Skip to main content
Cancer Medicine logoLink to Cancer Medicine
. 2023 May 18;12(13):14498–14510. doi: 10.1002/cam4.6102

Insights into the leveraging of GABAergic signaling in cancer therapy

Tian‐Jiao Li 1, Jian Jiang 2, Ya‐Ling Tang 3,, Xin‐Hua Liang 1,
PMCID: PMC10358225  PMID: 37199392

Abstract

Gamma‐aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the brain of adult mammals. Several studies have demonstrated that the GABAergic system may regulate tumor development via GABA receptors, downstream cyclic adenosine monophosphate (cAMP) pathway, epithelial growth factor receptor (EGFR) pathway, AKT pathway, mitogen‐activated protein kinase (MAPK) or extracellular signal‐related kinases (ERK) pathway, and matrix metalloproteinase (MMP) pathway, although the exact mechanism is unclear. Pioneering studies reported that GABA signaling exists and functions in the cancer microenvironment and has an immunosuppressive effect that contributes to metastasis and colonization. This article reviews the molecular structures and biological functions of GABAergic components correlated with carcinogenesis, the mechanisms underlying GABAergic signaling that manipulate the proliferation and invasion of cancer cells, and the potential GABA receptor agonists and antagonists for cancer therapy. These molecules may provide an avenue for the development of specific pharmacological components to prevent the growth and metastasis of various cancers.

Keywords: cancer, carcinogenesis, GABA, GABA receptor, neurotransmitter

1. INTRODUCTION

Gamma‐aminobutyric acid (GABA) is the principal inhibitory neurotransmitter in the central nervous system (CNS) that acts via the activation of specific GABA receptors. GABA is synthesized from glutamate, glutamine, and glucose by the glutamic acid decarboxylase enzymes GAD65 and GAD67 in the CNS. 1 , 2 GABA is activated by combining ionotropic GABAA receptors or metabotropic GABAB receptors 3 and controls the proliferation, differentiation, migration, and death of cells during nervous system development. 4 Recent studies revealed that GABA and its receptors also express and have diverse physiological functions in the development and maturation of nonneuronal peripheral tissue, 5 including the palate, 6 , 7 lungs, 8 , 9 digestive tract, 10 , 11 pancreas, 12 liver, 13 testicular cells, 14 , 15 and even stem cells. 16

Neurotransmitter systems contribute to multiple malignancies. Increasing reports demonstrated that the expression of GAD, GABA, and GABA receptors was significantly higher in cancers, including colon cancer, lung cancer, and gastric cancer than in normal tissues. 17 , 18 , 19 , 20 , 21 , 22 , 23 Cancer cell proliferation and invasion are regulated by the binding of GABA to GABAA or GABAB receptors, thus, opening new avenues to develop pharmacological agonists and antagonists for cancer therapy. This review describes the potential mechanisms of GABAergic signaling in cancer development and progression and the possible treatment of cancer using GABA receptor‐associated drugs.

2. THE GABAergic SYSTEM

The GABAergic system functions as an inhibitory neurotransmitter in the CNS and mainly contains GABA, GABA transporters, GABA receptors, and GABAergic neurons. 24 In GABAergic neurons, GABA can be directly synthesized by the decarboxylation of glutamate in the cytosol. 25 Alternatively, GABA can be indirectly formed from glutamate by bypassing the tricarboxylic acid (TCA) cycle. This process is called the GABA shunt, a closed‐loop system in charge of the production and conservation of GABA supply 25 (Figure 1). GABA synthesis is mediated by two GAD isoforms, namely, GAD65 and GAD67. GAD65 mainly participates in GABAergic synaptic transmission and plasticity, whereas GAD67 manipulates metabolic GABA synthesis. 26 Notably, GAD67 is upregulated in tumors and contributes to cancer progression, whereas GAD65 remains unreported. 27 GAD67 facilitates tumor progression as it is overexpressed in tumor tissues with greater proliferative and invasive potential. 28 , 29 , 30 Moreover, high DNA hypermethylation, mRNA expression, and protein expression of GAD67 have been correlated with advanced tumor status, which makes it a potentially important prognostic indicator of poor outcomes in several cancer types. 27 , 31 , 32 , 33 The GABA transaminase 4‐aminobutyrate aminotransferase (ABAT) catabolizes GABA into succinic semialdehyde. Reduced expression of ABAT in some cancer cells may promote cancer cell proliferation and migration due to the accumulation of GABA. 34 , 35 , 36 Therefore, the inhibition of GAD67 expression and promotion of ABAT expression may reduce cancer progression.

FIGURE 1.

FIGURE 1

The schematic diagram of GABA synthesis and the potential mechanisms of GABAergic components in cancer proliferation and invasion. In cancer cells, GABA is formed from glutamate directly or via TCA cycle metabolism, and high level of GABA in the tumor microenvironment due to increased GABA production from GAD and decreased GABA degradation by ABAT. Then the synthesized GABA activates the GABA receptors by combining with the extracellular binding domains of GABAA receptors or GABAB receptors to regulate cancer cell proliferation and/or invasion. ABAT, 4‐aminobutyrate aminotransferase; cAMP; cyclic adenosine monophosphate; EGFR, epithelial growth factor receptor; GABA, gamma‐aminobutyric acid; GAD, glutamic acid decarboxylase enzyme; MAPK/ERK, mitogen‐activated protein kinase/extracellular signal‐related kinase; MMP, matrix metalloproteinase; TCA, tricarboxylic acid.

GABA functions via two types of receptors, namely, ionotropic GABAA and GABAC receptors and metabotropic GABAB receptors, which have different structures and functions (Figure 2). GABAA receptors (GABAAR) are members of ligand‐gated chloride channels families consisting of the assembly of multiple subunit subtypes (α1–6, β1–3, γ1–3, δ, ϵ, θ, π, ρ1–3) into a pentamer. The pharmacological and functional properties of GABAAR channels closely depend on their subunit composition. The most abundant GABAAR subtype consists of two α, two β, and one γ subunits. 37 GABAAR is activated by binding with GABA to open the Cl channel, subsequently inducing the hyperpolarization of the postsynaptic membrane. Contrary to GABA‐binding sites, GABAAR possesses several allosteric binding sites for anxiolytic, hypnotic, anesthetic, and anticonvulsant drugs. 38 For instance, bicuculline and picrotoxin are antagonists of GABAAR, whereas propofol and benzodiazepines are its agonists. 39 , 40 , 41 GABAB receptors (GABABR) are members of the family of heterodimeric G‐protein‐coupled receptors, which are composed of two main GABABR subunits termed GABAB receptor 1 (GABABR1) subunit and GABABR2 subunit. 42 Each subunit contains a C‐terminal intracellular domain, an N‐terminal extracellular domain, a heptahelical transmembrane domain, and an extracellular Venus flytrap domain. The subunits interact with each other via an intracellular coiled‐coil domain near the C terminus, which can mask the reticulum retention signal. GABABR can be activated by GABA to regulate Ca2+ channels and K+ channels. 43 , 44 GABA activates serious proteins connecting with the C‐termini of GABABR by binding with the extracellular domain of GABABR1, whereas the extracellular domain of GABABR2 has no ligand‐binding activity. Some allosteric modulators, such as CGP7930, can bind the transmembrane domain of GABABR2 to modulate the binding affinity of the ligand with GABABR1, as well as the efficacy of signal transduction after binding. 45 However, the mechanisms governing downstream signaling remain unclear.

FIGURE 2.

