1
|
Liao D, He Y, He B, Zeng S, Cui Y, Li C, Huang H. Inhibiting SNX10 induces autophagy to suppress invasion and EMT and inhibits the PI3K/AKT pathway in cervical cancer. Clin Transl Oncol 2025; 27:2084-2094. [PMID: 39367898 DOI: 10.1007/s12094-024-03715-x] [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: 06/02/2024] [Accepted: 09/02/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE Cervical cancer (CC) is a prevalent malignancy among women with high morbidity and poor prognosis. Sorting nexin 10 (SNX10) is a newly recognized cancer regulatory factor, while its action on CC progression remains elusive. Hence, this study studied the effect of SNX10 on CC development and investigated the mechanism. METHODS The SNX10 level in CC and the overall survival of CC cases with different SNX10 expressions were determined by bioinformatics analysis in GEPIA. The SNX10 expression in tumor tissues and clinical significance were studied in 64 CC cases. The overall survival was assessed using Kaplan-Meier analysis. The formation of LC3 was evaluated using immunofluorescence. Cell invasion was measured using the Transwell assay. Epithelial-to-mesenchymal transition (EMT) was determined by observing cell morphology and assessing EMT marker levels. A xenograft tumor was constructed to evaluate tumor growth. RESULTS SNX10 was elevated in CC tissues and cells, and the CC cases with high SNX10 levels exhibited poor overall survival. Besides, SNX10 correlated with the FIGO stage, lymph node invasion, and stromal invasion of CC. SNX10 silencing induced CC cell autophagy and suppressed CC cell invasion and EMT. Meanwhile, silenced SNX10 could suppress invasion and EMT via inducing autophagy. Furthermore, SNX10 inhibition suppressed the PI3K/AKT pathway. Moreover, silenced SNX10 restrained the tumor growth, autophagy, and EMT of CC in vivo. CONCLUSION SNX10 was enhanced in CC and correlated with poor prognosis. Silenced SNX10 induced autophagy to suppress invasion and EMT and inhibited the PI3K/AKT pathway in CC, making SNX10 a valuable molecule for CC therapy.
Collapse
Affiliation(s)
- Dan Liao
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| | - Yanxian He
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Bin He
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Saitian Zeng
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Yejia Cui
- Department of Clinical Laboratory, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Cuifen Li
- Department of Gynaecology, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China
| | - Haohai Huang
- Clinical Translational Medical Center, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan, Guangdong, China.
- Department of Clinical Pharmacy, Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, No.1, Huangzhou Xianglong Road of Shilong Town, Dongguan, 523326, Guangdong, China.
| |
Collapse
|
2
|
Li H, Li Y, Chen Z, He C. HOXA3 activates USP15 to suppress autophagy and promote M2-type macrophage polarization in renal cell carcinoma via facilitating the deubiquitination of SQSTM1. Am J Physiol Cell Physiol 2025; 328:C576-C594. [PMID: 39740793 DOI: 10.1152/ajpcell.00712.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/27/2024] [Accepted: 12/08/2024] [Indexed: 01/02/2025]
Abstract
The disease burden of renal cell carcinoma (RCC) has decreased in recent years with advances in treatment, but its pathogeny still remains elusive. We aim to study the role of homeobox A3 (HOXA3)/ubiquitin-specific peptidase 15 (USP15)/SQSTM1 axis on autophagy and M2-type macrophage polarization in RCC. In this study, cell apoptosis and proliferation were assessed by flow cytometry and CCK-8. Autolysosome fusion was observed by immunofluorescence detection of LC3 and LAMP2. The binding between HOXA3 and USP15 promoter was tested by chromatin immunoprecipitation (ChIP), EMSA, and dual-luciferase reporter assays. Also, the interaction between deubiquitinated enzyme (DUB) USP15 and SQSTM1, and ubiquitinated level of SQSTM1 were determined by co-immunoprecipitation (Co-IP) assay. Expression levels of HOXA3, USP15, C-C motif chemokine 2 (CCL2), CCL2 receptor (CCR2), M2-type macrophages, and autophagy-related markers were measured by Western blot, quantitative reverse transcription PCR (RT-qPCR), ELISA, and immunohistochemistry. Role of HOXA3/USP15 axis was verified by xenograft tumor experiment in vivo. We showed upregulated HOXA3 in RCC tissues and cells, and RCC tissues with metastasis showed higher HOXA3 level. The higher HOXA3 expression was relevant to worse overall survival in patients with RCC. HOXA3 induced RCC cell proliferation, and suppressed autophagy and apoptosis via transcriptionally activating USP15 expression. USP15 then induced deubiquitination modification of SQSTM1 in RCC cells. SQSTM1 supported M2-type macrophage polarization by inducing CCL2 secretion. HOXA3 or USP15 knockdown suppressed tumor growth and M2-type macrophage infiltration in vivo. In conclusion, HOXA3 transcriptionally activates USP15 expression, and upregulated USP15 facilitates the deubiquitination of SQSTM1 in RCC. This process on the one hand suppresses autophagy, on the other hand increases M2-type macrophage polarization through stimulating the secretion of CCL2.NEW & NOTEWORTHY We report a novel finding that highly expressed homeobox A3 (HOXA3) transcriptionally activates the expression of ubiquitin-specific peptidase 15 (USP15), resulting in the promotion of deubiquitination of SQSTM1. This process on the one hand suppresses autophagy in renal cell carcinoma (RCC), on the other hand increases M2-type macrophage polarization in the tumor microenvironment through stimulating the secretion of C-C motif chemokine 2 (CCL2).
Collapse
Affiliation(s)
- Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhiyong Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Cheng He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
3
|
Hu J, Cao J, Huang S, Chen Y. ITGAX promotes gastric cancer progression via epithelial-mesenchymal transition pathway. Front Pharmacol 2025; 15:1536478. [PMID: 39845786 PMCID: PMC11750855 DOI: 10.3389/fphar.2024.1536478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Gastric cancer is the fifth most common cancer and the fourth leading cause of cancer-related deaths worldwide, accounting for nearly 800,000 fatalities annually. ITGAX (Integrin alpha X) is closely associated with immune cells, such as macrophages and dendritic cells. Its involvement in gastric cancer was identified through an analysis of The Gene Expression Omnibus (GEO) database, which highlighted ITGAX as one of four key gastric cancer-related genes. Our study demonstrates that ITGAX expression is significantly elevated in tumor tissues compared to normal tissues and is positively correlated with clinical prognosis in gastric cancer patients from the GEO database. Moreover, ITGAX enhanced cell proliferation, invasion, and tumorigenic capacity in mouse models. Furthermore, we explored the underlying role of ITGAX using Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction networks (PPI) analysis. Our findings reveal that ITGAX promotes gastric cancer progression by driving epithelial-mesenchymal transition pathway (EMT), suggesting its potential as a biomarker for early diagnosis and prognosis in gastric cancer.
