Pages
Products

PRKCQ

Official Full Name
protein kinase C theta
Organism
Homo sapiens
GeneID
5588
Background
Protein kinase C (PKC) is a family of serine- and threonine-specific protein kinases that can be activated by calcium and the second messenger diacylglycerol. PKC family members phosphorylate a wide variety of protein targets and are known to be involved in diverse cellular signaling pathways. PKC family members also serve as major receptors for phorbol esters, a class of tumor promoters. Each member of the PKC family has a specific expression profile and is believed to play a distinct role. The protein encoded by this gene is one of the PKC family members. It is a calcium-independent and phospholipid-dependent protein kinase. This kinase is important for T-cell activation. It is required for the activation of the transcription factors NF-kappaB and AP-1, and may link the T cell receptor (TCR) signaling complex to the activation of the transcription factors. [provided by RefSeq, Jul 2008]
Synonyms
PRKCT; nPKC-theta;
Bio Chemical Class
Kinase
Protein Sequence
MSPFLRIGLSNFDCGSCQSCQGEAVNPYCAVLVKEYVESENGQMYIQKKPTMYPPWDSTFDAHINKGRVMQIIVKGKNVDLISETTVELYSLAERCRKNNGKTEIWLELKPQGRMLMNARYFLEMSDTKDMNEFETEGFFALHQRRGAIKQAKVHHVKCHEFTATFFPQPTFCSVCHEFVWGLNKQGYQCRQCNAAIHKKCIDKVIAKCTGSAINSRETMFHKERFKIDMPHRFKVYNYKSPTFCEHCGTLLWGLARQGLKCDACGMNVHHRCQTKVANLCGINQKLMAEALAMIESTQQARCLRDTEQIFREGPVEIGLPCSIKNEARPPCLPTPGKREPQGISWESPLDEVDKMCHLPEPELNKERPSLQIKLKIEDFILHKMLGKGSFGKVFLAEFKKTNQFFAIKALKKDVVLMDDDVECTMVEKRVLSLAWEHPFLTHMFCTFQTKENLFFVMEYLNGGDLMYHIQSCHKFDLSRATFYAAEIILGLQFLHSKGIVYRDLKLDNILLDKDGHIKIADFGMCKENMLGDAKTNTFCGTPDYIAPEILLGQKYNHSVDWWSFGVLLYEMLIGQSPFHGQDEEELFHSIRMDNPFYPRWLEKEAKDLLVKLFVREPEKRLGVRGDIRQHPLFREINWEELERKEIDPPFRPKVKSPFDCSNFDKEFLNEKPRLSFADRALINSMDQNMFRNFSFMNPGMERLIS
Open
Disease
Postoperative inflammation, Transplant rejection
Approved Drug
0
Clinical Trial Drug
1 +
Discontinued Drug
3 +

Cat.No. Product Name Price
Cat.No. Product Name Price
Cat.No. Product Name Price
Cat.No. Product Name Price

Detailed Information

PRKCQ (Protein Kinase C Theta) encodes the PKCθ isoform, a member of the novel, calcium-independent PKC (nPKC) subfamily. The gene is located on human chromosome 10p15-p14, contains 18 exons, and encodes a 706-amino-acid protein with a molecular weight of approximately 82 kDa. PRKCQ protein consists of an N-terminal regulatory domain, including a C1 domain that mediates diacylglycerol (DAG) and phospholipid binding, and a C-terminal catalytic domain with serine/threonine kinase activity. Unlike calcium-dependent PKC isoforms, PRKCQ activation requires only DAG and phosphatidylserine, giving it unique functional advantages in immune cell signaling. PRKCQ expression is highly tissue-specific, enriched in T lymphocytes, skeletal muscle, platelets, and dendritic cells, while being low in most non-immune tissues. Gene regulation studies show that the PRKCQ promoter contains multiple key transcription factor binding sites (e.g., NFAT, AP-1, NF-κB), and its expression is positively regulated by T-cell receptor (TCR) activation. Epigenetic mechanisms, such as DNA methylation and histone modifications, also contribute to tissue-specific expression. Notably, the gene produces a natural antisense transcript, PRKCQ-AS1, a long non-coding RNA (lncRNA) that is dysregulated in diseases such as thyroid cancer and modulates PRKCQ function post-transcriptionally.

Figure 1. Schematic representation of PKCθ structure, showing its regulatory and catalytic domains.Figure 1. Schematic representation of PKCθ structure, showing its regulatory and catalytic domains. (Nicolle A, et al., 2021)

Biological Functions and Immune Regulatory Mechanisms

PRKCQ is a core component of TCR signaling and is indispensable for immune synapse formation and T-cell activation. Upon TCR-CD3/CD28 co-stimulation, PRKCQ is recruited to the immunological synapse center, where it phosphorylates CARD11/CARMA1 at multiple serine residues, activating the canonical NF-κB pathway. Phosphorylated CARD11 associates with lipid rafts, recruiting the BCL10-MALT1 complex and the IKK kinase complex, leading to IκB degradation and NF-κB nuclear translocation. PRKCQ also phosphorylates the mediator STK39/SPAK to activate the JUN pathway independently of the MAPK cascade. In calcium signaling, PRKCQ participates in TCR/CD28-induced NFATC1/NFATC2 transcriptional activation by modulating inositol trisphosphate (IP3) generation and calcium mobilization, influencing T-cell functional differentiation. PRKCQ selectively regulates T-cell subset differentiation, being critical for Th2 and Th17 cell development during inflammation and immune responses, while having minimal impact on Th1 cells. Mechanistically, it drives Th2 differentiation via IL-4 receptor signaling and GATA3 expression, and Th17 differentiation through RORγt activation. PRKCQ also provides survival signals by phosphorylating BAD to inhibit apoptosis and upregulating BCL-XL via NF-κB and JUN pathways, protecting T cells from activation-induced cell death.

