Abstract
GABAB receptors mediate prolonged inhibition in the brain and are important for keeping neuronal excitation and inhibition in a healthy balance. However, under excitotoxic/ischemic conditions, GABAB receptors are downregulated by dysregulated endocytic trafficking and can no longer counteract the severely enhanced excitation, eventually triggering neuronal death. Recently, we developed interfering peptides targeting protein-protein interactions involved in downregulating the receptors. Treatment with these peptides restored GABAB receptor expression after an ischemic insult and thereby inhibited neuronal overexcitation and progressive neuronal death. In this study, we searched for GABAB receptor interactions that specifically occur under ischemic conditions. We found that the E3 ubiquitin ligase MARCH1 is specifically upregulated under ischemic/excitotoxic conditions. Upregulated MARCH1 interacts with GABAB receptors and triggered downregulation of plasma membrane GABAB receptors by inhibiting fast recycling of the receptors. We developed an interfering peptide that inhibits the MARCH1/GABAB receptor interaction. Treatment of cultured neurons subjected to ischemic stress with this peptide restored receptor expression and as a consequence stopped progressive neuronal death. Thus, inhibiting the interaction of GABAB receptors with MARCH1 to restore cell surface receptor expression might be a promising strategy to prevent progressive neuronal death induced by ischemic conditions.
Similar content being viewed by others
Introduction
A precise balance between neuronal excitation and inhibition is fundamental to proper brain function. In various neurological disorders, the excitation/inhibition balance is disturbed resulting most commonly in over-excitation of neurons. Prolonged neuronal over-excitation triggers detrimental pathways leading to excitotoxic neuronal death. One important component controlling neuronal excitation is the GABAB receptor1. GABAB receptors are heterodimeric G-protein coupled receptors (GPCR) consisting of the two subunits GABAB1 (exists as GABAB1a and GABAB1b variants) and GABAB22. GABAB receptors are expressed at pre- and postsynaptic locations in most neurons3. The binding of the neurotransmitter GABA to GABAB receptors activates Gi/o-proteins and induces prolonged neuronal inhibition by regulating the activity of several effector systems. In this regard, the most prominent ones are the activation of G protein-coupled inwardly rectifying potassium (GIRK or Kir3) channels at postsynaptic sites causing the hyperpolarization of the neuronal membrane and the inhibition of voltage-gated Ca2+ channels at presynaptic sites which reduces neurotransmitter release4.
In many neurological diseases with a disturbed excitation/inhibition balance - like addiction, anxiety, depression, neurodegenerative diseases, and cerebral ischemia - GABAB receptors are downregulated from the plasma membrane5,6,7,8. This withdraws an important contributor to neuronal inhibition, who normally ensures that neurons do not shift into a state of over-excitation. The mechanisms responsible for the pathological downregulation of plasma membrane GABAB receptors identified so far are caused by deviations from normal trafficking pathways of the receptors. Under physiological conditions, GABAB receptors are constitutively internalized and most receptors are recycled to the plasma membrane while a fraction is degraded in lysosomes9. The degraded receptors are then replaced by newly synthetized receptors exported from the endoplasmic reticulum (ER). Pathological conditions leading to neuronal over-excitation, such as cerebral ischemia, are associated with ER stress which induces the expression of the pro-apoptotic transcription factor CHOP. Upregulated CHOP bind to the GABAB2 subunit in the ER, prevents their heterodimerization with the GABAB1 subunit and thereby the exit of the heterodimeric receptor complex from the ER10. This mechanism inhibits the supply of new receptors and plasma membrane downregulates GABAB receptor expression because constitutively degraded receptors cannot be replaced. In addition, constitutively internalized GABAB receptors are dephosphorylated at GABAB2(S783) by protein phosphatase 2 A (PP2A), which inhibits fast recycling of internalized receptors to the plasma membrane11,12. Subsequent phosphorylation of GABAB1(S867/T872) mediated by calcium calmodulin-dependent kinase II β (CaMKIIβ) and extracellular-signal regulated kinases 1 and 2 (ERK1/2) triggers the sorting of the receptors to the lysosomal degradation pathway13,14. Preventing the interaction of GABAB receptors with either CHOP, CaMKIIβ or PP2A using interfering peptides restored plasma membrane receptor expression and normal GABAB receptor mediated inhibition after ischemic/excitotoxic stress, resulting in the inhibition of progressive neuronal death12,15,16. Hence, interfering with the interaction of disease relevant GABAB receptor interactions might be promising strategy for the development of specific therapeutic interventions. In case the interactions are specifically associated with the disease state, it might even be possible to selectively target the receptor population in diseased neurons. This should minimize unwanted side effects.
In the search for new protein-protein interactions that might be selectively occurring under pathological conditions and are involved in the downregulation of plasma membrane GABAB receptors, we screened for ubiquitin E3 ligases that affect the cell surface expression of the receptors. We focused on E3 ligases because the expression, trafficking and degradation of many GPCRs had been shown to be regulated by ubiquitination, which often acts as master signal to decide the fate of proteins to specific cellular destinations17. Ubiquitination of a target protein involves an enzymatic cascade consisting of E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme) and E3 ligase enzymes, where the E3 ligase largely determines the substrate specificity. Hence, we screened various transmembrane E3 ligases for their ability to affect GABAB receptor cell surface expression. We concentrated on transmembrane E3 ligases because they most likely reside in the same cellular compartment(s) as the receptors. We found that the E3 ligase MARCH1 (membrane-associated RING CH-type finger 1) downregulated cell surface expression of the receptors when co-expressed with GABAB receptors in HEK293 cells. We characterized the effect of MARCH1 on GABAB receptors and developed an interfering peptide (M1-Pep) that inhibits the interaction of GABAB receptors with MARCH1. M1-Pep inhibits the interaction of GABAB receptors with MARCH1 after ischemic stress, restored cell surface expression of the receptors and inhibited progressive neuronal death in cultured cortical neurons.
Results
Screening for transmembrane RING domain E3 ligases regulating GABAB receptor expression
Transmembrane RING domain E3 ubiquitin ligases have been shown to regulate the cell surface expression of membrane proteins such as AMPA receptors18, insulin receptors19, CD8620 and major histocompatibility complex class II (MHC II)21. Therefore, we screened various transmembrane RING domain E3 ligases (MARCH1, MARCH5, RNF112, RNF144, RNF152, RNF16718, MARCH822 and RNF13323) for their ability to affect the cell surface expression of GABAB receptors. For this, we transfected HEK-293 cells with GABAB1 and GABAB2 subunits along with wildtype E3 ligases or with their corresponding functionally inactive mutants. The cell surface expression of GABAB1 and GABAB2 was determined by immunofluorescence staining using antibodies directed against the extracellular located N-terminal domains of the receptor subunits. Among the tested transmembrane RING domain E3 ligases, only the co-expression of MARCH1 affected the cell surface expression of GABAB receptors. Co-expression of wildtype MARCH1 severely downregulated GABAB1 (Fig. 1a) and GABAB2 (Fig. 1b), whereas the inactive MARCH1 mutant (MARCH1(DN)) had no effect on GABAB receptor cell surface expression. These results indicated that MARCH1 is involved in regulating cell surface expression of GABAB receptors presumably by ubiquitination.
Screening for transmembrane RING domain E3 ligases regulating GABAB receptor cell surface expression. HEK-293 cells were transfected with plasmids for GABAB1, GABAB2 and EGFP as a control (Ctrl) or with GABAB1, GABAB2 and the indicated E3 ligases. After 2 days, the cells were tested for cell surface expression of GABAB1 (a) and GABAB2 (b) by immunofluorescence staining using antibodies directed against GABAB1 and GABAB2. (a) Co-expression of MARCH1 downregulated the cell surface expression of GABAB1.Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 85–90 cells per condition, 4 independent experiments). (b) Co-expression of MARCH1 downregulated the cell surface expression of GABAB2. Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 80–132 cells per condition, 4 independent experiments). Brown-Forsythe and Welch’s one-way ANOVA followed by Games-Howell’s multiple comparisons test (****, p < 0.0001).
MARCH1 downregulated cell surface expression of GABAB receptor by mono- or multi-monoubiquitination
To gain more insights into the mechanism of MARCH1 mediated downregulation of cell surface GABAB receptors, we transfected HEK-293 cells with GABAB1 and GABAB2 subunits along with wildtype MARCH1 or with its corresponding mutants MARCH1(DN) and MARCH1(KKXX). MARCH1(DN) is a dominant negative mutant of MARCH1, in which conserved histidine residues within the RING domain were replaced by tryptophan, preventing zinc coordination and thereby ubiquitin ligase activity18. MARCH1(KKXX) lacks 40 amino acids at the C-terminus, creating a classical C-terminal di-lysine ER-retention motive, which prevents MARCH1 to leave the ER24. Co-expression of GABAB receptors with MARCH1(DN) did not affect cell surface expression of the receptors, suggesting that E3-ligase activity – i.e. ubiquitination – is required for downregulating the receptors (Fig. 2a). Similarly, co-expression of GABAB receptors with MARCH1(KKXX) did not affect cell surface expression of GABAB1 and GABAB2 (Fig. 2a), indicating that MARCH1 induces downregulation of cell surface GABAB receptors in compartments outside the ER, e.g. the plasma membrane, early endosomes, recycling endosomes or Golgi apparatus. Interestingly, MARCH1 downregulated the surface expression of GABAB receptors (Fig. 2a) without affecting the total expression of GABAB1 and GABAB2 (Fig. 2b). Thus, MARCH1 is not primarily be involved in the degradation pathway of GABAB receptors.
