1
|
Koll KK, Zimmermann MM, Will PA, Kneser U, Hirche C. The Transcription Factor SOX18 Inhibitor Small Molecule 4 Is a Potential Treatment of Cancer-Induced Lymphatic Metastasis and Lymphangiosarcoma. Cancer Rep (Hoboken) 2025; 8:e70110. [PMID: 39791369 PMCID: PMC11726641 DOI: 10.1002/cnr2.70110] [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: 12/25/2023] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Malignant tumors release growth factors, promoting lymphangiogenesis in primary tumors and draining sentinel lymph nodes, ultimately facilitating lymph node metastasis. As a malignant lymphatic tumor entity, lymphangiosarcomas are characterized by low survival rates and limited treatment options. The transcription factor SOX18 plays a crucial role in both lymphatic endothelial cell differentiation and cancer-induced lymphangiogenesis. AIMS In this in vitro study, we investigated the potential therapeutic effect of a small molecule called Sm4, which inhibits SOX18, on lymphatic endothelial and lymphangiosarcoma cells in vitro. METHODS AND RESULTS Human dermal lymphatic endothelial cells (HDLECs), lymphangiosarcoma cells (MO-LAS), and other endothelial cell lines were cultured. We found that Sox18 exhibited high mRNA expression levels in both HDLEC and MO-LAS. Sm4 treatment decreased the Sox18 expression level at the mRNA and protein levels in both HDLEC and MO-LAS significantly, a phenomenon confirmed through immunofluorescence images. Additionally, Sm4 treatment suppressed the expression of key lymphatic phenotype markers (Prox1, Flt4, and Lyve1) and hindered migration in both HDLEC and MO-LAS, all while maintaining cell viability. CONCLUSION These findings suggest that targeting SOX18 with Sm4 may hold potential as a therapeutic strategy for lymphangiosarcoma and cancer-induced lymphatic metastasis. Further in vitro studies are warranted to investigate the mechanisms and conduct dose-response analyses to evaluate Sm4's potential as a targeted therapy for lymphangiosarcoma and cancer-induced lymphangiogenesis in the future.
Collapse
Affiliation(s)
- Katja K. Koll
- Department of Hand, Plastic and Reconstructive Surgery, MicrosurgeryBurn Centre BG Klinik LudwigshafenLudwigshafenGermany
- Medical Faculty of the University HeidelbergHeidelbergGermany
| | - Martin M. Zimmermann
- Department of Hand, Plastic and Reconstructive Surgery, MicrosurgeryBurn Centre BG Klinik LudwigshafenLudwigshafenGermany
- Medical Faculty of the University HeidelbergHeidelbergGermany
| | - Patrick A. Will
- Department of Plastic and Hand SurgeryFaculty of Medicine and University Hospital Carl Gustav Carus, TU DresdenDresdenGermany
| | - Ulrich Kneser
- Department of Hand, Plastic and Reconstructive Surgery, MicrosurgeryBurn Centre BG Klinik LudwigshafenLudwigshafenGermany
- Medical Faculty of the University HeidelbergHeidelbergGermany
| | - Christoph Hirche
- Department of Plastic, Hand, and Reconstructive Microsurgery, BG Unfallklinik Frankfurt Am MainAffiliated Hospital of Goethe‐UniversityFrankfurt am MainGermany
| |
Collapse
|
2
|
Huaqi Y, Bingqi D, Yanhui Z, Yongkang M, Shiming Z, Zhenghui S, Zheng D, Jiangshan P, Tiejun Y. Hyperthermia inhibits cellular function and induces immunogenic cell death in renal cell carcinoma. BMC Cancer 2023; 23:972. [PMID: 37828458 PMCID: PMC10568811 DOI: 10.1186/s12885-023-11106-8] [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: 01/30/2023] [Accepted: 06/23/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND In recent years, hyperthermia has been widely applied as a novel strategy for cancer treatment due to its multiple antitumour effects. In particular, the potential influences of hyperthermia on the tumour immune microenvironment may improve the efficacy of immunotherapies. However, the effect of hyperthermia on renal cell carcinoma (RCC) has not been well characterized until now. METHODS In the present study, we primarily evaluated the effects of hyperthermia on cellular function via cellular proliferation, migration, invasion and apoptosis assays. In addition, the influence of hyperthermia on the immunogenicity of RCC cells was analysed using flow cytometry analysis, enzyme-linked immunosorbent assays, and immunofluorescent (IF) staining. RESULTS Our results demonstrate that hyperthermia significantly inhibits RCC cell proliferation, migration, and invasion and promotes cell apoptosis. In addition, we verified that hyperthermia improves the immunogenicity of RCC cells by inducing immunogenic cell death. CONCLUSION Our findings suggest that hyperthermia is a promising therapeutic strategy for RCC.
