1
|
Nałęcz D, Świętek A, Hudy D, Wiczkowski K, Złotopolska Z, Strzelczyk JK. Assessment of Concentration KRT6 Proteins in Tumor and Matching Surgical Margin from Patients with Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:7356. [PMID: 39000463 PMCID: PMC11242288 DOI: 10.3390/ijms25137356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are one of the most frequently detected cancers in the world; not all mechanisms related to the expression of keratin in this type of cancer are known. The aim of this study was to evaluate type II cytokeratins (KRT): KRT6A, KRT6B, and KRT6C protein concentrations in 54 tumor and margin samples of head and neck squamous cell carcinoma (HNSCC). Moreover, we examined a possible association between protein concentration and the clinical and demographic variables. Protein concentrations were measured using enzyme-linked immunosorbent assay (ELISA). Significantly higher KRT6A protein concentration was found in HNSCC samples compared to surgical margins. An inverse relationship was observed for KRT6B and KRT6C proteins. We showed an association between the KRT6C protein level and clinical parameters T and N in tumor and margin samples. When analyzing the effect of smoking and drinking on KRT6A, KRT6B, and KRT6C levels, we demonstrated a statistically significant difference between regular or occasional tobacco and alcohol habits and patients who do not have any tobacco and alcohol habits in tumor and margin samples. Moreover, we found an association between KRT6B and KRT6C concentration and proliferative index Ki-67 and HPV status in tumor samples. Our results showed that concentrations of KRT6s were different in the tumor and the margin samples and varied in relation to clinical and demographic parameters. We add information to the current knowledge about the role of KRT6s isoforms in HNSCC. We speculate that variations in the studied isoforms of the KRT6 protein could be due to the presence and development of the tumor and its microenvironment. It is important to note that the analyses were performed in tumor and surgical margins and can provide more accurate information on the function in normal and cancer cells and regulation in response to various factors.
Collapse
Affiliation(s)
- Dariusz Nałęcz
- Department of Otolaryngology and Maxillofacial Surgery, St. Vincent De Paul Hospital, 1 Wójta Radtkego St., 81-348 Gdynia, Poland
| | - Agata Świętek
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
- Silesia LabMed Research and Implementation Centre, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Dorota Hudy
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Karol Wiczkowski
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
- Students' Scientific Association, Department of Medical and Molecular Biology, Medical University of Silesia, Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| | - Zofia Złotopolska
- Department of Otolaryngology and Maxillofacial Surgery, St. Vincent De Paul Hospital, 1 Wójta Radtkego St., 81-348 Gdynia, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 19 Jordana St., 41-808 Zabrze, Poland
| |
Collapse
|
2
|
Melekoglu R, Yasar S, Colak C, Kasap M, Dogan UK, Yologlu S, Yilmaz E, Shazly S. Determination of biomarker candidates for the placenta accreta spectrum by plasma proteomic analysis. Sci Rep 2024; 14:2803. [PMID: 38307924 PMCID: PMC10837117 DOI: 10.1038/s41598-024-53324-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
Placenta accreta spectrum (PAS) presents a significant obstetric challenge, associated with considerable maternal and fetal-neonatal morbidity and mortality. Nevertheless, it is imperative to acknowledge that a noteworthy subset of PAS cases remains undetected until the time of delivery, thereby contributing to an augmented incidence of morbidity among the affected individuals. The delayed identification of PAS not only hinders timely intervention but also exacerbates the associated health risks for both the maternal and fetal outcomes. This underscores the urgency to innovate strategies for early PAS diagnosis. In this study, we aimed to explore plasma proteins as potential diagnostic biomarkers for PAS. Integrated transcriptome and proteomic analyses were conducted to establish a novel diagnostic approach. A cohort of 15 pregnant women diagnosed with PAS and delivering at Inonu University Faculty of Medicine between 01/04/2021 and 01/01/2023, along with a matched control group of 15 pregnant women without PAS complications, were enrolled. Plasma protein identification utilized enzymatic digestion and liquid chromatography-tandem mass spectrometry techniques. Proteomic analysis identified 228 plasma proteins, of which 85 showed significant differences (P < 0.001) between PAS and control cases. We refined this to a set of 20 proteins for model construction, resulting in a highly accurate classification model (96.9% accuracy). Notable associations were observed for proteins encoded by P01859 (Immunoglobulin heavy constant gamma 2), P02538 (Keratin type II cytoskeletal 6A), P29622 [Kallistatin (also known as Serpin A4)], P17900 (Ganglioside GM2 activator Calmodulin-like protein 5), and P01619 (Immunoglobulin kappa variable 3-20), with fold changes indicating their relevance in distinguishing PAS from control groups. In conclusion, our study has identified novel plasma proteins that could serve as potential biomarkers for early diagnosis of PAS in pregnant women. Further research and validation in larger PAS cohorts are necessary to determine the clinical utility and reliability of these proteomic biomarkers for diagnosing PAS.
