1
|
Ren Z, Zheng Y, Liu J, Liu Z, Chen J, Liu H, Qi R, Ma H. Cadmium induces the secretion of SASP factors regulated by MAPK and NF-κB signaling pathways in HEK293 cells: A possible mechanism of acute kidney damage induced by cadmium. Toxicology 2025; 515:154166. [PMID: 40288561 DOI: 10.1016/j.tox.2025.154166] [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: 03/06/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Cadmium (Cd) is a highly toxic environmental pollutant, which can accumulate in the kidney, induce cell damage and trigger inflammatory responses. However, the specific regulation mechanism of nephrotoxicity induced by Cd remains unclear. This study was conducted to investigate the toxic effects of Cd on human embryonic kidney 293 (HEK293) cells and explore its potential mechanisms. Cell viability was assessed with MTT assay. Reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP) were evaluated through DCFH-DA staining and Rhodamine staining. Apoptosis was detected with Hoechst 33258 staining. The expression of the DNA damage biomarker 8-hydroxy-2'-deoxyguanosine (8-OHdG) was detected with the 8-OHdG ELISA kit. Senescence-associated secretory phenotype (SASP) factors and signaling pathways were analyzed by Western blot. The results showed that Cd exposure could induce oxidative stress and cellular inflammation. It could also impair MMP, contribute to cell apoptosis and activate MAPK and NF-κB signaling pathways. Finally, exposure to Cd triggered DNA damage and SASP production. However, NF-κB inhibitor BAY11-7082 and antioxidant NAC could inhibit these effects by suppressing NF-κB and MAPK signaling pathways. The present study revealed the specific mechanisms of Cd toxicity in HEK293 cells and provided useful information for elucidating the nephrotoxicity of Cd.
Collapse
Affiliation(s)
- Ziqi Ren
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Yuanchen Zheng
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Jianli Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China.
| | - Zhicun Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Jiahe Chen
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Haotian Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Ruiquan Qi
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang 110036, P.R. China
| | - Huiping Ma
- Shenyang Women's and Children's Hospital, Center of Reproductive Medicine, Shenyang 110011, P.R. China.
| |
Collapse
|
2
|
Li Y, Chen Y, Tang Y, Yang T, Zhou P, Miao L, Chen H, Deng Y. Breaking the barriers in effective and safe Toll-like receptor stimulation via nano-immunomodulators for potent cancer immunotherapy. J Control Release 2025; 382:113667. [PMID: 40157608 DOI: 10.1016/j.jconrel.2025.113667] [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: 12/02/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy is an emerging strategy that awakens the intrinsic immune system for cancer treatment. Generally, successful immunotherapy of malignant tumours relies on the effective production of tumour-associated antigens and their lymph node delivery, antigen processing and presentation for T-cell activation, and the dismantling of the immunosuppressive tumour microenvironment. Toll-like receptor (TLR) agonists are potent stimulants in cancer immunotherapy, which can directly activate antigen-presenting cells (APCs) and further induce T cell activation for antitumour immune response and convert immunosuppressive tumour microenvironment to an immunogenic one for cooperative tumour ablation. However, TLR agonists for effective cancer immunotherapy have encountered essential challenges, such as insufficient immune activation and systemic side effects. In recent years, nano-immunomodulators with TLR agonists have been employed for tumour- and/or lymph node-targeted immune activation to improve the antitumour immune response and alleviate their systemic toxicities, providing a promising strategy for enhanced cancer immunotherapy. Herein, we introduce the recent progress in developing various TLR nano-immunomodulators for cancer immunotherapy via APC activation and tumour microenvironment remodelling. Upon elucidating the rational design principles of nano-immunomodulators, we elucidate the advancement of TLR nanoagonists to break the barriers in effective and safe Toll-like receptor stimulation for potent cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoqi Li
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou 215006, China.
| | - Huabing Chen
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
3
|
Liu X, Huang L, Zhu Y, Wu P. Elemental comparative analysis of 18 elements reveal distinct patterns in benign and malignant thyroid tissues. Biometals 2025; 38:873-886. [PMID: 40299266 DOI: 10.1007/s10534-025-00682-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
This study aims to compare variations in 18 trace elements (Al, Na, K, Mg, Ca, Ti, Ba, Sn, Cr, Mn, Fe, Cu, Zn, Se, Mo, Cd, Sr, and Tl) between benign and malignant thyroid tissues. Post-operative thyroid tissue samples were collected from 106 patients (34 benign, 72 malignant), and elemental concentrations were quantified using inductively coupled plasma mass spectrometry. Spearman's correlation analysis revealed positive correlations among these trace elements. Notably, the malignant group exhibited significantly higher concentrations in eight elements (Mg, Al, Fe, Cr, Ti, Sr, Sn, and Ba) compared to the benign group, while levels of six elements (Na, Mn, Cu, Zn, Cd, and Mo) were significantly lower. Orthogonal partial least squares discriminant analysis distinguished three elements (Al, Ti, Sn) for the malignant group and six elements (Na, Mn, Cu, Zn, Cd, and Mo) for the benign group. Multivariate logistic regression further revealed associations between thyroid cancer and levels of Al, Cr, Ti, Sr, Sn, Ba, Mn, Cu, Zn, and Cd. Considering with each elemental biological funcions, these findings suggest that Cu, Mn, and particularly Zn may act as essential antitumor elements with synergistic effects, whereas elevated Ba, Cr, and Al levels are closely related to thyroid malignancies. However deficiencies and excesses of elements may be the consequences of malignant tissues. In conclusion, benign and malignant thyroid tumors exhibit different trace-element profiles.
Collapse
Affiliation(s)
- Xueying Liu
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, China
- Department of Endocrinology, Fuqing City Hospital, Fujian Medical University, Fuzhou, 350300, Fujian, China
| | - Linjing Huang
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, China
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Peiwen Wu
- Department of Endocrinology, the First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, Fujian, China.
- Department of Endocrinology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, Fujian, China.
- Clinical Research Center for Metabolic Diseases of Fujian Province, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
- Fujian Key Laboratory of Glycolipid and Bone Mineral Metabolism, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China.
| |
Collapse
|
4
|
Jayab NA, Abed A, Talaat IM, Hamoudi R. The molecular mechanism of NF-κB dysregulation across different subtypes of renal cell carcinoma. J Adv Res 2025; 72:501-514. [PMID: 39094893 DOI: 10.1016/j.jare.2024.07.030] [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: 04/17/2024] [Revised: 06/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The nuclear factor kappa B (NF-κB) is a critical pathway that regulates various cellular functions, including immune response, proliferation, growth, and apoptosis. Furthermore, this pathway is tightly regulated to ensure stability in the presence of immunogenic triggers or genotoxic stimuli. The lack of control of the NF-κB pathway can lead to the initiation of different diseases, mainly autoimmune diseases and cancer, including Renal cell carcinoma (RCC). RCC is the most common type of kidney cancer and is characterized by complex genetic composition and elusive molecular mechanisms. AIM OF REVIEW The current review summarizes the mechanism of NF-κB dysregulation in different subtypes of RCC and its impact on pathogenesis. KEY SCIENTIFIC CONCEPT OF REVIEW This review highlights the prominent role of NF-κB in RCC development and progression by driving the expression of multiple genes and interplaying with different pathways, including the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. In silico analysis of RCC cohorts and molecular studies have revealed that multiple NF-κB members and target genes are dysregulated. The dysregulation includes receptors such as TLR2, signal-transmitting members including RelA, and target genes, for instance, HIF-1α. The lack of effective regulatory mechanisms results in a constitutively active NF-κB pathway, which promotes cancer growth, migration, and survival. In this review, we comprehensively summarize the role of dysregulated NF-κB-related genes in the most common subtypes of RCC, including clear cell RCC (ccRCC), chromophobe RCC (chRCC), and papillary RCC (PRCC).
Collapse
Affiliation(s)
- Nour Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Alaa Abed
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, 21131 Alexandria, Egypt.
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Center of Excellence for Precision Medicine, Research Institute of Medical and Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272 Sharjah, United Arab Emirates; BIMAI-Lab, Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, 27272 Sharjah, United Arab Emirates; Division of Surgery and Interventional Science, University College London, London, United Kingdom; ASPIRE Precision Medicine Research Institute Abu Dhabi, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
5
|
Wang W, Liu M, Wu H, Li J, Lv W, Li C, Du C, Feng C, Zhang Y, Cai Y, Jia Y, Hu Y, Qu J, Zhang S, Wu F. The oncogenic role of TIMM8A in cancer and the mechanistic insights into the function in breast cancer cells. Sci Rep 2025; 15:18374. [PMID: 40419670 DOI: 10.1038/s41598-025-03331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 05/20/2025] [Indexed: 05/28/2025] Open
Abstract
The expression of TIMM8A, a molecular chaperone involved in mitochondrial protein translocation, was observed to be significantly elevated in various tumor types. However, the specific role of TIMM8A in cancer development and its underlying molecular mechanism remains inadequately understood. In this study, our primary objective was to investigate the functional implications of TIMM8A in breast cancer development, while also assessing its expression levels and prognostic relevance in pan-cancer. Notably, TIMM8A exhibited high expression in nearly 33 different cancer types, which was consistently associated with unfavorable clinical outcomes. In breast cancer, TIMM8A exhibited a strong association with prognosis and demonstrated its potential as an independent prognostic indicator. Additionally, the inhibition of TIMM8A effectively impeded the proliferation of MCF-7 and MDA-MB-231 cells, while also suppressing their migratory and invasive capabilities in vitro. Mechanistically, the downregulation of NF-κB p65, upregulation of pro-apoptotic proteins, and regulation of EMT-related proteins were observed upon TIMM8A knockdown. Furthermore, the over-expression of miR-10b-5p effectively hindered the expression of TIMM8A. Collectively, TIMM8A, a critical oncogene, was regulated by miR-10b-5p and could activate NF-κB signaling cascade response, promote apoptosis inhibition and regulate EMT-related protein expression, thereby stimulating proliferation, migration, and invasion of breast cancer cells.
Collapse
Affiliation(s)
- Weiwei Wang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Mengjie Liu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Huizi Wu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Jia Li
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Wei Lv
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Chaofan Li
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Chong Du
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Cong Feng
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Yu Zhang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Yifan Cai
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Yiwei Jia
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Yijian Hu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Jingkun Qu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China
| | - Shuqun Zhang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China.
| | - Fei Wu
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, West 5Th Road 157, 710004, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
6
|
Roy A, Roy R, Bhattacharya P, Borah A. The Vicious Consequences of Chronic Kidney Disease on Cognitive Impairment and Alzheimer's Disease. ACS Chem Neurosci 2025; 16:1847-1859. [PMID: 40340356 DOI: 10.1021/acschemneuro.5c00050] [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] [Indexed: 05/10/2025] Open
Abstract
Chronic kidney disease (CKD) and Alzheimer's disease (AD) are two prevalent and debilitating conditions that frequently coexist, with CKD contributing to cognitive decline and potentially exacerbating AD pathology. In CKD, irreversible changes in the structure or function of the kidneys are observed, while AD is primarily marked by amyloid deposition and tau pathology. Both conditions involve complex and multifactorial pathophysiology affecting brain functioning, highlighting the need for comprehensive research to understand their potential crosstalk. This review articulates the possible molecular mechanisms underlying both diseases, focusing on key pathways, including oxidative stress, inflammation, vascular dysfunction, hypertension, and uremic toxin accumulation. These interconnected mechanisms suggest a potential bidirectional relationship where kidney dysfunction accelerates cognitive decline and vice versa. Additionally, we examine critical risk factors implicated in both CKD and AD, for instance, vitamin D deficiency, erythropoietin dysregulation, endothelin action, klotho gene expression, and the role of the extracellular vesicle, which may influence disease progression through their effects on the kidney and brain, influencing cognitive function. Further, we emphasized potential biomarkers that could aid in diagnosing and monitoring disease progression in these comorbid conditions, like amyloid beta, tau, homocysteine, cystatin C, creatinine, proteinuria, and estimated glomerular filtration rate. Lastly, the review highlights treatment strategies for managing CKD and AD concurrently, focusing on therapeutic approaches that address common pathophysiological mechanisms. These strategies not only aim to address the underlying causes of both conditions but also offer the potential to slow or even prevent the progression of cognitive impairment. Moreover, we recommend further research to refine these approaches, execute correlational studies on disease progression, and design clinical trials that address both conditions, aiming to establish effective, tailored treatments for this dual burden of disease.
Collapse
Affiliation(s)
- Abhideep Roy
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011, Assam, India
| | - Rubina Roy
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar 382355, Gujarat, India
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011, Assam, India
| |
Collapse
|
7
|
Khan S, Anwer A, Sevak JK, Abbas Z, Fatima K, Islam M, Jehan T, Trehanpati N, Kazim SN. Expression variation of Akt and cytokines in lamivudine-resistant HBV rare mutation that prematurely truncates the overlapping surface protein. Hum Immunol 2025; 86:111328. [PMID: 40382902 DOI: 10.1016/j.humimm.2025.111328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/24/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025]
Abstract
The sW196*(Stop) mutation of HBV is the rarest variant of sW196L. It is associated with rapid progression of chronic hepatitis B to end-stage liver disease and hepatocellular carcinoma. Information on proliferative vs. apoptotic cells and immunological manifestations related to the expression of HBV-associated cytokines in the presence of the lamivudine-resistant mutation sW196L and the rarest variant of sW196L, sW196*(Stop), has been limited. Therefore, the current investigation aimed to examine the molecular and immunological basis of lamivudine-resistant mutations with the help of transfection studies carried out in cell lines of hepatic origin. sW196*(Stop) transfected HepG2 cells showed IFN-gamma was extremely downregulated by 20.96 folds, and IL-6 was upregulated by 4.5 folds which had not been seen in any of the constructs. The current study reveals for the first time the underlying immunological processes that render detrimental effects to lamivudine-resistant rare mutations, in particular sW196*(Stop) of HBV that can be explained by downregulation of IFN-gamma, a known cytokine associated with efficient viral clearance, together with upregulation of IL-6 known to be associated with disease progression and HCC. Understanding the molecular and immunological mechanisms in such HBV mutations is furthered by the choreographed event of increased TNF-alpha expression, which is known to be linked to liver damage.
