rats with autism spectrum-like disorder [63], and (5) Alzheimer’s patients who were given homotaurine had reduced levels of circulating inflammatory cytokines and improved short-term memory performance [68]. Thus, homotaurine has the potential to reduce dysregulated immune responses to SARS-CoV-2, reduce viral loads, and act directly in the CNS to limit the inflammatory activities of glial cells and infiltrating immune cells. As noted above, homotaurine was found to be very safe in a large long-term phase III clinical study with Alzheimer’s patients [64,65,66,67].

9. Prospects for GABA-R-Based Therapies in Clinical Applications

GABA seems well suited as a preventive or interventive therapy for new-onset T1D given its effectiveness in many studies of T1D rodents, its anti-inflammatory effects on human immune cells in vitro, and the results of a small clinical trial, as discussed above. Moreover, the combination of GABA treatment with other types of T1D therapeutic approaches such as antigen-specific immunotherapy or immunosuppressive agents yields enhanced beneficial effects over those of either monotherapy in diabetic mice [23,28,45,146,147,148,149,150]. Although not discussed herein, GABA treatment can also promote the proliferation of human and rodent insulin-producing ß-cells in diabetic animals (reviewed in [151]).
Second, the ability of homotaurine to (1) inhibit neuroinflammation in animal models of MS and autism, and (2) reduce circulating levels of IL-18 in a small clinical trial with Alzheimer’s patients, together with its excellent safety profile in a phase III Alzheimer’s disease clinical trial, makes homotaurine a promising candidate to be an adjunctive treatment for MS. The advancement of GABA and homotaurine toward clinical trials for autoimmune diseases, however, faces challenges since these compounds are available over the counter in many countries, which limits pharmaceutical companies’ interest in their further development.
It is widely opined that additional therapeutic approaches are urgently needed for the treatment of coronavirus infections because new SARS-CoV-2 variants are sure to arise, some of which may evade vaccine-induced immune responses or resist antiviral medications. Our observations in mice infected with two very different types of coronaviruses suggest that GABA treatment could be a generalizable off-the-shelf inexpensive treatment to help reduce the severity of illness caused by new SARS-CoV-2 variants and novel coronaviruses. The human equivalents of the doses of GABA and homotaurine that were used in the preclinical studies of autoimmune diseases and COVID-19 are within the levels already deemed to be safe for human use. Since GABA and homotaurine are stable at room temperature and inexpensive, they are promising candidates to help treat COVID-19, especially in developing countries. Perhaps GABAA-R ligands will augment the therapeutic efficacy of other treatments for COVID-19. We also speculate that homotaurine, or other BBB-permeable GABAA-R-specific ligands, may limit the neurological sequelae of COVID-19. It must be emphasized, however, that these contentions are largely based on results from preclinical models. The impact of GABA-R agonists on human immune cells, lung cells, and cells in the CNS differs in important ways from that of rodents. Indeed, the ability of GABA-R ligands to limit inflammatory immune responses could interfere with immune responses against SARS-CoV-2 and exacerbate COVID-19. Therefore, careful clinical trials are needed to ascertain whether, and at what stage of the disease process, treatment with a GABA-R agonist might be beneficial for COVID-19 patients.
Table 1. Outstanding Questions.
Table 1. Outstanding Questions.
How do GABAA-R and GABAB-R signaling pathways modulate intracellular signaling pathways and metabolome in different types of immune cells?
How does GABA act in paracrine and autocrine fashions on different immune cell functions?
Will tumor-reactive T cells that have been engineered to be GABA-R-deficient have a greater ability to destroy certain types of tumors?
What roles do GABA and GABA-Rs play in the progression of glioblastomas and neuroblastomas?
How do GABAA-R agonists reduce coronavirus load in the lungs?
Do GABA-R agonists modulate ACE2 expression, surfactant production/absorption, and/or inflammatory responses, and autophagy in human lungs?
How does GABA-R signaling modulate the innate immune responses of SARS-CoV-2 infected lung cells?
Can the anti-inflammatory effects of GABAA-R agonists on immune cells and CNS glial cells be exploited to reduce neuroinflammation and help ameliorate disorders such as MS and long COVID?

Author Contributions

Wrote the manuscript; D.L.K., J.T. All authors have read and agreed to the published version of the manuscript.

Funding

We thank the National Institutes of Health, Juvenile Diabetes Research Foundation, National Multiple Sclerosis Society, and the UCLA DGSOM-Broad Stem Cell Research Center for their financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank past members of the Kaufman lab for their many contributions. We thank Jessica Castillo and CTSI grant UL TR001881 for graphics support.

Conflicts of Interest

D.L.K. and J.T. are inventors of GABA-related patents. D.L.K. is an inventor of GAD-related patents and serves on the Scientific Advisory Board of Diamyd Medical.

