1
|
Zhang M, Zhang Y, Peng J, Huang Y, Gong Z, Lu H, Han L, Wang D. Gastrodin against oxidative stress-inflammation crosstalk via inhibiting mtDNA/TLR9 and JAK2/STAT3 signaling to ameliorate ischemic stroke injury. Int Immunopharmacol 2024; 141:113012. [PMID: 39182268 DOI: 10.1016/j.intimp.2024.113012] [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: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
The pathway of Janus-activated kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3) (termed as JAK2/STAT3) plays an active role in stroke-related inflammation induced by ischemic stress. Gastrodin, the primary compound in Gastrodia elata Bl, has been identified for its notable neuroprotective effects and demonstrated to ameliorate cerebral ischemia-reperfusion but its exact mechanisms governing this defense are still unclear. This study aims to investigate whether gastrodin can regulate mitochondrial function via the JAK2/STAT3 pathway to limit cerebral ischemia-reperfusion. In vivo, gastrodin significantly reduced infarct volume, improved neurobiological function, attenuated neuronal apoptosis, oxidative stress, mitochondrial impairment, mtDNA leakage, and inflammatory responses. At the cellular level, gastrodin administration rescued OGD/R-induced cell apoptosis, oxidative stress, and mitochondrial dysfunction. Mechanistically, gastrodin notably suppressed Toll-like receptor 9 (TLR9) expression, important for the recognition of disrupted endogenous DNA to produce inflammatory reactions. Furthermore, gastrodin mitigated inflammation by inhibiting JAK2/STAT3 signaling, influencing inflammatory factors to aggravate inflammation. Notably, the effects of gastrodin were abolished by Coumermycin A1 (C-A1), a JAK2 agonist, validating the role of JAK2/STAT3 signaling. In summary, gastrodin enhances the protective effect against mitochondrial damage in ischemic stroke by inhibiting JAK2/STAT3 signaling. Gastrodin is a possible therapy for cerebral ischemia.
Collapse
Affiliation(s)
- Menglian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Yaowen Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Jinyong Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Yingying Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Zipeng Gong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
| | - Huixin Lu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China
| | - Lan Han
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China.
| | - Dandan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Institute for the Evaluation of the Efficacy and Safety of Chinese Medicines, Anhui Academy of Chinese Medicine, Hefei 230011, China.
| |
Collapse
|
2
|
Shaharudin NS, Surindar Singh GK, Kek TL, Sultan S. Targeting signaling pathways with andrographolide in cancer therapy (Review). Mol Clin Oncol 2024; 21:81. [PMID: 39301125 PMCID: PMC11411607 DOI: 10.3892/mco.2024.2779] [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: 03/19/2024] [Accepted: 07/10/2024] [Indexed: 09/22/2024] Open
Abstract
Terpenoids are a large group of naturally occurring organic compounds with a wide range of components. A phytoconstituent in this group, andrographolide, which is derived from a plant called Andrographis paniculate, offers a number of advantages, including anti-inflammatory, anticancer, anti-angiogenesis and antioxidant effects. The present review elucidates the capacity of andrographolide to inhibit signaling pathways, namely the nuclear factor-κB (NF-κB), hypoxia-inducible factor 1 (HIF-1), the Janus kinase (JAK)/signal transducer and activator of transcription (STAT), phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), Wnt/β-catenin and mitogen-activated protein kinase (MAPK) pathways, which are involved in cellular processes and responses such as the inflammatory response, apoptosis and angiogenesis. Inhibiting pathways enables andrographolide to exhibit its anticancer effects against breast, colorectal and lung cancer. The present review focuses on the anticancer effects of andrographolide, specifically in breast, colorectal and lung cancer through the NF-κB, HIF-1 and JAK/STAT signaling pathways. Therefore, the Google Scholar, PubMed and ScienceDirect databases were used to search for references to these prevalent types of cancer and the anticancer mechanisms of andrographolide associated with them. The following key words were used: Andrographolide, anticancer, JAK/STAT, HIF-1, NF-κB, PI3K/AKT/mTOR, Wnt/β-catenin and MAPK pathways, and the literature was limited to studies published between 2010 to 2023. The present review article provides details about the different involvements of signaling pathways in the anticancer mechanisms of andrographolide.
Collapse
Affiliation(s)
- Nur Shahirah Shaharudin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
| | - Gurmeet Kaur Surindar Singh
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
- Faculty of Pharmacy, Brain Degeneration and Therapeutics Research Center, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| | - Teh Lay Kek
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
| | - Sadia Sultan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300, Malaysia
- Faculty of Pharmacy, Biotransformation Research Center, Universiti Teknologi MARA, Shah Alam, Selangor 40450, Malaysia
| |
Collapse
|
3
|
Joghataie P, Ardakani MB, Sabernia N, Salary A, Khorram S, Sohbatzadeh T, Goodarzi V, Amiri BS. The Role of Circular RNA in the Pathogenesis of Chemotherapy-Induced Cardiotoxicity in Cancer Patients: Focus on the Pathogenesis and Future Perspective. Cardiovasc Toxicol 2024; 24:1151-1167. [PMID: 39158829 DOI: 10.1007/s12012-024-09914-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Cardiotoxicity is a serious challenge cancer patients face today. Various factors are involved in cardiotoxicity. Circular RNAs (circRNAs) are one of the effective factors in the occurrence and prevention of cardiotoxicity. circRNAs can lead to increased proliferation, apoptosis, and regeneration of cardiomyocytes by regulating the molecular pathways, as well as increasing or decreasing gene expression; some circRNAs have a dual role in cardiomyocyte regeneration or death. Identifying each of the pathways related to these processes can be effective on managing patients and preventing cardiotoxicity. In this study, an overview of the molecular pathways involved in cardiotoxicity by circRNAs and their effects on the downstream factors have been discussed.
Collapse
Affiliation(s)
- Pegah Joghataie
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Neda Sabernia
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Bahareh Shateri Amiri
- Assistant Professor of Internal Medicine, Department of Internal Medicine, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Jiang RY, Zhu JY, Zhang HP, Yu Y, Dong ZX, Zhou HH, Wang X. STAT3: Key targets of growth-promoting receptor positive breast cancer. Cancer Cell Int 2024; 24:356. [PMID: 39468521 PMCID: PMC11520424 DOI: 10.1186/s12935-024-03541-9] [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: 07/26/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Breast cancer has become the malignant tumor with the first incidence and the second mortality among female cancers. Most female breast cancers belong to luminal-type breast cancer and HER2-positive breast cancer. These breast cancer cells all have different driving genes, which constantly promote the proliferation and metastasis of breast cancer cells. Signal transducer and activator of transcription 3 (STAT3) is an important breast cancer-related gene, which can promote the progress of breast cancer. It has been proved in clinical and basic research that over-expressed and constitutively activated STAT3 is involved in the progress, proliferation, metastasis and chemotherapy resistance of breast cancer. STAT3 is an important key target in luminal-type breast cancer and HER2-positive cancer, which has an important impact on the curative effect of related treatments. In breast cancer, the activation of STAT3 will change the spatial position of STAT3 protein and cause different phenotypic changes of breast cancer cells. In the current basic research and clinical research, small molecule inhibitors activated by targeting STAT3 can effectively treat breast cancer, and enhance the efficacy level of related treatment methods for luminal-type and HER2-positive breast cancers.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Jia-Yu Zhu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Huan-Ping Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
- Department of Graduate Student, Wenzhou Medical University, No.270, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
| | - Yuan Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Zhi-Xin Dong
- Department of Oncology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, No.89-9, Dongge Road, Qingxiu District, Nanning, 530000, Guangxi, China
| | - Huan-Huan Zhou
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, NO.548, Binwen Road, Binjiang District, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Xiaojia Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
5
|
Chen Y, Zheng Z, Wang L, Chen R, He M, Zhao X, Jin L, Yao J. Deciphering STAT3's negative regulation of LHPP in ESCC progression through single-cell transcriptomics analysis. Mol Med 2024; 30:192. [PMID: 39468431 PMCID: PMC11520558 DOI: 10.1186/s10020-024-00962-0] [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: 06/15/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Esophageal Squamous Cell Carcinoma (ESCC) remains a predominant health concern in the world, characterized by high prevalence and mortality rates. Advances in single-cell transcriptomics have revolutionized cancer research by enabling a precise dissection of cellular and molecular diversity within tumors. OBJECTIVE This study aims to elucidate the cellular dynamics and molecular mechanisms in ESCC, focusing on the transcriptional influence of STAT3 (Signal Transducer and Activator of Transcription 3) and its interaction with LHPP, thereby uncovering potential therapeutic targets. METHODS Single-cell RNA sequencing was employed to analyze 44,206 cells from tumor and adjacent normal tissues of ESCC patients, identifying distinct cell types and their transcriptional shifts. We conducted differential gene expression analysis to assess changes within the tumor microenvironment (TME). Validation of key regulatory interactions was performed using qPCR in a cohort of 21 ESCC patients and further substantiated through experimental assays in ESCC cell lines. RESULTS The study revealed critical alterations in cell composition and gene expression across identified cell populations, with a notable shift towards pro-tumorigenic states. A significant regulatory influence of STAT3 on LHPP was discovered, establishing a novel aspect of ESCC pathogenesis. Elevated levels of STAT3 and suppressed LHPP expression were validated in clinical samples. Functional assays confirmed that STAT3 directly represses LHPP at the promoter level, and disruption of this interaction by promoter mutations diminished STAT3's repressive effect. CONCLUSION This investigation underscores the central role of STAT3 as a regulator in ESCC, directly impacting LHPP expression and suggesting a regulatory loop crucial for tumor behavior. The insights gained from our comprehensive cellular and molecular analysis offer a deeper understanding of the dynamics within the ESCC microenvironment. These findings pave the way for targeted therapeutic interventions focusing on the STAT3-LHPP axis, providing a strategic approach to improve ESCC management and prognosis.
Collapse
Affiliation(s)
- Yurao Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Zemao Zheng
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Luoshai Wang
- Department of Thoracic Surgery, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
| | - Ronghuai Chen
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Ming He
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Xiang Zhao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China
| | - Liyan Jin
- Department of Oncology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213000, Jiangsu, China.
- Department of Oncology, The Wujin Clinical college of Xuzhou Medical University, Changzhou, 213000, Jiangsu, China.
| | - Juan Yao
- Department of Radiation Oncology, Huaian Hospital of Huaian City, Huaian, 223299, Jiangsu, China.
- Department of Radiation Oncology, Huaian Cancer Hospital, Huaian, 223299, Jiangsu, China.
| |
Collapse
|
6
|
Gnanagurusamy J, Krishnamoorthy S, Muthusami S. Transforming growth factor-β micro-environment mediated immune cell functions in cervical cancer. Int Immunopharmacol 2024; 140:112837. [PMID: 39111147 DOI: 10.1016/j.intimp.2024.112837] [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: 02/19/2024] [Revised: 07/02/2024] [Accepted: 07/28/2024] [Indexed: 09/01/2024]
Abstract
Propensity to develop cervical cancer (CC) in human papilloma virus (HPV) infected individual could potentially involve the impaired immune functioning. Several stages of HPV surveillance by immune cells in tumor micro-environment (TME) is regulated mainly by transforming growth factor-beta (TGF-β) and is crucial for the establishment of CC. The role of TGF-β in the initiation and progression of CC is very complex and involve different suppressor of mothers against decapentaplegic homolog (SMAD) dependent and SMAD independent signaling mechanism(s). This review summarizes the handling of HPV by immune cells such as T lymphocytes, B lymphocytes, natural killer cells (NK), dendritic cells (DC), monocytes, macrophages, myeloid derived suppressor cells (MDSC) and their regulation by TGF-β. The hijack mechanisms adapted by HPV to evade this surveillance process is discussed. Biomarkers indicating the stages of CC and immune checkpoints that can be targeted for improved outcome are included for immune-based theragnostics. This review also addresses the direct actions of TGF-β on CC cells and tumor/immune cell interactions. Therapies focused on targeting TGF-β using small molecule inhibitors, monoclonal antibodies and TGF-β chimeric antigen receptor (CAR)T cells are collated to understand the current strategies related to TGF-β in the management of CC.
Collapse
Affiliation(s)
- Jayapradha Gnanagurusamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India.
| |
Collapse
|
7
|
XU SULING, LI XIAO, MA WENXUE. Redefining the tumor microenvironment with emerging therapeutic strategies. Oncol Res 2024; 32:1701-1708. [PMID: 39449800 PMCID: PMC11497178 DOI: 10.32604/or.2024.055161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/23/2024] [Indexed: 10/26/2024] Open
Abstract
The environment surrounding a tumor, known as the tumor microenvironment (TME), plays a role in how cancer progresses and responds to treatment. It poses both challenges and opportunities for improving cancer therapy. Recent progress in understanding the TME complexity and diversity has led to approaches for treating cancer. This perspective discusses the strategies for targeting the TME, such as adjusting networks using extracellular vesicles to deliver drugs and enhancing immune checkpoint inhibitors (ICIS) through combined treatments. Furthermore, it highlights adoptive cell transfer (ACT) therapies as an option for tumors. By studying how components of the TME interact and utilizing technologies like single-cell RNA sequencing and spatial transcriptomics, we can develop more precise and efficient treatments for cancer. This article emphasizes the need to reshape the TME to boost antitumor immunity and overcome resistance to therapy, providing guidance for research and clinical practices in precision oncology.