Collapse
Affiliation(s)
- Rauf Melekoglu
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, 44280, Malatya, Turkey.
| | - Seyma Yasar
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280, Malatya, Turkey
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280, Malatya, Turkey
| | - Murat Kasap
- Department of Medical Biology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Umran Karabulut Dogan
- Clinic of Obstetrics and Gynecology, Malatya Education and Research Hospital, Malatya, Turkey
| | - Saim Yologlu
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, 44280, Malatya, Turkey
| | - Ercan Yilmaz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Inonu University, 44280, Malatya, Turkey
| | - Sherif Shazly
- Department of Obstetrics and Gynecology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
3
|
Yang Y, Zeng Z, Li L, Lei S, Wu Y, Chen T, Zhang J. Sinapine thiocyanate exhibited anti-colorectal cancer effects by inhibiting KRT6A/S100A2 axis. Cancer Biol Ther 2023; 24:2249170. [PMID: 37647260 PMCID: PMC10469431 DOI: 10.1080/15384047.2023.2249170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 04/18/2023] [Accepted: 06/02/2023] [Indexed: 09/01/2023] Open
Abstract
Sinapine thiocyanate (ST), an alkaloid existed extensively in seeds of cruciferous plants, exhibits a number of pharmacological effects, including anti-inflammatory and anti-malignancy properties. However, it is still unknown what effects and molecular mechanisms ST has on colorectal cancer (CRC). In the current study, it was indicated that ST inhibited proliferation, colony formation, and apoptosis in vitro, as well as arrested the G1 phase of CRC cells. There was a significant repressive effects of ST on invasion and migration of CRC cells in vitro. RNA-sequencing indicated that 750 differentially expressed genes existed in CRC cells after ST treatment, and enrichment analysis demonstrated that ST obviously decreased the activation of keratinization pathways. Among DEGs enriched in keratinization, keratin 6A (KRT6A) was decreased the most significant, as well as its target gene S100 calcium-binding protein A2 (S100A2). Low expression of KRT6A and S100A2 signatures indicated a favorable prognosis in CRC patients. Moreover, we found overexpression of KRT6A relieved the inhibitory effects of ST in CRC cells. Furthermore, ST inhibited the CRC cell proliferation in vivo, and reduced KRT6A and KI67 expression in xenograft tumor. Taken together, we demonstrated that ST exhibited anti-CRC properties by inhibiting KRT6A/S100A2 axis. It is possible that ST can be used as a treatment for CRC.
Collapse
Affiliation(s)
- Yan Yang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Internal medicine, The Third Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhirui Zeng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lian Li
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shan Lei
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yingmin Wu
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tengxiang Chen
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jinjuan Zhang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Ergology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
4
|
Terekhanova NV, Karpova A, Liang WW, Strzalkowski A, Chen S, Li Y, Southard-Smith AN, Iglesia MD, Wendl MC, Jayasinghe RG, Liu J, Song Y, Cao S, Houston A, Liu X, Wyczalkowski MA, Lu RJH, Caravan W, Shinkle A, Naser Al Deen N, Herndon JM, Mudd J, Ma C, Sarkar H, Sato K, Ibrahim OM, Mo CK, Chasnoff SE, Porta-Pardo E, Held JM, Pachynski R, Schwarz JK, Gillanders WE, Kim AH, Vij R, DiPersio JF, Puram SV, Chheda MG, Fuh KC, DeNardo DG, Fields RC, Chen F, Raphael BJ, Ding L. Epigenetic regulation during cancer transitions across 11 tumour types. Nature 2023; 623:432-441. [PMID: 37914932 PMCID: PMC10632147 DOI: 10.1038/s41586-023-06682-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
Chromatin accessibility is essential in regulating gene expression and cellular identity, and alterations in accessibility have been implicated in driving cancer initiation, progression and metastasis1-4. Although the genetic contributions to oncogenic transitions have been investigated, epigenetic drivers remain less understood. Here we constructed a pan-cancer epigenetic and transcriptomic atlas using single-nucleus chromatin accessibility data (using single-nucleus assay for transposase-accessible chromatin) from 225 samples and matched single-cell or single-nucleus RNA-sequencing expression data from 206 samples. With over 1 million cells from each platform analysed through the enrichment of accessible chromatin regions, transcription factor motifs and regulons, we identified epigenetic drivers associated with cancer transitions. Some epigenetic drivers appeared in multiple cancers (for example, regulatory regions of ABCC1 and VEGFA; GATA6 and FOX-family motifs), whereas others were cancer specific (for example, regulatory regions of FGF19, ASAP2 and EN1, and the PBX3 motif). Among epigenetically altered pathways, TP53, hypoxia and TNF signalling were linked to cancer initiation, whereas oestrogen response, epithelial-mesenchymal transition and apical junction were tied to metastatic transition. Furthermore, we revealed a marked correlation between enhancer accessibility and gene expression and uncovered cooperation between epigenetic and genetic drivers. This atlas provides a foundation for further investigation of epigenetic dynamics in cancer transitions.
Collapse
Affiliation(s)
- Nadezhda V Terekhanova
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Alla Karpova
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Wen-Wei Liang
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | | | - Siqi Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Yize Li
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Austin N Southard-Smith
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Michael D Iglesia
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Michael C Wendl
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Jingxian Liu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Yizhe Song
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Song Cao
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andrew Houston
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Xiuting Liu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Rita Jui-Hsien Lu
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Wagma Caravan
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Andrew Shinkle
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
| | - Nataly Naser Al Deen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - John M Herndon
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Jacqueline Mudd
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Kazuhito Sato
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Omar M Ibrahim
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Chia-Kuei Mo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA
| | - Sara E Chasnoff
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Barcelona Supercomputing Center, Barcelona, Spain
| | - Jason M Held
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Russell Pachynski
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University in St Louis, St Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Albert H Kim
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
- Department of Neurological Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Ravi Vij
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - John F DiPersio
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Sidharth V Puram
- Department of Otolaryngology-Head & Neck Surgery, Washington University in St Louis, St Louis, MO, USA
| | - Milan G Chheda
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Katherine C Fuh
- Department of Obstetrics and Gynecology, University of California, San Francisco, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, MO, USA
| | - David G DeNardo
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA
| | - Ryan C Fields
- Department of Surgery, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Feng Chen
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| | - Li Ding
- Department of Medicine, Washington University in St Louis, St Louis, MO, USA.
