Supporting Information

10.1073/pnas.0908359107 SI Methods reverse PCR primer using the BigDye Terminator Cycle Se- Subjects. This study includes a three-generation dominant quencing Kit v3.1 (Applied Biosystems), purified with Sephadex migraine family referred to Vall d’Hebron University Hospital, G-50 (Amersham Biosciences) using MultiScreen Plates (Milli- Barcelona. Seven affected and three healthy individuals were pore), and run in an ABI PRISM 3700 DNA analyzer (Applied directly interviewed and examined by one of the authors (A.M.), Biosystems). Sequence chromatograms were inspected manually following the guidelines of the International Classification of and analyzed with the SeqMan v3.6 software (DNASTAR, Inc). Headache Disorders of the International Headache Society (1). CACNA1A mutation (c.1360G > A; A454T) was con- For the genetic studies, the recruited control individuals and firmed by sequencing reverse DNA strand and HphI restriction their first-degree relatives lacked any history of recurrent or analysis of exon 11 PCR product. Restriction analysis was also disabling headache. used to check for the presence of the mutation in all of the members of the pedigree and in a group of 150 unrelated Linkage Analysis. Genomic DNA was isolated from consenting healthy Spanish individuals. The mutation identified was named subjects, 10 family members, and 150 healthy unrelated indi- viduals using the QIAamp DNA Blood Maxi Kit (QIAGEN) according to Variation Society guidelines (approved by Universitat de Barcelona’s Ethical Committee (http://www.hgvs.org/) using RefSeq entry NM_001127221 as a following the Helsinki Declaration). Eight microsatellite markers cDNA reference sequence, with nucleotide 237A (ATG) cor- (synthesized using sequence data from the GDB (http://www. responding to +1, and NP_001120693 as the reference gdb.org/) and CEPH (http://www.cephb.fr/) databases) flanking sequence. Both sequences correspond to the CACNA1A tran- the CACNA1A (D19S586, D19S1165, D19S714), ATP1A2 script variant 3. (D1S3466, D1S1595, D1S1677) and SLC1A3 (D5S426, D5S418) , and three SNPs (rs8191987, rs994399, rs1542484) located Voltage Protocols and Fittings. Recordings were obtained with a D- fi fi within the SCN1A gene were genotyped in all of the available 6100 Darmstadt ampli er (List Medical) ltered at 1 kHz [only family members to test potential linkage of migraine to these for short (ms) pulses] and corrected for leak and capacitive loci. The genetic distances (in cM) between the markers are currents using the leak subtraction procedure (P/8 for short pulses according to the Marshfield map (available at http://www. and P/4 for long pulses). Currents were acquired at 33 kHz (for marshfieldclinic.org/research/pages/index.aspx). The haplotypes short pulses) or 5 kHz (for long pulses). Series-resistance com- were constructed manually by assuming the minimum number of pensation ≥80% was employed. The pClamp8 software (Axon recombinations (Fig. S1). Instruments) was used for pulse generation, data acquisition, and PCR reactions were performed under the following conditions: subsequent analysis. 50 ng genomic DNA, 0.2 mM dNTPs, 2.5 pmol each primer, 2 mM Peak inward Ca2+ currents were measured from cells clamped MgCl2, 0.5 U AmpliTaq Gold DNA polymerase (Applied Bio- at –80 mV and pulsed for 20 ms from –60 mV to +70 mV in 5-mV × fi μ systems), and 1 PCR Gold buffer in a nal volume of 25 L. steps. The following modified Boltzmann equation (Eq. 1) was fi Ampli cation conditions were 10 min at 94 °C, 35 cycles with 1 fitted to normalized current voltage (I–V) to obtain the voltage fi min at 94 °C, 1 min at 56 °C, 1 min at 72 °C, and a nal extension dependence of activation step of 10 min at 72 °C. For the microsatellite markers, the PCR products were separated on a 6% acrylamide/bisacrylamide 19:1 I ¼ GmaxðV − VrevÞ=ð1 þ expð − ðV − V1=2;actÞ=kactÞÞ; [1] gel and visualized by silver staining (2). For the SNPs, we per- formed direct sequencing of the PCR products, following the where I is thepeak current, Gmax is themaximum conductance ofthe procedure indicated below. cell, V is the membrane potential, Vrev is the extrapolated reversal potential of ICa, V1/2, act is the voltage for