51
|
Tian Y, Zhang L, Ping Y, Zhang Z, Yao C, Shen C, Li F, Wen C, Zhang Y. CCR5 and IL-12 co-expression in CAR T cells improves antitumor efficacy by reprogramming tumor microenvironment in solid tumors. Cancer Immunol Immunother 2025; 74:55. [PMID: 39751840 PMCID: PMC11699016 DOI: 10.1007/s00262-024-03909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy for solid tumors faces significant challenges, including inadequate infiltration, limited proliferation, diminished effector function of CAR T cells, and an immunosuppressive tumor microenvironment (TME). In this study, we utilized The Cancer Genome Atlas database to identify key chemokines (CCL4, CCL5, and CCR5) associated with T cell infiltration across various solid tumor types. The CCL4/CCL5-CCR5 axis emerged as significantly correlated with the presence of T cells within tumors, and enhancing the expression of CCR5 in CAR T cells bolstered their migratory capacity. Furthermore, single-cell immunoprofiling of tumor tissues revealed that macrophages within the TME primarily interact with CD8+ T cells, impeding their tumor response. However, CAR T cells engineered to secrete Interleukin (IL)-12 can counteract macrophage-mediated immunosuppression and augment T cell functionality. To address these obstacles, we employed esophageal carcinoma as a model to develop mesothelin-targeted CAR T cells co-expressing CCR5 and IL-12 (CARTmeso-5-12), subsequently assessing their antitumor capabilities in vitro and in vivo. The CARTmeso-5-12 cells demonstrated enhanced tumor infiltration due to overexpression of CCR5, and IL-12 secretion further amplified CAR T cell efficacy by attenuating the suppressive influence of tumor-infiltrating macrophages, thus improving tumor eradication.
Collapse
Affiliation(s)
- Yonggui Tian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chang Yao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunyi Shen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chunli Wen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
52
|
Lin S, He J, Zhou Y, Bao Y, Feng X, Cheng H, Cai H, Hu S, Wang L, Zheng Y, Zhang M, Fan Q, Wen S, Lin Y, Liu C, Chen X, Wang F, Ge X, Yang X. Cross-sectional and Longitudinal Associations Between Metal Mixtures and Serum C3, C4: Result from the Manganese‑exposed Workers Healthy Cohort. Biol Trace Elem Res 2025; 203:18-29. [PMID: 38492120 DOI: 10.1007/s12011-024-04143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Exposure to metal mixtures compromises the immune system, with the complement system connecting innate and adaptive immunity. Herein, we sought to explore the relationships between blood cell metal mixtures and the third and fourth components of serum complement (C3, C4). A total of 538 participants were recruited in November 2017, and 289 participants were followed up in November 2021. We conducted a cross-sectional analysis at baseline and a longitudinal analysis over 4 years. Least Absolute Shrinkage and Selection Operator (LASSO) was employed to identify the primary metals related to serum C3, C4; generalized linear model (GLM) was further used to evaluate the cross-sectional associations of the selected metals and serum C3, C4. Furthermore, participants were categorized into three groups according to the percentage change in metal concentrations over 4 years. GLM was performed to assess the associations between changes in metal concentrations and changes in serum C3, C4 levels. At baseline, each 1-unit increase in log10-transformed in magnesium, manganese, copper, rubidium, and lead was significantly associated with a change in serum C3 of 0.226 (95% CI: 0.146, 0.307), 0.055 (95% CI: 0.022, 0.088), 0.113 (95% CI: 0.019, 0.206), - 0.173 (95% CI: - 0.262, - 0.083), and - 0.020 (95% CI: - 0.039, - 0.001), respectively. Longitudinally, decreased copper concentrations were negatively associated with an increment in serum C3 levels, while decreased lead concentrations were positively associated with an increment in serum C3 levels. However, no metal was found to be primarily associated with serum C4 in LASSO, so we did not further explore the relationship between them. Our research indicates that copper and lead may affect complement system homeostasis by influencing serum C3 levels. Further investigation is necessary to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Sencai Lin
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Junxiu He
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Yinghua Zhou
- School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China
| | - Yu Bao
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xiuming Feng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Hong Cheng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Haiqing Cai
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Sihan Hu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Lin Wang
- School of Science, Guangxi University of Science and Technology, Liuzhou, 545006, Guangxi, China
| | - Yuan Zheng
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Mengdi Zhang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Qinghua Fan
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Shifeng Wen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Yuanxin Lin
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Chaoqun Liu
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xing Chen
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Fei Wang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoting Ge
- School of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China.
| | - Xiaobo Yang
- School of Public Health, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
53
|
Gorla M, Guleria DS. Rho GTPase Signaling: A Molecular Switchboard for Regulating the Actin Cytoskeleton in Axon Guidance. J Cell Physiol 2025; 240:e70005. [PMID: 39888031 DOI: 10.1002/jcp.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/11/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
Axon pathfinding is a highly dynamic process regulated by the interactions between cell-surface guidance receptors and guidance cues present in the extracellular environment. During development, precise axon pathfinding is crucial for the formation of functional neural circuits. The spatiotemporal expression of axon guidance receptors helps the navigating axon make correct decisions in a complex environment comprising both attractive and repulsive guidance cues. Axon guidance receptors initiate distinct signaling cascades that eventually influence the cytoskeleton at the growing tip of an axon, called the growth cone. The actin cytoskeleton is the primary target of these guidance signals and plays a key role in growth cone motility, exploration, and behavior. Of the many regulatory molecules that modulate the actin cytoskeleton in response to distinct guidance signals, Rho GTPases play central roles. Rho GTPases are molecular switchboards; their ON (GTP-bound) and OFF (GDP-bound) switches are controlled by their interactions with proteins that regulate the exchange of GDP for GTP or with the proteins that promote GTP hydrolysis. Various upstream signals, including axon guidance signals, regulate the activity of these Rho GTPase switch regulators. As cycling molecular switches, Rho GTPases interact with and control the activities of downstream effectors, which directly influence actin reorganization in a context-dependent manner. A deeper exploration of the spatiotemporal dynamics of Rho GTPase signaling and the molecular basis of their involvement in regulating growth cone actin cytoskeleton can unlock promising therapeutic strategies for neurodevelopmental disorders linked to dysregulated Rho GTPase signaling. This review not only provides a comprehensive overview of the field but also highlights recent discoveries that have considerably advanced our understanding of the complex regulatory roles of Rho GTPases in modulating actin cytoskeleton arrangement at the growth cone during axon guidance.
Collapse
Affiliation(s)
- Madhavi Gorla
- National Institute of Animal Biotechnology, Hyderabad, India
| | | |
Collapse
|
54
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
55
|
Ma R, Liang S, Zeng W, Li J, Lai Y, Yang X, Diao F. Single-cell RNA sequencing reveals the important role of Dcaf17 in spermatogenesis of golden hamsters†. Biol Reprod 2024; 111:1326-1340. [PMID: 39239833 DOI: 10.1093/biolre/ioae132] [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: 03/08/2024] [Revised: 07/10/2024] [Accepted: 09/05/2024] [Indexed: 09/07/2024] Open
Abstract
Dcaf17, also known as DDB1- and CUL4-associated factor 17, is a member of the DCAF family and acts as the receptor for the CRL4 ubiquitin E3 ligase complex. Several previous studies have reported that mutations in Dcaf17 cause Woodhouse-Sakati syndrome, which results in oligoasthenoteratozoospermia and male infertility. As a model to explore the role of Dcaf17 in the male reproductive system, we created Dcaf17-deficient male golden hamsters using CRISPR-Cas9 technology; the results of which demonstrate that deletion of Dcaf17 led to abnormal spermatogenesis and infertility. To uncover the underlying molecular mechanisms involved, we conducted single cell Ribonucleic Acid sequencing analysis to evaluate the effect of Dcaf17 deficiency on transcriptional levels in spermatogenic cells during various stages of spermatogenesis. These data emphasize the significant regulatory role played by Dcaf17 in early spermatogenic cells, with many biological processes being affected, including spermatogenesis and protein degradation. Dysregulation of genes associated with these functions ultimately leads to abnormalities. In summary, our findings highlight the critical function of Dcaf17 in spermatogenesis and clarify the specific stage at which Dcaf17 exerts its effects, while simultaneously providing a novel animal model for the study of Dcaf17.
Collapse
Affiliation(s)
- Rongzhu Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Jiangsu Province, China
| | - Shuang Liang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing China
| | - Wentao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing China
| | - Jianmin Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing China
| | - Yana Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing China
| | - Xiaoyu Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Jiangsu Province, China
| | - Feiyang Diao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Jiangsu Province, China
| |
Collapse
|
56
|
Winge MCG, Nasrallah M, Jackrazi LV, Guo KQ, Fuhriman JM, Szafran R, Ramanathan M, Gurevich I, Nguyen NT, Siprashvili Z, Inayathullah M, Rajadas J, Porter DF, Khavari PA, Butte AJ, Marinkovich MP. Repurposing an epithelial sodium channel inhibitor as a therapy for murine and human skin inflammation. Sci Transl Med 2024; 16:eade5915. [PMID: 39661704 DOI: 10.1126/scitranslmed.ade5915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/12/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024]
Abstract
Inflammatory skin disease is characterized by a pathologic interplay between skin cells and immunocytes and can result in disfiguring cutaneous lesions and systemic inflammation. Immunosuppression is commonly used to target the inflammatory component; however, these drugs are often expensive and associated with side effects. To identify previously unidentified targets, we carried out a nonbiased informatics screen to identify drug compounds with an inverse transcriptional signature to keratinocyte inflammatory signals. Using psoriasis, a prototypic inflammatory skin disease, as a model, we used pharmacologic, transcriptomic, and proteomic characterization to find that benzamil, the benzyl derivative of the US Food and Drug Administration-approved diuretic amiloride, effectively reversed keratinocyte-driven inflammatory signaling. Through three independent mouse models of skin inflammation (Rac1G12V transgenic mice, topical imiquimod, and human skin xenografts from patients with psoriasis), we found that benzamil disrupted pathogenic interactions between the small GTPase Rac1 and its adaptor NCK1. This reduced STAT3 and NF-κB signaling and downstream cytokine production in keratinocytes. Genetic knockdown of sodium channels or pharmacological inhibition by benzamil prevented excess Rac1-NCK1 binding and limited proinflammatory signaling pathway activation in patient-derived keratinocytes without systemic immunosuppression. Both systemic and topical applications of benzamil were efficacious, suggesting that it may be a potential therapeutic avenue for treating skin inflammation.
Collapse
Affiliation(s)
- Mårten C G Winge
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mazen Nasrallah
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leandra V Jackrazi
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Konnie Q Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica M Fuhriman
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca Szafran
- Unit of Dermatology, ME GHR, Karolinska University Hospital, SE-17176 Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Muthukumar Ramanathan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irina Gurevich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ngon T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory, Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Douglas F Porter
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - M Peter Marinkovich
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Dermatology Service, Veterans Affairs Medical Center, Palo Alto, CA 94304, USA
| |
Collapse
|
57
|
Xing X, Zhou Z, Peng H, Cheng S. Anticancer role of flubendazole: Effects and molecular mechanisms (Review). Oncol Lett 2024; 28:558. [PMID: 39355784 PMCID: PMC11443308 DOI: 10.3892/ol.2024.14691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Flubendazole, an anthelmintic agent with a well-established safety profile, has emerged as a promising anticancer drug that has demonstrated efficacy against a spectrum of cancer types over the past decade. Its anticancer properties encompass a multifaceted mechanism of action, including the inhibition of cancer cell proliferation, disruption of microtubule dynamics, regulation of cell cycle, autophagy, apoptosis, suppression of cancer stem cell characteristics, promotion of ferroptosis and inhibition of angiogenesis. The present review aimed to provide a comprehensive overview of the molecular underpinnings of the anticancer activity of flubendazole, highlighting key molecules and regulatory pathways. Given the breadth of the potential of flubendazole, further research is imperative to identify additional cancer types sensitive to flubendazole, refine experimental methodologies for enhancing its reliability, uncover synergistic drug combinations, improve its bioavailability and explore innovative administration methods. The present review provided a foundation for future studies on the role of flubendazole in oncology and described its molecular mechanisms of action.