- McDonnell Genome Institute, Washington University in St Louis, St Louis, MO, USA.
- Siteman Cancer Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Genetics, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
5
|
Sun K, Zhang Z, Wang D, Huang Y, Zhang J, Lian C. Regulation of early diagnosis and prognostic markers of lung adenocarcinoma in immunity and hypoxia. Sci Rep 2023; 13:6459. [PMID: 37081097 PMCID: PMC10119119 DOI: 10.1038/s41598-023-33404-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 04/12/2023] [Indexed: 04/22/2023] Open
Abstract
Lung adenocarcinoma is still cancer with the highest mortality. Hypoxia and immunity play an essential role in the occurrence and development of tumors. Therefore, this study is mainly to find new early diagnosis and prognosis markers and explore the relationship among the markers and immunity and hypoxia, to improve the prognosis of patients. Firstly, based on the clinical database in TCGA, we determined the most critical clinicopathological parameters affecting the prognosis of patients through a variety of analysis methods. According to pathological parameters, logistic most minor absolute contraction selection operator (lasso), univariate and multivariate regression analysis, the risk genes related to early prognosis were screened, and the risk model was established. Then, in different risk groups, GSEA and CIBERSORT algorithms were used to analyze the distribution and enrichment of the immune cells and hypoxia, to study the effects of early prognostic indicators on hypoxia and immunity. At the same time, we analyzed the different levels of risk genes in normal cells (BSEA-2B) and tumor cells (H1299, A549, PC9, and H1975). Finally, A549 and PC9 cells were induced by CoCl2 to establish a hypoxic environment, and the correlation between risk genes and HIF1A was analyzed. The risk model based on risk genes (CYP4B1, KRT6A, and FAM83A) was accurate and stable for the prognosis of patients. It is closely related to immunity and hypoxia. In BSEA-2B cells, the mRNA and protein expression of CYP4B1 was higher, while the expression of KRT6A and FAM83A was lower. Finally, we found that FAM83A and HIF1A showed a significant positive correlation when A549 and PC9 cells were exposed to hypoxia. The discovery of early diagnostic markers related to immunity, hypoxia, and prognosis, provides a new idea for early screening and prognostic treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Kang Sun
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Zhiqiang Zhang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Dongqin Wang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China
| | - Yinlong Huang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical College, Bengbu, 233000, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, 233030, China.
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, China.
| |
Collapse
|
6
|
Zhu Q, Rao B, Chen Y, Jia P, Wang X, Zhang B, Wang L, Zhao W, Hu C, Tang M, Yu K, Chen W, Pan L, Xu Y, Luo H, Wang K, Li B, Shi H. In silico development and in vitro validation of a novel five-gene signature for prognostic prediction in colon cancer. Am J Cancer Res 2023; 13:45-65. [PMID: 36777511 PMCID: PMC9906087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/24/2022] [Indexed: 02/14/2023] Open
Abstract
Colon cancer is one of the most common cancers in digestive system, and its prognosis remains unsatisfactory. Therefore, this study aimed to identify gene signatures that could effectively predict the prognosis of colon cancer patients by examining the data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. LASSO-Cox regression analysis generated a five-gene signature (DCBLD2, RAB11FIP1, CTLA4, HOXC6 and KRT6A) that was associated with patient survival in the TCGA cohort. The prognostic value of this gene signature was further validated in two independent GEO datasets. GO enrichment revealed that the function of this gene signature was mainly associated with extracellular matrix organization, collagen-containing extracellular matrix, and extracellular matrix structural constituent. Moreover, a nomogram was established to facilitate the clinical application of this signature. The relationships among the gene signature, mutational landscape and immune infiltration cells were also investigated. Importantly, this gene signature also reliably predicted the overall survival in IMvigor210 anti-PD-L1 cohort. In addition to the bioinformatics study, we also conducted a series of in vitro experiments to demonstrate the effect of the signature genes on the proliferation, migration, and invasion of colon cancer cells. Collectively, our data demonstrated that this five-gene signature might serve as a promising prognostic biomarker and shed light on the development of personalized treatment in colon cancer patients.