Collapse
Affiliation(s)
- Jiali Hu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Cao
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Shanshan Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yang Chen
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
- Key Laboratory of Phytochemistry and Natural Medicines, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| |
Collapse
|
4
|
Demir K, Turgut R, Şentürk S, Işıklar H, Günalan E. The Therapeutic Effects of Bioactive Compounds on Colorectal Cancer via PI3K/Akt/mTOR Signaling Pathway: A Critical Review. Food Sci Nutr 2024; 12:9951-9973. [PMID: 39723045 PMCID: PMC11666977 DOI: 10.1002/fsn3.4534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 12/28/2024] Open
Abstract
Understanding the molecular signaling pathways of colorectal cancer (CRC) can be accepted as the first step in treatment strategy. Permanent mTOR signaling activation stimulates the CRC process via various biological processes. It supplies the survival of CRC stem cells, tumorigenesis, morbidity, and decreased response to drugs in CRC pathogenesis. Therefore, inhibition of the mTOR signaling by numerous bioactive components may be effective against CRC. The study aims to discuss the therapeutic capacity of various polyphenols, terpenoids, and alkaloids on CRC via the PI3K/Akt/mTOR pathway. The potential molecular effects of bioactive compounds on the mTOR pathway's upstream and downstream targets are examined. Each bioactive component causes various physiological processes, such as triggering free radical production, disruption of mitochondrial membrane potential, cell cycle arrest, inhibition of CRC stem cell migration, and suppression of glycolysis through mTOR signaling inhibition. As a result, carcinogenesis is inhibited by inducing apoptosis and autophagy. However, it should be noted that studies are primarily in vitro dose-dependent treatment researchers. This study raises awareness about the role of phenolic compounds in treating CRC, contributing to their future use as anticancer agents. These bioactive compounds have the potential to be developed into food supplementation to prevent and treat various cancer types including CRC. This review has the potential to lead to further development of clinical studies. In the future, mTOR inhibition by applying several bioactive agents using advanced drug delivery systems may contribute to CRC treatment with 3D cell culture and in vivo clinical studies.
Collapse
Affiliation(s)
- Kübra Demir
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
- Faculty of Health Science, Department of Nutrition and DieteticsSabahattin Zaim UniversityIstanbulTürkiye
| | - Rana Turgut
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Selcen Şentürk
- Institute of Graduate EducationIstanbul Health and Technology UniversityIstanbulTürkiye
| | - Handan Işıklar
- Faculty of Medicine, Department of Internal MedicineYalova UniversityYalovaTürkiye
| | - Elif Günalan
- Faculty of Health Science, Department of Nutrition and DieteticsIstanbul Health and Technology UniversityIstanbulTürkiye
| |
Collapse
|
5
|
Andretta E, Costa A, Ventura E, Quintiliani M, Damiano S, Giordano A, Morrione A, Ciarcia R. Capsaicin Exerts Antitumor Activity in Mesothelioma Cells. Nutrients 2024; 16:3758. [PMID: 39519591 PMCID: PMC11547426 DOI: 10.3390/nu16213758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Mesothelioma is an aggressive cancer with limited treatment options. Mesothelioma therapy often involves a multimodal approach including surgery, radiotherapy and chemotherapy. However, the prognosis for patients remains poor. Difficult diagnosis, late symptoms when the tumor is in an advanced stage and the onset of chemotherapy resistance make mesothelioma difficult to treat. For this reason, it is essential to discover new pharmacological approaches. Capsaicin (CAPS) is the active compound of chili peppers. Based on CAPS's anticancer properties on various tumor lines and its chemo-sensitizing action on resistant cells, in this study, we evaluated the effects of CAPS on mesothelioma cells to assess its potential use in mesothelioma therapy. METHODS To evaluate antiproliferative effects of CAPS, we performed MTS assays on various mesothelioma cells, representative of all major mesothelioma subtypes. Transwell migration and wound-healing assays were used to examine the effect of CAPS on mesothelioma cell migration. We also determined the effects of CAPS on oncogenic signaling pathways by assessing the levels of AKT and MAPK activation. RESULTS In this study, we show that CAPS significantly reduces proliferation of both parental and cisplatin-resistant mesothelioma cells. CAPS promotes S-phase cell cycle arrest and inhibits lateral motility and migration of mesothelioma cells. Accordingly, CAPS suppresses AKT and ERK1/2 activation in MSTO-211H and NCI-H2052 cells. Our results support an antitumor effect of CAPS on cisplatin-resistant mesothelioma cells, suggesting that it may reduce resistance to cisplatin. CONCLUSIONS Our results could pave the way for further studies to evaluate the use of CAPS for mesothelioma treatment.
Collapse
Affiliation(s)
- Emanuela Andretta
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.A.); (A.C.); (E.V.); (A.G.)
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (R.C.)
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 4, 80126 Naples, Italy
| | - Aurora Costa
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.A.); (A.C.); (E.V.); (A.G.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Elisa Ventura
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.A.); (A.C.); (E.V.); (A.G.)
| | | | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (R.C.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.A.); (A.C.); (E.V.); (A.G.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA; (E.A.); (A.C.); (E.V.); (A.G.)