Collapse
Affiliation(s)
- Yin Huaqi
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Dong Bingqi
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Zhao Yanhui
- Department of Urology, Qingdao Central Hospital, No. 127, Si Liu Nan Road, Qingdao, 266042, China
| | - Ma Yongkang
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Zhao Shiming
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Sun Zhenghui
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Du Zheng
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Peng Jiangshan
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China
| | - Yang Tiejun
- Department of Urology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No. 127, Dong Ming Road, Zhengzhou, 450000, China.
| |
Collapse
|
3
|
Rodak O, Mrozowska M, Rusak A, Gomułkiewicz A, Piotrowska A, Olbromski M, Podhorska-Okołów M, Ugorski M, Dzięgiel P. Targeting SOX18 Transcription Factor Activity by Small-Molecule Inhibitor Sm4 in Non-Small Lung Cancer Cell Lines. Int J Mol Sci 2023; 24:11316. [PMID: 37511076 PMCID: PMC10379584 DOI: 10.3390/ijms241411316] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
The transcription factor SOX18 has been shown to play a crucial role in lung cancer progression and metastasis. In this study, we investigated the effect of Sm4, a SOX18 inhibitor, on cell cycle regulation in non-small cell lung cancer (NSCLC) cell lines LXF-289 and SK-MES-1, as well as normal human lung fibroblast cell line IMR-90. Our results demonstrated that Sm4 treatment induced cytotoxic effects on all three cell lines, with a greater effect observed in NSCLC adenocarcinoma cells. Sm4 treatment led to S-phase cell accumulation and upregulation of p21, a key regulator of the S-to-G2/M phase transition. While no significant changes in SOX7 or SOX17 protein expression were observed, Sm4 treatment resulted in a significant upregulation of SOX17 gene expression. Furthermore, our findings suggest a complex interplay between SOX18 and p21 in the context of lung cancer, with a positive correlation observed between SOX18 expression and p21 nuclear presence in clinical tissue samples obtained from lung cancer patients. These results suggest that Sm4 has the potential to disrupt the cell cycle and target cancer cell growth by modulating SOX18 activity and p21 expression. Further investigation is necessary to fully understand the relationship between SOX18 and p21 in lung cancer and to explore the therapeutic potential of SOX18 inhibition in lung cancer.
Collapse
Affiliation(s)
- Olga Rodak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Monika Mrozowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Gomułkiewicz
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Mateusz Olbromski
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Maciej Ugorski
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
4
|
Su P, Zhang M, Kang X. Targeting c-Met in the treatment of urologic neoplasms: Current status and challenges. Front Oncol 2023; 13:1071030. [PMID: 36959792 PMCID: PMC10028134 DOI: 10.3389/fonc.2023.1071030] [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: 10/15/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
At present, studies have found that c-Met is mainly involved in epithelial-mesenchymal transition (EMT) of tumor tissues in urologic neoplasms. Hepatocyte growth factor (HGF) combined with c-Met promotes the mitosis of tumor cells, and then induces motility, angiogenesis, migration, invasion and drug resistance. Therefore, c-Met targeting therapy may have great potential in urologic neoplasms. Many strategies targeting c-Met have been widely used in the study of urologic neoplasms. Although the use of targeting c-Met therapy has a strong biological basis for the treatment of urologic neoplasms, the results of current clinical trials have not yielded significant results. To promote the application of c-Met targeting drugs in the clinical treatment of urologic neoplasms, it is very important to study the detailed mechanism of c-Met in urologic neoplasms and innovate c-Met targeted drugs. This paper firstly discussed the value of c-Met targeted therapy in urologic neoplasms, then summarized the related research progress, and finally explored the potential targets related to the HGF/c-Met signaling pathway. It may provide a new concept for the treatment of middle and late urologic neoplasms.