Collapse
Affiliation(s)
- SULING XU
- Department of Dermatology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - XIAO LI
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - WENXUE MA
- Department of Medicine, Sanford Stem Cell Institute and Moores Cancer Center, University of California San Diego, La Jolla, CA92093, USA
| |
Collapse
|
8
|
Xie C, Wang T, Liu A, Huang B, Zeng W, Li Z, Peng S, Wu S. Sirt4 Overexpression Modulates the JAK2/STAT3 and PI3K/AKT/mTOR Axes to Alleviate Sepsis-Induced Acute Lung Injury. Cell Biochem Biophys 2024:10.1007/s12013-024-01588-z. [PMID: 39400781 DOI: 10.1007/s12013-024-01588-z] [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] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is a severe organ dysfunction characterized by lung inflammation and apoptosis. The mechanisms underlying sepsis-induced ALI remain poorly understood. Here, we determined the effects of sirtuin 4 (SIRT4) on sepsis-induced ALI. METHODS Lipopolysaccharide (LPS)-induced injury cell and cecal ligation and puncture (CLP) animal models were established. Overexpression vectors and lentiviral transfections were used to upregulate SIRT4 expression. Lung cell apoptosis, inflammation, and the levels of associated factors were evaluated. Changes in the PI3K/AKT/mTOR and JAK2/STAT3 pathways were measured, and their potential involvement was examined using LY294002 (PI3K inhibitor), 740 Y-P (PI3K agonist), AG490 (JAK2 inhibitor), and coumermycin A1 (JAK2 agonist). RESULTS Lower SIRT4 expression was observed in LPS-exposed A549 cells and CLP rats. In LPS-induced A549 cells, Sirt4 overexpression enhanced cell viability, resisted apoptosis, restored the expression of apoptosis-associated proteins (HMB1, cleaved CASP3, BAX, and BCL), and reduced the secretion of pro-inflammatory cytokines (IL-6, IL-1β, and TNF-α). In CLP rats, Sirt4 overexpression prolonged survival time, alleviated lung histopathological damage, reduced pulmonary edema, mitigated lung infection, decreased lung apoptosis, and lowered serum levels of inflammatory cytokines. Furthermore, Sirt4 overexpression blocked JAK2/STAT3/AKT/mTOR phosphorylation. 740 Y-P and coumermycin A1 reversed the protective effects of Sirt4 overexpression in LPS-treated A549 cells, resulting in decreased cell viability and increased apoptosis. LY294002 and AG490 enhanced the protective effects of Sirt4 overexpression in LPS-treated A549 cells. CONCLUSION SIRT4 alleviates sepsis-induced ALI by inhibiting JAK2/STAT3/PI3K/AKT/mTOR signaling. Upregulating SIRT4 expression may serve as an innovative therapeutic approach for lung injury management in sepsis.
Collapse
Affiliation(s)
- Cancan Xie
- Department of Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Ting Wang
- Department of Rehabilitation Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Anmin Liu
- Department of Emergency, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Bing Huang
- Department of Respiratory Medicine, Zhuzhou Central Hospital, Central South University, Zhuzhou, Hunan, China
| | - Weizhong Zeng
- Department of Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Zhengrong Li
- Department of Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Suna Peng
- Department of Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Shuanghua Wu
- Department of Critical Care Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
9
|
Sun Y, Wang Q, Zhang Z, Wang Q, Cen J, Zhu M, Pan J, Liu D, Shen H, Cai Y, Chen S. Distinct clinical profiles and patient outcomes in aCML and CNL. Ann Hematol 2024:10.1007/s00277-024-06032-z. [PMID: 39375227 DOI: 10.1007/s00277-024-06032-z] [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: 09/01/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
The classification of atypical chronic myeloid leukemia (aCML) and chronic neutrophilic leukemia (CNL) as a single disease entity remains a topic of debate. To elucidate the characteristics of both entities, this retrospective cohort study was conducted, encompassing 36 cases of aCML and 18 cases of CNL. We discovered that aCML and CNL presented distinct blood counts, genetics, molecular profiles and outcomes. Specifically, hemoglobin levels (P < 0.001) and platelet counts (P < 0.001) were significantly lower in aCML cases than in CNL cases, with no significant difference in mean white blood cells (P = 0.637). The proportion of abnormal karyotypes was higher in aCML cases compared with CNL cases (P = 0.010). Notably, we found that aCML and CNL showed distinct gene expression profiles by transcriptome sequencing technology. The median follow-up duration for the entire cohort was 8 months (rang 0.4 to 36.6 months), and the median overall survival (OS) was significantly shorter in aCML cases (7.3 months, 95%CI 5.4 to 20.5 months) than in CNL cases (median OS not reached). The one-year OS rate for aCML patients was 31.0% (9/29), compared to 92.9% (13/14) for CNL patients. In conclusion, our study supports the notion that aCML and CNL are indeed distinct disease entities characterized by unique hematological features and clinical outcomes.
Collapse
Affiliation(s)
- Yingxin Sun
- Affiliated Hospital of Nantong University, Nantong, China
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Qinrong Wang
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Zhiyu Zhang
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Qian Wang
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Jiannong Cen
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Mingqing Zhu
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Jinlan Pan
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Dandan Liu
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Hongjie Shen
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Yifeng Cai
- Affiliated Hospital of Nantong University, Nantong, China.
| | - Suning Chen
- The First Affiliated Hospital of Soochow University, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China.
| |
Collapse
|
10
|
Liu YY, Li N, Chen XY, Wang H, Zhu SW, Yang L, Quan FY, Ma JC, Dai JW, Jiang YL, Xiang ZF, Cheng Q, Zhang WH, Chen KH, Hou W, Xiong HR. MicroRNA let-7a regulation of Hantaan virus replication by Targeting FAS Signaling Pathways. Virology 2024; 600:110254. [PMID: 39383773 DOI: 10.1016/j.virol.2024.110254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
Hantaan virus (HTNV) infection in humans can cause hemorrhagic fever and renal syndrome (HFRS). Understanding host responses to HTNV infection is crucial for developing effective disease intervention strategies. Previous RNA-sequencing studies have investigated the role of microRNAs (miRNAs) in the post-transcriptional regulation of host genes in response to HTNV infection. In this study, we demonstrated that HTNV infection induces let-7a expression in human umbilical vein endothelial cells (HUVEC) and that HTNV G protein upregulates the expression of let-7a. miRNA let-7a mimics and inhibitors validated the predicted targets, including cell apoptosis genes (FAS, caspase-8, and caspase-3) and inflammatory factors (IL-6 and its related factors). Modulation of miRNA let-7a levels by miRNA mimics and inhibitors affected HTNV replication, indicating that HTNV modulates host miRNA expression to affect the outcome of the antiviral host response.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ning Li
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China
| | - Xing-Yuan Chen
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Hui Wang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; School of Ecology and Environment, Tibet University, Lhasa, 850000, Tibet Autonomous Region, China
| | - Shao-Wei Zhu
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Lan Yang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Fang-Yi Quan
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Jian-Chun Ma
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Jian-Wei Dai
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ya-le Jiang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China
| | - Zhou-Fu Xiang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China
| | - Qi Cheng
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Wei-Hao Zhang
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China
| | - Ke-Han Chen
- School of Public Health, Wuhan University, Wuhan, 430071, Hubei Province, China
| | - Wei Hou
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China; School of Ecology and Environment, Tibet University, Lhasa, 850000, Tibet Autonomous Region, China; Shenzhen Research Institute, Wuhan University, Shenzhen, 518057, Guangdong Province, China; School of Public Health, Wuhan University, Wuhan, 430071, Hubei Province, China.
| | - Hai-Rong Xiong
- State Key Laboratory of Virology/ Institute of Medical Virology, School of Basic Medical Sciences, Wuhan University, 185 Donghu Road, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
11
|
Ni R, Hu Z, Tao R. Advances of immune-checkpoint inhibition of CTLA-4 in pancreatic cancer. Biomed Pharmacother 2024; 179:117430. [PMID: 39260322 DOI: 10.1016/j.biopha.2024.117430] [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: 06/13/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024] Open
Abstract
Targeting checkpoints for immune cell activation has been acknowledged known as one of the most effective way to activate anti-tumor immune responses. Among them, drugs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are approved for clinical treatment though several more are in advanced stages of development, which demonstrated durable response rates and manageable safety profile. However, its therapy efficacy is unsatisfactory in pancreatic cancer (PC), which can be limited by the overall condition of patients, the pathological type of PC, the expression level of tumor related genes, etc. To improve clinical efficiency, various researches have been conducted, and the efficacy of combination therapy showed significantly improvement compared to monotherapy. This review analyzed current strategies based on anti-CTLA-4 combination immunotherapy, providing totally new idea for future research.
Collapse
Affiliation(s)
- Ran Ni
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhiming Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Hepatobiliary & Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China.
| | - Ran Tao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
12
|
Metwally YF, Elsaid AM, Elsadda RR, Refaat S, Zahran RF. Impact of IL-6 and IL-1β Gene Variants on Non-small-cell Lung Cancer Risk in Egyptian Patients. Biochem Genet 2024; 62:3367-3388. [PMID: 38103126 PMCID: PMC11427554 DOI: 10.1007/s10528-023-10596-2] [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: 07/26/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Lung cancer is a serious health and life issue, with the fastest-growing incidence and fatality rates worldwide. It is now clear that inflammation is a key factor involved in all aspects of carcinogenesis, notably lung cancer development. Genetic changes, including polymorphisms in inflammatory genes, are supposed to be a significant cause of increased lung cancer risk. The main idea of this research was to disclose the linkage between both IL-6 rs1800795 and IL-1β rs16944 variants and susceptibility to non-small-cell lung cancer (NSCLC) in Egyptians. This case-control design was composed of 127 cases and 138 controls, which were genotyped using the ARMS-PCR technique. To examine the NSCLC susceptibility under various genetic models, the odds ratio (OR) and 95% confidence intervals (CIs) were determined by logistic regression. Rs1800795 of the IL-6 gene was linked to higher odds of NSCLC under the allele model (adjusted, OR 2.28; 95% CI 1.2-4.33; p = 0.011). In the genetic models, IL-6 rs1800795 elevated the odds of NSCLC, while IL-1β rs16944 decreased the odds of NSCLC. Stratification analysis showed that IL-6 rs1800795 greatly increased the NSCLC risk in females and adenocarcinoma subtypes, whereas IL-1β rs16944 largely decreased the NSCLC risk for males, patients aged < 55, and nonsmokers. Regarding clinical data, the IL-6 variant was remarkably correlated with tumor size. This work primarily established that IL-6 and IL-1β variants have a great impact on NSCLC development in the Egyptian population; thus, it may be a supportive guide for earlier NSCLC prevention.
Collapse
Affiliation(s)
- Yomna F Metwally
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt.
| | - Afaf M Elsaid
- Genetics Unit, Children Hospital, Mansoura University, Mansoura, Egypt
| | - Rana R Elsadda
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Sherif Refaat
- Oncology Department, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Rasha F Zahran
- Biochemistry Department, Faculty of Science, Damietta University, Damietta, Egypt
| |
Collapse
|
13
|
Ahmad MS, Braoudaki M, Siddiqui SS. Differential expression of ST6GALNAC1 and ST6GALNAC2 and their clinical relevance to colorectal cancer progression. PLoS One 2024; 19:e0311212. [PMID: 39348343 PMCID: PMC11441655 DOI: 10.1371/journal.pone.0311212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024] Open
Abstract
Colorectal cancer (CRC) has become a significant global health concern and ranks among the leading causes of morbidity and mortality worldwide. Due to its malignant nature, current immunotherapeutic treatments are used to tackle this issue. However, not all patients respond positively to treatment, thereby limiting clinical effectiveness and requiring the identification of novel therapeutic targets to optimise current strategies. The putative ligand of Siglec-15, Sialyl-Tn (STn), is associated with tumour progression and is synthesised by the sialyltransferases ST6GALNAC1 and ST6GALNAC2. However, the deregulation of both sialyltransferases within the literature remain limited, and the involvement of microRNAs (miRNAs) in STn production require further elucidation. Here, we identified miRNAs involved in the regulation of ST6GALNAC1 via a computational approach and further analysis of miRNA binding sites were determined. In silico tools predicted miR-21, miR-30e and miR-26b to regulate the ST6GALNAC1 gene, all of which had shown significant upregulated expression in the tumour cohort. Moreover, each miRNA displayed a high binding affinity towards the seed region of ST6GALNAC1. Additionally, enrichment analysis outlined pathways associated with several cancer hallmarks, including epithelial to mesenchymal transition (EMT) and MYC targets associated with tumour progression. Furthermore, our in silico findings demonstrated that the ST6GALNAC1 expression profile was significantly downregulated in CRC tumours, and its low expression correlated with poor survival outcomes when compared with patient survival data. In comparison to its counterpart, there were no significant differences in the expression of ST6GALNAC2 between normal and malignant tissues, which was further evidenced in our immunohistochemistry analysis. Immunohistochemistry staining highlighted significantly higher expression was more prevalent in normal human tissues with regard to ST6GALNAC1. In conclusion, the integrated in silico analysis highlighted that STn production is not reliant on deregulated sialyltransferase expression in CRC, and ST6GALNAC1 expression is regulated by several oncomirs. We proposed the involvement of other sialyltransferases in the production of the STn antigen and CRC progression via the Siglec-15/Sia axis.