Collapse
Affiliation(s)
- Saniya Khan
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India
| | - Ayesha Anwer
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India
| | - Jayesh Kumar Sevak
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India; Institute of Liver and Biliary Sciences, New Delhi, India
| | - Zahra Abbas
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India
| | - Kaneez Fatima
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India
| | - Mahvish Islam
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India
| | - Tabassum Jehan
- Zakir Husain Delhi College, University of Delhi, New Delhi, India
| | | | - Syed Naqui Kazim
- Centre for Interdisciplinary Research in Basic Sciences, New Delhi, India.
| |
Collapse
|
8
|
Carballo-López GI, Ojeda-González J, Martínez-García KD, Cervantes-Luevano KE, Moreno-Ulloa A, Castro-Ceseña AB. Enhanced anti-inflammatory and anti-fibrotic effects of nanoparticles loaded with a combination of Aloe vera- Moringa oleifera extracts. Mol Omics 2025; 21:185-201. [PMID: 39878065 DOI: 10.1039/d4mo00195h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Metabolic associated steatohepatitis characterized by lipid accumulation, inflammation and fibrosis, is a growing global health issue, contributing to severe liver-related mortality. With limited effective treatments available, there is an urgent need for novel therapeutic strategies. Moringa oleifera, rich in antioxidants, offers potential for combating steatohepatitis, but its cytotoxicity presents challenges. Aloe vera, renowned for its cytocompatibility and anti-inflammatory effects, shows promise in mitigating these risks. Using infrared spectrometry and mass spectrometry, we identified 1586 metabolites from both plants across 84 chemical classes. By encapsulating these phytochemicals in nanoparticles, we achieved increased solubility, cytocompatibility, and gene modulation to hepatic stellate cells affected by steatohepatitis. Chemoinformatic analysis revealed bioactive metabolites, including hesperetin analogs, known to inhibit TGF-β. Our results demonstrate that these nanoparticles not only improved gene expression modulation related to metabolic associated steatohepatitis, particularly TGF-β and COL1A1, but also outperformed free compounds, highlighting their potential as a novel therapeutic approach.
Collapse
Affiliation(s)
- Gabriela I Carballo-López
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| | - Jhordan Ojeda-González
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| | - Kevin D Martínez-García
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| | - Karla E Cervantes-Luevano
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| | - Aldo Moreno-Ulloa
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
| | - Ana B Castro-Ceseña
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico.
- CONAHCYT - Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada, Baja California (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, C.P. 22860, Ensenada, Baja California, Mexico
| |
Collapse
|
9
|
Yao Y, Yang X, Fu Y, Zhang Y. Immunological features of various molecular subtypes of cervical cancer and their prognostic implications in the context of disulfidptosis. Front Oncol 2025; 15:1574911. [PMID: 40438679 PMCID: PMC12116334 DOI: 10.3389/fonc.2025.1574911] [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: 02/11/2025] [Accepted: 04/25/2025] [Indexed: 06/01/2025] Open
Abstract
Objective Cervical cancer ranks among the most prevalent malignancies impacting women globally. Disulfidptosis represents a recently identified pathway of cellular demise, although its role in the context of cervical cancer is not well elucidated. This research investigates the significance of Disulfidptosis-Related Genes (DRGs) within cervical cancer. Furthermore, it aims to analyze the differences in prognosis and immune infiltration among different molecular subtypes. Methods We compiled genes associated with cervical cancer and disulfidptosis from a variety of databases to perform a differential expression analysis. Subsequently, the samples are grouped through consensus clustering. To evaluate immune cell infiltration, we employed CIBERSORT. Additionally, immune checkpoint genes (ICGs) were gathered from existing literature and databases, enabling statistical analyses of two subtype samples of cervical cancer (CESC). Following our analyses using GO, KEGG, and GSEA to compare the differences between the two subtypes. Lastly, a prognostic risk model was constructed using LASSO regression and validated using ROC. Results This study identified seven key genes: PCBP3, ARNT, ANP32E, DSTN, CD2AP, EPAS1, and ACTN1.The consensus clustering analysis showed differences in immune cell infiltration and DFS(disease-free survival) among the various clusters. The immune checkpoint gene CXCL1 displayed highly significant statistical differences between subtype A (Cluster 1) and subtype B (Cluster 2) in cervical cancer (CESC) samples. The gene set enrichment analysis identified the negative regulation of peptidase activity and the IL-17 signaling pathway, which link to subtype-specific differentially expressed genes (DEGs). Conclusion Statistical analysis of the various subtypes of CESC samples highlighted the importance of subtype-specific therapeutic targets. Additionally, it seeks to enhance the accuracy of prognostic predictions, thereby establishing a foundation for the formulation of personalized treatment approaches.
Collapse
Affiliation(s)
- Yadan Yao
- Department of Gynecology, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Xiaomin Yang
- Department of Gynecology, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Yuanxin Fu
- Department of Acupuncture and Massage, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| | - Yinmin Zhang
- Department of Pediatrics, Jiaxing University Affiliated Traditional Chinese Medicine (TCM) Hospital, Jiaxing, Zhejiang, China
| |
Collapse
|
10
|
Dong S, Chen Y, Li Y, Liu X, Yan J, Xie M, Wu F, Niu M, Shang F, Huang H, Wu W, Guo S, Du Y, Hua M, Song C. Estradiol conjugation to estrogen receptorα upregulates Brms1 expression mediating M2 polarization of alveolar macrophages and exacerbating airway inflammation in asthmatic mice. Mol Immunol 2025; 181:84-92. [PMID: 40106959 DOI: 10.1016/j.molimm.2025.03.002] [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: 12/14/2024] [Revised: 02/22/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Asthma is a common condition involving chronic airway inflammation that primarily affects women and boys. Estrogen levels correlate with the observed differences in the prevalence of asthma between the sexes, but the exact mechanism is unclear. This study established a castration mice (OVX) model through bilateral ovariectomy surgery, and subcutaneously injected estradiol (E2) into OVX asthmatic mice to analyze the effect of E2 on the onset of asthma. Then, airway inflammation was evaluated in the mice using airway resistance measurements, lung tissue hematoxylin and eosin staining, and eosinophil counts. Furthermore, the proportion of CD206-positive cells and the expression of M2 polarization markers, such as Arg1 and YM1, were detected in alveolar macrophages (AMs). The effects of different concentrations of E2 on M2 polarization of AMs were examined in vitro, and the types of estrogen receptors (ERs) involved were investigated. Transcriptome analysis combined with volcano plots and heatmaps were used to compare the differentially expressed genes to investigate the mechanism by which E2 affects M2 polarization of AMs. The results showed that female asthmatic mice had more severe airway inflammation and higher airway responsiveness than male asthmatic mice. E2 increased airway inflammation and airway resistance in asthmatic mice. E2 not only promoted M2 polarization of AMs in asthmatic mice in vivo, but also increased the expression of M2 markers, such as Arg1 and YM1, by AMs in vitro. The use of ERα antagonist AZD9496 reduced the effect of E2 on the promotion of M2 polarization in AMs. Analysis of transcriptome differences indicated that E2 upregulated expression of M2 breast cancer metastasis suppressor gene 1 (Brms1) in AMs. Notably, antagonism of ERα inhibited this upregulation of Brms1 gene expression. Interference with Brms1 mRNA production reduced the gene expression of Arg1 and YM1 in AMs undergoing M2 polarization after E2 stimulation. In summary, E2 exacerbates airway inflammation in asthmatic mice and binds to ERα, upregulating Brms1 expression and mediating M2 polarization of AMs.
Collapse
Affiliation(s)
- Shu Dong
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Yingzi Chen
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Ya Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Xingyue Liu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Jiaqi Yan
- School of Clinical Medicine, Bengbu Medical University, Anhui 233030, China
| | - Minyu Xie
- School of Clinical Medicine, Bengbu Medical University, Anhui 233030, China
| | - Fan Wu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Minzhu Niu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Feifei Shang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Han Huang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Wenwen Wu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Shujun Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Yulin Du
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China
| | - Mengqing Hua
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China.
| | - Chuanwang Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Anhui 233030, China.
| |
Collapse
|
11
|
Chang W, Feng K, Zhou P, Gong D, Wang K, Huang A, Wang K, Tang N. SPOP Suppresses Hepatocellular Carcinoma Growth and Metastasis by Ubiquitination and Proteasomal Degradation of TRAF6. Cancer Sci 2025; 116:1295-1307. [PMID: 39962908 PMCID: PMC12044664 DOI: 10.1111/cas.70025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor necrosis factor receptor-associated factor-6 (TRAF6) is a well-established upstream regulator of the IKK complex, essential for the modulation of the NF-κB (nuclear factor kappa B) signaling pathway. Aberrant activation of TRAF6 has been strongly implicated in the pathogenesis of various cancers, including hepatocellular carcinoma (HCC). The speckle type BTB/POZ protein (SPOP), an E3 ubiquitin ligase substrate-binding adapter, constitutes a significant component of the CUL3/SPOP/RBX1 complex, which is closely linked to tumorigenesis. In this study, we demonstrated that the E3 ubiquitin ligase SPOP shielded TRAF6 from proteasomal degradation, leading to the hyperactivation of the NF-κB pathway. Notably, a liver cancer-associated S119N mutation in SPOP resulted in a failure to mediate the ubiquitination and subsequent degradation of TRAF6. Moreover, both gain-of-function and loss-of-function experiments revealed that SPOP inhibits the proliferation and invasion of HCC cells through the TRAF6-NF-κB axis in vitro and in vivo. Taken together, our findings elucidate the underpinning mechanism by which SPOP negatively regulates the stability of the TRAF6 oncoprotein, thus offering a new therapeutic target for HCC intervention.
Collapse
Affiliation(s)
- Wenyi Chang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Kaiying Feng
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Peng Zhou
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Deao Gong
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Ke Wang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Kai Wang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| | - Ni Tang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital Chongqing Medical UniversityChongqingChina
| |
Collapse
|
12
|
Sui Q, Zhu C, Shi S, Xu J, Zhang J, Wang A, Chen P, Liang G, Zhang Y. Ganoderic acid A: an in-depth review of pharmacological effects and molecular docking analysis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119868. [PMID: 40316150 DOI: 10.1016/j.jep.2025.119868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ganoderic acid A (GAA, C30H44O7) is one of the most abundant and active components of Ganoderic acids (GAs). GAs are highly oxidized tetracyclic triterpenoid compounds mainly derived from Ganoderma lucidum (Curtis) P. Karst (Chinese: ). GAA is primarily isolated from the fruiting body of Ganoderma lucidum. Modern pharmacological investigations have established the broad pharmacological effects of GAA, highlighting its notable influence on managing various conditions, including inflammatory diseases, neurodegenerative diseases, and cancer. This review provides a comprehensive summary of GAA's pharmacological activities. MATERIAL AND METHODS The literature in this review were searched in PubMed and China National Knowledge Infrastructure (CNKI) using the keywords "Ganoderic acid A″, "Pharmacology" and "Pharmacokinetics". The literature cited in this review dates from 2000 to 2024. RESULTS According to the data, GAA exerts anti-inflammatory, antioxidant, antitumor, neuropsychopharmacological, hepatoprotective, cardiovascular, renoprotective, and lung protective effects by regulating a variety of signal transduction pathways, such as nuclear factor kappa-B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), Toll-like receptor 4 (TLR4), nuclear factor erythroid 2-related factor-2 (Nrf2), phosphoinositide-3-kinase (PI3K)/AKT, mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK), and Notch. Given its promising pharmacological activity, GAA holds excellent potential for treating human diseases. The pharmacokinetic properties of GAA have also been reviewed, revealing low bioavailability but high absorption and elimination rates. In addition, network pharmacology and molecular docking analyses verified that GAA plays a role in multiple diseases through MAPK3, tumor necrosis factor (TNF), caspase-3 (CASP3), peroxisome proliferator-activated receptor gamma (PPARG), and β-catenin (CTNNB1) signaling pathways. CONCLUSION GAA plays a pivotal role in various pathological and physiological processes, boasting broad application prospects. Furthermore, the network pharmacological results reveal the mechanisms of GAA in the treatment of multiple diseases. In the future, it is necessary to conduct further experiments to elucidate its specific mechanism of action, thus laying the foundation for the scientific utilization of GAA.
Collapse
Affiliation(s)
- Qi Sui
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Chengkai Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Sha Shi
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jiaqi Xu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jingnan Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Ao Wang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Department of Pharmacy, School of Medicine, Hangzhou City University, 50 Huzhou Rd, Hangzhou, Zhejiang, 310015, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Peng Chen
- Department of Pharmacy, School of Medicine, Hangzhou City University, 50 Huzhou Rd, Hangzhou, Zhejiang, 310015, China.
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China.
| |
Collapse
|
13
|
Lee S, Ho YY, Hao S, Ouyang Y, Liew UL, Goyal A, Li S, Barbour JA, He M, Huang Y, Wong JWH. A tumour necrosis factor-α responsive cryptic promoter drives overexpression of the human endogenous retrovirus ERVK-7. J Biol Chem 2025:108568. [PMID: 40316021 DOI: 10.1016/j.jbc.2025.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/09/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025] Open
Abstract
Endogenous retroviruses (ERVs) shape human genome functionality and influence disease pathogenesis, including cancer. ERVK-7, a significant ERV, acts as an immune modulator and prognostic marker in lung adenocarcinoma (LUAD). Although ERVK-7 overexpression has been linked to the amplification of the 1q22 locus in approximately 10% of LUAD cases, it predominantly arises from alternative regulatory mechanisms. Our findings indicate that the canonical 5' long terminal repeat (LTR) of ERVK-7 is methylated and inactive, necessitating the use of alternative upstream promoters. We identified two novel transcripts, ERVK-7.long and ERVK-7.short, arising from distinct promoters located 2.8 kb and 13.8 kb upstream of the 5'LTR of ERVK-7, respectively. ERVK-7.long is predominantly overexpressed in LUAD. Through comprehensive epigenetic mapping and single-cell transcriptomics, we demonstrate that ERVK-7.long activation is predetermined by cell lineage, specifically in small airway epithelial cells (SAECs), where its promoter displays tumor-specific H3K4me3 modifications. Single-cell RNA sequencing further reveals a distinct enrichment of ERVK-7.long in LUAD tumor cells and alveolar type 2 epithelial cells, underscoring a cell-type-specific origin. Additionally, inflammatory signaling significantly influences ERVK-7 expression; TNF-α enhances ERVK-7.long, while interferon signaling preferentially augments ERVK-7.short by differential recruitment of NF-κB/RELA and IRF to their respective promoters. This differential regulation clarifies the elevated ERVK-7 expression in LUAD compared to lung squamous cell carcinoma (LUSC). Our study elucidates the complex regulatory mechanisms governing ERVK-7 in LUAD and proposes these transcripts as potential biomarkers and therapeutic targets, offering new avenues to improve patient outcomes.