References

  1. Erlander, M.G.; Tillakaratne, N.J.; Feldblum, S.; Patel, N.; Tobin, A.J. Two genes encode distinct glutamate decarboxylases. Neuron 1991, 7, 91–100. [Google Scholar] [CrossRef] [PubMed]
  2. Soghomonian, J.J.; Martin, D.L. Two isoforms of glutamate decarboxylase: Why? Trends Pharmacol. Sci. 1998, 19, 500–505. [Google Scholar] [CrossRef] [PubMed]
  3. Kaufman, D.L.; Houser, C.R.; Tobin, A.J. Two forms of the gamma-aminobutyric acid synthetic enzyme glutamate decarboxylase have distinct intraneuronal distributions and cofactor interactions. J. Neurochem. 1991, 56, 720–723. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, S.E.; Lee, Y.; Lee, G.H. The regulation of glutamic acid decarboxylases in GABA neurotransmission in the brain. Arch. Pharm. Res. 2019, 42, 1031–1039. [Google Scholar] [CrossRef]
  5. Kanaani, J.; Kolibachuk, J.; Martinez, H.; Baekkeskov, S. Two distinct mechanisms target GAD67 to vesicular pathways and presynaptic clusters. J. Cell Biol. 2010, 190, 911–925. [Google Scholar] [CrossRef] [Green Version]
  6. Olsen, R.W.; Sieghart, W. GABA A receptors: Subtypes provide diversity of function and pharmacology. Neuropharmacology 2009, 56, 141–148. [Google Scholar] [CrossRef] [Green Version]
  7. Ben-Ari, Y. Excitatory actions of gaba during development: The nature of the nurture. Nat. Rev. Neurosci. 2002, 3, 728–739. [Google Scholar] [CrossRef] [PubMed]
  8. Deidda, G.; Bozarth, I.F.; Cancedda, L. Modulation of GABAergic transmission in development and neurodevelopmental disorders: Investigating physiology and pathology to gain therapeutic perspectives. Front. Cell. Neurosci. 2014, 8, 119. [Google Scholar] [CrossRef] [Green Version]
  9. Couve, A.; Moss, S.J.; Pangalos, M.N. GABAB receptors: A new paradigm in G protein signaling. Mol. Cell. Neurosci. 2000, 16, 296–312. [Google Scholar] [CrossRef]
  10. Evenseth, L.S.M.; Gabrielsen, M.; Sylte, I. The GABAB Receptor-Structure, Ligand Binding and Drug Development. Molecules 2020, 25, 3093. [Google Scholar] [CrossRef]
  11. Kuriyama, K.; Sze, P.Y. Blood-brain barrier to H3-gamma-aminobutyric acid in normal and amino oxyacetic acid-treated animals. Neuropharmacology 1971, 10, 103–108. [Google Scholar] [CrossRef] [PubMed]
  12. Oldendorf, W.H. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am. J. Physiol. 1971, 221, 1629–1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Loscher, W.; Frey, H.H. Transport of GABA at the blood-CSF interface. J. Neurochem. 1982, 38, 1072–1079. [Google Scholar] [CrossRef] [PubMed]
  14. Al-Sarraf, H. Transport of 14C-gamma-aminobutyric acid into brain, cerebrospinal fluid and choroid plexus in neonatal and adult rats. Brain Res. Dev. Brain Res. 2002, 139, 121–129. [Google Scholar] [CrossRef] [PubMed]
  15. Tian, J.; Chau, C.; Hales, T.G.; Kaufman, D.L. GABA(A) receptors mediate inhibition of T cell responses. J. Neuroimmunol. 1999, 96, 21–28. [Google Scholar] [CrossRef]
  16. Tian, J.; Lu, Y.; Zhang, H.; Chau, C.H.; Dang, H.N.; Kaufman, D.L. Gamma-aminobutyric acid inhibits T cell autoimmunity and the development of inflammatory responses in a mouse type 1 diabetes model. J. Immunol. 2004, 173, 5298–5304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Alam, S.; Laughton, D.L.; Walding, A.; Wolstenholme, A.J. Human peripheral blood mononuclear cells express GABAA receptor subunits. Mol. Immunol. 2006, 43, 1432–1442. [Google Scholar] [CrossRef]
  18. Prud’homme, G.J.; Glinka, Y.; Hasilo, C.; Paraskevas, S.; Li, X.; Wang, Q. GABA protects human islet cells against the deleterious effects of immunosuppressive drugs and exerts immunoinhibitory effects alone. Transplantation 2013, 96, 616–623. [Google Scholar] [CrossRef]
  19. Zhang, B.; Vogelzang, A.; Miyajima, M.; Sugiura, Y.; Wu, Y.; Chamoto, K.; Nakano, R.; Hatae, R.; Menzies, R.J.; Sonomura, K.; et al. B cell-derived GABA elicits IL-10(+) macrophages to limit anti-tumour immunity. Nature 2021, 599, 471–476. [Google Scholar] [CrossRef]
  20. Soltani, N.; Qiu, H.; Aleksic, M.; Glinka, Y.; Zhao, F.; Liu, R.; Li, Y.; Zhang, N.; Chakrabarti, R.; Ng, T.; et al. GABA exerts protective and regenerative effects on islet beta cells and reverses diabetes. Proc. Natl. Acad. Sci. USA 2011, 108, 11692–11697. [Google Scholar] [CrossRef] [PubMed]
  21. Tian, J.; Yong, J.; Dang, H.; Kaufman, D.L. Oral GABA treatment downregulates inflammatory responses in a mouse model of rheumatoid arthritis. Autoimmunity 2011, 44, 465–470. [Google Scholar] [CrossRef] [Green Version]
  22. Tian, J.; Dang, H.N.; Yong, J.; Chui, W.S.; Dizon, M.P.; Yaw, C.K.; Kaufman, D.L. Oral treatment with gamma-aminobutyric acid improves glucose tolerance and insulin sensitivity by inhibiting inflammation in high fat diet-fed mice. PLoS ONE 2011, 6, e25338. [Google Scholar] [CrossRef] [Green Version]
  23. Tian, J.; Dang, H.; O’Laco, K.; Song, M.; Tiu, B.-C.; S, G.; Zakarian, C.; Kaufman, D. Homotaurine treatment enhances CD4+ and CD8+ Treg responses and synergizes with low-dose anti-CD3 to enhance diabetes remission in type 1 diabetic mice. ImmuoHorizons 2019, 3, 498–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tian, J.; Dang, H.; Wallner, M.; Olsen, R.; Kaufman, D.L. Homotaurine, a safe blood-brain barrier permeable GABAA-R-specific agonist, ameliorates disease in mouse models of multiple sclerosis. Sci. Rep. 2018, 8, 16555. [Google Scholar] [CrossRef] [Green Version]
  25. Tian, J.; Song, M.; Kaufman, D.L. Homotaurine limits the spreading of T cell autoreactivity within the CNS and ameliorates disease in a model of multiple sclerosis. Sci. Rep. 2021, 11, 5402. [Google Scholar] [CrossRef] [PubMed]