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, 80137 Naples, Italy; (S.D.); (R.C.)
| |
Collapse
|
6
|
Zhang Z, Shan X, Li S, Chang J, Zhang Z, Dong Y, Wang L, Liang F. Retinal light damage: From mechanisms to protective strategies. Surv Ophthalmol 2024; 69:905-915. [PMID: 39053594 DOI: 10.1016/j.survophthal.2024.07.004] [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: 04/23/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Visible light serves as a crucial medium for vision formation.;however, prolonged or excessive exposure to light is recognized as a significant etiological factor contributing to retinal degenerative diseases. The retina, with its unique structure and adaptability, relies on the homeostasis of cellular functions to maintain visual health. Under normal conditions, the retina can mount adaptive responses to various insults, including light-induced damage. Unfortunately, exposure to intense and excessive light triggers a cascade of pathological alterations in retinal photoreceptor cells, pigment epithelial cells, ganglion cells, and glial cells. These alterations encompass disruption of intracellular REDOX and Ca²⁺ homeostasis, pyroptosis, endoplasmic reticulum stress, autophagy, and the release of inflammatory cytokines, culminating in irreversible retinal damage. We first delineate the mechanisms of retinal light damage through 4 main avenues: mitochondria function, endoplasmic reticulum stress, cell autophagy, and inflammation. Subsequently, we discuss protective strategies against retinal light damage, aiming to guide research toward the prevention and treatment of light-induced retinal conditions.
Collapse
Affiliation(s)
- Zhao Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Xiaoqian Shan
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shujiao Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100040, China
| | - Jun Chang
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Zhenhua Zhang
- Tongliang District Hospital of Traditional Chinese Medicine, Chongqing 402560, China
| | - Yang Dong
- Ji'nan Hospital of Traditional Chinese Medicine, Jinan, 250002, China
| | - Li Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Fengming Liang
- Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
7
|
Penugurti V, Manne RK, Bai L, Kant R, Lin HK. AMPK: The energy sensor at the crossroads of aging and cancer. Semin Cancer Biol 2024; 106-107:15-27. [PMID: 39197808 PMCID: PMC11625618 DOI: 10.1016/j.semcancer.2024.08.002] [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: 07/21/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024]
Abstract
AMP-activated protein kinase (AMPK) is a protein kinase that plays versatile roles in response to a variety of physiological stresses, including glucose deprivation, hypoxia, and ischemia. As a kinase with pleiotropic functions, it plays a complex role in tumor progression, exhibiting both tumor-promoting and tumor-suppressing activities. On one hand, AMPK enhances cancer cell proliferation and survival, promotes cancer metastasis, and impairs anti-tumor immunity. On the other hand, AMPK inhibits cancer cell growth and survival and stimulates immune responses in a context-dependent manner. Apart from these functions, AMPK plays a key role in orchestrating aging and aging-related disorders, including cardiovascular diseases (CVD), Osteoarthritis (OA), and Diabetes. In this review article, we summarized the functions of AMPK pathway based on its oncogenic and tumor-suppressive roles and highlighted the importance of AMPK pathway in regulating cellular aging. We also spotlighted the significant role of various signaling pathways, activators, and inhibitors of AMPK in serving as therapeutic strategies for anti-cancer and anti-aging therapy.
Collapse
Affiliation(s)
- Vasudevarao Penugurti
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Rajesh Kumar Manne
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Ling Bai
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Rajni Kant
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States
| | - Hui-Kuan Lin
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27710, United States.
| |
Collapse
|
8
|
Yuan Z, He J, Li Z, Fan B, Zhang L, Man X. Targeting autophagy in urological system cancers: From underlying mechanisms to therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189196. [PMID: 39426690 DOI: 10.1016/j.bbcan.2024.189196] [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: 07/29/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The urological system, including kidneys, ureters, bladder, urethra and prostate is known to be vital for blood filtration, waste elimination and electrolyte balance. Notably, urological system cancers represent a significant portion of global cancer diagnoses and mortalities. The current therapeutic strategies for early-stage cancer primarily involve resection surgery, which significantly affects the quality of life of patients, whereas advanced-stage cancer often relies on less effective chemo- or radiotherapy. Recently, accumulating evidence has revealed that autophagy, a crucial process in which excess organelles or inclusions within cells are removed to maintain cell homeostasis, has numerous links to urological system cancers. In this review, we focus on summarizing the underlying two-sided mechanisms of autophagy in urological system cancers. We also review the current clinical drugs targeting autophagy, which demonstrate significant potential in improving treatment outcomes for urological system cancers. In addition, we provide an overview of the research status of novel small molecule compounds targeting autophagy that are in the preclinical stages of investigation. Furthermore, drug combinations based on autophagy modulation strategies in urological system cancers are systematically summarized and discussed. These findings provide comprehensive new insight for the future discovery of more autophagy-related drug candidates.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiani He
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Fan
- Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Xiaojun Man
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
9
|
Zhou X, Liu Q, Li Z, Liu X, Zhao Q, Wang Y, Wu F, Zhao G, Sun R, Guo X. The activation of adenosine monophosphate-activated protein kinase inhibits the migration of tongue squamous cell carcinoma cells by targeting Claudin-1 via epithelial-mesenchymal transition. Animal Model Exp Med 2024; 7:606-616. [PMID: 39017036 PMCID: PMC11528389 DOI: 10.1002/ame2.12444] [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: 02/26/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND The role of Claudin-1 in tongue squamous cell carcinoma (TSCC) metastasis needs further clarification, particularly its impact on cell migration. Herein, our study aims to investigate the role of Claudin-1 in TSCC cell migration and its underlying mechanisms. METHODS 36 TSCC tissue samples underwent immunohistochemical staining for Claudin-1. Western blotting and immunofluorescence analyses were conducted to evaluate Claudin-1 expression and distribution in TSCC cells. Claudin-1 knockdown cell lines were established using short hairpin RNA transfection. Migration effects were assessed through wound healing assays. Furthermore, the expression of EMT-associated molecules was measured via western blotting. RESULTS Claudin-1 expression decreased as TSCC malignancy increased. Adenosine monophosphate-activated protein kinase (AMPK) activation led to increased Claudin-1 expression and membrane translocation, inhibiting TSCC cell migration and epithelial-mesenchymal transition (EMT). Conversely, Claudin-1 knockdown reversed these inhibitory effects on migration and EMT caused by AMPK activation. CONCLUSIONS Our results indicated that AMPK activation suppresses TSCC cell migration by targeting Claudin-1 and EMT pathways.