Next, we tested whether MARCH1 ubiquitinates GABAB receptors. For this HEK-293 cells were transfected with GABAB1 and GABAB2 along with wild type MARCH1 or the dominant negative mutant MARCH1(DN) and tested for Lys63 -and Lys48-polyubiquitination - which are the most common types of polyubiquitination - by in situ PLA using selective antibodies. However, MARCH1 did not enhance Lys63- and Lys48-polyubiquitination of GABAB receptors (Fig. 2c). Then, we tested whether MARCH1 may mediate mono/multi-monoubiquitination by which a single ubiquitin moiety is attached to one (monoubiquitination) or multiple (multi-monoubiquitination) lysine residues in the target protein. For this, we used the same experimental setup described above but in addition co-transfected a mutant of ubiquitin (ubiquitin(KO)) in which all lysine residues were mutated so that it is unable to build polyubiquitin chains. Under control conditions (only GABAB receptors transfected) and upon transfection with the inactive MARCH1 mutant (MARCH1(DN)) we detected only very few dots, which most likely represented nonspecific PLA signals. However, in the presence of functional MARCH1 numerous PLA signals were observed, indicating that MARCH1 most likely mediates mono- or multi-monoubiquitination of the receptors (Fig. 2c).
It is reasonable to assume that MARCH1 need to associate with GABAB receptors for their ubiquitination. Using in situ PLA, we therefore tested whether MARCH1 interacts with GABAB receptors when co-expressed in HEK-293 cells. As expected, co-expression of MARCH1 led to numerous PLA signals, suggesting the direct or indirect interaction of MARCH1 with the receptor (Fig. 2d).
We then tested whether MARCH1 affects the expression of GABAB1 or GABAB2 subunits when expressed alone without its counterpart. For this, HEK-293 cells were transfected with MARCH1 and either GABAB1(RSAR), in which the ER retention signal was mutated to allow its exit from ER and cell surface expression in the absence of GABAB2, or with GABAB2. Interestingly, MARCH1 does not appear to substantially affect cell surface expression of GABAB1 in the absence of GABAB2 and vice versa (Fig. 2e). This finding suggests that MARCH1 only affects the expression of the assembled GABAB receptor heterodimer.
MARCH1 induced downregulation of cell surface GABAB receptors requires functional MARCH1 located outside the ER and mono/multi-monoubiquitination. (a, b) HEK-293 cells transfected with GABAB1, GABAB2 and EGFP as a control (Ctrl) or with GABAB1, GABAB2 and wildtype MARCH1 (M1) or with the non-functional mutant MARCH1(DN) or the mutant MARCH1(KKXX) that cannot leave the ER were tested for cell surface (a) or total receptor expression (b). Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 84–85 cells per condition, 3 independent experiments). Kruskal-Wallis test followed by Dunn’s multiple comparisons test (ns, p > 0.05; **, < 0.01; ****, p < 0.0001). (c) MARCH1 mono/multiubiquitinates GABAB receptors. HEK-293 cells expressing GABAB1/GABAB2/EGFP as a control (Ctrl) or GABAB1/GABAB2/MARCH1 or GABAB1/GABAB2/MARCH1(DN) with or without a mutant of HA-tagged ubiquitin that cannot form polyubiquitin chains (Ub(KO)) were tested for ubiquitination of GABAB receptors by in situ PLA using antibodies directed against GABAB1 and Lys63 polyubiquitin or Lys48 polyubiquitin or HA. Top: representative images (in situ PLA signals: white dots, scale bar: 10 μm). Bottom: quantification of in situ PLA signals (mean ± SD of 40–50 cells per condition, 2 independent experiments). Lys48: one way ANOVA followed by Tukey’s multiple comparison test; Lys63 and Ub(KO): Brown-Forsythe and Welch’s one-way ANOVA followed by Games-Howell’s multiple comparisons test (ns, p > 0.05; ****, p < 0.0001). (d) MARCH1 interacts with GABAB receptors. HEK-293 cells expressing GABAB1/GABAB2 (control for non-specific PLA signal (Ctrl)) or GABAB1/GABAB2/MARCH1 were tested for interaction by in situ PLA using antibodies directed against MARCH1 and GABAB1 or GABAB2 (top images: total GABAB staining, bottom images: PLA signals; scale bar: 5 μm). The images shown are representative for 90 cells analyzed per condition from 3 independent experiments. (e) MARCH1 does not affect the cell surface expression of single GABAB receptor subunits. HEK-293 cells were transfected with MARCH1/GABAB1/GABAB2 as a control (Ctrl) or with MARCH1/GABAB2 or with MARCH1 and a GABAB1 mutant that can leave the ER (GABAB1(RSAR)) and tested for cell surface expression of GABAB1 and GABAB2. Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 75 cells per condition, 3 independent experiments). Unpaired t-test with Welch’s correction (****, p < 0.0001).
MARCH1 was upregulated under ischemic conditions in neurons
MARCH1 mRNA has been found to be highly expressed in spleen and lung tissue and comparatively less in muscle, liver, and brain25,26. To verity the expression of MARCH1 protein in neurons, we co-stained neuron/glia cultures with antibodies directed against MARCH1 and the neuron-specific marker protein NeuN (Fig. 3a). We found that all neurons tested also expressed MARCH1 signals (1074 neurons analyzed). Thus, MARCH1 appears to be ubiquitously expressed in neurons.
GABAB receptors are significantly downregulated under ischemic/excitotoxic conditions11,12,13,14,15,16,27,28. Hence, it is plausible that upregulation of MARCH1 under ischemic condition might contribute to this pathological pathway. To test for this, we subjected cultured neurons to ischemia-like oxygen and glucose deprivation (OGD) for different time intervals and evaluated the expression of MARCH1 by immunofluorescence staining. The expression of MARCH1 was significantly upregulated after 1 h of OGD (Fig. 3b). Hence, the upregulation of MARCH1 could be involved in the pathway downregulating the GABAB receptors under ischemic conditions.
MARCH1 is expressed in neurons and is upregulated under ischemic conditions. (a) All neurons tested expressed MARCH1. The expression of MARCH1 in neuron/glia co-cultures was tested by immunofluorescence staining using antibodies directed against NeuN (marker protein for neurons) and MARCH1 (scale bar: 50 μm). (b) MARCH1 is significantly upregulated after 1 h of OGD. Neuron/glia co-cultures were subjected to OGD for 30 min–1 h and tested for MARCH1 expression by immunofluorescence staining using antibodies directed against MARCH1. Cultures not treated with OGD served as a control. Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 15 cells per condition, 2 independent experiments). One-way ANOVA followed by Dunnett’s multiple comparison test (ns, p > 0.05; **, p < 0.01).
Identification of interfering peptides for restoring MARCH1 induced downregulation of cell surface GABAB receptors
To further elucidate the function of MARCH1 on GABAB receptors, we screened a set of synthetic overlapping peptides covering the C-terminal domains of GABAB1 and GABAB2 for their ability to inhibit the interaction of GABAB receptors with MARCH1, thereby restoring MARCH1 induced downregulation of plasma membrane GABAB receptors (Fig. 4a). All peptides contained 8 arginine at their N-terminus to render them cell permeable. The screening yielded 2 peptides, one comprising the GABAB1 sequence AEKEERVSELRHQLQSRQQL (1–4) and one comprising the GABAB2 sequence DKDLEEVTMQLQDTPEKTTY (2–5). However, the GABAB2 derived peptide (2–5) restored the downregulated GABAB receptor more reliably than the GABAB1 derived peptide (1–4). This observation was substantiated in dose-response experiments of these two peptides (Fig. 4b). We found that 10 µg/ml peptide exerted the maximum effect on restoring GABAB receptor expression with peptide 2–5 being more effective than peptide 1–4 (Fig. 4b).
Because the GABAB2 peptide sequence 2–5 is in proximity to the GABAB receptor/CHOP interaction site16,29, we checked the specificity of peptide 2–5 by testing its ability to reverse CHOP-induced downregulation of plasma membrane GABAB receptors in a co-transfection assay in HEK 293 cells. Peptide 2–5 did not restore the CHOP mediated downregulation of cell surface GABAB receptors and therefore did not affect the interaction of CHOP with GABAB receptors (Fig. 4c).
In all further experiments, we used peptide 2–5 tagged at the N-terminus with a peptide sequence derived from the Rabies virus glycoprotein (RVG), which renders it cell permeable in a neuron-specific manner via a receptor-mediated uptake mechanism15,30,31. The RVG-2-5 peptide is named M1-Pep in the rest of the study. In major experiments, the specificity of M1-Pep was verified against a control peptide (Ctrl-Pep). Ctrl-Pep consists of the RVG sequence followed by the 2–5 amino acids in a random sequence.
