1
|
Cheng HS, Tey YH, Hu SY, Yeo AYN, Ngo ZH, Kim JHS, Tan NS. Advancements and Challenges in Modeling Mechanobiology in Intestinal Host-Microbiota Interaction. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40382722 DOI: 10.1021/acsami.4c20961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
The gastrointestinal tract is a dynamic biomechanical environment where physical forces, cellular processes, and microbial interactions converge to shape the gut health and disease. In this review, we examine the unique mechanical properties of the gut, including peristalsis, viscoelasticity, shear stress, and tissue stiffness, and their roles in modulating host mechanosignaling and microbial behavior under physiological and pathological conditions. We discuss how these mechanical forces regulate gut epithelial integrity, immune responses, and microbial colonization, leading to distinct ecological niches across different intestinal segments. Furthermore, we highlight recent advancements in 3D culture systems and gut-on-a-chip models that accurately recapitulate the complex interplay between biomechanics and gut microbiota. By elucidating the intricate relationship between mechanobiology and gut function, this review underscores the potential for mechanotherapeutic strategies to modulate host-microbe interactions, offering promising avenues for the prevention and treatment of disorders such as inflammatory bowel disease, irritable bowel syndrome, and colorectal cancer.
Collapse
Affiliation(s)
- Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
| | - Yee Han Tey
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Si Yuan Hu
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Alethea Yen Ning Yeo
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Zong Heng Ngo
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| | - Joseph Han Sol Kim
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore 637551, Singapore
| |
Collapse
|
2
|
Di J, Zhao Z, Xia M, Gao K, Chai K, Zhu B, Sun W, Zhang Y, Zheng J, Liu Y. Rap2B drives tumorigenesis and progression of colorectal cancer through intestinal cytoskeleton remodeling. Cell Death Dis 2025; 16:290. [PMID: 40223002 PMCID: PMC11994759 DOI: 10.1038/s41419-025-07627-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Ras family protein plays a key role in transducing signals involved in cytoskeletal remodeling and cell adhesion, which are particularly important in the development of colorectal cancer (CRC). While Rap2B, a member of the Ras superfamily, has been linked to cancer malignancy in vitro, its exact role in tumorigenesis remains unclear. In this study, we demonstrated that intestine-specific knockout of Rap2B suppresses the initiation and progression of CRC. Mechanistically, Rap2B interacts with plectin and enhances its expression, which in turn inhibits plectin-mediated F-actin assembly. Deletion of Rap2B resulted in a remodeling of the intestinal cytoskeleton, leading to reduced tumorigenesis and diminished metastatic potential. Clinically, there is a positive correlation between the expression levels of Rap2B and plectin in human CRC tissues, and higher levels of Rap2B and plectin predicting poorer clinical outcome in CRC patients. These findings underscore a critical role of Rap2B in CRC progression and highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jiehui Di
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Zhongjun Zhao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyi Xia
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Keyu Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Keli Chai
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Bao Zhu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wanping Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Yanping Zhang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Yong Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu, 221002, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
3
|
Seo Y, Jang J, Ko KP, Zou G, Huang Y, Zhang S, Zhang J, Jun S, Chu W, Venkatesan V, Dhakshinamoorthy S, Park JI. Actin dysregulation induces immune evasion via oxidative stress-activated PD-L1 in gastric cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.629227. [PMID: 39763993 PMCID: PMC11702617 DOI: 10.1101/2024.12.18.629227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Diffuse gastric adenocarcinoma (DGAC) is an aggressive malignancy with limited therapeutic options, poor prognosis, and poorly understood biology. CRACD, an actin polymerization regulator, is often inactivated in gastric cancer, including DGAC. We found that genetic engineering of murine gastric organoids with Cracd ablation combined with Kras mutation and Trp53 loss induced aberrant cell plasticity, hyperproliferation, and hypermucinosis, the features that recapitulate DGAC transcriptional signatures. Notably, CRACD inactivation remodeled the immune landscape for immune evasion through PD-L1 enrichment in tumor cells. Mechanistically, CRACD loss disrupted actin dynamics, generating reactive oxygen species that activated HIF1α, which transactivated PD-L1 . Pharmacologic inhibition of HIF1α or PD-L1 restored immune surveillance and suppressed tumorigenesis. These findings reveal a novel role of actin homeostasis in limiting cell plasticity and immune evasion, position CRACD as a potential biomarker for stratifying patients with DGAC, and highlight HIF1α and PD-L1 as actionable therapeutic targets.
Collapse
|
4
|
Yin QZ, Liu YJ, Zhang Q, Xi SY, Yang TB, Li JP, Gao J. Overexpression of Basonuclin Zinc Finger Protein 2 in stromal cell is related to mesenchymal phenotype and immunosuppression of mucinous colorectal adenocarcinoma. Int Immunopharmacol 2024; 142:113184. [PMID: 39306894 DOI: 10.1016/j.intimp.2024.113184] [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: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Mucinous carcinoma (MC) is a distinct histologic subtype of colorectal cancer (CRC) that is less studied and associated with poor prognosis. This study aimed to identify MC-specific therapeutic targets and biomarkers to improve the prognosis of this aggressive disease. METHODS CRC samples from The Cancer Genome Atlas (TCGA) were categorized into MC and non-MC (NMC) groups based on histologic type. A multi-scale embedded gene co-expression network analysis (MEGENA) was constructed to identify gene modules associated with the MC group. The potential functions of Basonuclin Zinc Finger Protein 2 (BNC2) were further analyzed using the Biomarker Exploration for Solid Tumors (BEST) database. In vivo and in vitro experiments were conducted to validate the predicted results. RESULTS We identified the stromal component-related gene, BNC2, in the MC population. This gene is associated with a shorter progression-free interval (PFI) in CRC patients. BNC2 promotes FAP (encoding Fibroblast Activation Protein Alpha) transcription in cancer-associated fibroblasts (CAFs) and is involved in angiogenesis through two pathways. Additionally, BNC2 enhances tumor cell invasiveness in a CAF-dependent manner. Patients with high BNC2 expression benefited less from immunotherapy compared to those with low BNC2 expression. CONCLUSIONS Our study highlights the clinical importance of BNC2 in MC, and targeting BNC2 on stromal cells (fibroblasts and endothelial cells) may be an effective strategy for treating MC.
Collapse
Affiliation(s)
- Qing-Zhong Yin
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuan-Jie Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Qian Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Song-Yang Xi
- Zhenjiang Hospital of Chinese Traditional and Western Medicine, Zhenjiang, Jiangsu 212000, China
| | - Tian-Bao Yang
- Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jie-Pin Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Ju Gao
- The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, Jiangsu 225009, China; Northern Jiangsu People's Hospital, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
5
|
Seo Y, Zhang S, Jang J, Ko KP, Kim KB, Huang Y, Kim DW, Kim B, Zou G, Zhang J, Jun S, Chu W, Kirk NA, Hwang YE, Ban YH, Dhar SS, Chan JM, Lee MG, Rudin CM, Park KS, Park JI. Actin Dysregulation Induces Neuroendocrine Plasticity and Immune Evasion: A Vulnerability of Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.15.528365. [PMID: 36824957 PMCID: PMC9949038 DOI: 10.1101/2023.02.15.528365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Small cell lung cancer (SCLC) is aggressive with limited therapeutic options. Despite recent advances in targeted therapies and immunotherapies, therapy resistance is a recurring issue, which might be partly due to tumor cell plasticity, a change in cell fate. Nonetheless, the mechanisms underlying tumor cell plasticity and immune evasion in SCLC remain elusive. CRACD, a capping protein inhibitor that promotes actin polymerization, is frequently inactivated in SCLC. Cracd knockout (KO) transforms preneoplastic cells into SCLC tumor-like cells and promotes in vivo SCLC development driven by Rb1, Trp53, and Rbl2 triple KO. Cracd KO induces neuroendocrine (NE) plasticity and increases tumor cell heterogeneity of SCLC tumor cells via dysregulated NOTCH1 signaling by actin cytoskeleton disruption. CRACD depletion also reduces nuclear actin and induces EZH2-mediated H3K27 methylation. This nuclear event suppresses the MHC-I genes and thereby depletes intratumoral CD8+ T cells for accelerated SCLC tumorigenesis. Pharmacological blockade of EZH2 inhibits CRACD-negative SCLC tumorigenesis by restoring MHC-I expression and immune surveillance. Unsupervised single-cell transcriptomics identifies SCLC patient tumors with concomitant inactivation of CRACD and downregulated MHC-I pathway. This study defines CRACD, an actin regulator, as a tumor suppressor that limits cell plasticity and immune evasion and proposes EZH2 blockade as a viable therapeutic option for CRACD-negative SCLC.
Collapse
Affiliation(s)
- Yoojeong Seo
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinho Jang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dong-Wook Kim
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wonhong Chu
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicole A. Kirk
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ye Eun Hwang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Young Ho Ban
- Hamatovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Shilpa S. Dhar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph M. Chan
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Min Gyu Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
6
|
Peng C, Lu W, An R, Li X, Sun C, Fang Y. Resistant Starch Nanoparticles Induce Colitis through Lysosomal Exocytosis in Mice. ACS NANO 2024; 18:30749-30760. [PMID: 39442088 DOI: 10.1021/acsnano.4c10481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Resistant starch (RS) is present in various natural and processed foods as well as medications. It has garnered significant attention from both scientists and consumers due to its notable health benefits. However, there is a limited understanding of how RS particles are absorbed at the cellular level and their metabolic behavior, resulting in a lack of clarity regarding the intestinal safety implications of prolonged RS exposure. Here, we demonstrate that rice-derived RS nanoparticles (RSNs) can lead to colitis in mice by triggering lysosomal exocytosis. The research shows that RSNs enter the cells through macropinocytosis and clathrin- and caveolin-mediated endocytosis and activate TRPML1 thereafter, causing the release of lysosomal calcium ions. This, in turn, triggered the TFEB signaling pathway and thus upregulated the lysosomal exocytosis level, leading to lysosomal enzymes to be released to the intestinal lumen. As a result, a decreased number of intestinal goblet cells, diminished tight junction protein expression, and imbalanced intestinal flora in mice were observed. These damages to the intestinal barrier ultimately led to the occurrence of colitis. Our study offers important insights into the cellular bioeffects and detrimental effects on intestinal health caused by RS particles and emphasizes the need to re-evaluate the safety of long-term RS consumption.