Collapse
Affiliation(s)
- Xin‐Yue Zhou
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
- Hubei Shizhen LaboratoryWuhanHubeiChina
| | - Qiu‐Ming Liu
- Sino‐German Biomedical CenterHubei University of TechnologyWuhanChina
- Center of Applied BiotechnologyWuhan Institute of BioengineeringWuhanChina
| | - Zhuang Li
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Xia‐Yang Liu
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Qi‐Wei Zhao
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Yu Wang
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Feng‐Hua Wu
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
- Hubei Shizhen LaboratoryWuhanHubeiChina
| | - Gang Zhao
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
| | - Rui Sun
- Department of Stomatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
- Department of Oral and Maxillofacial SurgeryShanxi Provincial People's HospitalTaiyuanChina
| | - Xiao‐Hong Guo
- Department of Basic MedicineHubei University of Chinese MedicineWuhanChina
- Hubei Shizhen LaboratoryWuhanHubeiChina
| |
Collapse
|
10
|
Al Azzani M, Nizami ZN, Magramane R, Sekkal MN, Eid AH, Al Dhaheri Y, Iratni R. Phytochemical-mediated modulation of autophagy and endoplasmic reticulum stress as a cancer therapeutic approach. Phytother Res 2024; 38:4353-4385. [PMID: 38961675 DOI: 10.1002/ptr.8283] [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: 01/20/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Autophagy and endoplasmic reticulum (ER) stress are conserved processes that generally promote survival, but can induce cell death when physiological thresholds are crossed. The pro-survival aspects of these processes are exploited by cancer cells for tumor development and progression. Therefore, anticancer drugs targeting autophagy or ER stress to induce cell death and/or block the pro-survival aspects are being investigated extensively. Consistently, several phytochemicals have been reported to exert their anticancer effects by modulating autophagy and/or ER stress. Various phytochemicals (e.g., celastrol, curcumin, emodin, resveratrol, among others) activate the unfolded protein response to induce ER stress-mediated apoptosis through different pathways. Similarly, various phytochemicals induce autophagy through different mechanisms (namely mechanistic target of Rapamycin [mTOR] inhibition). However, phytochemical-induced autophagy can function either as a cytoprotective mechanism or as programmed cell death type II. Interestingly, at times, the same phytochemical (e.g., 6-gingerol, emodin, shikonin, among others) can induce cytoprotective autophagy or programmed cell death type II depending on cellular contexts, such as cancer type. Although there is well-documented mechanistic interplay between autophagy and ER stress, only a one-way modulation was noted with some phytochemicals (carnosol, capsaicin, cryptotanshinone, guangsangon E, kaempferol, and δ-tocotrienol): ER stress-dependent autophagy. Plant extracts are sources of potent phytochemicals and while numerous phytochemicals have been investigated in preclinical and clinical studies, the search for novel phytochemicals with anticancer effects is ongoing from plant extracts used in traditional medicine (e.g., Origanum majorana). Nonetheless, the clinical translation of phytochemicals, a promising avenue for cancer therapeutics, is hindered by several limitations that need to be addressed in future studies.
Collapse
Affiliation(s)
- Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rym Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed N Sekkal
- Department of Surgery, Specialty Orthopedic, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
11
|
Li Z, Zhao Q, Liu X, Zhou X, Wang Y, Zhao M, Wu F, Zhao G, Guo X. Capsaicin combined with cisplatin inhibits TGF-β1-induced EMT and TSCC cells migration via the Claudin-1/PI3K/AKT/mTOR signaling pathway. Cancer Cell Int 2024; 24:300. [PMID: 39198820 PMCID: PMC11360848 DOI: 10.1186/s12935-024-03485-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Tongue squamous cell carcinoma (TSCC) is one of the most common malignant tumors among oral cancers, and its treatment is based on radio-chemotherapy and surgery, which always produces more serious side effects and sequelae. Traditional medicine can compensate for the shortcomings of modern medical treatments and play a better therapeutic role. Currently, active ingredients derived from plants are attracting the attention of researchers and clinical professionals. We examined capsaicin (CAP), an active ingredient isolated from Capsicum annuum (family Solanaceae), and explored the effect of CAP combined with cisplatin (DDP) on epithelial-mesenchymal transition (EMT) and TSCC cells migration. Our results demonstrated that Transforming growth factor-β1(TGF-β1) induced EMT and promoted cell migration in TSCC cells. CAP combined with DDP inhibits non-TGF-β1-induced or TGF-β1-induced EMT and migration. Mechanistically, the inhibition of non-TGF-β1-induced EMT and migration by CAP combined with DDP was mediated by the AMPK/mTOR pathway, whereas TGF-β1-induced EMT and migration were regulated by the Claudin-1/PI3K/AKT/mTOR pathway. A nude lung metastasis mouse model was established for in vivo validation. These results support our hypothesis that the combination of CAP and DDP inhibits TSCC metastasis. These data set the stage for further studies aimed at validating CAP as an effective active ingredient for enhancing chemotherapy efficacy and reducing the dosage and toxicity of chemotherapeutic drugs, ultimately paving the way for translational research and clinical trials for TSCC eradication.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Qiwei Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Xiayang Liu
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Xinyue Zhou
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Yu Wang
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Min Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Fenghua Wu
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China
| | - Gang Zhao
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China
| | - Xiaohong Guo
- Department of Medical Biology, School of Basic Medicine Sciences, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Wuhan, 430065, Hubei, P.R. China.
- Hubei Shizhen Laboratory, Wuhan, 430065, Hubei, P.R. China.
| |
Collapse
|
12
|
Petran EM, Periferakis A, Troumpata L, Periferakis AT, Scheau AE, Badarau IA, Periferakis K, Caruntu A, Savulescu-Fiedler I, Sima RM, Calina D, Constantin C, Neagu M, Caruntu C, Scheau C. Capsaicin: Emerging Pharmacological and Therapeutic Insights. Curr Issues Mol Biol 2024; 46:7895-7943. [PMID: 39194685 DOI: 10.3390/cimb46080468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Capsaicin, the most prominent pungent compound of chilli peppers, has been used in traditional medicine systems for centuries; it already has a number of established clinical and industrial applications. Capsaicin is known to act through the TRPV1 receptor, which exists in various tissues; capsaicin is hepatically metabolised, having a half-life correlated with the method of application. Research on various applications of capsaicin in different formulations is still ongoing. Thus, local capsaicin applications have a pronounced anti-inflammatory effect, while systemic applications have a multitude of different effects because their increased lipophilic character ensures their augmented bioavailability. Furthermore, various teams have documented capsaicin's anti-cancer effects, proven both in vivo and in vitro designs. A notable constraint in the therapeutic effects of capsaicin is its increased toxicity, especially in sensitive tissues. Regarding the traditional applications of capsaicin, apart from all the effects recorded as medicinal effects, the application of capsaicin in acupuncture points has been demonstrated to be effective and the combination of acupuncture and capsaicin warrants further research. Finally, capsaicin has demonstrated antimicrobial effects, which can supplement its anti-inflammatory and anti-carcinogenic actions.