Identification of an interfering peptide for restoring MARCH1 induced downregulation of plasma membrane GABAB receptors. (a) Screening for interfering peptides. HEK 293-cells expressing GABAB1/GABAB2 as a control (Ctrl) or GABAB1/GABAB2/MARCH1 were treated with the indicated peptides (10 µg/ml) and tested for cell surface expression of receptors using antibodies directed against and GABAB1 and GABAB2. Peptides 1–4 and 2–5 restored the cell surface expression of the receptors. Left: sequences of peptides used for screening derived from the C-terminal domains of GABAB1 and GABAB2. Right: quantification of fluorescence intensities (mean ± SD of 40–112 cells per condition, 2 independent experiments). Kruskal-Wallis test followed by Dunn’s multiple comparison test (ns, p > 0.05; *, p < 0.05; **, p < 0.01 ***, p < 0.001; ****, p < 0.0001). (b) Dose-response of peptides 1–4 and 2–5. HEK-293 cells expressing GABAB1/GABAB2/MARCH1 were treated with increasing concentrations of the respective peptides and were tested the following day for cell surface expression of GABAB2. The data represents the mean ± SD of 30 cells per condition from 2 independent experiments. One-way ANOVA followed by Dunnett’s multiple comparison test (ns, p > 0.05; ****, p < 0.0001) (c) Peptide 2–5 did not restore the CHOP mediated downregulation of GABAB receptors. HEK-293 cells expressing GABAB1/GABAB2 as a control (Ctrl) or GABAB1/GABAB2/CHOP were treated with peptide 2–5 (10 µg) and tested for cell surface expression of GABAB2 the next day. The data represents the mean ± SD of 93–103 cells per condition from 3 independent experiments. Brown-Forsythe and Welch’s one-way ANOVA followed by Games-Howell’s multiple comparisons test (****, p < 0.0001) (d) Treatment of neurons with M1-Pep does not affect cell surface expression of native GABAB receptors under physiological conditions. Neuron/glia co-cultures were treated for 16 h with or without M1-Pep or Ctrl-Pep and tested for cell surface (using GABAB2 antibodies) and total GABAB receptor (using GABAB1 antibodies) expression. Top: representative images (scale bar: 10 μm). Bottom: quantification of fluorescence intensities (mean ± SD of 25 cells per condition, 2 independent experiments). One-way ANOVA followed by Tukey’s multiple comparison test (ns, p > 0.05). (e) M1-Pep marginally affects constitutive internalization of GABAB receptors. Neurons transfected with GABAB2 tagged at the N-terminus with the minimum binding site for α-bungarotoxin (GABAB2(BBS)) were labeled with AlexaFluor 555 conjugated α-bungarotoxin and then analyzed by live cell imaging for internalization of labeled receptors. M1-Pep was added 10 min after starting the recording. Left: Representative images (scale bar: 5 μm). Right: Quantification of the immunofluorescence signal and statistical analysis. The statistical evaluation was performed on data recorded 15 min and 20 min after starting the recording. The data represent the mean ± SD of 7–8 neurons derived from three independent experiments. Unpaired t-test (ns, p > 0.05; *p < 0.05).
M1-Pep did not affect cell surface expression of GABAB receptor under physiological conditions
To test whether a potential interaction of MARCH1 with GABAB receptors affects receptor expression under physiological conditions, neuron/glia cultures were treated with M1-Pep and analyzed for GABAB receptor expression (Fig. 4d). However, under this condition, treatment with M1-Pep at a concentration of 10 µg/ml neither affected cell surface nor total expression of GABAB receptor. Thus, under physiological conditions GABAB receptor expression may only marginally, if at all, affected by MARCH1.
Next, we tested for potential subtle effects of MARCH1 on constitutive internalization of GABAB receptors. For this, neurons were transfected with GABAB2 tagged at the extracellularly located N-terminus with the minimum binding site for α-bungarotoxin (GABAB2(BBS)). After labeling cell surface receptors containing GABAB2(BBS) with AF555-conjugated α-bungarotoxin, neurons were analyzed by live cell imaging. Cell surface GABAB receptors were continually reduced during the time span of the measurement (20 min) demonstrating constitutive internalization of the receptors (Fig. 4e). Adding M1-Pep after 10 min slightly reduced loss of cell surface receptors. However, this effect became statistically significant only after longer treatment times with M1-Pep (20 min, Fig. 4e).
M1-Pep restored cell surface expression of GABAB receptors after OGD
Because MARCH1 was upregulated under ischemic conditions (Fig. 3b) we tested the effect of M1-Pep in restoring cell surface expression of GABAB receptor after OGD. For this, we subjected neuron/glia cultures for 1 h to OGD stress followed by treatment with M1-Pep and determination of GABAB receptor expression. OGD stress increased MARCH1 expression, as observed before, and concomitantly reduced cell surface expression of the receptors (Fig. 5a, Fig. S2). Treatment of cultures with M1-Pep after OGD normalized cell surface and total expression of GABAB receptors (Fig. 5a, b). The OGD induced downregulation of GABAB receptors was associated with an interaction of the receptors with MARCH1 as determined by in situ PLA. Under control conditions, we observed only few in situ PLA signals, most likely representing non-specific background (Fig. 5c). However, the PLA signals drastically increased after OGD stress, indicating the interaction of the receptors with MARCH1. Treatment of cultures with M1-Pep reduced PLA signals to control levels whereas the inactive Ctrl-Pep had no effect (Fig. 5c).
M1-Pep restored cell surface expression of GABAB receptors after OGD by inhibiting the MARCH1/GABAB receptor interaction. (a) Neurons/glia co-cultures were subjected to 1 h of OGD, then treated with M1-Pep or Ctrl-Pep for 16 h and analyzed for cell surface expression of GABAB receptors and MARCH1 expression. Left: representative images (scale bar: 10 μm). Right: quantification of fluorescence intensities (mean ± SD of 36 neurons per condition, 3 independent experiments). One-way ANOVA followed by Tukey’s multiple comparison test (ns, p > 0.05; ***, p < 0.001; ****, p < 0.0001). (b) Western blot analysis of total GABAB receptor expression using GABAB2 antibodies. Left: representative Western blot. Cultures were subjected for 1 h to OGD, then immediately treated or not with M1-Pep or Ctrl-Pep and harvested after 16–24 h for analysis. Right: quantification of Western blot signals (mean ± SD of 3 independent cultures per condition and one technical replicate). Signals were normalized to untreated control cultures (Ctrl). One-way ANOVA followed by Tukey’s multiple comparison test (ns, p > 0.05; ***, p < 0.001; ****, p < 0.0001). (c) M1-Pep inhibited the increased interaction between GABAB receptors and MARCH1 after OGD as analyzed by in situ PLA using antibodies directed against GABAB2 and MARCH1. Signals were normalized to untreated cultures (Ctrl). Left: representative images; top images: in situ PLA signals (white dots), bottom images: MARCH1 staining (scale bar: 10 μm). Right: quantification of in situ PLA and MARCH1 signals (mean ± SD of 25 neurons per condition, 3 independent experiments). One-way ANOVA followed by Tukey’s multiple comparison test (ns, p > 0.05; ***, ****, p < 0.0001). (d) M1-Pep induced lysosomal degradation MARCH1. Cultures were subjected to OGD for 1 h and immediately thereafter remained untreated or were treated with M1-Pep in the presence of proteasomal inhibitors (50 µM lactacystin or 50 µM MG132) or the lysosomal inhibitor leupeptin (50 µM). Neurons were tested for MARCH1 expression. Left: representative images (scale bar: 10 μm). Right: quantification of fluorescence intensities (mean ± SD of 30 neurons per condition, 2 independent experiments). The fluorescence intensity of untreated neurons served as control. Brown-Forsythe and Welch’s one-way ANOVA followed by Dunnett’s T3 multiple comparisons test (ns, p > 0.05; ****, p < 0.0001).
Interestingly, treatment of neuron/glia cultures with M1-Pep after OGD reduced the expression of MARCH1, suggesting that MARCH1 was degraded after binding to M1-Pep (Fig. 5c). To test for this, neuron/glia cultures were subjected to OGD, treated with M1-Pep and in addition with inhibitors for proteasomal (MG132, lactacyctin) or lysosomal (leupeptin) degradation (Fig. 5d). Inhibition of proteasomal degradation did not affect the downregulation of MARCH1, whereas blocking lysosomal degradation prevented it. Thus, binding of M1-Pep to MARCH1 most likely induced lysosomal degradation of a complex of M1-Pep bound to MARCH1.
We hypothesized that MARCH1 downregulates cell surface expression of GABAB receptors by permanently altering trafficking pathways of the receptor, such as internalization or recycling. If this is the case, M1-Pep should be able to quickly restore normal receptor expression. To test for how fast M1-Pep exerts its effect, we performed live cell imaging experiments as described for Fig. 4e, except that downregulation of cell surface receptors was induced by treating the cultures with glutamate. Cell surface GABAB receptors were reduced to 64 ± 12% 13 min after glutamate application (Fig. 6). Adding M1-Pep after 13 min completely restored cell surface expression of the receptors (96 ± 15%) within 1 min. The Ctrl-Pep was without effect (70 ± 18%). This finding supports our hypothesis that MARCH1 affects trafficking of GABAB receptors.