Collapse
Affiliation(s)
- Xing Xing
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Zongning Zhou
- Human Genetic Resources Preservation Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Hongwei Peng
- Human Genetic Resources Preservation Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
- Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Shaoping Cheng
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
58
|
Grützmann K, Kraft T, Meinhardt M, Meier F, Westphal D, Seifert M. Network-based analysis of heterogeneous patient-matched brain and extracranial melanoma metastasis pairs reveals three homogeneous subgroups. Comput Struct Biotechnol J 2024; 23:1036-1050. [PMID: 38464935 PMCID: PMC10920107 DOI: 10.1016/j.csbj.2024.02.013] [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: 11/06/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 03/12/2024] Open
Abstract
Melanoma, the deadliest form of skin cancer, can metastasize to different organs. Molecular differences between brain and extracranial melanoma metastases are poorly understood. Here, promoter methylation and gene expression of 11 heterogeneous patient-matched pairs of brain and extracranial metastases were analyzed using melanoma-specific gene regulatory networks learned from public transcriptome and methylome data followed by network-based impact propagation of patient-specific alterations. This innovative data analysis strategy allowed to predict potential impacts of patient-specific driver candidate genes on other genes and pathways. The patient-matched metastasis pairs clustered into three robust subgroups with specific downstream targets with known roles in cancer, including melanoma (SG1: RBM38, BCL11B, SG2: GATA3, FES, SG3: SLAMF6, PYCARD). Patient subgroups and ranking of target gene candidates were confirmed in a validation cohort. Summarizing, computational network-based impact analyses of heterogeneous metastasis pairs predicted individual regulatory differences in melanoma brain metastases, cumulating into three consistent subgroups with specific downstream target genes.
Collapse
Affiliation(s)
- Konrad Grützmann
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Theresa Kraft
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
| | - Matthias Meinhardt
- Department of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| | - Dana Westphal
- Department of Dermatology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry, Faculty of Medicine, TU Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), D-01307 Dresden, Germany
| |
Collapse
|
59
|
Berry JS, Tarn J, Casement J, Lendrem D, Thompson K, Mariette X, Gottenberg JE, Ng WF. Stability of symptom-based subtypes in Sjogren's disease. RMD Open 2024; 10:e004914. [PMID: 39581689 PMCID: PMC11590857 DOI: 10.1136/rmdopen-2024-004914] [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: 08/26/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024] Open
Abstract
OBJECTIVES The Newcastle Sjogren's Stratification Tool (NSST) stratifies Sjogren's disease patients into four subtypes. Understanding the stability of the subtypes is vital if symptom-based stratification is to be more broadly adopted. In this study, we stratify patients longitudinally to understand how symptom-based subtypes vary over time and factors influencing subtype change. METHODS 274 patients from the United Kingdom Primary Sjögren's Syndrome Registry (UKPSSR) with data permitting NSST subtype assignment from two study visits were included. The French Assessment of Systemic Signs and Evolution of Sjogren's Syndrome (ASSESS) cohort (n=237) acted as an independent comparator. Group analyses of significant differences were performed, with logistic regression models used to assess covariates of subtype stability. RESULTS UKPSSR and ASSESS cohorts showed a broadly similar proportion of subjects in each subtype and similar baseline clinical characteristics except body mass index (BMI). Several baseline characteristics differ significantly between the subtypes, most notably anti-Ro status and BMI. Subtype membership was reasonably stable in both cohorts with 60% and 57% retaining subtype. The high-symptom burden subtype was the most stable over time with 70% and 67% retaining subtype. Higher baseline probability score was the greatest predictor of subtype stability with higher C4 levels, antidepressant use, and a higher CCI score also predicting increased stability. CONCLUSION NSST subtype membership remains stable over time in a large proportion of patients. When subtype transition is associated with factors at baseline, it is most strongly associated with an uncertain subtype allocation. Our findings support the hypothesis that symptom-based subtypes reflect genuine pathobiological endotypes and therefore maybe important to consider in trial design and clinical management.
Collapse
Affiliation(s)
- Joe Scott Berry
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
- Clinical and Translational Medicine Research Centre, University of Sunderland, Sunderland, UK
| | - Jessica Tarn
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - John Casement
- Bioinformatics Support Unit, Newcastle University, Newcastle upon Tyne, UK
| | - Dennis Lendrem
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Kyle Thompson
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Xavier Mariette
- Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux universitaires Paris-Sud – Hôpital Bicêtre, Le Kremlin Bicêtre, France
- Université Paris-Sud, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | | | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
- HRB Clinical Research Facility, University College Cork, Cork, Ireland
| |
Collapse
|
60
|
Zhou Y, Song L, Li H. Full-resolution HLA and KIR gene annotations for human genome assemblies. Genome Res 2024; 34:1931-1941. [PMID: 38839374 PMCID: PMC11610593 DOI: 10.1101/gr.278985.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
The human leukocyte antigen (HLA) genes and the killer cell immunoglobulin-like receptor (KIR) genes are critical to immune responses and are associated with many immune-related diseases. Located in highly polymorphic regions, it is difficult to study them with traditional short-read alignment-based methods. Although modern long-read assemblers can often assemble these genes, using existing tools to annotate HLA and KIR genes in these assemblies remains a nontrivial task. Here, we describe Immuannot, a new computation tool to annotate the gene structures of HLA and KIR genes and to type the allele of each gene. Applying Immuannot to 56 regional and 212 whole-genome assemblies from previous studies, we annotate 9931 HLA and KIR genes and found that almost half of these genes, 4068, have novel sequences compared with the current Immuno Polymorphism Database (IPD). These novel gene sequences are represented by 2664 distinct alleles, some of which contained nonsynonymous variations, resulting in 92 novel protein sequences. We demonstrate the complex haplotype structures at the two loci and report the linkage between HLA/KIR haplotypes and gene alleles. We anticipate that Immuannot will speed up the discovery of new HLA/KIR alleles and enable the association of HLA/KIR haplotype structures with clinical outcomes in the future.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Li Song
- Department of Biomedical Data Science, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Heng Li
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA;
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
61
|
Cho CH, Sim WJ, Cho NC, Lim W, Lim TG. Structure-based virtual screening of natural compounds in preventing skin senescence: The role of epigallocatechin gallate in protein kinase C alpha-specific inhibition against UV-induced photoaging. Heliyon 2024; 10:e39933. [PMID: 39553571 PMCID: PMC11567019 DOI: 10.1016/j.heliyon.2024.e39933] [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: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
This study combines high-throughput screening and virtual molecular docking to identify natural compounds targeting PKC in skin aging. Go 6983, a PKC inhibitor, showed potent suppression of MMP-1 transcription. EGCG was one of the candidates that showed it could significantly lower UVB-induced MMP-1 expression in HaCaT cells, and it had a strong affinity for PKCα. Interestingly, EGCG is exclusively bound to PKCα, not the δ and ζ isoforms. Blocking PKCα did not elevate UVB-induced MMP-1 expression in HaCaT cells. In a model of human skin, EGCG stopped collagen breakdown and changes in epidermal thickness that were caused by UV light from the sun. This suggests that EGCG could be useful in dermatology and drug development. These findings highlight the role of structure-based screening in identifying candidate compounds with applications in the cosmetic, dermatological, preventive health, and pharmaceutical fields.
Collapse
Affiliation(s)
- Cheol Hyeon Cho
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Woo-Jin Sim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Nam-Chul Cho
- Korea Chemical Bank, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Wonchul Lim
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
62
|
Shi Y, Shi M, Wang Y, You J. Progress and prospects of mRNA-based drugs in pre-clinical and clinical applications. Signal Transduct Target Ther 2024; 9:322. [PMID: 39543114 PMCID: PMC11564800 DOI: 10.1038/s41392-024-02002-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 11/17/2024] Open
Abstract
In the last decade, messenger ribonucleic acid (mRNA)-based drugs have gained great interest in both immunotherapy and non-immunogenic applications. This surge in interest can be largely attributed to the demonstration of distinct advantages offered by various mRNA molecules, alongside the rapid advancements in nucleic acid delivery systems. It is noteworthy that the immunogenicity of mRNA drugs presents a double-edged sword. In the context of immunotherapy, extra supplementation of adjuvant is generally required for induction of robust immune responses. Conversely, in non-immunotherapeutic scenarios, immune activation is unwanted considering the host tolerability and high expression demand for mRNA-encoded functional proteins. Herein, mainly focused on the linear non-replicating mRNA, we overview the preclinical and clinical progress and prospects of mRNA medicines encompassing vaccines and other therapeutics. We also highlight the importance of focusing on the host-specific variations, including age, gender, pathological condition, and concurrent medication of individual patient, for maximized efficacy and safety upon mRNA administration. Furthermore, we deliberate on the potential challenges that mRNA drugs may encounter in the realm of disease treatment, the current endeavors of improvement, as well as the application prospects for future advancements. Overall, this review aims to present a comprehensive understanding of mRNA-based therapies while illuminating the prospective development and clinical application of mRNA drugs.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Meixing Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China
| | - Yi Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, P. R. China.
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, P. R. China.
- The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, P. R. China.
- Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, P. R. China.
| |
Collapse
|
63
|
Maiti S, Rangarajan A, Kareenhalli V. Experimentally-driven mathematical model to understand the effects of matrix deprivation in breast cancer metastasis. NPJ Syst Biol Appl 2024; 10:132. [PMID: 39532909 PMCID: PMC11557960 DOI: 10.1038/s41540-024-00443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Normal epithelial cells receive proper signals for growth and survival from attachment to the underlying extracellular matrix (ECM). They perceive detachment from the ECM as a stress and die - a phenomenon termed as 'anoikis'. However, metastatic cancer cells acquire anoikis-resistance and circulate through the blood and lymphatics to seed metastasis. Under normal (adherent) growth conditions, the serine-threonine protein kinase Akt stimulates protein synthesis and cell growth, maintaining an anabolic state in the cancer cell. In contrast, previously we showed that the stress due to matrix deprivation is sensed by yet another serine-threonine kinase, AMP-activated protein kinase (AMPK), that inhibits anabolic pathways while promoting catabolic processes. We illustrated a switch from Akthigh/AMPKlow in adherent condition to AMPKhigh/Aktlow in matrix-detached condition, with consequent metabolic switching from an anabolic to a catabolic state, which aids cancer cell stress-survival. In this study, we utilized these experimental data and developed a deterministic ordinary differential equation (ODE)-based mechanistic mathematical model to mimic attachment-detachment signaling network. To do so, we used the framework of insulin-glucagon signaling with consequent metabolic shifts to capture the pathophysiology of matrix-deprived state in breast cancer cells. Using the developed metastatic breast cancer signaling (MBCS) model, we identified perturbation of several signaling proteins such as IRS, PI3K, PKC, GLUT1, IP3, DAG, PKA, cAMP, and PDE3 upon matrix deprivation. Further, in silico molecular perturbations revealed that several feedback/crosstalks like DAG to PKC, PKC to IRS, S6K1 to IRS, cAMP to PKA, and AMPK to Akt are essential for the metabolic switching in matrix-deprived cancer cells. AMPK knockdown simulations identified a crucial role for AMPK in maintaining these adaptive changes. Thus, this mathematical framework provides insights on attachment-detachment signaling with metabolic adaptations that promote cancer metastasis.
Collapse
Affiliation(s)
- Sayoni Maiti
- Interdisciplinary Programme in Mathematical Sciences, IISc Mathematics Initiative, Indian Institute of Science, Bengaluru, India
| | - Annapoorni Rangarajan
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, India.
| | - Venkatesh Kareenhalli
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
64
|
Khuu A, Verreault M, Colin P, Tran H, Idbaih A. Clinical Applications of Antisense Oligonucleotides in Cancer: A Focus on Glioblastoma. Cells 2024; 13:1869. [PMID: 39594617 PMCID: PMC11592788 DOI: 10.3390/cells13221869] [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: 09/09/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are promising drugs capable of modulating the protein expression of virtually any target with high specificity and high affinity through complementary base pairing. However, this requires a deep understanding of the target sequence and significant effort in designing the correct complementary drug. In addition, ASOs have been demonstrated to be well tolerated during their clinical use. Indeed, they are already used in many diseases due to pathogenic RNAs of known sequences and in several neurodegenerative diseases and metabolic diseases, for which they were given marketing authorizations (MAs) in Europe and the United States. Their use in oncology is gaining momentum with several identified targets, promising preclinical and clinical results, and recent market authorizations in the US. However, many challenges remain for their clinical use in cancer. It seems necessary to take a step back and review our knowledge of ASOs and their therapeutic uses in oncology. The objectives of this review are (i) to summarize the current state of the art of ASOs; (ii) to discuss the therapeutic use of ASOs in cancer; and (iii) to focus on ASO usage in glioblastoma, the challenges, and the perspective ahead.