Collapse
Affiliation(s)
- Mohammed Saqif Ahmad
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Maria Braoudaki
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Shoib Sarwar Siddiqui
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
14
|
Brust LA, Linxweiler M, Schnatmann J, Kühn JP, Knebel M, Braun FL, Wemmert S, Menger MD, Schick B, Holick MF, Kuo F, Morris LGT, Körner S. Effects of Vitamin D on tumor cell proliferation and migration, tumor initiation and anti-tumor immune response in head and neck squamous cell carcinomas. Biomed Pharmacother 2024; 180:117497. [PMID: 39341078 DOI: 10.1016/j.biopha.2024.117497] [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: 07/06/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinomas (HNSCCs) are among the six most common cancers, with a constantly poor prognosis. Vitamin D has been found to have antineoplastic and immunomodulatory properties in various cancers. This study investigated the impact of Vitamin D on the initiation and progression as well as antitumor immune response in HNSCCs, both in vitro and in vivo. METHODS An immunocompetent, orthotopic oral carcinogenesis mouse model was used to examine the influence of Vitamin D3 substitution on HNSCC initiation and progression in vivo. Tumor immune infiltration was analyzed by immunohistochemistry targeting CD3, CD8, NKR-P1C, FOXP3, and CD163. Two HPV- and two HPV+ HNSCC cell lines were treated with 1,25-dihydroxyvitamin D3 to analyze effects on tumor cell proliferation, migration and transcriptomic changes using RNA-sequencing, differential gene expression and gene set enrichment analysis. RESULTS Vitamin D3 treatment led to a significant suppression of HNSCC initiation and progression, while also stimulating tumor immune infiltration with CD3+, CD8+ and NKR-P1C+ cells and lowering levels of M2 macrophages and Treg cells in vivo. In vitro experiments showed an inhibition of HNSCC cell proliferation and migration in HPV+ and HPV- cell lines. RNA-sequencing showed significant regulations in IL6 JAK STAT3, hypoxia signaling and immunomodulatory pathways upon Vitamin D3 treatment. CONCLUSION The findings of our study highlight the promising potential of Vitamin D in the therapeutic repertoire for HNSCC patients given its immune modulating, anti-proliferative and anti-migratory properties. Clinical transferability of those in vitro and in vivo effects should be further validated in clinical trials.
Collapse
Affiliation(s)
- Lukas A Brust
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Jana Schnatmann
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Jan-Philipp Kühn
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Moritz Knebel
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Felix L Braun
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Silke Wemmert
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Michael D Menger
- Institute of Clinical and Experimental Surgery; Saarland University, Homburg, Germany
| | - Bernhard Schick
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany
| | - Michael F Holick
- Department of Physiology and Biophysics; Boston University School of Medicine, Boston, MA, USA
| | - Fengshen Kuo
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G T Morris
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Head and Neck Service, Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sandrina Körner
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg, Germany; Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
15
|
Ke F, Zhang R, Chen R, Guo X, Song C, Gao X, Zeng F, Liu Q. The role of Rhizoma Paridis saponins on anti-cancer: The potential mechanism and molecular targets. Heliyon 2024; 10:e37323. [PMID: 39296108 PMCID: PMC11407946 DOI: 10.1016/j.heliyon.2024.e37323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer is a disease characterized by uncontrolled cell proliferation, leading to excessive growth and invasion that can spread to other parts of the body. Traditional Chinese medicine has made new advancements in the treatment of cancer, providing new perspectives and directions for cancer treatment. Rhizoma Paridis is a widely used Chinese herbal medicine with documented anti-cancer effects dating back to ancient times. Modern research has shown that Rhizoma Paridis saponins (RPS) have various pharmacological activities. RPS can inhibit cancer in multiple ways, such as suppressing tumor growth, inducing cell cycle arrest, promoting cell apoptosis, enhancing cell autophagy, inducing ferroptosis, reducing inflammation, inhibiting angiogenesis, as well as inhibiting metastasis and invasion, and these findings demonstrate the potent anti-cancer activity of RPS. Polyphyllin I, polyphyllin II, polyphyllin VI, and polyphyllin VII have been widely reported as the main active ingredients with anti-cancer properties. Polyphyllin D, polyphyllin E, and polyphyllin G have also been confirmed to possess strong anti-cancer activity in recent years. Therefore, this review dives deep into the molecular mechanisms underlying the anti-cancer effects of RPS to serve as a valuable reference for future scientific research and their potential applications in cancer treatment.
Collapse
Affiliation(s)
- Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Can Song
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiaowei Gao
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| |
Collapse
|
16
|
Chen D, Tong W, Ang B, Bai Y, Dong W, Deng X, Wang C, Zhang Y. Revealing the crosstalk between LOX + fibroblast and M2 macrophage in gastric cancer by single-cell sequencing. BMC Cancer 2024; 24:1117. [PMID: 39251966 PMCID: PMC11382413 DOI: 10.1186/s12885-024-12861-y] [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: 06/26/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND/AIMS Gastric cancer (GC) ranks among the prevalent types of cancer, and its progression is influenced by the tumor microenvironment (TME). A comprehensive comprehension of the TME associated with GC has the potential to unveil therapeutic targets of significance. METHODS The complexity and heterogeneity of TME interactions were revealed through our investigation using an integrated analysis of single-cell and bulk-tissue sequencing data. RESULTS We constructed a single-cell transcriptomic atlas of 150,913 cells isolated from GC patients. Our analysis revealed the intricate nature and heterogeneity of the GC TME and the metabolic properties of major cell types. Furthermore, two cell subtypes, LOX+ Fibroblasts and M2 Macrophages, were enriched in tumor tissue and related to the outcome of GC patients. In addition, LOX+ Fibroblasts were significantly associated with M2 macrophages. immunofluorescence double labeling indicated LOX+ Fibroblasts and M2 Macrophages were tightly localized in GC tissue. The two cell subpopulations strongly interacted in a hypoxic microenvironment, yielding an immunosuppressive phenotype. Our findings further suggest that LOX+ Fibroblasts may act as a trigger for inducing the differentiation of monocytes into M2 Macrophages via the IL6-IL6R signaling pathway. CONCLUSIONS Our study revealed the intricate and interdependent communication network between the fibroblast and macrophage subpopulations, which could offer valuable insights for targeted manipulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Dapeng Chen
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Wen Tong
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Bing Ang
- Oncology Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Yi Bai
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Wenhui Dong
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xiyue Deng
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300192, China
| | - Chunjiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yamin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China.
| |
Collapse
|
17
|
Liu R, Yu Y, Wang Q, Zhao Q, Yao Y, Sun M, Zhuang J, Sun C, Qi Y. Interactions between hedgehog signaling pathway and the complex tumor microenvironment in breast cancer: current knowledge and therapeutic promises. Cell Commun Signal 2024; 22:432. [PMID: 39252010 PMCID: PMC11382420 DOI: 10.1186/s12964-024-01812-6] [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: 05/24/2024] [Accepted: 08/31/2024] [Indexed: 09/11/2024] Open
Abstract
Breast cancer ranks as one of the most common malignancies among women, with its prognosis and therapeutic efficacy heavily influenced by factors associated with the tumor cell biology, particularly the tumor microenvironment (TME). The diverse elements of the TME are engaged in dynamic bidirectional signaling interactions with various pathways, which together dictate the growth, invasiveness, and metastatic potential of breast cancer. The Hedgehog (Hh) signaling pathway, first identified in Drosophila, has been established as playing a critical role in human development and disease. Notably, the dysregulation of the Hh pathway is recognized as a major driver in the initiation, progression, and metastasis of breast cancer. Consequently, elucidating the mechanisms by which the Hh pathway interacts with the distinct components of the breast cancer TME is essential for comprehensively evaluating the link between Hh pathway activation and breast cancer risk. This understanding is also imperative for devising novel targeted therapeutic strategies and preventive measures against breast cancer. In this review, we delineate the current understanding of the impact of Hh pathway perturbations on the breast cancer TME, including the intricate and complex network of intersecting signaling cascades. Additionally, we focus on the therapeutic promise and clinical challenges of Hh pathway inhibitors that target the TME, providing insights into their potential clinical utility and the obstacles that must be overcome to harness their full therapeutic potential.
Collapse
Affiliation(s)
- Ruijuan Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Yang Yu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Qingyang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qianxiang Zhao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yan Yao
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China
| | - Mengxuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
| | - Yuanfu Qi
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
18
|
Yang Y, Chen Q, Zhong W. The role of cytokines in the pathogenesis of SAPHO syndrome. Front Immunol 2024; 15:1427784. [PMID: 39286247 PMCID: PMC11402674 DOI: 10.3389/fimmu.2024.1427784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/09/2024] [Indexed: 09/19/2024] Open
Abstract
SAPHO syndrome is a complex inflammatory disorder affecting the skin and bones, characterized by osteomyelitis, acne, and pustulosis. Cytokines play a pivotal role in the pathogenesis of SAPHO syndrome, especially in inflammatory responses and immune regulation. This article reviews the cytokines involved in the pathogenesis of SAPHO syndrome, such as tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), IL-6, IL-10, and transforming growth factor-β (TGF-β), and discusses their potential as intervention points for treatment. These findings elucidate the intricate immune regulatory network of SAPHO syndrome and provide a theoretical foundation for the development of new targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yi Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Qianzhu Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| | - Weiyang Zhong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol 2024; 15:1404687. [PMID: 39286635 PMCID: PMC11402718 DOI: 10.3389/fphar.2024.1404687] [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: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Tumor-associated macrophages (TAMs), fundamental constituents of the tumor microenvironment (TME), significantly influence cancer development, primarily by promoting epithelial-mesenchymal transition (EMT). EMT endows cancer cells with increased motility, invasiveness, and resistance to therapies, marking a pivotal juncture in cancer progression. The review begins with a detailed exposition on the origins of TAMs and their functional heterogeneity, providing a foundational understanding of TAM characteristics. Next, it delves into the specific molecular mechanisms through which TAMs induce EMT, including cytokines, chemokines and stromal cross-talking. Following this, the review explores TAM-induced EMT features in select cancer types with notable EMT characteristics, highlighting recent insights and the impact of TAMs on cancer progression. Finally, the review concludes with a discussion of potential therapeutic targets and strategies aimed at mitigating TAM infiltration and disrupting the EMT signaling network, thereby underscoring the potential of emerging treatments to combat TAM-mediated EMT in cancer. This comprehensive analysis reaffirms the necessity for continued exploration into TAMs' regulatory roles within cancer biology to refine therapeutic approaches and improve patient outcomes.
Collapse
Affiliation(s)
- Yijia Zhang
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiaofei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| | - Xue Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ye Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Rui Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Hai-Jun Li
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Guang Chen
- Department of Pharmacy, Taizhou Second People's Hospital (Mental Health Center affiliated to Taizhou University School of Medicine), Taizhou University, Taizhou, Zhejiang, China
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang, China
| |
Collapse
|
20
|
Wang Z, Liao X, He H, Guo X, Chen J. Targeting the STAT3 pathway with STAT3 degraders. Trends Pharmacol Sci 2024; 45:811-823. [PMID: 39117533 DOI: 10.1016/j.tips.2024.07.003] [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: 06/27/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Signal transducer and activator of transcription 3 (STAT3) has been widely considered as a therapeutic target for various diseases, especially tumors. Thus far, several STAT3 inhibitors have been advanced to clinical trials; however, the development of STAT3 inhibitors is hindered by numerous dilemmas. Fortunately, STAT3 degraders represent an alternative and promising strategy to block STAT3, attracting extensive research interest. Here, we analyze the recent advancements of STAT3 degraders, including proteolysis targeting chimeras (PROTACs) and small-molecule natural products, focusing on their structures, mechanisms, and biological activities. We discuss the potential opportunities and challenges for developing STAT3 degraders. It is hoped that this Review will provide insights into the discovery of potent STAT3-targeting drugs.
