CLCN1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery time may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Chloride channel protein 1 antibody; Chloride channel protein antibody; Chloride channel protein skeletal muscle antibody; CIC 1 antibody; CIC1 antibody; ClC-1 antibody; CLC1 antibody; Clcn1 antibody; CLCN1_HUMAN antibody; MGC138361 antibody; MGC142055 antibody; skeletal muscle antibody
Target Names
CLCN1
Uniprot No.

Target Background

Function
Voltage-gated chloride channel. Plays a crucial role in membrane repolarization within skeletal muscle cells subsequent to muscle contraction. The CLC channel family encompasses both chloride channels and proton-coupled anion transporters that engage in chloride or other anion exchange for protons. The lack of conserved gating glutamate residues is characteristic of family members functioning as channels.
Gene References Into Functions
  1. Novel missense CLCN1 mutations have been associated with a family affected by myotonia congenita. PMID: 29896741
  2. Loss-of-function mutations within the CLCN1 gene, located on chromosome 7q35, are the primary contributors to the pathogenesis of Thomsen myotonia. PMID: 27639085
  3. Regulation of CLC-1 chloride channel biosynthesis by FKBP8 and Hsp90beta has been described as a molecular model for myotonia congenita. PMID: 27580824
  4. The spectrum of CLCN1 mutations in patients with Myotonia Congenita has been characterized. PMID: 28427807
  5. Combining our results with previous research on Chinese populations indicates that 21 mutations in CLCN1 have been linked to myotonia congenital, while 7 mutations in SCN4A have been associated with paramyotonia congenita, and 2 mutations in SCN4A have been linked to sodium channel myotonias. PMID: 27415035
  6. A novel ClC-1 mutation, T335N, has been reported and associated with a mild phenotype. PMID: 27324117
  7. This study provides the first demonstration of ClC-1 regulation in active human muscle and offers a detailed description of the involvement of PKC and ClC-1 in the down-regulation of Gm during AP-firing activity in human skeletal muscle fibers. PMID: 26857341
  8. Novel mutations in CLCN1 were detected, and the spectrum of CLCN1 mutations known to be associated with MC was expanded. PMID: 27118449
  9. This study confirms the presence of the myotonia-causing CLCN1 mutations p.F167L and p.R105C in the Costa Rican population. PMID: 26510092
  10. Three novel mutations were found in myotonia congenita patients, including 2 missense and one splicing mutation. PMID: 26260254
  11. Three additional myotonic ClC-1 mutations have been characterized. PMID: 26096614
  12. In 4 patients (3 families) with recessive MC, 4 CLCN1 variants were found, 3 of which are novel. c.244A>G (p.T82A) and c.1357C>T (p.R453W) were found as compound heterozygotes with c.568GG>TC (p.G190S). The novel c.809G>T (p.G270V) was homozygous. PMID: 26007199
  13. Our data are consistent with the notion that the CUL4A/B-DDB1-CRBN complex catalyzes the polyubiquitination and subsequently regulates the degradation of CLC-1 channels. PMID: 26021757
  14. Sequences of PRRT2 and CLCN1 were investigated in a proband diagnosed with paroxysmal kinesigenic dyskinesia and suspected myotonia congenita; the proband and his father carried a PRRT2 c.649dupC mutation, and CLCN1 c.1723C>T and c.2492A>G mutations. This represents the first report demonstrating the coexistence of PRRT2 and CLCN1 mutations. PMID: 25205014
  15. Electrophysiological and clinical observations suggest that heterozygous CLCN1 mutations can modify the clinical and electrophysiological expression of SCN4A mutations. PMID: 25088311
  16. Splicing mutations accounted for 23 percent of all pathogenic variants in the cohort of myotonia congenita patients; 4 were heterozygous mutations in 4 unrelated individuals: c.563G>T in exon 5; c.1169-5T>G in intron 10; c.1251+1G>A in intron 11 and c.1931-2A>G in intron 16. PMID: 24452722
  17. Six mutant forms of human CLC1 demonstrate modifications of channel gating behaviors and reduced chloride conductances that likely contribute to the physiological changes of myotonia congenita. PMID: 24625573
  18. In Czech patients with myotonia congenita (MC), 34 different CLCN1 mutations were identified in 51 MC probands (14 new mutations); structural analysis of mutations in the homology model of the dimeric ClC-1 protein was performed; results demonstrate structure-function relationships in the ClC-1 protein relevant to understanding the molecular pathogenesis of MC. PMID: 24349310
  19. Early truncating mutations of CLCN1 that preclude dimerization are expected to express an autosomal recessive phenotype in patients with myotonia congenita. PMID: 23893571
  20. The common CIC-1 gate likely occludes the channel pore via interaction of E232 with a highly conserved tyrosine, Y578, at the central anion-binding site. PMID: 24064982
  21. Two mutations in the CN1 gene, inherited on the same allele as a heterozygous trait, abrogate disease expression, although when inherited singularly they were pathogenic. PMID: 23739125
  22. A family reveals that compound heterozygosity of two dominantly inheritable disease mutations exacerbates myotonia, suggesting a dosage effect of CLCN1 mutation responsible for Thomsen-type myotonia congenita. PMID: 23603549