Collapse
Affiliation(s)
- Alexandre Khuu
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Maïté Verreault
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| | - Philippe Colin
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Helene Tran
- Institut de Recherche Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France;
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Hôpitaux Universitaires La Pitié Salpêtrière, Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, 75013 Paris, France; (A.K.); (M.V.)
| |
Collapse
|
65
|
Carmona-Carmona CA, Zini P, Velasco-Sampedro EA, Cózar-Castellano I, Perdomo G, Caloca MJ. β2-Chimaerin, a GTPase-Activating Protein for Rac1, Is a Novel Regulator of Hepatic Insulin Signaling and Glucose Metabolism. Molecules 2024; 29:5301. [PMID: 39598690 PMCID: PMC11597029 DOI: 10.3390/molecules29225301] [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: 10/10/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Glucose homeostasis is a complex process regulated by multiple organs and hormones, with insulin playing a central role. Recent evidence underscores the role of small GTP-binding proteins, particularly Rac1, in regulating insulin secretion and glucose uptake. However, the role of Rac1-regulatory proteins in these processes remains largely unexplored. In this study, we investigated the role of β2-chimaerin, a Rac1-specific GTPase-activating protein (GAP), in glucose homeostasis using whole-body β2-chimaerin knockout mice. Our data revealed that β2-chimaerin deficiency results in improved glucose tolerance and enhanced insulin sensitivity in mice. These metabolic effects were associated with increased insulin-induced AKT phosphorylation in the liver and activation of downstream pathways that regulate gluconeogenesis and glycogen synthesis. We show that insulin activates Rac1 in the liver. However, β2-chimaerin deletion did not significantly alter Rac1 activation in this organ, suggesting that β2-chimaerin regulates insulin signaling via a Rac1-independent mechanism. These findings expand our understanding of Rac1 regulation in glucose metabolism, and identify β2-chimaerin as a novel modulator of hepatic insulin signaling, with potential implications for the development of insulin resistance and diabetes.
Collapse
Affiliation(s)
- Cristian Andrés Carmona-Carmona
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
| | - Pablo Zini
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
| | - Eladio A. Velasco-Sampedro
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
| | - Irene Cózar-Castellano
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Germán Perdomo
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
| | - María J. Caloca
- Instituto de Biomedicina y Genética Molecular (IBGM), CSIC-UVA, 47003 Valladolid, Spain; (C.A.C.-C.); (P.Z.); (E.A.V.-S.); (I.C.-C.); (G.P.)
| |
Collapse
|
66
|
Oakes A, Liu Y, Dubielecka PM. Complement or insult: the emerging link between complement cascade deficiencies and pathology of myeloid malignancies. J Leukoc Biol 2024; 116:966-984. [PMID: 38836653 PMCID: PMC11531810 DOI: 10.1093/jleuko/qiae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
The complement cascade is an ancient and highly conserved arm of the immune system. The accumulating evidence highlights elevated activity of the complement cascade in cancer microenvironment and emphasizes its effects on the immune, cancer, and cancer stroma cells, pointing to a role in inflammation-mediated etiology of neoplasms. The role the cascade plays in development, progression, and relapse of solid tumors is increasingly recognized, however its role in hematological malignancies, especially those of myeloid origin, has not been thoroughly assessed and remains obscure. As the role of inflammation and autoimmunity in development of myeloid malignancies is becoming recognized, in this review we focus on summarizing the links that have been identified so far for complement cascade involvement in the pathobiology of myeloid malignancies. Complement deficiencies are primary immunodeficiencies that cause an array of clinical outcomes including an increased risk of a range of infectious as well as local or systemic inflammatory and thrombotic conditions. Here, we discuss the impact that deficiencies in complement cascade initiators, mid- and terminal-components and inhibitors have on the biology of myeloid neoplasms. The emergent conclusions indicate that the links between complement cascade, inflammatory signaling, and the homeostasis of hematopoietic system exist, and efforts should continue to detail the mechanistic involvement of complement cascade in the development and progression of myeloid cancers.
Collapse
Affiliation(s)
- Alissa Oakes
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
| | - Yuchen Liu
- Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201-1595, USA
| | - Patrycja M Dubielecka
- Department of Medicine, Alpert Medical School, Brown University, 69 Brown St, Providence, RI 02906, USA
- Division of Hematology/Oncology, Rhode Island Hospital, 69 Brown St, Providence, RI 02906, USA
- Therapeutic Sciences Graduate program, Brown University, 69 Brown St, Providence, RI 02906, USA
- Legorreta Cancer Center, Brown University, One Hoppin St., Coro West, Suite 5.01, Providence, RI 02903, USA
| |
Collapse
|
67
|
Shiryagin VV, Devyatkin AA, Fateev OD, Petriaikina ES, Bogdanov VP, Antysheva ZG, Volchkov PY, Yudin SM, Woroncow M, Skvortsova VI. Genomic complexity and clinical significance of the RCCX locus. PeerJ 2024; 12:e18243. [PMID: 39512309 PMCID: PMC11542561 DOI: 10.7717/peerj.18243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/15/2024] [Indexed: 11/15/2024] Open
Abstract
Nearly identical, repetitive elements in the genome contribute to the variability in genetic inheritance patterns, particularly in regions like the RCCX locus, where such repeats can lead to structural variations. In addition, during the formation of gametes as a result of meiosis, variants of loci with repetitive elements that do not code for the required proteins may occur. As a result, an individual with certain genetic rearrangements in this region may have an increased risk of developing a congenital disorder, particularly in cases where the non-functional allele is inherited dominantly. At the same time, there is still no routine or generally recognized diagnostic method to determine the sequence of the repetitive fragments. The functionally important RCCX locus consists of such repetitive fragments. The available knowledge about the genomic variants of the RCCX locus is fragmented, as there is no standardized method to determine its structure. It should be noted that in some structural variants of the RCCX locus, the sequence of protein-coding genes is disrupted, leading to the development of diseases such as congenital adrenal hyperplasia (CAH). Although genetic testing is generally accepted as a gold standard for CAH diagnosis, there are a myriad of strategies on which exact methods to use and in which order. The reason for this inconsistency lies in the complexity of the RCCX locus and the fact that each patient or carrier may have a highly individualized mutation or combination thereof. In this review, we have discussed all known methods that can be used to study the structure of the RCCX locus. As a result, optimal approaches are proposed for the diagnosis of the most common disease caused by lesions in the RCCX-CAH due to CYP21A2 deficiency.
Collapse
Affiliation(s)
- Vladimir V. Shiryagin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency (Centre for Strategic Planning of FMBA of Russia), Moscow, Russia
| | - Andrey A. Devyatkin
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Oleg D. Fateev
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency (Centre for Strategic Planning of FMBA of Russia), Moscow, Russia
| | - Ekaterina S. Petriaikina
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency (Centre for Strategic Planning of FMBA of Russia), Moscow, Russia
| | - Viktor P. Bogdanov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Zoia G. Antysheva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
| | - Pavel Yu Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, Russia
- Department of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Sergey M. Yudin
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency (Centre for Strategic Planning of FMBA of Russia), Moscow, Russia
| | - Mary Woroncow
- Department of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | |
Collapse
|
68
|
Zhang H, Fan K, Chen Y, Xu P, Zhang Z, Mo X, Guo Y. Genome-Wide Identification of Cell Type-Specific Susceptibility Genes for SLE Through the Analysis of RNA Modification-Associated SNPs. Immunol Invest 2024; 53:1264-1278. [PMID: 39230170 DOI: 10.1080/08820139.2024.2399577] [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: 09/05/2024]
Abstract
INTRODUCTION This study aimed to elucidate the functional genes associated with systemic lupus erythematosus (SLE) in various cell types through the utilization of RNAm-SNPs. METHODS Utilizing large-scale genetic data, we identified associations between RNAm-SNPs and SLE. The association between RNAm-SNPs and bulk and single-cell mRNA expression (eQTL) and protein levels (pQTL) were examined. Mendelian randomization and differential expression analyses were conducted to explore the links between gene expression, protein levels, and SLE. RESULTS We identified 41 RNAm-SNPs that were significantly associated with SLE. The GWAS signals exhibited notable enrichment in m6A-SNPs and m7G-SNPs. These RNAm-SNPs showed both eQTL and pQTL effects. In our single-cell analysis, 16 RNAm-SNPs exhibited associations with gene expression levels across 13 distinct cell types, including HLA-A, HLA-B, HLA-C, HLA-DQA1, HLA-DQB1, HLA-DRB1 and IRF7. We identified 58 noteworthy associations between the expression levels of 20 genes and SLE across 12 distinct immune cell types. Notably, HLA-DQB1, HLA-DRB1 and IRF7 exhibited abnormalities in CD8+ T cells, IRF7 displayed abnormal expression in CD4+ T cells, while HLA-DRB1 and IRF7 were also distinctly perturbed in natural killer cells. DISCUSSION This study advances our understanding of the genetic basis of SLE by highlighting the significance of RNAm-SNPs and immune cell gene expression in SLE.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Kedi Fan
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yuxi Chen
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Peng Xu
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Zhentao Zhang
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Xingbo Mo
- Department of Epidemiology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, MOE Key Laboratory of Geriatric Diseases and Immunology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yufan Guo
- Department of Rheumatology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| |
Collapse
|
69
|
Zhang W, Li M, Zhao Z, Xu J, Liu J, Feng P, Zhang B, Huang Z, Kong QQ, Lin Y. Tetrahedral Framework Nucleic Acid-Loaded Retinoic Acid Promotes Osteosarcoma Stem Cell Clearance. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58452-58463. [PMID: 39425646 DOI: 10.1021/acsami.4c14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Metastatic osteosarcoma is a commonly seen malignant tumor in adolescents, with a five year survival rate of approximately 20% and a lack of treatment options. Osteosarcoma cancer stem cells are considered to be important drivers of the metastasis of osteosarcoma, and therefore their clearance is considered a promising strategy for treating metastatic osteosarcoma. In the relevant literature, retinoic acid (ATRA) is considered effective for eliminating osteosarcoma stem cells, but it has some inherent disadvantages, including poor solubility, difficulty in entering cells, and structural instability. Tetrahedral framework nucleic acids (tFNAs) are a type of nanoparticles that can carry small-molecule drugs into cells to exert therapeutic effects. Therefore, we designed and synthesized a nanoparticle named T-ATRA by using tFNAs to load ATRA and studied its effect in a nude mouse model. T-ATRA is more effective than ATRA in the clearance of osteosarcoma stem cells and in inhibiting osteosarcoma cell metastasis via the Wnt signaling pathway, thus prolonging the survival time of nude mice with osteosarcoma.
Collapse
Affiliation(s)
- Weifei Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengqing Li
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Zhen Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiangshan Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junlin Liu
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Pin Feng
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Bin Zhang
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Zhangheng Huang
- Department of Orthopaedics (Spine Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qing-Quan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Orthopedics Surgery, Hospital of Chengdu Office of People's Government of Tibetan Autonomous Region, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
70
|
Wolf HN, Guempelein L, Schikora J, Pauly D. C3a Mediates Endothelial Barrier Disruption in Brain-Derived, but Not Retinal, Human Endothelial Cells. Int J Mol Sci 2024; 25:11240. [PMID: 39457022 PMCID: PMC11508547 DOI: 10.3390/ijms252011240] [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: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is associated with pathological aquaporin-4 immunoglobulin G (AQP4-IgG), which cause brain damage. However, the impact of AQP4-IgG on retinal tissue remains unclear. Additionally, dysregulated complement anaphylatoxins C3a and C5a, known to modulate the endothelial barrier, are implicated in NMOSD. This study evaluates the susceptibility of human brain microvascular endothelial cells (HBMEC) and human retinal endothelial cells (HREC) to C3a- and C5a-mediated stress using real-time cell barrier analysis, immunocytochemical staining, qPCR and IgG transmigration assays. The findings reveal that C3a induced a concentration-dependent paracellular barrier breakdown and increased transcellular permeability in HBMEC, while HREC maintained barrier integrity under the same conditions. C5a attenuated C3a-induced disruption in HBMEC, indicating a protective role. Anaphylatoxin treatment elevated transcript levels of complement component C3 and increased C5 gene and protein expression in HREC, with no changes observed in HBMEC. In HBMEC, C5a treatment led to a transient upregulation of C3a receptor (C3AR) mRNA and an early decrease in C5a receptor 1 (C5AR1) protein detection. Conversely, HREC exhibited a late increase in C5aR1 protein levels. These results indicate that the retinal endothelial barrier is more stable under anaphylatoxin-induced stress compared to the brain, potentially offering better protection against paracellular AQP4-IgG transport.