Collapse
Affiliation(s)
- Zhijie Wang
- Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China; Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaotong Liao
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haiqi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xia Guo
- Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
21
|
Jiang W, Yu W, Tan Y. Activation of GPR55 alleviates neuropathic pain and chronic inflammation. Biotechnol Appl Biochem 2024. [PMID: 39219239 DOI: 10.1002/bab.2656] [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: 05/23/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
Neuropathic pain (NP) significantly impacts the quality of life due to its prolonged duration and lack of effective treatment. Recent findings suggest that targeting neuroinflammation is a promising approach for treating NP. G protein-coupled receptor 55 (GPR55), a member of the GPCR family, plays an important role in neuroinflammatory regulation. CID16020046, a GPR55 agonist, possesses promising anti-neuroinflammatory effects. Herein, the therapeutic effect of CID16020046 on NP was investigated in an NP rat model. The NP model was established using the unilateral sciatic nerve chronic constriction injury (CCI) assay. Both sham and CCI rats were intraperitoneally administered with 20 mg/kg CID16020046. NP was assessed using paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). First, we showed that GPR55 was downregulated in the spinal dorsal horn of CCI rats. After CCI rats were treated with CID16020046, the values of PWT and PWL were increased, indicating their effect on pain relief. The treated rats had attenuated release of inflammatory cytokines in the spinal cord, decreased spinal malondialdehyde (MDA) levels, and increased spinal glutathione peroxidase (GSH-PX) activity. Additionally, the increased levels of phosphorylated nuclear factor (NF)-κB p65 in CCI rats were significantly alleviated by CID16020046 treatment. Mechanistically, we showed that CID16020046 significantly suppressed the activation of the Janus kinase (JAK2)/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway in the spinal cord of CCI-treated rats. However, Colivelin TFA (a STAT3 agonist) abolished the effect of CID16020046 on JAK2/STAT3 activation. In conclusion, our data demonstrate that the activation of GPR55 by CID16020046 alleviates NP and neuroinflammation in CCI rats by mediating the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Weiqun Jiang
- Department of Anesthesiology, Nanchang First Hospital, Nanchang, Jiangxi, China
| | - Wenbin Yu
- Department of Anesthesiology, Nanchang First Hospital, Nanchang, Jiangxi, China
| | - Yu Tan
- Department of Anesthesiology, Nanchang First Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
22
|
Matuszewska J, Krawiec A, Radziemski A, Uruski P, Tykarski A, Mikuła-Pietrasik J, Książek K. Alterations of receptors and insulin-like growth factor binding proteins in senescent cells. Eur J Cell Biol 2024; 103:151438. [PMID: 38945074 DOI: 10.1016/j.ejcb.2024.151438] [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: 02/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
The knowledge about cellular senescence expands dynamically, providing more and more conclusive evidence of its triggers, mechanisms, and consequences. Senescence-associated secretory phenotype (SASP), one of the most important functional traits of senescent cells, is responsible for a large extent of their context-dependent activity. Both SASP's components and signaling pathways are well-defined. A literature review shows, however, that a relatively underinvestigated aspect of senescent cell autocrine and paracrine activity is the change in the production of proteins responsible for the reception and transmission of SASP signals, i.e., receptors and binding proteins. For this reason, we present in this article the current state of knowledge regarding senescence-associated changes in cellular receptors and insulin-like growth factor binding proteins. We also discuss the role of these alterations in senescence induction and maintenance, pro-cancerogenic effects of senescent cells, and aging-related structural and functional malfunctions.
Collapse
Affiliation(s)
- Julia Matuszewska
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Artur Radziemski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Paweł Uruski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Andrzej Tykarski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland.
| |
Collapse
|
23
|
Lopresti L, Tatangelo V, Baldari CT, Patrussi L. Rewiring the T cell-suppressive cytokine landscape of the tumor microenvironment: a new frontier for precision anti-cancer therapy. Front Immunol 2024; 15:1418527. [PMID: 39281678 PMCID: PMC11392891 DOI: 10.3389/fimmu.2024.1418527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
T lymphocytes that infiltrate the tumor microenvironment (TME) often fail to function as effective anti-cancer agents. Within the TME, cell-to-cell inhibitory interactions play significant roles in dampening their anti-tumor activities. Recent studies have revealed that soluble factors released in the TME by immune and non-immune cells, as well as by tumor cells themselves, contribute to the exacerbation of T cell exhaustion. Our understanding of the cytokine landscape of the TME, their interrelationships, and their impact on cancer development is still at its early stages. In this review, we aim to shed light on Interleukin (IL) -6, IL-9, and IL-10, a small group of JAK/STAT signaling-dependent cytokines harboring T cell-suppressive effects in the TME and summarize their mechanisms of action. Additionally, we will explore how advancements in scientific research can help us overcoming the obstacles posed by cytokines that suppress T cells in tumors, with the ultimate objective of stimulating further investigations for the development of novel therapeutic strategies to counteract their tumor-promoting activities.
Collapse
Affiliation(s)
| | | | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| |
Collapse
|
24
|
Gu M, Liu Y, Zheng W, Jing Z, Li X, Guo W, Zhao Z, Yang X, Liu Z, Zhu X, Gao W. Combined targeting of senescent cells and senescent macrophages: A new idea for integrated treatment of lung cancer. Semin Cancer Biol 2024; 106-107:43-57. [PMID: 39214157 DOI: 10.1016/j.semcancer.2024.08.006] [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: 07/17/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer is one of the most common cancers worldwide and a leading cause of cancer-related deaths. Macrophages play a key role in the immune response and the tumour microenvironment. As an important member of the immune system, macrophages have multiple functions, including phagocytosis and clearance of pathogens, modulation of inflammatory responses, and participation in tissue repair and regeneration. In lung cancer, macrophages are considered to be the major cellular component of the tumor-associated inflammatory response and are closely associated with tumorigenesis, progression and metastasis. However, macrophages gradually undergo a senescence process with age and changes in pathological states. Macrophage senescence is an important change in the functional and metabolic state of macrophages and may have a significant impact on lung cancer development. In lung cancer, senescent macrophages interact with other cells in the tumor microenvironment (TME) by secreting senescence-associated secretory phenotype (SASP) factors, which can either promote the proliferation, invasion and metastasis of tumor cells or exert anti-tumor effects through reprogramming or clearance under specific conditions. Therefore, senescent macrophages are considered important potential targets for lung cancer therapy. In this paper, a systematic review of macrophages and their senescence process, and their role in tumors is presented. A variety of inhibitory strategies against senescent macrophages, including enhancing autophagy, inhibiting SASP, reducing DNA damage, and modulating metabolic pathways, were also explored. These strategies are expected to improve lung cancer treatment outcomes by restoring the anti-tumor function of macrophages.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xinwang Zhu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Wei Gao
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
25
|
Sukhamwang A, Inthanon S, Dejkriengkraikul P, Semangoen T, Yodkeeree S. Anti-Cancer Potential of Isoflavone-Enriched Fraction from Traditional Thai Fermented Soybean against Hela Cervical Cancer Cells. Int J Mol Sci 2024; 25:9277. [PMID: 39273231 PMCID: PMC11395072 DOI: 10.3390/ijms25179277] [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: 07/15/2024] [Revised: 08/17/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Cervical cancer is a leading cause of gynecological malignancies and cancer-related deaths among women worldwide. This study investigates the anti-cancer activity of Thua Nao, a Thai fermented soybean, against HeLa cervical carcinoma cells, and explores its underlying mechanisms. Our findings reveal that the ethyl acetate fraction of Thua Nao (TN-EA) exhibits strong anti-cancer potential against HeLa cells. High-performance liquid chromatography (HPLC) analysis identified genistein and daidzein as the major isoflavones in TN-EA responsible for its anti-cancer activity. TN-EA and genistein reduced cell proliferation and induced G2/M phase arrest, while daidzein induced G1 arrest. These responses were associated with the downregulation of cell cycle regulators, including Cyclin B1, cycle 25C (Cdc25C), and phosphorylated cyclin-dependent kinase 1 (CDK-1), and the upregulation of the cell cycle inhibitor p21. Moreover, TN-EA and its active isoflavones promoted apoptosis in HeLa cells through the intrinsic pathway, evidenced by increased levels of cleaved Poly (ADP-ribose) polymerase (PARP) and caspase-3, loss of mitochondrial membrane potential, and the downregulation of anti-apoptotic proteins B-cell leukemia/lymphoma 2 (Bcl-2), B-cell lymphoma-extra-large (Bcl-xL), cellular inhibitor of apoptosis proteins 1 (cIAP), and survivin. Additionally, TN-EA and its active isoflavones effectively reduced cell invasion and migration by downregulating extracellular matrix degradation enzymes, including Membrane type 1-matrix metalloproteinase (MT1-MMP), urokinase-type plasminogen activator (uPA), and urokinase-type plasminogen activator receptor (uPAR), and reduced the levels of the mesenchymal marker N-cadherin. At the molecular level, TN-EA suppressed STAT3 activation via the regulation of JNK and Erk1/2 signaling pathways, leading to reduced proliferation and invasion of HeLa cells.
Collapse
Affiliation(s)
- Amonnat Sukhamwang
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirinada Inthanon
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tistaya Semangoen
- Department of Medical Technology, Faculty of Allied Health Sciences, Burapha University, Chonburi 20131, Thailand
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
26
|
Wang G, Hiramoto K, Ma N, Ohnishi S, Morita A, Xu Y, Yoshikawa N, Chinzei Y, Murata M, Kawanishi S. Immunohistochemical analyses reveal FoxP3 expressions in spleen and colorectal cancer in mice treated with AOM/DSS, and their suppression by glycyrrhizin. PLoS One 2024; 19:e0307038. [PMID: 39150932 PMCID: PMC11329161 DOI: 10.1371/journal.pone.0307038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/27/2024] [Indexed: 08/18/2024] Open
Abstract
We previously demonstrated that glycyrrhizin (GL) suppressed inflammation and carcinogenesis in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced murine model of colorectal cancer (CC). In this study, we found an accumulation of regulatory T cells (Tregs) in the spleen and suppression by GL in model mice. ICR mice were divided into four groups: Control, GL, CC, and GL-treated CC (CC+GL), and were sacrificed 20 weeks after AOM/DSS treatment. We measured spleen weight, areas of white and red pulp, and CD8+ T cells (cytotoxic T lymphocytes, CTL), and CD11c-positive cells (dendritic cells) in splenic tissues and forkhead box protein 3 (FoxP3)-positive cells (Tregs) in colorectal and splenic tissues. In all cases, the CC group showed a significant increase compared with those in Control group, and GL administration significantly attenuated this increase. These results indicate that Tregs accumulated in the spleen may participate in inflammation-related carcinogenesis by suppressing CTL. We also suggest that GL which binds to high-mobility group box 1 (HMGB1), suppresses carcinogenesis with decreasing Tregs in the spleen. Furthermore, there was an expression of FoxP3 in cancer cells, indicating that it may be involved in the malignant transformation of cancer cells.
Collapse
Affiliation(s)
- Guifeng Wang
- Department of Acupuncture and Moxibustion Medical Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
- Institute of Traditional Chinese Medicine, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Shiho Ohnishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Akihiro Morita
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Yifei Xu
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | | | - Yasuo Chinzei
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Shosuke Kawanishi
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| |
Collapse
|
27
|
Regua AT, Bindal S, Najjar MK, Zhuang C, Khan M, Arrigo ABJ, Gonzalez AO, Zhang XR, Zhu JJ, Watabe K, Lo HW. Dual inhibition of the TrkA and JAK2 pathways using entrectinib and pacritinib suppresses the growth and metastasis of HER2-positive and triple-negative breast cancers. Cancer Lett 2024; 597:217023. [PMID: 38852701 DOI: 10.1016/j.canlet.2024.217023] [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: 04/01/2024] [Revised: 05/26/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
HER2-positive and triple-negative breast cancers (TNBC) are difficult to treat and associated with poor prognosis. Despite showing initial response, HER2-positive breast cancers often acquire resistance to HER2-targeted therapies, and TNBC lack effective therapies. To overcome these clinical challenges, we evaluated the therapeutic utility of co-targeting TrkA and JAK2/STAT3 pathways in these breast cancer subtypes. Here, we report the novel combination of FDA-approved TrkA inhibitors (Entrectinib or Larotrectinib) and JAK2 inhibitors (Pacritinib or Ruxolitinib) synergistically inhibited in vitro growth of HER2-positive breast cancer cells and TNBC cells. The Entrectinib-Pacritinib combination inhibited the breast cancer stem cell subpopulation, reduced expression of stemness genes, SOX2 and MYC, and induced apoptosis. The Entrectinib-Pacritinib combination suppressed orthotopic growth of HER2-positive Trastuzumab-refractory breast cancer xenografts and basal patient-derived xenograft (PDXs), reduced tumoral SOX2 and MYC, and induced apoptosis in both mouse models. The Entrectinib-Pacritinib combination inhibited overall metastatic burden, and brain and bone metastases of intracardially inoculated TNBC cells without toxicity. Together, our results demonstrate for the first time that co-inhibition of TrkA and JAK2 synergistically suppresses breast cancer growth and metastasis, thereby providing preclinical evidence that supports future clinical evaluations.
Collapse
Affiliation(s)
- Angelina T Regua
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shivani Bindal
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariana K Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Munazza Khan
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Austin B J Arrigo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anneliese O Gonzalez
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xinhai R Zhang
- Department of Pathology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hui-Wen Lo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
28
|
Youn JK, Lee HR, Ko D, Kim HY. Attenuation of esophageal anastomotic stricture through remote ischemic conditioning in a rat model. Sci Rep 2024; 14:18481. [PMID: 39122787 PMCID: PMC11315918 DOI: 10.1038/s41598-024-69386-4] [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: 04/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Anastomotic stricture is a typical complication of esophageal atresia surgery. Remote ischemic conditioning (RIC) has demonstrated multiorgan benefits, however, its efficacy in the esophagus remains unclear. This study aimed to investigate whether applying RIC after esophageal resection and anastomosis in rats could attenuate esophageal stricture and improve inflammation. Sixty-five male Sprague-Dawley rats were categorized into the following groups: controls with no surgery, resection and anastomosis only, resection and anastomosis with RIC once, and resection and anastomosis with RIC twice. RIC included three cycles of hind-limb ischemia followed by reperfusion. Inflammatory markers associated with the interleukin 6/Janus kinase/ signal transducer and activator of transcription 3 (IL-6/JAK/STAT3) and tumor necrosis factor-alpha/nuclear factor-κB (TNF-α/NF-kB) signaling pathways were evaluated with RNA and protein works. The RIC groups showed significantly lower stricture rates, lower inflammatory markers levels than the resection and anastomosis-only group. The RIC groups had significantly lower IL-6 and TNFa levels than the resection and anastomosis-only group, confirming the inhibitory role of remote ischemic conditioning in the IL-6/JAK/STAT3 and TNF-α/NF-kB signaling pathways. RIC after esophageal resection and anastomosis can reduce the inflammatory response, improving strictures at the esophageal anastomosis site, to be a novel noninvasive intervention for reducing esophageal anastomotic strictures.