  26. Song, M.; Tian, J.; Middleton, B.; Nguyen, C.Q.; Kaufman, D.L. GABA Administration Ameliorates Sjogren’s Syndrome in Two Different Mouse Models. Biomedicines 2022, 10, 129. [Google Scholar] [CrossRef]
  27. Bhat, R.; Axtell, R.; Mitra, A.; Miranda, M.; Lock, C.; Tsien, R.W.; Steinman, L. Inhibitory role for GABA in autoimmune inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 2580–2585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Tian, J.; Dang, H.; Nguyen, A.V.; Chen, Z.; Kaufman, D.L. Combined therapy with GABA and proinsulin/alum acts synergistically to restore long-term normoglycemia by modulating T-cell autoimmunity and promoting beta-cell replication in newly diabetic NOD mice. Diabetes 2014, 63, 3128–3134. [Google Scholar] [CrossRef] [Green Version]
  29. Mendu, S.K.; Akesson, L.; Jin, Z.; Edlund, A.; Cilio, C.; Lernmark, A.; Birnir, B. Increased GABAA channel subunits expression in CD8+ but not in CD4+ T cells in BB rats developing diabetes compared to their congenic littermates. Mol. Immunol. 2011, 48, 399–407. [Google Scholar] [CrossRef] [Green Version]
  30. Fanger, C.M.; Neben, A.L.; Cahalan, M.D. Differential Ca2+ influx, KCa channel activity, and Ca2+ clearance distinguish Th1 and Th2 lymphocytes. J. Immunol. 2000, 164, 1153–1160. [Google Scholar] [CrossRef]
  31. Weber, K.S.; Miller, M.J.; Allen, P.M. Th17 cells exhibit a distinct calcium profile from Th1 and Th2 cells and have Th1-like motility and NF-AT nuclear localization. J. Immunol. 2008, 180, 1442–1450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Orban, C.; Bajnok, A.; Vasarhelyi, B.; Tulassay, T.; Toldi, G. Different calcium influx characteristics upon Kv1.3 and IKCa1 potassium channel inhibition in T helper subsets. Cytom. A 2014, 85, 636–641. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, K.D.; Srikanth, S.; Tan, Y.V.; Yee, M.K.; Jew, M.; Damoiseaux, R.; Jung, M.E.; Shimizu, S.; An, D.S.; Ribalet, B.; et al. Calcium signaling via Orai1 is essential for induction of the nuclear orphan receptor pathway to drive Th17 differentiation. J. Immunol. 2014, 192, 110–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Woo, J.S.; Srikanth, S.; Kim, K.D.; Elsaesser, H.; Lu, J.; Pellegrini, M.; Brooks, D.G.; Sun, Z.; Gwack, Y. CRACR2A-Mediated TCR Signaling Promotes Local Effector Th1 and Th17 Responses. J. Immunol. 2018, 201, 1174–1185. [Google Scholar] [CrossRef] [Green Version]
  35. Srikanth, S.; Woo, J.S.; Sun, Z.; Gwack, Y. Immunological Disorders: Regulation of Ca2+ Signaling in T Lymphocytes. Adv. Exp. Med. Biol. 2017, 993, 397–424. [Google Scholar] [CrossRef]
  36. Januzi, L.; Poirier, J.W.; Maksoud, M.J.E.; Xiang, Y.Y.; Veldhuizen, R.A.W.; Gill, S.E.; Cregan, S.P.; Zhang, H.; Dekaban, G.A.; Lu, W.Y. Autocrine GABA signaling distinctively regulates phenotypic activation of mouse pulmonary macrophages. Cell. Immunol. 2018, 332, 7–23. [Google Scholar] [CrossRef]
  37. Wheeler, D.W.; Thompson, A.J.; Corletto, F.; Reckless, J.; Loke, J.C.; Lapaque, N.; Grant, A.J.; Mastroeni, P.; Grainger, D.J.; Padgett, C.L.; et al. Anaesthetic impairment of immune function is mediated via GABAA receptors. PLoS ONE 2011, 6, e17152. [Google Scholar] [CrossRef] [Green Version]
  38. Reyes-Garcia, M.G.; Hernandez-Hernandez, F.; Hernandez-Tellez, B.; Garcia-Tamayo, F. GABAA receptor subunits RNA expression in mice peritoneal macrophages modulate their IL-6/IL-12 production. J. Neuroimmunol. 2007, 188, 64–68. [Google Scholar] [CrossRef]
  39. Bhandage, A.K.; Friedrich, L.M.; Kanatani, S.; Jakobsson-Bjorken, S.; Escrig-Larena, J.I.; Wagner, A.K.; Chambers, B.J.; Barragan, A. GABAergic signaling in human and murine NK cells upon challenge with Toxoplasma gondii. J. Leukoc. Biol. 2021, 110, 617–628. [Google Scholar] [CrossRef] [PubMed]
  40. Kim, J.K.; Kim, Y.S.; Lee, H.M.; Jin, H.S.; Neupane, C.; Kim, S.; Lee, S.H.; Min, J.J.; Sasai, M.; Jeong, J.H.; et al. GABAergic signaling linked to autophagy enhances host protection against intracellular bacterial infections. Nat. Commun. 2018, 9, 4184. [Google Scholar] [CrossRef]
  41. Bhandage, A.K.; Olivera, G.C.; Kanatani, S.; Thompson, E.; Lore, K.; Varas-Godoy, M.; Barragan, A. A motogenic GABAergic system of mononuclear phagocytes facilitates dissemination of coccidian parasites. Elife 2020, 9, e60528. [Google Scholar] [CrossRef]
  42. Huang, S.; Mao, J.; Wei, B.; Pei, G. The anti-spasticity drug baclofen alleviates collagen-induced arthritis and regulates dendritic cells. J. Cell. Physiol. 2015, 230, 1438–1447. [Google Scholar] [CrossRef]
  43. Duthey, B.; Hubner, A.; Diehl, S.; Boehncke, S.; Pfeffer, J.; Boehncke, W.H. Anti-inflammatory effects of the GABA(B) receptor agonist baclofen in allergic contact dermatitis. Exp. Dermatol. 2010, 19, 661–666. [Google Scholar] [CrossRef]
  44. Beales, P.E.; Hawa, M.; Williams, A.J.; Albertini, M.C.; Giorgini, A.; Pozzilli, P. Baclofen, a gamma-aminobutyric acid-b receptor agonist, delays diabetes onset in the non-obese diabetic mouse. Acta Diabetol. 1995, 32, 53–56. [Google Scholar] [CrossRef]
  45. Tian, J.; Middleton, B.; Lee, V.S.; Park, H.W.; Zhang, Z.; Kim, B.; Lowe, C.; Nguyen, N.; Liu, H.; Beyer, R.S.; et al. GABAB-Receptor Agonist-Based Immunotherapy for Type 1 Diabetes in NOD Mice. Biomedicines 2021, 9, 43. [Google Scholar] [CrossRef]