Collapse
Affiliation(s)
| | | | | | - Diana Pauly
- Department of Experimental Ophthalmology, University Marburg, 35043 Marburg, Germany
| |
Collapse
|
71
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
72
|
Yu L, Zang C, Ye Y, Liu H, Eucker J. Effects of BYL-719 (alpelisib) on human breast cancer stem cells to overcome drug resistance in human breast cancer. Front Pharmacol 2024; 15:1443422. [PMID: 39469631 PMCID: PMC11514072 DOI: 10.3389/fphar.2024.1443422] [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: 06/04/2024] [Accepted: 09/06/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction Breast cancer continues to be a major health concern and is currently the most commonly diagnosed cancer worldwide. Relapse, metastasis, and therapy resistance are major clinical issues that doctors need to address. We believe BYL-719, which is PI3 kinase p110а inhibitor, could also inhibit the breast cancer stem cell phenotype and epithelial-to-mesenchymal transition (EMT). In addition to the PI3K/AKT signaling pathway, BYL-719 can also inhibit essential cancer-related signaling pathways, all of which would ultimately act on the microenvironment of cancer stem cells, which is quite complicated and regulates the characteristics of tumors. These include the stemness and resistance of malignant tumors, plasticity of cancer stem cells, and anti-apoptotic features. Materials and methods A three-dimensional (3D) mammosphere culture method was used in vitro to culture and collect breast cancer stem cells (BCSCs). MTT, clonogenic, and cell apoptosis assays were used to detect cell viability, self-renewal, and differentiation abilities. A sphere formation assay under 3D conditions was used to detect the mammophore inhibition rate of BYL-719. The subpopulation of CD44+CD24- was detected using flow cytometry analysis while EMT biomarkers and essential signaling pathways were detected using western blotting. All the data were analyzed using GraphPad Prism 9 software. Results BCSC-like cells were obtained by using the 3D cell culture method in vitro. We confirmed that BYL-719 could inhibit BCSC-like cell proliferation in 3D cultures and that the stemness characteristics of BCSC-like cells were inhibited. The PI3K/AKT/mTOR signaling pathway could be inhibited by BYL-719, and the Notch, JAK-STAT and MAPK/ERK signaling pathways which have crosstalk in the tumor microenvironment (TME) are also inhibited. By comparing eribulin-resistant breast cancer cell lines, we confirmed that BYL-719 could effectively overcome drug resistance. Summary/conclusion The 3D cell culture is a novel and highly effective method for enriching BCSCs in vitro. Furthermore, the stemness and EMT of BCSCs were inhibited by BYL-719 by acting on various signaling pathways. Finally, we believe that drug resistance can be overcome by targeting the BCSCs. Conjugation of BYL-719 with other anti-neoplastic agents may be a promising treatment for this in clinic.
Collapse
Affiliation(s)
- Leinan Yu
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
| | - Chuanbing Zang
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
| | - Yuanchun Ye
- School of Science, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Hongyu Liu
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
| | - Jan Eucker
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin Charité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu Berlin, Berlin, Germany
- Department of Oncology, Rheumatology and Gastroenterology, Vivantes Klinikum Spandau, Berlin, Germany
| |
Collapse
|
73
|
Liu B, Wang X, Yang Z, Yin Z, Tang C, He Y, Ling Q, Huang Z, Feng S. A genetic study to identify pathogenic mechanisms and drug targets for benign prostatic hyperplasia: a multi-omics Mendelian randomization study. Sci Rep 2024; 14:23120. [PMID: 39367121 PMCID: PMC11452698 DOI: 10.1038/s41598-024-73466-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) as a common geriatric disease in urology, the incidence and prevalence are rapidly increasing with the aging society, prompting an urgent need for effective prevention and treatment of BPH. However, limited therapeutic efficacy and higher risk of complications result in the treatment of BPH remaining challenging. The unclear pathogenic mechanism also hampers further exploration of therapeutic approaches for BPH. In this study, we used multi-omics methods to integrate genomics, transcriptomics, immunomics, and metabolomics data and identify biomolecules associated with BPH. We performed transcriptomic imputation, summary data-based Mendelian randomization (SMR), joint/conditional analysis, colocalization analysis, and FOCUS to explore high-confidence genes associated with BPH in blood and prostate tissue. Subsequently, three-step SMR was used to identify the DNA methylation sites regulating high-confidence genes to improve the pathogenic pathways of BPH. We also used cis-instruments of druggable genes to conduct SMR analysis to find potential drug targets for BPH. Finally, we used MR analysis to explore the immune pathways and metabolomics related to BPH. Multiple analytical methods identified BTN3A2 (Blood: TWAS Z score = 5.02912, TWAS P = 4.93 × 10-7; Prostate: TWAS Z score = 4.89, TWAS P = 1.01 × 10-6) and C4A (Blood: TWAS Z score = 4.90754, TWAS P = 9.22 × 10-7; Prostate: TWAS Z score = 5.084, TWAS P = 3.70 × 10-7) as high-confidence genes for BPH and identified the cg14345882-BTN3A2-BPH pathogenic pathway. We also used druggable gene data to identify 30 promising therapeutic target genes, including BTN3A2 and C4A. For MR analysis of immune pathways, we identified immune cell surface molecules as well as the inflammatory factor IL-17 (OR = 1.25, 95% CI = 1.09-1.43, PFDR = 0.12, Maximum likelihood) as risk factors for BPH. In addition, we found that disulfide levels of cysteinylglycine (OR = 1.11, 95% CI = 1.05-1.18, P = 5.18 × 10-4, Weighted median), oxidation levels of cysteinylglycine (OR = 1.09, 95% CI = 1.04-1.14, P = 3.87 × 10-4, Weighted median), and sebacate levels (OR = 1.05, 95% CI = 1.02-1.08, P = 3.0 × 10-4, Maximum likelihood) increase the risk of BPH. This multi-omics study explored biomolecules associated with BPH, improved the pathogenic pathways of BPH, and identified promising therapeutic targets. Our results provide evidence for future studies aimed at developing appropriate therapeutic interventions.
Collapse
Affiliation(s)
- Bohan Liu
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinyi Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zerui Yang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhaofa Yin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Cai Tang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yushi He
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qi Ling
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
| | - Zhongli Huang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Shijian Feng
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology) and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
74
|
Wang YD, Meng X, Guan YC, Zhao ZL, Tao LT, Gong JS, Liu XL, Zhao Y, Shan XF. The effects of dietary supplementation of ginseng stem and leaf saponins on the antioxidant capacity, immune response, and disease resistance of crucian carp, Carassius auratus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1915-1930. [PMID: 36414818 DOI: 10.1007/s10695-022-01142-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/11/2022] [Indexed: 06/16/2023]
Abstract
This is the first study to explore the positive effects of ginseng stem and leaf saponins (GSLS) on antioxidant capability, immunity, and disease resistance of crucian carp. Seven hundred fifty crucian carps (initial body weight: 25 ± 0.15 g (mean ± SE)) were randomly allocated into five groups with three replicates each; five diets supplemented with the final concentration of 0, 1, 2, 4, and 8 g/kg GSLS were fed to crucian carp for 5 weeks. The results demonstrated that, at a concentration of 8 g/kg, the contents of IgM, C4, SOD, GSH-Px, and the activity of AKP in serum of crucian carp gradually increased at 7, 14, 21, 28, and 35 days, and the expression of immune-relative cytokine genes (TNF-α, IL-10, IFN-γ) in the liver, spleen, and the intestinal tract also had a significant up-regulation (P < 0.05), and which were significant difference compared with control (P < 0.05). The above results demonstrated that dietary GSLS showed enhancement effects on the antioxidant and anti-inflammatory capability, and innate immune response of crucian carp. The feed of 8 g/kg GSLS for 1 week could improve the survival rate 44% more than the control group when crucian carp infected Aeromonas hydrophila (A. hydrophila). In conclusion, the addition of GSLS at a concentration of 8 g/kg in the diet improve immune-related enzyme activity better, immune-relative cytokine expression, and disease resistance.
Collapse
Affiliation(s)
- Ying-da Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xin Meng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yong-Chao Guan
- College of Life Sciences, Jilin Agricultural University, Changchun, 130118, Jilin Province, China
| | - Ze-Lin Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Luo-Tao Tao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jin-Shuo Gong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xin-Lan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agriculture University, Changchun, 130118, Jilin, China
| | - Xiao-Feng Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
75
|
Long Y, Dai W, Cai K, Xiao Y, Luo A, Lai Z, Wang J, Xu L, Nie H. Systemic Immune Factors and Risk of Allergic Contact Dermatitis: A Bidirectional Mendelian Randomization Study. Int J Mol Sci 2024; 25:10436. [PMID: 39408763 PMCID: PMC11476522 DOI: 10.3390/ijms251910436] [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: 08/29/2024] [Revised: 09/16/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Skin inflammation and immune regulation have been suggested to be associated with allergic contact dermatitis (ACD) progression, but whether the system's immune regulation is a cause or a potential mechanism is still unknown. This study aims to assess the upstream and downstream of systemic immune factors on ACD within a bidirectional Mendelian-randomization design. A bidirectional two-sample MR analysis was employed to implement the results from genome-wide association studies for 52 system immune factors and ACD. Genetic associations with systemic immune factors and ACD were obtained from the IEU Open GWAS project database. The inverse-variance weighted (IVW) method was adopted as the primary MR analysis, MR-Egger, weighted median, MR-pleiotropy residual sum, and outlier (MR-PRESSO) was also used as the sensitivity analyses. Only Tumor necrosis factor ligand superfamily member 11 (TNFS11) from among 52 systemic immune factors was associated with a protective effect of ACD. However, ACD was associated with a decrease in Interleukin-9 (IL9) and an increase in C-X-C motif chemokine 1 (GROα), Tumor necrosis factor ligand superfamily member 10 (TRAIL), C4, and complement factor B of the assessed systemic immune factors. This study identified TNFS11 as the upstream regulator and IL9, GROα, TRAIL, C4, and complement factor B as the downstream regulator of ACD, providing opportunities for new therapeutic exploitation of ACD. Nonetheless, these associations of systemic immune factors need to be verified in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lipeng Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; (Y.L.); (W.D.); (K.C.); (Y.X.); (A.L.); (Z.L.); (J.W.)
| | - Hong Nie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China; (Y.L.); (W.D.); (K.C.); (Y.X.); (A.L.); (Z.L.); (J.W.)
| |
Collapse
|
76
|
Bae ES, Hong J, Lim Y, Byun WS, Chun S, Hong S, Lee SK. Evo312: An Evodiamine Analog and Novel PKCβI Inhibitor with Potent Antitumor Activity in Gemcitabine-Resistant Pancreatic Cancer. J Med Chem 2024; 67:14885-14911. [PMID: 39151060 DOI: 10.1021/acs.jmedchem.4c00213] [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: 08/18/2024]
Abstract
As an obstinate cancer pancreatic cancer (PC) poses a major challenge due to limited treatment options which include resection surgery, radiation therapy, and gemcitabine-based chemotherapy. In cancer cells, protein kinase C βI (PKCβI) participates in diverse cellular processes, including cell proliferation, invasion, and apoptotic pathways. In the present study, we created a scaffold to develop PKCβI inhibitors using evodiamine-based synthetic molecules. Among the candidate inhibitors, Evo312 exhibited the highest antiproliferative efficacy against PC cells, PANC-1, and acquired gemcitabine-resistant PC cells, PANC-GR. Additionally, Evo312 robustly inhibited PKCβI activity. Mechanistically, Evo312 effectively suppressed the upregulation of PKCβI protein expression, leading to the induction of cell cycle arrest and apoptosis in PANC-GR cells. Furthermore, Evo312 exerted an antitumor activity in a PANC-GR cell-implanted xenograft mouse model. These findings position Evo312 as a promising lead compound for overcoming gemcitabine resistance in PC through novel mechanisms.