FIGURE 2

GABAA receptor and GABAB receptor molecular and functional structure. (A) GABAAR is a pentameric ligand‐gated ion chloride channels consisting of five transmembrane subunits. The most abundant GABAAR subtype consists of 2α, 2β, and 1γ subunits. GABAAR is activated by binding with GABA to open Cl channel. (B) GABABR is a heterodimeric G‐protein‐coupled receptor composed of the GABABR1 subunit and GABABR2 subunit. The subunits interact with each other via intracellular coiled‐coil domains near the C terminus. The intracellular loops of GABABR2 are coupled to Gi/o‐type G‐proteins. The extracellular domain of GABABR1 can bind GABA, whereas the extracellular domain of GABABR2 has no ligand‐binding activity. GABABR is activated by binding with GABA to regulate K+ channels and Ca2+ channels. GABAAR, GABAA receptor; GABABR, GABAB receptor; GABABR1, GABAB receptor 1; GABABR2, GABAB receptor 2.

All components of the GABAergic system have also been reported to be highly expressed in peripheral organs such as the liver, 46 pancreas, 47 , 48 prostate, 49 kidneys, 50 intestines, 51 testes, 52 and ovaries, 53 thus highlighting the pivotal effect of the GABAergic system in human development.

3. GABAergic SIGNALING IN CANCER

3.1. The effects of GABA and its receptors in different cancers

Recent studies have demonstrated that GABA exerts crucial regulatory effects on various types of cancers by binding with its receptors. In most cases, the expression levels of GABA and its receptors showed significant changes in tumor tissues compared with normal tissues, with GABA affecting cellular proliferation through GABAAR and cellular invasion through GABABR. However, it is unclear as to whether GABAergic signaling could serve a positive or negative role in the regulation of cancer cell behavior (Table 1). GABAergic signaling may have different functions that depend on the origin of the tumor and the type of receptor subunits, which underscore the need to understand the GABA crosstalk in cancer.

TABLE 1.

The expression, effect, and pathways of GABA and GABA receptors in different cancers.

Region Cancer Expression Cancer development Mechanisms Studies
Brain Glioblastoma GABAAR1 Negative GABAAR, miR‐155 104, 105
Head and neck Oral cancer GAD1↑, GABAARP Positive GAD1, GABAARP 56, 81
Nasopharyngeal carcinoma GAD1↑ Positive GAD1 31
Lung NSCLC GABA↓, GABAARA3↑, GABAARE↑, GABABR2 Negative GABABR, cAMP 57, 106
PAC GABA↓ Negative GABABR, cAMP, Erk1/2 58
Breast Breast cancer GABA↓ Negative GABAAR 66, 99
Stomach Gastric cancer GABA↑, GAD65(+), GABAAR (+), GABABR (−) Positive GABAAR, MAPK/ERK 23
Liver Hepatocellular carcinoma (HCC) GABAARβ3↓ Negative GABAARβ3, GABABR cAMP, ERK1/2 62, 63, 96
GABAARQ↑, GABAARA3 Positive GABAAR 17, 61
Cholangiocarcinoma GABAARβ3 (+), GABABR1(+), GABACR (+) Negative GABAR, cAMP, MAPK/ERK 60
Colon Colon cancer GABAR (+) Negative GABAR, cAMP, MMP 19, 59
Pancreas PDAC GABA↓ Negative GABABR, cAMP, ERK1/2 64
GABAARP Positive GABAARP, MAPK/ERK 30
Prostate Prostate cancer GABA↑, GAD67↑, GABAAR↑ Positive GABAAR and GABABR, MMP 18, 22
Ovarian Ovarian cancer GABA↑ Positive GABAARP 55, 107
Thyroid Thyroid tumors GABAARB2↑, GABAARA2 (+), GABABR2 Positive GABAARβ2, GABAARα2, GABABR2 108

Abbreviations: GAD, glutamic acid decarboxylase enzymes; GABAR, GABA receptor; GABAAR1, GABAA receptor 1; GABAARA2, GABAA receptor subunit α2; GABAARA3, GABAA receptor subunit α3; GABAARE, GABAA receptor subunit ε; GABABR2, GABAB receptor subunit 2; GABAARQ, GABAA receptor subunit θ; GABAARP, GABAA receptor subunit π; GABAARβ2, GABAA receptor subunit β2; GABAARβ3, GABAA receptor subunit β3.

3.1.1. The effects of GABA on cancers

The GABA pathway plays a dual role and can be differentially regulated in different cell populations. GABA promotes tumor development in prostate cancer, 18 , 22 , 54 gastric cancer, 23 ovarian cancer, 55 and oral squamous cell carcinoma (OSCC). 56 In contrast, GABA exerts inhibitory effects on non‐small cell lung cancer (NSCLC), 57 pulmonary adenocarcinoma (PAC), 58 colon cancer, 59 and cholangiocarcinoma. 60 Notably, GABA and its receptors have a dual function in hepatocellular carcinoma (HCC), 17 , 61 , 62 , 63 pancreatic ductal adenocarcinoma (PDAC), 30 , 64 and breast cancer. 65 , 66 , 67 , 68 In HCC, Li et al 17 found that GABA promotes the proliferation of hepatoma cells through the GABAAR θ subunit (GABAARQ), while Yan et al 61 found that this effect occurs through the GABAAR α3 subunit (GABAARA3). However, other subunits of GABA receptors may play an opposite role. Minuk et al 62 reported that the expression of hepatic GABAAR β3 subunit (GABAARB3) was downregulated in human HCC, and its restoration can attenuate tumor growth in nude mice. In PDAC, Takehara et al 30 observed that GABA promoted the proliferation of PDAC cells through the GABAAR π subunit (GABAARP), whereas Schuller et al 64 demonstrated that GABA inhibited PDAC cell proliferation and migration by inhibiting the isoproterenol‐stimulated pathway, which may be suppressed in human PDAC tissue. In breast cancer, GABA promotes the proliferation and invasion of breast cancer cells through GABABR1e, GABAARA3, 68 and GABAARP. 67 However, Drell et al 66 reported that GABA inhibited the norepinephrine‐induced migration of breast cancer cells, which may be mediated by GABABR, as confirmed by the use of a specific GABABR agonist, baclofen. The dichotomous role of GABA on cancers may be attributed to the diverse subunits of GABA receptors.

3.1.2. The effect of GABAA receptors on cancers

Several studies have reported that GABA can promote cell proliferation through the GABAAR in cancers such as HCC, 17 , 61 prostate cancer, 22 , 54 gastric cancer, 23 breast cancer, 67 , 68 PDAC, 30 and OSCC, 56 and inhibit cell proliferation through the GABAAR in cancers such as HCC 62 and cholangiocarcinoma. 60 The GABAARQ and GABAARA3 subunits of GABAAR, which mediate inhibitory synaptic transmission in the CNS, were overexpressed in HCC tissues compared with adjacent nontumor liver tissues, and the knockdown of GABAARQ and GABAARA3 expression in malignant hepatocytes resulted in attenuated tumor growth both in vitro and in vivo. 17 , 61 In contrast, GABAARB3 exerts an opposite function. 62 The overexpression of GABAARA3 also promoted the migration, invasion, and metastasis of breast cancer cells. 68 GABAARP, a subunit of GABAAR, has been reported to promote tumorigenesis, proliferation, and migration of PDAC cells, basal‐like breast cancer, ovarian cancer, and OSCC, 30 , 55 , 56 , 67 which confirms the function of GABAARP in the initiation and progression of cancer. In human thyroid cancer, the expression of GABA receptors was higher in tumors than in normal thyroid tissue, and GABAAR appeared to play a vital role. GABAARB2 was detected in the blood vessels of normal thyroid and thyroid tumors but not in thyroid cancer cells, suggesting that GABA signaling contributes to angiogenesis in thyroid cancer. GABAARA2 was detected in metastasis‐derived but not in primary‐tumor‐derived cell lines, suggesting its possible role in the development of metastases. 69 Although the research by Roberts documents the expression of the GABAergic system in human thyroid tumors, further functional studies are needed.

