CKMT1A catalyzes the reversible transfer of ATP’s γ-phosphate to creatine (Cr), generating phosphocreatine (PCr) and ADP . This reaction is central to the "creatine shuttle," which:
Links mitochondrial ATP production with cytosolic energy utilization .
Supports tissues with fluctuating energy needs (e.g., brain, cardiac muscle) .
A. Cancer Associations
CKMT1A overexpression correlates with aggressive tumors and poor prognosis:
Mechanistically, CKMT1A supports tumorigenesis by:
Cardiomyopathy: Impaired CKMT1A activity disrupts cardiac energetics .
Neurodegeneration: Oxidative damage to CKMT1A exacerbates metabolic failure in Alzheimer’s and Parkinson’s diseases .
Key Findings from Pan-Cancer Analysis
Expression Patterns: Elevated in 18/33 cancer types (e.g., LUAD, UCEC) but reduced in COAD and KIRC.
Immune Microenvironment: Negative correlation with CD8+ T-cell infiltration in multiple cancers.
Pathway Enrichment: Strong association with glycolysis, amino acid biosynthesis, and pentose phosphate pathways.
CKMT1A (mitochondrial creatine kinase 1A) is an enzyme primarily located in the mitochondrial intermembrane space where it catalyzes the reversible transfer of phosphate from phosphocreatine to ADP, generating ATP and creatine. This reaction is a critical component of the creatine-phosphocreatine shuttle system that facilitates energy transport from mitochondria to sites of cellular energy consumption. In research contexts, CKMT1A is studied for its role in maintaining energy balance in cells with high and fluctuating energy demands, including cancer cells that require substantial energy for proliferation and survival under challenging conditions such as hypoxia .
In normal human physiology, CKMT1A shows tissue-specific expression patterns, with highest levels typically observed in tissues with high energy demands like cardiac muscle, skeletal muscle, and brain. The regulation primarily responds to energy metabolism needs and mitochondrial function.
In contrast, cancer tissues show aberrant CKMT1A expression patterns. According to pan-cancer analysis data, CKMT1A is highly expressed in most cancer types including liver, lung, and breast cancers . This upregulation appears to be driven by both genetic factors and microenvironmental conditions:
Hypoxia induces CKMT1A expression through HIF-1α (Hypoxia-Inducible Factor 1-alpha) signaling
Experimental evidence shows that exposing lung cancer cell lines to hypoxic conditions (1% O₂) significantly increases CKMT1A protein expression, with levels peaking at approximately 24 hours of hypoxia
The regulation involves direct binding of HIF-1α to the CKMT1A promoter region, as confirmed by luciferase reporter assays
This differential expression pattern makes CKMT1A a potential biomarker and therapeutic target in oncology.
Several complementary analytical methods can be employed to detect and quantify CKMT1A expression in human samples:
RNA-level detection methods:
Quantitative real-time PCR (qRT-PCR): Provides sensitive measurement of CKMT1A mRNA expression in tissue samples and cell lines
RNA-sequencing (RNA-seq): Enables comprehensive transcriptomic profiling and comparative analysis across multiple cancer types
In situ hybridization: Allows visualization of CKMT1A mRNA within tissue sections while preserving spatial information
Protein-level detection methods:
Western blotting: Quantifies CKMT1A protein levels in cell lysates or tissue homogenates and can detect post-translational modifications
Immunohistochemistry (IHC): Visualizes CKMT1A protein in tissue sections, providing information about expression levels and localization patterns
Immunofluorescence: Offers higher resolution imaging of CKMT1A subcellular localization
Functional assays:
Enzymatic activity assays: Measure the catalytic activity of CKMT1A by detecting the conversion of creatine to phosphocreatine or vice versa
Metabolomic approaches: Gas or liquid chromatography coupled with mass spectrometry can profile metabolites affected by CKMT1A activity
The choice of method should be determined by the specific research question, sample availability, and whether the goal is to assess expression, localization, or functional activity of CKMT1A.