  46. Crowley, T.; Fitzpatrick, J.M.; Kuijper, T.; Cryan, J.F.; O’Toole, O.; O’Leary, O.F.; Downer, E.J. Modulation of TLR3/TLR4 inflammatory signaling by the GABAB receptor agonist baclofen in glia and immune cells: Relevance to therapeutic effects in multiple sclerosis. Front. Cell. Neurosci. 2015, 9, 284. [Google Scholar] [CrossRef] [Green Version]
  47. Bhandage, A.K.; Cunningham, J.L.; Jin, Z.; Shen, Q.; Bongiovanni, S.; Korol, S.V.; Syk, M.; Kamali-Moghaddam, M.; Ekselius, L.; Birnir, B. Depression, GABA, and Age Correlate with Plasma Levels of Inflammatory Markers. Int. J. Mol. Sci. 2019, 20, 6172. [Google Scholar] [CrossRef] [Green Version]
  48. David, M.; Richer, M.; Mamarbachi, A.M.; Villeneuve, L.R.; Dupre, D.J.; Hebert, T.E. Interactions between GABA-B1 receptors and Kir 3 inwardly rectifying potassium channels. Cell Signal 2006, 18, 2172–2181. [Google Scholar] [CrossRef]
  49. Richer, M.; David, M.; Villeneuve, L.R.; Trieu, P.; Ethier, N.; Petrin, D.; Mamarbachi, A.M.; Hebert, T.E. GABA-B1 receptors are coupled to the ERK1/2 MAP kinase pathway in the absence of GABA-B2 subunits. J. Mol. Neurosci. 2009, 38, 67–79. [Google Scholar] [CrossRef]
  50. Baloucoune, G.A.; Chun, L.; Zhang, W.; Xu, C.; Huang, S.; Sun, Q.; Wang, Y.; Tu, H.; Liu, J. GABAB receptor subunit GB1 at the cell surface independently activates ERK1/2 through IGF-1R transactivation. PLoS ONE 2012, 7, e39698. [Google Scholar] [CrossRef]
  51. Sanchez, G.M.; Incedal, T.C.; Prada, J.; O’Callaghan, P.; Dyachok, O.; Echeverry, S.; Dumral, O.; Nguyen, P.M.; Xie, B.; Barg, S.; et al. The beta-cell primary cilium is an autonomous Ca2+ compartment for paracrine GABA signaling. J. Cell. Biol. 2023, 222, e202108101. [Google Scholar] [CrossRef]
  52. McRae, B.L.; Vanderlugt, C.L.; Dal Canto, M.C.; Miller, S.D. Functional evidence for epitope spreading in the relapsing pathology of experimental autoimmune encephalomyelitis. J. Exp. Med. 1995, 182, 75–85. [Google Scholar] [CrossRef] [Green Version]
  53. Targoni, O.S.; Baus, J.; Hofstetter, H.H.; Hesse, M.D.; Karulin, A.Y.; Boehm, B.O.; Forsthuber, T.G.; Lehmann, P.V. Frequencies of neuroantigen-specific T cells in the central nervous system versus the immune periphery during the course of experimental allergic encephalomyelitis. J. Immunol. 2001, 166, 4757–4764. [Google Scholar] [CrossRef] [Green Version]
  54. Kuerten, S.; Rottlaender, A.; Rodi, M.; Velasco, V.B., Jr.; Schroeter, M.; Kaiser, C.; Addicks, K.; Tary-Lehmann, M.; Lehmann, P.V. The clinical course of EAE is reflected by the dynamics of the neuroantigen-specific T cell compartment in the blood. Clin. Immunol. 2010, 137, 422–432. [Google Scholar] [CrossRef]
  55. McMahon, E.J.; Bailey, S.L.; Castenada, C.V.; Waldner, H.; Miller, S.D. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat. Med. 2005, 11, 335–339. [Google Scholar] [CrossRef]
  56. Chastain, E.M.; Duncan, D.S.; Rodgers, J.M.; Miller, S.D. The role of antigen presenting cells in multiple sclerosis. Biochim. Biophys. Acta 2011, 1812, 265–274. [Google Scholar] [CrossRef]
  57. Almolda, B.; Gonzalez, B.; Castellano, B. Antigen presentation in EAE: Role of microglia, macrophages and dendritic cells. Front. Biosci. 2011, 16, 1157–1171. [Google Scholar] [CrossRef] [Green Version]
  58. Kuhn, S.A.; van Landeghem, F.K.; Zacharias, R.; Farber, K.; Rappert, A.; Pavlovic, S.; Hoffmann, A.; Nolte, C.; Kettenmann, H. Microglia express GABAB receptors to modulate interleukin release. Mol. Cell. Neurosci. 2004, 25, 312–322. [Google Scholar] [CrossRef]
  59. Mead, E.L.; Mosley, A.; Eaton, S.; Dobson, L.; Heales, S.J.; Pocock, J.M. Microglial neurotransmitter receptors trigger superoxide production in microglia; consequences for microglial-neuronal interactions. J. Neurochem. 2012, 121, 287–301. [Google Scholar] [CrossRef]
  60. Lee, M.; Schwab, C.; McGeer, P.L. Astrocytes are GABAergic cells that modulate microglial activity. Glia 2011, 59, 152–165. [Google Scholar] [CrossRef]
  61. Lee, M.; McGeer, E.G.; McGeer, P.L. Mechanisms of GABA release from human astrocytes. Glia 2011, 59 Pt A, 1600–1611. [Google Scholar] [CrossRef] [PubMed]
  62. Nicolini, C.; Fahnestock, M. The valproic acid-induced rodent model of autism. Exp. Neurol. 2018, 299, 217–227. [Google Scholar] [CrossRef]
  63. Singla, R.; Mishra, A.; Joshi, R.; Sarma, P.; Kumar, R.; Kaur, G.; Sharma, A.R.; Jain, A.; Prakash, A.; Bhatia, A.; et al. Homotaurine ameliorates the core ASD symptomatology in VPA rats through GABAergic signaling: Role of GAD67. Brain Res. Bull. 2022, 190, 122–133. [Google Scholar] [CrossRef]
  64. Manzano, S.; Aguera, L.; Aguilar, M.; Olazaran, J. A Review on Tramiprosate (Homotaurine) in Alzheimer’s Disease and Other Neurocognitive Disorders. Front. Neurol. 2020, 11, 614. [Google Scholar] [CrossRef] [PubMed]
  65. Aisen, P.S.; Gauthier, S.; Ferris, S.H.; Saumier, D.; Haine, D.; Garceau, D.; Duong, A.; Suhy, J.; Oh, J.; Lau, W.C.; et al. Tramiprosate in mild-to-moderate Alzheimer’s disease—A randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch. Med. Sci. 2010, 7, 102–111. [Google Scholar] [CrossRef] [Green Version]
  66. Tsolaki, M. Future strategies of management of Alzheimer’s Disease. The role of homotaurine. Hell. J. Nucl. Med. 2019, 22, 82–94. [Google Scholar]
  67. Gauthier, S.; Aisen, P.S.; Ferris, S.H.; Saumier, D.; Duong, A.; Haine, D.; Garceau, D.; Suhy, J.; Oh, J.; Lau, W.; et al. Effect of tramiprosate in patients with mild-to-moderate Alzheimer’s disease: Exploratory analyses of the MRI sub-group of the Alphase study. J. Nutr. Health Aging 2009, 13, 550–557. [Google Scholar] [CrossRef]