  23. Patients with myotonia congenita exhibit a wide range of phenotypes depending on the specific ClC-1 mutation. PMID: 23933576
  24. Non-dystrophic myotonias are characterized by muscle stiffness during voluntary movement due to delayed skeletal muscle relaxation caused by mutations in the CLCN1 skeletal muscle channel genes. PMID: 23417379
  25. Genetic testing in both patients reveals previously unidentified mutations in the CLCN1 gene specific to Myotonia Congenita. PMID: 23483815
  26. Non-dystrophic myotonias are rare diseases caused by mutations in skeletal muscle CLCN1. PMID: 23771340
  27. Functional and biochemical characterization of a myotonia mutation has been conducted. PMID: 23424641
  28. Our findings predict a dramatic reduction of the muscle fiber resting chloride conductance, fully explaining the disease-causing effects of mutations C277R and C277Y of CLC1 in myotonia congenita. PMID: 22641783
  29. The co-segregation of Myotonic dystrophy type 2 with a recessive CLCN1 mutation provided the explanation for the unusual clinical findings of juvenile onset of myotonia in a 14-year-old female with Myotonic dystrophy type 2 and her affected mother. PMID: 22407275
  30. Molecular localization reveals the presence of CLCN1 mRNA transcripts in the brain, previously thought to be absent in neurons. PMID: 23408874
  31. A single heterozygous missense CLCLN1 mutation, G233S, was associated with autosomal dominant myotonia congenita in a father and son. It affected the fast gate but not the common gate. PMID: 22790975
  32. CLCN1 mutations can cause a decrease in macroscopic chloride currents, impairing membrane potential repolarization and leading to hyperexcitability in muscle membranes. PMID: 22521272
  33. ClC1 mutations exert gene dose effects and enhance myotonia and pain in DM2 in Germany. Additionally, the ClC1(236X) splice variant may contribute to myotonia in DM2. PMID: 23097607
  34. The spectrum of CLCN1 gene mutations in patients with non-dystrophic Thomsen's and Becker's myotonias in Russia has been characterized. PMID: 23113340
  35. Identification of new mutations leads to the reclassification of Trp-118-Gly in CLCN1 as a moderately pathogenic mutation and confirmation of recessive (Becker) myotonia congenita. PMID: 23152584
  36. This study describes a large Brazilian consanguineous kindred with 3 clinically affected patients with a Thomsen myotonia phenotype; they carry a novel homozygous nonsense mutation in the CLCN1 gene (K248X); none of the 6 heterozygote carriers show any signs of myotonia. PMID: 22246887
  37. A new biological role for NAD suggests that the main physiological relevance may be the exquisite sensitivity to intracellular pH that NAD(+) inhibition imparts to ClC-1 gating. PMID: 22689570
  38. Copy number variation in CLCN1 may be a significant cause of recessive myotonia congenita. PMID: 22649220
  39. This study demonstrates that 26 different mutations in the CLCN1 gene, including 13 not previously reported, have been identified. Among those 26 mutations, c.180+3A>T in intron 1 is present in nearly half of the Spanish families in this series. PMID: 22094069
  40. Elucidating the pathophysiology of myotonia mutations requires understanding multiple molecular/cellular mechanisms of CLC-1 channels in skeletal muscles, including molecular operation, protein synthesis, and membrane trafficking mechanisms. PMID: 22187529
  41. Mutations in CLCN1 contribute to the pronounced variability in the myotonic phenotype. PMID: 21204798
  42. A novel p.N440K sodium channel mutation located at the inner end of segment S6 of repeat I has been identified. PMID: 22106717
  43. Our data show that slow activation gating of the P480T ClC-1 mutation impaired the increment of chloride conductance during repetitive depolarization. PMID: 21385601
  44. Sequencing of the CLCN1 gene in 2 families with myotonia congenita revealed a heterozygous mutation (892G>A, resulting in A298T) in 1 family and compound heterozygous mutations (782A>G, resulting in Y261C; 1679T>C, resulting in M560T) in the other family. PMID: 21045501
  45. Closure of the common gate in ClC-1 channels is accompanied by a separation of the carboxyl-termini, whereas, with opening, the carboxyl-termini approach each other more closely. PMID: 21413926
  46. A novel p.L2871 mutation in the CLCN1 gene is described in a Honduran boy with myotonia congenita; his unaffected father carries the same mutation, most likely reflecting autosomal-recessive myotonia congenita. PMID: 20399394
  47. hClC-1 mutants expressed in whole-cell patch-clamped HEK293 cells showed that elimination of the carboxyl side chain in the E232Q mutation prevents fast gate closure at all voltages, but common gating is also eliminated. PMID: 20398785
  48. A novel mutation, 568GG>TC(G190S) in the CLCN1 gene, is responsible for autosomal dominant myotonia congenita with a variable phenotypic spectrum. PMID: 19697366
  49. We investigated a 62-year-old man who had mild clinical features of myotonia congenita. He was found to have a novel heterozygous G-to-A nucleotide substitution at position 1652 in exon 15 of the CLCN1 gene. PMID: 20120005
  50. CLCN1 dysfunction alone does not result in skeletal muscle morphological changes, even in advanced stages of myotonia congenita, and MRI skeletal muscle alterations in myotonic dystrophy must be clear consequences of the dystrophic disease process. PMID: 20047568