Collapse
Affiliation(s)
- Qiankun Zhu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Benqiang Rao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Yongbing Chen
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Pingping Jia
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Xin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Bingdong Zhang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Lin Wang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Wanni Zhao
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Chunlei Hu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Meng Tang
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Kaiying Yu
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| | - Wei Chen
- Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China,Department of Intensive Care Unit, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China
| | - Lei Pan
- Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China,Department of Respiratory and Critical Care, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China
| | - Yu Xu
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical UniversityKunming 650032, Yunnan, The People’s Republic of China
| | - Huayou Luo
- Department of General Surgery, The First Affiliated Hospital of Kunming Medical UniversityKunming 650032, Yunnan, The People’s Republic of China
| | - Kunhua Wang
- Yunnan UniversityKunming 650091, Yunnan, The People’s Republic of China
| | - Bo Li
- Department of General Surgery, The Affiliated Hospital of Yunnan UniversityKunming 650091, Yunnan, The People’s Republic of China
| | - Hanping Shi
- Department of General Surgery, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical UniversityBeijing 100038, The People’s Republic of China,Beijing International Science and Technology Cooperation Base for Cancer Metabolism and NutritionBeijing 100038, The People’s Republic of China,Key Laboratory of Cancer FSMP for State Market RegulationBeijing 100038, The People’s Republic of China,Ninth School of Clinical Medicine, Peking UniversityBeijing 100038, The People’s Republic of China
| |
Collapse
|
7
|
Barbero G, Castro MV, Quezada MJ, Lopez-Bergami P. Bioinformatic analysis identifies epidermal development genes that contribute to melanoma progression. Med Oncol 2022; 39:141. [PMID: 35834068 DOI: 10.1007/s12032-022-01734-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/12/2022] [Indexed: 06/15/2023]
Abstract
Several diagnostic and prognostic markers for melanoma have been identified in last few years. However, their actual contribution to melanoma progression have not been investigated in detail. This study was aimed to identify genes, biological processes, and signaling pathways implicated in melanoma progression by applying bioinformatics analysis. We identified nine differentially expressed genes (DEGs) (IL36RN, KRT6A, KRT6B, KRT16, S100A7, SPRR1A, SPRR1B, SPRR2B, and KLK7) that were upregulated in primary melanoma compared with metastatic melanoma in all five datasets analyzed. All these genes except IL36RN, both form a protein-protein interaction network and have cellular functions associated with constitutive processes of keratinocytes. Thus, they were generically termed Epidermal Development and Cornification (EDC) genes. The differential expression of these genes in primary and metastatic melanoma was confirmed in the TCGA-SKCM cohort. High expression of the EDC genes correlated with reduced tumor thickness in primary melanoma and shorter survival in metastatic melanoma. Analysis of DEGs from primary melanoma patients displaying high or low expression of all eight EDC revealed that the upregulated genes are enriched in biological process related to cell migration, extracellular matrix organization, invasion, and Epithelial-Mesenchymal Transition. Further analysis of enriched curated oncogenic genesets together with RPPA data of phosphorylated proteins revealed the activation of MEK, ATF2, and EGFR pathways in tumors displaying high expression of EDC genes. Thus, EDC genes may contribute to melanoma progression by promoting the activation of MEK, ATF2, and EGFR pathways together with biological processes associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Gastón Barbero
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405, Buenos Aires, Argentina
| | - María Victoria Castro
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405, Buenos Aires, Argentina
| | - María Josefina Quezada
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405, Buenos Aires, Argentina
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405, Buenos Aires, Argentina.
| |
Collapse
|
8
|
A Novel 2-Metagene Signature to Identify High-Risk HNSCC Patients amongst Those Who Are Clinically at Intermediate Risk and Are Treated with PORT. Cancers (Basel) 2022; 14:cancers14123031. [PMID: 35740697 PMCID: PMC9221048 DOI: 10.3390/cancers14123031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: Patients with locally advanced head and neck squamous cell carcinoma (HNSCC) who are biologically at high risk for the development of loco−regional recurrences after postoperative radiotherapy (PORT) but at intermediate risk according to clinical risk factors may benefit from additional concurrent chemotherapy. In this matched-pair study, we aimed to identify a corresponding predictive gene signature. (2) Methods: Gene expression analysis was performed on a multicenter retrospective cohort of 221 patients that were treated with postoperative radiochemotherapy (PORT-C) and 283 patients who were treated with PORT alone. Propensity score analysis was used to identify matched patient pairs from both cohorts. From differential gene expression analysis and Cox regression, a predictive gene signature was identified. (3) Results: 108 matched patient pairs were selected. We identified a 2-metagene signature that stratified patients into risk groups in both cohorts. The comparison of the high-risk patients between the two types of treatment showed higher loco−regional control (LRC) after treatment with PORT-C (p < 0.001), which was confirmed by a significant interaction term in Cox regression (p = 0.027), i.e., the 2-metagene signature was indicative for the type of treatment. (4) Conclusion: We have identified a novel gene signature that may be helpful to identify patients with high-risk HNSCC amongst those at intermediate clinical risk treated with PORT, who may benefit from additional concurrent chemotherapy.
Collapse
|
9
|
Mukhtar U, Goyal A, Luthra-Guptasarma M, Gauba K, Kapur A, Thakur AK. Label-free quantitative proteomics reveals molecular correlates of altered biomechanical properties in molar incisor hypomineralization (MIH): an in vitro study. Eur Arch Paediatr Dent 2022; 23:179-191. [PMID: 35013981 DOI: 10.1007/s40368-021-00687-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/13/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE Molar-incisor hypomineralization (MIH) is a qualitative developmental defect of enamel that affects first permanent molars with or without affecting permanent incisors. We aimed to carry out a quantitative proteomics-based study to compare and evaluate proteins in sound and MIH-affected enamel. MATERIALS AND METHODS Ten blocks each of the MIH-affected enamel and sound enamel were processed and prepared for LC-MS/MS analysis. Label-free quantitation was carried out to evaluate the differentially expressed proteins in the two groups of samples. RESULTS A significant increase in the number of proteins in MIH-affected enamel (50.3 ± 29.6) was observed compared to the sound enamel (21.4 ± 3.2). While proteins like collagens, α1-anti-trypsin, kallikrein-4 (KLK4), matrix metalloprotease-20 (MMP-20), alpha-2-macroglobulin, and alpha-2-HS-glycoprotein were upregulated in sound enamel, there was over-expression of albumin, calcium-binding proteins, anti-thrombin III, and dentin sialophosphoprotein (DSPP), along with proteins implicated in stress response and inflammatory processes in MIH. CONCLUSION We propose that altered biomechanical properties of the enamel in MIH samples arise due to (i) down-regulation of proteins contributing to collagen biosynthesis and fibril formation; (ii) an overall imbalance in required levels of proteases (KLK4 and MMP-20) and anti-proteases (anti-thrombin-III which inhibits KLK-4), essential for optimal mineralization; (iii) very low levels of alpha-2-macroglobulin with important consequences in enamel mineralization and amelogenesis; and (iv) increased albumin in MIH, preventing proper growth of hydroxyapatite crystals. Increased inflammatory component was also seen in MIH; however, whether inflammation is a cause or consequence of the poor mineralization process needs to be assessed.