Hypoxia is a prominent feature of solid tumors and significantly influences CKMT1A expression through several molecular mechanisms:
HIF-1α-Dependent Regulation:
Under hypoxic conditions (1% O₂), cancer cells show increased expression of both HIF-1α and CKMT1A proteins
Time-course experiments in H1650 and H1299 lung cancer cell lines demonstrated that CKMT1A protein levels peak at approximately 24 hours of hypoxia exposure
HIF-1α directly binds to specific sequences in the CKMT1A promoter region to enhance transcription, as demonstrated by luciferase reporter assays
When HIF-1α binding sites in the CKMT1A promoter are mutated, hypoxia-induced expression is significantly reduced, though not completely eliminated
HIF-1α-Independent Mechanisms:
The research indicates that even with mutated HIF-1 binding sites, hypoxia still induced some CKMT1A expression, suggesting additional regulatory mechanisms beyond HIF-1α
These may include other transcription factors activated under hypoxia, post-transcriptional regulation of mRNA stability, or alterations in protein translation efficiency under hypoxic stress
Experimental Validation Approaches:
The use of LW6, a specific inhibitor of HIF-1α, confirmed the role of HIF-1α in regulating CKMT1A expression
This approach provides a methodological strategy for researchers studying hypoxia-induced gene expression
Understanding these regulatory mechanisms may provide insights for developing therapeutic strategies targeting hypoxia-induced metabolic adaptations in cancer.
CKMT1A plays several crucial roles in cancer metabolism that support tumor growth and survival:
Metabolic Pathway Involvement:
Enrichment analysis reveals CKMT1A involvement in "Glycolysis/Gluconeogenesis" and broader "metabolic pathways"
As a mitochondrial creatine kinase, CKMT1A facilitates energy transport from mitochondria to cytosolic sites of ATP consumption via the creatine phosphate shuttle
Contribution to Metabolic Reprogramming:
CKMT1A may support the Warburg effect (cancer cells' preference for glycolysis even in the presence of oxygen) by:
Ensuring efficient energy transport from mitochondria to cytosolic sites
Supporting the integrated function of mitochondrial and glycolytic metabolism
Maintaining energy homeostasis when oxidative phosphorylation is compromised
Experimental Evidence:
Knockdown of CKMT1 in lung cancer cell lines (H1650 and H1299) inhibited proliferation, colony formation, and invasion, suggesting that CKMT1A supports the metabolic requirements for cancer cell growth and metastatic potential
Hypoxia Adaptation:
Upregulation of CKMT1A under hypoxic conditions indicates its role in metabolic adaptation to low oxygen environments, which is critical for tumor regions experiencing hypoxia due to rapid growth and insufficient vascularization
This metabolic role makes CKMT1A a potential target for therapeutic strategies aimed at disrupting cancer cell metabolism, particularly in tumors that show high expression of this enzyme.
The relationship between CKMT1A expression and immune cell infiltration represents an emerging area of research with important implications for understanding tumor immunology:
Key Correlations with Immune Components:
CKMT1A expression is negatively correlated with the infiltration of cancer-associated fibroblasts (CAFs) in most tumor types
More significantly, CKMT1A expression shows negative association with CD8+ T-cell infiltration in several tumor types
Since CD8+ T cells are critical for anti-tumor immunity, this negative association suggests that CKMT1A might contribute to immune evasion mechanisms
Potential Mechanisms:
Metabolic competition: CKMT1A's role in cancer cell metabolism might create conditions unfavorable for immune cell function
Signaling effects: Beyond its metabolic role, CKMT1A might influence pathways that affect cytokine production or immune cell recruitment
Indirect effects: CKMT1A-mediated hypoxia adaptation might secondarily influence immune cell infiltration and function
Therapeutic Implications:
The negative correlation with CD8+ T-cell infiltration suggests that targeting CKMT1A might potentially enhance T cell infiltration and function
Combination approaches targeting both CKMT1A and immune checkpoints could be worth investigating
Understanding metabolic interactions between CKMT1A-expressing cancer cells and immune cells could inform the development of metabolism-targeted immunotherapies
This emerging area highlights the complex interplay between cancer metabolism and the immune microenvironment, with potential implications for immunotherapy development.
Genetic alterations of CKMT1A can significantly impact its function and contribute to cancer development:
Types of Genetic Alterations:
Functional Consequences:
Alterations may affect:
Catalytic activity through mutations in the active site
Protein stability and folding
Subcellular localization, particularly mitochondrial targeting
Protein-protein interactions, especially octamer formation and association with mitochondrial membrane proteins
Methodological Approaches for Study:
Site-directed mutagenesis to introduce cancer-specific alterations
Expression of wild-type and mutant CKMT1A in cellular models
Enzymatic activity assays to assess functional consequences
Protein localization studies using fluorescently tagged constructs
CRISPR-Cas9 genome editing to introduce or correct alterations
Implications for Cancer Research:
Specific alterations might serve as biomarkers for prognosis or treatment response
Understanding how alterations affect function can inform targeted therapeutic approaches
CKMT1A alterations might create specific vulnerabilities that could be therapeutically exploited through synthetic lethality approaches
Understanding the implications of CKMT1A genetic alterations contributes to a more complete picture of its role in cancer and may reveal new opportunities for therapeutic intervention.