M1-Pep restored glutamate induced loss of cell surface GABAB receptors. Neurons were transfected with GABAB2 tagged at the N-terminus with the minimum binding site for α-bungarotoxin (GABAB2(BBS)) together with EGFP (for quick identification of transfected neurons) and GABAB1. Cell surface receptors containing GABAB2(BBS) were labeled with AlexaFluor 555 conjugated α-bungarotoxin and then neurons were treated with 50 µM glutamate to induce downregulation of the receptors. Neurons were analyzed by live cell imaging for internalization of labeled receptors. M1-Pep and Crtl-Pep were added 13 min after adding glutamate. Top: Representative images (scale bar: 10 μm). Bottom: Quantification of the immunofluorescence signal and statistical analysis. The statistical evaluation was performed on data recorded 15 min after glutamate treatment. The data represent the mean ± SD of 4–6 neurons derived from three independent experiments. One-way ANOVA followed by Tukey’s multiple comparison test (ns, p > 0.05; **p < 0.01).
M1-Pep increased fast recycling of GABAB receptors
To investigate the pathway underlying the M1-Pep induced restoration of cell surface GABAB receptors after OGD, we analyzed the colocalization of the receptors with marker proteins for early endosomes (EEA1)32, fast recycling endosomes (Rab4)33,34, slow recycling endosomes (Rab11)35and late endosomes (Rab7)36using in situ PLA (Fig. 7).
Treatment of neuron/glia co-cultures with M1-Pep after OGD increased colocalization of GABAB receptors with the marker proteins for early endosomes (EEA1, Fig. 7a) and for fast recycling endosomes (Rab4, Fig. 7b). In contrast, M1-Pep normalized the OGD induced increased colocalization of the receptors with slow recycling endosomes (Rab11, Fig. 7c) and late endosomes (Rab7, Fig. 7d). These results suggest that after an ischemic insult the treatment with M1-Pep restored GABAB expression by increasing fast recycling of the receptors and thereby reducing the pathological sorting to lysosomal degradation.
Treatment of OGD-stressed neurons with M1-Pep increased fast recycling of GABAB receptors and inhibited their targeting to the lysosomal pathway. Cultures were subjected to OGD for 1 h followed by treatment with M1-Pep. Neurons were analyzed about 16 h after OGD for the colocalization of GABAB receptors with endosomal markers by in situ PLA (upper panels) and for expression of the marker proteins (lower panels) using antibodies directed against GABAB2 and for the early endosome marker EEA1 (a), the fast recycling endosome marker Rab4 (b), the slow recycling endosome marker Rab11 (c), and the late endosome marker Rab7 (d). Scale bars, 10 µm. The data represent the mean ± SD of 25-30 neurons per condition derived from three independent experiments. One-way ANOVA followed by Tukey’s multiple comparison test (ns, p>0.05; *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001).
M1-Pep protects neurons from OGD stress induced death
Since M1-Pep restored the expression of GABAB receptors after OGD, we tested its ability to limit excitotoxic neuronal death. For this, neuron/glia co-cultures were subjected for 1 h to OGD and then immediately treated with M1-Pep. Determination of the number of surviving neurons after 24 h revealed that M1-Pep completely prevented neuronal death whereas the control peptide showed no effect (Fig. 8a).
Next, we tested the neuroprotective efficacy of M1-Pep when added at different time intervals after OGD (0, 3, 6, 9, 12 and 24 h). The number of surviving neurons were counted after a further 24 h (Fig. 8b). M1-Pep inhibited progressive neuronal death when added up to 9 h after OGD (Fig. 8b). At later time points, the neuroprotective effect of M1-Pep was no longer visible due to the already massive death of neurons. Thus, restoring the GABAB receptor expression by M1-Pep treatment after an ischemic insult limits progressing neuronal death within a wide time window.
M1-Pep inhibits progressive neuronal death after OGD. (a) M1-Pep completely prevents neuronal death when added directly after OGD. Neuron/glia cultures were subjected to OGD for 1 h and immediately thereafter treated with M1-Pep or Crtl-Pep. Next day, the number of neurons was determined by immunostaining for the neuronal marker protein NeuN (green) and total cell number by DAPI staining (blue). Left: representative images (scale bar: 50 µm). Right: quantification of surviving neurons (ratio of neurons to total cells). Untreated cultures served as control. The data represent the mean ± SD of the analysis of 31 field of views per condition derived from three independent experiments. One-way ANOVA followed by Tukey’s multiple comparison test (ns, p>0.05; ****p<0.0001). (b) M1-Pep inhibited OGD induced progressive neuronal death. Neurons were subjected to OGD for 1 h and were then treated with M1-Pep-Pep or Ctrl-Pep at different time intervals. Twenty-four hours after peptide treatment, neurons were tested for the number of neurons and total cells. Top: scheme of experimental design. Bottom: quantification of surviving neurons. Untreated cultures served as control. The data represent the mean ± SD of the analysis of 27 field of views per condition derived from three independent experiments. Brown-Forsythe and Welch’s one-way ANOVA followed by Games-Howell’s multiple comparisons test (ns, p>0.05; ****p<0.0001).
Discussion
In this study, we explored for membrane associated E3 ubiquitin ligases that are involved in regulating cell surface expression of GABAB receptors specifically under pathological conditions. We identified MARCH1 as a critical element in downregulating plasma membrane expression of GABAB receptors under ischemic/excitotoxic conditions (Fig. 9). MARCH1 (also known as RNF171) is a transmembrane E3 ligase, whose mRNA is particularly well expressed in secondary lymphoid tissues (spleen and lymph nodes) and at lower levels in other tissues including the brain25,26. MARCH1 has been shown to downregulate cell surface expression of MHC class II molecules, CD86, transferrin receptors and insulin receptors by targeting them to lysosomal degradation19,24,26,37,38.
We found that co-expression of GABAB receptors with MARCH1 in HEK-293 cells downregulated the cell surface receptors and required - in addition to the association of MARCH1 with the heterodimeric receptor - intact ubiquitin ligase activity, i.e. ubiquitination of the receptor. So far, it has been shown that targeting of GABAB receptors to lysosomal degradation involves Lys63-linked polyubiquitination of least three lysine residues in GABAB1 by the E3 ligase Mind Bomb-2 39,40. In addition, long-term changes in neuronal activity alters the level of cell surface GABAB receptors by regulating the number of newly synthesized receptors in the ER via Lys48-linked polyubiquitination of the receptors by the ER E3 ligase Hrd1 and proteasomal degradation41,42. Here, we found that MARCH1 is not involved in Lys48- or Lys63-linked polyubiquitination of GABAB receptors but most likely in mono- or mult-monoiubiquitination of GABAB receptors at yet undefined sites. This is in line with reports that implicated MARCH 1 in mono-, Lys63- and Lys48-linked polyubiquitination of target proteins43,44,45,46.
For gaining more insights into the mechanism of MARCH induced downregulation of GABAB receptors, we developed an interfering peptide (M1-Pep) inhibiting the interaction of the receptors with MARCH1. Treatment of cultured neurons with M1-Pep did not appreciably affect cell surface expression of the receptor under physiological conditions. This finding was not surprising in view of the very low expression of MARCH1 protein reported in primary cells19,44,47 and the lack of interaction we detected under physiological conditions. However, in live cell imaging experiments, testing for a potential effect of MARCH1 on constitutive internalization of GABAB receptors, we observed a slightly lower loss of cell surface receptors after prolonged incubation with M1-Pep. It is very unlikely that this effect is related to a direct action of MARCH1 on internalization of the receptors. We rather attribute this observation to an upregulation of MARCH1 under the suboptimal culture conditions during microscopy, which might induce cellular stress at longer time points.
Only after upregulation of MARCH1 by subjecting neurons to excitotoxic or ischemic stress, GABAB receptors significantly interacted with MARCH1 and robustly downregulated plasma membrane expression of the receptors. It is well established that MARCH1 is largely localized to the endocytic pathway where MARCH1 does not affect internalization of target proteins but triggers their lysosomal degradation26,38,48. In our experiments, co-expression of GABAB receptors with MARCH1 in HEK293 cells solely downregulated cell surface receptors but not total receptor levels arguing against a direct involvement of MARCH1 in targeting the receptors to lysosomal degradation. Instead, MARCH1 appears to inhibit fast recycling of the receptors, as concluded from colocalization experiments of the receptors with marker proteins of the endocytic pathway in neurons subjected to ischemic stress. Inhibition of the MARCH1/GABAB receptors interaction by M1-Pep restored fast recycling of the receptors and as a consequence also normalized enhanced lysosomal degradation of the receptors. This effect restored normal cell surface expression levels of the receptors and thereby prevented ischemia-induced neuronal death.