Collapse
Affiliation(s)
- U Mukhtar
- Oral Health Sciences Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - A Goyal
- Oral Health Sciences Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - M Luthra-Guptasarma
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - K Gauba
- Oral Health Sciences Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - A Kapur
- Oral Health Sciences Center, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - A K Thakur
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
10
|
Zhou J, Jiang G, Xu E, Zhou J, Liu L, Yang Q. Identification of SRXN1 and KRT6A as Key Genes in Smoking-Related Non-Small-Cell Lung Cancer Through Bioinformatics and Functional Analyses. Front Oncol 2022; 11:810301. [PMID: 35071014 PMCID: PMC8767109 DOI: 10.3389/fonc.2021.810301] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/14/2021] [Indexed: 11/15/2022] Open
Abstract
Background Lung cancer is the leading cause of cancer-related mortality worldwide. Although cigarette smoking is an established risk factor for lung cancer, few reliable smoking-related biomarkers for non-small-cell lung cancer (NSCLC) are available. An improved understanding of these biomarkers would further the development of new biomarker-targeted therapies and lead to improvements in overall patient survival. Methods We performed bioinformatic analysis to screened potential target genes, then quantitative PCR, western, siRNA, CCK-8, flow cytometry, tumorigenicity assays in nude mice were performed to validated the function. Results In this study, we identified 83 smoking-related genes (SRGs) based on an integration analysis of two Gene Expression Omnibus (GEO) datasets, and 27 hub SRGs with potential carcinogenic effects by analyzing a dataset of smokers with NSCLC in The Cancer Genome Atlas (TCGA) database. A survival analysis revealed three genes with potential prognostic value, namely SRXN1, KRT6A and JAKMIP3. A univariate Cox analysis revealed significant associations of elevated SRXN1 and KRT6A expression with prognosis. A receiver operating characteristic (ROC) curve analysis indicated the high diagnostic value of SRXN1 and KRT6A for smoking and cancer. Quantitative PCR and western blotting validated the increased expression of SRXN1 and KRT6A mRNA and protein, respectively, in lung cancer cell lines and NSCLC tissues. In patients with NSCLC, SRXN1 and KRT6A expression was associated with the tumor–node–metastasis (TNM) stage, presence of metastasis, history of smoking and daily smoking consumption. Furthermore, inhibition of SRXN1 or KRT6A suppressed viability and enhanced apoptosis in the A549 human lung carcinoma cell line. Tumorigenicity assays in nude mice confirmed that the siRNA-mediated downregulation of SRXN1 and KRT6A expression inhibited tumor growth in vivo. Conclusions In summary, SRXN1 and KRT6A act as oncogenes in NSCLC and might be potential biomarkers of smoking exposure and the early diagnosis and prognosis of NSCLC in smokers, which is vital for lung cancer therapy.
Collapse
Affiliation(s)
- Jiazhen Zhou
- The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Guanqing Jiang
- The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Enwu Xu
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, People's Liberation Army (PLA), Guangzhou, China
| | - Jiaxin Zhou
- The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Lili Liu
- Guangdong Provincial Key Laboratory of Occupational Disease Prevention and Treatment, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, China
| | - Qiaoyuan Yang
- The Institute for Chemical Carcinogenesis, School of Public Health, Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Lung adenocarcinoma and lung squamous cell carcinoma cancer classification, biomarker identification, and gene expression analysis using overlapping feature selection methods. Sci Rep 2021; 11:13323. [PMID: 34172784 PMCID: PMC8233431 DOI: 10.1038/s41598-021-92725-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is one of the deadliest cancers in the world. Two of the most common subtypes, lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), have drastically different biological signatures, yet they are often treated similarly and classified together as non-small cell lung cancer (NSCLC). LUAD and LUSC biomarkers are scarce, and their distinct biological mechanisms have yet to be elucidated. To detect biologically relevant markers, many studies have attempted to improve traditional machine learning algorithms or develop novel algorithms for biomarker discovery. However, few have used overlapping machine learning or feature selection methods for cancer classification, biomarker identification, or gene expression analysis. This study proposes to use overlapping traditional feature selection or feature reduction techniques for cancer classification and biomarker discovery. The genes selected by the overlapping method were then verified using random forest. The classification statistics of the overlapping method were compared to those of the traditional feature selection methods. The identified biomarkers were validated in an external dataset using AUC and ROC analysis. Gene expression analysis was then performed to further investigate biological differences between LUAD and LUSC. Overall, our method achieved classification results comparable to, if not better than, the traditional algorithms. It also identified multiple known biomarkers, and five potentially novel biomarkers with high discriminating values between LUAD and LUSC. Many of the biomarkers also exhibit significant prognostic potential, particularly in LUAD. Our study also unraveled distinct biological pathways between LUAD and LUSC.