Collapse
Affiliation(s)
- Sojung Lee
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yin Yee Ho
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Suyu Hao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yingqi Ouyang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - U Ling Liew
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Ashish Goyal
- Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jayne A Barbour
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Mu He
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuanhua Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jason W H Wong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China; Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
14
|
Li X, Wang Z, Chen Y, Yang Y, Shao H, Feng X, Ren Y. Regulation of monocyte polarization through nuclear factor Kappa B /inhibitor of Kappa B Alpha pathway by Cuscuta chinensis Lam. In postmenopausal osteoporosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119710. [PMID: 40154899 DOI: 10.1016/j.jep.2025.119710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cuscuta chinensis Lam. (CCL), is made from the dried mature seeds of a plant in the Convolvulaceae family. Predominantly distributed in China and several Asian countries, it has long been used to treat osteoporosis (OP) and other aging-related diseases. However, studies on the mechanisms of anti-OP compounds in CCL remain limited. AIM OF THE STUDY The objective of this study is to determine the bioactive constituents present in CCL and to elucidate their mechanisms of action in the prevention and treatment of postmenopausal osteoporosis (PMOP). This will be achieved by investigating the modulation of bone marrow macrophage polarization via the Nuclear Factor Kappa B (NF-κB)/Inhibitor of Kappa B Alpha (IκBα) signaling cascade. MATERIALS AND METHODS CCL's chemical components were identified using UPLC-Q-TOF-MS. Blood components were analyzed for targets using databases. Pathway enrichment was performed via network pharmacology. We used an ovariectomy (OVX)-induced OP rat model to assess the effects of CCL extracts in comparison to a positive control drug. Osteogenic markers were analyzed. We utilized flow cytometry to assess macrophage marker expression, while quantitative PCR (qPCR) and Western blotting were employed to identify targets within the signaling pathways. RESULTS Seventeen chemical components were identified in CCL extracts, of which 14 were identified as prototype compounds absorbed into the bloodstream. Pathway enrichment analysis revealed that CCL's therapeutic effects on PMOP were closely associated with the NF-κB signaling pathway, specifically targeting NF-κB and IκBα proteins. Animal studies showed that high-dose CCL significantly lowered serum levels of tartrate-resistant acid phosphatase (TRACP) and c-terminal telopeptide of type I collagen (CTX) (p < 0.01) and increased levels of bone-specific alkaline phosphatase (BALP) and procollagen I c-terminal propeptide (PICP) (p < 0.01), indicating effective inhibition of bone resorption and promotion of bone formation. CCL treatment improved the microstructure of trabecular bone at the distal femur by reducing bone cavity spaces, increasing trabecular thickness, and enhancing trabecular alignment. CCL markedly enhanced the expression levels of osteoprotegerin (OPG), runt-related transcription factor 2 (Runx2), and alkaline phosphatase (ALP) genes and their corresponding proteins in the tibial tissue (p < 0.01), promoting osteoblast differentiation and function. Flow cytometry analysis showed that CCL modulated immune cell markers CD86 and CD163, supporting its anti-inflammatory effects in PMOP treatment. Furthermore, CCL regulated the NF-κB/IκBα signaling pathway by significantly decreasing NF-κB expression and increasing IκBα expression, thereby modulating inflammatory responses and bone metabolism. CONCLUSION The active components in CCL effectively prevent and treat PMOP by modulating bone marrow macrophage polarization through the NF-κB/IκBα signaling pathway.
Collapse
Affiliation(s)
- Xiaochen Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| | - Zhimin Wang
- Liaoning University of Traditional Chinese Medicine Affiliated Hospital, Shenyang, 110000, China.
| | - Yiran Chen
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| | - Ying Yang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| | - Hanrui Shao
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| | - Xiuzhi Feng
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| | - Yanling Ren
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| |
Collapse
|
15
|
Binan L, Jiang A, Danquah SA, Valakh V, Simonton B, Bezney J, Manguso RT, Yates KB, Nehme R, Cleary B, Farhi SL. Simultaneous CRISPR screening and spatial transcriptomics reveal intracellular, intercellular, and functional transcriptional circuits. Cell 2025; 188:2141-2158.e18. [PMID: 40081369 DOI: 10.1016/j.cell.2025.02.012] [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: 11/30/2023] [Revised: 10/24/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025]
Abstract
Pooled optical screens have enabled the study of cellular interactions, morphology, or dynamics at massive scale, but they have not yet leveraged the power of highly plexed single-cell resolved transcriptomic readouts to inform molecular pathways. Here, we present a combination of imaging spatial transcriptomics with parallel optical detection of in situ amplified guide RNAs (Perturb-FISH). Perturb-FISH recovers intracellular effects that are consistent with single-cell RNA-sequencing-based readouts of perturbation effects (Perturb-seq) in a screen of lipopolysaccharide response in cultured monocytes, and it uncovers intercellular and density-dependent regulation of the innate immune response. Similarly, in three-dimensional xenograft models, Perturb-FISH identifies tumor-immune interactions altered by genetic knockout. When paired with a functional readout in a separate screen of autism spectrum disorder risk genes in human-induced pluripotent stem cell (hIPSC) astrocytes, Perturb-FISH shows common calcium activity phenotypes and their associated genetic interactions and dysregulated molecular pathways. Perturb-FISH is thus a general method for studying the genetic and molecular associations of spatial and functional biology at single-cell resolution.
Collapse
Affiliation(s)
- Loϊc Binan
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aiping Jiang
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Serwah A Danquah
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vera Valakh
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brooke Simonton
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jon Bezney
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Robert T Manguso
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen B Yates
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA 02215, USA; Department of Biology, Boston University, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA.
| | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
16
|
Wu S, Luo Y, Wei F, Li Y, Fan J, Chen Y, Zhang W, Li X, Xu Y, Chen Z, Xia C, Hu M, Li P, Gu Q. Lactic acid bacteria target NF-κB signaling to alleviate gastric inflammation. Food Funct 2025; 16:3101-3119. [PMID: 40152095 DOI: 10.1039/d4fo06308b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Helicobacter pylori (H. pylori) infection and the resulting gastric inflammation are major contributors to gastric cancer development. Probiotics, particularly Lactobacillus, are promising for their anti-inflammatory potential, yet their exact mechanisms in inhibiting H. pylori-induced inflammation are unclear. In our previous study, Lactiplantibacillus plantarum ZJ316 (L. plantarum ZJ316) demonstrated strong anti-inflammatory effects against H. pylori infection in vivo, but its precise mechanisms were not fully understood. Here, we aimed to investigate how L. plantarum ZJ316 inhibits the inflammatory response to H. pylori infection. Our results demonstrated that L. plantarum ZJ316 effectively reduced the expression of pro-inflammatory cytokines in H. pylori-infected AGS cells. Mechanistically, L. plantarum ZJ316 inhibited the NF-κB signaling pathway by preventing the degradation of IκBα, suppressing p65 phosphorylation, and blocking the nuclear translocation of phosphorylated p65. Treatment with the NF-κB inhibitor BAY 11-7082 further decreased tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and interleukin-1β (IL-1β) levels, confirming the inhibitory effect of L. plantarum ZJ316 on the NF-κB pathway. In H. pylori-infected mice, oral administration of L. plantarum ZJ316 significantly alleviated inflammatory cell infiltration, reduced TNF-α and pepsinogen II (PGII) levels, and increased interleukin-10 (IL-10) levels in serum. A comparative metagenomic analysis of the gastric microbiota revealed a decrease in Prevotella and Desulfovibrio, alongside an increase in Ligilactobacillus and Akkermansia, supporting the protective effects of L. plantarum ZJ316 and correlating with their decreased inflammatory response. In summary, administration of L. plantarum ZJ316 demonstrated robust anti-inflammatory effects against H. pylori infection by suppressing NF-κB signaling and promoting favorable changes in the gastric microbiota composition. Therefore, L. plantarum ZJ316 holds promise as a novel functional food for protecting the body against H. pylori infection.
Collapse
Affiliation(s)
- Shiying Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yuenuo Luo
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Fangtong Wei
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Jiayi Fan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Yongqiang Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Wenjie Zhang
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Xuelong Li
- Hangzhou Helixinjian Industry Co., Ltd, No. 48 Zijinghua Road, Gudang Street, Xihu District, Hangzhou, Zhejiang 310050, China
| | - Yang Xu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ziqi Chen
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Chenlan Xia
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Mingyang Hu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
17
|
Canak HN, Bas K, Yağmur EA, Karakurt S. Mesobuthus eupeus venom modulates colorectal carcinoma signaling pathways and induces apoptosis. Med Oncol 2025; 42:163. [PMID: 40229568 PMCID: PMC11996983 DOI: 10.1007/s12032-025-02689-2] [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: 01/13/2025] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Colorectal cancer (CRC) is a significant global health concern, often challenging to treat effectively with conventional methods and burdened by adverse effects. Scorpion venoms offer a unique avenue for exploration, given their ability to disrupt the cell cycle, inhibit growth, and trigger apoptosis. This study delves into the impact of Mesobuthus eupeus (M. eupeus) scorpion venom on the proliferation and progression of colorectal cancer at the molecular level. The total protein concentration in the venom (607.5 µg/mL) also emphasized the rich composition and potential for therapeutic applications. The study reveals that M. eupeus venom effectively reduced the proliferation of DLD-1 and HT-29 colorectal cancer cells in a dose-dependent manner with IC50 values of 4.32 and 7.61 µg/mL, respectively. The venom also impedes cell migration, diminishes colony formation, and triggers apoptosis in the cancer cells. The venom also induced early and late apoptosis in the two cancer cell lines. The human colorectal cancer and apoptotic pathways were clarified at the molecular level using pathway panels, which revealed that 16 genes involved in colorectal cancer increased while 23 decreased. In the HT-29 cell line, 57 genes increased, and 1 decreased following venom treatment. Besides, the mRNA expression of 19 genes involved in the apoptotic pathway was increased, while 22 were reduced in DLD-1 cells. This study underscores the potential of M. eupeus venom as a natural therapeutic approach in the quest for cancer treatments.
Collapse
Affiliation(s)
- Havva Nur Canak
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye
| | - Kemal Bas
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye
| | - Ersen Aydın Yağmur
- Department of Plant and Animal Production, Alasehir Vocational High School, Manisa Celal Bayar University, Manisa, Türkiye
| | - Serdar Karakurt
- Faculty of Science, Department of Biochemistry, Selcuk University, Konya, Türkiye.
| |
Collapse
|
18
|
Zhao B, Wu J, Zhang T, Han M, Zhang C, Rong X, Zhang R, Chen X, Peng F, Jin J, Liu S, Dong X, Zhao S. A spatial transcriptomics study of MES-like and mono/macro cells in gliomas. Sci Rep 2025; 15:12730. [PMID: 40222970 PMCID: PMC11994772 DOI: 10.1038/s41598-025-95277-3] [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: 11/15/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Gliomas, including both glioblastoma multiforme (GBM) and lower-grade glioma (LGG), present a substantial challenge in neuro-oncology because of genetic heterogeneity and unsatisfactory prognosis. This study aimed to conduct a comprehensive multi-omics analysis of gliomas using various bioinformatics approaches to identify potential therapeutic targets and prognostic markers. A comprehensive analysis was conducted on 1327 sequencing data samples alongside their relevant clinical information sourced from The Cancer Genome Atlas (TCGA) pertaining to glioblastoma (GBM), low-grade glioma (LGG), the Chinese Glioma Genome Atlas (CCGA) and University of California Santa Cruz Xena (UCSC Xena) datasets. These tools were employed for gene expression profiling, survival analysis, and cell communication mapping. Spatial transcriptomics revealed the localization of mesenchymal (MES)-like malignant tumors, and drug sensitivity analysis was performed to evaluate responses to quinpirole and meropenem. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) framework was utilized to gauge the responsiveness to immunotherapy. The MES-like malignant and monocyte/macrophage (mono/macro) cell subsets showed high hallmark scores, playing key roles in the tumor microenvironment. MES-like malignant marker gene scores correlated with overall survival across datasets, whereas mono/macro marker gene scores were significant in the TCGA-LGG and CCGA datasets. Key interactions between these cell types were found, especially with CD14-ITGB2, LGALS1-CD69, and APOE-TREM2. The mono/macro cell subset demonstrated better immune therapy responsiveness, as indicated by lower TIDE scores. Spatial transcriptomics revealed that MES-like malignant tumors are predominantly localized in four distinct regions, with the marker genes CHI3L1 and ADM confirming these locations. Drug sensitivity analysis revealed differential responses of the MES-like malignant cell subset to quinpirole and meropenem. Our results offer fresh perspectives on the differential roles of MES-like malignant and monocyte/macrophage cell subsets in tumor progression and immune modulation, providing novel insights into glioma biology.
Collapse
Affiliation(s)
- Boyan Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Tiehui Zhang
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong, China
| | - Mingyang Han
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Cheng Zhang
- University of Toronto Scarborough 1265 Military Trail, Scarborough, ON, M1C 1A4, Canada
| | - Xuan Rong
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Ruotian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Key Laboratory of Neurosurgery of Colleges and Universities in Heilongjiang Province, Harbin, 150001, Heilongjiang, China
| | - Fei Peng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jin Jin
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Shiya Liu
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China
| | - Xingli Dong
- Central Laboratory, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China.
- Shenzhen University School of Medicine, Shenzhen, 518000, Guangdong, China.
- Department of Neurosurgery, Shenzhen University General Hospital, 1088 Xueyuan Avenue, Nanshan District, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
19
|
Popovici IA, Orasanu CI, Cozaru GC, Ionescu AC, Kajanto L, Cimpineanu B, Chisoi A, Mitroi AN, Poinareanu I, Voda RI, Ursica OA, Pundiche MB. An Overview of the Etiopathogenic Mechanisms Involved in the Expression of the Oral Microbiota. Clin Pract 2025; 15:80. [PMID: 40310312 PMCID: PMC12026067 DOI: 10.3390/clinpract15040080] [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: 02/13/2025] [Revised: 03/17/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Background/Objectives: The diversity of the oral microbiota exerts its effects in maintaining dental and overall health. The unique genetic profile of each individual influences the composition of the oral microbiota, determining susceptibility to certain diseases. The aim is to observe its role by highlighting the pathogenic mechanisms involved in oral dysbiosis and identify genetic determinism's influence in maintaining balance. Methods: This study was designed as a narrative review of the oral microbiota, utilizing some of the principles and guidelines of systematic review to increase methodological rigor. We examined 121 articles such as reviews, meta-analyses, editorials, and observational studies, which met the inclusion and exclusion criteria. The inclusion criteria for studies were as follows: (1) studies that evaluated the impact of the microbiota in oral or/and systemic diseases; (2) studies that observed pathogenic mechanisms in the oral microbiota; (3) studies that evaluated the interaction of the microbiota with the immune system (4); studies that evaluated genetic implications in the microbiota. Results: Host genes regulate inflammatory and immunological reactions that play a role in microbiological balance. This explains the increased resistance of some to diseases, including gingivitis or periodontitis. Also, the implications of oral dysbiosis are reflected not only locally, but also generally, being associated with various systemic conditions. Conclusions: Understanding the pathogenic mechanisms and genetic determinants involved in oral dysbiosis may help create individualized therapies for preventing and managing oral and systemic disorders. A healthy lifestyle and adequate oral hygiene can facilitate a diverse and balanced microbiome, crucial for overall health.