Collapse
Affiliation(s)
- Joong Kee Youn
- Department of Pediatric Surgery, Seoul National University Hospital, Seoul, Korea
- Department of Pediatric Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongro-gu, Seoul, 03080, Korea
| | - Hye-Rim Lee
- Department of Pediatric Surgery, Seoul National University Hospital, Seoul, Korea
| | - Dayoung Ko
- Department of Pediatric Surgery, Seoul National University Hospital, Seoul, Korea
- Department of Pediatric Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongro-gu, Seoul, 03080, Korea
| | - Hyun-Young Kim
- Department of Pediatric Surgery, Seoul National University Hospital, Seoul, Korea.
- Department of Pediatric Surgery, Seoul National University College of Medicine, 101 Daehak-ro, Jongro-gu, Seoul, 03080, Korea.
| |
Collapse
|
29
|
Chen X, Ma C, Li Y, Liang Y, Chen T, Han D, Luo D, Zhang N, Zhao W, Wang L, Chen B, Guo H, Yang Q. Trim21-mediated CCT2 ubiquitination suppresses malignant progression and promotes CD4 +T cell activation in breast cancer. Cell Death Dis 2024; 15:542. [PMID: 39079960 PMCID: PMC11289294 DOI: 10.1038/s41419-024-06944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Breast cancer remains a significant global health challenge, and its mechanisms of progression and metastasis are still not fully understood. In this study, analysis of TCGA and GEO datasets revealed a significant increase in CCT2 expression in breast cancer tissues, which was associated with poor prognosis in breast cancer patients. Functional analysis revealed that CCT2 promoted breast cancer growth and metastasis through activation of the JAK2/STAT3 signaling pathway. Additionally, the E3 ubiquitin ligase Trim21 facilitated CCT2 ubiquitination and degradation, significantly reversing the protumor effects of CCT2. Most interestingly, we discovered that exosomal CCT2 derived from breast cancer cells suppressed the activation and proinflammatory cytokine secretion of CD4+ T cell. Mechanistically, exosomal CCT2 constrained Ca2+-NFAT1 signaling, thereby reducing CD40L expression on CD4+ T cell. These findings highlight CCT2 upregulation as a potential driver of breast cancer progression and immune evasion. Our study provides new insights into the molecular mechanisms underlying breast cancer progression, suggesting that CCT2 is a promising therapeutic target and prognostic predictor for breast cancer.
Collapse
Affiliation(s)
- Xi Chen
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chenao Ma
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yaming Li
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yiran Liang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tong Chen
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dianwen Han
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dan Luo
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ning Zhang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenjing Zhao
- Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lijuan Wang
- Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Bing Chen
- Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hong Guo
- Shandong Desheng Bioengineering Company Limited, Jinan, Shandong, China
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Research Institute of Breast Cancer, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
30
|
Dawalibi A, Alosaimi AA, Mohammad KS. Balancing the Scales: The Dual Role of Interleukins in Bone Metastatic Microenvironments. Int J Mol Sci 2024; 25:8163. [PMID: 39125732 PMCID: PMC11311339 DOI: 10.3390/ijms25158163] [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: 06/30/2024] [Revised: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Bone metastases, a common and debilitating consequence of advanced cancers, involve a complex interplay between malignant cells and the bone microenvironment. Central to this interaction are interleukins (ILs), a group of cytokines with critical roles in immune modulation and inflammation. This review explores the dualistic nature of pro-inflammatory and anti-inflammatory interleukins in bone metastases, emphasizing their molecular mechanisms, pathological impacts, and therapeutic potential. Pro-inflammatory interleukins, such as IL-1, IL-6, and IL-8, have been identified as key drivers in promoting osteoclastogenesis, tumor proliferation, and angiogenesis. These cytokines create a favorable environment for cancer cell survival and bone degradation, contributing to the progression of metastatic lesions. Conversely, anti-inflammatory interleukins, including IL-4, IL-10, and IL-13, exhibit protective roles by modulating immune responses and inhibiting osteoclast activity. Understanding these opposing effects is crucial for developing targeted therapies aimed at disrupting the pathological processes in bone metastases. Key signaling pathways, including NF-κB, JAK/STAT, and MAPK, mediate the actions of these interleukins, influencing tumor cell survival, immune cell recruitment, and bone remodeling. Targeting these pathways presents promising therapeutic avenues. Current treatment strategies, such as the use of denosumab, tocilizumab, and emerging agents like bimekizumab and ANV419, highlight the potential of interleukin-targeted therapies in mitigating bone metastases. However, challenges such as therapeutic resistance, side effects, and long-term efficacy remain significant hurdles. This review also addresses the potential of interleukins as diagnostic and prognostic biomarkers, offering insights into patient stratification and personalized treatment approaches. Interleukins have multifaceted roles that depend on the context, including the environment, cell types, and cellular interactions. Despite substantial progress, gaps in research persist, particularly regarding the precise mechanisms by which interleukins influence the bone metastatic niche and their broader clinical implications. While not exhaustive, this overview underscores the critical roles of interleukins in bone metastases and highlights the need for continued research to fully elucidate their complex interactions and therapeutic potential. Addressing these gaps will be essential for advancing our understanding and treatment of bone metastases in cancer patients.
Collapse
Affiliation(s)
- Ahmad Dawalibi
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Amal Ahmed Alosaimi
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
31
|
Zhou L, Le MNU, Du Y, Chen X, Jin M, Xiang H, Xia LE, Zhou J, He J, Ning Y. A novel cancer-germline gene DAZL promotes progression and cisplatin resistance of non-small cell lung cancer by upregulating JAK2 and MCM8. Gene 2024; 916:148449. [PMID: 38588931 DOI: 10.1016/j.gene.2024.148449] [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: 12/24/2023] [Revised: 03/05/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024]
Abstract
Germline-specific genes are usually activated in cancer cells and drive cancer progression; such genes are called cancer-germline or cancer-testis genes. The RNA-binding protein DAZL is predominantly expressed in germ cells and plays a role in gametogenesis as a translational activator or repressor. However, its expression and role in non-small cell lung cancer (NSCLC) are unknown. Here, mining of RNA-sequencing data from public resources and immunohistochemical analysis of tissue microarrays showed that DAZL was expressed exclusively in testis among normal human tissues but ectopically expressed in NSCLC tissues. Testis and NSCLC cells expressed the shorter and longer transcript variants of the DAZL gene, respectively. Overexpression of the longer DAZL transcript promoted tumor growth in a mouse xenograft model. Silencing of DAZL suppressed cell proliferation, colony formation, migration, invasion, and cisplatin resistance in vitro and tumor growth in vivo. Quantitative proteomic analysis based on tandem mass tag and Western blot analysis showed that DAZL upregulated the expression of JAK2 and MCM8. RNA-binding protein immunoprecipitation assays showed that DAZL bound to the mRNA of JAK2 and MCM8. The JAK2 inhibitor fedratinib attenuated the oncogenic outcomes induced by DAZL overexpression, whereas silencing MCM8 counteracted the effects of DAZL overexpression on cisplatin-damaged DNA synthesis and half-maximal inhibitory concentration of cisplatin. In conclusion, DAZL was identified as a novel cancer-germline gene that enhances the translation of JAK2 and MCM8 to promote NSCLC progression and resistance to cisplatin, respectively. These findings suggest that DAZL is a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Lin Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Hunan Normal University, Changsha 41007, Hunan, China; College of Clinical Laboratory, Changsha Medical University, Changsha, Hunan, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Mai Nhu Uyen Le
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Yan Du
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Xi Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Miao Jin
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Hu Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Liang E Xia
- Chongzuo Key Laboratory of Biomedical Clinical Transformation, The People's Hospital of Chongzuo, Youjiang Medical University for Nationalities, Chongzuo 532200, Guangxi, China.
| | - Jianlin Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, Hunan, China.
| | - Jun He
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Hunan Normal University, Changsha 41007, Hunan, China.
| | - Yichong Ning
- Chongzuo Key Laboratory of Biomedical Clinical Transformation, The People's Hospital of Chongzuo, Youjiang Medical University for Nationalities, Chongzuo 532200, Guangxi, China.
| |
Collapse
|
32
|
Zhang Y, Li J, Feng L, Cheng Y, Shi L, Yang Q, Wang Z, Yi X, Zhong G, Sun X, Cheng Z, Guo M. STAC3 as a poor prognostic biomarker in renal clear cell carcinoma: relationship with immune infiltration. Am J Cancer Res 2024; 14:3294-3316. [PMID: 39113874 PMCID: PMC11301302 DOI: 10.62347/eaqw3113] [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: 04/02/2024] [Accepted: 06/17/2024] [Indexed: 08/10/2024] Open
Abstract
Calcium ions (Ca2+) are crucial in tumorigenesis and progression, with their elevated levels indicating a negative prognosis in Kidney Renal Clear Cell Carcinoma (KIRC). The influence of genes regulating calcium ions on the survival outcomes of KIRC patients and their interaction with the tumor's immune microenvironment is yet to be fully understood. This study analyzed gene expression data from KIRC tumor and adjacent non-tumor tissues using the TCGA-KIRC dataset to pinpoint genes that are differentially expressed in KIRC. Intersection of these genes with those regulating calcium ions highlighted specific calcium ion-regulating genes that exhibit differential expression in KIRC. Subsequently, prognostic risk models were developed using univariate Cox and LASSO-Cox regression analyses to verify their diagnostic precision. Additionally, the study investigated the correlation between tumor immunity and KIRC patient outcomes, assessing the contribution of STAC3 genes to tumor immunity. Further exploration entailed SSGASE, single-cell analysis, pseudotime analysis and both in vivo and in vitro experiments to evaluate STAC3's role in tumor immunity and progression. Notably, STAC3 was significantly overexpressed in tumor specimens and positively correlated with the degree of malignancy of KIRC, affecting patients' prognosis. Elevated STAC3 expression correlated with enhanced immune infiltration in KIRC tumors. Furthermore, silencing STAC3 curtailed KIRC cell proliferation, migration, invasion, and stemness properties. Experimental models in mice confirmed that STAC3 knockdown led to a reduction in tumor growth. Elevated STAC3 expression is intricately linked with immune infiltration in KIRC tumors, as well as with the aggressive biological behaviors of tumor cells, including their proliferation, migration, and invasion. Targeting STAC3 presents a promising strategy to augment the efficacy of current therapeutic approaches and to better the survival outcomes of patients with KIRC.
Collapse
Affiliation(s)
- Yingnan Zhang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Jingtao Li
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Luwen Feng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Yue Cheng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Linlin Shi
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Qian Yang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Zhaoyang Wang
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Xuan Yi
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Guocai Zhong
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Xueying Sun
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Zhifeng Cheng
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| | - Min Guo
- The Fourth Affiliated Hospital of Harbin Medical University Harbin 150001, Heilongjiang, China
| |
Collapse
|
33
|
Yu Y, Li X, Ying Q, Zhang Z, Liu W, Su J. Synergistic Effects of Shed-Derived Exosomes, Cu 2+, and an Injectable Hyaluronic Acid Hydrogel on Antibacterial, Anti-inflammatory, and Osteogenic Activity for Periodontal Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:33053-33069. [PMID: 38899855 DOI: 10.1021/acsami.4c05062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The primary pathology of periodontitis involves the gradual deterioration of periodontal tissues resulting from the inflammatory reaction triggered by bacterial infection. In this study, a novel drug for periodontal pocket injection, known as the Shed-Cu-HA hydrogel, was developed by incorporating copper ions (Cu2+) and Shed-derived exosomes (Shed-exo) inside the hyaluronic acid (HA) hydrogel. Suitable concentrations of Cu2+ and Shed-exo released from Shed-Cu-HA enhanced cell viability and cell proliferation of human periodontal ligament stem cells. Additionally, the Shed-Cu-HA demonstrated remarkable antibacterial effects against the key periodontal pathogen (Aa) owing to the synergistic effect of Cu2+ and HA. Furthermore, the material effectively suppressed macrophage inflammatory response via the IL-6/JAK2/STAT3 pathway. Moreover, the Shed-Cu-HA, combining the inflammation-regulating properties of HA with the synergistic osteogenic activity of Shed-exo and Cu2+, effectively upregulated the expression of genes and proteins associated with osteogenic differentiation. The experimental findings from a mouse periodontitis model demonstrated that the administration of Shed-Cu-HA effectively reduced the extent of inflammatory cell infiltration and bacterial infections in gingival tissues and facilitated the regeneration of periodontal bone tissues and collagen after 2 and 4 weeks of injection. Consequently, it holds significant prospects for future applications in periodontitis treatment.