Collapse
|
12
|
Che D, Wang M, Sun J, Li B, Xu T, Lu Y, Pan H, Lu Z, Gu X. KRT6A Promotes Lung Cancer Cell Growth and Invasion Through MYC-Regulated Pentose Phosphate Pathway. Front Cell Dev Biol 2021; 9:694071. [PMID: 34235156 PMCID: PMC8255478 DOI: 10.3389/fcell.2021.694071] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/30/2021] [Indexed: 12/25/2022] Open
Abstract
Keratin 6A (KRT6A) belongs to the keratin protein family which is a critical component of cytoskeleton in mammalian cells. Although KRT6A upregulation in non-small cell lung cancer (NSCLC) has been reported, the regulatory mechanism and functional role of KRT6A in NSCLC development have been less well investigated. In this study, KRT6A was confirmed to be highly expressed in NSCLC tissue samples, and its high expression correlated with poor patient prognosis. Furthermore, overexpression of KRT6A promotes NSCLC cell proliferation and invasion. Mechanistically, KRT6A overexpression is sufficient to upregulate glucose-6-phosphate dehydrogenase (G6PD) levels and increase the pentose phosphate pathway flux, an essential metabolic pathway to support cancer cell growth and invasion. In addition, we discovered that lysine-specific demethylase 1A (LSD1) functions upstream to promote KRT6A gene expression. We also found that the MYC family members c-MYC/MYCN are involved in KRT6A-induced G6PD upregulation. Therefore, this study reveals an underappreciated mechanism that KRT6A acts downstream of LSD1 and functions as a pivotal driver for NSCLC progression by upregulating G6PD through the MYC signaling pathway. Together, KRT6A and LSD1 may serve as potential prognostic indictors and therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Di Che
- Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mingshuo Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Juan Sun
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bo Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tao Xu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuxiong Lu
- Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Haiyan Pan
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China
| | - Zhaoliang Lu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoqiong Gu
- Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Yu P, Tong L, Song Y, Qu H, Chen Y. Systematic profiling of invasion-related gene signature predicts prognostic features of lung adenocarcinoma. J Cell Mol Med 2021; 25:6388-6402. [PMID: 34060213 PMCID: PMC8256358 DOI: 10.1111/jcmm.16619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Due to the high heterogeneity of lung adenocarcinoma (LUAD), molecular subtype based on gene expression profiles is of great significance for diagnosis and prognosis prediction in patients with LUAD. Invasion-related genes were obtained from the CancerSEA database, and LUAD expression profiles were downloaded from The Cancer Genome Atlas. The ConsensusClusterPlus was used to obtain molecular subtypes based on invasion-related genes. The limma software package was used to identify differentially expressed genes (DEGs). A multi-gene risk model was constructed by Lasso-Cox analysis. A nomogram was also constructed based on risk scores and meaningful clinical features. 3 subtypes (C1, C2 and C3) based on the expression of 97 invasion-related genes were obtained. C3 had the worst prognosis. A total of 669 DEGs were identified among the subtypes. Pathway enrichment analysis results showed that the DEGs were mainly enriched in the cell cycle, DNA replication, the p53 signalling pathway and other tumour-related pathways. A 5-gene signature (KRT6A, MELTF, IRX5, MS4A1 and CRTAC1) was identified by using Lasso-Cox analysis. The training, validation and external independent cohorts proved that the model was robust and had better prediction ability than other lung cancer models. The gene expression results showed that the expression levels of MS4A1 and KRT6A in tumour tissues were higher than in normal tissues, while CRTAC1 expression in tumour tissues was lower than in normal tissues. The 5-gene signature prognostic stratification system based on invasion-related genes could be used to assess prognostic risk in patients with LUAD.
Collapse
Affiliation(s)
- Ping Yu
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
- Liaoning Province Clinical Research Center for CancerShenyangChina
| | - Linlin Tong
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
- Liaoning Province Clinical Research Center for CancerShenyangChina
| | - Yujia Song
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangChina
| | - Hui Qu
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangChina
| | - Ying Chen
- Department of Medical OncologyThe First Hospital of China Medical UniversityShenyangChina
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning ProvinceThe First Hospital of China Medical UniversityShenyangChina
- Liaoning Province Clinical Research Center for CancerShenyangChina
| |
Collapse
|
14
|
Transcript levels of keratin 1/5/6/14/15/16/17 as potential prognostic indicators in melanoma patients. Sci Rep 2021; 11:1023. [PMID: 33441834 PMCID: PMC7806772 DOI: 10.1038/s41598-020-80336-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keratins (KRTs), the intermediate filament-forming proteins of epithelial cells, are extensively used as diagnostic biomarkers in cancers and associated with tumorigenesis and metastasis in multiple cancers. However, the diverse expression patterns and prognostic values of KRTs in melanoma have yet to be elucidated. In the current study, we examined the transcriptional and clinical data of KRTs in patients with melanoma from GEO, TCGA, ONCOMINE, GEPIA, cBioPortal, TIMER and TISIDB databases. We found that the mRNA levels of KRT1/2/5/6/8/10/14/15/16/17 were significantly differential expressed between primary melanoma and metastatic melanoma. The expression levels of KRT1/2/5/6/10/14/15/16/17 were correlated with advanced tumor stage. Survival analysis revealed that the high transcription levels of KRT1/5/6/14/15/16/17 were associated with low overall survival in melanoma patients. GSEA analysis indicated that the most involved hallmarks pathways were P53 pathway, KRAS signaling, estrogen response early and estrogen response late. Furthermore, we found some correlations among the expression of KRTs and the infiltration of immune cells. Our study may provide novel insights for the selection of prognostic biomarkers for melanoma.