Collapse
Affiliation(s)
- Elena Madalina Petran
- Department of Biochemistry, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Toxicology, Grigore Alexandrescu Emergency Children's Hospital, 011743 Bucharest, Romania
| | - Argyrios Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs (P.O.E.P), 17236 Athens, Greece
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, The "Carol Davila" Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, "Titu Maiorescu" University, 031593 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Romina-Marina Sima
- Department of Obstetrics and Gynecology, The "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- The "Bucur" Maternity, "Saint John" Hospital, 040294 Bucharest, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Carolina Constantin
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
- Faculty of Biology, University of Bucharest, 76201 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, "Prof. N.C. Paulescu" National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, "Foisor" Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
13
|
Wang H, Zhang Z, Wu S, Zhu Y, Liang T, Huang X, Yao J. Dietary patterns suggest that dark chocolate intake may have an inhibitory effect on oral cancer: a Mendelian randomization study. Front Nutr 2024; 11:1342163. [PMID: 39027665 PMCID: PMC11255456 DOI: 10.3389/fnut.2024.1342163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/22/2024] [Indexed: 07/20/2024] Open
Abstract
Background Previous studies reported that variations in dietary intake patterns substantially impact human health, specifically tumorigenesis. However, confounding factors in previous cohort studies have obscured the relationship between dietary differences and the risk of oral cancer (OC). Materials and methods We developed an outcome dataset from genome-wide association studies (GWAS) data on three OCs within the GAME-ON project, using GWAS-META merging. We extracted 21 dietary exposures, including 10 dietary patterns, 6 vitamins, and 5 micronutrients, from the UK Biobank database, using the inverse variance weighting method as the primary statistical method. Sensitivity analysis was conducted to detect heterogeneity and pleiotropy. Serum metabolite concentrations were adjusted using multivariate Mendelian randomization. Results Of the 10 analyzed dietary patterns, 8 showed no significant association with the risk of developing OC. Consumption of dark chocolate (inverse variance weighted [IVW]: Odds ratio (OR) = 0.786, 95% confidence interval [CI]: 0.622-0.993, p = 0.044) and sweet pepper exhibited an inverse relationship with OC risk (IVW: OR = 0.757, 95% CI: 0.574-0.997, p = 0.048). Reverse MR analysis revealed no reverse causality. Furthermore, no significant correlation was observed between the intake of 6 vitamins and 5 micronutrients and the risk of developing OC. After using multivariable MR to adjust for serum caffeine, linoleate, theophylline, and theobromine metabolism levels, consuming dark chocolate was unrelated to a decreased risk of OC. After adjusting each serum metabolite individually, the observed p-values deviated from the original values to varying degrees, indicating that the components of dark chocolate could have different effects. Among these components, theophylline demonstrated the most significant inhibitory effect. Conclusion This study demonstrated a causal relationship between the intake of dark chocolate and sweet peppers and a lower risk of OC. The components of dark chocolate could have different effects.
Collapse
Affiliation(s)
- Hongwei Wang
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Youjiang Medical University for Nationalities, Baise, China
- Department of Tumor Pathology, The Key Laboratory of Molecular Pathology (Hepatobiliary Diseases) of Guangxi, Baise, Guangxi, China
| | - Zhaoyin Zhang
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Youjiang Medical University for Nationalities, Baise, China
- Department of Tumor Pathology, The Key Laboratory of Molecular Pathology (Hepatobiliary Diseases) of Guangxi, Baise, Guangxi, China
| | - Sijie Wu
- Guilin Medical University, Guilin, Guangxi, China
| | - Yuanzhi Zhu
- Youjiang Medical University for Nationalities, Baise, China
| | - Tao Liang
- Youjiang Medical University for Nationalities, Baise, China
| | - Xiong Huang
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Youjiang Medical University for Nationalities, Baise, China
- Department of Tumor Pathology, The Key Laboratory of Molecular Pathology (Hepatobiliary Diseases) of Guangxi, Baise, Guangxi, China
| | - Jinguang Yao
- Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
- Youjiang Medical University for Nationalities, Baise, China
- Department of Tumor Pathology, The Key Laboratory of Molecular Pathology (Hepatobiliary Diseases) of Guangxi, Baise, Guangxi, China
| |
Collapse
|
14
|
Zhang Z, Liang F, Chang J, Shan X, Yin Z, Wang L, Li S. Autophagy in dry AMD: A promising therapeutic strategy for retinal pigment epithelial cell damage. Exp Eye Res 2024; 242:109889. [PMID: 38593971 DOI: 10.1016/j.exer.2024.109889] [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: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
Dry age-related macular degeneration (AMD) is a prevalent clinical condition that leads to permanent damage to central vision and poses a significant threat to patients' visual health. Although the pathogenesis of dry AMD remains unclear, there is consensus on the role of retinal pigment epithelium (RPE) damage. Oxidative stress and chronic inflammation are major contributors to RPE cell damage, and the NOD-like receptor thermoprotein structural domain-associated protein 3 (NLRP3) inflammasome mediates the inflammatory response leading to apoptosis in RPE cells. Furthermore, lipofuscin accumulation results in oxidative stress, NLRP3 activation, and the development of vitelliform lesions, a hallmark of dry AMD, all of which may contribute to RPE dysfunction. The process of autophagy, involving the encapsulation, recognition, and transport of accumulated proteins and dead cells to the lysosome for degradation, is recognized as a significant pathway for cellular self-protection and homeostasis maintenance. Recently, RPE cell autophagy has been discovered to be closely linked to the development of macular degeneration, positioning autophagy as a cutting-edge research area in the realm of dry AMD. In this review, we present an overview of how lipofuscin, oxidative stress, and the NLRP3 inflammasome damage the RPE through their respective causal mechanisms. We summarized the connection between autophagy, oxidative stress, and NLRP3 inflammatory cytokines. Our findings suggest that targeting autophagy improves RPE function and sustains visual health, offering new perspectives for understanding the pathogenesis and clinical management of dry AMD.