half-maximal current Mutation Analysis. SCN1A and SLC1A3 were not considered activation, and kact is the slope factor of the Boltzmann term. disease-causing genes in this pedigree because several hemi- Time constant for activation (τact), deactivation (τdeact), and plegic patients did not share the same haplotype. Analysis of inactivation (τinact) were obtained from single exponential fits of markers flanking the ATP1A2 gene indicated that all of the af- ICa activation phase, tail currents obtained with 20-ms prepulse fected individuals shared at least part of the genomic segment to +20 mV (to maximally open the voltage-gated P/Q channels) including the gene. The analysis of markers around the CAC- and followed by test pulses between –80 mV and +10 mV (in 5 mV NA1A gene showed that individual I.2 transmitted the same steps) for 30 ms, and the inactivation phase of I during a 3-s pulse combination of alleles to probands II.3, II.5, II.6, III.1, and III.2 Ca from a holding potential of –80 mV to a test potential of +20 mV, (all displaying FHM or MA with tongue/facial paresthesia), and respectively. a different combination of alleles was passed to proband II.1 Time course of I recovery from inactivation (τ ) was who, like her mother, presents MA without sensorimotor Ca recinact tested, applying a second pulse of 50 ms to +20 mV at increasing symptoms. Accordingly, the 47 exons of the CACNA1A gene and ATP1A2 time intervals (1-106 s) after the inactivating prepulse. Normal- the 23 exons of the gene and their corresponding exon/ fi intron junctions, including splice sites and branch points, were ized peak ICa at different times was tted to a single exponential. PCR amplified (following a standard procedure similar to that Voltage dependence of steady-state inactivation was estimated describe previously) and sequenced from individual II.5 (hemi- by measuring peak ICa currents at +20 mV following 30-s steps to – plegic migraine) and a healthy individual in 42 and 16 in- various holding potentials (from 100 to +10 mV;). During the – dependent PCR products, respectively. The coding region of the time interval between test pulses (20 ms), cells were held at 80 CACNA1A gene was sequenced also in individual II.1 (migraine mV. ICa was normalized to maximal ICa and half-maximal voltage with visual aura). The PCR products were column purified using (V1/2, inact), and slope factor for steady-state inactivation (kinact) the GFX PCR DNA and Gel Band Purification Kit (Amersham was obtained by fitting the data to the following Boltzmann Pharmacia Biotech), sequenced from either the forward or the equation (Eq. 2): www.pnas.org/cgi/content/short/0908359107 1of7 ÈÀ = = − = : [2] Total Protein Extraction. Forty-eight hours after transfection, cells I Imax ¼ 1 1 þ exp½ðV V1=2; inactÞ kinactg were washed twice with PBS solution, and whole-cell extracts were obtained by resuspension with cell scraper in BNP40 lysis buffer [50 Alternatively, voltage dependence of steady-state inactivation was mM Tris-HCl (pH 7.4), 150 mM NaCl, 0.5% Nonidet P-40, 1 mM evaluated from a 20-ms test pulse to +20 mV following a 30-s μ – – DTT, 5 mM EDTA, 0.1 mM Na3VO4, 0.1 mM PMSF, 2 g/mL prepulse to 80 mV or 20 mV (currents were normalized to aprotinin, 10 μg/mL leupeptin, 10 mM β−glycerolphospahte, the current following the –80 mV prepulse) or by the application × – 10 mM NaF] containing 1 Complete Mini inhibitor mixture of a 200-Hz train of 2-ms depolarizations from 80 mV to +20 (Roche). Cell extracts were vigorously vortexed for 15 min at 4 °C mV (in this case, currents were normalized to the current evoked and centrifuged at 18,000 × g for 30 min at 4 °C. by the first pulse of the train). Western Blotting. One hundred micrograms of total protein or 20 μg Capacitance and Amperometric Measurements. Capacitance meas- – fi of were eletrophoresed in NuPAGE Novex 3 urements were performed using an EPC-9 patch-clamp ampli er 8% Tris-Acetate precast gels (Invitrogen) or 8% Tris-glicine gels, (HEKA Electronics) using the “sine + dc” software lock-in fi respectively, and transferred onto nitrocellulose membranes us- ampli er method implemented in PULSE software. The pipette ing the iBlot Dry Blotting System (Invitrogen). Membranes were solution was similar to that indicated for ICa records, and in some 2+ blocked for 1 h at room temperature in TTBS [100 mM Tris-HCl experiments the Ca buffering was changed to 0.1 or 5 mM (pH 7.5) 150 mM NaCl, 0.1% Tween 20] containing 5% skim milk EGTA to modify the magnitude of Ca2+ microdomains. Cells – powder (blocking solution). The appropriate primary and sec- were held at 80 mV except during membrane depolarization. ondary antibodies (see below) were incubated in blocking solution The assumed reversal potential was 0 mV, and the sinusoid had overnight at 4 °C or 1 h at room temperature, respectively. an amplitude of 25 mV and a frequency of 1 kHz. The stim- Membranes were subjected to chemiluminiscence analysis using fi ulation protocol was a train of ve-step depolarizations to +20 SuperSignal West Pico Chemiluminiscent Substrate (Pierce Bio- mV for 200 ms delivered at 20 Hz, as reported previously (3). technology) and detected on Curix RP2 Plus films (Agfa). Primary Single-pulse current data were leak subtracted by hyperpo- antibodies (concentration, source, and supplier) include anti- larizing sweeps. syntaxin (1:2,000, mouse monoclonal; Sigma), anti-GFP/CFP Amperometric recordings were carried out on MPC 9/3L-AH (1:500, mouse monoclonal; Clontech), and anti-α-tubulin/β-actin fi fi cells triggered to secrete by local puf ng of a modi ed extracellular (1:3,000, mouse monoclonal; Sigma). Anti-mouse and rabbit IgG solution, where 50 mM KCl replaced equimollarly TEACl. Cells secondary antibodies (Amersham Biosciences) from a sheep and were loaded with 70 mM dopamine, following a 3-h incubation donkey source, respectively, were used at 1:2,000 dilution. period at 37 °C before the start of the experiment. The onset of the stimulus was recorded digitally via a TTL signal delivered by RNA Extraction, RT-PCR, and Semiquantitative PCR. Total RNA from the electrovalve system (Harvard Apparatus). The carbon fiber mouse cerebellum and MPC cells was extracted using Nuclospin electrodes, prepared from 10-μm diameter fibers as described RNA II Kit (Macherey-Nagel) following manufacturer’s in- previously (4), were held at 700 mV over the bath Ag/AgCl structions. RT-PCR was performed with 1 μg of total RNA using electrode by an Axoptach-200B (Molecular Devices) and pressed random primers (Promega) by the SuperScript Reverse Tran- against the cell membrane. Currents were filtered at 5 kHz scriptase System (Invitrogen). Semiquantitative PCR analyses of bandwidth and acquired using Strathclyde Electrophysiology the four different beta subunits were conducted using the fol- Data Recorder (EDR) software (provided by John Dempster, lowing primer pairs: for Cacnb1 (Gene ID: 12295, NCBI Gene University of Strathclyde, Strathclyde, UK; http://spider.science. database), forward 5′-TCAAAAGGTCAGACGGGAGT-3′ and strath.ac.uk/sipbs/page.php?page=software_ses). Analysis was reverse 5′-GTTTGGTCTTGGCTTTCTCG-3′;forCacnb2a (Gene performed using Igor Pro software version 6.0 (Wavemetrics). ID: 12296, NCBI Gene database), forward 5′-GATGGAAGCA- CATCGTCAGA-3′ and reverse 5′-TTTTTCCAACTGTGCCT- Membrane Protein Extraction. Forty-eight hours after transfection, GTG-3′;forCacnb3 (Gene ID: 12297, NCBI Gene database), cells were washed twice, detached with PBS solution, and cen- forward 5′-ACTGTGGGGTTCTGGATGAG-3′ and reverse trifuged at 335 × g for 5 min. Cell pellets were homogenized in 5′-GGCCTTCTGTTCCTGTTTGA-3′; and for Cacnb4 (Gene 20 mM Tris-HCl (pH 7.4), 140 mM NaCl, and 5 mM EDTA ID: 12298, NCBI Gene database), forward 5′-CAGGAACATT- and centrifuged twice for 10 min at 1,000 × g. The supernatant CCGAGCAACT-3′ and reverse 5′-CAAAGAGGGCTTTCT- was then transferred to polyallomer tubes and centrifuged at GCATC-3′. PCR conditions were 94 °C for 5 min; 94 °C for 30 s; 100,000 × g for 30 min. The membrane fraction obtained in the 60 °C for 30 s; 72 °C for 30 s; and 72 °C for 7 min, with 35 cycles of pellet was solubilized in 25 mM Hepes, 150 mM NaCl, 1% amplification. The PCR-derived cDNAs were analyzed by agarose CHAPS. All steps were carried out on ice, and all buffers con- gel electrophoresis, and the identity of the product was confirmed tained protease inhibitors. by sequencing.