Show More

Hide All

Database Links

HGNC: 2019

OMIM: 118425

KEGG: hsa:1180

STRING: 9606.ENSP00000339867

UniGene: Hs.121483

Involvement In Disease
Myotonia congenita, autosomal dominant (MCAD); Myotonia congenita, autosomal recessive (MCAR)
Protein Families
Chloride channel (TC 2.A.49) family, ClC-1/CLCN1 subfamily
Subcellular Location
Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Predominantly expressed in skeletal muscles.

Q&A

What is CLCN1 and what role does it play in skeletal muscle physiology?

CLCN1, also known as ClC-1, is a voltage-gated chloride channel that generates most of the plasma membrane chloride conductance in skeletal muscle fibers. It serves crucial functions in:

  • Stabilizing resting membrane potential

  • Contributing to repolarization during action potential firing

  • Maintaining normal muscle excitability and contraction-relaxation cycles

CLCN1 forms a homodimeric channel structure where each subunit contains its own ion conduction pathway. It conducts double-barreled currents controlled by two types of gates: fast glutamate gates that independently control each subunit and a slow common gate that simultaneously regulates both subunits . The channel demonstrates significant open probability at muscle resting potential and becomes further activated upon membrane depolarization. CLCN1 shows ion selectivity in the order of chloride > thiocyanate > bromide > nitrate > iodide .

What experimental techniques can effectively detect CLCN1 protein expression?