Collapse
Affiliation(s)
- Chenglu Peng
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Lu
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ran An
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoyang Li
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Cuixia Sun
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yapeng Fang
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
7
|
López-Valverde L, Vázquez-Mosquera ME, Colón-Mejeras C, Bravo SB, Barbosa-Gouveia S, Álvarez JV, Sánchez-Martínez R, López-Mendoza M, López-Rodríguez M, Villacorta-Argüelles E, Goicoechea-Diezhandino MA, Guerrero-Márquez FJ, Ortolano S, Leao-Teles E, Hermida-Ameijeiras Á, Couce ML. Characterization of the plasma proteomic profile of Fabry disease: Potential sex- and clinical phenotype-specific biomarkers. Transl Res 2024; 269:47-63. [PMID: 38395389 DOI: 10.1016/j.trsl.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/25/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
Fabry disease (FD) is a X-linked rare lysosomal storage disorder caused by deficient α-galactosidase A (α-GalA) activity. Early diagnosis and the prediction of disease course are complicated by the clinical heterogeneity of FD, as well as by the frequently inconclusive biochemical and genetic test results that do not correlate with clinical course. We sought to identify potential biomarkers of FD to better understand the underlying pathophysiology and clinical phenotypes. We compared the plasma proteomes of 50 FD patients and 50 matched healthy controls using DDA and SWATH-MS. The >30 proteins that were differentially expressed between the 2 groups included proteins implicated in processes such as inflammation, heme and haemoglobin metabolism, oxidative stress, coagulation, complement cascade, glucose and lipid metabolism, and glycocalyx formation. Stratification by sex revealed that certain proteins were differentially expressed in a sex-dependent manner. Apolipoprotein A-IV was upregulated in FD patients with complications, especially those with chronic kidney disease, and apolipoprotein C-III and fetuin-A were identified as possible markers of FD with left ventricular hypertrophy. All these proteins had a greater capacity to identify the presence of complications in FD patients than lyso-GB3, with apolipoprotein A-IV standing out as being more sensitive and effective in differentiating the presence and absence of chronic kidney disease in FD patients than renal markers such as creatinine, glomerular filtration rate and microalbuminuria. Identification of these potential biomarkers can help further our understanding of the pathophysiological processes that underlie the heterogeneous clinical manifestations associated with FD.
Collapse
Affiliation(s)
- Laura López-Valverde
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - María E Vázquez-Mosquera
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Cristóbal Colón-Mejeras
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Susana B Bravo
- Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Proteomic Platform, University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Sofía Barbosa-Gouveia
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - J Víctor Álvarez
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain
| | - Rosario Sánchez-Martínez
- Internal Medicine Department, Alicante General University Hospital-Alicante Institute of Health and Biomedical Research (ISABIAL), Pintor Baeza 12, Alicante 03010, Spain
| | - Manuel López-Mendoza
- Department of Nephrology, Hospital Universitario Virgen del Rocío, Manuel Siurot s/n, Sevilla 41013, Spain
| | - Mónica López-Rodríguez
- Internal Medicine Department, Hospital Universitario Ramón y Cajal, IRYCIS, Colmenar Viejo, Madrid 28034, Spain; Faculty of Medicine and Health Sciences, Universidad de Alcalá (UAH), Av. de Madrid, Alcalá de Henares 28871, Spain
| | - Eduardo Villacorta-Argüelles
- Department of Cardiology, Complejo Asistencial Universitario de Salamanca, P°. de San Vicente 58, Salamanca 37007, Spain
| | | | - Francisco J Guerrero-Márquez
- Department of Cardiology, Internal Medicine Service, Hospital de la Serranía, San Pedro, Ronda, Málaga 29400, Spain
| | - Saida Ortolano
- Rare Diseases and Pediatric Medicine Research Group, Galicia Sur Health Research Institute-SERGAS-UVIGO, Clara Campoamor 341, Vigo 36213, Spain
| | - Elisa Leao-Teles
- Centro de Referência de Doenças Hereditárias do Metabolismo, Centro Hospitalar Universitário de São João, Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Álvaro Hermida-Ameijeiras
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain.
| | - María L Couce
- Unit of Diagnosis and Treatment of Congenital Metabolic Diseases. RICORS-SAMID, CIBERER. University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain; Health Research Institute of Santiago de Compostela (IDIS), University Clinical Hospital of Santiago de Compostela, Choupana s/n, Santiago de Compostela, A Coruña 15706, Spain.
| |
Collapse
|
8
|
Kim B, Zhang S, Huang Y, Ko KP, Jung YS, Jang J, Zou G, Zhang J, Jun S, Kim KB, Park KS, Park JI. CRACD loss induces neuroendocrine cell plasticity of lung adenocarcinoma. Cell Rep 2024; 43:114286. [PMID: 38796854 PMCID: PMC11216895 DOI: 10.1016/j.celrep.2024.114286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 03/01/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Tumor cell plasticity contributes to intratumoral heterogeneity and therapy resistance. Through cell plasticity, some lung adenocarcinoma (LUAD) cells transform into neuroendocrine (NE) tumor cells. However, the mechanisms of NE cell plasticity remain unclear. CRACD (capping protein inhibiting regulator of actin dynamics), a capping protein inhibitor, is frequently inactivated in cancers. CRACD knockout (KO) is sufficient to de-repress NE-related gene expression in the pulmonary epithelium and LUAD cells. In LUAD mouse models, Cracd KO increases intratumoral heterogeneity with NE gene expression. Single-cell transcriptomic analysis showed that Cracd KO-induced NE cell plasticity is associated with cell de-differentiation and stemness-related pathway activation. The single-cell transcriptomic analysis of LUAD patient tumors recapitulates that the distinct LUAD NE cell cluster expressing NE genes is co-enriched with impaired actin remodeling. This study reveals the crucial role of CRACD in restricting NE cell plasticity that induces cell de-differentiation of LUAD.
Collapse
Affiliation(s)
- Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Youn-Sang Jung
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinho Jang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Zou G, Huang Y, Zhang S, Ko KP, Kim B, Zhang J, Venkatesan V, Pizzi MP, Fan Y, Jun S, Niu N, Wang H, Song S, Ajani JA, Park JI. E-cadherin loss drives diffuse-type gastric tumorigenesis via EZH2-mediated reprogramming. J Exp Med 2024; 221:e20230561. [PMID: 38411616 PMCID: PMC10899090 DOI: 10.1084/jem.20230561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/27/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Diffuse-type gastric adenocarcinoma (DGAC) is a deadly cancer often diagnosed late and resistant to treatment. While hereditary DGAC is linked to CDH1 mutations, the role of CDH1/E-cadherin inactivation in sporadic DGAC tumorigenesis remains elusive. We discovered CDH1 inactivation in a subset of DGAC patient tumors. Analyzing single-cell transcriptomes in malignant ascites, we identified two DGAC subtypes: DGAC1 (CDH1 loss) and DGAC2 (lacking immune response). DGAC1 displayed distinct molecular signatures, activated DGAC-related pathways, and an abundance of exhausted T cells in ascites. Genetically engineered murine gastric organoids showed that Cdh1 knock-out (KO), KrasG12D, Trp53 KO (EKP) accelerates tumorigenesis with immune evasion compared with KrasG12D, Trp53 KO (KP). We also identified EZH2 as a key mediator promoting CDH1 loss-associated DGAC tumorigenesis. These findings highlight DGAC's molecular diversity and potential for personalized treatment in CDH1-inactivated patients.
Collapse
Affiliation(s)
- Gengyi Zou
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuanjian Huang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shengzhe Zhang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kyung-Pil Ko
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bongjun Kim
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Zhang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vishwa Venkatesan
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa P. Pizzi
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yibo Fan
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sohee Jun
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Na Niu
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Huamin Wang
- Division of Pathology/Lab Medicine, Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumei Song
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A. Ajani
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jae-Il Park
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Osman AE, Alharbi S, Ahmed AA, Elbagir AA. Single nucleotide polymorphism within chromosome 8q24 is associated with prostate cancer development in Saudi Arabia. Asian J Urol 2024; 11:26-32. [PMID: 38312824 PMCID: PMC10837665 DOI: 10.1016/j.ajur.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Genome-wide association studies have demonstrated that single nucleotide polymorphisms (SNPs) are important risk factors for the development of prostate cancer (PCa). Preliminary studies have suggested that the incidence of PCa in Saudi males is low but is probably familial or genetically related. Methods To identify any possible association of SNP with PCa development in Saudi patients, we investigated a group of SNPs in Saudi PCa patients (n=85) and compared the outcomes to healthy normal controls (n=115) and nodular hyperplasia patients (n=120). DNA was extracted from paraffin-embedded formalin fixed tissue or whole blood from both patients' groups and healthy control group. A total of thirteen SNPs were genotyped using TaqMan® minor groove binder polymerase chain reaction assay. Results The rs16901979A, s629242T and rs1447295A alleles were found at significantly higher frequency in PCa patients than controls (p<0.05). The rs16901979 CA genotype was found at significantly greater frequency in PCa patients than in healthy controls (43% vs. 14%, odds ratio=4.6, p=0.0001) and benign hyperplasia group (43% vs. 25%, odds ratio=2.2, p=0.009). Conclusion Our study has highlighted the association of rs16901979 SNP with PCa in Saudi males. Such findings have important implications in the PCa diagnosis and in screening unaffected family members of Saudi patients.
