C1QBP Human

Complement Component 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mitochondrial Oxidative Phosphorylation (OXPHOS)

C1QBP is essential for mitochondrial energy production:

  • Respiratory Chain Support: Maintains translation of mitochondrial-encoded subunits of complexes I, III, IV, and V .

  • PDH Regulation: Binds to the dihydrolipoyl-lysine acetyltransferase (DLAT) subunit of PDH, enhancing pyruvate-to-acetyl-CoA conversion and TCA cycle activity .

  • Mitochondrial Morphology: Prevents fragmentation and swelling by inhibiting OMA1-mediated proteolytic processing of OPA1 .

Experimental Evidence:

  • C1QBP-deficient mice exhibit disordered mitochondrial alignment, reduced OXPHOS activity, and embryonic lethality .

  • Knockdown in cardiac myocytes causes complex I/IV deficiencies and cardiomyopathy .

Autophagy and Mitochondrial Quality Control

C1QBP regulates mitophagy via interaction with ULK1:

  • ULK1 Stabilization: Prevents polyubiquitination and proteasomal degradation of ULK1, preserving mitophagy under stress .

  • Starvation Response: Enhances cellular survival by balancing OXPHOS and autophagy .

Immune Modulation

C1QBP influences immune cell metabolism and function:

  • T Cell Differentiation: Required for effector CD8⁺ T cell differentiation by regulating acetyl-CoA, fumarate, and 2-HG production, affecting histone acetylation and transcriptional programs .

  • Dendritic Cell (DC) Maturation: Modulates mitochondrial metabolism to control DC activation and immune responses .

Disease Associations

Biallelic C1QBP mutations cause mitochondrial cardiomyopathy and multisystem disorders:

MutationPhenotypeReference
L275FCardiac hypertrophy, exercise intolerance, progressive external ophthalmoplegia (PEO)
R218QIntrauterine growth restriction, hepatomegaly, cerebral edema
R270CVentricular arrhythmias, peripheral nervous system dysfunction

Clinical Features:

  • Cardiomyopathy: Left ventricular hypertrophy, cardiomegaly, and respiratory chain deficiencies .

  • Neuromuscular Involvement: PEO, ptosis, and skeletal muscle weakness .

  • Metabolic Dysregulation: Impaired urea cycle (elevated ornithine, reduced citrulline) .

Research Models and Therapeutic Implications

  • Mouse Models: Cardiomyocyte-specific C1QBP knockout mice show mitochondrial dysfunction and premature death .

  • iPSC-CMs: Patient-derived induced pluripotent stem cell-cardiomyocytes (iPSC-CMs) replicate hypertrophic phenotypes and mitochondrial disorganization .

  • Therapeutic Targets: Modulating C1QBP-PDH interactions or ULK1 stability may address mitochondrial diseases or cancer metabolism .

Functional Localization in Neurons

In the brain, C1QBP is neuron-specific and predominantly mitochondrial:

  • Key Brain Regions: Cerebral cortex, hypothalamic paraventricular nucleus (PVN), spinal trigeminal nucleus .

  • Role in Neuroplasticity: May regulate local energy supply and synaptic function, though mechanisms remain under investigation .

Product Specs

Introduction
C1QBP, with accession number NP_001203, binds to the globular heads of C1q, inhibiting C1 activation. This protein interacts with various ligands and plays a role in cell signaling. It associates with C1r and C1s to form the first component of the serum complement system. C1QBP is also recognized as the p32 subunit of pre-mRNA splicing factor SF2 and a hyaluronic acid-binding protein. Moreover, it serves as a marker for tumor cells and tumor-associated macrophages/myeloid cells in hypoxic/metabolically deprived tumor regions. Mitochondrial C1QBP is crucial in p14ARF-induced apoptosis. The protein acts as a chemotactic factor for immature dendritic cells, with migration occurring through the binding of both C1QBP and cC1qR/CR. Overexpression of C1QBP effectively hinders mRNA accumulation from the adenovirus major late transcription unit (MLTU) and promotes RNA polymerase II carboxy-terminal domain phosphorylation in virus-infected cells. Additionally, C1QBP interacts with the Hepacivirus core protein on CD8+ and CD4+ T cells, inactivating lck and akt.
Description
Recombinant human C1QBP, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 210 amino acids. It has a molecular weight of 23.9 kDa.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The C1QBP protein is supplied in a solution containing 20mM Tris-HCl at pH 7.5, 20% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 95.0% as determined by SDS-PAGE analysis.
Synonyms
p32, HABP1, gC1Qr, GC1QBP, SF2p32, gC1Q-R, Complement component 1 Q subcomponent-binding protein mitochondrial, Glycoprotein gC1qBP, C1qBP, GC1q-R protein, Hyaluronan-binding protein 1, Mitochondrial matrix protein p32, p33, C1QBP.
Source
Escherichia Coli.
Amino Acid Sequence
MLHTDGDKAF VDFLSDEIKE ERKIQKHKTL PKMSGGWELE LNGTEAKLVR KVAGEKITVT FNINNSIPPT FDGEEEPSQG QKVEEQEPEL TSTPNFVVEV IKNDDGKKAL VLDCHYPEDE VGQEDEAESD IFSIREVSFQ STGESEWKDT NYTLNTDSLD WALYDHLMDF LADRGVDNTF ADELVELSTA LEHQEYITFL EDLKSFVKSQ.

