Research Article | | Peer-Reviewed

Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines

Received: 17 April 2025     Accepted: 29 April 2025     Published: 29 May 2025
Views:       Downloads:
Abstract

Microsatellite instability (MSI) is a hallmark of mismatch repair (MMR) deficiency and characterizes a distinct subset of colorectal cancers (CRC). In parallel, telomere length dynamics have emerged as important contributors to genomic stability and tumorigenesis. However, the relationship between MSI status, MMR protein expression, and telomere maintenance remains poorly defined. This study aimed to investigate the association between MSI status and telomere length in CRC cell lines and to evaluate the expression of key MMR proteins (MLH1, MSH2, MSH6, PMS2) to elucidate molecular differences between MSI and microsatellite stable (MSS) phenotypes. A panel of CRC cell lines with known MSI and MSS statuses was used. Telomere length was quantified using real-time quantitative PCR (qPCR) based on the T/S ratio method. MSI status was confirmed via PCR using mononucleotide repeat markers. Western blotting was performed to assess protein expressions of MLH1, MSH2, MSH6, and PMS2. β-actin served as a loading control. qPCR analysis revealed that MSI cell lines exhibited significantly longer telomeres compared to MSS lines (P < 0.05). Western blot results showed complete loss of MSH6 protein expression and reduction in expression levels of PMS2 and MSH2 in MSI cell lines, confirming MMR deficiency. In contrast, MSS cell lines maintained normal expression of all tested MMR proteins. These findings suggest a link between defective MMR function and altered telomere dynamics in MSI-CRC. MSI CRC cell lines exhibit telomere elongation and loss of key MMR proteins, highlighting distinct molecular features compared to MSS counterparts. These insights may inform future strategies for personalized CRC diagnostics and therapeutics, particularly in the context of telomere-targeted or immunomodulatory treatments.

Published in Cancer Research Journal (Volume 13, Issue 2)
DOI 10.11648/j.crj.20251302.11
Page(s) 23-29
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2025. Published by Science Publishing Group

Keywords

Colorectal Cancer, Telomere Length, Microsatellite Instability, Mismatch Repair Pathway, MMqPCR, Western Blotting, HRM Analysis

1. Introduction
Colorectal cancer (CRC) ranks as the third most common cancer globally and is the second leading cause of cancer-related mortality, with an estimated 1.9 million new cases and 935,000 deaths in 2020 . Although CRC primarily affects individuals over the age of 50, an alarming rise in early-onset CRC has been noted in younger populations. The disease generally evolves from benign adenomatous or serrated polyps, progressing via a multi-step carcinogenic sequence driven by cumulative genetic and epigenetic alterations. The adenoma-carcinoma sequence, first conceptualized by Fearon and Vogelstein, continues to serve as a foundational framework for elucidating CRC pathogenesis .
A hallmark of CRC is genomic instability, which manifests through three major and often overlapping mechanisms: chromosomal instability (CIN), microsatellite instability (MSI), and the CpG island methylator phenotype (CIMP) . These molecular pathways not only fuel tumor heterogeneity and progression but also significantly influence therapeutic resistance .
1.1. Microsatellite Instability in CRC
Microsatellite instability (MSI) is defined by the accumulation of length variations in short tandem repeats due to defective DNA mismatch repair (MMR) . First reported in CRC in the early 1990s, MSI is present in approximately 15-20% of sporadic CRCs and more than 95% of Lynch syndrome-associated tumors . MSI is classified into three categories—MSI-high (MSI-H), MSI-low (MSI-L), and microsatellite stable (MSS)—based on the frequency and pattern of instability . MSI-H tumors are associated with high tumor mutational burden (TMB), enhanced neoantigen presentation, better prognosis, and improved response to immune checkpoint inhibitors .
1.2. Mismatch Repair (MMR) Pathway and CRC
The MMR pathway is vital for maintaining genomic fidelity by correcting base-base mismatches and insertion-deletion loops during DNA replication. Core MMR proteins include MLH1, PMS2, MSH2, and MSH6, which operate as heterodimers (e.g., MutSα, MutLα) to recognize and repair replication errors . In sporadic CRC, MSI is frequently attributed to the epigenetic silencing of the MLH1 gene through promoter hypermethylation . Dysfunctional MMR leads to an accumulation of somatic mutations, thereby disrupting oncogenes and tumor suppressor pathways .
1.3. Telomere Dysfunction and Its Link to MMR
Telomeres, composed of tandem TTAGGG repeats, protect chromosomal termini from degradation and fusion. Progressive telomere shortening, a natural consequence of cellular replication, can induce senescence or apoptosis . In CRC, telomere attrition contributes to chromosomal instability and tumor evolution. MSI-H tumors have been shown to possess shorter telomeres than MSS counterparts, suggesting a correlation between MMR deficiency and telomere dynamics . Moreover, loss of MMR proteins such as MLH1 and MSH2 promotes homologous recombination at telomeres, potentially enabling the alternative lengthening of telomeres (ALT) pathway .
1.4. Telomerase and ALT in CRC
Telomerase, a ribonucleoprotein complex composed of the catalytic subunit hTERT and the RNA component hTERC, is reactivated in nearly 90% of human cancers, including CRC . In contrast, a subset of tumors maintains telomeres via the telomerase-independent ALT mechanism, which relies on homologous recombination . Intriguingly, MSI-H CRCs frequently exhibit diminished hTERT expression, indicating potential reliance on ALT for telomere maintenance . Therefore, exploring the interactions between MMR deficiency and telomere regulation may offer novel insights into CRC biology and therapeutic targets.
2. Materials and Methods
2.1. Cell Culture
Human CRC cell lines CACO2 (MSS, RRID: CVCL_0025) and HCT15 (MSI, RRID: CVCL_0292) were procured and cultured under standard conditions at 37°C in a humidified 5% CO₂ atmosphere. CACO2 cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 20% fetal bovine serum (FBS), while HCT15 cells were grown in RPMI 1640 medium with 10% FBS. All protocols adhered to ATCC guidelines .
2.2. DNA and Protein Extraction
Cells were harvested at ~90% confluence using trypsin-EDTA. Genomic DNA was extracted by phenol-chloroform method , verified for integrity via 1% agarose gel electrophoresis, and quantified using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific). Protein extraction was performed using RIPA buffer, followed by quantification and analysis according to standard protocols .
2.3. Telomere Length Measurement
Telomere length was assessed using monochrome multiplex quantitative real-time PCR (MMqPCR) as described by Cawthon and refined by Hosen et al . Primer sequences were:
Telomere primers:
TelG: ACACTAAGGTTTGGGTTTGGGTTTGGGTTTGGGTTAGTGT
TelC: TGTTAGGTATCCCTATCCCTATCCCTATCCCTATCCCTAACA
Single-copy gene (albumin) primers:
AlbUgcr2: CGGCGGCGGGCGGCGCGGGCTGGGCGGAAATGCTGCACACAGAATCCTTG
AlbDgcr2: GCCCGGCCCGCCGCGCCCGTCCCGCCGGAAAAGCATGGTCGCCTGTT
Reactions were performed using the QuantStudio 6 Pro Real-Time PCR system. T/S ratios (telomere repeat copy number to single-copy gene number) were calculated from Ct values to estimate relative telomere length .
2.4. Microsatellite Instability Detection
MSI status was determined using high-resolution melting (HRM) analysis adapted from Raji et al . The pentaplex panel included five mononucleotide markers :
NR-27: F: AACCATGCTTGCAAACCACT | R: CGATAATACTAGCAATGACC
NR-21: F: GAGTCGCTGGCACAGTTCTA | R: CTGGTCACTCGCGTTTACAA
NR-24: F: GCTGAATTTTACCTCCTGAC | R: ATTGTGCCATTGCATTCCAA
BAT-25: F: TACCAGGTGGCAAAGGGCA | R: TCTGCATTTTAACTATGGCTC
BAT-26: F: CTGCGGTAATCAAGTTTTTAG | R: AACCATTCAACATTTTTAACCC
qPCR amplification was followed by HRM profiling to detect heteroduplexes indicative of MSI .
2.5. Western Blotting
Protein concentrations were determined using the BCA assay. Lysates were electrophoresed on SDS-PAGE and transferred to membranes via semi-dry transfer. Membranes were blocked with 5% BSA or milk, then probed overnight at 4°C with primary antibodies against hMLH1, hMSH2, hMSH6, hPMS2, and β-actin . HRP-conjugated secondary antibodies were used, and bands were visualized using DAB or luminol substrate. Images were captured using ImageQuant software.
3. Results
3.1. DNA Quality Assessment and Quantification
For the MSI HCT15 cell line, the DNA showed high purity with A260/A280 ratios ranging between 1.80 and 1.90, and DNA yields were consistently above 100 ng/µL. Similarly, MSS CACO2 DNA samples demonstrated excellent integrity, with no smearing observed on gels and comparable purity metrics. These results confirmed that the genomic DNA was of suitable quality for downstream applications such as qPCR and HRM analyses.
3.2. Telomere Length Estimation
MMqPCR analysis revealed a significantly reduced telomere length (T/S ratio) in MSI-H HCT15 cells compared to MSS CACO2 cells (p < 0.05). The mean T/S ratio in HCT15 was 0.71 ± 0.08, whereas CACO2 cells exhibited a higher T/S ratio of 1.26 ± 0.11 (Figure 1). This indicates accelerated telomere shortening in MSI-H cells, aligning with the hypothesis that MMR deficiency may contribute to telomere erosion.
Figure 1. Telomere length Analysis.
Table 1. Telomere Length Estimation in CRC Cell Lines via T/S Ratio Relative telomere length (T/S ratio) in HCT15 (MSI) and CACO2 (MSS) cell lines was determined using the ΔCt method (CtT - CtS) and calculated as 2ΔCt. Ct values were obtained via qPCR using TelG/C (telomeric) and AlbU/D (single-copy) primers. SD reflects variability across replicates.

Cell Line

CtT (TelG/C)

CtS (AlbU/D)

T/S Ratio (2ΔCt)

SD

HCT15 (MSI)

20.13

34.61

22,862.18

1.28

CACO2 (MSS)

20.60

38.72

284,641.89

1.63

3.3. Microsatellite Instability Analysis
HRM profiles generated from the five-marker pentaplex assay (BAT-25, BAT-26, NR-21, NR-24, and NR-27) showed significant heteroduplex formation and distinct melting curve shifts in HCT15, confirming its MSI-H status. In contrast, CACO2 cells showed stable melting profiles across all five markers, consistent with an MSS phenotype (Figure 2). These findings validate the suitability of HRM as a rapid, dye-based, high-throughput method for MSI detection in CRC cell lines.
Figure 2. Microsatellite Instabilty Analysis.
Table 2. MSI Marker Amplification in CRC Cell Lines by HRM-qPCR Mean Ct values and SDs for five MSI markers analyzed via HRM-qPCR in HCT15 (MSI) and CACO2 (MSS) cells. Ct shifts indicate differential amplification efficiency related to MSI status.

MSI Marker

HCT15 (MSI) - Mean Ct

SD

CACO2 (MSS) - Mean Ct

SD

NR27

26.37

0.77

30.80

1.18

NR21

45.97

0.46

25.42

0.08

NR24

47.58

0.75

27.87

0.35

BAT25

41.05

3.95

25.62

0.37

BAT26

40.00

0.00

39.76

0.35

3.4. Western Blotting of MMR Proteins
Western blot analysis revealed a complete loss of MSH6 protein expression in the MSI-H HCT15 cell line, whereas MSH6 was robustly expressed in the MSS CACO2 cells. The expression levels of MSH2 and PMS2 were also significantly reduced in HCT15 cells, indicating downstream destabilization of the MMR heterodimers due to the absence of MSH6. Interestingly, MLH1 expression showed a 1.7-fold increase in the MSI cell line, suggesting a potential compensatory response. β-actin expression remained consistent across both cell lines, confirming equal protein loading (Figure 3).
Table 3. Protein Quantification in CRC Cell Lysates via BCA Assay Total protein concentrations (μg/mL) in CACO2 and HCT15 lysates were determined using the BCA assay. Absorbance at 562 nm was blank-corrected and values calculated using a standard curve. Measurements were performed in duplicate.

Sample

Absorbance (562 nm)

Blank

Protein Concentration (μg/mL)

CACO2_P1

1.2322

0.9717

3190

CACO2_P2

1.3291

1.0686

3513

HCT15_P1

1.5952

1.3347

4400

HCT15_P2

1.6422

1.3817

4556.5

Figure 3. MMR Protein expression analysis.
4. Discussion
Colorectal cancer (CRC) pathogenesis involves diverse molecular subtypes, with microsatellite instability (MSI) and telomere dysfunction being pivotal contributors. This study demonstrates a significant correlation between MSI status and telomere shortening, coupled with downregulation of DNA mismatch repair (MMR) proteins, suggesting a mechanistic interplay between genomic instability and telomere biology in CRC. Our findings contribute to a growing body of evidence implicating the involvement of MSI and telomere dysfunction in CRC development.
MMR Deficiency and MSI in CRC: Our HRM-based MSI assay effectively differentiated between MSI-H (HCT15) and MSS (CACO2) phenotypes. This was in agreement with previous reports that highlight the critical role of MMR proteins in maintaining genomic stability . The analysis revealed that MSH6 was not expressed in the MSI cell line, and there was downregulation of MSH2 and PMS2. Conversely, MLH1 expression was upregulated. These results suggest that the MSI cell line's heightened mutational burden and disrupted telomere maintenance may be due to the compromised functionality of specific MMR proteins. In particular, this study highlights that the absence of MSH6 protein expression is a significant contributor to microsatellite instability (MSI) in the CRC cell line. This deficiency not only induces MSI due to the inability to repair MMR mismatches but also appears to play a crucial role in the accelerated telomere shortening observed in MSI CRC cell lines. The simultaneous loss or reduced expression of MSH6, PMS2, and MSH2 likely compromises DNA repair efficiency, thereby exacerbating genomic instability. The elevated MLH1 expression in the MSI cell line may indicate a compensatory response to the loss of other MMR proteins, although its precise role in this context requires further investigation. The general classification and characterization of MSI across various types of cancer have been systematically outlined in previous studies, and de la Chapelle (2003) provided the foundational understanding of MSI’s role in CRC .
Telomere Shortening in MSI-H CRC: Our data on telomere length reveals a significant reduction in telomere size in HCT15 cells, in agreement with previous studies suggesting that MSI-H tumors tend to harbor shorter telomeres compared to their MSS counterparts . This reduction in telomere length could be attributed to the loss of MMR function, which normally prevents aberrant homologous recombination at telomeres. The absence of functional MMR proteins likely leads to the destabilization of telomeres, promoting telomere attrition in MSI-H tumors. Moreover, the observed reduction in telomere length aligns with the hypothesis that MSI-H tumors may rely on alternative telomere lengthening (ALT) mechanisms in the absence of telomerase reactivation, a phenomenon well-documented in MMR-deficient cancers . These findings align with the systematic analysis of telomere lengths and somatic alterations in CRC, which reveal a close association between telomere attrition and tumor progression .
Therapeutic and Diagnostic Implications: The findings of this study support the growing paradigm that MSI status and telomere dynamics can be leveraged for CRC prognosis and therapy. MSI-H tumors, due to their high neo-antigen burden, are known to respond favorably to immune checkpoint blockade therapies . Understanding the interplay between telomere dysfunction and MSI may open new avenues for targeted therapeutic strategies. For example, telomere-based diagnostics could be utilized to stratify patients for telomerase-targeted or ALT-inhibitory therapies, both of which may hold promises for improving clinical outcomes in MSI-H CRC patients.
Limitations and Future Directions: While this study provides robust in vitro data, it is limited to only two cell lines, which may not fully capture the complexity of CRC heterogeneity. Future work should aim to include primary tumor samples from MSI-H and MSS CRC patients to validate these observations and assess their clinical relevance. Additionally, telomere length assays, telomerase activity assays, and ALT pathway analyses could provide further insights into the telomere maintenance strategies employed by different CRC subtypes. Expanding the study to encompass a broader range of CRC models will help to define the full extent of telomere dysfunction and its therapeutic implications in MSI-associated cancers.
Abbreviations

CRC

Colorectal Cancer

MSI

Microsatellite Instability

MSI-H

Microsatellite Instability - High

MMR

Mismatch Repair

MSS

Microsatellite Stable

CIN

Chromosomal Instability

CIMP

CpG island Methylator Phenotype

ALT

Alternative Lengthening of Telomeres

MMqPCR

Monochrome Multiplex Quantitative Real-Time PCR

T/S Ratio

Telomere Repeat Copy Number to Single-Copy Gene Number

HRM

High-Resolution Melting

Acknowledgments
The authors gratefully acknowledge the Manipal Academy of Higher Education (MAHE) and the Manipal School of Life Sciences for providing essential infrastructure and institutional support throughout the course of this research. Special thanks are extended to the TIFAC-CORE in Pharmacogenomics, MAHE, for facilitating access to advanced research facilities.
Author Contributions
Rajasekhar Moka: Conceptualization, Writing-Review and Editing, Supervision.
Shreya Upadhya: Investigation, Methodology, Visualization, Validation, Formal Analysis, Writing-Original Draft.
Conflicts of Interest
The authors declare no conflicts of interest.
References
[1] Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022; 72: 7-33.
[2] Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990; 61(5): 759-767.
[3] Yamagishi H, Kuroda H, Imai Y, Hiraishi H. Molecular pathogenesis of sporadic colorectal cancers. Chin J Cancer. 2016; 35: 4.
[4] Boland CR, Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010; 138(6): 2073-2087. e3.
[5] Umar A, Boland CR, Terdiman JP, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004; 96(4): 261-268.
[6] Dey A, Mitra A, Pathak S, Prasad S, Zhang AS, Zhang H, et al. Recent advancements, limitations, and future perspectives of the use of personalized medicine in treatment of colon cancer. Technol Cancer Res Treat. 2023; 22: 15330338231178404.
[7] Li GM. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008; 18(1): 85-98.
[8] Lao VV, Grady WM. Epigenetics and colorectal cancer. Nat Rev Gastroenterol Hepatol. 2011; 8(12): 686-700.
[9] Hause RJ, Pritchard CC, Shendure J, Salipante SJ. Classification and characterization of microsatellite instability across 18 cancer types. Nat Med. 2016; 22(11): 1342-1350.
[10] de Lange T. Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev. 2018; 32(7-8): 527-540.
[11] Takagi S, Kinouchi Y, Hiwatashi N, et al. Relationship between microsatellite instability and telomere shortening in colorectal cancer. Jpn J Cancer Res. 2000; 1(4): 399-407.
[12] Bozzao C, Lastella P, Ponz de Leon M, Pedroni M, Di Gregorio C, D’Ovidio FD, et al. Analysis of telomere dynamics in peripheral blood cells from patients with Lynch syndrome. Cancer. 2011; 117: 4325-4335.
[13] Armanios M, Blackburn EH. The telomere syndromes. Nat Rev Genet. 2012; 13(10): 693-704.
[14] Shay JW. Role of telomeres and telomerase in aging and cancer. Cancer Discov. 2016; 6(6): 584-593.
[15] Omori Y, Nakayama F, Li D, Kanemitsu K, Semba S, Ito A, et al. Alternative lengthening of telomeres frequently occurs in mismatch repair system-deficient gastric carcinoma. Cancer Sci. 2009; 100: 413-418.
[16] Freshney RI. Culture of Animal Cells: A Manual of Basic Technique and Specialized Applications. 6th ed. Wiley-Liss; 2010.
[17] Sambrook J, Russell DW. Molecular Cloning: A Laboratory Manual. 3rd ed. Cold Spring Harbor Laboratory Press; 2001.
[18] Harlow E, Lane D. Using Antibodies: A Laboratory Manual. Cold Spring Harbor Laboratory Press; 2006.
[19] Hosen I, Rachakonda PS, Heidenreich B, de Verdier PJ, Ryk C, Steineck G, et al. Mutations in TERT promoter and FGFR3 and telomere length in bladder cancer. Int J Cancer. 2015; 137: 1621-1629.
[20] Vasilishina A, Kropotov A, Spivak I, Bernadotte A. Relative human telomere length quantification by real-time PCR. Methods Mol Biol. 2019; 1973: 39-44.
[21] Raji N, Majidi Zadeh T, Babheidarian P, Houshmand M. Detection of Microsatellite Instability by High-Resolution Melting Analysis in Colorectal Cancer. Iran Biomed J. 2022 Jan 1; 26(1): 70-6.
[22] Suraweera N, Duval A, Reperant M, et al. Evaluation of tumor microsatellite instability using five quasi-monomorphic mononucleotide repeats and pentaplex PCR. Oncogene. 2002; 21(37): 6041-6047.
[23] Goel A, Nagasaka T, Hamelin R, Boland CR. An optimized pentaplex PCR for detecting DNA mismatch repair-deficient colorectal cancers. PLoS One. 2010; 5: e9393.
[24] Smith PK, Krohn RI, Hermanson GT, et al. Measurement of protein using bicinchoninic acid. Anal Biochem. 1985; 150(1): 76-85.
[25] Jiricny J. The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol. 2006; 7(5): 335-346.
[26] Martinez P, Siegl‐Cachedenier I, Flores JM, Blasco MA. MSH2 deficiency abolishes the anticancer and pro‐aging activity of short telomeres. Aging Cell. 2009; 8(1): 2-17.
[27] Mendez‐Bermudez A, Royle NJ. Deficiency in DNA mismatch repair increases the rate of telomere shortening in normal human cells. Hum Mutat. 2011; 32(8): 939-946.
[28] Vilar E, Gruber SB. Microsatellite instability in colorectal cancer—the stable evidence. Nat Rev Clin Oncol. 2010; 7(3): 153-162.
[29] Kinouchi Y, Hiwatashi N, Chida M, Nagashima F, Takagi S, Maekawa H, et al. Telomere shortening in the colonic mucosa of patients with ulcerative colitis. J Gastroenterol. 1998; 33: 343–348.
[30] de la Chapelle A. Microsatellite instability. N Engl J Med. 2003; 349(3): 209-210.
[31] Denchi EL. Give me a break: How telomeres suppress the DNA damage response. DNA Repair (Amst). 2009;8(9):1118–1126.
[32] Nersisyan L, Hopp L, Loeffler-Wirth H, Galle J, Loeffler M, Arakelyan A, et al. Telomere length maintenance and its transcriptional regulation in Lynch syndrome and sporadic colorectal carcinoma. Front Oncol. 2019; 9: 1172.
[33] Greider CW, Blackburn EH. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell. 1985; 43(2 Pt 1): 405-413.
[34] Barthel FP, Wei W, Tang M, et al. Systematic analysis of telomere length and somatic alterations in 31 cancer types. Nat Genet. 2017; 49(3): 349-357.
Cite This Article
  • APA Style

    Moka, R., Upadhya, S. (2025). Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines. Cancer Research Journal, 13(2), 23-29. https://doi.org/10.11648/j.crj.20251302.11

    Copy | Download

    ACS Style

    Moka, R.; Upadhya, S. Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines. Cancer Res. J. 2025, 13(2), 23-29. doi: 10.11648/j.crj.20251302.11

    Copy | Download

    AMA Style

    Moka R, Upadhya S. Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines. Cancer Res J. 2025;13(2):23-29. doi: 10.11648/j.crj.20251302.11

    Copy | Download

  • @article{10.11648/j.crj.20251302.11,
      author = {Rajasekhar Moka and Shreya Upadhya},
      title = {Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines
    },
      journal = {Cancer Research Journal},
      volume = {13},
      number = {2},
      pages = {23-29},
      doi = {10.11648/j.crj.20251302.11},
      url = {https://doi.org/10.11648/j.crj.20251302.11},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.crj.20251302.11},
      abstract = {Microsatellite instability (MSI) is a hallmark of mismatch repair (MMR) deficiency and characterizes a distinct subset of colorectal cancers (CRC). In parallel, telomere length dynamics have emerged as important contributors to genomic stability and tumorigenesis. However, the relationship between MSI status, MMR protein expression, and telomere maintenance remains poorly defined. This study aimed to investigate the association between MSI status and telomere length in CRC cell lines and to evaluate the expression of key MMR proteins (MLH1, MSH2, MSH6, PMS2) to elucidate molecular differences between MSI and microsatellite stable (MSS) phenotypes. A panel of CRC cell lines with known MSI and MSS statuses was used. Telomere length was quantified using real-time quantitative PCR (qPCR) based on the T/S ratio method. MSI status was confirmed via PCR using mononucleotide repeat markers. Western blotting was performed to assess protein expressions of MLH1, MSH2, MSH6, and PMS2. β-actin served as a loading control. qPCR analysis revealed that MSI cell lines exhibited significantly longer telomeres compared to MSS lines (P < 0.05). Western blot results showed complete loss of MSH6 protein expression and reduction in expression levels of PMS2 and MSH2 in MSI cell lines, confirming MMR deficiency. In contrast, MSS cell lines maintained normal expression of all tested MMR proteins. These findings suggest a link between defective MMR function and altered telomere dynamics in MSI-CRC. MSI CRC cell lines exhibit telomere elongation and loss of key MMR proteins, highlighting distinct molecular features compared to MSS counterparts. These insights may inform future strategies for personalized CRC diagnostics and therapeutics, particularly in the context of telomere-targeted or immunomodulatory treatments.
    },
     year = {2025}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Telomere Elongation and Mismatch Repair Deficiency Distinguish MSI Colorectal Cancer Cell Lines
    
    AU  - Rajasekhar Moka
    AU  - Shreya Upadhya
    Y1  - 2025/05/29
    PY  - 2025
    N1  - https://doi.org/10.11648/j.crj.20251302.11
    DO  - 10.11648/j.crj.20251302.11
    T2  - Cancer Research Journal
    JF  - Cancer Research Journal
    JO  - Cancer Research Journal
    SP  - 23
    EP  - 29
    PB  - Science Publishing Group
    SN  - 2330-8214
    UR  - https://doi.org/10.11648/j.crj.20251302.11
    AB  - Microsatellite instability (MSI) is a hallmark of mismatch repair (MMR) deficiency and characterizes a distinct subset of colorectal cancers (CRC). In parallel, telomere length dynamics have emerged as important contributors to genomic stability and tumorigenesis. However, the relationship between MSI status, MMR protein expression, and telomere maintenance remains poorly defined. This study aimed to investigate the association between MSI status and telomere length in CRC cell lines and to evaluate the expression of key MMR proteins (MLH1, MSH2, MSH6, PMS2) to elucidate molecular differences between MSI and microsatellite stable (MSS) phenotypes. A panel of CRC cell lines with known MSI and MSS statuses was used. Telomere length was quantified using real-time quantitative PCR (qPCR) based on the T/S ratio method. MSI status was confirmed via PCR using mononucleotide repeat markers. Western blotting was performed to assess protein expressions of MLH1, MSH2, MSH6, and PMS2. β-actin served as a loading control. qPCR analysis revealed that MSI cell lines exhibited significantly longer telomeres compared to MSS lines (P < 0.05). Western blot results showed complete loss of MSH6 protein expression and reduction in expression levels of PMS2 and MSH2 in MSI cell lines, confirming MMR deficiency. In contrast, MSS cell lines maintained normal expression of all tested MMR proteins. These findings suggest a link between defective MMR function and altered telomere dynamics in MSI-CRC. MSI CRC cell lines exhibit telomere elongation and loss of key MMR proteins, highlighting distinct molecular features compared to MSS counterparts. These insights may inform future strategies for personalized CRC diagnostics and therapeutics, particularly in the context of telomere-targeted or immunomodulatory treatments.
    
    VL  - 13
    IS  - 2
    ER  - 

    Copy | Download

Author Information
  • Abstract
  • Keywords
  • Document Sections

    1. 1. Introduction
    2. 2. Materials and Methods
    3. 3. Results
    4. 4. Discussion
    Show Full Outline
  • Abbreviations
  • Acknowledgments
  • Author Contributions
  • Conflicts of Interest
  • References
  • Cite This Article
  • Author Information