Collapse
|
5
|
Yang R, Zhan Y, Li Y, Dai SY, He SW, Ye CJ, Meng LD, Chen DQ, Dong CB, Chen L, Chen G, Dong KR, Li K, Zheng S, Li J, Yao W, Dong R. The Cellular and Molecular Landscape of Synchronous Pediatric Sialoblastoma and Hepatoblastoma. Front Oncol 2022; 12:893206. [PMID: 35860547 PMCID: PMC9289541 DOI: 10.3389/fonc.2022.893206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/31/2022] [Indexed: 01/05/2023] Open
Abstract
Sialoblastoma (SBL) is an infrequent embryonal malignant tumor originating from the salivary gland, resembling primitive salivary gland anlage, whereas hepatoblastoma (HB) is the most common pediatric liver malignancy. The simultaneous occurrence of both tumors is extremely rare. Here we reported a case of a 6-month-old infant diagnosed with synchronous SBL and HB. The patient received neoadjuvant chemotherapy followed by surgical resection. Fresh tissues of both tumors were collected before and after chemotherapy, which were further profiled by whole exome sequencing (WES) and single-cell RNA sequencing (scRNA-seq). WES analysis revealed potential somatic driver mutation PIK3CA p.Glu454Lys for SBL and canonical mutation CTNNB1 p.Ser45Pro for HB. No shared somatic variants or common copy number alterations were found between SBL and HB primary tumor samples. Though scRNA-seq, single-cell atlases were constructed for both tumors. SBL may recapitulate a pre-acinar stage in the development of salivary gland, including basaloid, duct-like, myoepithelial-like, and cycling phenotypes. In the meantime, HB was composed of tumor cells resembling different stages of the liver, including hepatocyte-like, hepatic progenitor-like, and hepatoblast-like cells. After chemotherapy, both tumors were induced into a more mature phenotype. In terms of transcriptional signatures, SBL and HB showed enhanced expression of epithelial markers KRT8, KRT18, and essential embryo development genes SDC1, MDK, indicating the disruption of normal embryo epithelium development. Finally, heterozygous deleterious germline mutation BLM and FANCI were identified which could predispose the patient to higher cancer risk. It partially explained the reason for the co-occurrence of SBL and HB. Taken together, we provided valuable resources for deciphering cellular heterogeneity and adaptive change of tumor cells after chemotherapy for synchronous SBL and HB, providing insights into the mechanisms leading to synchronous pediatric tumors.