Interestingly, enhanced MARCH1 expression in ischemia-stressed neurons was normalized to control levels by treatment with M1-Pep. MARCH1 has a short half-life of less than 30 min47 and its expression is tightly controlled by dimerization and trans-autoubiquitination44. Both, lysosomes and proteasomes have been implicated in the degradation of MARCH1 44,47. In our setting, inhibition of lysosomes prevented downregulation of MARCH1 by M1-Pep, whereas blockers of proteasomes were ineffective. Thus, under ischemic conditions, the upregulated MARCH1 presumably forms a complex of MARCH1 with M1-Pep and is selectively degraded by lysosomes. Whether or not an interfering peptide induces degradation of its target protein appears not to be easily predictable. The interfering peptides we previously developed to inhibit the interaction of GABAB receptors with CaMKIIβ15 and PP2A12 did not affect the expression levels of CaMKIIβ and PP2A, whereas the peptide interfering with the CHOP/GABAB receptor interaction induced proteasomal degradation of ischemia-mediated upregulated CHOP16. The similar properties of MARCH1 and CHOP are the tight regulation of their expression (they are marginally expressed under physiological conditions19,47,49,50,51) and their detrimental effect on cellular health when overexpressed. Hence, tightly regulated proteins that interfere with cellular health at higher expression levels might be more susceptible to degradation when interacting with an interfering peptide.
Proposed mechanism of MARCH1-induced downregulation of GABAB receptors under ischemic conditions. Under normal physiological conditions, GABAB receptors are constitutively endocytosed and most internalized receptors are recycled to the plasma membrane, whereas a fraction is degraded in lysosomes. Under ischemic conditions, upregulated MARCH1 interacts with GABAB receptors and ubiquitinate them. This inhibits recycling of endocytosed GABAB receptors. Blocking recycling causes rapid removal of GABAB receptors from the plasma membrane and triggers their lysosomal degradation. Application of M1-Pep after ischemic stress inhibits the interaction of MARCH1 with GABAB receptors. This re-establishes recycling of the receptors to the plasma membrane and normalizes enhanced lysosomal degradation of GABAB receptors, resulting in normal plasma membrane expression of GABAB receptors and inhibition progressive neuronal death. The figure was created using https://www.biorender.com/.
We have previously shown that restoring GABAB receptor expression after an ischemic insult using interfering peptides targeting the CaMKIIβ15, PP2A12, or CHOP16 interaction with the receptor re-established normal GABAB receptor mediated inhibition. This inhibited enhanced excitability of affected neurons and prevented progressing neuronal death. These peptides are promising candidates for the development of a neuroprotective treatment in the acute phase of stroke. However, the interfering peptides targeting CaMKIIβ and PP2A have the potential limitation that they inhibit interactions that are regulating GABAB receptor expression under physiological conditions as well. Treatment with these peptides could theoretically increase GABAB receptor expression in non-diseased neurons. By contrast, the interfering peptides inhibiting the interaction of the receptors with CHOP or MARCH1 target interactions that are not occurring under physiological conditions and require disease-induced upregulation of MARCH1 and CHOP. Thus, the interaction of CHOP and MARCH1 with GABAB receptors are specifically occurring in diseased neurons. Therefore, treatment with the respective interfering peptides should not affect non-diseased neurons and are expected to exhibit no or minimal side effects related to GABAB receptors. In a next step, the neuroprotective activity of CHOP and MARCH1 interfering peptides need to be tested in suitable animal models of cerebral ischemia.
Methods
Plasmids
The following plasmids were used for this study: hemagglutinin (HA)-tagged GABAB1a52; GABAB253; GABAB1a(RSAR)54; EYFP-MARCH1 18; EYFP-MARCH1(DN)18; EYFP-MARCH1-KKxx24; EYFP-MARCH5 18; EYFP-MARCH5(DN)18; EYFP-MARCH8 24; EYFP-MARCH8(DN)24; EYFP-RNF112 18; EYFP-RNF112(DN)18; EYFP-RNF133 18; EYFP-RNF133(DN)18; EYFP-RNF144 18; EYFP-RNF144(DN)18; EYFP-RNF152 18; EYFP-RNF152(DN)18; EYFP-RNF167 18; EYFP-RNF167(DN)18; CHOP29; ubiquitin(KO) (Addgene plasmid 17603 55), and HA-GABAB2(BBS) tagged the minimum α-bungarotoxin binding site (BBS)12.
Drugs
The following drugs were used for this study: MG-132 (Abcam, #ab141003), lactacystin (Abcam, #ab141411), leupeptin (Merck, #L2884), D-tubocurarine chloride (Abcam, #AB120073) and AlexaFlour 555-conjugated α-bungarotoxin (Invitrogen, #B35451).
Antibodies
The following antibodies were used for this study: mouse anti-MARCH1 (1:250 for Western blotting [WB], 1:250 for immunofluorescence staining [IF] and 1:50 for in situ proximity assay [PLA], Sigma Aldrich #WH0055016M2), rabbit anti-MARCH1 (1:250 for IF and 1:50 for PLA, Antibodies Online, #ABIN2705354), mouse anti-GABAB1 (1:250 for IF and, 1:50 for PLA; Abcam #ab55051), rabbit anti-GABAB1b directed against the N-terminus of GABAB1b (affinity-purified,1:100 for IF; custom made by GenScript56), rabbit anti-GABAB2 directed against the N-terminus of GABAB2 (affinity-purified, used for cell surface staining, 1:250 for IF; custom made by GenScript57), rabbit anti-GABAB2 (1:500 for IF, 1:100 for PLA, 1:800 for WB; Abcam #ab75838), rabbit anti-NeuN (1:400 for IF, Millipore #ABN78), rabbit anti-HA (1:500 for IF and 1:50 for PLA, Sigma-Aldrich #SAB5600116), rabbit anti-ubiquitin-Lys63 (1:50 for PLA, Millipore #05-1307), rabbit anti-ubiquitin-Lys48 (1:50 for PLA, Millipore #05-1308), mouse anti-EEA1 (1:50 for PLA, BD Biosciences #610456), mouse anti-Rab4 (1:50 for PLA, BD Biosciences #610888), mouse anti-Rab11 (1:50 for PLA, Millipore #05-853), and rabbit anti-Rab7 (1:50 for PLA, Abcam #ab137029). For immunofluorescence staining, secondary antibodies used were labeled with either Alexa Fluor Plus 488, 555, or 647 (1:2000, ThermoFisher). For Western blotting secondary antibodies were conjugated to IRDye800CW (LI-COR Biosciences).
Culture and transfection of HEK-293 cells
HEK-293 cells (Human Embryonic Kidney, ATCC) were cultured in DMEM (Gibco Life Technologies) containing 10% fetal bovine serum (FBS) (Gibco Life Technologies) and penicillin/streptomycin (Gibco Life Technologies). HEK-293 cells were plated at a density of 100,000 cells per ml into 12 well plates containing poly-D-lysine coated coverslips and kept overnight in incubator at 37 °C and 5% CO2. Next day, the cells were transfected with respective plasmids using the polyethyleneimine (PEI) method according to the jet-PEI protocol (Polyplus Transfection) and then kept in incubator at 37 °C and 5% CO2 for 24–48 h.
In situ proximity ligation assay (in situ PLA)
The in situ PLA was performed using the Duolink II kit (Sigma Aldrich) according to the instructions of the manufacturer. Briefly, the neurons or HEK-293 cells were washed for 5 min with PBS and then fixed with 4% PFA for 30 min at room temperature. Then, the coverslips were rinsed in PBS for 5 min and permeabilized for 15 min with 0.2% Triton X-100/PBS. After rinsing the coverslips in PBS, they were incubated with two primary antibodies between whom the interaction had to be observed (one antibody raised in rabbit and other in mouse) overnight in a humidity chamber at 4 °C. Subsequently, the cultures were washed four times for 5 min with PBS and incubated for 30 min at room temperature with the PLA probes (prepared by diluting anti-Mouse MINUS and anti-Rabbit PLUS (Duolink II) at a concentration of 1:5 in 10% NGS/PBS). Afterwards, 60 µl of the PLA probe solution were pipetted on top of each coverslip and incubated in a humidity chamber at 37 °C for 1 h. The coverslips were then washed in PBS two times for 5 min and then incubated at 37 °C for 30 min with ligation solution. Subsequently, the cells were washed two times in Duolink II Wash Buffer A and then incubated with the amplification solution at 37 °C for 100 min. Finally, the coverslips were washed two times for 10 min with Duolink II Wash Buffer B in the dark and then mounted onto microscope slides with DAKO fluorescent mounting medium.
Primary neuron-glia co-cultures
In this study, we used mixed neuron/glia co-cultures because they mimic the brain physiology more closely compared to the pure neurons. The neuron to glial cell ratio in our cultures was roughly 1:5. All procedures were carried out according to the national guidelines of the Swiss Federal act on animal protection and were approved by the Cantonal Veterinary Office Zurich (license ZH011/19 and ZH087/2022). We confirm compliance with the ARRIVE guidelines.