Collapse
Affiliation(s)
- Awad Elsid Osman
- Pathology and Clinical Laboratory Management Department (PCLM), King Fahad Medical City, Riyadh, Saudi Arabia
| | - Sahar Alharbi
- Pathology and Clinical Laboratory Management Department (PCLM), King Fahad Medical City, Riyadh, Saudi Arabia
| | - Atif Ali Ahmed
- Department of Pathology and Laboratory Medicine, University of Missouri at Children's Mercy Hospital, Kansas City, MO, USA
| | - Asim Ali Elbagir
- Pathology and Clinical Laboratory Management Department (PCLM), King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Zou G, Huang Y, Zhang S, Ko KP, Kim B, Zhang J, Venkatesan V, Pizzi MP, Fan Y, Jun S, Niu N, Wang H, Song S, Ajani JA, Park JI. CDH1 loss promotes diffuse-type gastric cancer tumorigenesis via epigenetic reprogramming and immune evasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533976. [PMID: 36993615 PMCID: PMC10055394 DOI: 10.1101/2023.03.23.533976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Diffuse-type gastric adenocarcinoma (DGAC) is a deadly cancer often diagnosed late and resistant to treatment. While hereditary DGAC is linked to CDH1 gene mutations, causing E-Cadherin loss, its role in sporadic DGAC is unclear. We discovered CDH1 inactivation in a subset of DGAC patient tumors. Analyzing single-cell transcriptomes in malignant ascites, we identified two DGAC subtypes: DGAC1 (CDH1 loss) and DGAC2 (lacking immune response). DGAC1 displayed distinct molecular signatures, activated DGAC-related pathways, and an abundance of exhausted T cells in ascites. Genetically engineered murine gastric organoids showed that Cdh1 knock-out (KO), KrasG12D, Trp53 KO (EKP) accelerates tumorigenesis with immune evasion compared to KrasG12D, Trp53 KO (KP). We also identified EZH2 as a key mediator promoting CDH1 loss-associated DGAC tumorigenesis. These findings highlight DGAC's molecular diversity and potential for personalized treatment in CDH1-inactivated patients.
Collapse
Affiliation(s)
- Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vishwa Venkatesan
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Melissa P. Pizzi
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yibo Fan
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Na Niu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Huamin Wang
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shumei Song
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaffer A. Ajani
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
12
|
Chen X, Xu Z, Tang K, Hu G, Du P, Wang J, Zhang C, Xin Y, Li K, Zhang Q, Hu J, Zhang Z, Yang M, Wang G, Tan Y. The Mechanics of Tumor Cells Dictate Malignancy via Cytoskeleton-Mediated APC/Wnt/β-Catenin Signaling. RESEARCH (WASHINGTON, D.C.) 2023; 6:0224. [PMID: 37746658 PMCID: PMC10513157 DOI: 10.34133/research.0224] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/20/2023] [Indexed: 09/26/2023]
Abstract
Tumor cells progressively remodel cytoskeletal structures and reduce cellular stiffness during tumor progression, implicating the correlation between cell mechanics and malignancy. However, the roles of tumor cell cytoskeleton and the mechanics in tumor progression remain incompletely understood. We report that softening/stiffening tumor cells by targeting actomyosin promotes/suppresses self-renewal in vitro and tumorigenic potential in vivo. Weakening/strengthening actin cytoskeleton impairs/reinforces the interaction between adenomatous polyposis coli (APC) and β-catenin, which facilitates β-catenin nuclear/cytoplasmic localization. Nuclear β-catenin binds to the promoter of Oct4, which enhances its transcription that is crucial in sustaining self-renewal and malignancy. These results demonstrate that the mechanics of tumor cells dictate self-renewal through cytoskeleton-APC-Wnt/β-catenin-Oct4 signaling, which are correlated with tumor differentiation and patient survival. This study unveils an uncovered regulatory role of cell mechanics in self-renewal and malignancy, and identifies tumor cell mechanics as a hallmark not only for cancer diagnosis but also for mechanotargeting.
Collapse
Affiliation(s)
- Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Kai Tang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Pengyu Du
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Junfang Wang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Cunyu Zhang
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Ying Xin
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Keming Li
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Qiantang Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jianjun Hu
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Zhuxue Zhang
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Mo Yang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
13
|
Ko KP, Huang Y, Zhang S, Zou G, Kim B, Zhang J, Jun S, Martin C, Dunbar KJ, Efe G, Rustgi AK, Nakagawa H, Park JI. Key Genetic Determinants Driving Esophageal Squamous Cell Carcinoma Initiation and Immune Evasion. Gastroenterology 2023; 165:613-628.e20. [PMID: 37257519 PMCID: PMC10527250 DOI: 10.1053/j.gastro.2023.05.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND & AIMS Despite recent progress in identifying aberrant genetic and epigenetic alterations in esophageal squamous cell carcinoma (ESCC), the mechanism of ESCC initiation remains unknown. METHODS Using CRISPR/Cas 9-based genetic ablation, we targeted 9 genes (TP53, CDKN2A, NOTCH1, NOTCH3, KMT2D, KMT2C, FAT1, FAT4, and AJUBA) in murine esophageal organoids. Transcriptomic phenotypes of organoids and chemokine released by organoids were analyzed by single-cell RNA sequencing. Tumorigenicity and immune evasion of organoids were monitored by allograft transplantation. Human ESCC single-cell RNA sequencing data sets were analyzed to classify patients and find subsets relevant to organoid models and immune evasion. RESULTS We established 32 genetically engineered esophageal organoids and identified key genetic determinants that drive ESCC initiation. A single-cell transcriptomic analysis uncovered that Trp53, Cdkn2a, and Notch1 (PCN) triple-knockout induces neoplastic features of ESCC by generating cell lineage heterogeneity and high cell plasticity. PCN knockout also generates an immunosuppressive niche enriched with exhausted T cells and M2 macrophages via the CCL2-CCR2 axis. Mechanistically, CDKN2A inactivation transactivates CCL2 via nuclear factor-κB. Moreover, comparative single-cell transcriptomic analyses stratified patients with ESCC and identified a specific subtype recapitulating the PCN-type ESCC signatures, including the high expression of CCL2 and CD274/PD-L1. CONCLUSIONS Our study unveils that loss of TP53, CDKN2A, and NOTCH1 induces esophageal neoplasia and immune evasion for ESCC initiation and proposes the CCL2 blockade as a viable option for targeting PCN-type ESCC.
Collapse
Affiliation(s)
- Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cecilia Martin
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Karen J Dunbar
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Gizem Efe
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas; Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
14
|
Tang X, Ren J, Wei X, Wang T, Li H, Sun Y, Liu Y, Chi M, Zhu S, Lu L, Zhang J, Yang B. Exploiting synergistic effect of CO/NO gases for soft tissue transplantation using a hydrogel patch. Nat Commun 2023; 14:2417. [PMID: 37105981 PMCID: PMC10140290 DOI: 10.1038/s41467-023-37959-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Autologous skin flap transplantation is a common method for repairing complex soft tissue defects caused by cancer, trauma, and congenital malformations. Limited blood supply range and post-transplantation ischemia-reperfusion injury can lead to distal necrosis of the flap and long-term functional loss, which severely restricts the decision-making regarding the optimal surgical plan. To address this issue, we develop a hydrogel patch that releases carbon monoxide and nitric oxide gases on demand, to afford a timely blood supply for skin flap transplantation during surgery. Using an ischemia-reperfusion dorsal skin flap model in rats, we show that the hydrogel patch maintains the immediate opening of blood flow channels in transplanted tissue and effective blood perfusion throughout the perioperative period, activating perfusion of the hemodynamic donor site. We demonstrate that the hydrogel patch promotes distal vascularization and long-term functional reconstruction of transplanted tissues by inhibiting inflammatory damage and accelerating blood vessel formation.
Collapse
Affiliation(s)
- Xiaoduo Tang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Jingyan Ren
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Xin Wei
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Tao Wang
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Haiqiu Li
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Yihan Sun
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| | - Yang Liu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Mingli Chi
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China.
| | - Laijin Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
| | - Junhu Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China.
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China.
| | - Bai Yang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun, PR China
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, PR China
| |
Collapse
|
15
|
Kim B, Zhang S, Huang Y, Ko KP, Zou G, Zhang J, Jun S, Kim KB, Jung YS, Park KS, Park JI. CRACD suppresses neuroendocrinal plasticity of lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537576. [PMID: 37131761 PMCID: PMC10153265 DOI: 10.1101/2023.04.19.537576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Tumor cell plasticity contributes to intratumoral heterogeneity and therapy resistance. Through cell plasticity, lung adenocarcinoma (LUAD) cells transform into neuroendocrinal (NE) tumor cells. However, the mechanisms of NE cell plasticity remain unclear. CRACD, a capping protein inhibitor, is frequently inactivated in cancers. CRACD knock-out (KO) de-represses NE-related gene expression in the pulmonary epithelium and LUAD cells. In LUAD mouse models, Cracd KO increases intratumoral heterogeneity with NE gene expression. Single-cell transcriptomic analysis showed that Cracd KO-induced NE plasticity is associated with cell de-differentiation and activated stemness-related pathways. The single-cell transcriptomes of LUAD patient tumors recapitulate that the distinct LUAD NE cell cluster expressing NE genes is co-enriched with SOX2, OCT4, and NANOG pathway activation, and impaired actin remodeling. This study reveals an unexpected role of CRACD in restricting NE cell plasticity that induces cell de-differentiation, providing new insights into cell plasticity of LUAD.