3.1.3. The effect of GABAB receptors on cancers

GABABR plays a positive role in cancer cell invasion in breast cancer 65 and prostate cancer 22 and a negative role in breast cancer, 66 colon carcinoma, 59 NSCLC, 57 HCC, 63 and cholangiocarcinoma. 60 GABABR1e is overexpressed in human breast cancer cell lines and tissues and promotes the malignancy of breast cancer cells both in vitro and in vivo. 65 The expression of the GABA receptor gene phenotype is associated with tumorigenesis and the clinical prognosis of NSCLC. A high expression of GABABR2 with a low expression of GABAARA3 may predict a better outcome. 57

The diverse roles of GABA and its receptors need further description, which would yield more insights into novel drugs for treating cancers by targeting the regulation of GABAergic signaling. Ongoing efforts to characterize the mechanisms of GABAAR and GABABR in cancers have revealed the potential downstream pathways of GABA receptors.

3.2. The mechanisms of GABAergic signaling in cancer progression and metastasis

GABAergic signaling contributes to tumorigenesis in systemic organs, and growing evidence has elucidated the potential pathways of tumor progression and metastasis. Moreover, the GABAergic system has recently been shown to exert immunosuppressive effects by disrupting the functions of various peripheral immune cells (Figure 1).

3.2.1. cAMP pathway

Cyclic adenosine monophosphate (cAMP) signaling can regulate the biological behavior, including proliferation, migration, invasion, and metabolism, of cancer cells. Notably, cAMP signaling can either promote or suppress tumors depending on the cell type and the specific environment. 70 Current reports have reported that cAMP plays a pivotal role in the inhibitory effect of GABA on tumors. GABA can suppress the proliferation and migration of NSCLC, PAC, and cholangiocarcinoma cells via cAMP signaling. 57 , 58 , 60 Similarly, GABA can also inhibit colon cancer and HCC migration mediated by GABABR and intracellularly transduced by a decrease in the cAMP concentration. 19 , 59 , 63 Cancer‐induced bone pain (CIBP) remains a major challenge in advanced cancer patients. Zhou et al 71 first described that the downregulation of GABABR contributed to the development and maintenance of CIBP, and the restoration of depleted GABABR attenuated CIBP‐induced pain behaviors at least partially by inhibiting the cAMP signaling pathway. Moreover, baclofen significantly inhibited mechanical allodynia and ambulatory pain induced by CIBP in a dose‐dependent manner.

3.2.2. EGFR pathway

The epithelial growth factor receptor (EGFR) family has been widely researched in pharmacology owing to its close association with malignant proliferation. 72 GABAAR can be activated by 5α‐androstane‐3α,17β‐diol (3α‐diol) in prostate cancer cells to transform androgen‐dependent EGFR pathways for the progression of castration‐resistant prostate cancer. 73 Moreover, Wu et al 54 demonstrated that increased GABAARA1 might participate in this tumor‐promoting action by activating EGFR and the downstream signaling molecule Src. GABABR can also transactivate EGFR through a ligand‐dependent mechanism, thus promoting the migration and invasion of prostate cancer cells. 74 Furthermore, the GABABR agonist baclofen selectively causes multisite phosphorylation of tyrosine residues associated with EGFR ubiquitination. 74 Further the research group 65 describes that GABABR1e favors EGFR signaling by displacing phosphatase nonreceptor type 12 (PTPN12) to disrupt the interaction between EGFR and PTPN12, which in turn promotes the growth and invasion of breast cancer cells.

EGFR‐mediated carcinogenesis may function by continuously phosphorylating downstream signaling effectors, including phosphatidylinositol‐3‐kinase (PI3K), protein‐serine–threonine kinase (AKT), or mitogen‐activated protein kinase (MAPK) pathways.

3.2.3. AKT pathway

AKT is a serine/threonine kinase that participates in the critical role of the PI3K signaling pathway, which regulates the survival, invasion, migration, and epithelial‐mesenchymal transition (EMT) of cells. The kinase activity of AKT is induced by various growth factors, including EGFR, while the translocation of AKT from the cytoplasm to the inner surface of the cell membrane, a key process of AKT phosphorylation, depends on PI3K‐generated phospholipids. GABA may trigger several signaling molecules upstream of AKT to influence carcinogenesis through AKT signaling. Although GABAAR has been shown to regulate EGFR activation in prostate cancer, this condition was not observed in breast cancer cells. The overexpression of GABARA3 promotes the migration and metastasis of breast cancer through AKT signaling. 68 GABARA3 may mediate signaling molecules upstream of the AKT pathway, like PI3K, rather than EGFR. Conversely, GABABR can mediate the EGFR‐AKT signaling pathway. Moreover, in breast cancer, GABABR1e can activate the EGFR‐AKT pathway to promote the malignancy of breast cancer cells both in vitro and in vivo. 65 Similarly, GABA enhances the malignancy of human high‐grade chondrosarcoma by promoting AKT signaling. 75

3.2.4. MAPK/ERK pathway

Mitogen‐activated protein kinases (MAPKs) or extracellular signal‐related kinases (ERKs) are serine/threonine kinases that transform extracellular stimuli into intracellular signals controlling cell proliferation and differentiation, and their dysregulation induces tumorigenesis. 76 , 77 Elevated intracellular Ca2+, induced by GABA to influence neurogenesis and synaptogenesis, activates the small guanine nucleotide‐binding protein Ras and stimulates the MAPK cascade. 78 GABAAR, particularly GABAARP, contributes to the proliferative and aggressive action of advanced tumors by activating MAPK/ERK signaling. GABA stimulates tumor growth in PDAC, basal‐like breast cancer, and ovarian cancer through GABAARP by activating the MAPK/ERK cascade by increasing intracellular Ca2+ levels. 30 , 55 , 67 Similarly, GABAARP promotes the proliferation of OSCC by activating the MAPK pathway. 56 GABA can also promote the growth of human gastric cancer cells in an autocrine or paracrine approach through GABAAR, followed by the activation of MAPK/ERK and an increase in cyclin D1. 23 Besides GABAAR, GABABR can also facilitate the invasion and metastasis of breast cancer mediated by promoting the phosphorylation of ERK1/2 and subsequently increasing the expression of MMP‐2. 79 The MAPK/ERK pathway also involves the tumor‐suppressive effect of GABA. GABA decreases the in vitro cholangiocarcinoma growth through protein kinase A‐ and D‐myo‐inositol‐1,4,5‐triphosphate/Ca2+‐dependent pathways followed by the downregulation of ERK‐1/2 phosphorylation and cAMP. 60

3.2.5. MMP pathway

Matrix metalloproteinases (MMPs) are critical to tumor invasion and metastasis, as they influence the shape, movement, growth, survival, and differentiation of cancer cells by mediating the cytoskeletal machinery and cell adhesion. 80 The overexpression of GAD1 is closely correlated with the invasion and metastasis of OSCC by the activation of MMP7. 81 GABA promotes the invasion of prostate and breast cancer cells through GABABR, and the increasing activities of MMPs, especially MMP‐2 and MMP‐3, are involved in the mechanism underlying this function. 18 , 79 Moreover, the MMP inhibitor GM6001 can significantly decrease GABA‐induced migration, 18 with MMPs also being involved in the inhibitory effect of GABA in cancers such as colon cancer. Nembutal, a GABA agonist, inhibits the primary growth and metastasis of colon cancer, as it can reduce MMP production. 19

Owing to the involvement of a complex cascade of events in cancer progression and metastasis, the above signaling pathways are intricately intertwined rather than independent of each other. Existing evidence has demonstrated that the activation of GABABR downregulates the phosphorylation of ERK1/2 via the cAMP/PKA‐dependent pathway 60 and consequently promotes the production of MMPs. 79 GABABR can reduce the activity of adenylyl cyclase and enhance the production of cAMP due to its association with Gi and Go. 82 Moreover, EGFR transactivated by GABABR induces the activation of ERK1/2 by a Gi/o‐dependent mechanism that requires MMP‐modulated pro‐ligand shedding. 74 Further details about the modulatory mechanism need further clarification.