Collapse
Affiliation(s)
- Ran Yang
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yong Zhan
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Yi Li
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shu-Yang Dai
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shi-Wei He
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Chun-Jing Ye
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Ling-Du Meng
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - De-Qian Chen
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Chen-Bin Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Lian Chen
- Department of Pathology, Children’s Hospital of Fudan University, Shanghai, China
| | - Gong Chen
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Kui-Ran Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Kai Li
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
| | - Jun Li
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- *Correspondence: Rui Dong, ; Wei Yao, ; Jun Li,
| | - Wei Yao
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- *Correspondence: Rui Dong, ; Wei Yao, ; Jun Li,
| | - Rui Dong
- Department of Pediatric Surgery, Children’s Hospital of Fudan University, Shanghai Key Laboratory of Birth Defect, Shanghai, China
- *Correspondence: Rui Dong, ; Wei Yao, ; Jun Li,
| |
Collapse
|
6
|
Gao F, Li C, Danopoulos S, Al Alam D, Peinado N, Webster S, Borok Z, Kohbodi GA, Bellusci S, Minoo P. Hedgehog-responsive PDGFRa(+) fibroblasts maintain a unique pool of alveolar epithelial progenitor cells during alveologenesis. Cell Rep 2022; 39:110608. [PMID: 35385750 PMCID: PMC9199394 DOI: 10.1016/j.celrep.2022.110608] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/06/2021] [Accepted: 03/11/2022] [Indexed: 01/08/2023] Open
Abstract
The lung alveolus is lined with alveolar type 1 (AT1) and type 2 (AT2) epithelial cells. During alveologenesis, increasing demand associated with expanding alveolar numbers is met by proliferating progenitor AT2s (pAT2). Little information exists regarding the identity of this population and their niche microenvironment. We show that during alveologenesis, Hedgehog-responsive PDGFRa(+) progenitors (also known as SCMFs) are a source of secreted trophic molecules that maintain a unique pAT2 population. SCMFs are in turn maintained by TGFβ signaling. Compound inactivation of Alk5 TβR2 in SCMFs reduced their numbers and depleted the pAT2 pool without impacting differentiation of daughter cells. In lungs of preterm infants who died with bronchopulmonary dysplasia, PDGFRa is reduced and the number of proliferative AT2s is diminished, indicating that an evolutionarily conserved mechanism governs pAT2 behavior during alveologenesis. SCMFs are a transient cell population, active only during alveologenesis, making them a unique stage-specific niche mesodermal cell type in mammalian organs.
Collapse
Affiliation(s)
- Feng Gao
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Changgong Li
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Soula Danopoulos
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Neil Peinado
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Sha Webster
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Zea Borok
- Hastings Center for Pulmonary Research, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego School of Medicine, San Diego, CA 92093, USA
| | - GoleNaz Adeli Kohbodi
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA
| | - Saverio Bellusci
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA; Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, German Center for Lung Research (DZL), 35390 Giessen, Germany
| | - Parviz Minoo
- Division of Neonatology, Department of Pediatrics, LAC+USC Medical Center and Childrens Hospital Los Angeles, Los Angeles, CA 90033, USA; Hastings Center for Pulmonary Research, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
7
|
Molecular landscape of c-Myc signaling in prostate cancer: A roadmap to clinical translation. Pathol Res Pract 2022; 233:153851. [DOI: 10.1016/j.prp.2022.153851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/02/2022] [Accepted: 03/17/2022] [Indexed: 12/16/2022]
|
8
|
Rezzola S, Sigmund EC, Halin C, Ronca R. The lymphatic vasculature: An active and dynamic player in cancer progression. Med Res Rev 2021; 42:576-614. [PMID: 34486138 PMCID: PMC9291933 DOI: 10.1002/med.21855] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/29/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022]
Abstract
The lymphatic vasculature has been widely described and explored for its key functions in fluid homeostasis and in the organization and modulation of the immune response. Besides transporting immune cells, lymphatic vessels play relevant roles in tumor growth and tumor cell dissemination. Cancer cells that have invaded into afferent lymphatics are propagated to tumor‐draining lymph nodes (LNs), which represent an important hub for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites. In recent years many studies have reported new mechanisms by which the lymphatic vasculature affects cancer progression, ranging from induction of lymphangiogenesis to metastatic niche preconditioning or immune modulation. In this review, we provide an up‐to‐date description of lymphatic organization and function in peripheral tissues and in LNs and the changes induced to this system by tumor growth and progression. We will specifically focus on the reported interactions that occur between tumor cells and lymphatic endothelial cells (LECs), as well as on interactions between immune cells and LECs, both in the tumor microenvironment and in tumor‐draining LNs. Moreover, the most recent prognostic and therapeutic implications of lymphatics in cancer will be reported and discussed in light of the new immune‐modulatory roles that have been ascribed to LECs.