Q&A

What is C1QBP and where is it primarily localized in human cells?

C1QBP (Complement C1q binding protein) is a multifunctional protein that predominantly resides in the mitochondria, directed there by its 33-residue N-terminal mitochondria-targeting signal (MTS) sequence. While initially identified for its role in complement activation, increasing evidence has demonstrated that C1QBP is essential for embryonic development through its functions in mitochondrial translation and oxidative phosphorylation (OXPHOS) . The protein is ubiquitously expressed across various cell types and has been shown to play critical roles in maintaining mitochondrial structure and function.

To study C1QBP localization, researchers typically employ immunofluorescence staining with mitochondrial markers, subcellular fractionation followed by Western blotting, and electron microscopy techniques to visualize its precise distribution within the mitochondrial compartments.

How does C1QBP contribute to mitochondrial morphology and dynamics?

C1QBP regulates mitochondrial morphology through multiple mechanisms including:

  • Regulation of OMA1-dependent proteolytic processing of OPA1, which influences mitochondrial fragmentation and swelling

  • Modulation of mitochondrial fusion proteins (Mfn1 and Mfn2) and fission protein Drp-1

Research has demonstrated that C1QBP overexpression increases mitochondrial fibrils, while C1QBP siRNA treatment results in predominantly small spherical mitochondria compared to the normal elongated mitochondria in control cells . This morphological shift is accompanied by detectable loss of Mfn1 and Mfn2 along with decreased Drp-1 levels.

Methodological approaches to study these dynamics include live-cell imaging with fluorescent mitochondrial markers, transmission electron microscopy for ultrastructural analysis, and biochemical assays measuring the expression and activity of fusion/fission proteins.

What is the relationship between C1QBP and mitochondrial quality control?

C1QBP plays a crucial role in mitochondrial quality control primarily through its regulation of mitophagy - the selective autophagy of damaged mitochondria. Research has shown that:

  • C1QBP controls mitochondrial autophagy to help cells adapt to challenging microenvironments

  • It forms a protein complex with Unc-51-like kinase-1 (ULK1), preventing ULK1's polyubiquitylation and proteasome-mediated degradation

  • The interaction between ULK1 and C1QBP is indispensable for maintaining steady-state levels of ULK1

Notably, mitophagy defects can be recovered by re-introducing ULK1 into C1QBP-deficient cells, suggesting that C1QBP protects mitophagy through the prevention of ULK1 degradation . This C1QBP-Ulk1-mitophagy axis provides a survival advantage when nutrients are scarce, which ultimately can promote tumorigenesis in cancer contexts.

To investigate this relationship, researchers utilize mitophagy flux assays, co-immunoprecipitation studies, protein stability assays, and fluorescent reporter systems that track mitochondrial degradation.

How is C1QBP expression altered in human cancers?

C1QBP is expressed at high levels in a significant number of tumor types compared with their nonmalignant counterparts, including:

  • Melanoma

  • Colon cancer

  • Ovarian cancer

  • Gastric cancer

  • Prostate cancer

  • Brain tumors

  • Breast cancer

Multiple clinical studies have demonstrated that C1QBP expression is positively linked to tumor stage and poor prognosis across various cancer types . High expression levels of C1QBP are inversely correlated with tumor patients' prognosis, making it a potentially valuable independent prognostic marker of outcomes in cancer patients.