In platelets, PRKCQ regulates downstream signals of ITGA2B, CD36, F2R/PAR1, and F2RL3/PAR4, contributing to inside-out signaling and granule secretion. By modulating the dissociation of WASP from WIPF1, PRKCQ influences Arp2/3-mediated actin nucleation and branching, regulating platelet shape change and aggregation. In metabolic regulation, PRKCQ phosphorylates IRS1 to block tyrosine phosphorylation, inhibiting insulin signaling and PI3K/AKT activation, mediating free fatty acid-induced insulin resistance. Recent studies show PRKCQ phosphorylates CCDC88A/GIV to inhibit its guanine nucleotide exchange factor activity, affecting G-protein signaling, and phosphorylates/activates LRRK1 to regulate intracellular transport via RAB protein phosphorylation, expanding the understanding of PRKCQ functions beyond immune cells.

Disease Associations and Pathological Mechanisms

PRKCQ plays a key role in multiple autoimmune disorders. In inflammatory bowel disease (IBD), genome-wide association studies (GWAS) identified PRKCQ as a susceptibility locus (IBD1). T-cell-specific PRKCQ knockout mice resist experimental colitis, associated with reduced Th17 responses and enhanced regulatory T-cell function. In rheumatoid arthritis, PRKCQ promotes synovial T-cell activation and inflammatory cytokine production (e.g., TNF-α, IL-17), contributing to joint inflammation. In systemic lupus erythematosus (SLE), aberrantly elevated PRKCQ activity in T cells leads to hyperactivation of autoreactive B cells. PRKCQ inhibitors have demonstrated efficacy in autoimmune disease models, reducing disease severity and tissue pathology. Due to PRKCQ’s selective effect on effector T cells while sparing regulatory T cells, inhibition offers a relatively targeted immunomodulatory strategy, minimizing broad immunosuppressive side effects.

In cancer immunity, PRKCQ has dual roles: enhancing T-cell antitumor responses while potentially contributing to immune evasion. In solid tumor microenvironments, PRKCQ expression affects tumor-infiltrating lymphocyte (TIL) function. High PRKCQ correlates with enhanced CD8⁺ T-cell effector activity, potentially improving immune checkpoint inhibitor efficacy. However, in certain tumors (e.g., gastrointestinal stromal tumors), PRKCQ signaling may suppress antitumor immunity by promoting immunosuppressive cytokine expression or regulatory T-cell activity. In papillary thyroid carcinoma (PTC), the antisense transcript PRKCQ-AS1 is significantly downregulated. Mechanistically, fasting or fasting-mimicking diets (FMD) upregulate PRKCQ-AS1, which interacts with IGF2BPs to stabilize PRMT7 mRNA, suppressing glycolysis and mitochondrial function. The PRKCQ-AS1/IGF2BPs/PRMT7 axis is a key metabolic reprogramming regulator in PTC, providing a novel intervention target. PRKCQ also influences tumor-associated platelet activation, potentially affecting microthrombosis and stromal remodeling in metastasis.

Targeted Therapy Strategies and Clinical Progress

PRKCQ-targeted strategies focus on small molecule inhibitors, gene silencing, and immunomodulation. Due to its central role in T-cell activation, inhibitors have been widely developed for autoimmune diseases. First-generation ATP-competitive inhibitors (e.g., AEB071) demonstrated efficacy in skin graft rejection and psoriasis but lacked selectivity. Next-generation allosteric inhibitors targeting the unique regulatory domains (e.g., C2 domain) improve isoform specificity. In cancer immunotherapy, combination strategies are emerging: PRKCQ inhibitors with PD-1/PD-L1 blockade can overcome T-cell exhaustion and enhance antitumor immunity. In thyroid cancer, targeting the PRKCQ-AS1/PRMT7 axis offers a novel approach to tumor metabolic intervention. CRISPR/Cas9-mediated PRKCQ editing in CAR-T cells can improve T-cell persistence and antitumor activity. In platelets, PRKCQ inhibition may reduce tumor-associated thrombotic events, providing additional clinical benefits for advanced cancer patients.

Reference

  1. Nicolle A, Zhang Y, Belguise K. The Emerging Function of PKCtheta in Cancer. Biomolecules. 2021 Feb 5;11(2):221.

  2. Zaid Y, Senhaji N, Naya A, et al. PKCs in thrombus formation. Pathol Biol (Paris). 2015 Dec;63(6):268-71.

Quick Inquiry

Interested in learning more?

Contact us today for a free consultation with the scientific team and discover how Creative Biogene can be a valuable resource and partner for your organization.

Request a quote today!

Inquiry