Collapse
Affiliation(s)
- Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Junhwa Hong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yijae Lim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Simin Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Suckchang Hong
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
77
|
Ayele K, Wakimoto H, Saha D. An oncolytic adenovirus co-expressing a bi-specific T cell engager and IL-2 for the treatment of ovarian cancer. Mol Ther 2024; 32:2810-2813. [PMID: 39178849 PMCID: PMC11403211 DOI: 10.1016/j.ymthe.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024] Open
Affiliation(s)
- Kalkidan Ayele
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA; Department of Biology, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, USA.
| |
Collapse
|
78
|
Basnet S, Van der Heijden M, Quixabeira DCA, Jirovec E, Grönberg-Vähä-Koskela SAM, Clubb JHA, Kanerva A, Pakola S, Haybout L, Arias V, Hemminki O, Kudling T, Zafar S, Cervera-Carrascon V, Santos JM, Hemminki A. Overcoming effector T cell exhaustion in ovarian cancer ascites with a novel adenovirus encoding for a MUC1 bispecific antibody engager and IL-2 cytokine. Mol Ther 2024; 32:3114-3127. [PMID: 38910324 PMCID: PMC11403222 DOI: 10.1016/j.ymthe.2024.06.029] [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: 11/17/2023] [Revised: 03/18/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024] Open
Abstract
T cell-focused cancer immunotherapy including checkpoint inhibitors and cell therapies has been rapidly evolving over the past decade. Nevertheless, there remains a major unmet medical need in oncology generally and immuno-oncology specifically. We have constructed an oncolytic adenovirus, Ad5/3-E2F-d24-aMUC1aCD3-IL-2 (TILT-322), which is armed with a human aMUC1aCD3 T cell engager and IL-2. TILT-322 treatment stimulated T cell cytotoxicity through the increased presence of granzyme B, perforin, and interferon-gamma. Additional immune profiling indicated TILT-322 increased gamma delta T cell activation and impacted other cell types such as natural killer cells and natural killer-like T cells that are traditionally involved in cancer immunotherapy. TILT-322 treatment also decreased the proportion of exhausted CD8+ T cells as demarked by immune checkpoint expression in ovarian ascites samples. Overall, our data showed that TILT-322 treatment led to an enhanced T cell activation and reversed T cell exhaustion translating into high antitumor efficacy when given locally or intravenously. The analysis of blood and tumors isolated from an in vivo patient-derived ovarian cancer xenograft model suggested TILT-322 mediated tumor control through improved T cell functions. Therefore, TILT-322 is a promising novel anti-tumor agent for clinical translation.
Collapse
Affiliation(s)
- Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Mirte Van der Heijden
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Susanna A M Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki University Hospital (HUS), Comprehensive Cancer Center, Helsinki, Finland
| | - James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland; Helsinki University Hospital (HUS), Comprehensive Cancer Center, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Otto Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Tatiana Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Sadia Zafar
- Applied Tumor Genomics HUS Comprehensive Cancer Center, Research Program, Research Program Unit, University of Helsinki, Helsinki, Finland; Department of Pathology, HUSLAB, HUS Diagnostic Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland; Helsinki University Hospital (HUS), Comprehensive Cancer Center, Helsinki, Finland.
| |
Collapse
|
79
|
Singh I, Rainusso N, Kurenbekova L, Nirala BK, Dou J, Muruganandham A, Yustein JT. Intrinsic Epigenetic State of Primary Osteosarcoma Drives Metastasis. Mol Cancer Res 2024; 22:864-878. [PMID: 38842581 DOI: 10.1158/1541-7786.mcr-23-0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/03/2023] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Osteosarcoma is the most common primary malignant bone tumor affecting the pediatric population with a high potential to metastasize. However, insights into the molecular features enabling its metastatic potential are limited. We mapped the active chromatin landscapes of osteosarcoma tumors by integrating histone H3 lysine-acetylated chromatin state (n = 13), chromatin accessibility profiles (n = 11), and gene expression (n = 13) to understand the differences in their active chromatin profiles and their impact on molecular mechanisms driving the malignant phenotypes. Primary osteosarcoma tumors from patients with metastasis (primary met) have a distinct active chromatin landscape compared with those without metastasis (localized). This difference shapes the transcriptional profile of osteosarcoma. We identified novel candidate genes, including PPP1R1B, PREX1, and IGF2BP1, that exhibit increased chromatin activity in primary met. Loss of PREX1 in primary met osteosarcoma cells significantly diminishes osteosarcoma proliferation, invasion, migration, and colony formation capacity. Differential chromatin activity in primary met is associated with genes regulating cytoskeleton organization, cellular adhesion, and extracellular matrix, suggesting their role in facilitating osteosarcoma metastasis. Chromatin profiling of tumors from metastatic lung lesions shows increased chromatin activity in genes involved in cell migration and Wnt pathway. These data demonstrate that metastatic potential is intrinsically present in primary met tumors, with cellular chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal site. Implications: Our study demonstrates that metastatic potential is intrinsic to primary metastatic osteosarcoma tumors, with chromatin profiles further adapting for successful dissemination, migration, and colonization at the distal metastatic site.
Collapse
Affiliation(s)
- Irtisha Singh
- Department of Cell Biology and Genetics, College of Medicine, Texas A&M University, Bryan, Texas
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas
| | - Nino Rainusso
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Lyazat Kurenbekova
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Bikesh K Nirala
- Winship Cancer Institute and Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, Georgia
| | - Juan Dou
- Winship Cancer Institute and Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, Georgia
| | - Abhinaya Muruganandham
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas
| | - Jason T Yustein
- Winship Cancer Institute and Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, Georgia
| |
Collapse
|
80
|
Kegyes D, Milea PA, Mazga AI, Tigu AB, Nistor M, Cenariu D, Tomai R, Buruiana S, Einsele H, Daniela Tănase A, Tomuleasa C. Looking ahead to targeting macrophages by CAR T- or NK-cells in blood cancers. Expert Opin Ther Targets 2024; 28:779-787. [PMID: 39235181 DOI: 10.1080/14728222.2024.2400075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The bone marrow microenvironment (BME) is critical for healthy hematopoiesis and is often disrupted in hematologic malignancies. Tumor-associated macrophages (TAMs) are a major cell type in the tumor microenvironment (TME) and play a significant role in tumor growth and progression. Targeting TAMs and modulating their polarization is a promising strategy for cancer therapy. AREAS COVERED In this review, we discuss the importance of TME and different multiple possible targets to modulate immunosuppressive TAMs such as: CD123, Sphingosine 1-Phosphate Receptors, CD19/CD1d, CCR4/CCL22, CSF1R (CD115), CD24, CD40, B7 family proteins, MARCO, CD47, CD163, CD204, CD206 and folate receptors. EXPERT OPINION Innovative approaches to combat the immunosuppressive milieu of the tumor microenvironment in hematologic malignancies are of high clinical significance and may lead to increased survival, improved quality of life, and decreased toxicity of cancer therapies. Standard procedures will likely involve a combination of CAR T/NK-cell therapies with other treatments, leading to more comprehensive cancer care.
Collapse
Affiliation(s)
- David Kegyes
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Paul Alexandru Milea
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Andreea-Isabella Mazga
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Adrian-Bogdan Tigu
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Madalina Nistor
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Cenariu
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Radu Tomai
- Department of Hematology, Ion Chiricuta Cancer Center, Cluj-Napoca, Romania
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemitanu State University of Medicine and Pharmacy, Chisinau, Moldova
| | - Hermann Einsele
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine II, Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Alina Daniela Tănase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Ciprian Tomuleasa
- Department of Hematology/Medfuture Research Center for Advanced Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Cancer Center, Cluj-Napoca, Romania
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
81
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
82
|
Jones GD, Ellisdon AM. Understanding P-Rex regulation: structural breakthroughs and emerging perspectives. Biochem Soc Trans 2024; 52:1849-1860. [PMID: 39023851 PMCID: PMC11668296 DOI: 10.1042/bst20231546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
Rho GTPases are a family of highly conserved G proteins that regulate numerous cellular processes, including cytoskeleton organisation, migration, and proliferation. The 20 canonical Rho GTPases are regulated by ∼85 guanine nucleotide exchange factors (GEFs), with the largest family being the 71 Diffuse B-cell Lymphoma (Dbl) GEFs. Dbl GEFs promote GTPase activity through the highly conserved Dbl homology domain. The specificity of GEF activity, and consequently GTPase activity, lies in the regulation and structures of the GEFs themselves. Dbl GEFs contain various accessory domains that regulate GEF activity by controlling subcellular localisation, protein interactions, and often autoinhibition. This review focuses on the two phosphatidylinositol (3,4,5)-trisphosphate (PI(3,4,5)P3)-dependent Rac exchangers (P-Rex), particularly the structural basis of P-Rex1 autoinhibition and synergistic activation. First, we discuss structures that highlight the conservation of P-Rex catalytic and phosphoinositide binding activities. We then explore recent breakthroughs in uncovering the structural basis for P-Rex1 autoinhibition and detail the proposed minimal two-step model of how PI(3,4,5)P3 and Gβγ synergistically activate P-Rex1 at the membrane. Additionally, we discuss the further layers of P-Rex regulation provided by phosphorylation and P-Rex2-PTEN coinhibitory complex formation, although these mechanisms remain incompletely understood. Finally, we leverage the available data to infer how cancer-associated mutations in P-Rex2 destabilise autoinhibition and evade PTEN coinhibitory complex formation, leading to increased P-Rex2 GEF activity and driving cancer progression and metastasis.
Collapse
Affiliation(s)
- Gareth D. Jones
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Andrew M. Ellisdon
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| |
Collapse
|
83
|
Limonta P, Chiaramonte R, Casati L. Unveiling the Dynamic Interplay between Cancer Stem Cells and the Tumor Microenvironment in Melanoma: Implications for Novel Therapeutic Strategies. Cancers (Basel) 2024; 16:2861. [PMID: 39199632 PMCID: PMC11352669 DOI: 10.3390/cancers16162861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Cutaneous melanoma still represents a significant health burden worldwide, being responsible for the majority of skin cancer deaths. Key advances in therapeutic strategies have significantly improved patient outcomes; however, most patients experience drug resistance and tumor relapse. Cancer stem cells (CSCs) are a small subpopulation of cells in different tumors, including melanoma, endowed with distinctive capacities of self-renewal and differentiation into bulk tumor cells. Melanoma CSCs are characterized by the expression of specific biomarkers and intracellular pathways; moreover, they play a pivotal role in tumor onset, progression and drug resistance. In recent years, great efforts have been made to dissect the molecular mechanisms underlying the protumor activities of melanoma CSCs to provide the basis for novel CSC-targeted therapies. Herein, we highlight the intricate crosstalk between melanoma CSCs and bystander cells in the tumor microenvironment (TME), including immune cells, endothelial cells and cancer-associated fibroblasts (CAFs), and its role in melanoma progression. Specifically, we discuss the peculiar capacities of melanoma CSCs to escape the host immune surveillance, to recruit immunosuppressive cells and to educate immune cells toward an immunosuppressive and protumor phenotype. We also address currently investigated CSC-targeted strategies that could pave the way for new promising therapeutic approaches for melanoma care.