Pregnant Wistar rats were purchased from ENVIGO, Netherlands. All cell culture media used were from Gibco. The pregnant rat was deeply anesthetized with isoflurane, euthanized by decapitation and embryos were extracted for preparation of primary cultures. Embryos (18 days old) were killed by decapitation and the cerebral cortices were dissected and then washed with 5 ml sterile-filtered PBGA buffer (PBS containing 10 mM glucose, 1 mg/ml bovine serum albumin and antibiotic-antimycotic 1:100 (10,000 units/ml penicillin; 10,000 µg/ml streptomycin; 25 µg/ml amphotericin B). The cortices were cut into small tissue pieces with a sterile scalpel and then digested by incubating in 5 ml sterile filtered papain solution for 15 min at 37 °C. The supernatant was then removed and the tissue was washed twice with complete DMEM/FBS medium (Dulbecco’s Modified Eagle’s Medium containing 10% Fetal Bovine Serum and penicillin/streptomycin, 1:100). Then, 3–4 ml of fresh DMEM/FBS was added, and the tissue was carefully and gently triturated and subsequently filtered through a 40 μm cell-strainer. Finally, the neurons were plated at a density of 75,000–90,000 cells per well onto the poly D-lysine (Gibco Life Technologies) coated coverslips in a 12-well culture dish and incubated for 3–4 h at 37 °C and 5% CO2. Then, the DMEM medium was exchanged with freshly prepared NU-medium (Minimum Essential Medium (MEM) with 15% NU serum, 2% B27 supplement, 15 mM HEPES, 0.45% glucose, 1 mM sodium pyruvate, 2 mM GlutaMAX). The cells were kept in incubator for 12–16 days at 37 °C and 5% CO2.
Transfection of primary Neuron/Glia cultures
After 7 to 12 days in vitro cultures were transfected with a total of 1 µg of the respective plasmid DNAs by the magnetofection method using Lipofectamine 2000 (Invitrogen) and CombiMag (OZ Biosciences) at 37 °C and 5% CO2, as described by Buerli et al.58.
Oxygen and glucose deprivation (OGD) stress
Neuronal cultures were subjected to OGD stress by exposure to OGD medium (DMEM lacking glucose, glutamine, sodium pyruvate, HEPES and phenol red) in a hypoxic incubator. The OGD medium was deprived from oxygen by equilibrating it with nitrogen for 15 min in a water bath at 37 °C. For IF, 1 ml of equilibrated OGD medium was added into each well of a 12-well culture plate and the coverslips containing the cultured neurons were transferred to the OGD medium. The culture plate was then incubated for 1 h in a hypoxic incubator at 1% O2, 5% CO2 and 37 °C. The coverslips were then transferred back to the culture plate containing the original conditioned culture medium and incubated at 37 °C and 5% CO2. For WB, 5 ml of equilibrated OGD medium was added into each 6 cm dishes followed by 1 h in a hypoxic incubator at 1% O2, 5% CO2 and 37 °C. The equilibrated OGD medium was then removed and 5 ml of original conditioned culture medium was added and incubated at 37 °C and 5% CO2. Unless stated otherwise, the neurons were analyzed after a recovery period of 16–24 h.
Interfering peptide (M1-Pep)
For identification of peptides interfering with the GABAB receptor/March1 interaction a small library of overlapping synthetic peptides (15–25 amino acids long, for sequences see Fig. 4) comprising the intracellularly located C-terminal amino acid sequences of GABAB1 and GABAB2 was generated. All peptides contained a sequence of eight arginine at their N-terminus to render them cell permeable. To enhance peptide uptake into transfected HEK-293 cells, the cultures were washed three times with PBS and then treated with 50 µM pyrene butyric acid for 5 min. After three additional washes with PBS, the cells were treated with 10 µg/ml peptide and tested for their ability to inhibit the downregulation of plasma membrane GABAB receptors.
From this screening resulted the interfering peptide DKDLEEVTMQLQDTPEKTTY (M1-Pep). For control, a peptide (Ctrl-Pep, QTDTYKMLDQELTTDPEEVK) was used which contains the same amino acids as M1-Pep but in a random manner. The cell permeability of the peptides was achieved by tagging the N-terminus of the respective peptide with a peptide sequence from Rabies virus glycoprotein followed by 9 arginine (RVG, YTIWMPENPRPGTPCDIFTNSRGKRASNGGGGRRRRRRRRR)30. The Peptides were custom-synthesized by Pepmic Co., Ltd, Suzhou, China. The peptides were used at a concentration of 10 µg/ml for all the experiments.
Immunofluorescence staining
For cell surface staining of GABAB receptors, an antibody directed against the N-terminus of GABAB2 (GABAB2N) was used. Coverslips containing the cultured neuron/glia cells were washed 3 times with cold buffer A (25 mM HEPES pH 7.4, 119 mM NaCl, 2.5 mM KCl, 2 mM CaCl2, 1 mM MgCl2 and 30 mM glucose). Then, the GABAB2N antibody (1: 250 dilutions in buffer A containing 10% normal donkey serum (NDS)) was added and incubated on ice for 90 min. The coverslips were subsequently washed 3 times for 5 min with buffer A, followed by incubation with Alexa Fluor Plus anti-rabbit secondary antibody (1:2000 in PBS/10% NDS) for 60 min on ice. Afterwards, the coverslips were washed again 3 times for 5 min with buffer A. For subsequent staining of total MARCH1, the cells were fixed with 4% PFA for 30 min at room temperature. After fixation, the cells were washed with PBS and permeabilized by incubation for 12 min in 0.2% Triton X-100/PBS. Then, MARCH1 antibody (1:250 in PBS/10% NDS) was added and incubated overnight at 4 °C. After incubation, the coverslips were washed 3 times for 5 min with PBS. Then, Alexa Fluor Plus anti-mouse secondary antibody (dilution 1:2000 in PBS/ 10% NDS) was added and incubated for 1 h at room temperature. Finally, the coverslips were washed again 3 times for 5 min with PBS and mounted in DAKO fluorescence mounting medium onto glass slides for confocal microscopy.
For standard immunofluorescence staining, the cultures were briefly washed in PBS and then fixed with 4% PFA for 30 min at room temperature. After fixation, the cells were washed with PBS and permeabilized by incubation for 12 min in 0.2% Triton X-100/PBS, followed by incubation with primary antibody (diluted in PBS/10% NDS) overnight at 4 °C. Next day, the coverslips were washed 3 times for 5 min with PBS and incubated for 1 h at room temperature with Alexa Fluor Plus secondary antibody (diluted in PBS/ 10% NDS). The coverslips were then washed again 3 times for 5 min with PBS and mounted in DAKO fluorescence mounting medium onto glass slides for confocal microscopy.
Live cell imaging for tracing internalization of GABAB receptors
The live cell imaging of GABAB receptors tagged with the α-bungarotoxin binding site (BBS) was performed for monitoring the internalization of the receptor exactly as described previously12. Neuron/glia cultures were transfected at DIV 7–12 with GABAB2(BBS) together with EGFP (for fast identification of transfected neurons). For live cell imaging, the cultures were washed with ice-cold Krebs solution (140 mM NaCl, 4.7 mM KCl, 1.2 mM MgCl2, 2.5 mM CaCl2, 11 mM glucose, and 5 mM HEPES pH 7.4), pre-incubated with 1 mM D-tubocurarine for 5 min to block endogenous nicotinic acetylcholine receptors containing the α7 subunit and then incubated on ice with 3 µg/ml AlexaFlour 555-conjugated α-bungarotoxin for 15 min to label the GABAB receptors containing GABAB2(BBS) expressed at the cell surface. Then, the cultures were washed with ice-cold Krebs solution to remove excess BTX and transferred to the imaging chamber with warm Krebs solution at room temperature to enable internalization of the receptors. The EGFP expressing cells were located as quickly as possible, and settings were optimized for acquiring BTX florescence at time zero (t0). The imaging of a single plane was performed at Nyquist sampling and eight times averaging to increase the signal/noise ratio. Glutamate (50 µM) was added at the start of the recording and M1-Pep (10 µg/ml) 13 min thereafter. Both were added directly to the imaging chamber. The rate of GABAB receptor internalization was evaluated by measuring the mean fluorescence intensity at the cell surface at every time-point. Measurements were normalized to the mean cell surface membrane fluorescence obtained at t0 time point.
Quantification of neuronal loss
Neuron-glia co-cultures were subjected to OGD and subsequently treated with M1-Pep or control peptide. Then, cultures were incubated with an antibody directed against the neuron-specific marker protein NeuN (1:400 in PBS/ 10% NDS), followed by staining with Alexa Fluor Plus 488 secondary antibodies (1:2000 in PBS/ 10% NDS). The total number of cells were determined by counting the cell nuclei stained with DAPI included in the fluorescent mounting medium. After Microscopy, the neuronal loss was counted by ratio of number of neurons over number of DAPI positive cells using ImageJ plugin “Cell Counter”.