For investigating C1QBP expression in tumors, researchers commonly employ immunohistochemistry on tissue microarrays, quantitative PCR, Western blotting, and analysis of publicly available genomic and transcriptomic cancer databases.

How does C1QBP influence metabolic reprogramming in cancer cells?

C1QBP mediates tumor cell metabolic reprogramming through several sophisticated mechanisms:

  • Regulation of oxidative phosphorylation: Knocking down C1QBP expression in human cancer cells strongly shifts their metabolism from oxidative phosphorylation (OXPHOS) to glycolysis . This metabolic shift occurs because C1QBP deficiency reduces the synthesis of mitochondrial-DNA-encoded OXPHOS polypeptides, impairing several electron transport complexes' functions .

  • Modulation of glutamine metabolism: C1QBP is involved in glutamine oxidation, which is essential for mammalian cell proliferation and associated with tumor progression . Research has shown that Myc can upregulate C1QBP transcription, resulting in enhanced glutamine metabolism, particularly in malignant brain cancers .

  • Metabolic flexibility: By maintaining mitochondrial function, C1QBP enables cancer cells to adapt to varying nutrient availability in the tumor microenvironment.

To study these metabolic alterations, researchers utilize metabolic flux analysis (such as Seahorse technology), stable isotope tracing with 13C-labeled glucose or glutamine, metabolomics profiling, and assessment of key metabolic enzyme activities under different C1QBP expression conditions.

What is the dual role of C1QBP in tumor and immune cell function?

C1QBP exhibits a fascinating dual role in cancer biology, affecting both tumor cells and immune cells:

In tumor cells:

  • Promotes mitochondrial plasticity and metabolic flexibility

  • Enhances tumor cell proliferation and metastatic potential

  • Contributes to therapeutic resistance

In immune cells:

  • Required for dendritic cell (DC) maturation through regulation of pyruvate dehydrogenase (PDH) activity

  • C1QBP binds to PDH-E2, promoting PDH activity and facilitating citrate production, which enhances fatty acid synthesis and endoplasmic reticulum expansion in DCs after lipopolysaccharide stimulation

  • Essential for CD8+ T cell differentiation by augmenting OXPHOS and controlling the production of metabolites like acetyl-CoA, fumarate, and 2-HG

This dual role creates a therapeutic conundrum: inhibiting C1QBP may reduce tumor growth but could simultaneously impair anti-tumor immune responses. Therefore, the manipulation of C1QBP must be carefully calibrated to "adjust the competitive balance between tumor cells and immune cells" .

Research approaches to investigate this dual role include co-culture systems, immune cell functional assays, tumor models in immunocompetent versus immunodeficient mice, and single-cell metabolic profiling.

How does C1QBP regulate CD8+ T cell differentiation and function?

C1QBP plays a critical role in CD8+ T cell biology through several mechanisms:

  • Mitochondrial respiratory capacity: C1QBP deficiency in T cells intrinsically impairs the differentiation of effector CD8+ T cells by preventing the increase in mitochondrial respiratory capacities required during activation .

  • Metabolite regulation: C1QBP controls the production of key metabolites in CD8+ T cells:

    • Acetyl-CoA: C1QBP deficiency decreases acetyl-CoA levels

    • Fumarate and 2-HG: Levels increase with C1QBP deficiency

  • Epigenetic programming: Decreased acetyl-CoA resulting from C1QBP deficiency dampens histone protein H3K27 acetylation, which enhances CpG methylation. This epigenetic alteration reduces expression of genes encoding master transcription factors that drive effector CD8+ T cell differentiation, including Id2, Prdm1, and Tbx21 .

  • Metabolic requirements: Research has shown that activated CD8+ T cells have greater glycolytic and OXPHOS requirements than activated CD4+ T cells and are more sensitive to OXPHOS impairment, making C1QBP particularly important for CD8+ T cell function .

To investigate these mechanisms, researchers use conditional knockout mouse models, chromatin immunoprecipitation sequencing (ChIP-seq), metabolomics, T cell transfer experiments, and functional T cell assays measuring cytokine production and cytotoxicity.

What experimental approaches can determine the role of C1QBP in dendritic cell maturation?