1. Lipton RB, Bigal ME, Steiner TJ, Silberstein SD, Olesen J (2004) Classification of primary 3. Harkins AB, Cahill AL, Powers JF, Tischler AS, Fox AP (2004) Deletion of the synaptic

headaches. Neurology 63:427–435. protein interaction site of the N-type (CaV2.2) inhibits secretion in 2. Chabás A, et al. (1996) Neuronopathic and non-neuronopathic presentation of mouse pheochromocytoma cells. Proc Natl Acad Sci USA 101:15219–15224. Gaucher disease in patients with the third most common mutation (D409H) in Spain. 4. Schulte A, Chow RH (1996) A simple method for insulating carbon fiber microelectrodes J Inherit Metab Dis 19:798–800. using anodic electrophoretic deposition of paint. Anal Chem 68:3054–3058.

www.pnas.org/cgi/content/short/0908359107 2of7 Fig. S1. Pedigree segregating the A454T mutation and gene location of the nucleotide change. (A) Clinical and genetic characterization of a three-generation migraineur pedigree segregating the A454T mutation. Affected individuals are denoted by solid symbols (squares indicate male family members; circles, fe- male family members; symbols with a slash, members who had died). The genotype for the 11 polymorphic markers of the four genes (ATP1A2, SCN1A, SLC1A3, and CACNA1A) previously involved in FHM used in linkage analysis, the CACNA1A mutation carrier status, and the main clinical characteristics (type of aura) are indicated below each individual. (Inset) Order of markers and genetic distances between them in cM. (B) Conservation of the A454 amino acid residue in α1 subunits of different human calcium channels (Upper) and in the α1A subunit of the P/Q type calcium channel of different species (Lower). Nonconserved residues are shadowed in gray. The alignments were performed with clustalW (http://www.ebi.ac.uk/Tools/clustalw2/index.html). (C) Location of the single- nucleotide substitution (c.1360G > A) in the CACNA1A gene.

www.pnas.org/cgi/content/short/0908359107 3of7 Fig. S2. A454T has no effect on current density, voltage-dependent activation, and inactivation kinetics of heterologously expressed P/Q channels. Current density-voltage relationships (Left) and normalized I-V curves (Right) for WT and A454T P/Q channels containing CaVβ2a (A)orCaVβ3 (B) subunits expressed in HEK 293 cells. Peak current densities were (in pA/pF): WTβ2a (○, n = 53) 52 ± 4.2; A454Tβ2a (•, n = 34) 53.9 ± 4.9; WTβ3 (△, n = 22) 94.8 ± 16.4; and A454Tβ3 (▲, n = 16) 100.7 ± 20.6. Maximum P/Q current density from cells expressing the “fast-inactivating” regulatory CaVβ3 subunit were significantly higher (P < 0.05) than that obtained in the presence of the “slow-inactivating” palmitoylated CaVβ2a subunit without differences between WT and A454T channels (P > 0.7). No 2+ differences between WT and A454T (ANOVA, P > 0.11) were observed regarding their voltage for half-maximal Ca current (ICa) activation, the steepness of 2+ the activation curve, or the apparent reversal potential (ruling out major changes in Ca permeability) (A and B Right). V1/2, act and kact values were (in mV): WTβ2a (○, n = 53) 7.9 ± 0.4 and 4.0 ± 0.3; A454Tβ2a (•, n = 34) 7.9 ± 0.4 and 3.9 ± 0.3; WTβ3 (△, n = 22) 6.9 ± 0.3 and 3.8 ± 0.2; and A454Tβ3 (▲, n = 16) 7.3 ± 0.3 and 3.6 ± 0.2, respectively. Average Vrev values in the different groups were between 64.7 mV and 65.3 mV, with no statistically significant differences between them (ANOVA, P = 0.41). Average τact (C) and τdeact (D)ofWT(○) and A454T channels (•), at the indicated voltage, illustrating that activation and tail currents deactivation time constants were similar for all combinations of P/Q channel subunits under study (only data for CaVβ2a-containing channels are shown, ANOVA, P > 0.1). (E) Representative current traces illustrating voltage-dependent inactivation of WT (blue line) and A454T (red line) channels comprising either the β2a (Upper)ortheβ3 (Lower) subunit, in response to a 3-s depolarizing pulse. As widely reported before (reviewed in refs. 12 and 15), P/Q channels containing CaVβ2a subunit inactivated slower (Upper) than those including CaVβ3 (Lower) without significant differences between WT and A454T channels (WTβ2a, 1.01 ± 0.1 s, n =27vs.WTβ3, 0.25 ± 0.03 s, n = 14, P < 0.001; A454Tβ2a, 0.96 ± 0.09 s, n = 21 vs. A454Tβ3, 0.23 ± 0.05 s, n =6,P < 0.01). (F) Similar time 2+ course of Ca current recovery from inactivation for WT and A454T channels in the presence of the β2a (Upper)orβ3 (Lower) channel subunit. Average τ of current recovery from inactivation for the different P/Q channels, obtained after fitting the data to a single exponential, were (in s): WTβ2a (○, n = 20) 18.54 ± 2.74; A454Tβ2a (•, n = 10) 22.09 ± 4.93; WTβ3 (△, n = 14) 14.16 ± 1.99; A454Tβ3 (▲, n = 8) 9.73 ± 1.66 (ANOVA, P = 0.093).