Collapse
Affiliation(s)
- Yiqiang Yu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Xuejing Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Qiao Ying
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Zhanwei Zhang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Weicai Liu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| | - Jiansheng Su
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai 200072, China
| |
Collapse
|
34
|
Boidin L, Moinard M, Moussaron A, Merlier M, Moralès O, Grolez GP, Baydoun M, Mohd-Gazzali A, Tazizi MHDM, Allah HHA, Kerbage Y, Arnoux P, Acherar S, Frochot C, Delhem N. Targeted Photodynamic Therapy using a Vectorized Photosensitizer coupled to Folic Acid Analog induces Ovarian Tumor Cell Death and inhibits IL-6-mediated Inflammation. J Control Release 2024; 371:351-370. [PMID: 38789088 DOI: 10.1016/j.jconrel.2024.05.033] [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: 12/15/2023] [Revised: 05/10/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Ovarian cancer (OC) is one of the most lethal cancers among women. Frequent recurrence in the peritoneum due to the presence of microscopic tumor residues justifies the development of new therapies. Indeed, our main objective is to develop a targeted photodynamic therapy (PDT) treatment of peritoneal carcinomatosis from OC to improve the life expectancy of cancer patients. Herein, we propose a targeted-PDT using a vectorized photosensitizer (PS) coupled with a newly folic acid analog (FAA), named PSFAA, in order to target folate receptor alpha (FRα) overexpressed on peritoneal metastasis. This PSFAA was the result of the coupling of pyropheophorbide-a (Pyro-a), as the PS, to a newly synthesized FAA via a polyethylene glycol (PEG) spacer. The selectivity and the PDT efficacy of PSFAA was evaluated on two human OC cell lines overexpressing FRα compared to fibrosarcoma cells underexpressing FRα. Final PSFAA, including the synthesis of a newly FAA and its conjugation to Pyro-a, was obtained after 10 synthesis steps, with an overall yield of 19%. Photophysical properties of PSFAA in EtOH were performed and showed similarity with those of free Pyro-a, such as the fluorescence and singlet oxygen quantum yields (Φf = 0.39 and ΦΔ = 0.53 for free Pyro-a, and Φf = 0.26 and ΦΔ = 0.41 for PSFAA). Any toxicity of PSFAA was noticed. After light illumination, a dose-dependent effect on PS concentration and light dose was shown. Furthermore, a PDT efficacy of PSFAA on OC cell secretome was detected inducing a decrease of a pro-inflammatory cytokine secretion (IL-6). This new PSFAA has shown promising biological properties highlighting the selectivity of the therapy opening new perspectives in the treatment of a cancer in a therapeutic impasse.
Collapse
Affiliation(s)
- Léa Boidin
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Morgane Moinard
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France
| | - Albert Moussaron
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France; Univ. Lorraine, CNRS, UMR7375 - LCPM - Laboratoire de Chimie-Physique Macromoléculaire, Nancy F-54000, France
| | - Margaux Merlier
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Olivier Moralès
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France; Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille F-59000, France
| | - Guillaume Paul Grolez
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Martha Baydoun
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Amirah Mohd-Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Hassan Hadi Abd Allah
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Yohan Kerbage
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France
| | - Philippe Arnoux
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France
| | - Samir Acherar
- Univ. Lorraine, CNRS, UMR7375 - LCPM - Laboratoire de Chimie-Physique Macromoléculaire, Nancy F-54000, France.
| | - Céline Frochot
- Univ. Lorraine, CNRS, UMR7274 - LRGP- Laboratoire des Réactions et Génie des Procédés, Nancy F-54000, France.
| | - Nadira Delhem
- Univ. Lille, Inserm, CHU Lille, U1189-ONCOTHAI-Assisted Laser Therapy and Immunotherapy for Oncology, Lille F-59000, France.
| |
Collapse
|
35
|
Weidle UH, Nopora A. CircRNAs in Pancreatic Cancer: New Tools for Target Identification and Therapeutic Intervention. Cancer Genomics Proteomics 2024; 21:327-349. [PMID: 38944427 PMCID: PMC11215428 DOI: 10.21873/cgp.20451] [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: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/01/2024] Open
Abstract
We have reviewed the literature for circular RNAs (circRNAs) with efficacy in preclinical pancreatic-cancer related in vivo models. The identified circRNAs target chemoresistance mechanisms (n=5), secreted proteins and transmembrane receptors (n=15), transcription factors (n=9), components of the signaling- (n=11), ubiquitination- (n=2), autophagy-system (n=2), and others (n=9). In addition to identifying targets for therapeutic intervention, circRNAs are potential new entities for treatment of pancreatic cancer. Up-regulated circRNAs can be inhibited by antisense oligonucleotides (ASO), small interfering RNAs (siRNAs), short hairpin RNAs (shRNAs) or clustered regularly interspaced short-palindromic repeats-CRISPR associated protein (CRISPR-CAS)-based intervention. The function of down-regulated circRNAs can be reconstituted by replacement therapy using plasmids or virus-based vector systems. Target validation experiments and the development of improved delivery systems for corresponding agents were examined.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
36
|
Xie W, Zhang Y, Zhang Z, Li Q, Tao L, Zhang R. ISG15 promotes tumor progression via IL6/JAK2/STAT3 signaling pathway in ccRCC. Clin Exp Med 2024; 24:140. [PMID: 38951255 PMCID: PMC11217101 DOI: 10.1007/s10238-024-01414-z] [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: 04/07/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Although renal cell carcinoma (RCC) is a prevalent type of cancer, the most common pathological subtype, clear cell renal cell carcinoma (ccRCC), still has poorly understood molecular mechanisms of progression. Moreover, interferon-stimulated gene 15 (ISG15) is associated with various types of cancer; however, its biological role in ccRCC remains unclear.This study aimed to explore the role of ISG15 in ccRCC progression.ISG15 expression was upregulated in ccRCC and associated with poor prognosis. RNA sequence analysis and subsequent experiments indicated that ISG15 modulated IL6/JAK2/STAT3 signaling to promote ccRCC proliferation, migration, and invasion. Additionally, our animal experiments confirmed that sustained ISG15 knockdown reduced tumor growth rate in nude mice and promoted cell apoptosis. ISG15 modulates the IL6/JAK2/STAT3 pathway, making it a potential therapeutic target and prognostic biomarker for ccRCC.
Collapse
Affiliation(s)
- Wei Xie
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road. 74, Jiangbei, Chongqing, China
| | - Yuanfeng Zhang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road. 74, Jiangbei, Chongqing, China
| | - Zhechuan Zhang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road. 74, Jiangbei, Chongqing, China
| | - Qinke Li
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road. 74, Yuzhong, Chongqing, China
| | - Lesha Tao
- Department of Urology, Chongqing People's Hospital, Xingguang Road.118, Chongqing, China
| | - Ronggui Zhang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Linjiang Road. 74, Jiangbei, Chongqing, China.
- Department of Urology, Chongqing People's Hospital, Xingguang Road.118, Chongqing, China.
| |
Collapse
|
37
|
Zhang H, Liu X, Shi J, Su X, Xie J, Meng Q, Dong H. Research progress on the mechanism of exosome-mediated virus infection. Front Cell Infect Microbiol 2024; 14:1418168. [PMID: 38988816 PMCID: PMC11233549 DOI: 10.3389/fcimb.2024.1418168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Exosomes are extracelluar vesicles that facilitate intercellular communication and are pivotal in post-transcriptional regulation within cellular gene regulatory networks, impacting pathogen dynamics. These vesicles serve as crucial regulators of immune responses, mediating cellular interactions and enabling the introduction of viral pathogenic regions into host cells. Exosomes released from virus-infected cells harbor diverse microRNAs (miRNAs), which can be transferred to recipient cells, thereby modulating virus infection. This transfer is a critical element in the molecular interplay mediated by exosomes. Additionally, the endosomal sorting complex required for transport (ESCRT) within exosomes plays a vital role in virus infection, with ESCRT components binding to viral proteins to facilitate virus budding. This review elucidates the roles of exosomes and their constituents in the invasion of host cells by viruses, aiming to shed new light on the regulation of viral transmission via exosomes.
Collapse
Affiliation(s)
- Hanjia Zhang
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Xuanyi Liu
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Jiuming Shi
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Xuan Su
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Jiayuan Xie
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
| | - Qingfeng Meng
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun, Jilin, China
- Engineering Research Center of Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun, China
| |
Collapse
|
38
|
Mohamed AH, Ahmed AT, Al Abdulmonem W, Bokov DO, Shafie A, Al-Hetty HRAK, Hsu CY, Alissa M, Nazir S, Jamali MC, Mudhafar M. Interleukin-6 serves as a critical factor in various cancer progression and therapy. Med Oncol 2024; 41:182. [PMID: 38900329 DOI: 10.1007/s12032-024-02422-5] [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: 04/24/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Interleukin-6 (IL-6), a pro-inflammatory cytokine, plays a crucial role in host immune defense and acute stress responses. Moreover, it modulates various cellular processes, including proliferation, apoptosis, angiogenesis, and differentiation. These effects are facilitated by various signaling pathways, particularly the signal transducer and activator of transcription 3 (STAT3) and Janus kinase 2 (JAK2). However, excessive IL-6 production and dysregulated signaling are associated with various cancers, promoting tumorigenesis by influencing all cancer hallmarks, such as apoptosis, survival, proliferation, angiogenesis, invasiveness, metastasis, and notably, metabolism. Emerging evidence indicates that selective inhibition of the IL-6 signaling pathway yields therapeutic benefits across diverse malignancies, such as multiple myeloma, prostate, colorectal, renal, ovarian, and lung cancers. Targeting key components of IL-6 signaling, such as IL-6Rs, gp130, STAT3, and JAK via monoclonal antibodies (mAbs) or small molecules, is a heavily researched approach in preclinical cancer studies. The purpose of this study is to offer an overview of the role of IL-6 and its signaling pathway in various cancer types. Furthermore, we discussed current preclinical and clinical studies focusing on targeting IL-6 signaling as a therapeutic strategy for various types of cancer.
Collapse
Affiliation(s)
- Asma'a H Mohamed
- Biomedical Engineering Department, College of Engineering and Technologies, Al-Mustaqbal University, Babil, Hilla, 51001, Iraq
| | - Abdulrahman T Ahmed
- Department of Nursing, Al-Maarif University College, Ramadi, AL-Anbar Governorate, Iraq.
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy named after A.P. Nelyubin, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow, Russian Federation, 119991
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow, Russian Federation, 109240
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | | | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, 85004, USA
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shahid Nazir
- School of Science and Technology, University of New England, Armidale, NSW, Australia
| | - Mohammad Chand Jamali
- Faculty of Medical and Health Sciences, Liwa College, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mustafa Mudhafar
- Department of Medical Physics, College of Applied Medical Sciences, University of Kerbala, Karbala, 56001, Iraq
- Department of Anesthesia Techniques and Intensive Care, Al-Taff University College, Kerbala, 56001, Iraq
| |
Collapse
|
39
|
Viken JK, Olsen T, Drevon CA, Hjorth M, Birkeland KI, Norheim F, Lee-Ødegård S. Intra-Individual Variations in How Insulin Sensitivity Responds to Long-Term Exercise: Predictions by Machine Learning Based on Large-Scale Serum Proteomics. Metabolites 2024; 14:335. [PMID: 38921470 PMCID: PMC11206077 DOI: 10.3390/metabo14060335] [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: 05/18/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Physical activity is effective for preventing and treating type 2 diabetes, but some individuals do not achieve metabolic benefits from exercise ("non-responders"). We investigated non-responders in terms of insulin sensitivity changes following a 12-week supervised strength and endurance exercise program. We used a hyperinsulinaemic euglycaemic clamp to measure insulin sensitivity among 26 men aged 40-65, categorizing them into non-responders or responders based on their insulin sensitivity change scores. The exercise regimen included VO2max, muscle strength, whole-body MRI scans, muscle and fat biopsies, and serum samples. mRNA sequencing was performed on biopsies and Olink proteomics on serum samples. Non-responders showed more visceral and intramuscular fat and signs of dyslipidaemia and low-grade inflammation at baseline and did not improve in insulin sensitivity following exercise, although they showed gains in VO2max and muscle strength. Impaired IL6-JAK-STAT3 signalling in non-responders was suggested by serum proteomics analysis, and a baseline serum proteomic machine learning (ML) algorithm predicted insulin sensitivity responses with high accuracy, validated across two independent exercise cohorts. The ML model identified 30 serum proteins that could forecast exercise-induced insulin sensitivity changes.
Collapse
Affiliation(s)
- Jonas Krag Viken
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0313 Oslo, Norway; (J.K.V.); (K.I.B.)
| | - Thomas Olsen
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0313 Oslo, Norway; (T.O.); (C.A.D.); (M.H.); (F.N.)
| | - Christian André Drevon
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0313 Oslo, Norway; (T.O.); (C.A.D.); (M.H.); (F.N.)
- Vitas Ltd., Oslo Science Park, 0349 Oslo, Norway
| | - Marit Hjorth
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0313 Oslo, Norway; (T.O.); (C.A.D.); (M.H.); (F.N.)
| | - Kåre Inge Birkeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0313 Oslo, Norway; (J.K.V.); (K.I.B.)
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0586 Oslo, Norway
| | - Frode Norheim
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, 0313 Oslo, Norway; (T.O.); (C.A.D.); (M.H.); (F.N.)
| | - Sindre Lee-Ødegård
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0313 Oslo, Norway; (J.K.V.); (K.I.B.)