To study C1QBP's role in dendritic cell maturation, researchers can employ several methodological approaches:

  • Genetic manipulation: Generate C1QBP knockout or knockdown in dendritic cells using CRISPR-Cas9 or RNAi technologies, followed by comparison with wild-type cells during maturation processes.

  • Metabolic profiling: Measure key metabolic parameters in wild-type versus C1QBP-deficient dendritic cells:

    • Pyruvate and lactate production

    • Citrate production after lipopolysaccharide stimulation

    • PDH activity assays

  • Protein interaction studies: Perform immunoprecipitation experiments to confirm C1QBP binding to PDH-E2 and identify other potential interaction partners in the PDH complex .

  • Morphological assessment: Monitor endoplasmic reticulum expansion (which occurs in wild-type but not C1QBP-deficient DCs after LPS stimulation) using electron microscopy or fluorescent ER markers .

  • Functional assays: Assess DC antigen presentation capabilities, T cell stimulatory capacity, and cytokine production profiles with and without C1QBP.

Research has revealed that C1QBP controls PDH activity by binding to PDH-E2, thereby regulating citrate production to support DC maturation . This mechanism impacts fatty acid synthesis and endoplasmic reticulum expansion, which are crucial for proper DC function.

How might C1QBP be therapeutically targeted in cancer treatment?

The dual role of C1QBP in tumor and immune cells presents both challenges and opportunities for therapeutic targeting:

  • Differential targeting approach: The ideal strategy would be to inhibit C1QBP specifically in tumor cells while preserving or enhancing its function in immune cells . This could potentially be achieved through:

    • Tumor-specific delivery systems (nanoparticles, antibody-drug conjugates)

    • Exploitation of differences in subcellular localization or binding partners between tumor and immune cells

    • Targeting tumor-specific C1QBP interactions or modifications

  • Combination therapy strategies: Since potentiation of immune cells through enhancement of mitochondrial plasticity can prevent immune exhaustion and promote durable antitumor immunity:

    • Combine C1QBP modulation with immune checkpoint inhibitors (anti-PD-L1, PD-1, CTLA-4)

    • Enhance T cell mitochondrial fitness to enable persistent immune function in the stressful tumor microenvironment

  • Metabolism-based interventions: Since C1QBP influences both fatty acid oxidation (FAO) and oxidative phosphorylation:

    • Target metabolic pathways differentially regulated by C1QBP in tumor versus immune cells

    • Explore whether metabolic reprogramming through modulation of C1QBP would impact the generation of long-lived memory T cells, as FAO engagement is critical for memory T cell formation

Research methods to investigate these approaches include in vitro drug screening, patient-derived xenograft models, immune-competent mouse models, and ex vivo testing on human tumor samples and immune cells.

What are the key experimental controls necessary when studying C1QBP function?

When investigating C1QBP function, researchers should implement several critical controls:

  • Genetic manipulation validation:

    • Confirm knockout/knockdown efficiency at both mRNA and protein levels

    • Use multiple siRNA/shRNA sequences to minimize off-target effects

    • Include rescue experiments by re-expressing C1QBP to verify observed phenotypes are specific to C1QBP loss

  • Subcellular localization controls:

    • Use mitochondrial markers to confirm C1QBP co-localization

    • Include non-mitochondrial markers to demonstrate specificity

    • Consider mitochondrial subcompartment markers to precisely locate C1QBP within mitochondria

  • Functional assays:

    • Include metabolic inhibitors as positive controls (e.g., oligomycin for OXPHOS inhibition)

    • Perform parallel experiments with established mitochondrial regulators

    • Use both gain- and loss-of-function approaches when possible

  • Cell type-specific considerations:

    • When studying C1QBP in immune cells, include proper immune cell activation controls

    • For tumor studies, compare matched tumor and adjacent normal tissue whenever possible

    • Use multiple cell lines from the same cancer type to account for heterogeneity

These controls help ensure the specificity and reliability of results when studying this multifunctional protein across different cellular contexts.

How can researchers accurately measure C1QBP-dependent changes in mitochondrial function?

