Gene Location: The CNN1 gene is located at 19p13.2-p13.1 on the human chromosome and consists of 7 exons encoding a 297-amino-acid protein .
Protein Properties:
Smooth Muscle Regulation: CNN1 inhibits actin-activated myosin ATPase, stabilizing the actin cytoskeleton and fine-tuning contractility in mature smooth muscle cells .
Developmental Role: Expression increases postnatally, correlating with smooth muscle differentiation .
Vascular Maturation: Loss of CNN1 in mice leads to impaired blood vessel maturation, reduced tumor vascular stability, and enhanced susceptibility to antiangiogenic therapies .
Cancer Microenvironment:
Therapeutic Potential:
Transcriptional Control: A single intronic CArG box (C2) is essential for smooth muscle-specific CNN1 expression, as shown in transgenic mouse models .
Post-Translational Modifications: Phosphorylation regulates its actin-binding activity and cellular localization .
CCN1 (also known as CYR61) is a matricellular protein that plays significant roles in cancer development and progression. In terms of nomenclature, it's important to note that in some research contexts, CNN1 specifically refers to 1,4-Naphthoquinone, a compound studied for its anticancer properties, while CCN1 refers to the gene/protein involved in cancer signaling pathways.
CCN1 has been found to both promote and suppress cancer growth depending on tumor cell context. For example, in non-small cell lung cancer (NSCLC), CCN1 has demonstrated context-dependent effects that can either promote or inhibit cancer growth and progression . In prostate cancer, CCN1 is detected in human prostate cancer cells 2-6 hours after the addition of lysophosphatidic acid (LPA), suggesting its involvement in LPA signaling pathways .
Research on CNN1/CCN1 spans multiple cancer types with varying incidence rates. According to epidemiological data, cancer frequencies where CNN1/CCN1 has been studied include:
Cancer Type (Males) | Frequency | Cancer Type (Females) | Frequency |
---|---|---|---|
Lung/bronchus/trachea | 1,368,500 | Breast | 2,088,800 |
Prostate | 1,276,100 | Colon/rectum/anus | 823,300 |
Colon/rectum/anus | 1,026,200 | Lung/bronchus/trachea | 725,400 |
Leukemia | 249,500 | Ovary | 295,400 |
CCN1 has been specifically studied in:
Prostate cancer, where it plays a role in lysophosphatidic acid signaling
Colorectal cancer, where it has been targeted by microRNA to inhibit tumor growth
Pancreatic cancer, where it promotes tumorigenicity through Rac1/Akt/NF-κB signaling
Leukemia, where the compound 1,4-Naphthoquinone (CNN1) has shown promising anticancer activity
Standard methodological approaches for studying CNN1/CCN1 in cancer research include:
Cell Culture: Using established cancer cell lines such as PC-3 (prostate cancer), K-562 and FEPS (leukemia) cultivated at 37°C with 5% CO₂ in appropriate media supplemented with fetal bovine serum and antibiotics .
Gene Expression Analysis: Employing RT-qPCR to evaluate gene expression using appropriate probes and primers. For example, H2AFX gene expression can be measured using TaqMan® Gene Expression Assays with Beta-Actin (ACTB) as an endogenous control .
Protein Detection: Performing immunoblotting to detect and quantify CCN1 protein expression in cancer cells under various treatment conditions .
Cell Viability Assays: Determining cytotoxicity using methods such as MTT assays to evaluate the effects of CNN1 on cancer cell proliferation .
CNN1 induces apoptosis through multiple molecular mechanisms:
DNA Damage and Fragmentation: CNN1 causes significant DNA damage and fragmentation in leukemia cell lines, leading to cell cycle arrest at the G2/M phase .
Mitochondrial Membrane Depolarization: Research demonstrates that CNN1 induces mitochondrial membrane depolarization in leukemia cell lines, a crucial step in the intrinsic apoptotic pathway .
H2AFX Upregulation: CNN1 treatment (0.1 μM) significantly increases H2AFX gene expression in K-562 and FEPS leukemia cell lines. H2AFX is involved in DNA damage response and repair mechanisms, and its upregulation indicates the presence of double-strand DNA breaks .