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, 0586 Oslo, Norway
| |
Collapse
|
40
|
Sasaki M, Kato D, Murakami K, Yoshida H, Takase S, Otsubo T, Ogiwara H. Targeting dependency on a paralog pair of CBP/p300 against de-repression of KREMEN2 in SMARCB1-deficient cancers. Nat Commun 2024; 15:4770. [PMID: 38839769 PMCID: PMC11153594 DOI: 10.1038/s41467-024-49063-w] [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: 06/19/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
SMARCB1, a subunit of the SWI/SNF chromatin remodeling complex, is the causative gene of rhabdoid tumors and epithelioid sarcomas. Here, we identify a paralog pair of CBP and p300 as a synthetic lethal target in SMARCB1-deficient cancers by using a dual siRNA screening method based on the "simultaneous inhibition of a paralog pair" concept. Treatment with CBP/p300 dual inhibitors suppresses growth of cell lines and tumor xenografts derived from SMARCB1-deficient cells but not from SMARCB1-proficient cells. SMARCB1-containing SWI/SNF complexes localize with H3K27me3 and its methyltransferase EZH2 at the promotor region of the KREMEN2 locus, resulting in transcriptional downregulation of KREMEN2. By contrast, SMARCB1 deficiency leads to localization of H3K27ac, and recruitment of its acetyltransferases CBP and p300, at the KREMEN2 locus, resulting in transcriptional upregulation of KREMEN2, which cooperates with the SMARCA1 chromatin remodeling complex. Simultaneous inhibition of CBP/p300 leads to transcriptional downregulation of KREMEN2, followed by apoptosis induction via monomerization of KREMEN1 due to a failure to interact with KREMEN2, which suppresses anti-apoptotic signaling pathways. Taken together, our findings indicate that simultaneous inhibitors of CBP/p300 could be promising therapeutic agents for SMARCB1-deficient cancers.
Collapse
Affiliation(s)
- Mariko Sasaki
- Division of Cancer Therapeutics, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Daiki Kato
- Cancer Research Unit, Sumitomo Pharma Co., Ltd, 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Karin Murakami
- Cancer Research Unit, Sumitomo Pharma Co., Ltd, 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shohei Takase
- Division of Cancer Therapeutics, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tsuguteru Otsubo
- Cancer Research Unit, Sumitomo Pharma Co., Ltd, 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Hideaki Ogiwara
- Division of Cancer Therapeutics, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
41
|
Du H, You L, Wu A, Wang F, Yu J, Chen C. Resolvin D1 Inhibits IL-6-Induced Epithelial-Mesenchymal Transition of Colorectal Cancer Cells by Targeting IL-6/STAT3 Signaling. Cell Biochem Biophys 2024; 82:1453-1461. [PMID: 38740668 DOI: 10.1007/s12013-024-01299-5] [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] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
Colorectal cancer (CRC) has emerged as a prevalent malignancy worldwide, exhibiting the high morbidity and mortality rates. Resolvin D1 (RvD1) can exert anti-inflammation and anti-cancer effects on various diseases. This study is aimed to explore the role of RvD1 in CRC cells. HCT15 and SW480 cells were stimulated with IL-6 in our study. A series of assays such as CCK-8, colony formation, wound healing, Transwell, Western blotting, and immunofluorescence staining were designed and conducted to figure out the role of RvD1 in CRC cells. RvD1 suppressed IL-6-induced SW480 and HCT15 cell proliferation. In addition, RvD1 inhibited IL-6-induced SW480 and HCT15 cell migration, invasion, and EMT process. In mechanism, RvD1 inhibited the activation of IL-6/STAT3 signaling in SW480 and HCT15 cells. Angoline strengthened the inhibitive effect of RvD1 on cell malignancy. RvD1 inhibited cell growth, migration, invasion and EMT process by inactivating IL-6/STAT3 signaling in CRC.
Collapse
Affiliation(s)
- Heng Du
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Lijuan You
- Department of Anesthesiology, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Anding Wu
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Fei Wang
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Jie Yu
- Department of Gestrointestinal Surgery, Huanggang Central Hospital Affiliated to Yangtze University, Changsha, 438000, China
| | - Chaowu Chen
- Department of Gestrointestinal Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 438000, China.
| |
Collapse
|
42
|
Gu D, Zhao X, Song J, Xiao J, Zhang L, Deng G, Li D. Expression and clinical significance of interleukin-6 pathway in cholangiocarcinoma. Front Immunol 2024; 15:1374967. [PMID: 38881895 PMCID: PMC11176422 DOI: 10.3389/fimmu.2024.1374967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Background Cholangiocarcinoma (CCA) is a typical inflammation-induced malignancy, and elevated serum interleukin-6 (IL-6) levels have been reported to be linked to the onset and progression of CCA. We aim to investigate the potential prognostic value of the IL-6 pathway for CCA. Methods We detected the expressions of IL-6, IL-6R, glycoprotein (gp130), C-reactive protein (CRP), Janus kinase 2 (JAK2), and signal transducer and activator of transcription 3 (STAT3) in CCA tissue microarray using multiplex immunofluorescence. Furthermore, the clinical associations and prognostic values were assessed. Finally, single-cell transcriptome analysis was performed to evaluate the expression level of IL-6 pathway genes in CCA. Results The results revealed that the expression of IL-6 was lower, while the expression of STAT3 was higher in tumor tissues compared to normal tissues. Especially in tumor microenvironment, the expression of IL-6 pathway genes was generally downregulated. Importantly, gp130 was strongly correlated with JAK2 in tumor tissues, while it was moderately correlated with JAK2 in normal tissue. Although none of the gene expressions were directly associated with overall survival and disease-free survival, our study found that IL-6, IL-6R, CRP, gp130, and JAK2 were inversely correlated with vascular invasion, which is a risk factor for poor prognosis in patients with CCA. Conclusion The findings from this study suggest that the IL-6 signaling pathway may have a potential prognostic value for CCA. Further investigation is needed to understand the underlying molecular mechanisms of the IL-6 pathway in CCA.
Collapse
Affiliation(s)
- Dongqing Gu
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Viral Infectious Diseases, Chongqing, China
| | - Xin Zhao
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jing Song
- Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University (CHCMU), Chongqing, China
| | - Jianmei Xiao
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Viral Infectious Diseases, Chongqing, China
| | - Leida Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Viral Infectious Diseases, Chongqing, China
| | - Dajiang Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
43
|
Wang Y, Chu Y, Dai H, Zheng Y, Chen R, Zhou C, Zhong Y, Zhan C, Luo J. Protective role of pretreatment with Anisodamine against sepsis-induced diaphragm atrophy via inhibiting JAK2/STAT3 pathway. Int Immunopharmacol 2024; 133:112133. [PMID: 38652962 DOI: 10.1016/j.intimp.2024.112133] [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: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
There is an increasing tendency for sepsis patients to suffer from diaphragm atrophy as well as mortality. Therefore, reducing diaphragm atrophy could benefit sepsis patients' prognoses. Studies have shown that Anisodamine (Anis) can exert antioxidant effects when blows occur. However, the role of Anisodamine in diaphragm atrophy in sepsis patients has not been reported. Therefore, this study investigated the antioxidant effect of Anisodamine in sepsis-induced diaphragm atrophy and its mechanism. We used cecal ligation aspiration (CLP) to establish a mouse septic mode and stimulated the C2C12 myotube model with lipopolysaccharide (LPS). After treatment with Anisodamine, we measured the mice's bodyweight, diaphragm weight, fiber cross-sectional area and the diameter of C2C12 myotubes. The malondialdehyde (MDA) levels in the diaphragm were detected using the oxidative stress kit. The expression of MuRF1, Atrogin1 and JAK2/STAT3 signaling pathway components in the diaphragm and C2C12 myotubes was measured by RT-qPCR and Western blot. The mean fluorescence intensity of ROS in C2C12 myotubes was measured by flow cytometry. Meanwhile, we also measured the levels of Drp1 and Cytochrome C (Cyt-C) in vivo and in vitro by Western blot. Our study revealed that Anisodamine alleviated the reduction in diaphragmatic mass and the loss of diaphragmatic fiber cross-sectional area and attenuated the atrophy of the C2C12 myotubes by inhibiting the expression of E3 ubiquitin ligases. In addition, we observed that Anisodamine inhibited the JAK2/STAT3 signaling pathway and protects mitochondrial function. In conclusion, Anisodamine alleviates sepsis-induced diaphragm atrophy, and the mechanism may be related to inhibiting the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yurou Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Chu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongkai Dai
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingfang Zheng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renyu Chen
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanxia Zhong
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengye Zhan
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlong Luo
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
44
|
Long J, Zhao W, Xiang Y, Wang Y, Xiang W, Liu X, Jiang M, Song Y, Hu J. STAT3 promotes cytoplasmic-nuclear translocation of RNA-binding protein HuR to inhibit IL-1β-induced IL-8 production. Int Immunopharmacol 2024; 133:112065. [PMID: 38608448 DOI: 10.1016/j.intimp.2024.112065] [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: 11/25/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Signal transducer and activator of transcription 3 (STAT3) functions to regulate inflammation and immune response, but its mechanism is not fully understood. We report here that STAT3 inhibitors Stattic and Niclosamide up-regulated IL-1β-induced IL-8 production in C33A, CaSki, and Siha cervical cancer cells. As expected, IL-1β-induced IL-8 production was also up-regulated through the molecular inhibition of STAT3 by use of CRISPR/Cas9 technology. Unexpectedly, IL-1β induced IL-8 production via activating ERK and P38 signal pathways, but neither STAT3 inhibitors nor STAT3 knockout affected IL-1β-induced signal transduction, suggesting that STAT3 decreases IL-8 production not via inhibition of signal transduction. To our surprise, STAT3 inhibition increased the stabilization, and decreased the degradation of IL-8 mRNA, suggesting a post-transcriptional regulation of IL-1β-induced IL-8. Moreover, Dihydrotanshinone I, an inhibitor of RNA-binding protein HuR, down-regulated IL-1β-induced IL-8 dose-dependently. HuR inhibition by CRISPR/Cas9 also decreased IL-8 production induced by IL-1β. Mechanistically, co-immunoprecipitation results showed that STAT3 did not react with HuR directly, but STAT3 inhibition increased the protein levels of HuR in cytoplasm. And IL-6 activation of STAT3 induced HuR cytoplasmic-nuclear transport. Taken together, these results suggest that STAT3 contributes to HuR nuclear localization and inhibits Il-1β-induced IL-8 production through this non-transcriptional mechanism.
Collapse
Affiliation(s)
- Jiangwen Long
- Department of Clinical Laboratory, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Wang Zhao
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Yangen Xiang
- Department of Clinical Laboratory, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Yufei Wang
- Department of Clinical Laboratory, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China; Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Wei Xiang
- Department of Clinical Laboratory, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China; Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Xueting Liu
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Manli Jiang
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China
| | - Yinghui Song
- Central Laboratory, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Jinyue Hu
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha 410004, China.
| |
Collapse
|
45
|
Liao Y, Xie X, Zhang C, Zhong H, Shan L, Yu P, Xu L. Quercetin exerts anti-tumor immune mechanism by regulating IL-6/JAK2/STAT3 signaling pathway to deplete Treg cells. Toxicon 2024; 243:107747. [PMID: 38714236 DOI: 10.1016/j.toxicon.2024.107747] [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: 12/19/2023] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/09/2024]
Abstract
Breast cancer is still the leading cause of death among women worldwide. Due to the lack of effective drug targets, triple-negative breast cancer has a worse prognosis and higher mortality compared with other types of breast cancer, and chemotherapy is still the main treatment for triple-negative breast cancer at present. Quercetin (QUE) is a flavonoid compound found in a variety of fruits and vegetables. The mechanism of QUE has been extensively studied, such as prostate cancer, colon cancer, ovarian cancer, etc. However, the anti-tumor immune mechanism of QUE in triple-negative breast cancer remains unclear. Therefore, we assessed the anti-tumor immune effects of QUE on triple-negative breast cancer using both 4T1 cells and a xenograft mouse model of 4T1 cells. In vitro, we examined the inhibitory effects of QUE on 4T1 cells and its molecular mechanisms through MTT, Transwell, ELISA, and Western blotting. In vivo, by establishing a xenograft mouse model, we utilized flow cytometry, immunohistochemistry, ELISA, and Western blotting to evaluate the anti-tumor immune effects of QUE on triple-negative breast cancer. The results indicate that QUE inhibits the proliferation, migration, and invasion of 4T1 cells, concurrently significantly suppressing the IL-6/JAK2/STAT3 signaling pathway. Furthermore, it depletes Treg cell content in 4T1 xenograft mice, thereby improving the tumor immune microenvironment and promoting the cytotoxicity of relevant tumor immune cells. These findings suggest that QUE may serve as a potential adjuvant for immune therapy in triple-negative breast cancer.