Western blotting
For Western blot analysis of neuron-glia co-cultures, the cells were grown for 12–16 days on 6 cm poly-D lysine coated culture dishes at a density of 500,000 cells per dish. The cultures were washed two times with ice-cold PBS, harvested, and homogenized by sonication. The samples were incubated with Laemmli sample buffer (Bio-Rad) for 1 h at 37 °C and aliquots containing 20 µg protein were subjected to sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) using 7.5% mini-gels (Mini Protean3; Bio-Rad). The proteins were transferred onto nitrocellulose membranes in a semi-dry transfer cell (Trans-Blot SD; Bio-Rad) at 15 V for 75 min. After blotting, the transferred total proteins were stained with REVERT 700 Total Protein Stain (LI-COR Biosciences) and detected by the ODYSSEY CLx scanner (LI-COR Biosciences). The blots were blocked for 1 h in PBS containing 5% nonfat dry milk at room temperature, followed by incubation with primary antibody overnight at 4 °C in PBS containing 5% nonfat dry milk. The blots were then washed five times for 5 min with TBST and incubated with secondary antibodies for 1 h at room temperature. The blots were washed again with TBST and immunoreactivity was detected by the ODYSSEY CLx scanner (LI-COR Biosciences). Immunoreactivity was quantified with the Image Studio software (LI-COR Biosciences) and normalized to total protein in the corresponding lanes.
Microscopy and image analysis
Images were taken with a Zeiss laser scanning confocal microscope (CLSM800 AiryScan) in the sequential mode using the Zeiss 40 × (1.3 NA) or 63 × (1.45 NA) Plan-Fluor objective. Signal saturation was avoided by adjusting the values of laser intensity and the detector gain accordingly. All images of one experiment was imaged in one continuous session with the same settings. The images were quantitatively analyzed using ImageJ (v1.54i). For quantification of cell surface staining, the outer and inner perimeter of the cell surface were exactly outlined. Then, the fluorescence intensity value obtained from the inner border was subtracted from the one of the outer border so that only the fluorescence present at the cell surface was determined and used for statistical evaluation. For quantification of the total cell staining, only the outer border of the cell was marked and the mean fluorescence intensity was measured. For quantification of in situ PLA signals, the soma of neurons or HEK-293 cells was surrounded and the fluorescent dots inside these borders were counted using the ImageJ option “Find maxima”. A fixed noise tolerance value was used for the analysis of all images of the same experiment. The PLA signals were normalized to the area analyzed and also normalized to the GABAB receptor expression level.
Statistics
The statistical evaluation of data was performed using the software GraphPad Prism (version 8.4.3). Results were given as mean value ± standard deviation (SD). All data sets were tested for normal or lognormal distributions and analyzed by One-Way ANOVA followed by appropriate post hoc tests or unpaired t-test. Data sets that did not show normal distribution were analyzed by the Kruskal-Wallis test. In case of significant deviation from homoscedasticity, Welch and Brown Forsythe variations of ANOVA was used. A p-value of < 0.05 was considered as statistically significant. Details of the statistics are given in the figure legends.
Data availability
The datasets analyzed for this study are deposited at ZENODO.org and are publicly available as of the date of publication (https://doi.org/10.5281/zenodo.14608218). Due to their large size raw images can be made available only upon reasonable request to the corresponding author.
References
Chalifoux, J. R. & Carter, A. G. GABAB receptor modulation of synaptic function. Curr. Opin. Neurobiol. 21, 1–6. https://doi.org/10.1016/j.conb.2011.02.004 (2011). [doi].
Terunuma, M. Diversity of structure and function of GABAB receptors: A complexity of GABAB-mediated signaling. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 94, 390–411, (2018). https://doi.org/10.2183/pjab.94.026
Gassmann, M. & Bettler, B. Regulation of neuronal GABAB receptor functions by subunit composition. Nat. Rev. Neurosci. 13, 380–394 (2012). doi:nrn3249 [pii] 10.1038/nrn3249 [doi].
Rose, T. R. & Wickman, K. Mechanisms and regulation of neuronal GABAB receptor-dependent signaling. Curr. Top. Behav. Neurosci. 52, 39–79. https://doi.org/10.1007/7854_2020_129/TABLES/2 (2022).
Jacobson, L. H., Vlachou, S., Slattery, D. A., Li, X. & Cryan, J. F. The g-aminobutyric acid B receptor in depression and reward. Biol. Psychiatry. 83, 963–976. https://doi.org/10.1016/j.biopsych.2018.02.006 (2018).
Princivalle, A. P. GABAB receptors in neurodegeneration. Curr. Top. Behav. Neurosci. 52, 267–290. https://doi.org/10.1007/7854_2021_222 (2022).
Vlachou, S. GABAB receptors and cognitive processing in health and disease. Curr. Top. Behav. Neurosci. https://doi.org/10.1007/7854_2021_231 (2021).
Bhat, M. A., Hleihil, M. & Benke, D. Restoration of GABAB receptor expression in cerebral ischemia: A promising novel neuroprotective strategy. Neural Regen Res. 18, 2659–2660. https://doi.org/10.4103/1673-5374.373704 (2023).
Benke, D. Mechanisms of GABAB receptor exocytosis, endocytosis, and degradation. Adv. Pharmacol. 58, 93–111 (2010).
Maier, P. J. et al. Ischemia-like oxygen and glucose deprivation mediates down-regulation of cell surface g-aminobutyric acidB receptors via the endoplasmic reticulum (ER) stress-induced transcription factor CCAAT/enhancer-binding protein (C/EBP)-homologous protein (CHOP). J. Biol. Chem. 289, 12896–12907. https://doi.org/10.1074/jbc.M114.550517 (2014).
Terunuma, M. et al. Prolonged activation of NMDA receptors promotes dephosphorylation and alters postendocytic sorting of GABAB receptors. Proc. Natl. Acad. Sci. USA 107, 13918–13923 (2010).
Hleihil, M., Balakrishnan, K. & Benke, D. Protein phosphatase 2A regulation of GABAB receptors normalizes ischemia-induced aberrant receptor trafficking and provides neuroprotection. Front. Mol. Neurosci. 15, 1015906. https://doi.org/10.3389/fnmol.2022.1015906 (2022).
Guetg, N. et al. NMDA receptor-dependent GABAB receptor internalization via CaMKII phosphorylation of serine 867 in GABAB1. Proc. Natl. Acad. Sci. USA 107, 13924–13929 (2010).
Bhat, M. A., Grampp, T. & Benke, D. ERK1/2-dependent phosphorylation of GABAB1(S867/T872), controlled by CaMKIIβ, is required for GABAB receptor degradation under physiological and pathological conditions. Int. J. Mol. Sci. 24 https://doi.org/10.3390/ijms241713436 (2023).
Balakrishnan, K. et al. Targeting the interaction of GABAB receptors with CaMKII with an interfering peptide restores receptor expression after cerebral ischemia and inhibits progressive neuronal death in mouse brain cells and slices. Brain Pathol. 33, e13099. https://doi.org/10.1111/bpa.13099 (2023).
Bhat, M. A., Esmaeili, A., Neumann, E., Balakrishnan, K. & Benke, D. Targeting the interaction of GABAB receptors with CHOP after an ischemic insult restores receptor expression and inhibits progressive neuronal death. Front. Pharmacol. 13, 870861. https://doi.org/10.3389/fphar.2022.870861 (2022).
Jean-Charles, P. Y., Snyder, J. C. & Shenoy, S. K. Chapter one - ubiquitination and deubiquitination of G protein-coupled receptors. Prog Mol. Biol. Transl Sci. 141, 1–55. https://doi.org/10.1016/bs.pmbts.2016.05.001 (2016).
Lussier, M. P. et al. Ubiquitin ligase RNF167 regulates AMPA receptor-mediated synaptic transmission. Proc. Natl. Acad. Sci. U S A 109, 19426–19431, (2012). https://doi.org/10.1073/pnas.1217477109
Nagarajan, A. et al. MARCH1 regulates insulin sensitivity by controlling cell surface insulin receptor levels. Nat. Commun. 7, 12639. https://doi.org/10.1038/ncomms12639 (2016).
Trenker, R. et al. Human and viral membrane-associated E3 ubiquitin ligases MARCH1 and MIR2 recognize different features of CD86 to downregulate surface expression. J. Biol. Chem. 297, 100900. https://doi.org/10.1016/j.jbc.2021.100900 (2021).
Wilson, K. R. et al. MARCH1-mediated ubiquitination of MHC II impacts the MHC I antigen presentation pathway. PLoS One. 13, e0200540. https://doi.org/10.1371/journal.pone.0200540 (2018).
Goto, E. et al. c-MIR, a human E3 ubiquitin ligase, is a functional homolog of herpesvirus proteins MIR1 and MIR2 and has similar activity. J. Bio Chem. 278, 14657–14668. https://doi.org/10.1074/jbc.M211285200 (2003).
Nian, H. et al. Mouse RING finger protein Rnf133 is a testis-specific endoplasmic reticulum-associated E3 ubiquitin ligase. Cell. Res. 18, 800–802. https://doi.org/10.1038/cr.2008.73 (2008).
Bourgeois-Daigneault, M. C. & Thibodeau, J. Identification of a novel motif that affects the conformation and activity of the MARCH1 E3 ubiquitin ligase. J. Cell. Sci. 126, 989–998. https://doi.org/10.1242/jcs.117804 (2013).
Kang, Z. et al. Goat membrane associated ring-CH-type finger 1 (MARCH1) mRNA expression and association with litter size. Theriogenology 128, 8–16. https://doi.org/10.1016/j.theriogenology.2019.01.014 (2019).