Collapse
Affiliation(s)
- Zhao Zhang
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Fengming Liang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Jun Chang
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Xiaoqian Shan
- Tianjin University of Chinese Medicine, Tianjin, 300193, China; The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Zhixian Yin
- Hebei University of Technology, School of Electronics and Information Engineering, Tianjin, 300401, China
| | - Li Wang
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center of Traditional Chinese Medicine and Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Shujiao Li
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, 100040, China
| |
Collapse
|
15
|
Ebrahimi N, Manavi MS, Faghihkhorasani F, Fakhr SS, Baei FJ, Khorasani FF, Zare MM, Far NP, Rezaei-Tazangi F, Ren J, Reiter RJ, Nabavi N, Aref AR, Chen C, Ertas YN, Lu Q. Harnessing function of EMT in cancer drug resistance: a metastasis regulator determines chemotherapy response. Cancer Metastasis Rev 2024; 43:457-479. [PMID: 38227149 DOI: 10.1007/s10555-023-10162-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/08/2023] [Indexed: 01/17/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a complicated molecular process that governs cellular shape and function changes throughout tissue development and embryogenesis. In addition, EMT contributes to the development and spread of tumors. Expanding and degrading the surrounding microenvironment, cells undergoing EMT move away from the main location. On the basis of the expression of fibroblast-specific protein-1 (FSP1), fibroblast growth factor (FGF), collagen, and smooth muscle actin (-SMA), the mesenchymal phenotype exhibited in fibroblasts is crucial for promoting EMT. While EMT is not entirely reliant on its regulators like ZEB1/2, Twist, and Snail proteins, investigation of upstream signaling (like EGF, TGF-β, Wnt) is required to get a more thorough understanding of tumor EMT. Throughout numerous cancers, connections between tumor epithelial and fibroblast cells that influence tumor growth have been found. The significance of cellular crosstalk stems from the fact that these events affect therapeutic response and disease prognosis. This study examines how classical EMT signals emanating from various cancer cells interfere to tumor metastasis, treatment resistance, and tumor recurrence.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | | | | | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Inland Norway University of Applied Sciences, Hamar, Norway
| | | | | | - Mohammad Mehdi Zare
- Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX, 77030, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Amir Reza Aref
- Translational Medicine Group, Xsphera Biosciences, 6 Tide Street, Boston, MA, 02210, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Chu Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye.
- Department of Biomedical Engineering, Erciyes University, Kayseri, 38039, Türkiye.
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China.
| |
Collapse
|
16
|
Musolino M, D’Agostino M, Zicarelli M, Andreucci M, Coppolino G, Bolignano D. Spice Up Your Kidney: A Review on the Effects of Capsaicin in Renal Physiology and Disease. Int J Mol Sci 2024; 25:791. [PMID: 38255865 PMCID: PMC10815060 DOI: 10.3390/ijms25020791] [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: 12/10/2023] [Revised: 12/31/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024] Open
Abstract
Capsaicin, the organic compound which attributes the spicy flavor and taste of red peppers and chili peppers, has been extensively studied for centuries as a potential natural remedy for the treatment of several illnesses. Indeed, this compound exerts well-known systemic pleiotropic effects and may thus bring important benefits against various pathological conditions like neuropathic pain, rhinitis, itching, or chronic inflammation. Yet, little is known about the possible biological activity of capsaicin at the kidney level, as this aspect has only been addressed by sparse experimental investigations. In this paper, we aimed to review the available evidence focusing specifically on the effects of capsaicin on renal physiology, as well as its potential benefits for the treatment of various kidney disorders. Capsaicin may indeed modulate various aspects of renal function and renal nervous activity. On the other hand, the observed experimental benefits in preventing acute kidney injury, slowing down the progression of diabetic and chronic kidney disease, ameliorating hypertension, and even delaying renal cancer growth may set the stage for future human trials of capsaicin administration as an adjuvant or preventive therapy for different, difficult-to-treat renal diseases.
Collapse
Affiliation(s)
- Michela Musolino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Mario D’Agostino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
| | | | - Michele Andreucci
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Giuseppe Coppolino
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Davide Bolignano
- Nephrology and Dialysis Unit, Magna Graecia University Hospital, 88100 Catanzaro, Italy; (M.M.); (M.D.); (M.A.); (G.C.)
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
17
|
Miao K, Liu W, Xu J, Qian Z, Zhang Q. Harnessing the power of traditional Chinese medicine monomers and compound prescriptions to boost cancer immunotherapy. Front Immunol 2023; 14:1277243. [PMID: 38035069 PMCID: PMC10684919 DOI: 10.3389/fimmu.2023.1277243] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
At present, cancer is the largest culprit that endangers human health. The current treatment options for cancer mainly include surgical resection, adjuvant radiotherapy and chemotherapy, but their therapeutic effects and long-term prognosis are unsatisfactory. Immunotherapy is an emerging therapy that has completely transformed the therapeutic landscape of advanced cancers, and has tried to occupy a place in the neoadjuvant therapy of resectable tumors. However, not all patients respond to immunotherapy due to the immunological and molecular features of the tumors. Traditional Chinese Medicine (TCM) provides a new perspective for cancer treatment and is considered to have the potential as promising anti-tumor drugs considering its immunoregulatory properties. This review concludes commonly used TCM monomers and compounds from the perspective of immune regulatory pathways, aiming to clearly introduce the basic mechanisms of TCM in boosting cancer immunotherapy and mechanisms of several common TCM. In addition, we also summarized closed and ongoing trials and presented prospects for future development. Due to the significant role of immunotherapy in the treatment of non-small cell lung cancer (NSCLC), TCM combined with immunotherapy should be emphasized in NSCLC.