To accurately assess C1QBP-mediated alterations in mitochondrial function, researchers should employ a comprehensive set of complementary techniques:

  • Respiratory capacity analysis:

    • Seahorse XF Analyzer to measure oxygen consumption rate (OCR) and extracellular acidification rate (ECAR)

    • High-resolution respirometry for detailed analysis of individual respiratory complexes

    • Clark-type electrode measurements of isolated mitochondria

  • Mitochondrial membrane potential:

    • Fluorescent probes (TMRM, JC-1) with appropriate controls

    • Flow cytometry for population analysis

    • Live-cell imaging for temporal dynamics

  • ROS production:

    • MitoSOX for mitochondrial superoxide

    • DCF-DA for generalized cellular ROS

    • Genetically encoded redox sensors for compartment-specific measurements

  • Mitochondrial dynamics assessment:

    • Live-cell imaging with mitochondrial markers (MitoTracker, mito-GFP)

    • Quantification of mitochondrial morphology parameters (length, branching, area)

    • Electron microscopy for ultrastructural analysis

  • Metabolomics approaches:

    • Targeted metabolomics focusing on TCA cycle intermediates

    • Stable isotope tracing (13C-glucose, 13C-glutamine) to track metabolic flux

    • Integration with proteomics data on mitochondrial proteins

These methodologies, when used in combination, provide a comprehensive picture of how C1QBP impacts various aspects of mitochondrial function in both normal and pathological conditions.

What are the potential pitfalls in interpreting C1QBP's role in cancer metabolism?

Researchers should be aware of several potential confounding factors when studying C1QBP in cancer metabolism:

  • Context-dependent effects:

    • C1QBP functions may vary significantly across cancer types

    • The same cancer type at different stages may exhibit different C1QBP dependencies

    • The tumor microenvironment can dramatically influence metabolic requirements and C1QBP function

  • Compensatory mechanisms:

    • Long-term C1QBP depletion may trigger compensatory metabolic rewiring

    • Acute versus chronic C1QBP inhibition may yield different phenotypes

    • Alternative pathways may mask the full impact of C1QBP modulation

  • Technical considerations:

    • In vitro culture conditions (particularly oxygen and nutrient levels) poorly recapitulate the tumor microenvironment

    • Standard cell culture media containing supraphysiological levels of glucose and glutamine may obscure metabolic dependencies

    • Immortalized cell lines may not accurately represent primary tumor metabolism

  • Translational challenges:

    • Mouse models may not fully recapitulate human tumor metabolism

    • Patient heterogeneity means findings in one cohort may not generalize

    • Xenograft models lack the immune component critical for understanding C1QBP's dual role

To address these pitfalls, researchers should:

  • Use multiple experimental models and approaches

  • Validate findings in primary patient samples when possible

  • Consider both acute and chronic C1QBP modulation

  • Perform studies under conditions that better mimic the tumor microenvironment (hypoxia, nutrient limitation)

  • Integrate multi-omics approaches to capture the full spectrum of C1QBP-dependent changes

How might C1QBP function in long-term immune memory formation?

An intriguing area for future investigation is C1QBP's potential role in immune memory formation. Current evidence suggests several promising research avenues:

  • Metabolic regulation of memory T cells:

    • Since fatty acid oxidation (FAO) engagement is critical for the generation of memory T cells , and C1QBP is involved in regulating lipid metabolism and homeostasis, researchers should investigate whether C1QBP influences memory T cell formation and persistence.

    • Experimental approaches could include tracking C1QBP expression during the transition from effector to memory T cells and examining the metabolic profiles of memory precursors with varying levels of C1QBP.

  • Epigenetic programming:

    • C1QBP's influence on acetyl-CoA production affects histone acetylation in effector CD8+ T cells . Similar epigenetic mechanisms might be involved in establishing the memory T cell transcriptional program.

    • ChIP-seq and ATAC-seq analyses comparing wild-type and C1QBP-deficient memory T cells could reveal critical epigenetic signatures.

  • Mitochondrial health maintenance:

    • Long-lived memory T cells require healthy mitochondria for their persistence and rapid recall response.

    • C1QBP's role in mitochondrial quality control through mitophagy might be essential for maintaining the mitochondrial network integrity in memory T cells over extended periods.

These investigations could have significant implications for vaccine development and cancer immunotherapy strategies aiming to generate durable immune responses.

What is the potential for developing selective C1QBP modulators for cancer treatment?