  68. Toppi, E.; Sireno, L.; Lembo, M.; Banaj, N.; Messina, B.; Golesorkhtafti, S.; Spalletta, G.; Bossu, P. IL-33 and IL-10 Serum Levels Increase in MCI Patients Following Homotaurine Treatment. Front. Immunol. 2022, 13, 813951. [Google Scholar] [CrossRef]
  69. Kang, S.; Liu, L.; Wang, T.; Cannon, M.; Lin, P.; Fan, T.W.; Scott, D.A.; Wu, H.J.; Lane, A.N.; Wang, R. GAB functions as a bioenergetic and signalling gatekeeper to control T cell inflammation. Nat. Metab. 2022, 4, 1322–1335. [Google Scholar] [CrossRef] [PubMed]
  70. Bhandage, A.K.; Jin, Z.; Korol, S.V.; Shen, Q.; Pei, Y.; Deng, Q.; Espes, D.; Carlsson, P.O.; Kamali-Moghaddam, M.; Birnir, B. GABA Regulates Release of Inflammatory Cytokines From Peripheral Blood Mononuclear Cells and CD4+ T Cells and Is Immunosuppressive in Type 1 Diabetes. EBioMedicine 2018, 30, 283–294. [Google Scholar] [CrossRef] [Green Version]
  71. Semyanov, A.; Walker, M.C.; Kullmann, D.M.; Silver, R.A. Tonically active GABA A receptors: Modulating gain and maintaining the tone. Trends Neurosci. 2004, 27, 262–269. [Google Scholar] [CrossRef]
  72. Glykys, J.; Mody, I. Activation of GABAA receptors: Views from outside the synaptic cleft. Neuron 2007, 56, 763–770. [Google Scholar] [CrossRef] [Green Version]
  73. Mody, I.; De Koninck, Y.; Otis, T.S.; Soltesz, I. Bridging the cleft at GABA synapses in the brain. Trends Neurosci. 1994, 17, 517–525. [Google Scholar] [CrossRef]
  74. Yoon, B.E.; Woo, J.; Chun, Y.E.; Chun, H.; Jo, S.; Bae, J.Y.; An, H.; Min, J.O.; Oh, S.J.; Han, K.S.; et al. Glial GABA, synthesized by monoamine oxidase B, mediates tonic inhibition. J. Physiol. 2014, 592, 4951–4968. [Google Scholar] [CrossRef]
  75. Birnir, B.; Korpi, E.R. The impact of sub-cellular location and intracellular neuronal proteins on properties of GABA(A) receptors. Curr. Pharm. Des. 2007, 13, 3169–3177. [Google Scholar] [CrossRef]
  76. Brickley, S.G.; Cull-Candy, S.G.; Farrant, M. Development of a tonic form of synaptic inhibition in rat cerebellar granule cells resulting from persistent activation of GABAA receptors. J. Physiol. 1996, 497 Pt 3, 753–759. [Google Scholar] [CrossRef]
  77. Wall, M.J.; Usowicz, M.M. Development of action potential-dependent and independent spontaneous GABAA receptor-mediated currents in granule cells of postnatal rat cerebellum. Eur. J. Neurosci. 1997, 9, 533–548. [Google Scholar] [CrossRef]
  78. Nusser, Z.; Sieghart, W.; Somogyi, P. Segregation of different GABAA receptors to synaptic and extrasynaptic membranes of cerebellar granule cells. J. Neurosci. 1998, 18, 1693–1703. [Google Scholar] [CrossRef] [Green Version]
  79. Richerson, G.B. Looking for GABA in all the wrong places: The relevance of extrasynaptic GABAA receptors to epilepsy. Epilepsy Curr. 2004, 4, 239–242. [Google Scholar] [CrossRef] [Green Version]
  80. Farrant, M.; Kaila, K. The cellular, molecular and ionic basis of GABAA receptor signalling. Prog. Brain Res. 2007, 160, 59–87. [Google Scholar] [CrossRef]
  81. Storustovu, S.I.; Ebert, B. Pharmacological characterization of agonists at delta-containing GABAA receptors: Functional selectivity for extrasynaptic receptors is dependent on the absence of gamma2. J. Pharmacol. Exp. Ther. 2006, 316, 1351–1359. [Google Scholar] [CrossRef] [PubMed]
  82. Meera, P.; Wallner, M.; Otis, T.S. Molecular basis for the high THIP/gaboxadol sensitivity of extrasynaptic GABAA receptors. J. Neurophysiol. 2011, 106, 2057–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Korol, S.V.; Jin, Z.; Jin, Y.; Bhandage, A.K.; Tengholm, A.; Gandasi, N.R.; Barg, S.; Espes, D.; Carlsson, P.O.; Laver, D.; et al. Functional Characterization of Native, High-Affinity GABA(A) Receptors in Human Pancreatic beta Cells. EBioMedicine 2018, 30, 273–282. [Google Scholar] [CrossRef] [Green Version]
  84. Bhandage, A.K.; Hellgren, C.; Jin, Z.; Olafsson, E.B.; Sundstrom-Poromaa, I.; Birnir, B. Expression of GABA receptors subunits in peripheral blood mononuclear cells is gender dependent, altered in pregnancy and modified by mental health. Acta Physiol. 2015, 213, 575–585. [Google Scholar] [CrossRef] [PubMed]
  85. Huang, D.; Wang, Y.; Thompson, J.W.; Yin, T.; Alexander, P.B.; Qin, D.; Mudgal, P.; Wu, H.; Liang, Y.; Tan, L.; et al. Cancer-cell-derived GABA promotes beta-catenin-mediated tumour growth and immunosuppression. Nat. Cell. Biol. 2022, 24, 230–241. [Google Scholar] [CrossRef] [PubMed]
  86. Kaufman, D.L. GABA molecules made by B cells can dampen antitumour responses. Nature 2021, 599, 374–376. [Google Scholar] [CrossRef]
  87. Huang, D.; Alexander, P.B.; Li, Q.J.; Wang, X.F. GABAergic signaling beyond synapses: An emerging target for cancer therapy. Trends Cell. Biol. 2022. [Google Scholar] [CrossRef]
  88. Kim, H.B.; Myung, S.K.; Park, Y.C.; Park, B. Use of benzodiazepine and risk of cancer: A meta-analysis of observational studies. Int. J. Cancer 2017, 140, 513–525. [Google Scholar] [CrossRef] [Green Version]
  89. Kleinerman, R.A.; Brinton, L.A.; Hoover, R.; Fraumeni, J.F., Jr. Diazepam use and progression of breast cancer. Cancer Res. 1984, 44, 1223–1225. [Google Scholar]
  90. Bonsack, F.; Sukumari-Ramesh, S. TSPO: An Evolutionarily Conserved Protein with Elusive Functions. Int. J. Mol. Sci. 2018, 19, 1694. [Google Scholar] [CrossRef] [Green Version]
  91. Jow, F.; Chiu, D.; Lim, H.K.; Novak, T.; Lin, S. Production of GABA by cultured hippocampal glial cells. Neurochem. Int. 2004, 45, 273–283. [Google Scholar] [CrossRef]
  92. Liu, Q.Y.; Schaffner, A.E.; Chang, Y.H.; Maric, D.; Barker, J.L. Persistent activation of GABAA receptor/Cl− channels by astrocyte-derived GABA in cultured embryonic rat hippocampal neurons. J. Neurophysiol. 2000, 84, 1392–1403. [Google Scholar] [CrossRef]