3.3. Cancer microenvironment

The GABAergic system is highly expressed in peripheral organs, and its control over the biological behavior of cells is also widespread through peripheral organs not only limited the CNS. The proliferation and metastasis of tumors benefit from GABA synthesized by not only cancer cells but also from the tumor microenvironment. Young et al 83 reviewed the similarity of the brain and stem cell niches of peripheral organs and reported that GABAergic components were expressed in both normal and tumor conditions in the brain and peripheral organs. Neman et al 84 found that the breast‐to‐brain metastatic tissue and cells displayed a GABAergic phenotype similar to that of neuronal cells. The GABA components, including GABAAR, GABA transporter, GABA transaminase, parvalbumin, and reelin, are all highly expressed in the metastasis of breast cancer to the brain. This indicates that breast cancers may metastasize to the brain when they escape their normative genetic constraints to adapt and coinhabit the neural microenvironment. GABAergic signaling not only provides the biosynthetic energy source for cancer proliferation but also disturbs normal cell proliferation, resulting in abnormal proliferation. All these findings pave the way for the development of a potential therapeutic target that disturbs the cancer microenvironment to inhibit tumor growth and metastasis for improved prognosis.

3.4. Cancer immunometabolism

Recent studies in the field of cancer immunometabolism have demonstrated that the production and consumption of metabolic products by different immune cells in various stages of differentiation and activation influence antitumor immune potential. 85 , 86 These small metabolites produced during the differentiation and activation of immune cells have better evolutionary potential as communication molecules than agents of classical signaling regulated by cytoplasmic, membrane‐bound, or secreted proteins. They can use fewer cellular resources for faster synthesis and secretion. Much attention has focused on the metabolism of glucose, amino acids, and fatty acids, although little is known about the role of metabolite GABA.

A recent study by Zhang et al 87 reported that GABA is synthesized and secreted by activated B cells and confirmed that it can increase the expression of IL‐10 receptors, promote the differentiation and survival of anti‐inflammatory macrophages, and regulate the antitumor responses of CD8+ T cells through GABAAR in a mouse model of colon cancer. Previous studies demonstrated that several immune cells express GABAAR, and the activation of GABAAR through GABA binding inhibits inflammation. The activation of GABAAR on T cells restrains inflammatory T cells, namely, helper CD4+ T cells and killer CD8+ T cells, 88 , 89 , 90 while augmenting the numbers of regulatory T cells, which restricts inflammation. 12 , 89 Also, the activation of GABAAR on antigen‐presenting cells reduces their pro‐inflammatory properties. 91 , 92 , 93 Zhang et al 87 found that a GABAAR agonist named muscimol can inhibit the activation and proliferation of tumor‐infiltrating CD8+ T cells, whereas the GABAAR antagonist picrotoxin can promote the cytotoxic activity of CD8+ T cells and limit tumor growth in vivo. The use of picrotoxin paves a way for targeting metabolite signaling to boost the efficacy of immune checkpoint interaction and chimeric antigen receptor T‐cell (CAR‐T) therapies.

A new study by Huang et al 27 more broadly reveals the molecular and functional mechanism of GABA in the inhibition of antitumor immunity. Rather than serving as metabolic fuel or building blocks, GABA derived from lung and colon cancers activates GABABR to stabilize β‐catenin induced by the ectopic expression of GAD1. Thus, it suppresses the expression of CCL4 and CCL5 to prevent the recruitment of dendritic cells, subsequently promoting tumor progression in an autocrine manner and inhibiting antitumor immune cell infiltration in a paracrine manner.

This emerging work reinforces the need to annotate metabolites within the cancer microenvironment, as it may boost the development of targeted therapy for the inhibition of tumor growth and metastasis through immunomodulation and metabolites.

4. TARGETING GABA RECEPTORS FOR CANCER THERAPY

With growing evidence implicating the role of GABA receptors in tumor progression and metastasis, a novel strategy for cancer treatment by manipulating GABA receptors comes in view. Hence, GABA agonists and antagonists represent direct therapeutic strategies for cancer treatment (Table 2). The functional activity of GABAAR can be enhanced by GABAAR agonists, including benzodiazepines, barbiturates, zolpidem, and propofol, and can be attenuated by GABAAR antagonists, including bicuculline and flumazenil (which attach at the GABAAR recognition site) and picrotoxin (which attaches elsewhere on the receptor complex). 94 Like GABAAR, the activity of GABABR is also regulated by certain drugs. For instance, it can be activated by baclofen and competitively antagonized by phaclofen. A growing body of selective GABABR agonists and antagonists has been developed, such as the agonist CGP44532 and the antagonists CGP55845, CGP54626, and CGP35348.

TABLE 2.

The potential GABA agonists and antagonists as cancer therapy drugs.

Classification Name Target Cancer Effect Researches
Agonists Benzodiazepines GABAAR Medulloblastoma Inhibitory 109
Nembutal GABAAR Colon cancer Inhibitory 19
Propofol GABAAR Lung cancer, breast cancer Promotive 98, 99
Baclofen GABABR Colon cancer, Hepatocellular carcinoma Inhibitory 59, 96
Breast cancer, prostate cancer Promotive 74
CGP7930 GABABR Prostate cancer Promotive 74
Antagonists Bicuculline GABAAR Pancreatic Cancer Inhibitory 30
Flumazenil GABAAR Metastatic neuroendocrine cancers Inhibitory 97
Picrotoxin GABAAR Prostate cancer Inhibitory 54
CGP55845 GABABR Breast cancer Inhibitory 79
CGP54626 GABABR Prostate cancer, high‐grade chondrosarcoma Inhibitory 74, 75
CGP35348 GABABR Prostate cancer Inhibitory 18

Nembutal, a barbiturate, exerts agonistic effects on GABA and is a common anesthetic, hypnotic, and anticonvulsive drug. 95 Thaker et al 19 first confirmed that nembutal effectively inhibits both primary development and metastasis of colon cancer by promoting apoptosis in vivo. Further, Joseph et al 59 used baclofen as a specific GABABR agonist to inhibit the norepinephrine‐induced migration of colon carcinoma cells. Wang et al 96 demonstrated that baclofen also suppresses HCC tumor cell growth both in vitro and in vivo, and these inhibitory effects can be abrogated by pretreatment with phaclofen. Baclofen may function by suppressing cell proliferation with G0/G1 phase arrest by inhibiting cyclic adenosine monophosphate (cAMP) and downregulating the protein level and phosphorylation of p21WAF1. 96 In prostate cancer, although baclofen had no effect on tumor growth, GABAAR antagonists such as picrotoxin have demonstrated antitumor efficacy. 54 , 73 Similar anti‐proliferative effects have been obtained using GABAAR antagonists such as flumazenil and bicuculline in cancers. 30 , 97 GABABR antagonists, including CGP55845, CGP54626, and CGP35348, can inhibit GABABR‐induced migration. 18 , 74 , 75 , 79 Kanbara et al 75 confirmed that these antitumor effects may be implemented via different signaling pathways, including Ca2+ channels, cell cycle arrest, and apoptosis in cancer cells.

Although GABA receptor agonists and antagonists have significant benefits on the behavior of tumors by suppressing the proliferation and metastasis of cancer cells, their dual effect on cancers cannot be ignored. Contrary to the inhibitory effect of baclofen (GABABR agonist) in colon and liver cancers, baclofen can exert the opposite effect on other cancers. Baclofen significantly promoted cell invasion and migration in vitro and metastasis in vivo. 74 , 79 The GABAAR agonist propofol activates GABAAR to downregulate the expression of the tripartite motif (TRIM)21, consequently upregulating the expression of Src, a protein that regulates cell adhesion and extension, causing the potentiation of tumor metastasis in vivo. 98 Similarly, breast carcinoma cells also responded to propofol with increased migration which was mediated by calcium influx and actin reorganization. 99 As baclofen and propofol are both commonly used anesthetics in surgical tumor resection, their pro‐metastatic effects need careful deliberation for the surgeon.