Bartee, E., Mansouri, M., Hovey Nerenberg, B. T., Gouveia, K. & Früh, K. Downregulation of major histocompatibility complex class I by human ubiquitin ligases related to viral immune evasion proteins. J. Virol. 78, 1109–1120. https://doi.org/10.1128/jvi.78.3.1109-1120.2004 (2004).
Maier, P. J., Marin, I., Grampp, T., Sommer, A. & Benke, D. Sustained glutamate receptor activation down-regulates GABAB receptors by shifting the balance from recycling to lysosomal degradation. J. Biol. Chem. 285, 35606–35614. https://doi.org/10.1074/jbc.M110.142406 (2010). [doi].
Hleihil, M., Vaas, M., Bhat, M. A., Balakrishnan, K. & Benke, D. Sustained baclofen-induced activation of GABAB receptors after cerebral ischemia restores receptor expression and function and limits progressing loss of neurons. Front. Mol. Neurosci. 14, 726133. https://doi.org/10.3389/fnmol.2021.726133 (2021).
Sauter, K. et al. Subtype-selective interaction with the transcription factor CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) regulates cell surface expression of GABAB receptors. J. Biol. Chem. 280, 33566–33572 (2005).
Kumar, P. et al. Transvascular delivery of small interfering RNA to the central nervous system. Nature 448, 39–43. https://doi.org/10.1038/nature05901 (2007).
Hleihil, M. & Benke, D. Restoring GABAB receptor expression in the ventral tegmental area of methamphetamine addicted mice inhibits locomotor sensitization and drug seeking behavior. Front. Mol. Neurosci. 17 https://doi.org/10.3389/fnmol.2024.1347228 (2024).
Wilson, J. M. et al. EEA1, a tethering protein of the early sorting endosome, shows a polarized distribution in hippocampal neurons, epithelial cells, and fibroblasts. Mol. Biol. Cell. 11, 2657–2671. https://doi.org/10.1091/mbc.11.8.2657 (2000).
Daro, E., van der Sluijs, P., Galli, T. & Mellman, I. Rab4 and cellubrevin define different early endosome populations on the pathway of transferrin receptor recycling. Proc. Natl. Acad. Sci. U S A. 93, 9559–9564. https://doi.org/10.1073/pnas.93.18.9559 (1996).
van der Sluijs, P. et al. The small GTP-binding protein rab4 controls an early sorting event on the endocytic pathway. Cell 70, 729–740. https://doi.org/10.1016/0092-8674(92)90307-x (1992).
Ullrich, O., Reinsch, S., Urbé, S., Zerial, M. & Parton, R. G. Rab11 regulates recycling through the pericentriolar recycling endosome. J. Cell. Biol. 135, 913–924. https://doi.org/10.1083/jcb.135.4.913 (1996).
Feng, Y., Press, B. & Wandinger-Ness, A. Rab 7: An important regulator of late endocytic membrane traffic. J. Cell. Biol. 131, 1435–1452. https://doi.org/10.1083/jcb.131.6.1435 (1995).
Walseng, E. et al. Ubiquitination regulates MHC class II-peptide complex retention and degradation in dendritic cells. Proc. Natl. Acad. Sci. U S A. 107, 20465–20470. https://doi.org/10.1073/pnas.1010990107 (2010).
Eyster, C. A. et al. MARCH ubiquitin ligases alter the itinerary of clathrin-independent cargo from recycling to degradation. Mol. Biol. Cell. 22, 3218–3230. https://doi.org/10.1091/mbc.E10-11-0874 (2011).
Zemoura, K., Trumpler, C. & Benke, D. Lys-63-linked ubiquitination of g-aminobutyric acid (GABA), type B1, at multiple sites by the E3 ligase mind Bomb-2 targets GABAB receptors to lysosomal degradation. J. Biol. Chem. 291, 21682–21693. https://doi.org/10.1074/jbc.M116.750968 (2016).
Zemoura, K., Balakrishnan, K., Grampp, T. & Benke, D. Ca2+/Calmodulin-dependent protein kinase II (CaMKII) b-dependent phosphorylation of GABAB1 triggers lysosomal degradation of GABAB receptors via mind bomb-2 (MIB2)-mediated Lys-63-linked ubiquitination. Mol. Neurobiol. 56, 1293–1309. https://doi.org/10.1007/s12035-018-1142-5 (2019).
Zemoura, K. et al. Endoplasmic reticulum-associated degradation (ERAD) controls cell surface expression of g-aminobutyric acid, type B receptors. J. Biol. Chem. 288, 34897–34905. https://doi.org/10.1074/jbc.M113.514745 (2013).
Zemoura, K. & Benke, D. Proteasomal degradation of g-aminobutyric acidB receptors is mediated by the interaction of the GABAB2 C terminus with the proteasomal ATPase Rtp6 and regulated by neuronal activity. J. Biol. Chem. 289, 7738–7746. https://doi.org/10.1074/jbc.M113.541987 (2014).
Gogoi, M., Ravikumar, V., Dixit, N. M. & Chakravortty, D. Salmonella escapes antigen presentation through K63 ubiquitination mediated endosomal proteolysis of MHC II via modulation of endosomal acidification in dendritic cells. Pathog Dis. 76, ftx128. https://doi.org/10.1093/femspd/ftx125 (2018).
Bourgeois-Daigneault, M. C. & Thibodeau, J. Autoregulation of MARCH1 expression by dimerization and autoubiquitination. J. Immunol. 188, 4959–4970. https://doi.org/10.4049/jimmunol.1102708 (2012).
Corcoran, K. et al. Ubiquitin-mediated regulation of CD86 protein expression by the ubiquitin ligase membrane-associated RING-CH-1 (MARCH1). J. Biol. Chem. 286, 37168–37180. https://doi.org/10.1074/jbc.M110.204040 (2011).
Thibodeau, J. et al. Interleukin-10-induced MARCH1 mediates intracellular sequestration of MHC class II in monocytes. Eur. J. Immunol. 38, 1225–1230. https://doi.org/10.1002/eji.200737902 (2008).
Jabbour, M., Campbell, E. M., Fares, H. & Lybarger, L. Discrete domains of MARCH1 mediate its localization, functional interactions, and posttranscriptional control of expression. J. Immunol. 183, 6500–6512. https://doi.org/10.4049/jimmunol.0901521 (2009).
Cho, K. J. & Roche, P. A. Regulation of MHC class II-peptide complex expression by ubiquitination. Front. Immunol. 4, 369. https://doi.org/10.3389/fimmu.2013.00369 (2013).
Oyadomari, S. & Mori, M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell. Death Differ. 11, 381–389 (2004).
Yang, Y. et al. Transcription Factor C/EBP Homologous Protein in Health and diseases. Front. Immunol. 8, 1612. https://doi.org/10.3389/fimmu.2017.01612 (2017).
Hu, H., Tian, M., Ding, C. & Yu, S. The C/EBP homologous protein (CHOP) transcription factor functions in endoplasmic reticulum stress-induced apoptosis and microbial infection. Front. Immunol. 9, 3083. https://doi.org/10.3389/fimmu.2018.03083 (2019).
Kaupmann, K. et al. Expression cloning of GABAB receptors uncovers similarity to metabotropic glutamate receptors. Nature 386, 239–246 (1997).
Kaupmann, K. et al. GABAB receptor subtypes assemble into functional heteromeric complexes. Nature 396, 683–687 (1998).
Pagano, A. et al. C-terminal interaction is essential for surface trafficking but not for heteromeric assembly of GABAB receptors. J. Neurosci. 21, 1189–1202 (2001).
Lim, K. L. et al. Parkin mediates nonclassical, proteasomal-independent ubiquitination of synphilin-1: Implications for Lewy body formation. J. Neurosci. 25, 2002–2009 (2005).
Benke, D., Honer, M., Michel, C., Bettler, B. & Mohler, H. g-Aminobutyric acid type B receptor splice variant proteins GBR1a and GBR1b are both associated with GBR2 in situ and display differential regional and subcellular distribution. J. Biol. Chem. 274, 27323–27330 (1999).
Benke, D., Michel, C. & Mohler, H. Structure of GABAB receptors in rat retina. J. Recept Signal. Transduc. 22, 253–266 (2002).
Buerli, T. et al. Efficient transfection of DNA or shRNA vectors into neurons using magnetofection. Nat. Protoc. 2, 3090–3101. https://doi.org/10.1038/nprot.2007.445 (2007). [doi].
Acknowledgements
The authors are grateful to Khaled Zemoura, Sara Heynen and Nicolas Kalt for initial preliminary experiments leading to this study. This research was funded by the Swiss National Science Foundation grant numbers 31003A_182325 and 310030_212221 to D.B.
Author information
Authors and Affiliations
Contributions
M.A.B. and D.B. designed the study, analyzed data, and wrote the manuscript. M.A.B., M.H., I.M. and T.G. performed experiments and analyzed data.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Bhat, M.A., Hleihil, M., Mondéjar, I. et al. The E3 ubiquitin ligase MARCH1 mediates downregulation of plasma membrane GABAB receptors under ischemic conditions by inhibiting fast receptor recycling. Sci Rep 15, 1330 (2025). https://doi.org/10.1038/s41598-025-85842-1
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41598-025-85842-1