Multiple validated techniques are available for CLCN1 detection in research settings:

TechniqueApplicationsRecommended DilutionsValidated Species
Western Blot (WB)Protein expression analysis1:200-1:1000Human, Mouse, Rat
Immunohistochemistry (IHC)Tissue localization1:50-1:500Human, Mouse
ELISAQuantitative analysisVariable (antibody-dependent)Human, Mouse

For optimal results in IHC applications, antigen retrieval with TE buffer (pH 9.0) is recommended, though citrate buffer (pH 6.0) may serve as an alternative . Always validate antibody dilutions for your specific experimental conditions.

What are the critical considerations for CLCN1 antibody storage and handling?

Proper storage and handling are essential for maintaining antibody functionality:

  • Lyophilized antibodies should be stored at -20°C upon arrival

  • Reconstituted solutions can be stored at 4°C for up to 1 week

  • For longer storage periods, prepare small aliquots and store at -20°C

  • Avoid multiple freeze-thaw cycles as they can compromise antibody integrity

  • Centrifuge all antibody preparations before use (10000 × g for 5 minutes)

Reconstitution should be performed with 50 μL or 0.2 mL double-distilled water, depending on sample size. Some formulations may be supplied in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3) .

How can CLCN1 antibodies be utilized to investigate myotonia congenita and related channelopathies?

Myotonia congenita is a muscle disorder resulting from mutations in the CLCN1 gene. Researchers investigating this condition can employ CLCN1 antibodies in multiple experimental approaches:

  • Mutation characterization: CLCN1 antibodies can evaluate protein expression levels of various mutants in heterologous expression systems. For example, studies have examined mutations like F167L, G190S, and A531V using patch-clamp techniques combined with immunodetection .

  • Functional correlation: Correlating electrophysiological data with protein expression patterns can reveal how specific mutations affect channel trafficking, membrane integration, and function. Patch-clamp recordings from HEK293 cells expressing wild-type versus mutant channels have demonstrated distinct current-voltage relationships and activation properties .

  • Animal model validation: In myotonia models, antibodies can confirm the presence and localization of mutant channels. Western blot analysis of skeletal muscle membranes provides critical validation of animal models .

  • Therapeutic development assessment: When evaluating potential therapeutics, antibodies help monitor changes in CLCN1 expression. For instance, antisense oligonucleotide therapies targeting CLCN1 alternative splicing can be validated using antibody-based detection methods .

What methodological approaches should researchers consider when optimizing immunohistochemistry protocols for CLCN1 detection?

Optimizing IHC protocols for CLCN1 requires careful consideration of several parameters:

  • Tissue preparation: Fresh skeletal muscle tissue should be rapidly fixed to preserve membrane protein structure. Flash-freezing in isopentane cooled with liquid nitrogen is recommended for cryosections.

  • Antigen retrieval: Heat-induced epitope retrieval using TE buffer (pH 9.0) typically provides optimal results, though some protocols may benefit from citrate buffer (pH 6.0) .

  • Antibody validation: Always include positive controls (skeletal muscle tissue) and negative controls (tissues known not to express CLCN1 or primary antibody omission).

  • Detection systems: For low abundance detection, amplification systems such as tyramide signal amplification may enhance sensitivity while maintaining specificity.

  • Cross-reactivity assessment: When examining tissues from different species, consider sequence homology at the epitope region. The N-terminal region (residues 102-117) has been used successfully for generating antibodies that work across species .

  • Counterstaining optimization: Minimal counterstaining is recommended to avoid masking specific CLCN1 signals, particularly when evaluating membrane localization.

What experimental strategies can be employed to study CLCN1 alternative splicing in myotonic dystrophy using antibody-based techniques?

Myotonic dystrophy type 1 (DM1) involves aberrant alternative splicing of CLCN1, leading to myotonia. Several antibody-based approaches can investigate this mechanism:

  • Isoform-specific antibodies: Developing antibodies that specifically recognize exon-included or exon-excluded CLCN1 variants enables quantification of splicing changes.

  • Combined RT-PCR and immunoblotting: Correlating mRNA splicing patterns with protein expression provides comprehensive insight into post-transcriptional regulation.