  93. Kozlov, A.S.; Angulo, M.C.; Audinat, E.; Charpak, S. Target cell-specific modulation of neuronal activity by astrocytes. Proc. Natl. Acad. Sci. USA 2006, 103, 10058–10063. [Google Scholar] [CrossRef] [Green Version]
  94. Jimenez-Gonzalez, C.; Pirttimaki, T.; Cope, D.W.; Parri, H.R. Non-neuronal, slow GABA signalling in the ventrobasal thalamus targets delta-subunit-containing GABAA receptors. Eur. J. Neurosci. 2011, 33, 1471–1482. [Google Scholar] [CrossRef] [Green Version]
  95. Le Meur, K.; Mendizabal-Zubiaga, J.; Grandes, P.; Audinat, E. GABA release by hippocampal astrocytes. Front. Comput. Neurosci. 2012, 6, 59. [Google Scholar] [CrossRef] [Green Version]
  96. Wang, C.M.; Chang, Y.Y.; Kuo, J.S.; Sun, S.H. Activation of P2X7 receptors induced [3H]GABA release from the RBA-2 type-2 astrocyte cell line through a Cl/HCO3-dependent mechanism. Glia 2002, 37, 8–18. [Google Scholar] [CrossRef] [PubMed]
  97. Doblas, S.; He, T.; Saunders, D.; Hoyle, J.; Smith, N.; Pye, Q.; Lerner, M.; Jensen, R.L.; Towner, R.A. In vivo characterization of several rodent glioma models by 1H MRS. NMR Biomed. 2012, 25, 685–694. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, G.; Wang, R.; Ferris, B.; Salit, J.; Strulovici-Barel, Y.; Hackett, N.R.; Crystal, R.G. Smoking-mediated up-regulation of GAD67 expression in the human airway epithelium. Respir. Res. 2010, 11, 150. [Google Scholar] [CrossRef] [Green Version]
  99. Yabumoto, Y.; Watanabe, M.; Ito, Y.; Maemura, K.; Otsuki, Y.; Nakamura, Y.; Yanagawa, Y.; Obata, K.; Watanabe, K. Expression of GABAergic system in pulmonary neuroendocrine cells and airway epithelial cells in GAD67-GFP knock-in mice. Med. Mol. Morphol. 2008, 41, 20–27. [Google Scholar] [CrossRef]
  100. Jin, S.; Merchant, M.L.; Ritzenthaler, J.D.; McLeish, K.R.; Lederer, E.D.; Torres-Gonzalez, E.; Fraig, M.; Barati, M.T.; Lentsch, A.B.; Roman, J.; et al. Baclofen, a GABABR agonist, ameliorates immune-complex mediated acute lung injury by modulating pro-inflammatory mediators. PLoS ONE 2015, 10, e0121637. [Google Scholar] [CrossRef]
  101. Wang, S.; Xiang, Y.Y.; Ellis, R.; Wattie, J.; Feng, M.; Inman, M.D.; Lu, W.Y. Effects of furosemide on allergic asthmatic responses in mice. Clin. Exp. Allergy 2011, 41, 1456–1467. [Google Scholar] [CrossRef]
  102. Jin, N.; Kolliputi, N.; Gou, D.; Weng, T.; Liu, L. A novel function of ionotropic gamma-aminobutyric acid receptors involving alveolar fluid homeostasis. J. Biol. Chem. 2006, 281, 36012–36020. [Google Scholar] [CrossRef]
  103. Huang, T.; Zhang, Y.; Wang, C.; Gao, J. Propofol reduces acute lung injury by up-regulating gamma-aminobutyric acid type a receptors. Exp. Mol. Pathol. 2019, 110, 104295. [Google Scholar] [CrossRef]
  104. Fortis, S.; Spieth, P.M.; Lu, W.Y.; Parotto, M.; Haitsma, J.J.; Slutsky, A.S.; Zhong, N.; Mazer, C.D.; Zhang, H. Effects of anesthetic regimes on inflammatory responses in a rat model of acute lung injury. Intensive Care Med. 2012, 38, 1548–1555. [Google Scholar] [CrossRef]
  105. Chintagari, N.R.; Liu, L. GABA receptor ameliorates ventilator-induced lung injury in rats by improving alveolar fluid clearance. Crit. Care 2012, 16, R55. [Google Scholar] [CrossRef] [Green Version]
  106. Voigtsberger, S.; Lachmann, R.A.; Leutert, A.C.; Schlapfer, M.; Booy, C.; Reyes, L.; Urner, M.; Schild, J.; Schimmer, R.C.; Beck-Schimmer, B. Sevoflurane ameliorates gas exchange and attenuates lung damage in experimental lipopolysaccharide-induced lung injury. Anesthesiology 2009, 111, 1238–1248. [Google Scholar] [CrossRef] [Green Version]
  107. Faller, S.; Strosing, K.M.; Ryter, S.W.; Buerkle, H.; Loop, T.; Schmidt, R.; Hoetzel, A. The volatile anesthetic isoflurane prevents ventilator-induced lung injury via phosphoinositide 3-kinase/Akt signaling in mice. Anesth. Analg. 2012, 114, 747–756. [Google Scholar] [CrossRef] [PubMed]
  108. Taniguchi, T.; Yamamoto, K.; Ohmoto, N.; Ohta, K.; Kobayashi, T. Effects of propofol on hemodynamic and inflammatory responses to endotoxemia in rats. Crit. Care Med. 2000, 28, 1101–1106. [Google Scholar] [CrossRef]
  109. Lin, X.; Ju, Y.N.; Gao, W.; Li, D.M.; Guo, C.C. Desflurane Attenuates Ventilator-Induced Lung Injury in Rats with Acute Respiratory Distress Syndrome. Biomed. Res. Int. 2018, 2018, 7507314. [Google Scholar] [CrossRef] [Green Version]
  110. Mahmoud, K.; Ammar, A. Immunomodulatory Effects of Anesthetics during Thoracic Surgery. Anesthesiol. Res. Pract. 2011, 2011, 317410. [Google Scholar] [CrossRef] [Green Version]
  111. De Conno, E.; Steurer, M.P.; Wittlinger, M.; Zalunardo, M.P.; Weder, W.; Schneiter, D.; Schimmer, R.C.; Klaghofer, R.; Neff, T.A.; Schmid, E.R.; et al. Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology 2009, 110, 1316–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Schilling, T.; Kozian, A.; Kretzschmar, M.; Huth, C.; Welte, T.; Buhling, F.; Hedenstierna, G.; Hachenberg, T. Effects of propofol and desflurane anaesthesia on the alveolar inflammatory response to one-lung ventilation. Br. J. Anaesth. 2007, 99, 368–375. [Google Scholar] [CrossRef]
  113. Kochiyama, T.; Li, X.; Nakayama, H.; Kage, M.; Yamane, Y.; Takamori, K.; Iwabuchi, K.; Inada, E. Effect of Propofol on the Production of Inflammatory Cytokines by Human Polarized Macrophages. Mediat. Inflamm. 2019, 2019, 1919538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Forkuo, G.S.; Nieman, A.N.; Kodali, R.; Zahn, N.M.; Li, G.; Rashid Roni, M.S.; Stephen, M.R.; Harris, T.W.; Jahan, R.; Guthrie, M.L.; et al. A Novel Orally Available Asthma Drug Candidate That Reduces Smooth Muscle Constriction and Inflammation by Targeting GABAA Receptors in the Lung. Mol. Pharm. 2018, 15, 1766–1777. [Google Scholar] [CrossRef] [PubMed]