These studies provide a novel‐targeted treatment of individuals using GABA receptor agonists or antagonists modulating GABA for the prevention of several cancers, as well as suggest clinical considerations for cancer metastasis. While several existing agonists and antagonists can selectively target GABAAR and GABABR, 100 , 101 , 102 it is vital to maximize the utilization of appropriate compositions to optimize antitumor efficacy, owing to the significantly varying expressions of different receptor subtypes on cancer cells. These studies are needed to facilitate the development of various structural types of GABA agonists or antagonists for the treatment of cancers.

5. CONCLUSIONS AND PERSPECTIVE

GABAergic agents significantly vary from cancer tissues to paired noncancerous tissues and exert regulatory effects on the biological characteristics of cancer cells. The distinct effects of GABA receptors on cancer progression in multiple tumor types depend on both receptor type and physiological context. In most cases, GABA regulates cancer cell proliferation through the GABAAR pathway but regulates cancer cell invasion through the GABABR pathway. Moreover, the regulatory signaling downstream of GABA receptors includes cAMP, EGFR, AKT, MAPK/ERK, and MMP. Presumably, the disparate subunits of GABA receptors target their specific downstream signaling to modulate cancer progression. The function and mechanism of GABAARP, a subunit of GABA, are thoroughly described in multiple cancers (including PDAC, breast cancer, ovarian cancer, and OSCC), which underscores the pivotal role of GABAAR in cancer progression, with more subunits needing similar elucidation. A normal human body can maintain a balance in the GABAergic system. However, the altered expression of GABAergic components in the peripheral organs indicated the control of the cancer microenvironment on the proliferation of normal and cancer cells. Studies by Zhang et al and Huang et al 27 add insights into how GABAergic components influence the immune microenvironment and tumor progression. This provides evidence for a novel therapeutic strategy for the treatment of cancers by modulating the GABAergic system.

Given the widespread distribution of GABAergic neurons in the nervous system, the most common side effects of drugs associated with GABA receptors include impaired CNS functions, such as sedation, ataxia, motor incoordination, anterograde amnesia, and paradoxical excitement. For instance, existing GABAAR inhibitors such as picrotoxin and bicuculline can induce severe convulsions in vivo due to their effect on GABAAR in the CNS. 103 Therefore, an important strategy to avoid the risk of severe adverse reactions involves the development of selective GABA receptor agonists and antagonists that are highly specific to the receptor subunits (e.g., GABAARP) and cannot penetrate the blood–brain barrier. Anesthetic drugs such as baclofen and propofol have an indispensable effect on tumor growth and metastasis, which is related to GABA receptors. These findings will attract more attention to the correlation between anesthesia and cancer metastasis, facilitating better prognosis for patients undergoing tumor resection.

In conclusion, the expression and molecular functions of GABA and its receptors in various cancers suggest that the GABAergic system is a potential target for anticancer therapy. This review provides an avenue for the development of more selective drugs that manipulate the activity of GABA receptors.

AUTHOR CONTRIBUTIONS

Tianjiao Li: Conceptualization (lead); data curation (equal); methodology (equal); project administration (equal); resources (equal); visualization (equal); writing – original draft (lead); writing – review and editing (lead). Jian Jiang: Conceptualization (equal); investigation (equal); methodology (equal); writing – original draft (supporting); writing – review and editing (supporting). Yaling Tang: Conceptualization (lead); formal analysis (equal); funding acquisition (lead); writing – review and editing (lead). Xinhua Liang: Conceptualization (lead); funding acquisition (lead); investigation (lead); resources (equal); writing – review and editing (lead).

CONFLICT OF INTEREST STATEMENT

The authors have no conflict of interest to declare.

ACKNOWLEDGMENTS

This work was supported by National Natural Science Foundation of China grants (Nos. 82073000, 82173326), National Science Foundation of Sichuan Province (Nos. 2022YFS0289 and 2022NSFSC0687), and Exploration and Research Projects of West China College of Stomatology, Sichuan University (LCYJ2020‐YJ‐1).

Li T‐J, Jiang J, Tang Y‐L, Liang X‐H. Insights into the leveraging of GABAergic signaling in cancer therapy. Cancer Med. 2023;12:14498‐14510. doi: 10.1002/cam4.6102

Tian‐Jiao Li and Jian Jiang contributed equally to this work.

Contributor Information

Ya‐Ling Tang, Email: tangyaling@scu.edu.cn.

Xin‐Hua Liang, Email: lxh88866@scu.edu.cn.