Apoptotic Pathway Activation: Flow cytometry analysis using Annexin V®488 and propidium iodide has confirmed that CNN1 exposure (0.1 μM) causes a significant increase in early apoptotic cells (Annexin-V positive/PI negative) and late apoptotic cells (Annexin V-positive/PI-positive) in leukemia cell lines compared to negative controls .
CCN1 has significant implications for drug resistance in cancer treatment:
Multidrug Resistant (MDR) Leukemia Cells: Studies show that 1,4-Naphthoquinone (CNN1) demonstrates potent cytotoxic activity against multidrug resistant leukemia cell lines. CNN1 has an IC₅₀ value of 0.90 μM (CI 95% 0.34–1.27) against K-562-Lucena-1 and 0.60 μM (CI 95% 0.48–0.80) against FEPS cell lines .
Comparison with Standard Treatments: When compared to standard treatments like Imatinib Mesylate (IM), CNN1 requires significantly lower concentrations to achieve cytotoxic effects in MDR cell lines. IM showed an IC₅₀ of 4.97 μM (CI 95% 3.69–5.70) for K-562-Lucena-1 and 9.66 μM (CI 95% 8.45–11.1) for FEPS - concentrations much higher than those required for CNN1 .
Potential for Overcoming Treatment Resistance: The efficacy of CNN1 against multidrug resistant leukemia cells suggests its potential as a lead compound for developing new treatments for chronic myeloid leukemia (CML), particularly in cases where resistance to standard therapies has developed .
CCN1 participates in multiple signaling networks that influence cancer progression:
Lysophosphatidic Acid (LPA) Signaling: In prostate cancer cells, CCN1 is induced 2-6 hours after LPA exposure, suggesting its role as a downstream effector in LPA signaling pathways .
MACC1 and TSP1 Regulation: Research has identified interactions between CCN1 and MACC1 (Metastasis Associated in Colon Cancer 1), a key regulator of HGF-MET signaling that predicts colon cancer metastasis. CCN1 knockdown affects LPA-induced upregulation of MACC1 and TSP1 .
Rac1/Akt/NF-κB Pathway: In pancreatic cancer, CCN1 promotes tumorigenicity through the Rac1/Akt/NF-κB signaling pathway, contributing to cancer progression .
EMT Regulation: CCN1 has been implicated in epithelial-mesenchymal transition (EMT) through interactions with EMT regulators such as SNAI1, which is crucial for cancer metastasis .
When designing experiments to study CNN1 in drug-resistant cancer models, researchers should consider the following methodological approaches:
Cell Line Selection: Use paired sensitive and resistant cell lines such as K-562 (sensitive) and K-562-Lucena-1 or FEPS (resistant) leukemia cells to properly evaluate differential responses .
Concentration Determination: Establish appropriate concentrations based on IC₅₀ values. For CNN1, previous research has used 0.1 μM for mechanistic studies based on previously determined IC₅₀ values (0.60-1.12 μM range depending on cell line) .
Time Course Experiments: Conduct time-dependent experiments to capture both early and late effects. For example, gene expression studies are typically performed after 18 hours of treatment (a time that does not significantly modify cellular viability), while apoptosis assays may require longer incubation periods .
Comprehensive Cell Death Analysis: Employ multiple complementary assays including:
Include Appropriate Controls: Use established drugs like Imatinib Mesylate as positive controls and vehicle controls (DMSO) as negative controls to validate experimental findings .
The scientific literature contains contradictory findings regarding CNN1/CCN1's role in cancer, with reports indicating both pro-tumorigenic and anti-tumorigenic effects. To address these contradictions, researchers should:
Context-Specific Analysis: Acknowledge that CCN1 can both promote and suppress cancer growth depending on the tumor cell context. For instance, in NSCLC, CCN1 has demonstrated dual roles .
Standardize Experimental Protocols: Poor reproducibility of results and failure to translate findings to clinical trials may stem from inadequate experimental design. Follow guidelines like ARRIVE (Animal Research: Reporting of In Vivo Experiments) to improve standardization .
Pre-registration: Consider pre-registering experimental protocols to reduce reporting bias and increase transparency in research .