Collapse
Affiliation(s)
- Ion Alexandru Popovici
- Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 010221 Bucharest, Romania;
| | - Cristian Ionut Orasanu
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University of Constanta, 900591 Constanta, Romania; (G.-C.C.); (A.C.); (R.I.V.)
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
| | - Georgeta-Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University of Constanta, 900591 Constanta, Romania; (G.-C.C.); (A.C.); (R.I.V.)
- “Sf. Apostol Andrei” County Emergency Clinical Hospital, 900591 Constanta, Romania
| | - Anita-Cristina Ionescu
- Oncological Institute “Prof. Dr. Alexandru Trestioreanu”, 022328 Bucharest, Romania; (A.-C.I.); (L.K.)
| | - Lidia Kajanto
- Oncological Institute “Prof. Dr. Alexandru Trestioreanu”, 022328 Bucharest, Romania; (A.-C.I.); (L.K.)
| | - Bogdan Cimpineanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
- “Sf. Apostol Andrei” County Emergency Clinical Hospital, 900591 Constanta, Romania
| | - Anca Chisoi
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University of Constanta, 900591 Constanta, Romania; (G.-C.C.); (A.C.); (R.I.V.)
- “Sf. Apostol Andrei” County Emergency Clinical Hospital, 900591 Constanta, Romania
| | - Adrian Nelutu Mitroi
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
- Railway Clinical Hospital, 900123 Constanta, Romania
| | - Ionut Poinareanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
| | - Raluca Ioana Voda
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology (CEDMOG), “Ovidius” University of Constanta, 900591 Constanta, Romania; (G.-C.C.); (A.C.); (R.I.V.)
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
| | - Oana Andreea Ursica
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
- “Sf. Apostol Andrei” County Emergency Clinical Hospital, 900591 Constanta, Romania
| | - Mihaela Butcaru Pundiche
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (B.C.); (A.N.M.); (I.P.); (O.A.U.); (M.B.P.)
- “Sf. Apostol Andrei” County Emergency Clinical Hospital, 900591 Constanta, Romania
| |
Collapse
|
20
|
Zhou Y, Ding Y, Xu B, Fei H, Wang Z. Genetically druggable targets for MAPK-activated colorectal cancer by a two-sample mendelian randomization analysis. Sci Rep 2025; 15:12239. [PMID: 40210889 PMCID: PMC11986099 DOI: 10.1038/s41598-024-82567-5] [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] [Received: 02/18/2024] [Accepted: 12/06/2024] [Indexed: 04/12/2025] Open
Abstract
Colorectal cancer (CRC) is a significant worldwide health issue, ranking second in women and third in men. Predictions estimate a rise to 2.5 million cases by 2035, with CRC being the fourth deadliest cancer due to delayed diagnosis and the scarcity of effective treatment options. Over 60% of CRC cases involve MAPK-activated signal pathways, particularly driven by RAS oncogene mutations, which hinder treatment responses, making them 'undruggable.' This study conducts a two-sample Mendelian randomization protein quantitative trait loci (pQTL) analysis to investigate the causal association between plasma proteins and MAPK-activated CRCs. The study indicates that four plasma proteins-MHC class I polypeptide-related sequence B (MICB), complement C4A, C4B, and interleukin-21 (IL-21) are associated with an increased risk of MAPK-activated CRCs. These findings highlight the possibility of utilizing plasma proteins as therapeutic targets and diagnostic markers to advance the fight against CRCs, indicating promising results for more effective interventions. To ascertain and expand upon these discoveries, further research is imperative to fully harness the potential of these discoveries.
Collapse
Affiliation(s)
- Yuxuan Zhou
- Department of Gastrointestinal Surgery/Hernia Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China
| | - Yunlong Ding
- Department of Emergency General Surgery, Weifang People's Hospital, Weifang, Shandong, China
| | - Bangyue Xu
- Jilin Central General Hospital, Changchun, Jilin, China
| | - Hongyang Fei
- Department of Hepatobiliary and Pancreatic Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery/Hernia Surgery, Jilin Province People's Hospital, No. 1183 Gongnong Road, Changchun, Jilin, China.
| |
Collapse
|
21
|
Mulla AN, Thorat ST, Chandramore K, Kumar P, Reddy KS, Kumar N. Curcumin as a protective agent against chromium and ammonia toxicity using molecular and biochemical approaches in fish. Sci Rep 2025; 15:12023. [PMID: 40200001 PMCID: PMC11978759 DOI: 10.1038/s41598-025-95369-0] [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: 01/06/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
In aquatic ecosystems, metal and ammonia pollution pose major concern as they contaminate the environment and induces toxicity in fish. The present study addresses the toxicity induced by chromium (Cr) and ammonia (NH3) toxicity in fish and investigates the potential of dietary curcumin in mitigating the effects of concurrent exposure to theses stressors in Pangasianodon hypophthalmus. Three isonitrogenous and isocaloric diets were formulated: a control diet (0% curcumin) and two curcumin-supplemented diets containing 0.1% and 0.2% curcumin. Four experimental groups were designed in a completely randomized design: (1) control, (2) concurrent exposure to Cr and NH3 toxicity and fed with control diet, (3) 0.1% curcumin with Cr and NH3 exposure, and (4) 0.2% curcumin with Cr and NH3 exposure. Fish fed with 0.2% curcumin diet, followed by the 0.1% curcumin diet under Cr and NH3 stress, exhibited significantly reduced cortisol levels compared to the control and Cr + NH3 groups. Similarly, the expression of HSP70 and iNOS genes in liver tissue was significantly downregulated in the 0.1% and 0.2% curcumin-fed groups compared to other groups. Concurrent exposure to Cr and NH3 led to a considerable increase in oxidative stress enzyme in liver and kidney tissues, including glutathione S-transferase (GST), catalase (CAT), and superoxide dismutase (SOD). However, dietary supplementation with 0.1% curcumin significantly reduced oxidative stress enzyme activities. The stressors markedly reduced acetylcholinesterase (AChE) activity, but supplementation with 0.1% curcumin restored AChE activity. The expression of stress-related genes such as cytochrome P450 (CYP450), caspase-3a (Cas3a), and tumor necrosis factor-alpha (TNF-α) was noticeably downregulated in the curcumin-fed groups, reducing the impact of Cr + NH3 stress. Furthermore, total immunoglobulin (Ig) levels and growth-related gene expression, including growth hormone (GH) and growth hormone receptor 1 (GHR1), were significantly upregulated in the 0.1% curcumin-fed group under Cr + NH3 stress compared to all other groups. Additionally, myostatin (MYST) gene expression was significantly downregulated in the 0.1% curcumin-fed group. Activities of cellular metabolic and digestive enzymes were significantly improved with curcumin supplementation, mitigating the adverse effects of Cr + NH3 stress compared to the control and other groups. Moreover, Cr bioaccumulation in different fish tissues was reduced in the 0.1% curcumin-fed group. This study highlights the potential of dietary curcumin in mitigating the adverse effects of concurrent Cr and NH3 exposure through gene regulation, thereby improving the physiological and productive performance of Pangasianodon hypophthalmus.
Collapse
Affiliation(s)
- Amir Najir Mulla
- ICAR-National Institute of Abiotic Stress Management, Malegaon, Pune, Baramati, 413115, India
- Vidya Pratishthan's Arts, Commerce and Science College Baramati, Pune, Baramati, 413133, India
| | - Supriya Tukaram Thorat
- ICAR-National Institute of Abiotic Stress Management, Malegaon, Pune, Baramati, 413115, India
| | - Kalpana Chandramore
- Vidya Pratishthan's Arts, Commerce and Science College Baramati, Pune, Baramati, 413133, India
| | - Prem Kumar
- ICAR-Central Institute of Fisheries Education, Versova, Mumbai, 400061, India
| | - Kotha Sammi Reddy
- ICAR-National Institute of Abiotic Stress Management, Malegaon, Pune, Baramati, 413115, India
| | - Neeraj Kumar
- ICAR-National Institute of Abiotic Stress Management, Malegaon, Pune, Baramati, 413115, India.
| |
Collapse
|
22
|
Zamanian MY, Maleki S, Oghenemaro EF, Singh M, Mohammadi M, Alkhayyat AH, Sapaev IB, Kaur P, Shirsalimi N, Nagarwal A. Omentin-1 as a promising biomarker and therapeutic target in hypertension and heart failure: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04008-y. [PMID: 40126671 DOI: 10.1007/s00210-025-04008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
Omentin-1, a novel adipocytokine predominantly secreted by visceral adipose tissue, has emerged as a significant factor in cardiovascular health, particularly regarding hypertension (HTN) and heart failure (HF). This manuscript investigates the multifaceted roles of omentin-1 in these conditions, emphasizing its protective effects on vascular function and its potential as both a biomarker and therapeutic target. Clinical studies indicate that reduced circulating levels of omentin-1 are associated with metabolic syndrome (MetS) and increased cardiovascular risk, while animal studies demonstrate its ability to ameliorate endothelial dysfunction and lower blood pressure. Omentin-1 exerts its beneficial effects through various signaling pathways, including AMP-activated protein kinase (AMPK) and protein kinase B (Akt), thereby promoting vasodilation, enhancing insulin sensitivity, and mitigating inflammation. In the context of HF, particularly heart failure with preserved ejection fraction (HFpEF), omentin-1 levels exhibit a negative correlation with diastolic dysfunction and inflammatory markers, suggesting its role in cardiac protection. Additionally, the manuscript discusses the implications of omentin-1 in managing obesity-related cardiovascular diseases and its potential utility as a prognostic marker for adverse outcomes in HF patients. Collectively, omentin-1 represents a promising avenue for research in cardiovascular health, with the potential to inform novel therapeutic strategies aimed at improving outcomes in patients with HTN and HF. Further research is necessary to elucidate the details of omentin-1 function and evaluate its potential in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Saba Maleki
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Guilan University of Medical Sciences (GUMS), Rasht, Guilan Province, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mandeep Singh
- Directorate of Sports and Physical Education, University of Jammu, Jammu, India
| | - Maryam Mohammadi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hussen Alkhayyat
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ibrokhim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan
- Scientific Researcher, University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Niyousha Shirsalimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Amritesh Nagarwal
- Department of Cardiology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| |
Collapse
|
23
|
Zhang W, Wang J, Li H, Zhang X, Yao D, Zhang H, Zhou X, Nie J, Lai T, Zhu H, Gong Y, Tanaka Y, Li X, Liao X, Su L. TAF7 directly targets SAA1 to enhance triple-negative breast cancer metastasis via phosphorylating E-cadherin and N-cadherin. iScience 2025; 28:111989. [PMID: 40083715 PMCID: PMC11903838 DOI: 10.1016/j.isci.2025.111989] [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: 08/27/2024] [Revised: 12/10/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Identification of metastasis drivers of triple-negative breast cancer (TNBC) is a multifaceted challenge. Here, we identified TATA-box binding protein associated factor 7 (TAF7) as a candidate to modulate TNBC metastasis. TAF7 exhibited high expression in metastatic TNBC patients, and its elevated expression showed a negative correlation with overall survival in TNBC patients. The knockdown of TAF7 suppressed the migration and invasion of TNBC, suggesting TAF7 plays a role in the metastatic processes. Further, TAF7 was enhancing serum amyloid A1 (SAA1) transcription by binding to a specific motif in the SAA1 gene promoter. The elevated SAA1 in TNBC cells directly increased E-cadherin and N-cadherin phosphorylation thereby regulating cell adhesion. Mechanistically, TAF7 modulated cell invasion, migration, and lung metastasis through an SAA1-dependent manner in vitro and in vivo experiments. Taken together, it is likely that TAF7 could directly act on the SAA1 gene promoter, upregulating SAA1 and consequently promoting TNBC metastasis.
Collapse
Affiliation(s)
- Wanjun Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Hanning Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Dunjie Yao
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Huimin Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Xinhong Zhou
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiaqi Nie
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tongxing Lai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yiping Gong
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Yoshimasa Tanaka
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida, Sakyo-ku, Kyoto 606-8501, Japan
| | - Xingrui Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinghua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Li Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
24
|
Lim DM, Kim D, Ju HM, Jeong SH, Kim YH, Ok SM, Park HR. Distinct Immunological Features Compared to Lichen Planus and Oral Lichen Planus. J Inflamm Res 2025; 18:4037-4056. [PMID: 40125076 PMCID: PMC11929516 DOI: 10.2147/jir.s506313] [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: 11/26/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Lichen planus (LP) and oral lichen planus (OLP) share clinical and histological similarities, yet their distinct immunopathological mechanisms make differentiation and management challenging. Clarifying these differences is essential for accurate diagnosis and treatment. This study aimed to investigate the systemic immune profile of OLP using single-cell transcriptomics, identifying distinct immune cell subsets and signaling pathways contributing to its chronic inflammatory state. Additionally, it sought to compare the inflammatory lesion microenvironments of OLP and LP by analyzing key immune pathways and cellular interactions. Methods Peripheral blood mononuclear cells (PBMCs) were obtained from 16 OLP patients and 5 healthy controls. Single-cell transcriptomic data from PBMCs and lesion tissues of OLP and LP were analyzed to profile immune and inflammatory signatures. Key molecular findings were validated using independent datasets and enzyme-linked immunosorbent assays (ELISA). Results Prostaglandin D2 synthase (PTGDS), a pivotal enzyme in prostaglandin metabolism, emerged as a diagnostic marker with elevated expression in NK cells from OLP patients. Additionally, a novel CXCR4 high-TSC22D3 high CD4 cytotoxic T cell subset with enhanced cytotoxicity was identified, potentially contributing to OLP pathogenesis. OLP blood samples also demonstrated significant upregulation of TNF and TLR signaling in NK cells, indicating a heightened chronic inflammatory state. Comparative tissue analysis revealed intensified TNF-driven inflammation and a disrupted HIF1A- vascular endothelial growth factor (VEGF) interactions in OLP, contrasting with LP's robust VEGF-mediated angiogenesis. Discussion These findings highlight distinct immunopathogenic mechanisms between OLP and LP. The upregulation of PTGDS in NK cells and CXCR4 high-TSC22D3 high CD4 cytotoxic T cells in PBMCs indicates systemic immune dysregulation in OLP, while tissue-level differences suggest impaired vascular remodeling and chronic inflammation. These insights underscore the need for targeted immunomodulatory therapies. Conclusion This study identifies distinct immune signatures that differentiate OLP from LP, highlighting potential therapeutic targets that require further validation for personalized treatment strategies.