Collapse
Affiliation(s)
- Yupei Liao
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Xiaoqing Xie
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Chu Zhang
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Haijing Zhong
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Pei Yu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China
| | - Lipeng Xu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese. Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) /State Key Laboratory of Bioactive Molecules and Druggability Assessment of China, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
46
|
Yang P, He S, Ye L, Weng H. Transcription Factor ETV4 Activates AURKA to Promote PD-L1 Expression and Mediate Immune Escape in Lung Adenocarcinoma. Int Arch Allergy Immunol 2024; 185:910-920. [PMID: 38781935 DOI: 10.1159/000537754] [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: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 05/25/2024] Open
Abstract
INTRODUCTION The occurrence and progression of lung adenocarcinoma (LUAD) impair T-cell immune responses, causing immune escape and subsequently affecting the efficacy of immunotherapy in patients. Aurora kinase A (AURKA) is upregulated in varying cancers, but its role in LUAD immune escape is elusive. This work attempted to explore molecular mechanisms of AURKA regulation in LUAD immune escape. METHODS Through bioinformatics analysis, AURKA level in LUAD was evaluated, and potential upstream transcription factors of AURKA were predicted using hTFtarget. ETS variant transcription factor 4 (ETV4) expression in LUAD was analyzed through The Cancer Genome Atlas. Pearson's correlation analysis was then utilized to test the correlation between AURKA and ETV4. Interaction and binding between AURKA and ETV4 were validated through dual-luciferase assay and chromatin immunoprecipitation. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) tested relative mRNA expression of AURKA and ETV4 in LUAD cells, cell counting kit-8 assayed cell viability, and Western blot analysis was conducted to determine the protein level of programmed death-ligand 1 (PD-L1). Coculture of LUAD cells with activated CD8+ T cells was carried out, and an LDH assay was used to assess the cytotoxicity of CD8+ T cells against LUAD cells. Interferon-γ (IFN-γ), interleukin-2 (IL-2), and tumor necrosis factor-α (TNF-α) levels in the coculture system were assessed by enzyme-linked immunosorbent assay (ELISA). Western blot assessed protein levels of JAK2, p-JAK2, STAT3, and p-STAT3. RESULTS Compared to normal tissues, AURKA and ETV4 were upregulated in tumor tissues, and AURKA presented a negative association with CD8+ T-cell immune infiltration but a positive association with PD-L1. qRT-PCR unveiled significantly upregulated mRNA of AURKA and ETV4 in LUAD cells compared to normal lung epithelial cells. Knockdown of AURKA significantly decreased cell viability and PD-L1 protein level in LUAD cells, enhanced cytotoxicity of CD8+ T cells against LUAD cells and IFN-γ, IL-2, and TNF-α expression, while overexpression of AURKA yielded opposite results. Furthermore, the knockdown of ETV4 could reverse the oncogenic characteristics of cells caused by AURKA overexpression. CONCLUSION Our study illustrated that ETV4/AURKA axis promoted PD-L1 expression, suppressed CD8+ T-cell activity, and mediated immune escape in LUAD by regulating the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ping Yang
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fujian Province, Fuzhou, China
| | - Shangxiang He
- Department of Medical Oncology, Shanghai Artemed Hospital, Shanghai, China
| | - Ling Ye
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fujian Province, Fuzhou, China
| | - Heng Weng
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fujian Province, Fuzhou, China
| |
Collapse
|
47
|
Ji L, Lou S, Fang Y, Wang X, Zhu W, Liang G, Lee K, Luo W, Zhuang Z. Patchouli Alcohol Protects the Heart against Diabetes-Related Cardiomyopathy through the JAK2/STAT3 Signaling Pathway. Pharmaceuticals (Basel) 2024; 17:631. [PMID: 38794201 PMCID: PMC11124524 DOI: 10.3390/ph17050631] [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: 02/08/2024] [Revised: 04/20/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) represents a common pathological state brought about by diabetes mellitus (DM). Patchouli alcohol (PatA) is known for its diverse advantageous effects, notably its anti-inflammatory properties and protective role against metabolic disorders. Despite this, the influence of PatA on DCM remains relatively unexplored. To explore the effect of PatA on diabetes-induced cardiac injury and dysfunction in mice, streptozotocin (STZ) was used to mimic type 1 diabetes in mice. Serological markers and echocardiography show that PatA treatment protects the heart against cardiomyopathy by controlling myocardial fibrosis but not by reducing hyperglycemia in diabetic mice. Discovery Studio 2017 software was used to perform reverse target screening of PatA, and we found that JAK2 may be a potential target of PatA. RNA-seq analysis of heart tissues revealed that PatA activity in the myocardium was primarily associated with the inflammatory fibrosis through the Janus tyrosine kinase 2 (JAK2)/signal transducer and activator of the transcription 3 (STAT3) pathway. In vitro, we also found that PatA alleviates high glucose (HG) + palmitic acid (PA)-induced fibrotic and inflammatory responses via inhibiting the JAK2/STAT3 signaling pathway in H9C2 cells. Our findings illustrate that PatA mitigates the effects of HG + PA- or STZ-induced cardiomyopathy by acting on the JAK2/STAT3 signaling pathway. These insights indicate that PatA could potentially serve as a therapeutic agent for DCM treatment.
Collapse
Affiliation(s)
- Lijun Ji
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea;
| | - Shuaijie Lou
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
| | - Yi Fang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
| | - Xu Wang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
| | - Weiwei Zhu
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
| | - Guang Liang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Kwangyoul Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea;
| | - Wu Luo
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Zaishou Zhuang
- The Affiliated Cangnan Hospital and Chemical Biology Research Center, Wenzhou Medical University, Wenzhou 325000, China; (L.J.); (S.L.); (Y.F.); (X.W.); (W.Z.); (G.L.)
| |
Collapse
|
48
|
Hendawi NY, Crane HL, Mehanna H, Bolt R, Lambert DW, Hunter KD. Fibroblasts from HPV-negative oropharynx squamous cell carcinomas stimulate the release of osteopontin from cancer cells via the release of IL-6. FRONTIERS IN ORAL HEALTH 2024; 5:1390081. [PMID: 38803348 PMCID: PMC11128591 DOI: 10.3389/froh.2024.1390081] [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: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction HPV-associated oropharyngeal squamous cell carcinoma (OPSCC) shows distinct biological and clinical behaviour when compared to HPV-negative OPSCC. The overall role of the tumour microenvironment (TME) in head and neck cancer progression and metastasis has been studied intensively, but differences in HPV-negative and HPV-positive OPSCCs are less understood. Objective To investigate the role of cancer-associated fibroblasts (CAFs) and the functional interactions of normal tonsil fibroblasts (NTFs) and OP CAFs with HPV+ and HPV- OPSCC cells and explore novel candidates in tumour-fibroblast crosstalk. Materials and methods A retrospective cohort of 143 primary OPSCCs was characterised using HPV16/18 RNAScope assay, p16 IHC and ɑ-SMA. Four OPSCC, three NTF and 2 new OPSCC CAF cultures were used to assess the cytokine-based interactions using cytokine arrays on conditioned media (CM), followed by co-culture approaches to identify the role of individual cell types and the role of OPN (SPP1) and IL-6 in SCC/fibroblast communication. Results HPV status was associated with better overall survival. Although ɑ-SMA expression was observed in both OPSCC subtypes, it provided survival stratification only in the HPV-positive group (Log-Rank p = 0.02). Three normal tonsillar fibroblast cultures (NTFs) were characterised by induction of myofibroblastic and senescent phenotypes with similar reactivity to our published NOF phenotype. The OPSCC-derived CAF cultures were characterised and their baseline myofibroblastic and senescence phenotypes varied. Cytokine array analysis of CM to identify novel candidates in the crosstalk between OPSCC tumour cells and NTFs/CAFs identified differences in the cytokine profiles on comparison of HPV+ and HPV- OPSCC cells. Osteopontin (OPN/SPP1) was identified, particularly in HPV-negative OPSCC cell analyses. We have demonstrated that OPN was produced by the OPSCC cells and revealed an associated upregulation of IL-6 in fibroblasts. Treatment of NTFs with rOPN showed alteration in phenotype, including increased contraction and IL-6 production. Antibody-mediated inhibition of CD44v6 attenuated the production of IL-6 by OPN in NTFs. Conclusion This investigation with OPSCC fibroblasts provides novel insights into the role of CAFs in OPSCC mediated by IL-6 stimulated release of OPN from HPV negative OPSCC cells. The details of HPV-positive SCC cell/fibroblast cytokine crosstalk remain elusive.
Collapse
Affiliation(s)
- Naeima Yahia Hendawi
- Academic Unit of Oral Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- Faculty of Dentistry, University of Benghazi, Benghazi, Libya
| | - Hannah L. Crane
- Academic Unit of Oral Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Hisham Mehanna
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Robert Bolt
- Academic Unit of Oral Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Daniel W. Lambert
- Academic Unit of Oral Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Keith D. Hunter
- Academic Unit of Oral Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
- Liverpool Head and Neck Centre, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
49
|
Yeo D, Yun YG, Shin SJ, Dashnyam K, Khurelbaatar A, Lee JH, Kim HW. Chaga mushroom extract suppresses oral cancer cell growth via inhibition of energy metabolism. Sci Rep 2024; 14:10616. [PMID: 38720012 PMCID: PMC11078932 DOI: 10.1038/s41598-024-61125-z] [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: 10/04/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
Oral cancer stands as a prevalent maligancy worldwide; however, its therapeutic potential is limited by undesired effects and complications. As a medicinal edible fungus, Chaga mushroom (Inonotus obliquus) exhibits anticancer effects across diverse cancers. Yet, the precise mechanisms underlying its efficacy remain unclear. We explored the detailed mechanisms underlying the anticancer action of Chaga mushroom extract in oral cancer cells (HSC-4). Following treatment with Chaga mushroom extracts, we analyzed cell viability, proliferation capacity, glycolysis, mitochondrial respiration, and apoptosis. Our findings revealed that the extract reduced cell viability and proliferation of HSC-4 cells while arresting their cell cycle via suppression of STAT3 activity. Regarding energy metabolism, Chaga mushroom extract inhibited glycolysis and mitochondrial membrane potential in HSC-4 cells, thereby triggering autophagy-mediated apoptotic cell death through activation of the p38 MAPK and NF-κB signaling pathways. Our results indicate that Chaga mushroom extract impedes oral cancer cell progression, by inhibiting cell cycle and proliferation, suppressing cancer cell energy metabolism, and promoting autophagy-mediated apoptotic cell death. These findings suggest that this extract is a promising supplementary medicine for the treatment of patients with oral cancer.
Collapse
Affiliation(s)
- Donghyeon Yeo
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119, Dandae-ro, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Yeo Gyun Yun
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119, Dandae-ro, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119, Dandae-ro, Cheonan, 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Khandmaa Dashnyam
- Drug Research Institute, Mongolian University of Pharmaceutical Science, Ulaanbaatar, 18130, Mongolia
| | - Anand Khurelbaatar
- Drug Research Institute, Mongolian University of Pharmaceutical Science, Ulaanbaatar, 18130, Mongolia
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119, Dandae-ro, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, 119, Dandae-ro, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science and BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
50
|
Panda SP, Kesharwani A, Datta S, Prasanth DSNBK, Panda SK, Guru A. JAK2/STAT3 as a new potential target to manage neurodegenerative diseases: An interactive review. Eur J Pharmacol 2024; 970:176490. [PMID: 38492876 DOI: 10.1016/j.ejphar.2024.176490] [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: 12/01/2023] [Revised: 02/06/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Neurodegenerative diseases (NDDs) are a collection of incapacitating disorders in which neuroinflammation and neuronal apoptosis are major pathological consequences due to oxidative stress. Neuroinflammation manifests in the impacted cerebral areas as a result of pro-inflammatory cytokines stimulating the Janus Kinase2 (JAK2)/Signal Transducers and Activators of Transcription3 (STAT3) pathway via neuronal cells. The pro-inflammatory cytokines bind to their respective receptor in the neuronal cells and allow activation of JAK2. Activated JAK2 phosphorylates tyrosines on the intracellular domains of the receptor which recruit the STAT3 transcription factor. The neuroinflammation issues are exacerbated by the active JAK2/STAT3 signaling pathway in conjunction with additional transcription factors like nuclear factor kappa B (NF-κB), and the mammalian target of rapamycin (mTOR). Neuronal apoptosis is a natural process made worse by persistent neuroinflammation and immunological responses via caspase-3 activation. The dysregulation of micro-RNA (miR) expression has been observed in the consequences of neuroinflammation and neuronal apoptosis. Neuroinflammation and neuronal apoptosis-associated gene amplification may be caused by dysregulated miR-mediated aberrant phosphorylation of JAK2/STAT3 signaling pathway components. Therefore, JAK2/STAT3 is an attractive therapeutic target for NDDs. Numerous synthetic and natural small molecules as JAK2/STAT3 inhibitors have therapeutic advances against a wide range of diseases, and many are now in human clinical studies. This review explored the interactive role of the JAK2/STAT3 signaling system with key pathological factors during the reinforcement of NDDs. Also, the clinical trial data provides reasoning evidence about the possible use of JAK2/STAT3 inhibitors to abate neuroinflammation and neuronal apoptosis in NDDs.
Collapse
Affiliation(s)
- Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Adarsh Kesharwani
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Samaresh Datta
- Department of Pharmaceutical Chemistry, Birbhum Pharmacy School, Sadaipur, Birbhum, West Bengal, India
| | - D S N B K Prasanth
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Polepally SEZ, TSIIC, Jadcherla, Mahbubnagar, Hyderabad, 509301, India
| | | | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|