Collapse
|
15
|
Yang B, Zhang W, Zhang M, Wang X, Peng S, Zhang R. KRT6A Promotes EMT and Cancer Stem Cell Transformation in Lung Adenocarcinoma. Technol Cancer Res Treat 2021; 19:1533033820921248. [PMID: 32329414 PMCID: PMC7225834 DOI: 10.1177/1533033820921248] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: Keratin 6A is a type II cytokeratin which is important in forming nail bed, filiform papillae, the epithelial lining of oral mucosa, and esophagus; recently, keratin 6A was found hyperexpressed in different types of cancer. But, the biological function of keratin 6A in lung adenocarcinoma still remains unclear. Therefore, in current study, we investigated the biological role of keratin 6A in lung adenocarcinoma. Methods: By utilizing The Cancer Genome Atlas database, we investigated the expression profile of keratin 6A and its relationship with other clinical parameters in lung adenocarcinoma. The biological function of keratin 6A in lung adenocarcinoma was also investigated by using A549 and PC-9 lung cancer cell lines in vitro. Results: Our data indicate that, compared with normal lung tissue samples, keratin 6A was hyperexpressed in lung adenocarcinoma. Moreover, keratin 6A hyperexpression was positively correlated with lymph node positive and aggressive tumor T stage. Keratin 6A knockdown inhibited the cell proliferation, migration, and colony formation ability but not cell death in lung adenocarcinoma cells. In addition, we found keratin 6A exerted its phenotype via promoting cancer stem cells (CXCR4high/CD133high) transformation and epithelial–mesenchymal transition. Conclusion: In conclusion, current study suggests that hyperexpressed keratin 6A in lung adenocarcinoma promotes lung cancer proliferation and metastasis via epithelial–mesenchymal transition and cancer stem cells transformation.
Collapse
Affiliation(s)
- Bin Yang
- Department of Thoracic Surgery (III), Shanxi Cancer Hospital, Taiyuan, People's Republic of China
| | - Wei Zhang
- Department of General Surgery (II), The 6th Division Hospital of Xinjiang Corps, Taiyuan, People's Republic of China
| | - Mengmeng Zhang
- Department of Gastrointestinal Surgery, Laboratory of Surgical Oncology, Peking University People's Hospital, Taiyuan, People's Republic of China
| | - Xuhong Wang
- Department of Head and Neck Surgery, Shanxi Cancer Hospital, Taiyuan, People's Republic of China
| | - Shengzu Peng
- Department of Thoracic Surgery (III), Shanxi Cancer Hospital, Taiyuan, People's Republic of China
| | - Rongsheng Zhang
- Department of Thoracic Surgery (III), Shanxi Cancer Hospital, Taiyuan, People's Republic of China
| |
Collapse
|
16
|
Zhang J, Sun H, Liu S, Huang W, Gu J, Zhao Z, Qin H, Luo L, Yang J, Fang Y, Ge J, Ni B, Wang H. Alteration of tumor-associated macrophage subtypes mediated by KRT6A in pancreatic ductal adenocarcinoma. Aging (Albany NY) 2020; 12:23217-23232. [PMID: 33221741 PMCID: PMC7746340 DOI: 10.18632/aging.104091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is severely affecting the health and lives of patients. Clarifying the composition and regulatory factors of tumor immune microenvironment (TIME) is helpful for the treatment of PDAC. We analyzed the unique TIMEs and gene expression patterns between PDAC and adjacent normal tissue (ANT) using Gene Expression Omnibus (GEO) to find new immunotherapy targets. The Cancer Genome Atlas (TCGA) datasets were used to elucidate the possible mechanism of which tumor-associated macrophages (TAMs) changed in PDAC. We found that the composition of TAMs subtypes, including M0, M1, and M2, was different between PDAC and ANT, which was validated in recently published single-cell RNA-seq data. Many immune cells interacted with each other to affect the TIME. There were many DEGs enriched in some pathways that could potentially change the immune cell composition. KRT6A was found to be a DEG between PDAC and ANT that overlapped with DEGs between the M0-high group and the M0-low group in TCGA datasets, and it might alter and regulate TAMs via a collection of genes including COL5A2, COL1A2, MIR3606, SPARC, and COL6A3. TAMs, which could be a target of immunotherapy, might be influenced by genes through KRT6A and indicate an undesirable prognosis in PDAC.