Collapse
Affiliation(s)
- Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences “R. Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy
| | - Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy;
| | - Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy;
| |
Collapse
|
84
|
Ravala SK, Tesmer JJG. New Mechanisms Underlying Oncogenesis in Dbl Family Rho Guanine Nucleotide Exchange Factors. Mol Pharmacol 2024; 106:117-128. [PMID: 38902036 PMCID: PMC11331503 DOI: 10.1124/molpharm.124.000904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
Transmembrane signaling is a critical process by which changes in the extracellular environment are relayed to intracellular systems that induce changes in homeostasis. One family of intracellular systems are the guanine nucleotide exchange factors (GEFs), which catalyze the exchange of GTP for GDP bound to inactive guanine nucleotide binding proteins (G proteins). The resulting active G proteins then interact with downstream targets that control cell proliferation, growth, shape, migration, adhesion, and transcription. Dysregulation of any of these processes is a hallmark of cancer. The Dbl family of GEFs activates Rho family G proteins, which, in turn, alter the actin cytoskeleton and promote gene transcription. Although they have a common catalytic mechanism exercised by their highly conserved Dbl homology (DH) domains, Dbl GEFs are regulated in diverse ways, often involving the release of autoinhibition imposed by accessory domains. Among these domains, the pleckstrin homology (PH) domain is the most commonly observed and found immediately C-terminal to the DH domain. The domain has been associated with both positive and negative regulation. Recently, some atomic structures of Dbl GEFs have been determined that reemphasize the complex and central role that the PH domain can play in orchestrating regulation of the DH domain. Here, we discuss these newer structures, put them into context by cataloging the various ways that PH domains are known to contribute to signaling across the Dbl family, and discuss how the PH domain might be exploited to achieve selective inhibition of Dbl family RhoGEFs by small-molecule therapeutics. SIGNIFICANCE STATEMENT: Dysregulation via overexpression or mutation of Dbl family Rho guanine nucleotide exchange factors (GEFs) contributes to cancer and neurodegeneration. Targeting the Dbl homology catalytic domain by small-molecule therapeutics has been challenging due to its high conservation and the lack of a discrete binding pocket. By evaluating some new autoinhibitory mechanisms in the Dbl family, we demonstrate the great diversity of roles played by the regulatory domains, in particular the PH domain, and how this holds tremendous potential for the development of selective therapeutics that modulate GEF activity.
Collapse
Affiliation(s)
- Sandeep K Ravala
- Departments of Biological Sciences and Medicinal Chemistry and Molecular Pharmacology (S.K.R., J.J.G.T.) and Purdue University Institute for Cancer Research (J.J.G.T.), Purdue University, West Lafayette, Indiana
| | - John J G Tesmer
- Departments of Biological Sciences and Medicinal Chemistry and Molecular Pharmacology (S.K.R., J.J.G.T.) and Purdue University Institute for Cancer Research (J.J.G.T.), Purdue University, West Lafayette, Indiana
| |
Collapse
|
85
|
Pinto-Souza CC, Kaihara JNS, Nunes PR, Mastella MH, Rossini BC, Cavecci-Mendonça B, Cavalli RDC, dos Santos LD, Sandrim VC. Different Proteomic Profiles Regarding Antihypertensive Therapy in Preeclampsia Pregnant. Int J Mol Sci 2024; 25:8738. [PMID: 39201423 PMCID: PMC11354552 DOI: 10.3390/ijms25168738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/22/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Preeclampsia (PE) is a hypertensive pregnancy syndrome associated with target organ damage and increased cardiovascular risks, necessitating antihypertensive therapy. However, approximately 40% of patients are nonresponsive to treatment, which results in worse clinical outcomes. This study aimed to compare circulating proteomic profiles and identify differentially expressed proteins among 10 responsive (R-PE), 10 nonresponsive (NR-PE) patients, and 10 healthy pregnant controls (HP). We also explored correlations between these proteins and clinical data. Plasma protein relative quantification was performed using mass spectrometry, followed by bioinformatics analyses with the UniProt database, PatternLab for Proteomics 4.0, and MetaboAnalyst software (version 6.0). Considering a fold change of 1.5, four proteins were differentially expressed between NR-PE and R-PE: one upregulated (fibronectin) and three downregulated (pregnancy-specific beta-1-glycoprotein 1, complement C4B, and complement C4A). Between NR-PE and HP, six proteins were differentially expressed: two upregulated (clusterin and plasmin heavy chain A) and four downregulated (apolipoprotein L1, heparin cofactor II, complement C4B, and haptoglobin-related protein). Three proteins were differentially expressed between R-PE and HP: one downregulated (transthyretin) and two upregulated (apolipoprotein C1 and hemoglobin subunit beta). These findings suggest a complex interplay of these proteins involved in inflammatory, immune, and metabolic processes with antihypertensive therapy responsiveness and PE pathophysiology.
Collapse
Affiliation(s)
- Caroline C. Pinto-Souza
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (C.C.P.-S.); (J.N.S.K.); (P.R.N.); (M.H.M.)
| | - Julyane N. S. Kaihara
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (C.C.P.-S.); (J.N.S.K.); (P.R.N.); (M.H.M.)
| | - Priscila R. Nunes
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (C.C.P.-S.); (J.N.S.K.); (P.R.N.); (M.H.M.)
| | - Moises H. Mastella
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (C.C.P.-S.); (J.N.S.K.); (P.R.N.); (M.H.M.)
| | - Bruno C. Rossini
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (B.C.R.); (B.C.-M.); (L.D.d.S.)
| | - Bruna Cavecci-Mendonça
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (B.C.R.); (B.C.-M.); (L.D.d.S.)
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu 18619-002, SP, Brazil
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto 14049-900, SP, Brazil;
| | - Lucilene D. dos Santos
- Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (B.C.R.); (B.C.-M.); (L.D.d.S.)
| | - Valeria C. Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (C.C.P.-S.); (J.N.S.K.); (P.R.N.); (M.H.M.)
| |
Collapse
|
86
|
Kumar P, Lakhera R, Aggarwal S, Gupta S. Unlocking the Therapeutic Potential of Oral Cancer Stem Cell-Derived Exosomes. Biomedicines 2024; 12:1809. [PMID: 39200273 PMCID: PMC11351673 DOI: 10.3390/biomedicines12081809] [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: 07/18/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Oral cancer (OC) presents a significant global health burden with rising incidence rates. Despite advancements in diagnosis and treatments, the survival rate for OC patients, particularly those with advanced or recurrent disease, remains low at approximately 20%. This poor prognosis is often due to a small population of cancer stem cells (CSCs) that are capable of self-renewal and immune evasion, playing pivotal roles in proliferation, tumor initiation, progression, metastasis, and therapy resistance. Exosomes, which are nano-sized extracellular vesicles (EVs), have emerged as crucial mediators of cell-to-cell communication within the tumor microenvironment (TME). These vesicles carry diverse molecules such as DNA, RNA, proteins, lipids, and metabolites, influencing various cellular processes. Emerging evidence suggests that CSC-derived EVs significantly promote tumor progression and metastasis and maintain the balance between CSCs and non-CSCs, which is vital for intracellular communication within the TME of oral cancer. Recent reports indicate that oral cancer stem cell-derived EVs (OCSC-EVs) influence stemness, immune evasion, metastasis, angiogenesis, tumor reoccurrence, and drug resistance. Understanding OCSC-EVs could significantly improve oral cancer diagnosis, prognosis, and therapy. In this mini-review, we explore OCSC-derived exosomes in oral cancer, examining their potential as diagnostic and prognostic biomarkers that reflect CSC characteristics, and delve into their therapeutic implications, emphasizing their roles in tumor progression and therapy resistance. However, despite their promising potential, several challenges remain, including the need to standardize isolation and characterization methods and to elucidate exosome-mediated mechanisms. Thus, a comprehensive understanding of OCSC-EVs could pave the way for innovative therapeutic strategies that have the potential to improve clinical outcomes for OC patients.
Collapse
Affiliation(s)
- Prabhat Kumar
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| | - Rishabh Lakhera
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| | - Sadhna Aggarwal
- Department of Radiation Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shilpi Gupta
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| |
Collapse
|
87
|
Kalinina E. Glutathione-Dependent Pathways in Cancer Cells. Int J Mol Sci 2024; 25:8423. [PMID: 39125992 PMCID: PMC11312684 DOI: 10.3390/ijms25158423] [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: 07/18/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The most abundant tripeptide-glutathione (GSH)-and the major GSH-related enzymes-glutathione peroxidases (GPxs) and glutathione S-transferases (GSTs)-are highly significant in the regulation of tumor cell viability, initiation of tumor development, its progression, and drug resistance. The high level of GSH synthesis in different cancer types depends not only on the increasing expression of the key enzymes of the γ-glutamyl cycle but also on the changes in transport velocity of its precursor amino acids. The ability of GPxs to reduce hydroperoxides is used for cellular viability, and each member of the GPx family has a different mechanism of action and site for maintaining redox balance. GSTs not only catalyze the conjugation of GSH to electrophilic substances and the reduction of organic hydroperoxides but also take part in the regulation of cellular signaling pathways. By catalyzing the S-glutathionylation of key target proteins, GSTs are involved in the regulation of major cellular processes, including metabolism (e.g., glycolysis and the PPP), signal transduction, transcription regulation, and the development of resistance to anticancer drugs. In this review, recent findings in GSH synthesis, the roles and functions of GPxs, and GST isoforms in cancer development are discussed, along with the search for GST and GPx inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Elena Kalinina
- T.T. Berezov Department of Biochemistry, Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| |
Collapse
|
88
|
Rokade S, Damani AM, Oft M, Emmerich J. IL-2 based cancer immunotherapies: an evolving paradigm. Front Immunol 2024; 15:1433989. [PMID: 39114660 PMCID: PMC11303236 DOI: 10.3389/fimmu.2024.1433989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Discovered over 4 decades ago in the supernatants of activated T cells, interleukin-2 (IL-2) is a potent pleiotropic cytokine involved in the regulation of immune responses. It is required for effector T cell expansion and differentiation as well as for peripheral tolerance induced by regulatory T cells. High-dose IL-2 treatment was the first FDA-approved immunotherapy for renal cell carcinoma and melanoma, achieving single agent complete and durable responses, albeit only in a small proportion of patients. The therapeutic potential of wild type IL-2 is clinically limited by its short half-life and severe vascular toxicity. Moreover, the activation of regulatory T cells and the terminal differentiation of effector T cells on IL-2 pose additional restrictions. To overcome the toxicity of IL-2 in order to realize its full potential for patients, several novel engineering strategies are being developed and IL-2 based immunotherapy for cancer has emerged as a burgeoning field of clinical and experimental research. In addition, combination of IL-2 with PD-1/L1 pathway blockade shows vastly improved anti-tumor efficacy over either monotherapy in preclinical tumor models. In this review we discuss the biological characteristics of IL-2 and its receptors, as well as its efficacy and treatment limiting toxicities in cancer patients. We also explore the efforts aimed at developing novel and safer IL-2 therapies to harness the full therapeutic potential of this cytokine.
Collapse
Affiliation(s)
- Sushama Rokade
- Development Department, Synthekine, Menlo Park, CA, United States
| | | | | | - Jan Emmerich
- Development Department, Synthekine, Menlo Park, CA, United States
| |
Collapse
|
89
|
Isca VMS, Bangay G, Princiotto S, Saraíva L, Dos Santos DJVA, García-Sosa AT, Rijo P. Extraction optimization and reactivity of 7α-acetoxy-6β-hydroxyroyleanone and ability of its derivatives to modulate PKC isoforms. Sci Rep 2024; 14:16990. [PMID: 39043734 PMCID: PMC11266714 DOI: 10.1038/s41598-024-67384-0] [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: 01/08/2023] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
Protein kinase C is a family of kinases that play important roles in carcinogenesis. Medicinal plants from Plectranthus spp. (Lamiaceae) are a well-known source of interesting abietanes, such as 7α-acetoxy-6β-hydroxyroyleanone (Roy). This study aimed to extract and isolate Roy from P. grandidentatus Gürke, comparing two extraction methods (CO2 supercritical and ultrasound-assisted acetonic extraction), and design new royleanone derivatives for PKC modulation focusing on breast cancer therapy. The concentration of Roy in the extracts was determined by HPLC-DAD. The supercritical extraction method yielded 3.6% w/w, with the presence of 42.7 μg mg-1 of Roy (yield of 0.13%), while ultrasound-assisted acetonic extraction yielded 2.3% w/w, with the presence of 55.2 μg mg-1 of Roy (yield of 0.15%). The reactivity of Roy was investigated aiming at synthetizing new ester derivatives through standard benzoylation and esterification reactions. The benzoylated (Roy-12-Bz) and acetylated (Roy-12-Ac) derivatives in the C12 position were consistently prepared with overall good yields (33-86%). These results indicate the 12-OH position as the most reactive for esterification, affording derivatives under mild conditions. The reported di-benzoylated (RoyBz) and di-acetylated (RoyAc) derivatives were also synthesized after increasing the temperature (50 °C), reaction time, and using an excess of reagents. The cytotoxic potential of Roy and its derivatives was assessed against breast cancer cell lines, with RoyBz emerging as the most promising compound. Derivatization at position C-12 did not offer advantages over di-esterification at positions C-12 and C-6 or over the parent compound Roy and the presence of aromatic groups favored cytotoxicity. Evaluation of royleanones as PKC-α, βI, δ, ε, and ζ activators revealed DeRoy's efficacy across all isoforms, while RoyPr showed promising activation of PKC-δ but not PKC-ζ, highlighting the influence of slight structural changes on isoform selectivity. Molecular docking analysis emphasized the importance of microenvironmental factors in isoform specificity, underscoring the complexity of PKC modulation and the need for further exploration.