Collapse
Affiliation(s)
- Dong Min Lim
- Interdisciplinary Program of Genomic Data Science, Pusan National University, Yangsan, 50612, Republic of Korea
| | - DoYeon Kim
- Department of Oral Pathology, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hye-Min Ju
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, 50612, Republic of Korea
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, 50612, Republic of Korea
| | - Sung-Hee Jeong
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, 50612, Republic of Korea
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, 50612, Republic of Korea
| | - Yun Hak Kim
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Soo-Min Ok
- Department of Oral Medicine, Dental and Life Science Institute, Pusan National University School of Dentistry, Yangsan, 50612, Republic of Korea
- Department of Oral Medicine, Dental Research Institute, Pusan National University Dental Hospital, Yangsan, 50612, Republic of Korea
| | - Hae Ryoun Park
- Department of Oral Pathology, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Periodontal Disease Signaling Network Research Center, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Periodontology and Dental Research Institute, Pusan National University Dental Hospital, Yangsan, 50612, Republic of Korea
| |
Collapse
|
25
|
Arias AM, Reinartz DM, Sairs C, Kumar SS, Wilson JE. Streptococcus anginosus Activates the NLRP3 Inflammasome to Promote Inflammatory Responses from Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642696. [PMID: 40161672 PMCID: PMC11952393 DOI: 10.1101/2025.03.12.642696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic inflammation and oral dysbiosis are common features of oral squamous cell carcinoma (OSCC). The commensal streptococci, S. anginosus, is increased in oral diseases including OSCC. Our previous work revealed that S. anginosus promotes inflammatory responses from macrophage cell lines, however the molecular mechanism by which S. anginosus interacts with macrophages to instigate this response remains to be investigated. Here, we expand on our previous findings by investigating the effects of S. anginosus infection of primary bone marrow derived macrophages (BMMs) and during in vivo infection. We found S. anginosus activated primary BMMs, which presented an enlarged cellular area, increased NF-κB activation and downstream inflammatory cytokines TNF⍰, IL-6 and IL-1β at 24 hours post infection. S. anginosus viability was dispensable for NF-κB activation, but essential for the induction of downstream inflammatory proteins and cytokines. S. anginosus persisted intracellularly within BMMs and induced the expression of inflammasome sensors AIM2, NLRC4 and NLRP3. Further, BMMs lacking the inflammasome adapter protein ASC ( Asc -/- ) had significantly diminished IL-1β production compared to wild type BMMs, indicating that S. anginosus activated the inflammasome. S. anginosus primarily triggered the inflammasome through NLRP3 as S. anginosus -infected Nlrp3 -/- BMMs and NLRP3 inhibitor (MCC950)-treated wild type BMMs displayed diminished IL-1β production compared to wild type controls. Lastly, S. anginosus -infected Asc -/- and Nlrp3 -/- mice displayed reduced weight loss compared to C57BL/6 mice. These overall findings indicate that S. anginosus replicates within macrophages and promotes a proinflammatory response in part through activation of the NLRP3 inflammasome. brief summary sentence: S. anginosus replicates intracellularly within macrophages and is sensed by the NLRP3 inflammasome to promote proinflammatory response.
Collapse
|
26
|
Alonso-Marañón J, Solé L, Álvarez-Villanueva D, Maqueda M, Lobo-Jarne T, Montoto Á, Yélamos J, Borràs E, Uraga L, Hooper C, Sabidó E, Miyamoto S, Bigas A, Espinosa L. NEMO is essential for directing the kinases IKKα and ATM to the sites of DNA damage. Sci Signal 2025; 18:eadr0128. [PMID: 40067909 PMCID: PMC12070652 DOI: 10.1126/scisignal.adr0128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025]
Abstract
The DNA damage repair kinase ATM is phosphorylated by the NF-κB pathway kinase IKKα, resulting in enhanced DNA damage repair through the nonhomologous end-joining pathway. Thus, inhibition of IKKα enhances the efficacy of cancer therapy based on inducing DNA damage. Here, we found a role for the IKK regulatory subunit NEMO in DNA damage repair mediated by ATM and IKKα. Exposure to damaging agents induced the interaction of NEMO with a preformed ATM-IKKα complex, which was required to target active ATM and IKKα to chromatin for efficient DNA damage repair but not for activating ATM. Recognition of damaged DNA by the IKKα-NEMO-ATM complex was facilitated by the interaction between NEMO and histones and depended on the ADP ribosylation of histones by the enzyme PARP1. NEMO-deficient cells showed increased activity of the kinase ATR, and inhibition of ATR potentiated the effect of chemotherapy in cells lacking NEMO or IKKα. Bioinformatic analysis of colorectal cancer datasets demonstrated that the expression of genes encoding IKKα, NEMO, and ATM correlated with poor patient prognosis, suggesting that the mechanism linking these three elements may be clinically relevant.
Collapse
Affiliation(s)
- Josune Alonso-Marañón
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Laura Solé
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Daniel Álvarez-Villanueva
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet del Llobregat, Barcelona 08908, Spain
| | - María Maqueda
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Teresa Lobo-Jarne
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Ángela Montoto
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Jose Yélamos
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
- Immunology Unit, Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Eva Borràs
- Proteomics Unit, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona 08003, Spain
- Proteomics Unit, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Leire Uraga
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| | - Christopher Hooper
- McArdle Laboratory for Cancer Research, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Wisconsin Institutes for Medical Research, Madison, WI 53705, USA
| | - Eduard Sabidó
- Proteomics Unit, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona 08003, Spain
- Proteomics Unit, Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Shigeki Miyamoto
- McArdle Laboratory for Cancer Research, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Wisconsin Institutes for Medical Research, Madison, WI 53705, USA
| | - Anna Bigas
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
- Josep Carreras Leukemia Research Institute, Barcelona 08916, Spain
| | - Lluís Espinosa
- Cancer Research Program, Hospital del Mar Research Institute, CIBERONC, Hospital del Mar, Doctor Aiguader 88, Barcelona 08003, Spain
| |
Collapse
|
27
|
Hu L, Luo Y, Yang J, Cheng C. Botanical Flavonoids: Efficacy, Absorption, Metabolism and Advanced Pharmaceutical Technology for Improving Bioavailability. Molecules 2025; 30:1184. [PMID: 40076406 PMCID: PMC11902153 DOI: 10.3390/molecules30051184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Flavonoids represent a class of natural plant secondary metabolites with multiple activities including antioxidant, antitumor, anti-inflammatory, and antimicrobial properties. However, due to their structural characteristics, they often exhibit low bioavailability in vivo. In this review, we focus on the in vivo study of flavonoids, particularly the effects of gut microbiome on flavonoids, including common modifications such as methylation, acetylation, and dehydroxylation, etc. These modifications aim to change the structural characteristics of the original substances to enhance absorption and bioavailability. In order to improve the bioavailability of flavonoids, we discuss two feasible methods, namely dosage form modification and chemical modification, and hope that these approaches will offer new insights into the application of flavonoids for human health. In this article, we also introduce the types, plant sources, and efficacy of flavonoids. In conclusion, this is a comprehensive review on how to improve the bioavailability of flavonoids.
Collapse
Affiliation(s)
- Lei Hu
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Yiqing Luo
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Jiaxin Yang
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| | - Chunsong Cheng
- Jiangxi Key Laboratory for Sustainable Utilization of Chinese Materia Medica Resources, Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China; (L.H.); (Y.L.); (J.Y.)
- Lushan Xinglin Institute for Medicinal Plants, Jiujiang Xinglin Key Laboratory for Traditional Chinese Medicines, Jiujiang 332900, China
| |
Collapse
|
28
|
Yin L, Wang H, Xu H, Lu H, Lv J, Lu C. Asperuloside suppresses the progression of depression through O-GlcNAcylation of IκBα and regulating NFκB signaling. J Pharmacol Sci 2025; 157:179-188. [PMID: 39929592 DOI: 10.1016/j.jphs.2025.01.010] [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: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 05/08/2025] Open
Abstract
Depression is a pervasive mental disorder that poses a significant threat to human health globally. Asperuloside (ASP), an iridoid glycoside extracted from Herba Paederiae, exhibits a range of pharmacological activities, including anti-tumor and anti-inflammatory effects. This study aims to explore the function and molecular mechanisms of ASP in alleviating depression. Chronic unpredictable mild stress (CUMS) was employed to establish a rat model of depression. Behavioral tests were conducted to evaluate the antidepressant effects of ASP. Apoptosis in hippocampal tissues was assessed using TUNEL assay. Primary hippocampal neuron apoptosis was assessed using Annexin V/PI staining and flow cytometry, while cell death was detected via PI staining. The expression levels of target mRNAs and proteins were analyzed by quantitative PCR (qPCR) and western blotting, respectively. Additionally, the levels of O-GlcNAcylation and ubiquitination were determined by western blot analysis following immunoprecipitation. Molecular docking was performed to elucidate the interaction mode between ASP and its target protein, O-linked β-N-acetylglucosamine transferase (OGT). Our findings revealed that ASP treatment significantly ameliorated depression-like behaviors and cognitive dysfunction, as well as inhibited hippocampus apoptosis in CUMS-induced rats, Moreover, ASP inhibited LPS-induced neuronal cell apoptosis and suppressed the activation of the NF-κB signaling pathway. Mechanistically, we demonstrated that ASP promoted O-GlcNAcylation of IκBα, and suppressed its ubiquitination and phosphorylation, thereby stabilizing IκBα protein. In conclusion, ASP exerts antidepressant effects by enhancing IκBα O-GlcNAcylation, thus inhibiting its ubiquitination and phosphorylation. These findings provide a novel therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Li Yin
- Zunyi Medical and Pharmaceutical College, No. 8, North Section of Ping'an Avenue, Honghuagang District, Zunyi City, Guizhou, China
| | - Huakun Wang
- Zunyi Medical and Pharmaceutical College, No. 8, North Section of Ping'an Avenue, Honghuagang District, Zunyi City, Guizhou, China
| | - Hong Xu
- Department of Scientific Research and Technology, Xinjiang Uyghur Autonomous Region Institute for Drug Control, No.518, Xiba Jiahu Road, Xinshi District, Urumqi, 830054, Xinjiang, China
| | - Haixiao Lu
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China; Bioengineering & Technology Center for Native Medicinal Resources Development, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China
| | - Jiayu Lv
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China
| | - Chengshu Lu
- Department of Biopharmaceutics, Yulin Normal University, No. 1303, East education road, Yulin, 537000, Guangxi, China.
| |
Collapse
|
29
|
Zhou M, Wang J, Peng Y, Tian X, Zhang W, Chen J, Wang Y, Wang Y, Yang Y, Zhang Y, Huo X, Wu Y, Yu Z, Xie T, Ma X. Elemene as a binding stabilizer of microRNA-145-5p suppresses the growth of non-small cell lung cancer. J Pharm Anal 2025; 15:101118. [PMID: 40161444 PMCID: PMC11953980 DOI: 10.1016/j.jpha.2024.101118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 04/02/2025] Open
Abstract
Elemene is widely recognized as an effective anti-cancer compound and is routinely administered in Chinese clinical settings for the management of several solid tumors, including non-small cell lung cancer (NSCLC). However, its detailed molecular mechanism has not been adequately demonstrated. In this research, it was demonstrated that elemene effectively curtailed NSCLC growth in the patient-derived xenograft (PDX) model. Mechanistically, employing high-throughput screening techniques and subsequent biochemical validations such as microscale thermophoresis (MST), microRNA-145-5p (miR-145-5p) was pinpointed as a critical target through which elemene exerts its anti-tumor effects. Interestingly, elemene serves as a binding stabilizer for miR-145-5p, demonstrating a strong binding affinity (dissociation constant (K D) = 0.39 ± 0.17 μg/mL) and preventing its degradation both in vitro and in vivo, while not interfering with the synthesis of the primary microRNA transcripts (pri-miRNAs) and precursor miRNAs (pre-miRNAs). The stabilization of miR-145-5p by elemene resulted in an increased level of this miRNA, subsequently suppressing NSCLC progression through the miR-145-5p/mitogen-activated protein kinase kinase kinase 3 (MAP3K3)/nuclear factor kappaB (NF-κB) pathway. Our findings provide a new perspective on revealing the interaction patterns between clinical anti-tumor drugs and miRNAs.
Collapse
Affiliation(s)
- Meirong Zhou
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Jiayue Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yulin Peng
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Xiangge Tian
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Wen Zhang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Junlin Chen
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Yue Wang
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yu Wang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Youjian Yang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Yongwei Zhang
- Research & Production Department, Dalian Huali Jingang Pharmaceutical Co., Ltd., Dalian, Liaoning, 116110, China
| | - Xiaokui Huo
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Yuzhuo Wu
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Tian Xie
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xiaochi Ma
- Second Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, 116000, China
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, 116044, China
- Shenzhen Bao'an Authentic TCM Therapy Hospital, Shenzhen, Guangdong, 518101, China
| |
Collapse
|
30
|
Chen X, Sun F, Wang X, Feng X, Aref AR, Tian Y, Ashrafizadeh M, Wu D. Inflammation, microbiota, and pancreatic cancer. Cancer Cell Int 2025; 25:62. [PMID: 39987122 PMCID: PMC11847367 DOI: 10.1186/s12935-025-03673-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
Pancreatic cancer (PC) is a malignancy of gastrointestinal tract threatening the life of people around the world. In spite of the advances in the treatment of PC, the overall survival of this disease in advanced stage is less than 12%. Moreover, PC cells have aggressive behaviour in proliferation and metastasis as well as capable of developing therapy resistance. Therefore, highlighting the underlying molecular mechanisms in PC pathogenesis can provide new insights for its treatment. In the present review, inflammation and related pathways as well as role of gut microbiome in the regulation of PC pathogenesis are highlighted. The various kinds of interleukins and chemokines are able to regulate angiogenesis, metastasis, proliferation, inflammation and therapy resistance in PC cells. Furthermore, a number of molecular pathways including NF-κB, TLRs and TGF-β demonstrate dysregulation in PC aggravating inflammation and tumorigenesis. Therapeutic regulation of these pathways can reverse inflammation and progression of PC. Both chronic and acute pancreatitis have been shown to be risk factors in the development of PC, further highlighting the role of inflammation. Finally, the composition of gut microbiota can be a risk factor for PC development through affecting pathways such as NF-κB to mediate inflammation.
Collapse
Affiliation(s)
- XiaoLiang Chen
- Department of General Surgery and Integrated Traditional Chinese and Western Medicine Oncology, Tiantai People'S Hospital of Zhejiang Province(Tiantai Branch of Zhejiang Provincial People'S Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| | - Feixia Sun
- Nursing Department, Shandong First Medical University Affiliated Occupational Disease Hospital (Shandong Provincial Occupational Disease Hospital), Jinan, China
| | - Xuqin Wang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, 525200, Guangdong, China
| | - Amir Reza Aref
- VitroVision Department, DeepkinetiX, Inc, Boston, MA, USA
| | - Yu Tian
- Research Center, the Huizhou Central People'S Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
- School of Public Health, Benedictine University, No. 5700 College Road, Lisle, IL, 60532, USA.