Collapse
Affiliation(s)
- Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena C Sigmund
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Jiang T, Li Z, Zhao D, Hui B, Zheng Z. SOX18 enhances the proliferation and migration of airway smooth muscle cells induced by tumor necrosis factor-α via the regulation of Notch1 signaling. Int Immunopharmacol 2021; 96:107746. [PMID: 34004439 DOI: 10.1016/j.intimp.2021.107746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 02/06/2023]
Abstract
Childhood asthma is a frequent chronic disease of pediatric populations. The excessive proliferation and migration of airway smooth muscle cells contribute to airway remodeling during asthma pathogenesis. Sex-determining region on the Y chromosome-related high mobility group box 18 (SOX18) has been reported to be over-expressed in asthma. However, whether SOX18 plays a role in modulating the airway remodeling of asthma is not fully understood. The purposes of this work were to assess the potential role of SOX18 in modulating airway remodeling using tumor necrosis factor-α (TNF-α)-stimulated airway smooth muscle cells in vitro. Our results showed that SOX18 expression was increased following TNF-α stimulation in airway smooth muscle cells. The silencing of SOX18 markedly prohibited the proliferation and migration of airway smooth muscle cells induced by TNF-α, whilst the over-expression of SOX18 produced the opposite effects. Further investigation revealed that SOX18 promoted the expression of Notch1, and enhanced the activation of Notch1 signaling in airway smooth muscle cells stimulated by TNF-α. The inhibition of Notch1 markedly diminished SOX18-over-expression-evoked promotion effects on TNF-α-induced proliferation and migration of airway smooth muscle cells. In addition, the reactivation of Notch1 signaling markedly reversed the SOX18-silencing-induced suppressive effect on the TNF-α-induced proliferation and the migration of airway smooth muscle cells. In summary, the findings of this work demonstrate that SOX18 regulates the proliferation and migration of airway smooth muscle cells induced by TNF-α via the modulation of Notch1 signaling. This study indicates a potential role for SOX18 in promoting airway remodeling during asthma pathogenesis.
Collapse
Affiliation(s)
- Te Jiang
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China
| | - Zhankui Li
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China.
| | - Di Zhao
- Pediatrics, Northwest Women's and Children's Hospital, Xi'an 610113, China
| | - Bengang Hui
- Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China
| | - Zhiyuan Zheng
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University and Shanghai Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
10
|
Wang L, Li Y, Lyu Y, Wen H, Feng C. Association between copy-number alteration of +20q, -14q and -18p and cross-sensitivity to tyrosine kinase inhibitors in clear-cell renal cell carcinoma. Cancer Cell Int 2020; 20:482. [PMID: 33041663 PMCID: PMC7541266 DOI: 10.1186/s12935-020-01585-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/29/2020] [Indexed: 12/30/2022] Open
Abstract
Background We aim to explore association between copy number alteration (CNA) and sensitivity to common tyrosine kinase inhibitors (TKIs) used in clear-cell renal cell carcinoma (ccRCC) treatment. Methods CNA with related sensitivity profiles were extracted from the Genomics of Drug Sensitivity in Cancer (GDSC) dataset and was cross-referenced with common CNA in ccRCC in the Cancer Genome Atlas (TCGA) dataset. Functional annotation was profiled using GSEA and NET-GE. Target genes within cytobands of interest were screened in silico and validated in vitro using proliferation assays in A498 and 786-O ccRCC cells. Results Four TKIs (Sunitinib, Cabozantinib, Axitinib and Sorafenib) that were clinically used in ccRCC were selected. In silico analysis showed gain of 20q (+20q) occurred in ~ 23% of cases and was associated with resistance to all four TKIs; loss of 14q (−14q) occurred in ~ 39% of cases and was associated with resistance to Sunitinib and Sorafenib; loss of 18p (−18p) occurred in ~ 39% of cases and was associated with sensitivity to Sunitinib and Sorafenib. All 3 CNAs were associated with worsened prognosis, respectively. Candidate target genes included of RBL1 on 20q, KLHL33 on 14q and ARHGAP28 on18q. In vitro validation showed RBL1 overexpression induced resistance to Sunitinib and Cabozantinib; KLHL33 silencing induced resistance to Sunitinib; ARHGAP28 silencing induced sensitivity to Cabozantinib. Functional annotation indicated FoxO signaling, hypoxic response and Wnt pathway, and Rho-related cellular adhesion were mechanistically associated with +20q, −14q and −18p, respectively. Conclusion Common CNAs in ccRCC are associated with cancer-intrinsic cross-sensitivity to common TKIs. Further validation and functional analyses are therefore needed.