www.pnas.org/cgi/content/short/0908359107 4of7 2+ Fig. S3. No substantial P/Q Ca currents are recorded from transfected HEK 293 cells in the absence of regulatory CaVβ subunits. (A) Current density-voltage 2+ relationships for WT P/Q channels expressed in HEK 293 in the presence or absence of CaVβ subunits, as indicated. (B) Representative Ca current traces (Upper) and average current densities (Lower) obtained in response to a 20-ms depolarizing pulse to +20 mV from HEK 293 cells expressing α1A, α2δ, and EGFP in the presence or absence of different CaVβ subunits.

www.pnas.org/cgi/content/short/0908359107 5of7 Fig. S4. Electrophysiological properties of WT and A454T PQ channels expressed in MPC 9/3L-AH cells. (ALeft) Current density-voltage relationships for WT and A454T P/Q channels expressed in MPC 9/3L-AH cells. Peak current densities were (in pA/pF): WT (○, n = 17) 132.8 ± 20.6; A454T (•, n = 9) 173.7 ± 69.3.

(Right) Normalized I-V curves for WT and A454T channels expressed in MPC 9/3L-AH cells. V1/2,act, kact, and Vrev values obtained by fitting the data to the modified Boltzmann equation (Eq. 1) were, respectively (in mV): WT (○, n = 17), 9.6 ± 0.4, 4.3 ± 0.3 and 65.8 ± 0.7; A454T (•, n =9)8.9± 0.4, 4.5 ± 0.3, and 66.1 ± 0.7. (B) Representative current traces illustrating voltage-dependent inactivation of WT (blue line) and A454T (red line) P/Q channels expressed in MPC 9/3L- AH cells, in response to a 3-s depolarizing pulse. As observed in transfected HEK 293 cells, no significant differences between WT and A454T P/Q channels expressed in MPC 9/3L-AH cells were observed regarding their maximum current density (P = 0.48; Fig. S4A Left), voltage for half-maximal activation (P = 0.29), steepness of the activation curve (P = 0.6), apparent reversal potential (P = 0.8; Fig. S4B Right), or inactivation kinetics (WT, 0.23 ± 0.05 s, n = 10 vs. A454T, 0.27 ± 0.09 s, n =8;P = 0.69; Fig. S4B). (C) Similar time course of Ca2+ current recovery from inactivation for WT and A454T P/Q channels expressed in MPC 9/3L-AH cells. Average τ of current recovery from inactivation for the different P/Q channels, obtained after fitting the data to a single exponential, were (in s): WT (○, n = 9) 13.96 ± 0.93; A454T (•, n = 6) 13.11 ± 1.09 (P = 0.56). (D) Detection of endogenous expression of Cacnb1, Cacnb2a, Cacnb3,andCacnb4 in MPC cells and mouse cerebellum by RT-PCR. Mouse brain expressed the four different beta subunits, whereas MPC cells only expressed β1 and β3 subunits. No amplification could be seen by using water as negative control (see SI Methods for methodological details).

www.pnas.org/cgi/content/short/0908359107 6of7 Fig. S5. Coupling of the human α1A channel subunit to exocytosis in MPC 9/3L-AH cells is reduced by the A454T mutation. (A) Currents from two MPC 9/3L-AH cells of similar size expressing either human WT (Upper) or human A454T α1A channel subunits (Lower) in response to a train of five successive 200-ms de- polarizing voltage steps to +20 mV delivered at 20 Hz. As observed for the rabbit α1A subunit, the A454T mutation did not affected the inactivation kinetics of P/Q currents through channels containing the human α1A subunit expressed in MPC 9/3L-AH cells in response to a 3-s depolarizing pulse (hWT, 0.14 ± 0.019 s, n = 5 vs. hA454T, 0.135 ± 0.019 s, n =5;P = 0.82). (B) Capacitance traces, plotted as a function of time, from the same cells showed in A. Human WT (hWT, □) and human A454T (hA454T, ▪) α1A expressing cells were stimulated at 20 Hz as described previously.

www.pnas.org/cgi/content/short/0908359107 7of7