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China.
| | - Dengfeng Wu
- Department of Emergency, The People'S Hospital of Gaozhou, No. 89 Xiguan Road, Gaozhou, 525200, Guangdong, China.
| |
Collapse
|
31
|
Liu X, Gong Z, Yang Y, Dong J, Zhang L, Li Z, Zhao F, Zhang J, Gao R. Chebulagic acid inhibits lipopolysaccharide-induced endometritis by regulating mitogen-activated protein kinase/nuclear factor-κB signaling. J Reprod Immunol 2025; 169:104464. [PMID: 40010025 DOI: 10.1016/j.jri.2025.104464] [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: 07/29/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
This study investigates the potential protective effects of chebulagic acid (CA) against endometritis and its underlying molecular mechanisms. Network pharmacology analysis identified 19 potential targets of CA related to endometritis, mainly associated with the mitogen-activated protein kinase (MAPK) signaling pathways. Molecular docking analysis further indicated that MAPK14 and MAPK3 are critical targets of CA, suggesting its potential role in modulating inflammatory responses. In vitro experiments demonstrated that CA at concentrations of 12.5, 25, and 50 µg/mL significantly inhibited the secretion of proinflammatory cytokines interleukin (IL)-1β and IL-6 in lipopolysaccharide (LPS)-stimulated bovine endometrial epithelial cells (bEECs), without affecting cell viability. In vivo, CA treatment mitigated uterine inflammation in an LPS-induced mouse model of endometritis by downregulating high-mobility group box protein 1 (HMGB1) expression and inhibiting the phosphorylation of key signaling molecules, including p65, extracellular signal-regulated kinase (ERK), and p38. These findings suggest that CA exerts significant anti-inflammatory effects in endometritis by modulating the MAPK/NF-κB signaling pathway. Given its potential to suppress excessive inflammatory responses, CA may serve as a promising candidate for the development of novel therapeutic strategies for endometritis.
Collapse
Affiliation(s)
- Xinyu Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhiguo Gong
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Ying Yang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Jinzhong Dong
- Guang Yilong Township Government, Chayouzhong Banne, Ulaanchabu City, Inner Mongolia Autonomous Region, China
| | - Lanxin Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhengyi Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Feifan Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Jianbing Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China.
| | - Ruifeng Gao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China.
| |
Collapse
|
32
|
Ha B, Kang JH, Kim DH, Lee MY. Lipopolysaccharide-Induced Inflammatory Response and Its Prominent Suppression by Paspalum thunbergii Extract. Int J Mol Sci 2025; 26:1611. [PMID: 40004077 PMCID: PMC11855676 DOI: 10.3390/ijms26041611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The extract of Paspalum thunbergii, a native perennial herb in Korea belonging to the rice family, was investigated for its anti-inflammatory activity and the underlying mechanisms driving its effects. Fifteen chemical components of the P. thunbergii extract, including rosmarinic acid and isoquercitrin, were identified using LC-MS. The extract showed antioxidative activity through DPPH and ABTS cation radical scavenging activity. The P. thunbergii extract significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production in macrophage RAW 264.7 cells. The extract inhibited the expression of lipopolysaccharide-induced iNOS and COX-2, which are inflammation-related enzymes. To explore the underlying anti-inflammatory mechanism, the expression levels of signal proteins related to MAPK, NF-κB, JAK/STAT, and Wnt/β-catenin signaling were measured. As a result, the P. thunbergii extract inhibited the expression of p-p38, and p-JNK increased by LPS in RAW 264.7 cells. Additionally, it decreased the expression of LPS-induced p-IKKβ and p-NF-κB p65 and prevented the migration of p-NF-κB into the nucleus caused by LPS. Notably, p-JAK1, p-STAT3, Wnt 3α, β-catenin, and p-GSK-3β protein expressions were also inhibited. Therefore, the prominent anti-inflammatory activity of the P. thunbergii extract may be via the MAPK, NF-κB, JAK/STAT, Wnt/β-catenin signal pathway.
Collapse
Affiliation(s)
- Bin Ha
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Ji-Hye Kang
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Do Hyun Kim
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| | - Mi-Young Lee
- Department of Medical Science, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Medical Biotechnology, College of Medical Science, Soonchunhyang University, Asan-si 31538, Chungcheongnam-do, Republic of Korea;
- Department of Research and Development, Eshel Biopharm Co., Ltd., Asan-si 31538, Chungcheongnam-do, Republic of Korea;
| |
Collapse
|
33
|
Zhang S, Huang J, Jiang Z, Tong H, Ma X, Liu Y. Tumor microbiome: roles in tumor initiation, progression, and therapy. MOLECULAR BIOMEDICINE 2025; 6:9. [PMID: 39921821 PMCID: PMC11807048 DOI: 10.1186/s43556-025-00248-9] [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: 08/02/2024] [Revised: 01/06/2025] [Accepted: 01/21/2025] [Indexed: 02/10/2025] Open
Abstract
Over the past few years, the tumor microbiome is increasingly recognized for its multifaceted involvement in cancer initiation, progression, and metastasis. With the application of 16S ribosomal ribonucleic acid (16S rRNA) sequencing, the intratumoral microbiome, also referred to as tumor-intrinsic or tumor-resident microbiome, has also been found to play a significant role in the tumor microenvironment (TME). Understanding their complex functions is critical for identifying new therapeutic avenues and improving treatment outcomes. This review first summarizes the origins and composition of these microbial communities, emphasizing their adapted diversity across a diverse range of tumor types and stages. Moreover, we outline the general mechanisms by which specific microbes induce tumor initiation, including the activation of carcinogenic pathways, deoxyribonucleic acid (DNA) damage, epigenetic modifications, and chronic inflammation. We further propose the tumor microbiome may evade immunity and promote angiogenesis to support tumor progression, while uncovering specific microbial influences on each step of the metastatic cascade, such as invasion, circulation, and seeding in secondary sites. Additionally, tumor microbiome is closely associated with drug resistance and influences therapeutic efficacy by modulating immune responses, drug metabolism, and apoptotic pathways. Furthermore, we explore innovative microbe-based therapeutic strategies, such as engineered bacteria, oncolytic virotherapy, and other modalities aimed at enhancing immunotherapeutic efficacy, paving the way for microbiome-centered cancer treatment frameworks.
Collapse
Affiliation(s)
- Shengxin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jing Huang
- Department of Medical Ultrasound, West China Hospital of Sichuan University, 37 Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Zedong Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Huan Tong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| | - Yang Liu
- Day Surgery Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
| |
Collapse
|
34
|
Miyazaki A, Yoshida S, Takeda Y, Tomaru U, Matsumoto M, Seya T. Th1 adjuvant ARNAX, in combination with radiation therapy, enhances tumor regression in mouse tumor-implant models. Immunol Lett 2025; 271:106947. [PMID: 39603425 DOI: 10.1016/j.imlet.2024.106947] [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: 08/04/2024] [Revised: 10/31/2024] [Accepted: 11/23/2024] [Indexed: 11/29/2024]
Abstract
Radiation therapy (RT) rarely induces tumor regression at untreated metastatic sites, the so-called abscopal effect. A syngeneic tumor (EG7) transplanted into a Th1-dominant mouse strain (C57BL/6) regressed significantly on the treated side and less on the contralateral side after RT. Additional subcutaneous administration of ARNAX, a non-inflammatory adjuvant, further accelerated tumor regression on the untreated side. This suggests that ARNAX after RT significantly enhances the tumor regression effect on the irrelevant tumor. Based on this setting, we next observed similar tumor shrinkage after RT and ARNAX by transplanting syngeneic breast cancer tumors (4T1) into a Th2-dominant mouse strain (BALB/c). The results were as follows: 1. ARNAX enhanced RT-mediated tumor shrinkage comparable to polyI:C; 2. In the Th2 mouse strain, little tumor regression occurred on the untreated side compared to tumor regression on the treated side after RT alone; 3. RT+ARNAX treatment caused additive regression on the treated side and induced slight tumor regression on the untreated side; 4. PD-L1 antibody + RT combination therapy caused tumor regression and further induced additive regression with ARNAX; 5. The combination of RT and ARNAX reduced the number and volume of lung metastases compared to RT alone. However, tumor regression was not always accompanied by a significant prolongation of survival in the mice receiving our regimen and protocol (one 10Gy radiation and a single ARNAX treatment). In conclusion, RT therapy promoted abscopal tumor regression in both Th2 and Th1 models with the addition of the non-inflammatory adjuvant ARNAX.
Collapse
Affiliation(s)
- Aya Miyazaki
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Sumito Yoshida
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Yohei Takeda
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Utano Tomaru
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Misako Matsumoto
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan; Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan.
| | - Tsukasa Seya
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan; Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; Nebuta Research Institute for Life Sciences, Aomori University, Aomori 030-0943, Japan.
| |
Collapse
|
35
|
Ece G, Aktaş A, Caner A, Sağlık İ, Kula Atik T, Ulusan Bağcı Ö, Bayındır Bilman F, Demirbakan H, Güdül Havuz S, Kaya E, Koyuncu Özyurt Ö, Yetkin G, Zorbozan O. The Urogenital System Microbiota: Is It a New Gamechanger in Urogenital Cancers? Microorganisms 2025; 13:315. [PMID: 40005682 PMCID: PMC11858393 DOI: 10.3390/microorganisms13020315] [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: 01/15/2025] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
The human microbiome, which encompasses microbial communities and their genetic material, significantly influences health and disease, including cancer. The urogenital microbiota, naturally present in the urinary and genital tracts, interact with factors such as age, lifestyle, and health conditions to affect homeostasis and carcinogenesis. Studies suggest that alterations in this microbiota contribute to the development and progression of genitourinary cancers, emphasizing the concept of oncobiome, which refers to microbial genetic contributions to cancer. Similarly, gut microbiota can influence hormone levels and systemic inflammation, impacting cancers such as cervical and prostate cancer. Advanced studies indicate that microbial communities in genitourinary cancers have distinct profiles that may serve as diagnostic biomarkers or therapeutic targets. Dysbiosis of the urinary microbiota correlates with bladder and kidney cancer. Additionally, gut microbiota influence the effectiveness of cancer treatments. However, further research is necessary to clarify causality, the role of microbial metabolites, and hormonal regulation. The aim of this review is to understand that these dynamics present opportunities for innovative cancer diagnostics and therapies, highlighting the need for integration of microbiology, oncology, and genomics to explore the role of microbiota in genitourinary cancers. For this, a comprehensive search of relevant databases was conducted, applying specific inclusion and exclusion criteria to identify studies examining the association between microbiota and urogenital cancers. Research into the mechanisms by which microbiota influence urogenital cancers may pave the way for new diagnostic and therapeutic approaches, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Gülfem Ece
- Department of Medical Microbiology, İzmir City Hospital, İzmir 35540, Türkiye; (G.E.); (F.B.B.)
| | - Ahmet Aktaş
- İstanbul Provincial Health Directorate, Istanbul Public Health Laboratory No. 2, İstanbul 34524, Türkiye;
| | - Ayse Caner
- Department of Parasitology, Faculty of Medicine, Department of Basic Oncology, Institute of Health Sciences, Ege University, Izmir 35100, Türkiye
| | - İmran Sağlık
- Department of Medical Microbiology, Faculty of Medicine, Uludag University, Bursa 16059, Türkiye;
| | - Tuğba Kula Atik
- Department of Microbiology, Faculty of Medicine, Balıkesir University, Balıkesir 10145, Türkiye;
| | - Özlem Ulusan Bağcı
- Department of Parasitology, Faculty of Medicine, Ankara University, Ankara 06230, Türkiye;
| | - Fulya Bayındır Bilman
- Department of Medical Microbiology, İzmir City Hospital, İzmir 35540, Türkiye; (G.E.); (F.B.B.)
| | - Hadiye Demirbakan
- Department of Medical Microbiology, Faculty of Medicine, Sanko University, Gaziantep 27090, Türkiye;
| | - Seda Güdül Havuz
- Samsun Provincial Health Directorate, Samsun Bafra State Hospital, Department of Medical Microbiology, Samsun 55400, Türkiye;
| | - Esra Kaya
- Department of Medical Microbiology, Kahramanmaraş Necip Fazıl City Hospital, Kahramanmaraş 46100, Türkiye;
| | - Özlem Koyuncu Özyurt
- Department of Medical Microbiology, Faculty of Medicine, Akdeniz Univertsity, Antalya 07070, Türkiye;
| | - Gülay Yetkin
- Bakırköy Dr Sadi Konuk Education and Research Hospital, Hamidiye Faculty of Medicine, Health Science University, İstanbul 34140, Türkiye;
| | - Orçun Zorbozan
- Department of Medical Microbiology, Faculty of Medicine, Bakircay University, İzmir 35665, Türkiye;
| |
Collapse
|
36
|
Zheng W, Tang S, Ren X, Song S, Ai C. Fucoidan alleviated colitis aggravated by fiber deficiency through protecting the gut barrier, suppressing the MAPK/NF-κB pathway, and modulating gut microbiota and metabolites. Front Nutr 2025; 11:1462584. [PMID: 39925971 PMCID: PMC11802440 DOI: 10.3389/fnut.2024.1462584] [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: 07/10/2024] [Accepted: 12/26/2024] [Indexed: 02/11/2025] Open
Abstract
Insufficient dietary fiber intake has become a global public health issue, affecting the development and management of various diseases, including intestinal diseases and obesity. This study showed that dietary fiber deficiency enhanced the susceptibility of mice to colitis, which could be attributed to the disruption of the gut barrier integrity, activation of the NF-κB pathway, and oxidative stress. Undaria pinnatifida fucoidan (UPF) alleviated colitis symptoms in mice that fed with a fiber deficient diet (FD), characterized by increased weight gain and reduced disease activity index, liver and spleen indexes, and histological score. The protective effect of UPF against FD-exacerbated colitis can be attributed to the alleviation of oxidative stress, the preservation of the gut barrier integrity, and inhibition of the MAPK/NF-κB pathway. UPF ameliorated the gut microbiota composition, leading to increased microbiota richness, as well as increased levels of Muribaculaceae, Lactobacillaceae, and Bifidobacterium and reduced levels of Proteobacteria, Bacteroidetes, and Bacteroides. Metabolomics analysis revealed that UPF improved the profile of microbiota metabolites, with increased levels of carnitine and taurine and decreased levels of tyrosine and deoxycholic acid. This study suggests that UPF has the potential to be developed as a novel prebiotic agent to enhance human health.