DATA AVAILABILITY STATEMENT

Data availability is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  • 1. Sieghart W. Structure and pharmacology of gamma‐aminobutyric acidA receptor subtypes. Pharmacol Rev. 1995;47(2):181‐234. [PubMed] [Google Scholar]
  • 2. Kerr DI, Ong J. GABAB receptors. Pharmacol Ther. 1995;67(2):187‐246. [DOI] [PubMed] [Google Scholar]
  • 3. Bowery N. GABAB receptors and their significance in mammalian pharmacology. Trends Pharmacol Sci. 1989;10(10):401‐407. [DOI] [PubMed] [Google Scholar]
  • 4. Owens DF, Kriegstein AR. Is there more to GABA than synaptic inhibition? Nat Rev Neurosci. 2002;3(9):715‐727. [DOI] [PubMed] [Google Scholar]
  • 5. Kempson SA, Zhou Y, Danbolt NC. The betaine/GABA transporter and betaine: roles in brain, kidney, and liver. Front Physiol. 2014;5:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Toso L, Roberson R, Abebe D, Spong CY. Neuroprotective peptides prevent some alcohol‐induced alteration in gamma‐aminobutyric acid A‐beta3, which plays a role in cleft lip and palate and learning in fetal alcohol syndrome. Am J Obstet Gynecol. 2007;196(3):259.e1‐259.e5. [DOI] [PubMed] [Google Scholar]
  • 7. Ding R, Tsunekawa N, Obata K. Cleft palate by picrotoxin or 3‐MP and palatal shelf elevation in GABA‐deficient mice. Neurotoxicol Teratol. 2004;26(4):587‐592. [DOI] [PubMed] [Google Scholar]
  • 8. Chintagari NR, Jin N, Gao L, Wang Y, Xi D, Liu L. Role of GABA receptors in fetal lung development in rats. PLoS One. 2010;5(11):e14171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Jin N, Guo Y, Sun P, et al. Ionotropic GABA receptor expression in the lung during development. Gene Expr Patterns. 2008;8(6):397‐403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Seifi M, Swinny JD. Developmental and age‐dependent plasticity of GABA(A) receptors in the mouse colon: implications in colonic motility and inflammation. Auton Neurosci. 2019;221:102579. [DOI] [PubMed] [Google Scholar]
  • 11. Wang FY, Watanabe M, Zhu RM, Maemura K. Characteristic expression of gamma‐aminobutyric acid and glutamate decarboxylase in rat jejunum and its relation to differentiation of epithelial cells. World J Gastroenterol. 2004;10(24):3608‐3611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Soltani N, Qiu H, Aleksic M, et al. GABA exerts protective and regenerative effects on islet beta cells and reverses diabetes. Proc Natl Acad Sci USA. 2011;108(28):11692‐11697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Shilpa J, Pretty MA, Anitha M, Paulose CS. Gamma aminobutyric acid B and 5‐hydroxy tryptamine 2A receptors functional regulation during enhanced liver cell proliferation by GABA and 5‐HT chitosan nanoparticles treatment. Eur J Pharmacol. 2013;715(1–3):154‐163. [DOI] [PubMed] [Google Scholar]
  • 14. Kanbara K, Okamoto K, Nomura S, et al. Cellular localization of GABA and GABAB receptor subunit proteins during spermiogenesis in rat testis. J Androl. 2005;26(4):485‐493. [DOI] [PubMed] [Google Scholar]
  • 15. Kanbara K, Okamoto K, Nomura S, Kaneko T, Watanabe M, Otsuki Y. The cellular expression of GABA(a) receptor alpha1 subunit during spermatogenesis in the mouse testis. Histol Histopathol. 2010;25(10):1229‐1238. [DOI] [PubMed] [Google Scholar]
  • 16. Andäng M, Hjerling‐Leffler J, Moliner A, et al. Histone H2AX‐dependent GABA (A) receptor regulation of stem cell proliferation. Nature. 2008;451(7177):460‐464. [DOI] [PubMed] [Google Scholar]
  • 17. Li YH, Liu Y, Li YD, et al. GABA stimulates human hepatocellular carcinoma growth through overexpressed GABAA receptor theta subunit. World J Gastroenterol. 2012;18(21):2704‐2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Azuma H, Inamoto T, Sakamoto T, et al. Gamma‐aminobutyric acid as a promoting factor of cancer metastasis; induction of matrix metalloproteinase production is potentially its underlying mechanism. Cancer Res. 2003;63(23):8090‐8096. [PubMed] [Google Scholar]
  • 19. Thaker PH, Yokoi K, Jennings NB, et al. Inhibition of experimental colon cancer metastasis by the GABA‐receptor agonist nembutal. Cancer Biol Ther. 2005;4(7):753‐758. [DOI] [PubMed] [Google Scholar]
  • 20. Al‐Wadei HA, Ullah MF, Al‐Wadei M. GABA (γ‐aminobutyric acid), a non‐protein amino acid counters the β‐adrenergic cascade‐activated oncogenic signaling in pancreatic cancer: a review of experimental evidence. Mol Nutr Food Res. 2011;55(12):1745‐1758. [DOI] [PubMed] [Google Scholar]
  • 21. Schuller HM, Al‐Wadei HA. Neurotransmitter receptors as central regulators of pancreatic cancer. Future Oncol. 2010;6(2):221‐228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Abdul M, McCray SD, Hoosein NM. Expression of gamma‐aminobutyric acid receptor (subtype A) in prostate cancer. Acta Oncol. 2008;47(8):1546‐1550. [DOI] [PubMed] [Google Scholar]
  • 23. Maemura K, Shiraishi N, Sakagami K, et al. Proliferative effects of gamma‐aminobutyric acid on the gastric cancer cell line are associated with extracellular signal‐regulated kinase 1/2 activation. J Gastroenterol Hepatol. 2009;24(4):688‐696. [DOI] [PubMed] [Google Scholar]
  • 24. Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic inhibitory interneurons. Physiol Rev. 2017;97(4):1619‐1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Petroff OA. GABA and glutamate in the human brain. Neuroscientist. 2002;8(6):562‐573. [DOI] [PubMed] [Google Scholar]
  • 26. Lange MD, Jüngling K, Paulukat L, et al. Glutamic acid decarboxylase 65: a link between GABAergic synaptic plasticity in the lateral amygdala and conditioned fear generalization. Neuropsychopharmacology. 2014;39(9):2211‐2220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Huang D, Wang Y, Thompson JW, et al. Cancer‐cell‐derived GABA promotes β‐catenin‐mediated tumour growth and immunosuppression. Nat Cell Biol. 2022;24(2):230‐241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kleinrok Z, Matuszek M, Jesipowicz J, Matuszek B, Opolski A, Radzikowski C. GABA content and GAD activity in colon tumors taken from patients with colon cancer or from xenografted human colon cancer cells growing as s.c. tumors in athymic nu/nu mice. J Physiol Pharmacol. 1998;49(2):303‐310. [PubMed] [Google Scholar]
  • 29. Opolski A, Mazurkiewicz M, Wietrzyk J, Kleinrok Z, Radzikowski C. The role of GABA‐ergic system in human mammary gland pathology and in growth of transplantable murine mammary cancer. J Exp Clin Cancer Res. 2000;19(3):383‐390. [PubMed] [Google Scholar]
  • 30. Takehara A, Hosokawa M, Eguchi H, et al. Gamma‐aminobutyric acid (GABA) stimulates pancreatic cancer growth through overexpressing GABAA receptor pi subunit. Cancer Res. 2007;67(20):9704‐9712. [DOI] [PubMed] [Google Scholar]
  • 31. Lee YY, Chao TB, Sheu MJ, et al. Glutamate decarboxylase 1 overexpression as a poor prognostic factor in patients with nasopharyngeal carcinoma. J Cancer. 2016;7(12):1716‐1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Soejima S, Kondo K, Tsuboi M, et al. GAD1 expression and its methylation as indicators of malignant behavior in thymic epithelial tumors. Oncol Lett. 2021;21(6):483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Yan H, Tang G, Wang H, et al. DNA methylation reactivates GAD1 expression in cancer by preventing CTCF‐mediated polycomb repressive complex 2 recruitment. Oncogene. 2016;35(30):3995‐4008. [DOI] [PubMed] [Google Scholar]
  • 34. Chen X, Cao Q, Liao R, et al. Loss of ABAT‐mediated GABAergic system promotes basal‐like breast cancer progression by activating Ca(2+)‐NFAT1 axis. Theranostics. 2019;9(1):34‐47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Han H, Zhou S, Chen G, Lu Y, Lin H. ABAT targeted by miR‐183‐5p regulates cell functions in liver cancer. Int J Biochem Cell Biol. 2021;141:106116. [DOI] [PubMed] [Google Scholar]