Collapse
Affiliation(s)
- Keyan Miao
- Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Weici Liu
- Department of Thoracic Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jingtong Xu
- The First School of Clinical Medicine, Nanjing Medical University. Nanjing, Jiangsu, China
| | - Zhengtao Qian
- Department of Clinical Laboratory, Changshu Medicine Examination Institute, Changshu, Jiangsu, China
| | - Qinglin Zhang
- Department of Gastroenterology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| |
Collapse
|
18
|
Vivanco PG, Taboada P, Coelho A. The Southern European Atlantic Diet and Its Supplements: The Chemical Bases of Its Anticancer Properties. Nutrients 2023; 15:4274. [PMID: 37836558 PMCID: PMC10574233 DOI: 10.3390/nu15194274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Scientific evidence increasingly supports the strong link between diet and health, acknowledging that a well-balanced diet plays a crucial role in preventing chronic diseases such as obesity, diabetes, cardiovascular issues, and certain types of cancer. This perspective opens the door to developing precision diets, particularly tailored for individuals at risk of developing cancer. It encompasses a vast research area and involves the study of an expanding array of compounds with multilevel "omics" compositions, including genomics, transcriptomics, proteomics, epigenomics, miRNomics, and metabolomics. We review here the components of the Southern European Atlantic Diet (SEAD) from both a chemical and pharmacological standpoint. The information sources consulted, complemented by crystallographic data from the Protein Data Bank, establish a direct link between the SEAD and its anticancer properties. The data collected strongly suggest that SEAD offers an exceptionally healthy profile, particularly due to the presence of beneficial biomolecules in its foods. The inclusion of olive oil and paprika in this diet provides numerous health benefits, and scientific evidence supports the anticancer properties of dietary supplements with biomolecules sourced from vegetables of the brassica genus. Nonetheless, further research is warranted in this field to gain deeper insights into the potential benefits of the SEAD's bioactive compounds against cancer.
Collapse
Affiliation(s)
- Pablo García Vivanco
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain
- Nutrition and Digestive Working Group, Spanish Society of Clinical, Family, and Community Pharmacy (SEFAC), 28045 Madrid, Spain
| | - Pablo Taboada
- Department of Condensed Matter Physics, Faculty of Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Alberto Coelho
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
19
|
Periferakis AT, Periferakis A, Periferakis K, Caruntu A, Badarau IA, Savulescu-Fiedler I, Scheau C, Caruntu C. Antimicrobial Properties of Capsaicin: Available Data and Future Research Perspectives. Nutrients 2023; 15:4097. [PMID: 37836381 PMCID: PMC10574431 DOI: 10.3390/nu15194097] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Capsaicin is a phytochemical derived from plants of the genus Capsicum and subject of intensive phytochemical research due to its numerous physiological and therapeutical effects, including its important antimicrobial properties. Depending on the concentration and the strain of the bacterium, capsaicin can exert either bacteriostatic or even bactericidal effects against a wide range of both Gram-positive and Gram-negative bacteria, while in certain cases it can reduce their pathogenicity by a variety of mechanisms such as mitigating the release of toxins or inhibiting biofilm formation. Likewise, capsaicin has been shown to be effective against fungal pathogens, particularly Candida spp., where it once again interferes with biofilm formation. The parasites Toxoplasma gondi and Trypanosoma cruzi have been found to be susceptible to the action of this compound too while there are also viruses whose invasiveness is significantly dampened by it. Among the most encouraging findings are the prospects for future development, especially using new formulations and drug delivery mechanisms. Finally, the influence of capsaicin in somatostatin and substance P secretion and action, offers an interesting array of possibilities given that these physiologically secreted compounds modulate inflammation and immune response to a significant extent.
Collapse
Affiliation(s)
- Aristodemos-Theodoros Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Argyrios Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs (P.O.E.P), 17236 Athens, Greece
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, ‘Prof. N.C. Paulescu’ National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| |
Collapse
|
20
|
Wang M, Kejian S, Ye L, Chen J, Ma L. Transforms of Cell Surface Glycoproteins Charge Influences Tumor Cell Metastasis via Atypically Inhibiting Epithelial-Mesenchymal Transition Including Matrix Metalloproteinases and Cell Junctions. Bioconjug Chem 2023; 34:1498-1507. [PMID: 37498932 DOI: 10.1021/acs.bioconjchem.3c00303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Cell communication and signal transduction rely heavily on the charge on the cell surface. The cell surface is negatively charged, with glycoproteins on the cell membrane providing a large percentage of the charge. Sialic acid is found on the outermost side of glycan chains and contributes to glycoprotein's negative charge. Sialic acid is highly expressed in tumor cells and plays an important role in tumor metastasis and immune escape by interacting with extracellular ligands. However, the specific effect of negative charge changes on glycoproteins is still poorly understood. In this study, we used 9-azido sialic acid (9Az-Sia) to create artificial epitopes on glycoproteins via metabolic glycan labeling, and we attached charged groups such as amino and carboxyl to 9Az-Sia via a click reaction with dibenzocyclooctyne (DBCO). The charge of glycoproteins was changed by metabolic glycan labeling and click modification. The results showed that the migration and invasion ability of the MDA-MB-231 cell labeled with 9Az-Sia was significantly reduced after the modification with amino groups rather than carboxyl groups. Epithelial-mesenchymal transition (EMT) is the biological process of metastatic tumor cells, with an increasing ability of tumor cells to migrate and invade. In particular, the expression of adhesion molecules increased in the amine-linked group, whereas the expression of matrix metalloproteinases (MMPs) increased significantly, which is not identical to EMT characteristics. In vivo experiments have demonstrated that the loss of negative charge on glycoproteins has an inhibitory effect on tumors. In conclusion, modifying the positive charge on the surface of glycoproteins can inhibit tumor cell metastasis and has great potential for tumor therapy.
Collapse
Affiliation(s)
- Mingzhe Wang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shi Kejian
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lei Ye
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Jiaqi Chen
- School of Mechanical Engineering, Chengdu University, Chengdu 610000, China
| | - Lan Ma
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen 518055, China
| |
Collapse
|
21
|
He P, Dai Q, Wu X. New insight in urological cancer therapy: From epithelial-mesenchymal transition (EMT) to application of nano-biomaterials. ENVIRONMENTAL RESEARCH 2023; 229:115672. [PMID: 36906272 DOI: 10.1016/j.envres.2023.115672] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 05/21/2023]
Abstract
A high number of cancer-related deaths (up to 90) are due to metastasis and simple definition of metastasis is new colony formation of tumor cells in a secondary site. In tumor cells, epithelial-mesenchymal transition (EMT) stimulates metastasis and invasion, and it is a common characteristic of malignant tumors. Prostate cancer, bladder cancer and renal cancer are three main types of urological tumors that their malignant and aggressive behaviors are due to abnormal proliferation and metastasis. EMT has been well-documented as a mechanism for promoting invasion of tumor cells and in the current review, a special attention is directed towards understanding role of EMT in malignancy, metastasis and therapy response of urological cancers. The invasion and metastatic characteristics of urological tumors enhance due to EMT induction and this is essential for ensuring survival and ability in developing new colonies in neighboring and distant tissues and organs. When EMT induction occurs, malignant behavior of tumor cells enhances and their tend in developing therapy resistance especially chemoresistance promotes that is one of the underlying reasons for therapy failure and patient death. The lncRNAs, microRNAs, eIF5A2, Notch-4 and hypoxia are among common modulators of EMT mechanism in urological tumors. Moreover, anti-tumor compounds such as metformin can be utilized in suppressing malignancy of urological tumors. Besides, genes and epigenetic factors modulating EMT mechanism can be therapeutically targeted for interfering malignancy of urological tumors. Nanomaterials are new emerging agents in urological cancer therapy that they can improve potential of current therapeutics by their targeted delivery to tumor site. The important hallmarks of urological cancers including growth, invasion and angiogenesis can be suppressed by cargo-loaded nanomaterials. Moreover, nanomaterials can improve chemotherapy potential in urological cancer elimination and by providing phototherapy, they mediate synergistic tumor suppression. The clinical application depends on development of biocompatible nanomaterials.