  115. Xiang, Y.Y.; Chen, X.; Li, J.; Wang, S.; Faclier, G.; Macdonald, J.F.; Hogg, J.C.; Orser, B.A.; Lu, W.Y. Isoflurane regulates atypical type-A gamma-aminobutyric acid receptors in alveolar type II epithelial cells. Anesthesiology 2013, 118, 1065–1075. [Google Scholar] [CrossRef]
  116. Boost, K.A.; Leipold, T.; Scheiermann, P.; Hoegl, S.; Sadik, C.D.; Hofstetter, C.; Zwissler, B. Sevoflurane and isoflurane decrease TNF-alpha-induced gene expression in human monocytic THP-1 cells: Potential role of intracellular IkappaBalpha regulation. Int. J. Mol. Med. 2009, 23, 665–671. [Google Scholar] [CrossRef] [Green Version]
  117. Chen, M.S.; Lin, W.C.; Yeh, H.T.; Hu, C.L.; Sheu, S.M. Propofol specifically reduces PMA-induced neutrophil extracellular trap formation through inhibition of p-ERK and HOCl. Life Sci. 2019, 221, 178–186. [Google Scholar] [CrossRef]
  118. Bredthauer, A.; Geiger, A.; Gruber, M.; Pfaehler, S.M.; Petermichl, W.; Bitzinger, D.; Metterlein, T.; Seyfried, T. Propofol Ameliorates Exaggerated Human Neutrophil Activation in a LPS Sepsis Model. J. Inflamm. Res. 2021, 14, 3849–3862. [Google Scholar] [CrossRef]
  119. Meier, A.; Chien, J.; Hobohm, L.; Patras, K.A.; Nizet, V.; Corriden, R. Inhibition of Human Neutrophil Extracellular Trap (NET) Production by Propofol and Lipid Emulsion. Front. Pharmacol. 2019, 10, 323. [Google Scholar] [CrossRef] [Green Version]
  120. Lin, K.H.; Lu, W.J.; Wang, S.H.; Fong, T.H.; Chou, D.S.; Chang, C.C.; Chang, N.C.; Chiang, Y.C.; Huang, S.Y.; Sheu, J.R. Characteristics of endogenous gamma-aminobutyric acid (GABA) in human platelets: Functional studies of a novel collagen glycoprotein VI inhibitor. J. Mol. Med. 2014, 92, 603–614. [Google Scholar] [CrossRef]
  121. De Albuquerque, N.; Baig, E.; Ma, X.; Zhang, J.; He, W.; Rowe, A.; Habal, M.; Liu, M.; Shalev, I.; Downey, G.P.; et al. Murine hepatitis virus strain 1 produces a clinically relevant model of severe acute respiratory syndrome in A/J mice. J. Virol. 2006, 80, 10382–10394. [Google Scholar] [CrossRef] [Green Version]
  122. Khanolkar, A.; Hartwig, S.M.; Haag, B.A.; Meyerholz, D.K.; Epping, L.L.; Haring, J.S.; Varga, S.M.; Harty, J.T. Protective and pathologic roles of the immune response to mouse hepatitis virus type 1: Implications for severe acute respiratory syndrome. J. Virol. 2009, 83, 9258–9272. [Google Scholar] [CrossRef] [PubMed]
  123. Khanolkar, A.; Hartwig, S.M.; Haag, B.A.; Meyerholz, D.K.; Harty, J.T.; Varga, S.M. Toll-like receptor 4 deficiency increases disease and mortality after mouse hepatitis virus type 1 infection of susceptible C3H mice. J. Virol. 2009, 83, 8946–8956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Khanolkar, A.; Fulton, R.B.; Epping, L.L.; Pham, N.L.; Tifrea, D.; Varga, S.M.; Harty, J.T. T cell epitope specificity and pathogenesis of mouse hepatitis virus-1-induced disease in susceptible and resistant hosts. J. Immunol. 2010, 185, 1132–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Tian, J.; Middleton, B.; Kaufman, D.L. GABAA-Receptor Agonists Limit Pneumonitis and Death in Murine Coronavirus-Infected Mice. Viruses 2021, 13, 966. [Google Scholar] [CrossRef]
  126. Korner, R.W.; Majjouti, M.; Alcazar, M.A.A.; Mahabir, E. Of Mice and Men: The Coronavirus MHV and Mouse Models as a Translational Approach to Understand SARS-CoV-2. Viruses 2020, 12, 880. [Google Scholar] [CrossRef]
  127. McCray, P.B., Jr.; Pewe, L.; Wohlford-Lenane, C.; Hickey, M.; Manzel, L.; Shi, L.; Netland, J.; Jia, H.P.; Halabi, C.; Sigmund, C.D.; et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007, 81, 813–821. [Google Scholar] [CrossRef] [Green Version]
  128. Moreau, G.B.; Burgess, S.L.; Sturek, J.M.; Donlan, A.N.; Petri, W.A.; Mann, B.J. Evaluation of K18-hACE2 Mice as a Model of SARS-CoV-2 Infection. Am. J. Trop. Med. Hyg. 2020, 103, 1215–1219. [Google Scholar] [CrossRef]
  129. Rathnasinghe, R.; Strohmeier, S.; Amanat, F.; Gillespie, V.L.; Krammer, F.; Garcia-Sastre, A.; Coughlan, L.; Schotsaert, M.; Uccellini, M.B. Comparison of transgenic and adenovirus hACE2 mouse models for SARS-CoV-2 infection. Emerg. Microbes Infect. 2020, 9, 2433–2445. [Google Scholar] [CrossRef]
  130. Golden, J.W.; Cline, C.R.; Zeng, X.; Garrison, A.R.; Carey, B.D.; Mucker, E.M.; White, L.E.; Shamblin, J.D.; Brocato, R.L.; Liu, J.; et al. Human angiotensin-converting enzyme 2 transgenic mice infected with SARS-CoV-2 develop severe and fatal respiratory disease. JCI Insight 2020, 5, e142032. [Google Scholar] [CrossRef]
  131. Winkler, E.S.; Bailey, A.L.; Kafai, N.M.; Nair, S.; McCune, B.T.; Yu, J.; Fox, J.M.; Chen, R.E.; Earnest, J.T.; Keeler, S.P.; et al. SARS-CoV-2 infection in the lungs of human ACE2 transgenic mice causes severe inflammation, immune cell infiltration, and compromised respiratory function. bioRxiv 2020. [Google Scholar] [CrossRef]
  132. Oladunni, F.S.; Park, J.G.; Pino, P.A.; Gonzalez, O.; Akhter, A.; Allue-Guardia, A.; Olmo-Fontanez, A.; Gautam, S.; Garcia-Vilanova, A.; Ye, C.; et al. Lethality of SARS-CoV-2 infection in K18 human angiotensin-converting enzyme 2 transgenic mice. Nat. Commun. 2020, 11, 6122. [Google Scholar] [CrossRef]