Collapse
Affiliation(s)
- Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong, China
| | - Shuangru Tang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Xiaomeng Ren
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian, China
| | - Shuang Song
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian, China
| | - Chunqing Ai
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
- National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
37
|
Liu D, Liu L, Che X, Wu G. Discovery of paradoxical genes: reevaluating the prognostic impact of overexpressed genes in cancer. Front Cell Dev Biol 2025; 13:1525345. [PMID: 39911323 PMCID: PMC11794808 DOI: 10.3389/fcell.2025.1525345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Oncogenes are typically overexpressed in tumor tissues and often linked to poor prognosis. However, recent advancements in bioinformatics have revealed that many highly expressed genes in tumors are associated with better patient outcomes. These genes, which act as tumor suppressors, are referred to as "paradoxical genes." Analyzing The Cancer Genome Atlas (TCGA) confirmed the widespread presence of paradoxical genes, and KEGG analysis revealed their role in regulating tumor metabolism. Mechanistically, discrepancies between gene and protein expression-affected by pre- and post-transcriptional modifications-may drive this phenomenon. Mechanisms like upstream open reading frames and alternative splicing contribute to these inconsistencies. Many paradoxical genes modulate the tumor immune microenvironment, exerting tumor-suppressive effects. Further analysis shows that the stage- and tumor-specific expression of these genes, along with their environmental sensitivity, influence their dual roles in various signaling pathways. These findings highlight the importance of paradoxical genes in resisting tumor progression and maintaining cellular homeostasis, offering new avenues for targeted cancer therapy.
Collapse
Affiliation(s)
| | | | - Xiangyu Che
- *Correspondence: Guangzhen Wu, ; Xiangyu Che,
| | | |
Collapse
|
38
|
Ma W, Yan H, Ma H, Xu Z, Dai W, Wu Y, Zhang H, Li Y. Roles of leukemia inhibitory factor receptor in cancer. Int J Cancer 2025; 156:262-273. [PMID: 39279155 DOI: 10.1002/ijc.35157] [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: 04/14/2024] [Revised: 06/19/2024] [Accepted: 07/29/2024] [Indexed: 09/18/2024]
Abstract
Leukemia inhibitory factor receptor (LIFR), in complex with glycoprotein 130 (gp130) as the receptor for leukemia inhibitory factor (LIF), can bind to a variety of cytokines and subsequently activate a variety of signaling pathways, including Janus kinase/signal transducer and activator of transcription 3. LIF, the most multifunctional cytokines of the interleukin-6 family acts as both a growth factor and a growth inhibitor in different types of tumors. LIF/LIFR signaling regulates a broad array of tumor-related processes including proliferation, apoptosis, migration, invasion. However, due to the activation of different signaling pathways, opposite regulatory effects are observed in certain tumor cells. Therefore, the role of LIFR in human cancers varies across different tumor and tissue, despite their recognized value in tumor treatment and prognosis observation is affirmed. Given its aberrant expression in numerous tumor cells and crucial regulatory function in tumorigenesis and progression, LIFR is considered as a promising targeted therapeutic agent. This review provides an overview of LIFR's initiating signaling pathway function as a cytokine receptor and summarize the current literature on the role of LIFR in cancer and its possible use in therapy.
Collapse
Affiliation(s)
- Wei Ma
- School of Stomatology, China Medical University, Shenyang, China
| | - Haixu Yan
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Haoyuan Ma
- Department of Clinical Medicine, China Medical University, Shenyang, China
| | - Zengyan Xu
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Wei Dai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yudan Wu
- School of Nursing, China Medical University, Shenyang, China
| | - Hongyan Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yanshu Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
39
|
Jain A, John S, Verma S, Gupta S. Navigating controversies in primary intraosseous carcinoma: A comprehensive literature review concerning the odontogenic origin and diagnostic challenges. Semin Diagn Pathol 2025; 42:50-54. [PMID: 39843327 DOI: 10.1053/j.semdp.2025.01.001] [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: 12/06/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025]
Abstract
Primary intraosseous carcinoma (PIOC) is a rare and challenging jawbone malignancy often linked to odontogenic cysts. With minimal connection to oral mucosa and a low incidence rate, PIOC presents significant diagnostic difficulties, often mimicking other odontogenic lesions. Histogenesis and the correct classification of the lesion remains debated, with theories suggesting origins from odontogenic epithelium or cysts. Chronic inflammation may contribute to malignant transformation, though genetic predispositions could also play a role in the pathogenesis. This review underscores the current knowledge of the lesion with the need for standardized diagnostic markers and an enhanced understanding of PIOC origin to improve diagnostic accuracy and treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Shalini Gupta
- Dept of Oral Pathology, King George's Medical University, Uttar Pradesh, India.
| |
Collapse
|
40
|
Dong Y, Zou YZ, Li T, Sun JH, Li H, Zhuang WY, Song Y, Wang CM. Schisandrol A Alleviates Allergic Asthma in Mice via Regulating the NF-κB/IκBα and Nrf2/HO-1 Signaling Pathways. J Med Food 2025; 28:28-37. [PMID: 39315928 DOI: 10.1089/jmf.2024.k.0117] [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] [Indexed: 09/25/2024] Open
Abstract
Schisandra chinensis (Turcz) Baill (S. chinensis) is the key traditional Chinese medicine for the treatment of asthma used by ancient and modern medical practitioners. However, the material basis and the main mechanism of its antiasthmatic effect remain unclear. Our preliminary results showed that schisandrol A (SCA), a representative monomer of Schisandra lignans, had the best relaxation effect on tracheal rings in isolated rats. In this research, a mouse asthma model was prepared by combining ovalbumin (OVA) with Al (OH)3 for exploring the antiasthmatic action and the underlying mechanism of SCA. The study results demonstrated that SCA improved the behavior of mice with asthma and pathological changes in their lung tissues and airways, decreased serum immunoglobulin E (IgE) and OVA-IgE levels, interleukin-4 (IL-4), IL-5, IL-13, and eotaxin contents, and leukocytes number in bronchoalveolar lavage fluid. SCA downregulated the gene expressions of keratinocyte-derived protein chemokines and ILs and reduced the expressions of phosphorylated IκB kinase α (p-IKKα) and p-nuclear factor kappa-B (NF-κB) proteins in lung tissues. In addition, it was found that SCA could significantly increase T-superoxide dismutase and catalase activities, decrease malondialdehyde content, and elevate p-IκBα, NF-E2-related-factor 2 (Nrf2), and heme oxygenase-1 (HO-1) protein expressions. In summary, SCA treatment resulted in a significant improvement in the allergic bronchial asthma in mice, and its mechanisms may involve the regulation of the NF-κB/IκBα pathway to reduce inflammatory response and the Nrf2/HO-1 pathway to improve the body's antioxidant capacity. These results suggest that SCA is a key component of S. chinensis in exerting antiasthmatic effects.
Collapse
Affiliation(s)
- Yang Dong
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| | - Yi-Zhuo Zou
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| | - Ting Li
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| | - Jing-Hui Sun
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| | - He Li
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| | - Wen-Yue Zhuang
- Department of Molecular Biology Test Technique, School of Medical Technology, Beihua University, Jilin, China
| | - Yan Song
- Department of Medical Nursing, School of Nursing, Beihua University, Jilin, China
| | - Chun-Mei Wang
- Department of Pharmacology, School of Pharmacy, Beihua University, Jilin, China
| |
Collapse
|
41
|
Liao AQ, Wen J, Wei JC, Xu BB, Jin N, Lin HY, Qin XY. Syntheses, crystal structures of copper (II)-based complexes of sulfonamide derivatives and their anticancer effects through the synergistic effect of anti-angiogenesis, anti-inflammation, pro-apoptosis and cuproptosis. Eur J Med Chem 2024; 280:116954. [PMID: 39406115 DOI: 10.1016/j.ejmech.2024.116954] [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: 04/27/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/25/2024]
Abstract
Three novel copper(II)-based complexes Cu-1, Cu-2, and Cu-3 containing sulfamethoxazole or sulfamethazine ligand were obtained, and their single structures were characterized. Both Cu-1 and Cu-3 show a broad spectrum of cytotoxicity than Cu-2, and Cu-1 is more cytotoxic than Cu-3. What's interesting is that Cu-1 can exhibit obvious inhibitory effect on the growth of human triple-negative breast cancer in vivo and vitro through anti-proliferative, anti-angiogenic, anti-inflammatory, pro-apoptotic and cuproptotic synergistic effects. Though Cu-3 shows no significant cytotoxicity against MDA-MB-231 cells, it can significantly inhibit the growth of SKOV3 cells in vitro by down-regulating the expression of some key proteins in the VEGF/VEGFR2 signaling pathway and the expression of some pro-inflammatory cytokines, and by disrupting the balance of intracellular reactive oxygen species levels.
Collapse
Affiliation(s)
- Ai-Qiu Liao
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Juan Wen
- Department of Pharmacy, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, 541001, China
| | - Jing-Chen Wei
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Bing-Bing Xu
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Nan Jin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Hong-Yu Lin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Xiu-Ying Qin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China.
| |
Collapse
|
42
|
Ahluwalia P, Mondal AK, Vashisht A, Singh H, Alptekin A, Ballur K, Omar N, Ahluwalia M, Jones K, Barrett A, Kota V, Kolhe R. Identification of a distinctive immunogenomic gene signature in stage-matched colorectal cancer. J Cancer Res Clin Oncol 2024; 151:9. [PMID: 39673574 PMCID: PMC11646222 DOI: 10.1007/s00432-024-06034-4] [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: 05/21/2024] [Accepted: 11/11/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Despite advances in diagnosis and treatment, including surgery, chemotherapy, and immunotherapy, accurate clinical markers are still lacking. The development of prognostic and predictive indicators, particularly in the context of personalized medicine, could significantly improve CRC patient management. METHOD In this retrospective study, we used FFPE blocks of tissue samples from CRC patients at Augusta University (AU) to quantify a custom 15-gene panel. To differentiate the tumor and adjacent normal regions (NAT), H&E staining was utilized. For the quantification of transcripts, we used the NanoString nCounter platform. Kaplan-Meier and Log-rank tests were used to perform survival analyses. Several independent datasets were explored to validate the gene signature. Orthogonal analyses included single-cell profiling, differential gene expression, immune cell deconvolution, neoantigen prediction, and biological pathway assessment. RESULTS A 3-gene signature (GTF3A, PKM, and VEGFA) was found to be associated with overall survival in the AU cohort (HR = 2.26, 95% CI 1.05-4.84, p = 0.02, 93 patients), TCGA cohort (HR = 1.57, 95% CI 1.05-2.35, p < 0.02, 435 patients) and four other GEO datasets. Independent single-cell analysis identified relatively higher expression of the 3-gene signature in the tumor region. Differential analysis revealed dysregulated tissue inflammation, immune dysfunction, and neoantigen load of cell cycle processes among high-risk patients compared to low-risk patients. CONCLUSION We developed a 3-gene signature with the potential for prognostic and predictive clinical assessment of CRC patients. This gene-based stratification offers a cost-effective approach to personalized cancer management. Further research using similar methods could identify therapy-specific gene signatures to strengthen the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashis K Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ahmet Alptekin
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | | | - Kimya Jones
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Amanda Barrett
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Vamsi Kota
- Georgia Cancer Center at Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA.
| |
Collapse
|
43
|
Jin C, Zhang F, Luo H, Li B, Jiang X, Pirozzi CJ, Liang C, Zhang M. The CCL5/CCR5/SHP2 axis sustains Stat1 phosphorylation and activates NF-κB signaling promoting M1 macrophage polarization and exacerbating chronic prostatic inflammation. Cell Commun Signal 2024; 22:584. [PMID: 39633456 PMCID: PMC11619290 DOI: 10.1186/s12964-024-01943-w] [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: 10/19/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Chronic prostatitis (CP) is a condition markered by persistent prostate inflammation, yet the specific cytokines driving its progression remain largely undefined. This study aims to identify key cytokines involved in CP and investigate their role in driving inflammatory responses through mechanistic and therapeutic exploration. METHODS A 48-cytokine panel test was conducted to compare the plasma cytokine profiles between participants with CP-like symptoms (CP-LS) and healthy controls. Experimental autoimmune prostatitis (EAP) models were used for functional validation, with further mechanistic studies performed through in vivo and in vitro assays. Pharmacological inhibition was applied using maraviroc, and pathway inhibitors to assess therapeutic potential. RESULTS Our analysis identified CCL5 as one of the most prominently elevated cytokines in CP-LS patients. Further validation in the EAP model mice confirmed elevated CCL5 levels, highlighting its role in driving prostatic inflammation. Mechanistic studies revealed that CCL5 interacts with the CCR5 receptor, promoting M1 macrophage polarization and activating key inflammatory signaling pathways, including Stat1 and NF-κB, as indicated by increased phosphorylation of Stat1 and p65. In vitro, CCL5 combined with LPS stimulation amplified these effects, further promoting M1 polarization. CCL5 also sustained Stat1 activation by inhibiting its dephosphorylation through reduced interaction with SHP2, leading to prolonged inflammatory signaling. Single-cell transcriptomics confirmed high CCR5 expression in macrophages, correlating with inflammatory pathways. Pharmacological inhibition of CCR5, or its downstream signaling, significantly reduced macrophage-driven inflammation both in vivo and in vitro. CONCLUSION These findings establish the CCL5/CCR5 axis as a critical driver of persistant prostatic inflammation and present it as a potential therapeutic target for CP.
Collapse
Affiliation(s)
- Chen Jin
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Fei Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China
| | - Hailang Luo
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China
| | - Boyang Li
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China
| | - Xue Jiang
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | | | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China.
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, P. R. China.
- Institute of Urology, Anhui Medical University, Hefei, Anhui, 230022, P. R. China.
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, 230022, P. R. China.
| |
Collapse
|
44
|
Liu J, Wei Z, Meng L, Wu L, Liu F, Sang M, Zhao L, Gu L, Shan B. CircJPH1 regulates the NF-κB/HERC5 axis to promote the malignant progression of esophageal squamous cell carcinoma through binding to XRCC6. Cell Signal 2024; 124:111403. [PMID: 39255925 DOI: 10.1016/j.cellsig.2024.111403] [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: 06/19/2024] [Revised: 09/02/2024] [Accepted: 09/07/2024] [Indexed: 09/12/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent and malignant cancer with an unknown pathogenesis and a poor prognosis; therefore, the identification of effective biomarkers and targets is crucial for its diagnosis and treatment. Circular (circ)RNAs are prominent functional biomarkers and therapeutic targets in various diseases, particularly cancer, due to their widespread expression and regulatory mechanisms. Our study aimed to investigate the therapeutic potential of circRNA for ESCC. We identified Hsa_circ_0137111 for the first time as one of the most significantly up-regulated genes in ESCC sequencing and named it circJPH1. The results of the present study demonstrated an enhanced expression of circJPH1 in ESCC tissues. Moreover, circJPH1-knockdown could significantly inhibit the proliferation, migration, and invasion of ESCC cells, while its overexpression promoted these characteristics. In addition, circJPH1 promoted ESCC cell tumor growth in vivo. For the first time, mass spectrometry and RNA pull-down analysis revealed the interaction of X-ray repair cross-complementary 6 (XRCC6) protein with circJPH1, thereby promoting its nuclear translocation. Consequently, the nuclear factor kappa-B (NF-κB) signaling pathway was activated, leading to an up-regulation of HECT and RLD domain containing E3 ubiquitin protein ligase 5 (HERC5), thereby promoting ESCC progression. In summary, the present study elucidated the regulatory impact of circJPH1 on ESCC progression in vitro and in vivo, thereby indicating its potential role in ESCC treatment.