  • Immunofluorescence colocalization: Determining subcellular localization of differentially spliced CLCN1 isoforms helps evaluate functional consequences of splicing alterations.

  • Therapeutic response assessment: Following antisense oligonucleotide treatment targeting CLCN1 exon 7a, antibodies can confirm the restoration of proper CLCN1 expression patterns. This approach has demonstrated that correction of CLCN1 alternative splicing increases glycolytic 2B fiber levels to ≥40% frequency, reduces muscle injury, and improves fiber hypertrophy .

  • Pulse-chase experiments: Antibody-based detection in pulse-chase studies can reveal differential stability and trafficking of alternatively spliced CLCN1 isoforms.

What approaches can be used to validate CLCN1 antibody specificity in experimental models?

Rigorous validation of antibody specificity is critical for reliable CLCN1 research:

  • Blocking peptide controls: Pre-incubation of the antibody with its immunizing peptide should abolish specific signals. Western blot analysis of rat skeletal muscle membranes has demonstrated this approach, where signals detected with anti-CLCN1 antibody (1:200 dilution) were eliminated when the antibody was pre-incubated with the corresponding blocking peptide .

  • Knockout/knockdown controls: Tissues or cells lacking CLCN1 expression (through genetic knockout or siRNA knockdown) provide definitive negative controls.

  • Heterologous expression systems: Overexpression of tagged CLCN1 in cell lines like HEK293 allows comparison between antibody detection and tag-based detection.

  • Cross-species reactivity testing: Evaluating reactivity across species with known sequence differences can confirm epitope specificity.

  • Multiple antibody concordance: Utilizing antibodies targeting different CLCN1 epitopes should yield consistent expression patterns if each is specific.

What are the critical parameters for successful co-immunoprecipitation experiments using CLCN1 antibodies?

Co-immunoprecipitation (co-IP) with CLCN1 antibodies requires careful optimization:

  • Membrane protein solubilization: CLCN1 is a transmembrane protein requiring appropriate detergents for solubilization without disrupting protein-protein interactions. Mild detergents like digitonin (0.5-1%) or CHAPS (0.5-2%) are recommended initial choices.

  • Antibody selection: Choose antibodies validated for immunoprecipitation applications, preferably targeting accessible epitopes like the cytoplasmic N-terminal region (residues 102-117) .

  • Control conditions: Include isotype controls, pre-immune serum controls, and when possible, CLCN1-deficient samples.

  • Buffer optimization: Consider ionic strength, pH, and divalent cation concentration, as these can affect channel conformation and protein interactions.

  • Crosslinking consideration: For transient interactions, membrane-permeable crosslinkers may stabilize complexes prior to solubilization.

  • Elution conditions: Native elution with competing peptides may preserve complex integrity better than denaturing conditions for downstream functional studies.

How can researchers distinguish between different CLCN1 splice variants using antibody-based techniques?

Distinguishing CLCN1 splice variants requires strategic experimental design:

  • Epitope selection: Antibodies raised against exon-exon junction sequences can specifically recognize particular splice variants.

  • Combined methodologies: Integrating RT-PCR results with protein detection provides validation of splice variant expression.

  • Size discrimination: Some splice variants display detectable size differences on Western blots that can be resolved with high-percentage or gradient gels.

  • Functional correlation: Correlating antibody detection with electrophysiological properties can confirm the presence of specific functional variants. For instance, in DM1 research, antisense oligonucleotide treatment targeting CLCN1 exon 7a corrects alternative splicing and restores normal muscle physiology, which can be monitored using appropriate antibodies .

  • Phosphorylation-state specific antibodies: Some splice variants may exhibit differential post-translational modification patterns that can be detected with phospho-specific antibodies.

How can CLCN1 antibodies contribute to therapeutic development for myotonic disorders?

CLCN1 antibodies play crucial roles in therapeutic development pipelines:

  • Target engagement verification: Antibodies can confirm whether potential therapeutics successfully modulate CLCN1 expression or localization.