Selecting appropriate experimental models is crucial for studying CKMT1A function, with each model offering distinct advantages:
Cell Culture Models:
Cancer cell lines with varying CKMT1A expression
3D culture systems (spheroids, organoids)
Genetic Manipulation Approaches:
RNA interference (siRNA, shRNA) has been successfully used to study CKMT1A function in cancer cell lines
CRISPR-Cas9 gene editing allows for complete knockout or introduction of specific mutations
Overexpression systems for studying gain-of-function effects and structure-function relationships
Animal Models:
Xenograft models: Useful for studying CKMT1A's role in tumor growth, metastasis, and microenvironment interactions
Genetically engineered mouse models (GEMMs): Allow study of CKMT1A in de novo tumor development with intact immune system
Patient-derived xenografts (PDXs): Maintain tumor heterogeneity and more closely recapitulate human disease
Ex Vivo Models:
Tissue slice cultures: Maintain original tumor architecture while allowing experimental manipulation
Explant cultures: Enable direct testing of therapies targeting CKMT1A on patient-derived material
Model Selection Considerations:
Match the model to specific aspects of CKMT1A biology being investigated
Consider cancer type specificity and endpoint measures
The most robust approaches often combine multiple model systems to validate findings and address limitations of individual models
Targeting CKMT1A represents a promising therapeutic approach based on its role in cancer metabolism and upregulation in various tumor types:
Gene Silencing Approaches:
RNA interference (RNAi): Studies have used siRNA or shRNA to knockdown CKMT1 expression in cancer cell lines
Experiments demonstrate that CKMT1 knockdown inhibits proliferation, colony formation, and invasion in non-small cell lung cancer cells
CRISPR-Cas9 gene editing provides an alternative approach for more complete knockout
Small Molecule Inhibitors:
Several creatine kinase inhibitors have been developed, though many lack specificity for CKMT1A
Cyclocreatine analogs function as alternative substrates that can disrupt the creatine-phosphocreatine shuttle
Structure-based drug design approaches use the crystal structure of CKMT1A to develop specific inhibitors
Combination Therapy Approaches:
Combining CKMT1A inhibition with hypoxia-targeted therapies may be particularly effective since CKMT1A is upregulated under hypoxic conditions
Potential synergy with glycolysis inhibitors, based on CKMT1A's involvement in glycolysis/gluconeogenesis pathways
Biomarker Development:
Challenges and Considerations:
Ensuring specificity for CKMT1A without affecting other creatine kinase isoforms
Developing effective delivery systems that reach tumor cells
Identifying and addressing potential resistance mechanisms
Understanding impacts on normal tissues with high energy demands
These approaches provide a framework for developing CKMT1A-targeted cancer therapies, though more research is needed to fully evaluate their clinical potential.
CKMT1A is encoded by the CKMT1A gene, which is located on chromosome 15. The gene produces a protein that exists in two isoenzymes: sarcomeric mitochondrial creatine kinase (sMtCK) and ubiquitous mitochondrial creatine kinase (uMtCK). These isoenzymes are encoded by separate genes but share a high degree of homology .
The primary function of CKMT1A is to facilitate the transfer of high-energy phosphate from mitochondria to the cytosolic carrier, creatine. This process is essential for maintaining cellular energy homeostasis, especially in tissues with high and fluctuating energy demands, such as muscles and the brain .
CKMT1A operates in two different oligomeric forms: dimers and octamers. This structural versatility allows it to efficiently manage energy transfer under varying cellular conditions. The enzyme’s activity is crucial for the rapid regeneration of ATP, which is vital for muscle contraction and other energy-intensive cellular processes .
CKMT1A is highly expressed in tissues with high energy demands, including skeletal muscle, heart muscle, and the brain. Its role in energy metabolism makes it a critical player in various physiological processes. Additionally, overexpression of CKMT1A has been observed in several malignant cancers with poor prognosis, suggesting a potential link between CKMT1A activity and cancer cell metabolism .
Mutations or dysregulation of the CKMT1A gene can lead to metabolic disorders and have been associated with conditions such as mitochondrial complex I deficiency and reticular dysgenesis. The enzyme’s role in energy metabolism also makes it a potential target for therapeutic interventions in metabolic diseases and cancer .
Human recombinant CKMT1A is used in various research applications to study its function and regulation. Recombinant proteins are produced through genetic engineering techniques, allowing researchers to investigate the enzyme’s properties and interactions in a controlled environment. This research is essential for developing targeted therapies and understanding the enzyme’s role in health and disease .