Collapse
Affiliation(s)
- Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kee-Beom Kim
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Kwon-Sik Park
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
16
|
Li C, Wang S, Chen S, Wang X, Deng X, Liu G, Chang W, Beckers Y, Cai H. Screening and Characterization of Pediococcus acidilactici LC-9-1 toward Selection as a Potential Probiotic for Poultry with Antibacterial and Antioxidative Properties. Antioxidants (Basel) 2023; 12:215. [PMID: 36829774 PMCID: PMC9952579 DOI: 10.3390/antiox12020215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/06/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Growing interest has been focused on lactic acid bacteria as alternatives to antimicrobial growth promoters, which are characterized by the production of various functional metabolites, such as antimicrobial and antioxidants compounds. The present study was undertaken to evaluate a potential probiotic from the antioxidant perspective. LC-9-1, screened from the intestines of healthy animals, was revealed to be Pediococcus acidilactici on the basis of its morphological, biochemical, and molecular characteristics. The strain has excellent properties, including acid-production efficiency, antibacterial performance and antioxidant activity. The safety of the strain was also evaluated. Furthermore, the experiments in broiler chickens suggested that dietary LC-9-1 supplementation improved the growth performance and decreased the abdominal fat, and enhanced the antioxidant capability and intestinal innate immunity of broilers. Analysis of intestinal microbiota showed that a higher community diversity (Shannon index) was achieved. In addition to the significantly increased relative abundances of Pediococcus spp., beneficial genera such as Rothia spp. and Ruminococcus spp. were abundant, while opportunistic pathogens such as Escherichia-Shigella spp. were significantly reduced in LC-9-1-supplemented broilers. Collectively, such in-depth characterization and the available data will guide future efforts to develop next-generation probiotics, and LC-9-1 could be considered a potential strain for further utilization in direct-fed microbial or starter culture for fermentation.
Collapse
Affiliation(s)
- Chong Li
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium
| | - Shaolong Wang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Si Chen
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Xiaoying Wang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Xuejuan Deng
- National Engineering Research Center of Biological Feed, Beijing 100081, China
| | - Guohua Liu
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Wenhuan Chang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
| | - Yves Beckers
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium
| | - Huiyi Cai
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agriculture Sciences, Beijing 100081, China
- National Engineering Research Center of Biological Feed, Beijing 100081, China
| |
Collapse
|
17
|
Zou G, Park JI. Wnt signaling in liver regeneration, disease, and cancer. Clin Mol Hepatol 2023; 29:33-50. [PMID: 35785913 PMCID: PMC9845677 DOI: 10.3350/cmh.2022.0058] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/30/2022] [Indexed: 02/02/2023] Open
Abstract
The liver exhibits the highest recovery rate from acute injuries. However, in chronic liver disease, the long-term loss of hepatocytes often leads to adverse consequences such as fibrosis, cirrhosis, and liver cancer. The Wnt signaling plays a pivotal role in both liver regeneration and tumorigenesis. Therefore, manipulating the Wnt signaling has become an attractive approach to treating liver disease, including cancer. Nonetheless, given the crucial roles of Wnt signaling in physiological processes, blocking Wnt signaling can also cause several adverse effects. Recent studies have identified cancer-specific regulators of Wnt signaling, which would overcome the limitation of Wnt signaling target approaches. In this review, we discussed the role of Wnt signaling in liver regeneration, precancerous lesion, and liver cancer. Furthermore, we summarized the basic and clinical approaches of Wnt signaling blockade and proposed the therapeutic prospects of cancer-specific Wnt signaling blockade for liver cancer treatment.
Collapse
Affiliation(s)
- Gengyi Zou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Corresponding author : Gengyi Zou Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd Unit 1054, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA,Jae-Il Park Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd. Unit 1052, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| |
Collapse
|
18
|
Li L, Wen Z, Kou N, Liu J, Jin D, Wang L, Wang F, Gao L. LIS1 interacts with CLIP170 to promote tumor growth and metastasis via the Cdc42 signaling pathway in salivary gland adenoid cystic carcinoma. Int J Oncol 2022; 61:129. [PMID: 36102310 PMCID: PMC9477107 DOI: 10.3892/ijo.2022.5419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/05/2022] [Indexed: 11/20/2022] Open
Abstract
Salivary gland adenoid cystic carcinoma (SACC) is one of the most common malignant tumors, with high aggressive potential in the oral and maxillofacial regions. Lissencephaly 1 (LIS1) is a microtubule-organizing center-associated protein that regulates the polymerization and stability of microtubules by mediating the motor function of dynein. Recent studies have suggested that LIS1 plays a potential role in the malignant development of tumors, such as in mitosis and migration. However, the role of LIS1 in SACC development and its related molecular mechanisms remain unclear. Thus, the effects of LIS1 on the proliferation, apoptosis, invasion and metastasis of SACC were studied, in vivo and in vitro. The results of immunohistochemical staining showed that LIS1 was highly expressed in SACC tissues, and its expression level was associated with malignant progression. In vitro, the results of CCK-8, TUNEL, wound healing and Transwell assays demonstrated that LIS1 promotes proliferation, inhibits apoptosis, and enhances the migration and invasion of SACC-LM cells. In vivo, knockdown of LIS1 effectively suppressed the growth of subcutaneous tumors in a mouse xenograft and distant metastasis of tumor cells in the metastasis model. The co-immunoprecipitation, immunofluorescence and western blot results also revealed that LIS1 binds to cytoplasmic linker protein 170 (CLIP170) to form a protein complex (LIS1/CLIP170), which activates the cell division control protein 42 homolog (Cdc42) signaling pathway to modulate the proliferation and anti-apoptosis of tumor cells, and enhanced invasion and metastasis by regulating the formation of invadopodia and the expression of MMPs in SACC-LM cells. Therefore, the present study demonstrated that LIS1 is a cancer promoter in SACC, and the molecular mechanism of the LIS1/CLIP170/Cdc42 signaling pathway is involved in the malignant progression, which offers a promising strategy for targeted therapy of SACC.
Collapse
Affiliation(s)
- Lijun Li
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhihao Wen
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ni Kou
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jing Liu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Dong Jin
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lina Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Fu Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Lu Gao
- School of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
19
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
20
|
Yang KT, Yen CC, Chang R, Wang JT, Chen JS. CAST as a Potential Oncogene, Identified by Machine Search, in Gastric Cancer Infiltrated with Macrophages and Associated with Lgr5. Biomolecules 2022; 12:biom12050670. [PMID: 35625600 PMCID: PMC9138541 DOI: 10.3390/biom12050670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/25/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the leading malignant diseases worldwide, especially in Asia. CAST is a potential oncogene in GC carcinogenesis. The character of macrophage infiltration in the GC microenvironment also remains unaddressed. Methods: We first applied machine searching to evaluate gene candidates for GC. CAST expression and pan-cancer surveyance were analyzed using the Human Protein Atlas (HPA) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) database. The protein–protein interaction (PPI) network was downloaded from STRING. We investigated the impact of CAST on clinical prognosis using a Kaplan–Meier plotter. The correlations between CAST and Lgr5 and macrophage infiltration in GC were determined using TIMER 2.0. Finally, GeneMANIA was also used to evaluate the possible functional linkages between genes. Results: After the machine-assisted search, CAST expression was found to significantly influence the overall survival of GC patients. STRING revealed CAST-related proteomic and transcriptomic associations, mainly concerning the CAPN family. Moreover, CAST significantly impacts the prognosis of GC based on the validation of other datasets. Notably, high CAST expression was correlated with worse overall survival in GC patients (hazard ratio = 1.59; log-rank P = 9.4 × 10−8). CAST and Lgr5 expression were both positively correlated with WNT 2 and WNT 2B. Among the GC patients in several datasets, CAST and macrophage infiltration, evaluated together, showed no obvious association with poor clinical overall survival. Conclusions: CAST plays an important role in the clinical prognosis of GC and is associated with WNT 2/WNT 2B/Lgr5. Our study demonstrates that CAST’s influence on overall survival in GC is regulated by macrophage infiltration.
Collapse
Affiliation(s)
- Kuang-Tsu Yang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802213, Taiwan;
- Division of Family Medicine, Department of Community Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802213, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chia-Chi Yen
- Superintendent’s Office, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 804201, Taiwan;
- Department of Nutrition, Institute of Biomedical Nutrition, Hung-Kuang University, Taichung 433304, Taiwan
- Department of Business Management, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Renin Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung 84001, Taiwan
- Department of Recreation and Sports Management, Tajen University, Pingtung 90741, Taiwan
| | - Jui-Tzu Wang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
| | - Jin-Shuen Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan; (R.C.); (J.-T.W.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Faculty of Medicine, School of Medicine, National Defense Medicine, Taipei 11490, Taiwan
- Correspondence: ; Tel.: +886-7-342-2121
| |
Collapse
|
21
|
Wang Z, Hou H, Zhang H, Duan X, Li L, Meng L. Effect of MUC16 mutations on tumor mutation burden and its potential prognostic significance for cutaneous melanoma. Am J Transl Res 2022; 14:849-862. [PMID: 35273689 PMCID: PMC8902552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES MUC16, a mucin marker with a high mutation probability, is closely related to the occurrence, development, response to treatment, and prognosis of melanoma. As melanoma has high immunogenicity, immunotherapy has become a routine treatment. Tumor mutation burden (TMB) is the most common indicator for determining appropriate immunotherapy. The relationship between the mutation and expression of MUC16 and the prognosis, TMB, level of immune infiltration, and drug sensitivity in melanoma was investigated in this study. METHODS Melanoma data were downloaded from the Cancer Genome Atlas and the International Cancer Genome Consortium database, and the "GenVisR" package was used to visualize the gene mutation types and frequencies. Intersections of the top 30 genes with the highest mutation frequencies were determined. Thereafter, we investigated the effects of MUC16 mutations on overall survival (OS) and TMB of melanoma patients by multivariate Cox regression and multivariate logistic analyses. Related pathways that were enriched by MUC16 and BRAF were investigated using gene-set enrichment analysis and gene-set variation analysis. The CIBERSORT calculation method was used to analyze the proportion of tumor-infiltrating immune subsets. The relationship between MUC16 expression and drug sensitivity was also discussed. RESULTS Twenty-two genes with high mutation frequencies were identified in both datasets. MUC16 and ADGRV1 mutations were associated with higher TMB and good clinical prognosis (P<0.05). Multivariate Cox regression analysis showed that age, clinical stage, and MUC16 mutations were independent prognostic factors affecting OS of melanoma patients. Multivariate logistic analysis showed that gender and MUC16 mutations were independent prognostic factors affecting the TMB. MUC16 mutations and high-expression groups were primarily enriched in immune-related pathways. Furthermore, T-cell CD4 memory activation and T-cell CD8 were positively correlated with MUC16 expression and activated dendritic cells were significantly enriched in the MUC16 mutant group. Abnormal MUC16 expression may be related to abnormal methylation and drug resistance. CONCLUSION MUC16 was found to have a higher mutation frequency in melanoma patients, which is associated with a higher TMB. The mutation and/or expression of MUC16 may affect immune-related pathways and tumor-infiltrating immune cell subsets, which may improve the prognosis for melanoma patients.