  • Mechanism validation: For splice-correction approaches, antibodies help verify that interventions restore proper CLCN1 isoform expression. Recent studies demonstrated that antisense oligonucleotides targeting CLCN1 exon 7a successfully correct alternative splicing, resulting in functional improvement and muscle fiber type restoration in DM1 models .

  • Biomarker development: CLCN1 expression patterns detected by antibodies may serve as biomarkers for disease progression or therapeutic response.

  • Pharmacodynamic assessment: Quantitative analysis of CLCN1 levels and distribution provides pharmacodynamic data for dose-optimization studies.

  • Safety evaluation: Monitoring potential off-target effects on CLCN1 expression in non-muscle tissues during pre-clinical safety studies.

What considerations should researchers make when comparing CLCN1 antibody data across different species models?

Cross-species research with CLCN1 antibodies requires careful interpretation:

  • Epitope conservation analysis: Evaluate sequence homology at the epitope region across target species. For instance, antibodies targeting the N-terminal region (residues 102-117) have demonstrated cross-reactivity between rat and other species .

  • Validation in each species: Even with predicted reactivity, experimental validation in each species is essential. Some antibodies have confirmed reactivity with human and mouse samples, while others may work with rat tissues as well .

  • Species-specific controls: Include appropriate positive and negative controls from each species under investigation.

  • Expression pattern differences: Normal expression patterns may vary between species; for example, CLCN1 expression levels and distribution patterns may differ between rodent and human skeletal muscle.

  • Experimental condition standardization: When comparing across species, standardize sample preparation, antibody concentrations, and detection methods to minimize technical variables.

What approaches can researchers use to quantitatively assess CLCN1 protein expression changes in disease models?

Accurate quantification of CLCN1 expression requires rigorous methodology:

MethodAdvantagesLimitationsBest Applications
Western BlotProtein size verification, semi-quantitativeLimited spatial informationExpression level changes
Quantitative IHCSpatial information, cell-specific expressionComplex standardizationLocalization changes
Flow CytometrySingle-cell quantificationRequires cell dissociationPopulation distribution analysis
ELISAHigh throughput, quantitativeLimited isoform discriminationLarge sample comparisons
Mass SpectrometryAbsolute quantificationComplex sample preparationDetailed proteomic analysis

For Western blot quantification, normalization to stable reference proteins (not affected by the experimental condition) is essential. When analyzing CLCN1 in skeletal muscle from disease models, consider the following:

  • Use loading controls appropriate for the subcellular fraction being analyzed

  • Implement technical replicates to account for transfer variability

  • Verify signals fall within the linear dynamic range of detection

  • Consider using fluorescent secondary antibodies for more accurate quantification

In studies of myotonic dystrophy, quantitative analysis of CLCN1 expression following corrective treatments has demonstrated significant restoration of proper expression patterns correlating with functional improvement .

How can researchers effectively combine electrophysiological techniques with antibody-based detection to study CLCN1 function?

Integrating electrophysiology with immunodetection provides powerful functional insights:

  • Patch-clamp with immunocytochemistry: Following electrophysiological recording from cells expressing wild-type or mutant CLCN1, fixation and immunolabeling can correlate functional data with expression levels and subcellular localization.

  • Structure-function correlation: Mutations like F167L, G190S, and A531V have been characterized using patch-clamp techniques in HEK293 cells, where voltage steps applied between -200 mV and +120 mV (or -120 mV and +200 mV for G190S) revealed distinct functional impairments that can be correlated with antibody-detected expression patterns .

  • Current density normalization: Normalizing electrophysiological currents to protein expression levels (quantified by antibody-based methods) provides more accurate functional comparisons between constructs with different expression efficiencies.

  • Single-cell correlation: Combining whole-cell patch-clamp with post-hoc immunolabeling permits direct correlation between a cell's functional properties and its CLCN1 expression level/pattern.

  • Activity-dependent trafficking: Antibody labeling following various stimulation protocols can reveal activity-dependent changes in CLCN1 surface expression and localization that explain functional observations.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.