Developing selective C1QBP modulators represents a challenging but promising avenue for cancer therapeutics:

  • Structure-based drug design approaches:

    • Detailed structural analysis of C1QBP interactions with different binding partners in tumor cells versus immune cells

    • Identification of druggable pockets that could selectively disrupt tumor-promoting interactions while preserving immune cell functions

    • Computational screening of compound libraries followed by biochemical validation

  • Cell type-specific delivery strategies:

    • Development of tumor-targeting nanoparticles carrying C1QBP inhibitors

    • Antibody-drug conjugates directed against tumor-specific surface markers

    • Exploitation of the tumor microenvironment (pH, hypoxia) for selective drug activation

  • Combination therapy potential:

    • Integration with immune checkpoint inhibitors to enhance T cell function while targeting tumor metabolism

    • Sequential treatment approaches that first target tumor cells and then boost immune function

    • Complementary metabolic interventions that enhance the effect of C1QBP modulation

  • Biomarker development:

    • Identification of patient populations most likely to benefit from C1QBP-targeted therapies

    • Development of companion diagnostics to monitor treatment efficacy

    • Establishment of predictive markers for potential resistance mechanisms

The ideal therapeutic strategy would involve inhibiting C1QBP specifically in tumor cells while preserving or enhancing its function in immune cells, as this would beneficially adjust the competitive balance between tumor and immune cells .

How does C1QBP interact with other mitochondrial quality control pathways?

Future research should explore the integration of C1QBP with broader mitochondrial quality control networks:

  • Interactions with the PINK1-Parkin pathway:

    • Investigation of potential crosstalk between C1QBP-ULK1 and PINK1-Parkin pathways in mitophagy regulation

    • Examination of mitochondrial ubiquitination patterns in the presence and absence of C1QBP

    • Assessment of how these pathways might cooperate or compensate for each other under different stress conditions

  • Relationship with mitochondrial unfolded protein response (UPRmt):

    • Exploration of whether C1QBP influences the activation of the UPRmt

    • Analysis of how C1QBP-mediated mitochondrial translation affects proteostasis

    • Investigation of potential protective mechanisms against mitochondrial stress

  • Role in mitochondrial dynamics beyond currently known interactions:

    • Detailed characterization of the C1QBP interactome in different cell types

    • Investigation of temporal dynamics of C1QBP localization during mitochondrial stress

    • Examination of potential post-translational modifications of C1QBP that regulate its function

  • Integration with cellular metabolic sensors:

    • Exploration of connections between C1QBP and nutrient-sensing pathways (mTOR, AMPK)

    • Investigation of how metabolic stress signals are transmitted to influence C1QBP function

    • Assessment of whether C1QBP directly responds to metabolic cues

These investigations would provide a more comprehensive understanding of how C1QBP functions within the broader network of mitochondrial homeostasis mechanisms.

Product Science Overview

Structure and Composition

C1 is a multimolecular complex composed of three subcomponents:

  1. C1q: This is the recognition protein that binds to antibodies complexed with antigens or directly to pathogen surfaces.
  2. C1r: This is a serine protease that, upon activation, cleaves and activates C1s.
  3. C1s: This is another serine protease that cleaves the next components in the pathway, C4 and C2 .

The C1 complex is structured such that a single C1q molecule is bound to two molecules each of C1r and C1s, forming a C1q(C1r)₂(C1s)₂ complex .

Function and Activation

The primary function of C1 is to initiate the classical pathway of complement activation. This process begins when C1q binds to the Fc region of antibodies (IgG or IgM) that are attached to antigens on the surface of pathogens. This binding induces a conformational change in the C1 complex, leading to the activation of C1r, which in turn activates C1s .

Once activated, C1s cleaves C4 into C4a and C4b. C4b then binds to the pathogen surface and subsequently binds C2, which is cleaved by C1s to form C2a and C2b. The C4bC2a complex, also known as C3 convertase, then cleaves C3 into C3a and C3b, leading to opsonization of the pathogen and further propagation of the complement cascade .

Recombinant Production

Recombinant human Complement Component 1 is produced using recombinant DNA technology, which involves inserting the gene encoding C1 into a suitable expression system, such as bacteria, yeast, or mammalian cells. This allows for the large-scale production of C1 for research and therapeutic purposes .

Clinical Relevance

Deficiencies or dysfunctions in any of the components of C1 can lead to immune system disorders. For example, C1q deficiency is associated with autoimmune diseases such as systemic lupus erythematosus (SLE). Understanding the structure and function of C1 is crucial for developing therapeutic interventions for such conditions .

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 2024 Thebiotek. All Rights Reserved.