Collapse
Affiliation(s)
- Zi Wang
- Department of Dermatology, Dong Zhimen Hospital Affiliated to Beijing University of Chinese MedicineBeijing 100700, China
| | - Huimin Hou
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijing 100730, China
| | - Haomin Zhang
- Department of Dermatology, Dong Zhimen Hospital Affiliated to Beijing University of Chinese MedicineBeijing 100700, China
| | - Xingwu Duan
- Department of Dermatology, Dong Zhimen Hospital Affiliated to Beijing University of Chinese MedicineBeijing 100700, China
| | - Lingling Li
- Dong Zhimen Hospital Affiliated to Beijing University of Chinese MedicineBeijing 100700, China
| | - Lingfeng Meng
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijing 100730, China
- Beijing Hospital Continence CenterBeijing 100730, China
| |
Collapse
|
22
|
Chang Y, Zhang J, Huo X, Qu X, Xia C, Huang K, Xie F, Wang N, Wei X, Jia Q. Substrate rigidity dictates colorectal tumorigenic cell stemness and metastasis via CRAD-dependent mechanotransduction. Cell Rep 2022; 38:110390. [PMID: 35172140 DOI: 10.1016/j.celrep.2022.110390] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 12/16/2022] Open
Abstract
Tumor physical microenvironment contributes greatly to the response of tumor cells. However, the mechanism of how extracellular substrate rigidity remodels colorectal cancer (CRC) cell fate and affects CRC progression remains elusive. Here, we show that F-actin regulator KIAA1211, also known as Capping protein inhibiting regulator of actin dynamics (CRAD), negatively correlates with CRC progression, stemness, and metastasis. Mechanistically, decreased CRAD in soft substrates induces Yes-associated protein (YAP) retention in the cytoplasm, restoring the repression effect on stemness markers NANOG and OCT4, thereby promoting CRC stemness and metastasis. Furthermore, CRAD deficiency promotes colorectal tumor cell softening and regulates epithelial-mesenchymal transition (EMT) states, contributing to its metastasis potential. Clinically, CRAD expression is correlated with malignant degrees and metastasis in CRC patients. Our work uncovers a role of CRAD in anticancer and mechanical signal transduction of the extracellular matrix in CRC.
Collapse
Affiliation(s)
- Yuhan Chang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Juan Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xinying Huo
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xinliang Qu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chunlei Xia
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Kaizong Huang
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Fuyang Xie
- Department of Radiotherapy, The Affiliated Lianshui People's Hospital of Kangda College of Nanjing Medical University, Jiangsu 223400, China
| | - Nuofan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| | - Qiong Jia
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China.
| |
Collapse
|
23
|
Abstract
Actin is a highly conserved protein in mammals. The actin dynamics is regulated by actin-binding proteins and actin-related proteins. Nuclear actin and these regulatory proteins participate in multiple nuclear processes, including chromosome architecture organization, chromatin remodeling, transcription machinery regulation, and DNA repair. It is well known that the dysfunctions of these processes contribute to the development of cancer. Moreover, emerging evidence has shown that the deregulated actin dynamics is also related to cancer. This chapter discusses how the deregulation of nuclear actin dynamics contributes to tumorigenesis via such various nuclear events.
Collapse
Affiliation(s)
- Yuanjian Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and Health Science Center, Houston, TX, USA.
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
24
|
Zheng ZM, Wang YY, Chen M, Yang HL, Lai ZZ, Li MQ, Shao J. FBXO17 Inhibits the Wnt/β-Catenin Pathway and Proliferation of Ishikawa Cells. Int J Med Sci 2022; 19:1430-1441. [PMID: 36035375 PMCID: PMC9413558 DOI: 10.7150/ijms.60335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/28/2022] [Indexed: 11/11/2022] Open
Abstract
Uterine corpus endometrial carcinoma (UCEC) is one of the most common types of cancer in women, and the incidence is rapidly increasing. Studies have shown that various signaling pathways serve crucial roles in the tumorigenesis of UCEC, amongst which the Wnt/β-catenin pathway is of great interest due to its crucial role in cell proliferation and the huge potential as a therapeutic target. In the present study, it was shown that FBXO17, which is a member of the F-box family, is abnormally downregulated in UCEC tissues compared with non-tumor endometrial tissues, and this was significantly associated with the clinical histological grade, as well as the abnormal proliferation of the UCEC cell line, Ishikawa, both in vitro and in vivo. Besides, the results suggested that FBXO17 may inhibit the Wnt/β-catenin signaling pathway and influence the expression of adhesion molecules, such as E-cadherin and N-cadherin in Ishikawa cells. In conclusion, these findings indicate that FBXO17 is a novel inhibitor of endometrial tumor development and it likely exerts effects via regulation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Zi-Meng Zheng
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China
| | - Ying-Ying Wang
- Department of Obstetrics and Gynecology, Yidu Central Hospital of Weifang, Weifang 262500, People's Republic of China
| | - Min Chen
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China
| | - Hui-Li Yang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China
| | - Zhen-Zhen Lai
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China
| | - Ming-Qing Li
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, People's Republic of China
| | - Jun Shao
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200082, People's Republic of China
| |
Collapse
|
25
|
A new murine esophageal organoid culture method and organoid-based model of esophageal squamous cell neoplasia. iScience 2021; 24:103440. [PMID: 34877497 PMCID: PMC8633967 DOI: 10.1016/j.isci.2021.103440] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/07/2021] [Accepted: 11/10/2021] [Indexed: 02/07/2023] Open
Abstract
Organoids mimic the physiologic and pathologic events of organs. However, no consensus on esophageal organoid (EO) culture methods has been reached. Moreover, organoid models reproducing esophageal squamous cell carcinoma (ESCC) initiation have been unavailable. Herein, we sought to develop an esophageal minimum essential organoid culture medium (E-MEOM) for culturing murine EOs and establishing an early ESCC model. We formulated E-MEOM to grow EOs from a single cell with clonal expansion, maintenance, and passage. We found that EOs cultured in E-MEOM were equivalent to the esophageal epithelium by histological analysis and transcriptomic study. Trp53 knockout and KrasG12D expression in EOs induced the development of esophageal squamous neoplasia, an early lesion of ESCC. Here we propose the new formula for EO culture with minimum components and the organoid model recapitulating ESCC initiation, laying the foundation for ESCC research and drug discovery. Identification of minimal components for murine EO growth and maintenance Mouse EOs morphologically and transcriptionally recapitulate the human esophagus Trp53 KO and KrasG12D induced esophageal neoplasia mimicking early ESCC
Collapse
|
26
|
Kwon M, Rubio G, Nolan N, Auteri P, Volmar JA, Adem A, Javidian P, Zhou Z, Verzi MP, Pine SR, Libutti SK. FILIP1L Loss Is a Driver of Aggressive Mucinous Colorectal Adenocarcinoma and Mediates Cytokinesis Defects through PFDN1. Cancer Res 2021; 81:5523-5539. [PMID: 34417201 DOI: 10.1158/0008-5472.can-21-0897] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/25/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022]
Abstract
Aneuploid mucinous colorectal adenocarcinoma (MAC) is an aggressive subtype of colorectal cancer with poor prognosis. The tumorigenic mechanisms in aneuploid MAC are currently unknown. Here we show that downregulation of Filamin A-interacting protein 1-like (FILIP1L) is a driver of MAC. Loss of FILIP1L increased xenograft growth, and, in colon-specific knockout mice, induced colonic epithelial hyperplasia and mucin secretion. The molecular chaperone prefoldin 1 (PFDN1) was identified as a novel binding partner of FILIP1L at the centrosomes throughout mitosis. FILIP1L was required for proper centrosomal localization of PFDN1 and regulated proteasome-dependent degradation of PFDN1. Importantly, increased PFDN1, caused by downregulation of FILIP1L, drove multinucleation and cytokinesis defects in vitro and in vivo, which were confirmed by time-lapse imaging and 3D cultures of normal epithelial cells. Overall, these findings suggest that downregulation of FILIP1L and subsequent upregulation of PFDN1 is a driver of the unique neoplastic characteristics in aggressive aneuploid MAC. SIGNIFICANCE: This study identifies FILIP1L as a tumor suppressor in mucinous colon cancer and demonstrates that FILIP1L loss results in aberrant stabilization of a centrosome-associated chaperone protein to drive aneuploidy and disease progression.