Collapse
Affiliation(s)
- Liang Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, 300052 People's Republic of China
| | - Yuqing Li
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Yinfeng Lyu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Hui Wen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| | - Chenchen Feng
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200040 People's Republic of China
| |
Collapse
|
11
|
Geng Q, Deng H, Fu J, Cui F. SOX18 exerts tumor-suppressive functions in papillary thyroid carcinoma through inhibition of Wnt/β-catenin signaling. Exp Cell Res 2020; 396:112249. [PMID: 32858034 DOI: 10.1016/j.yexcr.2020.112249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 01/16/2023]
Abstract
Sex-determining region on the Y chromosome-related high mobility group box 18 (SOX18) has emerged as a key tumor-related protein in a wide range of human tumors. Yet, the involvement of SOX18 in papillary thyroid carcinoma has not been determined. This study aimed to explore the expression and biological function of SOX18 in papillary thyroid carcinoma. There was a significant decrease in SOX18 expression in papillary thyroid carcinoma tissues compared with that in normal tissues. Low expression of SOX18 was also detected in papillary thyroid carcinoma cell lines and upregulation of SOX18 effectively repressed the proliferative, colony-forming and invasive abilities of papillary thyroid carcinoma cells in vitro. In contrast, knockdown of SOX18 in papillary thyroid carcinoma cells was associated with a significant increase in cell proliferation and invasion. Further studies revealed that SOX18 upregulation was associated with the reduced nuclear accumulation of β-catenin and the downregulation of Wnt/β-catenin signaling in thyroid carcinoma cells. Moreover, inhibition of Wnt/β-catenin signaling markedly attenuated SOX18 knockdown-evoked oncogenic effects in papillary thyroid carcinoma cells. In addition, SOX18 overexpression remarkably retarded the tumor growth of papillary thyroid carcinoma cell-derived xenograft tumors in nude mice. Taken together, these results demonstrate that SOX18 suppresses the proliferation and invasion of papillary thyroid carcinoma by inhibiting Wnt/β-catenin signaling. Our study reveals a tumor-suppressive role of SOX18 in papillary thyroid carcinoma and suggests that SOX18 is an attractive candidate target for treatment of papillary thyroid carcinoma.
Collapse
Affiliation(s)
- Qianqian Geng
- Nuclear Medicine Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Huixing Deng
- Nuclear Medicine Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jiao Fu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Feibo Cui
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
12
|
Rathi N, Maughan BL, Agarwal N, Swami U. Mini-Review: Cabozantinib in the Treatment of Advanced Renal Cell Carcinoma and Hepatocellular Carcinoma. Cancer Manag Res 2020; 12:3741-3749. [PMID: 32547210 PMCID: PMC7246323 DOI: 10.2147/cmar.s202973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
Cabozantinib is an oral, tyrosine-kinase inhibitor with potent activity against VEGFR2 and MET, along with multiple other tyrosine kinases involved in cancer development and progression. Herein, we will focus on preclinical and clinical studies leading to the approval of cabozantinib in advanced renal cell carcinoma and hepatocellular carcinoma. Covered studies include NCT01100619, CABOSUN, METEOR, NCT00940225 and the CELESTIAL trial. Finally, we review future directions of cabozantinib development by highlighting some ongoing clinical trials.
Collapse
Affiliation(s)
- Nityam Rathi
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L Maughan
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|