Collapse
Affiliation(s)
- Vera M S Isca
- CBIOS - Center for Research in Biosciences and Health Technologies, Universidade Lusófona, 1749-024, Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Farmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
| | - Gabrielle Bangay
- CBIOS - Center for Research in Biosciences and Health Technologies, Universidade Lusófona, 1749-024, Lisboa, Portugal
- Departamento de Ciencias Biomédicas (Área de Farmacología), Nuevos agentes antitumorales, Acción tóxica sobre células leucémicas, Facultad de Farmacia, Universidad de Alcalá de Henares, Ctra. Madrid-Barcelona Km. 33, 600 28805, Alcalá de Henares, Madrid, Spain
| | - Salvatore Princiotto
- CBIOS - Center for Research in Biosciences and Health Technologies, Universidade Lusófona, 1749-024, Lisboa, Portugal
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, via Celoria 2, 20133, Milan, Italy
| | - Lucília Saraíva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Daniel J V A Dos Santos
- CBIOS - Center for Research in Biosciences and Health Technologies, Universidade Lusófona, 1749-024, Lisboa, Portugal
| | | | - Patrícia Rijo
- CBIOS - Center for Research in Biosciences and Health Technologies, Universidade Lusófona, 1749-024, Lisboa, Portugal.
- Research Institute for Medicines (iMed.ULisboa), Faculty of Farmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal.
| |
Collapse
|
90
|
Chen H, Fang S, Zhu X, Liu H. Cancer-associated fibroblasts and prostate cancer stem cells: crosstalk mechanisms and implications for disease progression. Front Cell Dev Biol 2024; 12:1412337. [PMID: 39092186 PMCID: PMC11291335 DOI: 10.3389/fcell.2024.1412337] [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: 04/07/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
The functional heterogeneity and ecological niche of prostate cancer stem cells (PCSCs), which are major drivers of prostate cancer development and treatment resistance, have attracted considerable research attention. Cancer-associated fibroblasts (CAFs), which are crucial components of the tumor microenvironment (TME), substantially affect PCSC stemness. Additionally, CAFs promote PCSC growth and survival by releasing signaling molecules and modifying the surrounding environment. Conversely, PCSCs may affect the characteristics and behavior of CAFs by producing various molecules. This crosstalk mechanism is potentially crucial for prostate cancer progression and the development of treatment resistance. Using organoids to model the TME enables an in-depth study of CAF-PCSC interactions, providing a valuable preclinical tool to accurately evaluate potential target genes and design novel treatment strategies for prostate cancer. The objective of this review is to discuss the current research on the multilevel and multitarget regulatory mechanisms underlying CAF-PCSC interactions and crosstalk, aiming to inform therapeutic approaches that address challenges in prostate cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Hao Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
91
|
Zhang W, Xiao Y, Zhu X, Zhang Y, Xiang Q, Wu S, Song X, Zhao J, Yuan R, Li Q, Xiao B, Li L. Integrative Pan-Cancer Analysis Reveals the Oncogenic Role of MND1 and Validation of MND1's Role in Breast Cancer. J Inflamm Res 2024; 17:4721-4746. [PMID: 39051055 PMCID: PMC11268618 DOI: 10.2147/jir.s458832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose Meiotic nuclear division 1 (MND1) is a meiosis-specific protein that promotes lung adenocarcinoma progression. However, its expression and biological function across cancers remain largely unexplored. Patients and Methods The expression, prognostic significance, mutation status, and methylation profile of MND1 in various cancers were comprehensively analyzed using the TIMER, GTEX, Kaplan-Meier plotter, cBioPortal, and GSCA databases. Additionally, we constructed a PPI network, enrichment analysis and single-cell transcriptomic sequencing to elucidate the underlying mechanism of MND1. Furthermore, we investigated the association between MND1 expression and drug sensitivity using CellMiner. Moreover, we also explored the correlation between MND1 expression and immune infiltration. Finally, we validated the functional role of MND1 in breast cancer through IHC staining, CCK8, EdU, colony formation, and flow cytometry assays. Results MND1 has been reported to be highly expressed in Pan-cancer, High MND1 expression was significantly associated with poor prognosis in cancers. Additionally, MND1 mutation frequency is high in most cancers, and its expression correlates with methylation. Furthermore, MND1 expression significantly correlates with immune checkpoint blockade (ICB) markers, including PD-L1, PD-1, and CTLA-4. The PPI network reveals interactions between MND1 and PSMC3IP, BRCA1, and BRCA2. Enrichment analysis and single-cell sequencing indicate that MND1 positively correlates with cell cycle. ROC curve reveals favorable diagnostic efficacy of MND1 in breast cancer. In vitro, MND1 overexpression promotes breast cancer cell proliferation and increases the expression of key cell cycle regulators (CDK4, CDK6, and cyclin D3), accelerating the G1/S phase transition and leading to abnormal breast cancer cell proliferation. The immunohistochemical analysis revealed a robust expression of MND1 in breast cancer tissues, exhibiting a significant positive correlation with PD-L1 and FOXP3. Conclusion MND1 is an oncogene and may serve as a biomarker for cancer prognosis and immunotherapy. Targeting MND1 may be a potential tumor treatment strategy.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
- Department of Laboratory Medicine, Suzhou Municipal Hospital, Affiliated to Nanjing Medical University, Suzhou, 21500, People’s Republic of China
| | - Yuhan Xiao
- School of Public Health, Dali University, Dali, 671000, People’s Republic of China
| | - Xin Zhu
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Yanxia Zhang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Qin Xiang
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Xiaoyu Song
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Junxiu Zhao
- School of Public Health, Dali University, Dali, 671000, People’s Republic of China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Qiguang Li
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Bin Xiao
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| | - Linhai Li
- Department of Laboratory Medicine, The Affiliated Qingyuan Hospital (Qingyuan People’s Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People’s Republic of China
| |
Collapse
|
92
|
Yan Z, Zhang Z, Chen Y, Xu J, Wang J, Wang Z. Enhancing cancer therapy: the integration of oncolytic virus therapy with diverse treatments. Cancer Cell Int 2024; 24:242. [PMID: 38992667 PMCID: PMC11238399 DOI: 10.1186/s12935-024-03424-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
As one of the significant challenges to human health, cancer has long been a focal point in medical treatment. With ongoing advancements in the field of medicine, numerous methodologies for cancer therapy have emerged, among which oncolytic virus therapy has gained considerable attention. However, oncolytic viruses still exhibit limitations. Combining them with various therapies can further enhance the efficacy of cancer treatment, offering renewed hope for patients. In recent research, scientists have recognized the promising prospect of amalgamating oncolytic virus therapy with diverse treatments, potentially surmounting the restrictions of singular approaches. The central concept of this combined therapy revolves around leveraging oncolytic virus to incite localized tumor inflammation, augmenting the immune response for immunotherapeutic efficacy. Through this approach, the patient's immune system can better recognize and eliminate cancer cells, simultaneously reducing tumor evasion mechanisms against the immune system. This review delves deeply into the latest research progress concerning the integration of oncolytic virus with diverse treatments and its role in various types of cancer therapy. We aim to analyze the mechanisms, advantages, potential challenges, and future research directions of this combination therapy. By extensively exploring this field, we aim to instill renewed hope in the fight against cancer.
Collapse
Affiliation(s)
- Zhuo Yan
- Department of Clinical Medical Laboratory Center, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, Zhejiang, China
| | - Zhengbo Zhang
- Department of Clinical Medical Laboratory Center, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China
| | - Yanan Chen
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, Zhejiang, China
| | - Jianghua Xu
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, Zhejiang, China
| | - Jilong Wang
- Department of Clinical Medical Laboratory Center, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China.
- Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, Zhejiang, China.
| | - Zhangquan Wang
- Department of Clinical Medical Laboratory Center, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People's Hospital), Hangzhou Medical College, Taizhou, 317200, Zhejiang, China.
| |
Collapse
|
93
|
Yuan Z, Zhang Y, Wang X, Wang X, Ren S, He X, Su J, Zheng A, Guo S, Chen Y, Deng S, Wu X, Li M, Du F, Zhao Y, Shen J, Wang Z, Xiao Z. The investigation of oncolytic viruses in the field of cancer therapy. Front Oncol 2024; 14:1423143. [PMID: 39055561 PMCID: PMC11270537 DOI: 10.3389/fonc.2024.1423143] [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: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Oncolytic viruses (OVs) have emerged as a potential strategy for tumor treatment due to their ability to selectively replicate in tumor cells, induce apoptosis, and stimulate immune responses. However, the therapeutic efficacy of single OVs is limited by the complexity and immunosuppressive nature of the tumor microenvironment (TME). To overcome these challenges, engineering OVs has become an important research direction. This review focuses on engineering methods and multi-modal combination therapies for OVs aimed at addressing delivery barriers, viral phagocytosis, and antiviral immunity in tumor therapy. The engineering approaches discussed include enhancing in vivo immune response, improving replication efficiency within the tumor cells, enhancing safety profiles, and improving targeting capabilities. In addition, this review describes the potential mechanisms of OVs combined with radiotherapy, chemotherapy, cell therapy and immune checkpoint inhibitors (ICIs), and summarizes the data of ongoing clinical trials. By continuously optimizing engineering strategies and combination therapy programs, we can achieve improved treatment outcomes and quality of life for cancer patients.
Collapse
Affiliation(s)
- Zijun Yuan
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anfu Zheng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Sipeng Guo
- Research And Experiment Center, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zechen Wang
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
| | - Zhangang Xiao
- Gulin Traditional Chinese Medicine Hospital, Luzhou, China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
94
|
Gkouvi A, Tsiogkas SG, Bogdanos DP, Gika H, Goulis DG, Grammatikopoulou MG. Proteomics in Patients with Fibromyalgia Syndrome: A Systematic Review of Observational Studies. Curr Pain Headache Rep 2024; 28:565-586. [PMID: 38652420 PMCID: PMC11271354 DOI: 10.1007/s11916-024-01244-4] [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] [Accepted: 03/16/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE OF REVIEW Fibromyalgia syndrome (FMS) is a disease of unknown pathophysiology, with the diagnosis being based on a set of clinical criteria. Proteomic analysis can provide significant biological information for the pathophysiology of the disease but may also reveal biomarkers for diagnosis or therapeutic targets. The present systematic review aims to synthesize the evidence regarding the proteome of adult patients with FMS using data from observational studies. RECENT FINDINGS An extensive literature search was conducted in MEDLINE/PubMed, CENTRAL, and clinicaltrials.gov from inception until November 2022. The study protocol was published in OSF. Two independent reviewers evaluated the studies and extracted data. The quality of studies was assessed using the modified Newcastle-Ottawa scale adjusted for proteomic research. Ten studies fulfilled the protocol criteria, identifying 3328 proteins, 145 of which were differentially expressed among patients with FMS against controls. The proteins were identified in plasma, serum, cerebrospinal fluid, and saliva samples. The control groups included healthy individuals and patients with pain (inflammatory and non-inflammatory). The most important proteins identified involved transferrin, α-, β-, and γ-fibrinogen chains, profilin-1, transaldolase, PGAM1, apolipoprotein-C3, complement C4A and C1QC, immunoglobin parts, and acute phase reactants. Weak correlations were observed between proteins and pain sensation, or quality of life scales, apart from the association of transferrin and a2-macroglobulin with moderate-to-severe pain sensation. The quality of included studies was moderate-to-good. FMS appears to be related to protein dysregulation in the complement and coagulation cascades and the metabolism of iron. Several proteins may be dysregulated due to the excessive oxidative stress response.