Collapse
Affiliation(s)
- Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Hui Sun
- Department of Rheumatology, First Affiliated Hospital of Third Military Medical University, Chongqing 400038, P R China
| | - Songsong Liu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Wenjie Huang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Jianyou Gu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Zhiping Zhao
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Huan Qin
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Liwen Luo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Jiali Yang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Yongfei Fang
- Department of Rheumatology, First Affiliated Hospital of Third Military Medical University, Chongqing 400038, P R China
| | - Jiayun Ge
- Hepatopancreatobiliary Surgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming 650101, Yunnan Province, P R China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P R China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, P R China
- Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, P R China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| |
Collapse
|
17
|
Chandrashekar DS, Chakravarthi BVSK, Robinson AD, Anderson JC, Agarwal S, Balasubramanya SAH, Eich ML, Bajpai AK, Davuluri S, Guru MS, Guru AS, Naik G, Della Manna DL, Acharya KK, Carskadon S, Manne U, Crossman DK, Ferguson JE, Grizzle WE, Palanisamy N, Willey CD, Crowley MR, Netto GJ, Yang ES, Varambally S, Sonpavde G. Therapeutically actionable PAK4 is amplified, overexpressed, and involved in bladder cancer progression. Oncogene 2020; 39:4077-4091. [PMID: 32231273 DOI: 10.1038/s41388-020-1275-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/30/2022]
Abstract
Muscle-invasive bladder carcinomas (MIBCs) are aggressive genitourinary malignancies. Metastatic urothelial carcinoma of the bladder is generally incurable by current chemotherapy and leads to early mortality. Recent studies have identified molecular subtypes of MIBCs with different sensitivities to frontline therapy, suggesting tumor heterogeneity. We have performed multi-omic profiling of the kinome in bladder cancer patients with the goal of identify therapeutic targets. Our analyses revealed amplification, overexpression, and elevated kinase activity of P21 (RAC1) activated kinase 4 (PAK4) in a subset of Bladder cancer (BLCA). Using bladder cancer cells, we confirmed the role of PAK4 in BLCA cell proliferation and invasion. Furthermore, we observed that a PAK4 inhibitor was effective in curtailing growth of BLCA cells. Transcriptomic analyses identified elevated expression of another kinase, protein tyrosine kinase 6 (PTK6), upon treatment with a PAK4 inhibitor and RNA interference of PAK4. Treatment with a combination of kinase inhibitors (vandetanib and dasatinib) showed enhanced sensitivity compared with either drug alone. Thus, PAK4 may be therapeutically actionable for a subset of MIBC patients with amplified and/or overexpressed PAK4 in their tumors. Our results also indicate that combined inhibition of PAK4 and PTK6 may overcome resistance to PAK4. These observations warrant clinical investigations with selected BLCA patients.
Collapse
Affiliation(s)
| | | | - Alyncia D Robinson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua C Anderson
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Marie-Lisa Eich
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Maya S Guru
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Arjun S Guru
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gurudatta Naik
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Deborah L Della Manna
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kshitish K Acharya
- Shodhaka Life Sciences Private Limited, Bengaluru, India.,Institute of Bioinformatics and Applied Biotechnology (IBAB), Biotech Park, Electronic City, Bengaluru, 560100, Karnataka, India
| | - Shannon Carskadon
- Vattikuti Urology Institute, Department of Urology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James E Ferguson
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William E Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nallasivam Palanisamy
- Vattikuti Urology Institute, Department of Urology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Christopher D Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael R Crowley
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, USA.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA. .,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA. .,Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Guru Sonpavde
- Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
18
|
Hu HB, Yang XP, Zhou PX, Yang XA, Yin B. High expression of keratin 6C is associated with poor prognosis and accelerates cancer proliferation and migration by modulating epithelial-mesenchymal transition in lung adenocarcinoma. Genes Genomics 2019; 42:179-188. [PMID: 31768767 DOI: 10.1007/s13258-019-00889-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a more frequent subtype of lung cancer and most cases are discovered in the late stages. The proliferation and metastasis of LUAD are pivotal for disease progression. Despite unremitting deeper understanding of LUAD biology, the mechanisms involved in the proliferation and metastasis of LUAD remain unclear. The objective of our article was to inquiry the expression and the function of keratin 6C (KRT6C) in LUAD cells. METHODS First, the expression level and prognostic value of KRT6C in LUAD tissues were analyzed on the basis of the data acquired from TCGA database. Through qRT-PCR, the expression level of KRT6C on LUAD cell lines (A549, H1299, PC-9) and human normal lung cell line MRC-5 was tested. After that, CCK8 and colony formation assays was utilized to detect cell proliferation. In addition, to explore the influence of KRT6C on LUAD migration and invasion ability, scratch wound healing and transwell assays were utilized. Through western blotting, the protein expression levels of KRT6C, PCNA, E-cadherin, N-cadherin, Snail and Vimentin were detected. RESULTS The outcomes revealed that KRT6C was highly expressed in LUAD tissues and cell lines. Besides, elevated level of KRT6C was related to worse prognosis in LUAD patients. Ablation of KRT6C restrained proliferation, migration and invasion of A549 cells. KRT6C deficiency augmented the expression of E-cadherin as well as reduced the expression of N-cadherin, Snail and Vimentin. CONCLUSION Above all, these consequences indicated that depletion of KRT6C suppressed A549 cell proliferation, migration and invasion, which might be achieved by regulating EMT. In general, KRT6C is identified as a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Hai-Bo Hu
- Department of Respiration, Qingdao TCM Hospital, No. 4 Ren Min Road, Qingdao, 266033, Shandong, People's Republic of China
| | - Xiao-Ping Yang
- Department of Respiration, Qingdao TCM Hospital, No. 4 Ren Min Road, Qingdao, 266033, Shandong, People's Republic of China
| | - Pei-Xia Zhou
- Department of Respiration, Qingdao TCM Hospital, No. 4 Ren Min Road, Qingdao, 266033, Shandong, People's Republic of China
| | - Xin-Ai Yang
- Department of Respiration, Qingdao TCM Hospital, No. 4 Ren Min Road, Qingdao, 266033, Shandong, People's Republic of China
| | - Bin Yin
- Department of Respiration, Qingdao TCM Hospital, No. 4 Ren Min Road, Qingdao, 266033, Shandong, People's Republic of China.
| |
Collapse
|