Collapse
Affiliation(s)
- Mijung Kwon
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Genesaret Rubio
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Nicholas Nolan
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Peter Auteri
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Jean Arly Volmar
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Asha Adem
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Parisa Javidian
- Department of Pathology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Zhongren Zhou
- Department of Pathology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Michael P Verzi
- Department of Genetics, Rutgers University, Piscataway, New Jersey
| | - Sharon R Pine
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Steven K Libutti
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
| |
Collapse
|
27
|
Kundishora AJ, Peters ST, Pinard A, Duran D, Panchagnula S, Barak T, Miyagishima DF, Dong W, Smith H, Ocken J, Dunbar A, Nelson-Williams C, Haider S, Walker RL, Li B, Zhao H, Thumkeo D, Marlier A, Duy PQ, Diab NS, Reeves BC, Robert SM, Sujijantarat N, Stratman AN, Chen YH, Zhao S, Roszko I, Lu Q, Zhang B, Mane S, Castaldi C, López-Giráldez F, Knight JR, Bamshad MJ, Nickerson DA, Geschwind DH, Chen SSL, Storm PB, Diluna ML, Matouk CC, Orbach DB, Alper SL, Smith ER, Lifton RP, Gunel M, Milewicz DM, Jin SC, Kahle KT. DIAPH1 Variants in Non-East Asian Patients With Sporadic Moyamoya Disease. JAMA Neurol 2021; 78:993-1003. [PMID: 34125151 PMCID: PMC8204259 DOI: 10.1001/jamaneurol.2021.1681] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Importance Moyamoya disease (MMD), a progressive vasculopathy leading to narrowing and ultimate occlusion of the intracranial internal carotid arteries, is a cause of childhood stroke. The cause of MMD is poorly understood, but genetic factors play a role. Several familial forms of MMD have been identified, but the cause of most cases remains elusive, especially among non-East Asian individuals. Objective To assess whether ultrarare de novo and rare, damaging transmitted variants with large effect sizes are associated with MMD risk. Design, Setting, and Participants A genetic association study was conducted using whole-exome sequencing case-parent MMD trios in a small discovery cohort collected over 3.5 years (2016-2019); data were analyzed in 2020. Medical records from US hospitals spanning a range of 1 month to 1.5 years were reviewed for phenotyping. Exomes from a larger validation cohort were analyzed to identify additional rare, large-effect variants in the top candidate gene. Participants included patients with MMD and, when available, their parents. All participants who met criteria and were presented with the option to join the study agreed to do so; none were excluded. Twenty-four probands (22 trios and 2 singletons) composed the discovery cohort, and 84 probands (29 trios and 55 singletons) composed the validation cohort. Main Outcomes and Measures Gene variants were identified and filtered using stringent criteria. Enrichment and case-control tests assessed gene-level variant burden. In silico modeling estimated the probability of variant association with protein structure. Integrative genomics assessed expression patterns of MMD risk genes derived from single-cell RNA sequencing data of human and mouse brain tissue. Results Of the 24 patients in the discovery cohort, 14 (58.3%) were men and 18 (75.0%) were of European ancestry. Three of 24 discovery cohort probands contained 2 do novo (1-tailed Poisson P = 1.1 × 10-6) and 1 rare, transmitted damaging variant (12.5% of cases) in DIAPH1 (mammalian diaphanous-1), a key regulator of actin remodeling in vascular cells and platelets. Four additional ultrarare damaging heterozygous DIAPH1 variants (3 unphased) were identified in 3 other patients in an 84-proband validation cohort (73.8% female, 77.4% European). All 6 patients were non-East Asian. Compound heterozygous variants were identified in ena/vasodilator-stimulated phosphoproteinlike protein EVL, a mammalian diaphanous-1 interactor that regulates actin polymerization. DIAPH1 and EVL mutant probands had severe, bilateral MMD associated with transfusion-dependent thrombocytopenia. DIAPH1 and other MMD risk genes are enriched in mural cells of midgestational human brain. The DIAPH1 coexpression network converges in vascular cell actin cytoskeleton regulatory pathways. Conclusions and Relevance These findings provide the largest collection to date of non-East Asian individuals with sporadic MMD harboring pathogenic variants in the same gene. The results suggest that DIAPH1 is a novel MMD risk gene and impaired vascular cell actin remodeling in MMD pathogenesis, with diagnostic and therapeutic ramifications.
Collapse
Affiliation(s)
- Adam J. Kundishora
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Samuel T. Peters
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson
| | - Amélie Pinard
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Daniel Duran
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson
| | | | - Tanyeri Barak
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut
| | - Danielle F. Miyagishima
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut
- Yale Program on Neurogenetics, Yale School of Medicine, New Haven, Connecticut
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Hannah Smith
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Jack Ocken
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Ashley Dunbar
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | | | - Shozeb Haider
- Department of Pharmaceutical and Biological Chemistry, University College London School of Pharmacy, London, United Kingdom
| | - Rebecca L. Walker
- Department of Neurology, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles
| | - Boyang Li
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Dean Thumkeo
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Arnaud Marlier
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Phan Q. Duy
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Nicholas S. Diab
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Benjamin C. Reeves
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | | | | | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri
| | - Yi-Hsien Chen
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Shujuan Zhao
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Isabelle Roszko
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Qiongshi Lu
- Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Shrikant Mane
- Yale Center for Genome Analysis, West Haven, Connecticut
| | | | | | | | | | | | - Daniel H. Geschwind
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles
| | - Shih-Shan Lang Chen
- Division of Neurosurgery, Children's Hospital of Philadelphia, Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Phillip B. Storm
- Division of Neurosurgery, Children's Hospital of Philadelphia, Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Michael L. Diluna
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Charles C. Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
| | - Darren B. Orbach
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seth L. Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Edward R. Smith
- Department of Neurosurgery, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard P. Lifton
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Dianna M. Milewicz
- Department of Internal Medicine, McGovern Medical School, University of Texas Health Science Center, Houston
| | - Sheng Chih Jin
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Kristopher T. Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut
- Department of Pediatrics, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
28
|
Abstract
Mucins are high molecular-weight epithelial glycoproteins and are implicated in many physiological processes, including epithelial cell protection, signaling transduction, and tissue homeostasis. Abnormality of mucus expression and structure contributes to biological properties related to human cancer progression. Tumor growth sites induce inhospitable conditions. Many kinds of research suggest that mucins provide a microenvironment to avoid hypoxia, acidic, and other biological conditions that promote cancer progression. Given that the mucus layer captures growth factors or cytokines, we propose that mucin helps to ameliorate inhospitable conditions in tumor-growing sites. Additionally, the composition and structure of mucins enable them to mimic the surface of normal epithelial cells, allowing tumor cells to escape from immune surveillance. Indeed, human cancers such as mucinous carcinoma, show a higher incidence of invasion to adjacent organs and lymph node metastasis than do non-mucinous carcinoma. In this mini-review, we discuss how mucin provides a tumor-friendly environment and contributes to increased cancer malignancy in mucinous carcinoma.
Collapse
Affiliation(s)
- Dong-Han Wi
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| | - Jong-Ho Cha
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon 22212, Korea
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| |
Collapse
|
29
|
Abstract
Colorectal cancer has served as a genetic and biological paradigm for the evolution of solid tumors, and these insights have illuminated early detection, risk stratification, prevention, and treatment principles. Employing the hallmarks of cancer framework, we provide a conceptual framework to understand how genetic alterations in colorectal cancer drive cancer cell biology properties and shape the heterotypic interactions across cells in the tumor microenvironment. This review details research advances pertaining to the genetics and biology of colorectal cancer, emerging concepts gleaned from immune and single-cell profiling, and critical advances and remaining knowledge gaps influencing the development of effective therapies for this cancer that remains a major public health burden.
Collapse
Affiliation(s)
- Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xingdi Ma
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
30
|
Zhou Y, Long Y, Chen Y, Liu J, Pu D, Huang J, Bi F, Li Q, Gou H, Qiu M. First-line therapy of bevacizumab plus chemotherapy versus cetuximab plus chemotherapy for metastatic colorectal cancer patients with mucinous adenocarcinoma or mucinous component. Cancer Med 2021; 10:3388-3402. [PMID: 33939281 PMCID: PMC8124114 DOI: 10.1002/cam4.3876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To compare the efficacy of first-line bevacizumab plus chemotherapy with cetuximab plus chemotherapy based on the stratification of metastatic colorectal cancer (mCRC) patients with mucinous adenocarcinoma (MA) or mucinous component (MC). METHODS A retrospective study involving all mCRC patients receiving first-line bevacizumab-based or cetuximab-based chemotherapy at our hospital from September 2013 to January 2020 was conducted. Overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) were compared between the cetuximab-chemotherapy group and the bevacizumab-chemotherapy group on the basis of the conventional pathological classification of MA or MC. RESULTS A total of 620 patients with mCRC were included in our study, consisting of 141 (22.7%) patients with MA/MC and 479 (77.3%) patients with non-mucinous adenocarcinoma (NMA). In the MA/MC cohort, patients who were treated with bevacizumab-based chemotherapy were associated with significantly better OS than those treated with cetuximab-base chemotherapy (30.0 vs. 26.3 months, p = 0.002), irrespective of tumor sites. The efficacy of bevacizumab-based chemotherapy was higher in nearly all subgroups as shown in the subgroup analysis. In the NMA cohort, median OS was better in the cetuximab plus chemotherapy group than that in the bevacizumab plus chemotherapy group (32.2 vs. 27.0 months, p = 0.005) for left-side mCRC patients, whereas OS was significantly longer in the bevacizumab plus chemotherapy group for right-side mCRC patients (26.0 vs. 20.9 months, p = 0.013). CONCLUSION Conventional pathological classification (e.g. MA/MC) should be considered when tailoring the individualized optimal treatment for mCRC. Bevacizumab plus chemotherapy as first-line therapy may be the optimal option for patients with MA/MC.
Collapse
Affiliation(s)
- Yu‐Wen Zhou
- Department of BiotherapyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Yi‐Xiu Long
- Department of BiotherapyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Ye Chen
- Department of Abdominal OncologyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Ji‐Yan Liu
- Department of BiotherapyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Dan Pu
- Lung Cancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Jia‐Yan Huang
- Department of Radiology and Medical UltrasoundWest China Hospital of Sichuan UniversityChengduChina
| | - Feng Bi
- Department of Abdominal OncologyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Qiu Li
- Department of Abdominal OncologyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Hong‐Feng Gou
- Department of Abdominal OncologyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| | - Meng Qiu
- Department of Abdominal OncologyCancer CenterWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
31
|
Knockdown of CRAD suppresses the growth and promotes the apoptosis of human lung cancer cells via Claudin 4. Biosci Rep 2021; 40:226565. [PMID: 33006362 PMCID: PMC7560521 DOI: 10.1042/bsr20201140] [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: 04/09/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 11/21/2022] Open
Abstract
Non–small cell lung cancer (NSCLC) is one of the most common causes of cancer-related mortality globally. However, the mechanism underlying NSCLC is not fully understood. Here, we investigated the role of cancer-related regulator of actin dynamics (CRAD) in NSCLC. We showed that CRAD was up-regulated in human NSCLC tissues and lung cancer cell lines. Lentivirus-mediated knockdown of CRAD repressed the proliferation and colony growth of A549 and H1299 cells. Apoptosis was enhanced by CRAD silencing in both cells, implicating that CRAD might maintain the survival of lung cancer cells. Microarray and bioinformatic assay revealed that CRAD directly or indirectly regulated diverse genes, including those involved in cell cycle and DNA damage repair. qRT-PCR and Western blot results confirmed the dysregulated genes as shown in microarray analysis. Claudin 4 was up-regulated in CRAD silenced A549 cells. The knockdown of Claudin 4 blocked the effects of CRAD on the expression of cell cycle and apoptosis effectors and enhanced the viability of A549 cells with CRAD down-regulation. Taken together, our findings demonstrate that CRAD acts as an oncogene in NSCLC at least partly through repressing Claudin 4.