Collapse
Affiliation(s)
- Arriana Gkouvi
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Sotirios G Tsiogkas
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Dimitrios P Bogdanos
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece.
| | - Helen Gika
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Biomic_AUTh, Balkan Center Thermi B1.4, GR-57001, Thessaloniki, Greece
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, GR-54124, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria G Grammatikopoulou
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
95
|
Wang R, Lu JY, Herbert D, Lieberman JA, Meltzer HY, Tiwari AK, Remington G, Kennedy JL, Zai CC. Analysis of schizophrenia-associated genetic markers in the HLA region as risk factors for tardive dyskinesia. Hum Psychopharmacol 2024; 39:e2898. [PMID: 38676936 DOI: 10.1002/hup.2898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 04/29/2024]
Abstract
OBJECTIVES The pathology of Tardive Dyskinesia (TD) has yet to be fully understood, but there have been proposed hypotheses for the cause of this condition. Our team previously reported a possible association of TD with the Complement Component C4 gene in the HLA region. In this study, we explored the HLA region further by examining two previously identified schizophrenia-associated HLA-region single-nucleotide polymorphisms (SNPs), namely rs13194504 and rs210133. METHODS The SNPs rs13194504 and rs210133 were tested for association with the occurrence and severity of TD in a sample of 172 schizophrenia patients who were recruited for four studies from three different clinical sites in Canada and USA. RESULTS The rs13194504 AA genotype was associated with decreased severity for TD as measured by Abnormal Involuntary Movement Scale (AIMS) scores (p = 0.047) but not for TD occurrence. SNP rs210133 was not significantly associated with either TD occurrence or AIMS scores. CONCLUSION Our findings suggest that the rs13194504 AA genotype may play a role in TD severity, while SNP rs210133 may not have a major role in the risk or severity of TD.
Collapse
Affiliation(s)
- Ruoyu Wang
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Justin Y Lu
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Deanna Herbert
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jeffrey A Lieberman
- Columbia University, New York State Psychiatric Institute, New York City, New York, USA
| | - Herbert Y Meltzer
- Psychiatry and Behavioral Sciences, Pharmacology and Physiology, Chemistry of Life Processes Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Arun K Tiwari
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Gary Remington
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Clement C Zai
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
96
|
Prince A. Immunometabolites Direct the Pathogenesis of Bacterial Infection. J Innate Immun 2024; 16:367-369. [PMID: 38952110 PMCID: PMC11540411 DOI: 10.1159/000540093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- Alice Prince
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
97
|
Theofilou VI, Ghita I, Elnaggar M, Chaisuparat R, Papadimitriou JC, Bentzen SM, Dyalram D, Lubek JE, Ord RA, Younis RH. Histological pattern of tumor inflammation and stromal density correlate with patient demographics and immuno-oncologic transcriptional profile in oral squamous cell carcinoma. FRONTIERS IN ORAL HEALTH 2024; 5:1408072. [PMID: 38903181 PMCID: PMC11187265 DOI: 10.3389/froh.2024.1408072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/07/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction Oral squamous cell carcinoma (OSCC) is the most prevalent oral malignancy, with emerging interest in the characterization of its tumor microenvironment. Herein, we present a comprehensive histological analysis of OSCC stromal density and inflammation and their relationship with patient demographics, clinicopathologic features and immuno-oncologic signatures. Materials-methods Eighty-seven completely excised OSCC tissues were prospectively collected and scored for histopathologic inflammatory subtypes [HIS]-inflamed (INF), immune-excluded (IE) and immune-desert (ID), peritumoral stromal inflammation (PTSI), and peritumoral stromal fibrosis (PTSF). Scoring of inflammation was complemented by Semaphorin 4D immunohistochemistry. NanoString differential gene expression (DGE) analysis was conducted for eight OSCC cases representative of the inflammatory and stromal subtypes and the demographic groups. Results PTSF correlated with male gender (p = 0.0043), smoking (p = 0.0455), alcohol consumption (p = 0.0044), increased tumor size (p = 0.0054), and advanced stage (p = 0.002). On the contrary, PTSI occurred predominantly in females (p = 0.0105), non-drinkers (p = 0.0329), and small tumors (p = 0.0044). Transcriptionally, decreased cytokine signaling, and oncogenic pathway activation were observed in HIS-IE. Smokers and males displayed decreased global immune-cell levels and myeloid-cell predominance. Conclusion Our work describes OSCC stromal and inflammatory phenotypes in correlation with distinct patient groups and DGE, highlighting the translational potential of characterizing the tumor microenvironment for optimal patient stratification.
Collapse
Affiliation(s)
- Vasileios Ionas Theofilou
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Ioana Ghita
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Manar Elnaggar
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Risa Chaisuparat
- Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - John C. Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Soren M. Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Biostatistics Core, Institute of Clinical and Translational Research, University of Maryland, Baltimore, MD, United States
- Biostatistics Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center,Baltimore, MD, United States
| | - Donita Dyalram
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Joshua E. Lubek
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Robert A. Ord
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Rania H. Younis
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
- Department of Oral Pathology, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| |
Collapse
|
98
|
Govender S, David M, Naicker T. Is the Complement System Dysregulated in Preeclampsia Comorbid with HIV Infection? Int J Mol Sci 2024; 25:6232. [PMID: 38892429 PMCID: PMC11172754 DOI: 10.3390/ijms25116232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
South Africa is the epicentre of the global HIV pandemic, with 13.9% of its population infected. Preeclampsia (PE), a hypertensive disorder of pregnancy, is often comorbid with HIV infection, leading to multi-organ dysfunction and convulsions. The exact pathophysiology of preeclampsia is triggered by an altered maternal immune response or defective development of maternal tolerance to the semi-allogenic foetus via the complement system. The complement system plays a vital role in the innate immune system, generating inflammation, mediating the clearance of microbes and injured tissue materials, and a mediator of adaptive immunity. Moreover, the complement system has a dual effect, of protecting the host against HIV infection and enhancing HIV infectivity. An upregulation of regulatory proteins has been implicated as an adaptive phenomenon in response to elevated complement-mediated cell lysis in HIV infection, further aggravated by preeclamptic complement activation. In light of the high prevalence of HIV infection and preeclampsia in South Africa, this review discusses the association of complement proteins and their role in the synergy of HIV infection and preeclampsia in South Africa. It aims to identify women at elevated risk, leading to early diagnosis and better management with targeted drug therapy, thereby improving the understanding of immunological dysregulation.
Collapse
Affiliation(s)
| | | | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (S.G.); (M.D.)
| |
Collapse
|
99
|
Rahman MA, Apu EH, Rakib-Uz-Zaman SM, Chakraborti S, Bhajan SK, Taleb SA, Shaikh MH, Jalouli M, Harrath AH, Kim B. Exploring Importance and Regulation of Autophagy in Cancer Stem Cells and Stem Cell-Based Therapies. Cells 2024; 13:958. [PMID: 38891090 PMCID: PMC11171866 DOI: 10.3390/cells13110958] [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: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Autophagy is a globally conserved cellular activity that plays a critical role in maintaining cellular homeostasis through the breakdown and recycling of cellular constituents. In recent years, there has been much emphasis given to its complex role in cancer stem cells (CSCs) and stem cell treatment. This study examines the molecular processes that support autophagy and how it is regulated in the context of CSCs and stem cell treatment. Although autophagy plays a dual role in the management of CSCs, affecting their removal as well as their maintenance, the intricate interaction between the several signaling channels that control cellular survival and death as part of the molecular mechanism of autophagy has not been well elucidated. Given that CSCs have a role in the development, progression, and resistance to treatment of tumors, it is imperative to comprehend their biological activities. CSCs are important for cancer biology because they also show a tissue regeneration model that helps with organoid regeneration. In other words, the manipulation of autophagy is a viable therapeutic approach in the treatment of cancer and stem cell therapy. Both synthetic and natural substances that target autophagy pathways have demonstrated promise in improving stem cell-based therapies and eliminating CSCs. Nevertheless, there are difficulties associated with the limitations of autophagy in CSC regulation, including resistance mechanisms and off-target effects. Thus, the regulation of autophagy offers a versatile strategy for focusing on CSCs and enhancing the results of stem cell therapy. Therefore, understanding the complex interactions between autophagy and CSC biology would be essential for creating therapeutic treatments that work in both regenerative medicine and cancer treatment.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Global Biotechnology and Biomedical Research Network (GBBRN), Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Ehsanul Hoque Apu
- Department of Biomedical Sciences, College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA;
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - S. M Rakib-Uz-Zaman
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Somdeepa Chakraborti
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (S.M.R.-U.-Z.); (S.C.)
| | - Sujay Kumar Bhajan
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science & Technology University, Gopalganj 8100, Bangladesh;
| | - Shakila Afroz Taleb
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06510, USA;
| | - Mushfiq H. Shaikh
- Department of Otolaryngology—Head and Neck Surgery, Western University, London, ON N6A 4V2, Canada;
| | - Maroua Jalouli
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Abdel Halim Harrath
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, 1-5 Hoegidong, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
100
|
Mao X, Wang Y, Zhang T, Ma J, Zhao J, Xu D. Dietary arginine regulates the growth performance, antioxidant capacity, and immune response in Culter alburnus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1251-1264. [PMID: 38578588 DOI: 10.1007/s10695-024-01334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Culter alburnus is sensitive to stressors. Arginine is a precursor of nitric oxide, which can effectively relieve the level of oxidative stress and improve the antioxidant and immune capacity of fish. The effect of different arginine levels on topmouth culter (Culter alburnus) fry development performance, liver antioxidant capacity, and immune parameters were investigated in this study. Five diets (1.96%, ARG1, control group; 2.28%, ARG2; 2.52%, ARG3; 2.81%, ARG4; 3.09%, ARG5) were used to feed fry (initial weight 0.31 ± 0.01 g) for 8 weeks. The data showed that the final weight (FW), weight gain rate (WGR), and specific growth rate (SGR) of the ARG3 and ARG4 groups were significantly improved, while the feed conversion ratio (FCR) reduced significantly. Compared with the ARG1 group, all groups remarkably reduced the crude ash content of the whole body. The activity of hepatic superoxide dismutase (SOD) and the content of hepatic glutathione (GSH) were significantly increased in the ARG3 and ARG4 groups, while the malondialdehyde (MDA) content was significantly decreased. Compared with the ARG1 group, arginine levels in ARG2, ARG3, and ARG4 groups up-regulated the expression levels of Nrf2, down-regulated the gene expression level of Keap1 in the liver. And the expression of Nrf2/Keap1 pathway downstream genes Mn-SOD and CAT was up-regulated in ARG2 and ARG3 groups. Furthermore, the expression levels of MyD88 and IL-1β were down-regulated, and the anti-inflammatory gene TGF-β expression levels were up-regulated in the ARG2, ARG3, and ARG4 groups. Additionally, compared to the ARG1 group, there was a significant increase in the relative expression levels of the C3 and C4 genes in the ARG4 group. In conclusion, 2.28-2.81% dietary arginine levels improved the growth performance, promoted antioxidant capacity, and enhance immune response. The optimal level of arginine was determined by the quadratic regression analysis of SGR and FCR to be 2.55% of diet (5.43% of dietary protein) and 2.53% of diet (5.38% of dietary protein), accordingly.
Collapse
Affiliation(s)
- Xinyuan Mao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
- Graduate School of Chinese Academy of Agricultural Science, Beijing, 100081, China
| | - Yue Wang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
- Graduate School of Chinese Academy of Agricultural Science, Beijing, 100081, China
| | - Ting Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Junlei Ma
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China
| | - Jihao Zhao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
- Graduate School of Chinese Academy of Agricultural Science, Beijing, 100081, China
| | - Dongpo Xu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China.
| |
Collapse
|