Address Biases: Implement strategies to reduce various biases:
Cross-Validate Findings: Use multiple cell lines, techniques, and model systems to verify results and determine if contradictions are due to biological context or methodological differences .
For optimal detection and quantification of CCN1 in human cancer tissues, researchers should consider these methodological approaches:
mRNA Expression Analysis:
RT-qPCR using validated primers and probes (e.g., TaqMan® Gene Expression Assays)
RNA isolation using TRIzol Reagent® followed by quality assessment via Nanodrop
cDNA synthesis using HighCapacity cDNA Reverse Transcriptase kit
Data analysis using the 2^(-ΔΔCT) method with appropriate endogenous controls (e.g., ACTB, ABL)
Protein Detection:
Functional Assays:
Advanced Techniques:
CNN1 (1,4-Naphthoquinone) shows promising potential as a therapeutic target for multidrug-resistant leukemia based on several lines of evidence:
Superior Efficacy Against Resistant Cells: CNN1 demonstrates significantly lower IC₅₀ values against multidrug-resistant leukemia cell lines compared to standard treatments. Against K-562-Lucena-1, CNN1 has an IC₅₀ of 0.90 μM compared to Imatinib Mesylate's 4.97 μM. For FEPS cells, CNN1's IC₅₀ is 0.60 μM versus Imatinib's 9.66 μM .
Multiple Mechanism of Action: CNN1 induces cell death through multiple mechanisms including:
Selective Cytotoxicity: Importantly, CNN1 shows no genotoxicity on peripheral blood mononuclear cells (PBMC), suggesting potential selectivity for cancer cells over normal cells .
Lead Compound Potential: Research indicates that CNN1 has promising anticancer activity and potential as a lead compound for developing new treatments for chronic myeloid leukemia (CML), particularly for overcoming drug resistance .
While the search results don't provide direct correlations between CCN1 expression and clinical outcomes across all cancer types, several important relationships can be inferred:
Metastasis Prediction: CCN1 interacts with MACC1, which has been established as a predictor of colon cancer metastasis. MACC1 is a key regulator of HGF-MET signaling, and circulating MACC1 transcripts serve as diagnostic and prognostic biomarkers in various cancers including gastric cancer and non-small cell lung cancer .
Signaling Pathway Involvement: CCN1 promotes tumorigenicity through the Rac1/Akt/NF-κB signaling pathway in pancreatic cancer, suggesting its potential role in aggressive disease behavior .
Drug Resistance Implications: The interaction of CCN1 with multidrug resistance mechanisms suggests that its expression may correlate with treatment response and outcomes in certain contexts .
Context-Dependent Effects: Since CCN1 has been reported to have both pro-tumorigenic and anti-tumorigenic effects depending on cellular context, its correlation with clinical outcomes likely varies by cancer type and stage .
Developing CNN1-targeted therapies faces several significant challenges that researchers must address:
Dual Role Complexity: CCN1 exhibits both pro-tumorigenic and anti-tumorigenic effects depending on tumor cell context, complicating therapeutic targeting strategies. A targeted approach might benefit some cancer types while potentially harming others .
Resistance Mechanisms: While CNN1 (1,4-Naphthoquinone) shows efficacy against currently resistant cancer cells, the potential for developing new resistance mechanisms against CNN1-targeted therapies must be considered and investigated .
Delivery and Specificity: Ensuring selective delivery to tumor cells while minimizing effects on normal cells remains challenging, though preliminary data showing no genotoxicity on PBMCs is promising .
Reproducibility Issues: The scientific literature of laboratory animal research shows poor reproducibility of results and failure to translate to clinical trials in humans. This suggests that experimental design and conduct need refinement to improve clinical translation potential .
Signaling Network Complexity: CCN1 interacts with multiple signaling pathways including MACC1, TSP1, and the Rac1/Akt/NF-κB pathway. This complex network of interactions presents challenges for predicting therapeutic effects and potential compensatory mechanisms .
Several emerging technologies hold promise for advancing CNN1/CCN1 cancer research:
Global Proteomics Analysis: Advanced proteomics using LC-MS has already been applied to study lysophosphatidic acid signaling and the role of CCN1 in prostate cancer cells. Expanding this approach could identify novel protein interactions and signaling networks .