Collapse
|
32
|
Li Y, Bai L, Yu H, Cai D, Wang X, Huang B, Peng S, Huang M, Cao G, Kaz AM, Grady WM, Wang J, Luo Y. Epigenetic Inactivation of α-Internexin Accelerates Microtubule Polymerization in Colorectal Cancer. Cancer Res 2020; 80:5203-5215. [PMID: 33051252 DOI: 10.1158/0008-5472.can-20-1590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/26/2020] [Accepted: 10/08/2020] [Indexed: 11/16/2022]
Abstract
DNA methylation contributes to malignant transformation, but little is known about how the methylation drives colorectal cancer evolution at the early stages. Here we identify aberrant INA (α-internexin) gene methylation in colon adenoma and adenocarcinoma by filtering data obtained from a genome-wide screen of methylated genes. The gene encoding INA, a type IV intermediate filament, was frequently hypermethylated in CpG islands located in the promoter region. This hypermethylation preferentially occurred in large tumors and was a prognostic marker for poor overall survival in patients with colorectal cancer. This type of epigenetic alteration silenced INA expression in both adenoma and adenocarcinoma tissues. Gene silencing of INA in colorectal cancer cells increased cell proliferation, migration, and invasion. Restored INA expression blocked migration and invasion in vitro and reduced lung metastasis in vivo. Mechanistically, INA directly inhibited microtubule polymerization in vitro and decreased intracellular microtubule plus-end assembly rates. A peptide array screen surveying the tubulin-binding sites in INA identified a tubulin-binding motif located in the N-terminal head domain that plays a tumor-suppressive role by binding to unpolymerized tubulins and impeding microtubule polymerization. Thus, epigenetic inactivation of INA is an intermediate filament reorganization event that is essential to accelerate microtubule polymerization in the early stages of colorectal cancer. SIGNIFICANCE: This work provides insight into the epigenetic inactivation of INA, a novel identified tumor suppressor, which increases microtubule polymerization during colorectal cancer progression.
Collapse
Affiliation(s)
- Yingjie Li
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liangliang Bai
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huichuan Yu
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Du Cai
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaolin Wang
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Baoyuan Huang
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shaoyong Peng
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meijin Huang
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guangwen Cao
- Department of Epidemiology, Second Military Medical University, Shanghai, China
| | - Andrew M Kaz
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Gastroenterology Section, VA Puget Sound Health Care System, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - William M Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Jianping Wang
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanxin Luo
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
33
|
Snider PL, Simmons O, Conway SJ. Cracd Marks the First Wave of Meiosis during Spermatogenesis and Is Mis-Expressed in Azoospermia Mice. J Dev Biol 2020; 8:jdb8030021. [PMID: 32962040 PMCID: PMC7558608 DOI: 10.3390/jdb8030021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/04/2020] [Accepted: 09/06/2020] [Indexed: 12/16/2022] Open
Abstract
Testicular development starts in utero and maturation continues postnatally, requiring a cascade of gene activation and differentiation into different cell types, with each cell type having its own specific function. As we had previously reported that the Capping protein inhibiting regulator of actin (Cracd) gene was expressed in the adult mouse testis, herein we examine when and where the β-catenin associated Cracd is initially expressed during postnatal testis development. Significantly, Cracd mRNA is present in both the immature postnatal and adult testis in round spermatid cells, with highest level of expression occurring during the first wave of meiosis and spermatogenesis. In the juvenile testes, Cracd is initially expressed within the innermost region but as maturation occurs, Cracd mRNA switches to a more peripheral location. Thereafter, Cracd is downregulated to maintenance levels in the haploid male germ cell lineage. As Cracd mRNA was expressed within developing round spermatids, we tested its effectiveness as a biomarker of non-obstructive azoospermia using transgenic knockout mice models. Meaningfully, Cracd expression was absent in Deleted in azoospermia like (Dazl) null testis, which exhibit a dramatic germ cell loss. Moreover, Cracd was abnormally regulated and ectopically mis-expressed in Polypyrimidine tract binding protein-2 (Ptbp2) conditional germ cell restricted knockout testis, which exhibit a block during spermatid differentiation and a reduction in the number of late stage spermatocytes coincident with reduced β-catenin expression. Combined, these data suggest that Cracd is a useful first wave of spermatogenesis biomarker of azoospermia phenotypes, even prior to an overt phenotype being evident.
Collapse
|
34
|
Jung YS, Park JI. Wnt signaling in cancer: therapeutic targeting of Wnt signaling beyond β-catenin and the destruction complex. Exp Mol Med 2020; 52:183-191. [PMID: 32037398 PMCID: PMC7062731 DOI: 10.1038/s12276-020-0380-6] [Citation(s) in RCA: 314] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023] Open
Abstract
Wnt/β-catenin signaling is implicated in many physiological processes, including development, tissue homeostasis, and tissue regeneration. In human cancers, Wnt/β-catenin signaling is highly activated, which has led to the development of various Wnt signaling inhibitors for cancer therapies. Nonetheless, the blockade of Wnt signaling causes side effects such as impairment of tissue homeostasis and regeneration. Recently, several studies have identified cancer-specific Wnt signaling regulators. In this review, we discuss the Wnt inhibitors currently being used in clinical trials and suggest how additional cancer-specific regulators could be utilized to treat Wnt signaling-associated cancer.
Collapse
Affiliation(s)
- Youn-Sang Jung
- 0000 0001 2291 4776grid.240145.6Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Jae-Il Park
- 0000 0001 2291 4776grid.240145.6Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA ,0000 0001 2291 4776grid.240145.6Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA ,0000 0001 2291 4776grid.240145.6Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| |
Collapse
|
35
|
Catalano V, Bergamo F, Cremolini C, Vincenzi B, Negri F, Giordani P, Alessandroni P, Intini R, Stragliotto S, Rossini D, Borelli B, Santini D, Sarti D, Rocchi MBL, Lonardi S, Falcone A, Zagonel V, Mattioli R, Graziano F. Clinical impact of first-line bevacizumab plus chemotherapy in metastatic colorectal cancer of mucinous histology: a multicenter, retrospective analysis on 685 patients. J Cancer Res Clin Oncol 2020; 146:493-501. [PMID: 31691872 DOI: 10.1007/s00432-019-03077-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 11/02/2019] [Indexed: 01/26/2023]
Abstract
PURPOSE In metastatic colorectal cancer (MCRC), mucinous histology has been associated with poor response rate and prognosis. We investigated whether bevacizumab combined with different chemotherapy regimens may have an impact on clinical outcomes of MCRC patients with mucinous histology. METHODS 685 MCRC patients were classified in mucinous adenocarcinoma (MC) and non-mucinous adenocarcinoma (NMC) and were treated with first-line bevacizumab plus fluoropyrimidine (FP)-based, oxaliplatin (OXA)-based, irinotecan (IRI)-based, or FOLFOXIRI. RESULTS Ninety-four (13.7%) patients had MC. With a median follow-up of 50 months, MC patients had a median overall survival (OS) of 28.2 months compared with 27.7 months for the NMC group [hazard ratio (HR) = 0.92; 95% confidence interval (CI) 0.70-1.19, P = 0.530]. The overall response rates for MC and NMC were 41.5% (95% CI 31.5-51.4) and 62.4% (95% CI 58.4-66.3), respectively (Chi-square test, P <0.003). After correcting for significant prognostic factors by multivariate Cox regression analysis, age, resection of the primary tumour, and number of metastatic sites were found to be associated with poorer OS, but not mucinous histology. CONCLUSION Compared with NMC, MCRC patients with mucinous histology treated with bevacizumab plus chemotherapy had comparable OS despite lower overall response rate.