  • 36. Lu J, Chen Z, Zhao H, et al. ABAT and ALDH6A1, regulated by transcription factor HNF4A, suppress tumorigenic capability in clear cell renal cell carcinoma. J Transl Med. 2020;18(1):101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Connolly CN, Krishek BJ, McDonald BJ, Smart TG, Moss SJ. Assembly and cell surface expression of heteromeric and homomeric gamma‐aminobutyric acid type A receptors. J Biol Chem. 1996;271(1):89‐96. [DOI] [PubMed] [Google Scholar]
  • 38. Sieghart W. Allosteric modulation of GABAA receptors via multiple drug‐binding sites. Adv Pharmacol. 2015;72:53‐96. [DOI] [PubMed] [Google Scholar]
  • 39. Rebrov IG, Kryzhanovskiĭ GN, Belykh NP, Shukalova TF, Glebov RN. The effect of picrotoxin and bicuculline on desensitization of the GABA receptor/Cl‐ionophore complex. Biull Eksp Biol Med. 1994;118(8):160‐163. [PubMed] [Google Scholar]
  • 40. Ghansah E, Weiss DS. Modulation of GABA(A) receptors by benzodiazepines and barbiturates is autonomous of PKC activation. Neuropharmacology. 2001;40(3):327‐333. [DOI] [PubMed] [Google Scholar]
  • 41. Shiratsuchi H, Kouatli Y, Yu GX, Marsh HM, Basson MD. Propofol inhibits pressure‐stimulated macrophage phagocytosis via the GABAA receptor and dysregulation of p130cas phosphorylation. Am J Physiol Cell Physiol. 2009;296(6):C1400‐C1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Bettler B, Kaupmann K, Mosbacher J, Gassmann M. Molecular structure and physiological functions of GABA(B) receptors. Physiol Rev. 2004;84(3):835‐867. [DOI] [PubMed] [Google Scholar]
  • 43. Mintz IM, Bean BP. GABAB receptor inhibition of P‐type Ca2+ channels in central neurons. Neuron. 1993;10(5):889‐898. [DOI] [PubMed] [Google Scholar]
  • 44. Gähwiler BH, Brown DA. GABAB‐receptor‐activated K+ current in voltage‐clamped CA3 pyramidal cells in hippocampal cultures. Proc Natl Acad Sci USA. 1985;82(5):1558‐1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Urwyler S, Gjoni T, Koljatić J, Dupuis DS. Mechanisms of allosteric modulation at GABAB receptors by CGP7930 and GS39783: effects on affinities and efficacies of orthosteric ligands with distinct intrinsic properties. Neuropharmacology. 2005;48(3):343‐353. [DOI] [PubMed] [Google Scholar]
  • 46. Minuk GY. Gamma‐aminobutyric acid and the liver. Dig Dis. 1993;11(1):45‐54. [DOI] [PubMed] [Google Scholar]
  • 47. Kim J, Richter W, Aanstoot HJ, et al. Differential expression of GAD65 and GAD67 in human, rat, and mouse pancreatic islets. Diabetes. 1993;42(12):1799‐1808. [DOI] [PubMed] [Google Scholar]
  • 48. Yang W, Reyes AA, Lan NC. Identification of the GABAA receptor subtype mRNA in human pancreatic tissue. FEBS Lett. 1994;346(2–3):257‐262. [DOI] [PubMed] [Google Scholar]
  • 49. Napoleone P, Bronzetti E, Cavallotti C, Amenta F. Predominant epithelial localization of type A gamma‐aminobutyric acid receptor sites within rat seminal vesicles and prostate glands. Pharmacology. 1990;41(1):49‐56. [DOI] [PubMed] [Google Scholar]
  • 50. Whelan DT, Scriver CR, Mohyuddin F. Glutamic acid decarboxylase and gamma‐aminobutyric acid in mammalian kidney. Nature. 1969;224(5222):916‐917. [DOI] [PubMed] [Google Scholar]
  • 51. Poulter MO, Singhal R, Brown LA, Krantis A. GABA(A) receptor subunit messenger RNA expression in the enteric nervous system of the rat: implications for functional diversity of enteric GABA(A) receptors. Neuroscience. 1999;93(3):1159‐1165. [DOI] [PubMed] [Google Scholar]
  • 52. Geigerseder C, Doepner R, Thalhammer A, et al. Evidence for a GABAergic system in rodent and human testis: local GABA production and GABA receptors. Neuroendocrinology. 2003;77(5):314‐323. [DOI] [PubMed] [Google Scholar]
  • 53. Erdö SL, László A. High specific gamma‐aminobutyric acid binding to membranes of the human ovary. J Neurochem. 1984;42(5):1464‐1467. [DOI] [PubMed] [Google Scholar]
  • 54. Wu W, Yang Q, Fung KM, et al. Linking γ‐aminobutyric acid A receptor to epidermal growth factor receptor pathways activation in human prostate cancer. Mol Cell Endocrinol. 2014;383(1–2):69‐79. [DOI] [PubMed] [Google Scholar]
  • 55. Sung HY, Yang SD, Ju W, Ahn JH. Aberrant epigenetic regulation of GABRP associates with aggressive phenotype of ovarian cancer. Exp Mol Med. 2017;49(5):e335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Ma J, Zhang Y, Wang J, et al. Proliferative effects of gamma‐amino butyric acid on oral squamous cell carcinoma cells are associated with mitogen‐activated protein kinase signaling pathways. Int J Mol Med. 2016;38(1):305‐311. [DOI] [PubMed] [Google Scholar]
  • 57. Zhang X, Zhang R, Zheng Y, et al. Expression of gamma‐aminobutyric acid receptors on neoplastic growth and prediction of prognosis in non‐small cell lung cancer. J Transl Med. 2013;11:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Schuller HM, Al‐Wadei HA, Majidi M. Gamma‐aminobutyric acid, a potential tumor suppressor for small airway‐derived lung adenocarcinoma. Carcinogenesis. 2008;29(10):1979‐1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Joseph J, Niggemann B, Zaenker KS, Entschladen F. The neurotransmitter gamma‐aminobutyric acid is an inhibitory regulator for the migration of SW 480 colon carcinoma cells. Cancer Res. 2002;62(22):6467‐6469. [PubMed] [Google Scholar]
  • 60. Fava G, Marucci L, Glaser S, et al. Gamma‐Aminobutyric acid inhibits cholangiocarcinoma growth by cyclic AMP‐dependent regulation of the protein kinase A/extracellular signal‐regulated kinase 1/2 pathway. Cancer Res. 2005;65(24):11437‐11446. [DOI] [PubMed] [Google Scholar]
  • 61. Liu Y, Li YH, Guo FJ, et al. Gamma‐aminobutyric acid promotes human hepatocellular carcinoma growth through overexpressed gamma‐aminobutyric acid A receptor alpha 3 subunit. World J Gastroenterol. 2008;14(47):7175‐7182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Minuk GY, Zhang M, Gong Y, et al. Decreased hepatocyte membrane potential differences and GABAA‐beta3 expression in human hepatocellular carcinoma. Hepatology. 2007;45(3):735‐745. [DOI] [PubMed] [Google Scholar]
  • 63. Lodewyks C, Rodriguez J, Yan J, et al. GABA‐B receptor activation inhibits the in vitro migration of malignant hepatocytes. Can J Physiol Pharmacol. 2011;89(6):393‐400. [DOI] [PubMed] [Google Scholar]
  • 64. Schuller HM, Al‐Wadei HA, Majidi M. GABA B receptor is a novel drug target for pancreatic cancer. Cancer. 2008;112(4):767‐778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Wei B, Zhu Y, Yang P, et al. GABA(B1e) promotes the malignancy of human cancer cells by targeting the tyrosine phosphatase PTPN12. iScience. 2021;24(11):103311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Drell T, Joseph J, Lang K, Niggemann B, Zaenker KS, Entschladen F. Effects of neurotransmitters on the chemokinesis and chemotaxis of MDA‐MB‐468 human breast carcinoma cells. Breast Cancer Res Treat. 2003;80(1):63‐70. [DOI] [PubMed] [Google Scholar]
  • 67. Sizemore GM, Sizemore ST, Seachrist DD, Keri RA. GABA(A) receptor pi (GABRP) stimulates basal‐like breast cancer cell migration through activation of extracellular‐regulated kinase 1/2 (ERK1/2). J Biol Chem. 2014;289(35):24102‐24113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Gumireddy K, Li A, Kossenkov AV, et al. The mRNA‐edited form of GABRA3 suppresses GABRA3‐mediated Akt activation and breast cancer metastasis. Nat Commun. 2016;7:10715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Roberts SS, Mendonça‐Torres MC, Jensen K, Francis GL, Vasko V. GABA receptor expression in benign and malignant thyroid tumors. Pathol Oncol Res. 2009;15(4):645‐650. [DOI] [PubMed] [Google Scholar]
  • 70. Zhang H, Kong Q, Wang J, Jiang Y, Hua H. Complex roles of cAMP‐PKA‐CREB signaling in cancer. Exp Hematol Oncol. 2020;9(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Zhou YQ, Chen SP, Liu DQ, et al. The role of spinal GABAB receptors in cancer‐induced bone pain in rats. J Pain. 2017;18(8):933‐946. [DOI] [PubMed] [Google Scholar]