CRISPR-Cas9 Gene Editing: More precise genetic manipulation of CCN1 and related genes could help clarify context-dependent functions and identify synthetic lethal interactions that could be therapeutically exploited.
Patient-Derived Organoids and Xenografts: These models provide more physiologically relevant systems than traditional cell lines for studying CNN1/CCN1 function and therapeutic responses in personalized medicine contexts.
Single-Cell Analysis: Single-cell RNA sequencing and proteomics could reveal heterogeneity in CCN1 expression and function within tumors, potentially explaining some contradictory findings in the literature.
Computational Biology and AI: Machine learning approaches could help integrate diverse datasets (genomic, transcriptomic, proteomic) to build predictive models of CNN1/CCN1 function across cancer types and identify biomarkers of response to CNN1-targeted therapies.
Strategic design of combination therapies incorporating CNN1 could include:
Sequential Therapy Approaches: Using CNN1 after conventional therapy failure to target resistant cell populations. Research shows CNN1 has superior efficacy against Imatinib-resistant leukemia cells .
Simultaneous Targeting of Multiple Pathways: Combining CNN1 with inhibitors of complementary pathways. For example, since CNN1 affects H2AFX expression, combining it with other DNA damage response modulators might enhance efficacy .
Exploiting Synthetic Lethality: Identifying genes/pathways that, when inhibited alongside CNN1 treatment, produce synergistic cytotoxicity specifically in cancer cells.
Resistance Mechanism Targeting: Combining CNN1 with inhibitors of drug efflux pumps (e.g., ABCB1) or other resistance mechanisms to prevent development of resistance to CNN1 itself .
Immunotherapy Combinations: Investigating whether CNN1-induced cancer cell death promotes immunogenic responses that could be enhanced by immune checkpoint inhibitors.
Although the search results don't directly address CNN1/CCN1's role in cancer stem cells, several findings suggest potential connections:
Metastasis Regulation: CCN1 interactions with MACC1, a key predictor of colon cancer metastasis, suggest involvement in metastatic processes. MACC1 regulates HGF-MET signaling, which is crucial for cancer cell migration and invasion .
EMT Pathway Involvement: CCN1's interaction with EMT regulators like SNAI1 suggests a potential role in the epithelial-mesenchymal transition, a process critical for both metastasis and cancer stem cell generation .
Cancer Stemness Pathways: CCN1 promotes tumorigenicity through the Rac1/Akt/NF-κB signaling pathway in pancreatic cancer . These pathways are also implicated in cancer stem cell maintenance and self-renewal.
Cell Cycle Regulation: CNN1's ability to induce cell cycle arrest at G2/M phase may differentially affect cancer stem cells, which often have distinct cell cycle regulation compared to bulk tumor cells.
Future research should specifically investigate CNN1/CCN1's effects on cancer stem cell populations, including their self-renewal capacity, differentiation potential, and contribution to metastatic dissemination and therapy resistance.
The CNN1 gene is located on chromosome 19 (19p13.2-p13.1) and contains 7 exons . The human calponin 1 protein consists of 297 amino acids, with a molecular weight of approximately 33.2 kDa and an isoelectric point of 9.1 . This protein is also referred to as basic calponin due to its isoelectric point .
Calponin 1 is a thin filament-associated protein that binds to actin, calmodulin, troponin C, and tropomyosin . Its primary function is to inhibit the actomyosin Mg-ATPase activity, which is essential for muscle contraction . By binding to actin, calponin 1 modulates the interaction between actin and myosin, thereby regulating smooth muscle contraction .
The expression of CNN1 is specific to differentiated mature smooth muscle cells . It is up-regulated in smooth muscle tissues during postnatal development, with a higher content in phasic smooth muscle of the digestive tract . This suggests that calponin 1 plays a significant role in the contractile functions of smooth muscle cells .
Given its role in smooth muscle contraction, calponin 1 is implicated in various physiological and pathological processes. Understanding its function and regulation can provide insights into smooth muscle-related disorders and potential therapeutic targets.
Recombinant human calponin 1 is produced through recombinant DNA technology, allowing for the study of its structure, function, and interactions in a controlled environment. This recombinant protein is valuable for research in muscle physiology and related fields.