Collapse
Affiliation(s)
- Vincenzo Catalano
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy.
| | - Francesca Bergamo
- Medical Oncology Unit 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine, Azienda Ospedaliero-Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico, Università di Roma, Rome, Italy
| | - Francesca Negri
- Department of Oncology, Ospedale Universitario, Parma, Italy
| | - Paolo Giordani
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy
| | - Paolo Alessandroni
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy
| | - Rossana Intini
- Medical Oncology Unit 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Silvia Stragliotto
- Medical Oncology Unit 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Daniele Rossini
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine, Azienda Ospedaliero-Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Beatrice Borelli
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine, Azienda Ospedaliero-Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Daniele Santini
- Department of Medical Oncology, Campus Bio-Medico, Università di Roma, Rome, Italy
| | - Donatella Sarti
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy
| | - Marco B L Rocchi
- Unità di Statistica Medica e Biometria, Department of Biomolecular Sciences, Università "Carlo Bo", Urbino, Italy
| | - Sara Lonardi
- Medical Oncology Unit 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Alfredo Falcone
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine, Azienda Ospedaliero-Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Vittorina Zagonel
- Medical Oncology Unit 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Rodolfo Mattioli
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy
| | - Francesco Graziano
- Department of Oncology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Presidio San Salvatore, Via Lombroso 1, 61122, Pesaro, Italy
| |
Collapse
|
36
|
Zhou M, Fan J, Li Z, Li P, Sun Y, Yang Y, Zhou X, Wang J, Wang Y, Qi H, Cai W, Dai X, Hirsch FR. Prognostic impact of tumor mutation burden and the mutation in KIAA1211 in small cell lung cancer. Respir Res 2019; 20:248. [PMID: 31699089 PMCID: PMC6836503 DOI: 10.1186/s12931-019-1205-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022] Open
Abstract
Background Small cell lung cancer (SCLC) is a highly aggressive lung cancer subtype with poor survival and limited treatment options. Sequencing results have revealed gene mutations associated with SCLC, however, the correlation between the genomic alterations and clinical prognosis of SCLC is yet unclear. Methods Targeted next-generation sequencing of 62 cancer related genes was performed on 53 SCLC samples. The correlations between clinical outcomes and genomic alterations were analyzed. Results 38/62 (61.3%) candidate genes harbored some alterations, while all the SCLC samples carried at least 3 gene mutations. The most common nonsynonymous mutations included ERBB2 (95.9%), CREBBP (95.9%), and TP53 (77.6%). The median nonsynonymous tumor mutation burden (TMB) was 21.7 mutations/Mb (rang, 9.3–55.9). High TMB (> 21 mutations/Mb) was good prognostic factor in overall survival (OS) (21.7 vs. 10.4 months, P = 0.012). Multivariate analysis showed that high TMB was an independent prognostic factor. The overall survival (OS) of patients carrying KIAA1211 mutation was significantly longer than those with wild-type KIAA1211 (P < 0.001). Conclusions The current study highlights the potential role of genomic alterations for the prognosis of SCLC. Higher TMB was associated with a better prognosis, and KIAA1211 might be a good prognostic factor in SCLC.
Collapse
Affiliation(s)
- Mengting Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Jun Fan
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Zhenyu Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Pindong Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Yuhui Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Xiaoshu Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Jing Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Ye Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China
| | - Huiwei Qi
- Shanghai Tongshu Biotechnology Co., Ltd, Shanghai, 200120, People's Republic of China
| | - Weijing Cai
- Shanghai Tongshu Biotechnology Co., Ltd, Shanghai, 200120, People's Republic of China
| | - Xiaofang Dai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, People's Republic of China.
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Center Mount Sinai Health System; Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
37
|
Liu Z, Cao H, Shi Y, Yang R. KIAA1211 plays an oncogenic role in human non-small cell lung cancer. J Cancer 2019; 10:6747-6753. [PMID: 31777604 PMCID: PMC6856884 DOI: 10.7150/jca.35951] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/31/2019] [Indexed: 12/31/2022] Open
Abstract
One of the main causes of cancer disease and death worldwide is lung cancer. Our study focused on the function of KIAA1211 in non-small cell lung cancer (NSCLC). According to the data about NSCLC patients that from the Cancer Genome Atlas (TCGA), we found that KIAA1211 in NSCLC (P=5.06E-06) was significantly higher than the adjacent normal. Lentivirus-mediated short hairpin RNA (shRNA) was used to knockdown BATF expression in the human A549 NSCLC cell line and assessed by RT-qPCR and Western blot. Cell proliferation was evaluated by MTT assay and Celigo imaging cytometry. Cell apoptosis were detected by Annexin V staining. The test results showed that KIAA1211-shRNA A549 and SPC-A-1 cells can inhibit cell proliferation, and the apoptosis rate of KIAA1211-shRNA group was significantly higher than that of the control group. Knockdown of KIAA1211 inhibited NSCLC progression in xenograft tumor model. In conclusion, KIAA1211 could regulate NSCLC cells proliferation and apoptosis in vitro and in vivo. KIAA1211 may serve as a potent target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Zhengcheng Liu
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu, 210000, China
| | - Hui Cao
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu, 210000, China
| | - Ye Shi
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu, 210000, China
| | - Rusong Yang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing, Jiangsu, 210000, China
| |
Collapse
|
38
|
Cohen J, Luxenburg C. Wave of the future: involvement of actin polymerization in the regulation of tissue growth and shape. Mol Cell Oncol 2019; 6:e1609877. [PMID: 31528690 DOI: 10.1080/23723556.2019.1609877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 10/26/2022]
Abstract
The WASP-family verprolin-homologous (Wave) complex enhances actin-related protein 2/3 (Arp2/3)-mediated actin polymerization. Recently, we have identified a novel Wave complex-actin-Wnt/β-catenin-Sox9 pathway that regulates epidermal morphogenesis and proliferation. These findings highlight a mechanism by which actin polymerization impacts crucial signaling events in vivo.
Collapse
Affiliation(s)
- Jonathan Cohen
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Affiliation(s)
- George Eng
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Braverman
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
40
|
Snider PL, Snider E, Simmons O, Lilly B, Conway SJ. Analysis of Uncharacterized mKiaa1211 Expression during Mouse Development and Cardiovascular Morphogenesis. J Cardiovasc Dev Dis 2019; 6:jcdd6020024. [PMID: 31234534 PMCID: PMC6617212 DOI: 10.3390/jcdd6020024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/13/2019] [Accepted: 06/19/2019] [Indexed: 12/24/2022] Open
Abstract
Mammalian Kiaa1211 and Kiaa1211-like are a homologous pair of uncharacterized, highly conserved genes cloned from fetal and adult brain cDNA libraries. Herein we map the in utero spatiotemporal expression of mKiaa1211 and mKiaa1211L mRNA and their expression patterns in postnatal testis, skin, gastrointestinal, and adipose progenitor tissues. Significantly, mKiaa1211 is present throughout the early stages of mouse heart development, particularly in the second heart field (SHF) lineage as it differentiates from mesenchymal cells into cardiomyocytes. We also show that mKiaa1211 is expressed within several early neuronal tissues destined to give rise to central, peripheral, and sympathetic nervous system structures. Expression profiling revealed that the paralog mKiaa1211L is not expressed during the normal developmental process and that mKiaa1211 expression was noticeably absent from most adult terminally differentiated tissues. Finally, we confirm that a previously uncharacterized CRISPR/CAS-generated mKiaa1211 mouse mutant allele is hypomorphic.
Collapse
Affiliation(s)
- Paige L Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Elizabeth Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Biosciences, Indiana University, Bloomington, IN 47405, USA.
| | - Olga Simmons
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Brenda Lilly
- The Heart Center, Nationwide Children's Hospital, Columbus, OH 43205, USA.
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
41
|
Cohen J, Raviv S, Adir O, Padmanabhan K, Soffer A, Luxenburg C. The Wave complex controls epidermal morphogenesis and proliferation by suppressing Wnt-Sox9 signaling. J Cell Biol 2019; 218:1390-1406. [PMID: 30867227 PMCID: PMC6446834 DOI: 10.1083/jcb.201807216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/11/2018] [Accepted: 02/01/2019] [Indexed: 02/08/2023] Open
Abstract
The Wave complex promotes Arp2/3-mediated actin polymerization. Cohen et al. show that Wave complex activity regulates epidermal shape and growth. Without Wave complex activity, F-actin content is down-regulated and ectopic activity of the Wnt/β-catenin–SOX9 pathway is triggered. This activity induces epidermal hyperproliferation and disrupts tissue architecture. Development of the skin epidermis requires tight spatiotemporal control over the activity of several signaling pathways; however, the mechanisms that orchestrate these events remain poorly understood. Here, we identify a key role for the Wave complex proteins ABI1 and Wave2 in regulating signals that control epidermal shape and growth. In utero RNAi-mediated silencing of Abi1 or Wasf2 induced cellular hyperproliferation and defects in architecture of the interfollicular epidermis (IFE) and delayed hair follicle growth. Unexpectedly, SOX9, a hair follicle growth regulator, was aberrantly expressed throughout the IFE of the mutant embryos, and its forced overexpression mimicked the Wave complex loss-of-function phenotype. Moreover, Wnt signaling, which regulates SOX9+ cell specification, was up-regulated in Wave complex loss-of-function IFE. Importantly, we show that the Wave complex regulates filamentous actin content and that a decrease in actin levels is sufficient to elevate Wnt/β-catenin signaling. Our results identify a novel role for Wave complex– and actin-regulated signaling via Wnt and SOX9 in skin development.
Collapse
Affiliation(s)
- Jonathan Cohen
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shaul Raviv
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Adir
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Krishnanand Padmanabhan
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arad Soffer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
42
|
Friedman FB. So you always wanted to write about that patient who. Exp Mol Med 1981; 51:1-13. [PMID: 31827074 PMCID: PMC6906379 DOI: 10.1038/s12276-019-0349-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/11/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022] Open
Abstract
Small-cell lung cancer (SCLC) remains the deadliest of all the lung cancer types. Its high mortality is largely attributed to the invariable development of resistance to standard chemo/radiotherapies, which have remained unchanged for the past 30 years, underscoring the need for new therapeutic approaches. The discovery of molecular targets for chemoprevention and treatment has been hampered by the poor understanding of SCLC progression. In recent years, comprehensive omics-based analyses have led to the discovery of recurrent alterations in patient tumors, and functional studies using genetically engineered mouse models and patient-derived tumor models have provided information about the alterations critical for SCLC pathogenesis. Defining the somatic alterations scattered throughout the SCLC genome will help to understand the underlying mechanism of this devastating disease and pave the way for the discovery of therapeutic vulnerabilities associated with the genomic alterations. Alterations in the small cell lung cancer (SCLC) genome are critical for disease progression and relapse. A complete map of the genome in cancerous cells would greatly improve the chances of successfully treating this deadly disease. SCLC is often detected too late, and only five per cent of patients survive beyond five years after diagnosis. While the disease initially responds to standard chemotherapy, the cancer cells quickly build resistance and relapse follows. Kwon-Sik Park at the University of Virginia, Charlottesville, US, and co-workers reviewed current understanding of SCLC genome alterations. The latest research highlights substantial variations in the SCLC genome between patients, with implications for existing treatment regimens. Researchers have made considerable progress in profiling the genome, with significant alterations, mutations and potential therapeutic targets now being explored in genetically engineered mouse models and patient-derived tumor models.
Collapse
|