Collapse
Affiliation(s)
- Jingjing Liu
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Zishuan Wei
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Lingjiao Meng
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Lixia Wu
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Fei Liu
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Meixiang Sang
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Lianmei Zhao
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China
| | - Lina Gu
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China.
| | - Baoen Shan
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050017, China; Key Laboratory of Tumor Gene Diagnosis, Prevention and Therapy, Clinical Oncology Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province 050001, China.
| |
Collapse
|
45
|
Thiruvengadam R, Dareowolabi BO, Moon EY, Kim JH. Nanotherapeutic strategy against glioblastoma using enzyme inhibitors. Biomed Pharmacother 2024; 181:117713. [PMID: 39615164 DOI: 10.1016/j.biopha.2024.117713] [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: 07/22/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Glioblastoma is the most aggressive brain cancer and thus patients with glioblastoma have a severely low 5-year survival rate (<5 %). Glioblastoma damages neural centers, causing severe depression, anxiety, and cognitive disorders. Glioblastoma is highly resistant to most of available anti-tumor medications, due to heterogeneity of glioblastoma as well as the presence of stem-like cells. To overcome the challenges in the current medications against glioblastoma, novel medications that are effective in treating the aggressive and heterogenous glioblastoma should be developed. Enzyme inhibitor and nanomedicine have been getting attention because of effective anticancer efficacies of enzyme inhibitors and a role of nanomedicine as effective carrier of chemotherapeutic drugs by targeting specific tumor areas. Furthermore, a tumor-initiating neuroinflammatory microenvironment, which is crucial for glioblastoma progression, was linked with several carcinogenesis pathways. Therefore, in this review, first we summarize neuroinflammation and glioblastoma-related neuropathways. Second, we discuss the importance of enzyme inhibitors targeting specific proteins in relation with neuroinflammation and glioblastoma-related molecular mechanisms. Third, we summarize recent findings on the significance of nanotherapeutic anticancer drugs developed using natural or synthetic enzyme inhibitors against glioblastoma as well as currently available Food and Drug Administration (FDA)-approved drugs against glioblastoma.
Collapse
Affiliation(s)
- Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | | | - Eun-Yi Moon
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
46
|
Utpal BK, Dehbia Z, Zidan BMRM, Sweilam SH, Singh LP, Arunkumar MS, Sona M, Panigrahy UP, Keerthana R, Mandadi SR, Rab SO, Alshehri MA, Koula D, Suliman M, Nafady MH, Emran TB. Carotenoids as modulators of the PI3K/Akt/mTOR pathway: innovative strategies in cancer therapy. Med Oncol 2024; 42:4. [PMID: 39549201 DOI: 10.1007/s12032-024-02551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/29/2024] [Indexed: 11/18/2024]
Abstract
Cancer progression is primarily driven by the uncontrolled activation of cellular signaling pathways, with the PI3K/Akt/mTOR (PAMT) pathway playing a central role. This pathway significantly contributes to the proliferation and survival of cancer cells, and its hyperactivity is a major challenge in managing several types of malignancies. This article delves into the promising potential of carotenoids, natural pigments found in abundance in fruits and vegetables, as a novel therapeutic strategy for cancer treatment. By specifically targeting and inhibiting the PAMT pathway, carotenoids may effectively disrupt the growth and survival of cancer cells. The article examines the complex mechanisms underlying these interactions and highlights the obstacles faced in cancer treatment. It proposes a compelling approach to developing therapies that leverage natural products to target this critical pathway, offering a fresh perspective on cancer treatment. Further research is essential to enhance the therapeutic efficacy of these compounds.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Zerrouki Dehbia
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - B M Redwan Matin Zidan
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of Pharmacy, Gopal Narayan Singh University, Sasaram (Rohtas) Bihar, Jamuhar, 821305, India
| | - M S Arunkumar
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - M Sona
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Uttam Prasad Panigrahy
- Faculty of Pharmaceutical Science, Assam Down Town University, Gandhi Nagar, Sankar Madhab Path, Panikhaiti, Guwahati, Assam, India
| | - R Keerthana
- Faculty of Pharmacy, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, 641021, India
| | - Sandhya Rani Mandadi
- Department of Pharmaceutics, Vishnu Institute of Pharmaceutical Education and Research, Tuljaraopet, Telangana , 502313, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Doukani Koula
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
- Laboratory of Animal Production Sciences and Techniques, University of Abdelhamid Ibn Badis, Mostaganem, Algeria
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohamed H Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza, 12568, Egypt.
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|
47
|
Hareeri RH, Hofni A. Berberine Alleviates Uterine Inflammation in Rats via Modulating the TLR-2/p-PI3K/p-AKT Axis. Int Immunopharmacol 2024; 141:112931. [PMID: 39146781 DOI: 10.1016/j.intimp.2024.112931] [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: 06/04/2024] [Revised: 08/02/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Uterine inflammation affects 8% of women in the United States and 32% in developing nations, often caused by uncontrolled inflammation and oxidative stress. This condition significantly impacts women's health, productivity, and quality of life, and increases the risk of related morbidities leading to higher healthcare costs. Research now focuses on natural antioxidants and anti-inflammatory, particularly berberine (BBR), an isoquinoline alkaloid known for its antioxidant, anti-inflammatory, and antiapoptotic activities. The present study sought to examine the potential therapeutic efficacy of BBR against uterine inflammation induced by the intrauterine infusion of an iodine (I2) mixture in an experimental setting. Female Sprague Dawley rats (n = 6) were divided into five groups, control, sham, I2, I2 and BBR 10 mg/kg, and I2 and BBR 25 mg/kg-treated groups. Compared to I2 infusion, BBR treatment effectively restored normal uterine histopathology and reduced inflammatory markers such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), nuclear factor- kappa B (NF-κB), monocyte chemoattractant protein 1 (MCP1), and myeloperoxidase (MPO). It lowered oxidative markers like malondialdehyde (MDA), and increased antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD). It balanced apoptotic genes by upregulating B-cell lymphoma 2 (Bcl-2) and downregulating Bcl-2-associated X protein (Bax). Furthermore, BBR reduced the expression of Toll-like receptor 2 (TLR-2), phosphorylated phosphatidylinositol 3‑kinase (p-PI3K), and phosphorylated protein kinase B (p-AKT) in the rats treated with intrauterine I2. Ultimately, the therapeutic benefits of BBR can be attributed, to some extent, to its antioxidant, anti-inflammatory, and antiapoptotic properties, in addition to its ability to modulate the TLR-2/p-PI3K/p-AKT axis.
Collapse
Affiliation(s)
- Rawan H Hareeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Amal Hofni
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt
| |
Collapse
|
48
|
Compton SLE, Yang S, Maniscalco LS, Muhsen RA, Shrestha P, Wu X, Woodard KT, Zunica ERM, Cho E, Wall RL, Brown J, Jayaraman A, Kirby BJ, Gilmore LA, Greenway FL, Spielmann G, Brown JC. A randomized trial of aerobic exercise in colorectal cancer: Rationale, design, recruitment, and exercise adherence results. Contemp Clin Trials 2024; 146:107702. [PMID: 39362405 PMCID: PMC11531371 DOI: 10.1016/j.cct.2024.107702] [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: 04/21/2024] [Revised: 07/20/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Physical activity is associated with improved disease-free survival in colorectal cancer survivors. This report describes the purpose, design, recruitment, and exercise adherence results of the National Cancer Institute (NCI)-sponsored Exercise and Colorectal Cancer Treatment (EXACT) trial. METHODS The primary objective of the EXACT trial is to determine if randomization to 150 min per week of moderate-intensity aerobic exercise reduces systemic inflammation among stage I-III colorectal cancer survivors compared with a waitlist control group over 12 weeks. Participants were provided with an in-home treadmill and heart rate monitor. Characteristics associated with randomization were identified using χ2 or Fisher's exact test for categorical variables and t-tests or analysis of covariance (ANCOVA). Exercise adherence was calculated as the total minutes exercised by total minutes prescribed. RESULTS Between August 2019 and February 2023, 3082 colorectal cancer survivors were invited to participate, 89 were screened, and 60 were randomized to the study protocol. Younger age (P = 0.02), female sex (P = 0.002), white race (P = 0.01), proximal time since tumor resection (P = 0.02), and regional tumor stage (P < 0.001) were associated with study participation. Average exercise adherence was 92.2 % (95 % CI: 85.5, 98.8) and all study participants achieved ≥80 % exercise adherence. Endpoint data collection was completed for all participants in May 2023. CONCLUSION The results from the EXACT trial will characterize the changes that occur from exercise to advance our understanding of the biological mechanisms by which exercise may prevent tumor recurrence and death in colorectal cancer survivors.
Collapse
Affiliation(s)
| | - Shengping Yang
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA
| | - Lauren S Maniscalco
- LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Reem A Muhsen
- LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Pratibha Shrestha
- LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Xiaocheng Wu
- LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Kaylee T Woodard
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA; LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA
| | - Elizabeth R M Zunica
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA
| | - Eunhan Cho
- School of Kinesiology, College of Human Sciences & Education, Louisiana State University, Baton Rouge, LA 70802, USA
| | - Rachel L Wall
- School of Kinesiology, College of Human Sciences & Education, Louisiana State University, Baton Rouge, LA 70802, USA
| | - John Brown
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA; Louisiana State University Health Shreveport, School of Medicine, 1501 Kings Hwy., Shreveport, LA 71103, USA
| | | | - Brian J Kirby
- Cornell University, 377 Kimball Hall, Ithaca, NY 14853, USA
| | - L Anne Gilmore
- UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Frank L Greenway
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA
| | - Guillaume Spielmann
- School of Kinesiology, College of Human Sciences & Education, Louisiana State University, Baton Rouge, LA 70802, USA
| | - Justin C Brown
- Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA; LSU Health Sciences Center, New Orleans School of Public Health, Louisiana Tumor Registry, 2020 Gravier St., New Orleans, LA 70112, USA; Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 533 Bolivar St., New Orleans, LA 70112, USA; Louisiana Cancer Research Center, 1700 Tulane Ave., New Orleans, LA 70112, USA.
| |
Collapse
|
49
|
Cao Y, Yi Y, Han C, Shi B. NF-κB signaling pathway in tumor microenvironment. Front Immunol 2024; 15:1476030. [PMID: 39493763 PMCID: PMC11530992 DOI: 10.3389/fimmu.2024.1476030] [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: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
The genesis and progression of tumors are multifaceted processes influenced by genetic mutations within the tumor cells and the dynamic interplay with their surrounding milieu, which incessantly impacts the course of cancer. The tumor microenvironment (TME) is a complex and dynamic entity that encompasses not only the tumor cells but also an array of non-cancerous cells, signaling molecules, and the extracellular matrix. This intricate network is crucial in tumor progression, metastasis, and response to treatments. The TME is populated by diverse cell types, including immune cells, fibroblasts, endothelial cells, alongside cytokines and growth factors, all of which play roles in either suppressing or fostering tumor growth. Grasping the nuances of the interactions within the TME is vital for the advancement of targeted cancer therapies. Consequently, a thorough understanding of the alterations of TME and the identification of upstream regulatory targets have emerged as a research priority. NF-κB transcription factors, central to inflammation and innate immunity, are increasingly recognized for their significant role in cancer onset and progression. This review emphasizes the crucial influence of the NF-κB signaling pathway within the TME, underscoring its roles in the development and advancement of cancer. By examining the interactions between NF-κB and various components of the TME, targeting the NF-κB pathway appears as a promising cancer treatment approach.
Collapse
Affiliation(s)
- Yaning Cao
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Yanan Yi
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Chongxu Han
- Department of Laboratory Medicine, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingwei Shi
- Department of Blood Transfusion, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| |
Collapse
|
50
|
Hosseini FS, Nikparast A, Etesami E, Javaheri-Tafti F, Asghari G. The association between empirical dietary inflammatory pattern and risk of cancer and cancer-specific mortality: a systematic review and meta-analysis of prospective cohort studies. Front Nutr 2024; 11:1462931. [PMID: 39494310 PMCID: PMC11527705 DOI: 10.3389/fnut.2024.1462931] [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: 07/10/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Background/aim Current evidence indicates a correlation between the inflammatory potential of diet and the risk of cancer and cancer-specific mortality. This study aimed to assess the association between empirical dietary inflammatory pattern (EDIP), which has recently been designed based on the inflammatory potential of the diet, and the risk of cancer and cancer-specific mortality. Methods A systematic literature search was conducted across the PubMed/Medline, Scopus, and Web of Science databases from January 2016 to March 2024. A random effects model was used to calculate the pooled effect size (ES) and 95% confidence intervals (95% CI). Heterogeneity between studies was assessed using the Cochran Q test and the I 2 statistic. Results From the initial 229 records, 24 prospective cohort studies with 2,683,350 participants and 37,091 cancer incidence cases, as well as 20,819 cancer-specific mortality, were included in our study. Pooled results indicated a significant association between higher adherence to the EDIP and an increased risk of total cancer (ES: 1.10; 95% CI: 1.05-1.15; I 2 = 41.1), colorectal cancer (ES: 1.19; 95% CI: 1.11-1.27; I 2 = 41.1), and liver cancer (ES: 1.48; 95% CI: 1.14-1.94; I 2 = 36.9). However, no significant association between increased adherence to the EDIP and an increased risk of ovarian or endometrial cancer was found. Furthermore, greater adherence to the EDIP was significantly associated with an increased risk of cancer-specific mortality (ES: 1.18; 95% CI: 1.05-1.33; I 2 = 45.4). Conclusion Our results showed that a diet with higher inflammatory properties is associated with an increased risk of cancer and cancer-specific mortality. Systematic review registration PROSPERO registration no. CRD42024496912.
Collapse
Affiliation(s)
- Fatemeh S. Hosseini
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Nikparast
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Etesami
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Javaheri-Tafti
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Golaleh Asghari
- Department of Clinical Nutrition & Dietetics, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|