Collapse
Affiliation(s)
- Peng He
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qiang Dai
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaojun Wu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
22
|
Wang YX, Jin YY, Wang J, Zhao ZC, Xue KW, Xiong H, Che HL, Ge YJ, Wu GS. Icaritin Derivative IC2 Induces Cytoprotective Autophagy of Breast Cancer Cells via SCD1 Inhibition. Molecules 2023; 28:1109. [PMID: 36770781 PMCID: PMC9920188 DOI: 10.3390/molecules28031109] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Breast cancer is one of the most prevalent malignancies and the leading cause of cancer-associated mortality in China. Icaritin (ICT), a prenyl flavonoid derived from the Epimedium Genus, has been proven to inhibit the proliferation and stemness of breast cancer cells. Our previous study demonstrated that IC2, a derivative of ICT, could induce breast cancer cell apoptosis by Stearoyl-CoA desaturase 1 (SCD1) inhibition. The present study further investigated the mechanism of the inhibitory effects of IC2 on breast cancer cells in vitro and in vivo. Our results proved that IC2 could stimulate autophagy in breast cancer cells with the activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling and mitogen-activated protein kinase (MAPK) signaling. Combination treatment of the AMPK inhibitor decreased IC2-induced autophagy while it markedly enhanced IC2-induced apoptosis. In common with IC2-induced apoptosis, SCD1 overexpression or the addition of exogenous oleic acid (OA) could also alleviate IC2-induced autophagy. In vivo assays additionally demonstrated that IC2 treatment markedly inhibited tumor growth in a mouse breast cancer xenograft model. Overall, our study was the first to demonstrate that IC2 induced cytoprotective autophagy by SCD1 inhibition in breast cancer cells and that the autophagy inhibitor markedly enhanced the anticancer activity of IC2. Therefore, IC2 was a potential candidate compound in combination therapy for breast cancer.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Yi-Yuan Jin
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
- Taizhou Center for Disease Control and Prevention, Taizhou 318000, China
| | - Jie Wang
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Zi-Cheng Zhao
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Ke-Wen Xue
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - He Xiong
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Hui-Lian Che
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Yun-Jun Ge
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Guo-Sheng Wu
- Department of Basic Medical Science, Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| |
Collapse
|
23
|
Wang C, He Y, Zheng J, Wang X, Chen S. Dissecting order amidst chaos of programmed cell deaths: construction of a diagnostic model for KIRC using transcriptomic information in blood-derived exosomes and single-cell multi-omics data in tumor microenvironment. Front Immunol 2023; 14:1130513. [PMID: 37153569 PMCID: PMC10154557 DOI: 10.3389/fimmu.2023.1130513] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
Background Kidney renal clear cell carcinoma (KIRC) is the most frequently diagnosed subtype of renal cell carcinoma (RCC); however, the pathogenesis and diagnostic approaches for KIRC remain elusive. Using single-cell transcriptomic information of KIRC, we constructed a diagnostic model depicting the landscape of programmed cell death (PCD)-associated genes, namely cell death-related genes (CDRGs). Methods In this study, six CDRG categories, including apoptosis, necroptosis, autophagy, pyroptosis, ferroptosis, and cuproptosis, were collected. RNA sequencing (RNA-seq) data of blood-derived exosomes from the exoRBase database, RNA-seq data of tissues from The Cancer Genome Atlas (TCGA) combined with control samples from the GTEx databases, and single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) database were downloaded. Next, we intersected the differentially expressed genes (DEGs) of the KIRC cohort from exoRBase and the TCGA databases with CDRGs and DEGs obtained from single-cell datasets, further screening out the candidate biomarker genes using clinical indicators and machine learning methods and thus constructing a diagnostic model for KIRC. Finally, we investigated the underlying mechanisms of key genes and their roles in the tumor microenvironment using scRNA-seq, single-cell assays for transposase-accessible chromatin sequencing (scATAC-seq), and the spatial transcriptomics sequencing (stRNA-seq) data of KIRC provided by the GEO database. Result We obtained 1,428 samples and 216,155 single cells. After the rational screening, we constructed a 13-gene diagnostic model for KIRC, which had high diagnostic efficacy in the exoRBase KIRC cohort (training set: AUC = 1; testing set: AUC = 0.965) and TCGA KIRC cohort (training set: AUC = 1; testing set: AUC = 0.982), with an additional validation cohort from GEO databases presenting an AUC value of 0.914. The results of a subsequent analysis revealed a specific tumor epithelial cell of TRIB3high subset. Moreover, the results of a mechanical analysis showed the relatively elevated chromatin accessibility of TRIB3 in tumor epithelial cells in the scATAC data, while stRNA-seq verified that TRIB3 was predominantly expressed in cancer tissues. Conclusions The 13-gene diagnostic model yielded high accuracy in KIRC screening, and TRIB3high tumor epithelial cells could be a promising therapeutic target for KIRC.
Collapse
Affiliation(s)
- Chengbang Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Yuan He
- Department of Urology, The Second Nanning People’s Hospital, Nanning, China
- *Correspondence: Xiang Wang, ; Shaohua Chen, ; Yuan He,
| | - Jie Zheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiang Wang, ; Shaohua Chen, ; Yuan He,
| | - Shaohua Chen
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory for Genomic and Personalized Medicine, Center for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
- *Correspondence: Xiang Wang, ; Shaohua Chen, ; Yuan He,
| |
Collapse
|