  133. Tian, J.; Dillion, B.J.; Henley, J.; Comai, L.; Kaufman, D.L. A GABA-receptor agonist reduces pneumonitis severity, viral load, and death rate in SARS-CoV-2-infected mice. Front. Immunol. 2022, 13, 1007955. [Google Scholar] [CrossRef] [PubMed]
  134. Bansal, P.; Wang, S.; Liu, S.; Xiang, Y.Y.; Lu, W.Y.; Wang, Q. GABA coordinates with insulin in regulating secretory function in pancreatic INS-1 beta-cells. PLoS ONE 2011, 6, e26225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Rorsman, P.; Berggren, P.O.; Bokvist, K.; Ericson, H.; Mohler, H.; Ostenson, C.G.; Smith, P.A. Glucose-inhibition of glucagon secretion involves activation of GABAA-receptor chloride channels. Nature 1989, 341, 233–236. [Google Scholar] [CrossRef] [PubMed]
  136. Bai, D.; Fang, L.; Xia, S.; Ke, W.; Wang, J.; Wu, X.; Fang, P.; Xiao, S. Porcine deltacoronavirus (PDCoV) modulates calcium influx to favor viral replication. Virology 2020, 539, 38–48. [Google Scholar] [CrossRef]
  137. Kraeft, S.K.; Chen, D.S.; Li, H.P.; Chen, L.B.; Lai, M.M. Mouse hepatitis virus infection induces an early, transient calcium influx in mouse astrocytoma cells. Exp. Cell. Res. 1997, 237, 55–62. [Google Scholar] [CrossRef]
  138. Yang, A.C.; Kern, F.; Losada, P.M.; Agam, M.R.; Maat, C.A.; Schmartz, G.P.; Fehlmann, T.; Stein, J.A.; Schaum, N.; Lee, D.P.; et al. Dysregulation of brain and choroid plexus cell types in severe COVID-19. Nature 2021, 595, 565–571. [Google Scholar] [CrossRef]
  139. Douaud, G.; Lee, S.; Alfaro-Almagro, F.; Arthofer, C.; Wang, C.; McCarthy, P.; Lange, F.; Andersson, J.L.R.; Griffanti, L.; Duff, E.; et al. SARS-CoV-2 is associated with changes in brain structure in UK Biobank. Nature 2022, 604, 697–707. [Google Scholar] [CrossRef]
  140. Frere, J.J.; Serafini, R.A.; Pryce, K.D.; Zazhytska, M.; Oishi, K.; Golynker, I.; Panis, M.; Zimering, J.; Horiuchi, S.; Hoagland, D.A.; et al. SARS-CoV-2 infection results in lasting and systemic perturbations post recovery. bioRxiv 2022. [Google Scholar] [CrossRef]
  141. Crunfli, F.; Carregari, C.C.; Veras, F.P.; Vendramini, P.H.; Valença, A.G.F.; Antunes, A.S.L.M. Morphological, cellular and molecular basis of brain infection in COVID-19 patients. MedRxiv 2020, 16. [Google Scholar] [CrossRef] [PubMed]
  142. Virhammar, J.; Naas, A.; Fallmar, D.; Cunningham, J.L.; Klang, A.; Ashton, N.J.; Jackmann, S.; Westman, G.; Frithiof, R.; Blennow, K.; et al. Biomarkers for central nervous system injury in cerebrospinal fluid are elevated in COVID-19 and associated with neurological symptoms and disease severity. Eur. J. Neurol. 2021, 28, 3324–3331. [Google Scholar] [CrossRef]
  143. Kanberg, N.; Ashton, N.J.; Andersson, L.M.; Yilmaz, A.; Lindh, M.; Nilsson, S.; Price, R.W.; Blennow, K.; Zetterberg, H.; Gisslen, M. Neurochemical evidence of astrocytic and neuronal injury commonly found in COVID-19. Neurology 2020, 95, e1754–e1759. [Google Scholar] [CrossRef]
  144. Lee, M.H.; Perl, D.P.; Nair, G.; Li, W.; Maric, D.; Murray, H.; Dodd, S.J.; Koretsky, A.P.; Watts, J.A.; Cheung, V.; et al. Microvascular Injury in the Brains of Patients with COVID-19. N. Engl. J. Med. 2021, 384, 481–483. [Google Scholar] [CrossRef]
  145. Johansson, A.; Mohamed, M.S.; Moulin, T.C.; Schioth, H.B. Neurological manifestations of COVID-19: A comprehensive literature review and discussion of mechanisms. J. Neuroimmunol. 2021, 358, 577658. [Google Scholar] [CrossRef]
  146. Tian, J.; Dang, H.; Kaufman, D.L. Combining antigen-based therapy with GABA treatment synergistically prolongs survival of transplanted ss-cells in diabetic NOD mice. PLoS ONE 2011, 6, e25337. [Google Scholar] [CrossRef] [PubMed]
  147. Liu, W.; Lau, H.K.; Son, D.O.; Jin, T.; Yang, Y.; Zhang, Z.; Li, Y.; Prud’homme, G.J.; Wang, Q. Combined use of GABA and sitagliptin promotes human beta-cell proliferation and reduces apoptosis. J. Endocrinol. 2021, 248, 133–143. [Google Scholar] [CrossRef]
  148. Son, D.O.; Liu, W.; Li, X.; Prud’homme, G.J.; Wang, Q. Combined effect of GABA and glucagon-like peptide-1 receptor agonist on cytokine-induced apoptosis in pancreatic beta-cell line and isolated human islets. J. Diabetes 2019, 11, 563–572. [Google Scholar] [CrossRef]
  149. Prud’homme, G.J.; Glinka, Y.; Kurt, M.; Liu, W.; Wang, Q. The anti-aging protein Klotho is induced by GABA therapy and exerts protective and stimulatory effects on pancreatic beta cells. Biochem. Biophys. Res. Commun. 2017, 493, 1542–1547. [Google Scholar] [CrossRef]
  150. Tian, J.; Song, M.; Kaufman, D.L. Designing Personalized Antigen-Specific Immunotherapies for Autoimmune Diseases-The Case for Using Ignored Target Cell Antigen Determinants. Cells 2022, 11, 1081. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, Q.; Ren, L.; Wan, Y.; Prud’homme, G.J. GABAergic regulation of pancreatic islet cells: Physiology and antidiabetic effects. J. Cell. Physiol. 2019, 234, 14432–14444. [Google Scholar] [CrossRef] [PubMed]

Figure and Table

Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tian, J.; Kaufman, D.L. The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19. Biomedicines 2023, 11, 254. https://doi.org/10.3390/biomedicines11020254

AMA Style

Tian J, Kaufman DL. The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19. Biomedicines. 2023; 11(2):254. https://doi.org/10.3390/biomedicines11020254

Chicago/Turabian Style

Tian, Jide, and Daniel L. Kaufman. 2023. "The GABA and GABA-Receptor System in Inflammation, Anti-Tumor Immune Responses, and COVID-19" Biomedicines 11, no. 2: 254. https://doi.org/10.3390/biomedicines11020254

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop