Mol. Cells 2020; 43(7): 619-631
Published online July 8, 2020
https://doi.org/10.14348/molcells.2020.0075
© The Korean Society for Molecular and Cellular Biology
Correspondence to : sanghyuk@ewha.ac.kr (SL); jkim1964@ewha.ac.kr (JK)
In this study, we describe a novel function of TNNC1 (Troponin C1, Slow Skeletal and Cardiac Type), a component of actin-bound troponin, as a tumor suppressor of lung adenocarcinoma (LUAD). First, the expression of TNNC1 was strongly down-regulated in cancer tissues compared to matched normal lung tissues, and down-regulation of TNNC1 was shown to be strongly correlated with increased mortality among LUAD patients. Interestingly, TNNC1 expression was enhanced by suppression of KRAS, and ectopic expression of TNNC1 in turn inhibited KRASG12D-mediated anchorage independent growth of NIH3T3 cells. Consistently, activation of KRAS pathway in LUAD patients was shown to be strongly correlated with down-regulation of TNNC1. In addition, ectopic expression of TNNC1 inhibited colony formation of multiple LUAD cell lines and induced DNA damage, cell cycle arrest and ultimately apoptosis. We further examined potential correlations between expression levels of TNNC1 and various clinical parameters and found that low-level expression is significantly associated with invasiveness of the tumor. Indeed, RNA interference-mediated down-regulation of TNNC1 led to significant enhancement of invasiveness in vitro. Collectively, our data indicate that TNNC1 has a novel function as a tumor suppressor and is targeted for down-regulation by KRAS pathway during the carcinogenesis of LUAD.
Keywords invasion, KRAS, lung adenocarcinoma, TNNC1, tumor suppressor
Lung cancer represents one of the top-ranked cancers in terms of both incidence and mortality (Siegel et al., 2018). In United States, 234,030 new cases of lung and bronchus cancer had been predicted to occur in 2018 which in terms of incidence would represent the second highest among all types of cancer (Siegel et al., 2018). Approximately 83,550 men and 70,500 women were projected to die that year from this disease which has been the leading cause of cancer-related death for over 25 years (Siegel et al., 2018). Given the poor prospect with the overall five-year survival rate of less than 20%, there is an urgent need for development of new diagnostic and therapeutic strategies (Siegel et al., 2018).
The most significant contribution to this end is likely to come from large-scale high-throughput sequencing projects and systems biological analyses which have provided comprehensive molecular profiling of multiple types of cancer. For lung cancers, the most notable study was published in 2014 by The Cancer Genome Atlas Research Network, a genomics program aiming to molecularly characterize primary cancers and matched normal samples of multiple cancer types (Cancer Genome Atlas Research Network, 2014). This particular study used 230 resected surgical samples whose transcriptome and exome were subsequently profiled for somatic mutations, differentially expressed genes (DEGs) and altered pathways. Data from such studies are expected to bring new understanding to carcinogenesis at the molecular level and point to candidate actionable events. However, given that large scale studies typically result in hundreds of mutations and thousands of DEGs, there is a great need to distinguish the so-called drivers and passengers among mutations and DEGs based on functional analyses.
Troponin, a complex of three regulatory proteins Troponin C, Troponin I and Troponin T, is best known for regulating muscle contraction as a component of the thin filament (Johnston et al., 2018). Accordingly, they are abundantly expressed in cytoplasm of skeletal and cardiac muscle cells. Mounting evidence also suggests that various subunits of troponin are expressed in non-muscle tissues and cells. These include eye, brain, ovary, lung, bone, and liver (Berezowsky and Bag, 1992; Chen et al., 2014; Johnston et al., 2018; Leung et al., 2014; Moses et al., 1999; Schmidt et al., 2006). Interestingly, some of the troponin genes are found in nucleus rather than cytoplasm depending on cell types and were proposed to be involved in nuclear processes including transcription by RNA polymerase II (Casas-Tinto et al., 2016; Chase et al., 2013; Johnston et al., 2018; Sahota et al., 2009). Most intriguingly, troponin genes are expressed in diverse cancer cells and in immortalized cell lines and show oncogenic or tumor suppressor activities. For example, slow skeletal and cardiac type Troponin C1 (
Here, we present data indicating that
Tumor and normal tissue samples were obtained from patients who had undergone curative surgery at the Samsung Medical Center (Korea). Informed consents were obtained from patients, and all plans and procedures were approved by the Institutional Review Boards of Samsung Medical Center (IRB No. 2010-08-063-006) in accordance with the Declaration of Helsinki. RNAseq for the said samples has been described (Yu et al., 2019), and the data for the tumor and matched normal samples were processed according to the TCGA pipeline of MapSplice-RSEM (Li and Dewey, 2011; Wang et al., 2010) using Ensembl 81 genome and transcript models. Sequence reads were normalized within-samples to the upper quartile of total reads. DEGs were obtained using Voom (Law et al., 2014) with false discovery rate < 0.001. TCGA gene expression data were downloaded at level 3 from the Broad GDAC Firehose website (released on January 28, 2016) (Cerami et al., 2012; Gao et al., 2013).
The Hallmark gene sets and C2 gene sets from the molecular signature database (MSigDB) were used to examine the correlation between the expression level of
A549 and NCI-H2009, human LUAD cell lines, were obtained from the American Type Culture Collection (ATCC, USA). Cells were cultured in RPMI-1640 supplemented with 10% fetal bovine serum (Hyclone, USA). NIH3T3, mouse embryonic fibroblast cells were purchased from the ATCC and cultured in DMEM supplemented with 10% calf serum (Invitrogen, USA).
A549 or H2009 cells at 40% confluence were transfected with 40 nM
The coding regions of
Soft agar colony formation assay was carried out as described previously with minor modifications (Jung et al., 2015a; 2015b). NIH3T3 cells were seeded at a density of 3 × 104 per well in 12-well tissue culture plates and infected with the retrovirus. After two days, 2,000 cells were resuspended in complete media containing 0.35% Difco Noble agar (BD Biosciences, USA) and plated on the top of solidified 0.9% base agar containing complete media. Twenty-one days after plating the cells in soft agar, cells were stained with 1 mg/ml of MTT solution overnight. The colonies were counted using OpenCFU (Geissmann, 2013) version 3.9.
For conventional reverse transcription PCR (RT-PCR) analysis, cDNA was amplified using Platinum
A549 and H2009 cells were plated at a density of 2 × 104 and 3 × 104, respectively in 12-well tissue culture plates. After incubation for 24 h, cells were infected with the retrovirus. Two days after infection, 1,000 live A549 cells and 2,000 live H2009 cells were re-plated in 6-well tissue culture plates in duplicates. Nine days (A549) or 13 days (H2009) after incubation, colonies were stained with 0.1% Coomassie Blue in 45% methanol and 10% acetic acid solution. Colony numbers were determined using OpenCFU.
For apoptosis analyses, cells were seeded and infected with the retrovirus. After 96 h, trypsinized cells were collected and washed with cold PBS and resuspended in 1× Annexin V binding buffer (BD Biosciences) at a concentration of 1 × 106 cells/ml. After staining with propidium iodide and V450 annexin V (BD Biosciences), flow cytometric analyses typically using 10,000 cells were carried out using BD LSRFortessa cell analyzer. For cell cycle analyses, cells were seeded in 6-well plates and infected with the retrovirus. After 48 h, trypsinized cells were washed with cold PBS and fixed with 70% ethanol overnight. Subsequently, cells were stained with 50 µg/ml propidium iodide (PI; Sigma, USA) in PBS containing 10 μg/ml RNase A (Sigma) and 0.1% Triton X-100 at RT for 15 min. Flow cytometry for cell cycle analysis was carried out using BD LSRFortessa cell analyzer (BD Biosciences) and the distribution of a total of 10,000 nuclei was determined using BD FACSDiva software (BD Biosciences).
TNNC1-V5-expressing cells were harvested and lysed using RIPA buffer as described previously (Jung et al., 2015a). Primary antibodies used in immunoblot analyses are as follows: anti-V5 (Invitrogen), anti-p53 (Santa Cruz Biotechnology, USA), anti-p21 Waf1/Cip1 (Cell Signaling Technology, USA), anti-CDC25C (Cell Signaling Technology), anti-Cyclin B1 (Cell Signaling Technology), anti-α-tubulin (AbFrontier, Korea), and anti-γH2AX (Millipore, USA). Peroxidase-conjugated secondary antibodies were used for detection in combination with AbSignal Western Blotting Detection Reagent kit (Abclon, Korea) or enhanced chemiluminescence detection kit (Amersham-Pharmacia Biotech, USA) following the manufacturer’s protocols.
For immunofluorescence, the transduced cells were fixed in 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 in PBS and blocked with 1% BSA in PBS. Cells were first stained with anti-γH2AX antibody or anti-V5, followed by Alexa Fluor 594 goat anti-mouse (Invitrogen) and DAPI (4',6-diamidino-2-phenylindole) treatment and were examined by epifluorescence microscopy.
Cell invasion assay was performed with BD BioCoat Matrigel Invasion Chambers (BD Bioscience) according to the manufacturer’s instruction. siRNA duplexes of
The association between
We analyzed RNA-seq data from tumor and matched normal samples of 102 LUAD patients, and a total of 1,533 DEGs were identified as significantly down-regulated genes in tumor tissues. Tumor suppressor candidates were isolated based on several additional filtering criteria including FDR < 0.001 in Voom analysis, consistency of > 98%, Log2 fold change of median normalized reads (MNRs) in normal versus tumor tissue < –3, and MNRs of normal tissue > 1,500 and MNRs of tumor tissue < 1,000 (Fig. 1A). From the 28 genes thus found, we were able to obtain reliable results from real-time PCR analyses for 15 genes indicating sufficient levels of expression in A549 cells (Fig. 1B).
We proceeded to examine changes in their expression upon inhibition of
First, we investigated the association of
We further investigated the interaction between
We examined the expression of
Using colony formation assay, we examined if TNNC1 has an inhibitory effect on cell growth in two independent cell lines, A549 and H2009. Upon ectopic expression of
One frequently seen effect of tumor suppressor expression is the induction of cell cycle arrest. We examined the cell cycle progression of A549 and H2009 cells after ectopic expression of
The differential response based on p53 status suggested that TNNC1 possibly induces or mediates DNA damage response which would in turn normally induce p53 response and G1 arrest. We examined cells after ectopic expression of
We next used clinical data to test if any of the major clinical parameters were correlated with the expression level of
Multiple large-scale high throughput sequencing projects for diverse types of cancers have been reported recently. In the case of lung cancer, a comprehensive multi-layered profiling of 230 LUAD is the most representative (Cancer Genome Atlas Research Network, 2014). While numerous mutations, DEGs and altered pathways have been proposed for their potential involvement, each event clearly needs to be carefully re-examined for its significance in carcinogenesis by experimental validation as a large fraction of them is likely to be the so-called ‘passenger’ events.
Several studies have reported that various troponin genes are expressed in a variety of non-contracting cells and involved in cancer development although the exact function and mechanism for their activities are barely known (Johnston et al., 2018). From our sequencing analyses of matched normal and tumor samples of over 100 never-smoker female patients of LUAD,
Our data indicate that
It is also interesting that
Although our data together render
Authors thank all members of Ewha Research Center for Systems Biology for the helpful discussions. This research was supported by funding from the Ministry of Science and ICT via National Research Foundation, Republic of Korea (NRF-2015K1A4A3047851).
S.K., Jaewon K., Yeonjoo J., and Yukyung J. performed experiments and analyzed data. Yeonwha J., H.Y.L., and Juhee K. provided technical support and performed experiments. B.J.P. and J.L. prepared and provided samples and reagents. Jhingook K., S.L., and Jaesang K. conceived the study and wrote the manuscript.
The authors have no potential conflicts of interest to disclose.
Association of
Clinicopathological features | χ2 | P value | |||
---|---|---|---|---|---|
Low (n = 38) | High (n = 64) | ||||
Age (y) | < 62 | 19 | 31 | 0.02 | 0.879 |
≥ 62 | 19 | 33 | |||
Tumor size (cm) | < 3 | 21 | 47 | 6.52 | 0.038* |
3-5 | 16 | 12 | |||
≥ 5 | 1 | 4 | |||
Null | 0 | 1 | |||
Pathologic stagea | 1 | 23 | 41 | 1.89 | 0.595 |
2 | 8 | 8 | |||
3 | 7 | 14 | |||
4 | 0 | 1 | |||
T stagea | 1 | 15 | 41 | 13.2 | 0.004* |
2 | 23 | 17 | |||
3 | 0 | 5 | |||
4 | 0 | 1 | |||
N stagea | 0 | 24 | 44 | 4.7 | 0.094 |
1 | 7 | 3 | |||
2 | 7 | 14 | |||
Null | 0 | 3 | |||
Differentiation grade | Well differentiated | 1 | 8 | 2.7 | 0.257 |
Moderately differentiated | 29 | 47 | |||
Poorly differentiated | 3 | 4 | |||
Null | 5 | 5 | |||
Vascular invasion | No | 33 | 61 | 6.9 | 0.009* |
Yes | 4 | 0 | |||
Null | 1 | 3 | |||
Lymphatic invasion | No | 21 | 44 | 2.4 | 0.118 |
Yes | 16 | 17 | |||
Null | 1 | 3 | |||
Visceral pleural invasion | No | 21 | 53 | 8.1 | 0.004* |
Yes | 15 | 10 | |||
Null | 2 | 1 | |||
Recurrence status | No | 21 | 41 | 1 | 0.326 |
Yes | 17 | 22 | |||
Null | 0 | 1 |
Major clinicopathological features and subcategories therein are listed. Patients are grouped into either high or low TNNC1 expression groups as in Fig. 3C. The chi-squared test shows significant correlation with tumor size, tumor stage, vascular invasion, and visceral pleural invasion.
Null, no data.
*
a The clinical and pathological stage was determined according to the 7th edition of the American Joint Committee on Cancer.
Mol. Cells 2020; 43(7): 619-631
Published online July 31, 2020 https://doi.org/10.14348/molcells.2020.0075
Copyright © The Korean Society for Molecular and Cellular Biology.
Suyeon Kim1,2,5 , Jaewon Kim1,2,5
, Yeonjoo Jung1,2,5
, Yukyung Jun1,2
, Yeonhwa Jung2
, Hee-Young Lee2
, Juhee Keum2
, Byung Jo Park3
, Jinseon Lee4
, Jhingook Kim3
, Sanghyuk Lee1,2,*
, and Jaesang Kim1,2,*
1Department of Life Science, Ewha Womans University, Seoul 03760, Korea, 2Ewha Research Center for Systems Biology, Ewha Womans University, Seoul 03760, Korea, 3Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, 4Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea, 5These authors contributed equally to this work.
Correspondence to:sanghyuk@ewha.ac.kr (SL); jkim1964@ewha.ac.kr (JK)
In this study, we describe a novel function of TNNC1 (Troponin C1, Slow Skeletal and Cardiac Type), a component of actin-bound troponin, as a tumor suppressor of lung adenocarcinoma (LUAD). First, the expression of TNNC1 was strongly down-regulated in cancer tissues compared to matched normal lung tissues, and down-regulation of TNNC1 was shown to be strongly correlated with increased mortality among LUAD patients. Interestingly, TNNC1 expression was enhanced by suppression of KRAS, and ectopic expression of TNNC1 in turn inhibited KRASG12D-mediated anchorage independent growth of NIH3T3 cells. Consistently, activation of KRAS pathway in LUAD patients was shown to be strongly correlated with down-regulation of TNNC1. In addition, ectopic expression of TNNC1 inhibited colony formation of multiple LUAD cell lines and induced DNA damage, cell cycle arrest and ultimately apoptosis. We further examined potential correlations between expression levels of TNNC1 and various clinical parameters and found that low-level expression is significantly associated with invasiveness of the tumor. Indeed, RNA interference-mediated down-regulation of TNNC1 led to significant enhancement of invasiveness in vitro. Collectively, our data indicate that TNNC1 has a novel function as a tumor suppressor and is targeted for down-regulation by KRAS pathway during the carcinogenesis of LUAD.
Keywords: invasion, KRAS, lung adenocarcinoma, TNNC1, tumor suppressor
Lung cancer represents one of the top-ranked cancers in terms of both incidence and mortality (Siegel et al., 2018). In United States, 234,030 new cases of lung and bronchus cancer had been predicted to occur in 2018 which in terms of incidence would represent the second highest among all types of cancer (Siegel et al., 2018). Approximately 83,550 men and 70,500 women were projected to die that year from this disease which has been the leading cause of cancer-related death for over 25 years (Siegel et al., 2018). Given the poor prospect with the overall five-year survival rate of less than 20%, there is an urgent need for development of new diagnostic and therapeutic strategies (Siegel et al., 2018).
The most significant contribution to this end is likely to come from large-scale high-throughput sequencing projects and systems biological analyses which have provided comprehensive molecular profiling of multiple types of cancer. For lung cancers, the most notable study was published in 2014 by The Cancer Genome Atlas Research Network, a genomics program aiming to molecularly characterize primary cancers and matched normal samples of multiple cancer types (Cancer Genome Atlas Research Network, 2014). This particular study used 230 resected surgical samples whose transcriptome and exome were subsequently profiled for somatic mutations, differentially expressed genes (DEGs) and altered pathways. Data from such studies are expected to bring new understanding to carcinogenesis at the molecular level and point to candidate actionable events. However, given that large scale studies typically result in hundreds of mutations and thousands of DEGs, there is a great need to distinguish the so-called drivers and passengers among mutations and DEGs based on functional analyses.
Troponin, a complex of three regulatory proteins Troponin C, Troponin I and Troponin T, is best known for regulating muscle contraction as a component of the thin filament (Johnston et al., 2018). Accordingly, they are abundantly expressed in cytoplasm of skeletal and cardiac muscle cells. Mounting evidence also suggests that various subunits of troponin are expressed in non-muscle tissues and cells. These include eye, brain, ovary, lung, bone, and liver (Berezowsky and Bag, 1992; Chen et al., 2014; Johnston et al., 2018; Leung et al., 2014; Moses et al., 1999; Schmidt et al., 2006). Interestingly, some of the troponin genes are found in nucleus rather than cytoplasm depending on cell types and were proposed to be involved in nuclear processes including transcription by RNA polymerase II (Casas-Tinto et al., 2016; Chase et al., 2013; Johnston et al., 2018; Sahota et al., 2009). Most intriguingly, troponin genes are expressed in diverse cancer cells and in immortalized cell lines and show oncogenic or tumor suppressor activities. For example, slow skeletal and cardiac type Troponin C1 (
Here, we present data indicating that
Tumor and normal tissue samples were obtained from patients who had undergone curative surgery at the Samsung Medical Center (Korea). Informed consents were obtained from patients, and all plans and procedures were approved by the Institutional Review Boards of Samsung Medical Center (IRB No. 2010-08-063-006) in accordance with the Declaration of Helsinki. RNAseq for the said samples has been described (Yu et al., 2019), and the data for the tumor and matched normal samples were processed according to the TCGA pipeline of MapSplice-RSEM (Li and Dewey, 2011; Wang et al., 2010) using Ensembl 81 genome and transcript models. Sequence reads were normalized within-samples to the upper quartile of total reads. DEGs were obtained using Voom (Law et al., 2014) with false discovery rate < 0.001. TCGA gene expression data were downloaded at level 3 from the Broad GDAC Firehose website (released on January 28, 2016) (Cerami et al., 2012; Gao et al., 2013).
The Hallmark gene sets and C2 gene sets from the molecular signature database (MSigDB) were used to examine the correlation between the expression level of
A549 and NCI-H2009, human LUAD cell lines, were obtained from the American Type Culture Collection (ATCC, USA). Cells were cultured in RPMI-1640 supplemented with 10% fetal bovine serum (Hyclone, USA). NIH3T3, mouse embryonic fibroblast cells were purchased from the ATCC and cultured in DMEM supplemented with 10% calf serum (Invitrogen, USA).
A549 or H2009 cells at 40% confluence were transfected with 40 nM
The coding regions of
Soft agar colony formation assay was carried out as described previously with minor modifications (Jung et al., 2015a; 2015b). NIH3T3 cells were seeded at a density of 3 × 104 per well in 12-well tissue culture plates and infected with the retrovirus. After two days, 2,000 cells were resuspended in complete media containing 0.35% Difco Noble agar (BD Biosciences, USA) and plated on the top of solidified 0.9% base agar containing complete media. Twenty-one days after plating the cells in soft agar, cells were stained with 1 mg/ml of MTT solution overnight. The colonies were counted using OpenCFU (Geissmann, 2013) version 3.9.
For conventional reverse transcription PCR (RT-PCR) analysis, cDNA was amplified using Platinum
A549 and H2009 cells were plated at a density of 2 × 104 and 3 × 104, respectively in 12-well tissue culture plates. After incubation for 24 h, cells were infected with the retrovirus. Two days after infection, 1,000 live A549 cells and 2,000 live H2009 cells were re-plated in 6-well tissue culture plates in duplicates. Nine days (A549) or 13 days (H2009) after incubation, colonies were stained with 0.1% Coomassie Blue in 45% methanol and 10% acetic acid solution. Colony numbers were determined using OpenCFU.
For apoptosis analyses, cells were seeded and infected with the retrovirus. After 96 h, trypsinized cells were collected and washed with cold PBS and resuspended in 1× Annexin V binding buffer (BD Biosciences) at a concentration of 1 × 106 cells/ml. After staining with propidium iodide and V450 annexin V (BD Biosciences), flow cytometric analyses typically using 10,000 cells were carried out using BD LSRFortessa cell analyzer. For cell cycle analyses, cells were seeded in 6-well plates and infected with the retrovirus. After 48 h, trypsinized cells were washed with cold PBS and fixed with 70% ethanol overnight. Subsequently, cells were stained with 50 µg/ml propidium iodide (PI; Sigma, USA) in PBS containing 10 μg/ml RNase A (Sigma) and 0.1% Triton X-100 at RT for 15 min. Flow cytometry for cell cycle analysis was carried out using BD LSRFortessa cell analyzer (BD Biosciences) and the distribution of a total of 10,000 nuclei was determined using BD FACSDiva software (BD Biosciences).
TNNC1-V5-expressing cells were harvested and lysed using RIPA buffer as described previously (Jung et al., 2015a). Primary antibodies used in immunoblot analyses are as follows: anti-V5 (Invitrogen), anti-p53 (Santa Cruz Biotechnology, USA), anti-p21 Waf1/Cip1 (Cell Signaling Technology, USA), anti-CDC25C (Cell Signaling Technology), anti-Cyclin B1 (Cell Signaling Technology), anti-α-tubulin (AbFrontier, Korea), and anti-γH2AX (Millipore, USA). Peroxidase-conjugated secondary antibodies were used for detection in combination with AbSignal Western Blotting Detection Reagent kit (Abclon, Korea) or enhanced chemiluminescence detection kit (Amersham-Pharmacia Biotech, USA) following the manufacturer’s protocols.
For immunofluorescence, the transduced cells were fixed in 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 in PBS and blocked with 1% BSA in PBS. Cells were first stained with anti-γH2AX antibody or anti-V5, followed by Alexa Fluor 594 goat anti-mouse (Invitrogen) and DAPI (4',6-diamidino-2-phenylindole) treatment and were examined by epifluorescence microscopy.
Cell invasion assay was performed with BD BioCoat Matrigel Invasion Chambers (BD Bioscience) according to the manufacturer’s instruction. siRNA duplexes of
The association between
We analyzed RNA-seq data from tumor and matched normal samples of 102 LUAD patients, and a total of 1,533 DEGs were identified as significantly down-regulated genes in tumor tissues. Tumor suppressor candidates were isolated based on several additional filtering criteria including FDR < 0.001 in Voom analysis, consistency of > 98%, Log2 fold change of median normalized reads (MNRs) in normal versus tumor tissue < –3, and MNRs of normal tissue > 1,500 and MNRs of tumor tissue < 1,000 (Fig. 1A). From the 28 genes thus found, we were able to obtain reliable results from real-time PCR analyses for 15 genes indicating sufficient levels of expression in A549 cells (Fig. 1B).
We proceeded to examine changes in their expression upon inhibition of
First, we investigated the association of
We further investigated the interaction between
We examined the expression of
Using colony formation assay, we examined if TNNC1 has an inhibitory effect on cell growth in two independent cell lines, A549 and H2009. Upon ectopic expression of
One frequently seen effect of tumor suppressor expression is the induction of cell cycle arrest. We examined the cell cycle progression of A549 and H2009 cells after ectopic expression of
The differential response based on p53 status suggested that TNNC1 possibly induces or mediates DNA damage response which would in turn normally induce p53 response and G1 arrest. We examined cells after ectopic expression of
We next used clinical data to test if any of the major clinical parameters were correlated with the expression level of
Multiple large-scale high throughput sequencing projects for diverse types of cancers have been reported recently. In the case of lung cancer, a comprehensive multi-layered profiling of 230 LUAD is the most representative (Cancer Genome Atlas Research Network, 2014). While numerous mutations, DEGs and altered pathways have been proposed for their potential involvement, each event clearly needs to be carefully re-examined for its significance in carcinogenesis by experimental validation as a large fraction of them is likely to be the so-called ‘passenger’ events.
Several studies have reported that various troponin genes are expressed in a variety of non-contracting cells and involved in cancer development although the exact function and mechanism for their activities are barely known (Johnston et al., 2018). From our sequencing analyses of matched normal and tumor samples of over 100 never-smoker female patients of LUAD,
Our data indicate that
It is also interesting that
Although our data together render
Authors thank all members of Ewha Research Center for Systems Biology for the helpful discussions. This research was supported by funding from the Ministry of Science and ICT via National Research Foundation, Republic of Korea (NRF-2015K1A4A3047851).
S.K., Jaewon K., Yeonjoo J., and Yukyung J. performed experiments and analyzed data. Yeonwha J., H.Y.L., and Juhee K. provided technical support and performed experiments. B.J.P. and J.L. prepared and provided samples and reagents. Jhingook K., S.L., and Jaesang K. conceived the study and wrote the manuscript.
The authors have no potential conflicts of interest to disclose.
. Association of
Clinicopathological features | χ2 | P value | |||
---|---|---|---|---|---|
Low (n = 38) | High (n = 64) | ||||
Age (y) | < 62 | 19 | 31 | 0.02 | 0.879 |
≥ 62 | 19 | 33 | |||
Tumor size (cm) | < 3 | 21 | 47 | 6.52 | 0.038* |
3-5 | 16 | 12 | |||
≥ 5 | 1 | 4 | |||
Null | 0 | 1 | |||
Pathologic stagea | 1 | 23 | 41 | 1.89 | 0.595 |
2 | 8 | 8 | |||
3 | 7 | 14 | |||
4 | 0 | 1 | |||
T stagea | 1 | 15 | 41 | 13.2 | 0.004* |
2 | 23 | 17 | |||
3 | 0 | 5 | |||
4 | 0 | 1 | |||
N stagea | 0 | 24 | 44 | 4.7 | 0.094 |
1 | 7 | 3 | |||
2 | 7 | 14 | |||
Null | 0 | 3 | |||
Differentiation grade | Well differentiated | 1 | 8 | 2.7 | 0.257 |
Moderately differentiated | 29 | 47 | |||
Poorly differentiated | 3 | 4 | |||
Null | 5 | 5 | |||
Vascular invasion | No | 33 | 61 | 6.9 | 0.009* |
Yes | 4 | 0 | |||
Null | 1 | 3 | |||
Lymphatic invasion | No | 21 | 44 | 2.4 | 0.118 |
Yes | 16 | 17 | |||
Null | 1 | 3 | |||
Visceral pleural invasion | No | 21 | 53 | 8.1 | 0.004* |
Yes | 15 | 10 | |||
Null | 2 | 1 | |||
Recurrence status | No | 21 | 41 | 1 | 0.326 |
Yes | 17 | 22 | |||
Null | 0 | 1 |
Major clinicopathological features and subcategories therein are listed. Patients are grouped into either high or low TNNC1 expression groups as in Fig. 3C. The chi-squared test shows significant correlation with tumor size, tumor stage, vascular invasion, and visceral pleural invasion..
Null, no data..
*
a The clinical and pathological stage was determined according to the 7th edition of the American Joint Committee on Cancer..
Hongli Cao, Ping Zhang, Hong Yu*, and Jianing Xi*
Mol. Cells 2022; 45(6): 376-387 https://doi.org/10.14348/molcells.2022.2221Dandan Li, Weiyan Peng, Bin Wu, Huan Liu, Ruizhen Zhang, Ruiqin Zhou, Lijun Yao, and Lin Ye
Mol. Cells 2021; 44(4): 267-278 https://doi.org/10.14348/molcells.2021.2179Ji-hun Han, Jeong Seon Yoon, Da-Young Chang, Kyung Gi Cho, Jaejoon Lim, Sung-Soo Kim, and Haeyoung Suh-Kim
Mol. Cells 2020; 43(6): 539-550 https://doi.org/10.14348/molcells.2020.0098