  • 72. Gallick GE, Corn PG, Zurita AJ, Lin SH. Small‐molecule protein tyrosine kinase inhibitors for the treatment of metastatic prostate cancer. Future Med Chem. 2012;4(1):107‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Xia D, Lai DV, Wu W, et al. Transition from androgenic to neurosteroidal action of 5α‐androstane‐3α, 17β‐diol through the type A γ‐aminobutyric acid receptor in prostate cancer progression. J Steroid Biochem Mol Biol. 2018;178:89‐98. [DOI] [PubMed] [Google Scholar]
  • 74. Xia S, He C, Zhu Y, et al. GABA(B)R‐induced EGFR transactivation promotes migration of human prostate cancer cells. Mol Pharmacol. 2017;92(3):265‐277. [DOI] [PubMed] [Google Scholar]
  • 75. Kanbara K, Otsuki Y, Watanabe M, et al. GABA(B) receptor regulates proliferation in the high‐grade chondrosarcoma cell line OUMS‐27 via apoptotic pathways. BMC Cancer. 2018;18(1):263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Marshall CJ. Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal‐regulated kinase activation. Cell. 1995;80(2):179‐185. [DOI] [PubMed] [Google Scholar]
  • 77. Widmann C, Gibson S, Jarpe MB, Johnson GL. Mitogen‐activated protein kinase: conservation of a three‐kinase module from yeast to human. Physiol Rev. 1999;79(1):143‐180. [DOI] [PubMed] [Google Scholar]
  • 78. Rosen LB, Ginty DD, Weber MJ, Greenberg ME. Membrane depolarization and calcium influx stimulate MEK and MAP kinase via activation of Ras. Neuron. 1994;12(6):1207‐1221. [DOI] [PubMed] [Google Scholar]
  • 79. Zhang D, Li X, Yao Z, Wei C, Ning N, Li J. GABAergic signaling facilitates breast cancer metastasis by promoting ERK1/2‐dependent phosphorylation. Cancer Lett. 2014;348(1–2):100‐108. [DOI] [PubMed] [Google Scholar]
  • 80. Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174. [DOI] [PubMed] [Google Scholar]
  • 81. Kimura R, Kasamatsu A, Koyama T, et al. Glutamate acid decarboxylase 1 promotes metastasis of human oral cancer by β‐catenin translocation and MMP7 activation. BMC Cancer. 2013;13:555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Enna SJ. A GABA(B) mystery: the search for pharmacologically distinct GABA(B) receptors. Mol Interv. 2001;1(4):208‐218. [PubMed] [Google Scholar]
  • 83. Young SZ, Bordey A. GABA's control of stem and cancer cell proliferation in adult neural and peripheral niches. Physiology (Bethesda). 2009;24:171‐185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Neman J, Termini J, Wilczynski S, et al. Human breast cancer metastases to the brain display GABAergic properties in the neural niche. Proc Natl Acad Sci USA. 2014;111(3):984‐989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Geltink RIK, Kyle RL, Pearce EL. Unraveling the complex interplay between T cell metabolism and function. Annu Rev Immunol. 2018;36:461‐488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Makowski L, Chaib M, Rathmell JC. Immunometabolism: from basic mechanisms to translation. Immunol Rev. 2020;295(1):5‐14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Zhang B, Vogelzang A, Miyajima M, et al. B cell‐derived GABA elicits IL‐10(+) macrophages to limit anti‐tumour immunity. Nature. 2021;599(7885):471‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Mendu SK, Åkesson L, Jin Z, et al. Increased GABA(a) 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(4):399‐407. [DOI] [PubMed] [Google Scholar]
  • 89. Tian J, Dang H, O’Laco KA, et al. Homotaurine treatment enhances CD4(+) and CD8(+) regulatory T cell responses and synergizes with low‐dose anti‐CD3 to enhance diabetes remission in type 1 diabetic mice. Immunohorizons. 2019;3(10):498‐510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Tian J, Middleton B, Kaufman DL. GABA(A)‐receptor agonists limit pneumonitis and death in murine coronavirus‐infected mice. Viruses. 2021;13(6):966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Tian J, Song M, Kaufman DL. Homotaurine limits the spreading of T cell autoreactivity within the CNS and ameliorates disease in a model of multiple sclerosis. Sci Rep. 2021;11(1):5402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Bhat R, Axtell R, Mitra A, et al. Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci USA. 2010;107(6):2580‐2585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Tian J, Yong J, Dang H, Kaufman DL. Oral GABA treatment downregulates inflammatory responses in a mouse model of rheumatoid arthritis. Autoimmunity. 2011;44(6):465‐470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Möhler H, Fritschy JM, Crestani F, Hensch T, Rudolph U. Specific GABA(A) circuits in brain development and therapy. Biochem Pharmacol. 2004;68(8):1685‐1690. [DOI] [PubMed] [Google Scholar]
  • 95. Krampfl K, Wolfes H, Dengler R, Bufler J. Kinetic analysis of the agonistic and blocking properties of pentobarbital on recombinant rat alpha(1)beta(2)gamma(2S) GABA(A) receptor channels. Eur J Pharmacol. 2002;435(1):1‐8. [DOI] [PubMed] [Google Scholar]
  • 96. Wang T, Huang W, Chen F. Baclofen, a GABAB receptor agonist, inhibits human hepatocellular carcinoma cell growth in vitro and in vivo. Life Sci. 2008;82(9–10):536‐541. [DOI] [PubMed] [Google Scholar]
  • 97. Ippolito JE, Merritt ME, Bäckhed F, et al. Linkage between cellular communications, energy utilization, and proliferation in metastatic neuroendocrine cancers. Proc Natl Acad Sci USA. 2006;103(33):12505‐12510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Liu Q, Sheng Z, Cheng C, et al. Anesthetic propofol promotes tumor metastasis in lungs via GABA(a) R‐dependent TRIM21 modulation of Src expression. Adv Sci (Weinh). 2021;8(18):e2102079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Garib V, Lang K, Niggemann B, Zänker KS, Brandt L, Dittmar T. Propofol‐induced calcium signalling and actin reorganization within breast carcinoma cells. Eur J Anaesthesiol. 2005;22(8):609‐615. [DOI] [PubMed] [Google Scholar]
  • 100. Evenseth LSM, Gabrielsen M, Sylte I. The GABA(B) receptor‐structure, ligand binding and drug development. Molecules. 2020;25(13):3093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. George K, Preuss CV, Sadiq NM. GABA inhibitors. Treasure Island, FL; 2022. [PubMed] [Google Scholar]
  • 102. Jewett, B.E. , Sharma S. Physiology, GABA. StatPearls; 2018. [PubMed] [Google Scholar]
  • 103. Mares P, Chino M, Kubová H, Mathern P, Veliký M. Convulsant action of systemically administered glutamate and bicuculline methiodide in immature rats. Epilepsy Res. 2000;42(2–3):183‐189. [DOI] [PubMed] [Google Scholar]
  • 104. D'Urso PI, D'Urso OF, Storelli C, et al. miR‐155 is up‐regulated in primary and secondary glioblastoma and promotes tumour growth by inhibiting GABA receptors. Int J Oncol. 2012;41(1):228‐234. [DOI] [PubMed] [Google Scholar]
  • 105. Smits A, Jin Z, Elsir T, et al. GABA‐A channel subunit expression in human glioma correlates with tumor histology and clinical outcome. PLoS One. 2012;7(5):e37041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Al‐Wadei HA, Al‐Wadei MH, Schuller HM. Cooperative regulation of non‐small cell lung carcinoma by nicotinic and beta‐adrenergic receptors: a novel target for intervention. PLoS One. 2012;7(1):e29915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Nicholson‐Guthrie CS, Guthrie GD, Sutton GP, Baenziger JC. Urine GABA levels in ovarian cancer patients: elevated GABA in malignancy. Cancer Lett. 2001;162(1):27‐30. [DOI] [PubMed] [Google Scholar]
  • 108. Magnaghi V, Ballabio M, Cavarretta ITR, et al. GABAB receptors in Schwann cells influence proliferation and myelin protein expression. Eur J Neurosci. 2004;19(10):2641‐2649. [DOI] [PubMed] [Google Scholar]
  • 109. Sengupta S, Weeraratne SD, Sun H, et al. α5‐GABAA receptors negatively regulate MYC‐amplified medulloblastoma growth. Acta Neuropathol. 2014;127(4):593‐603. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data availability is not applicable to this article as no new data were created or analyzed in this study.


Articles from Cancer Medicine are provided here courtesy of Wiley

RESOURCES