1
|
Zhao Y, Xiang C, Roy BC, Bruce HL, Blecker C, Zhang Y, Liu C, Zhang D, Chen L, Huang C. Apoptosis and its role in postmortem meat tenderness: A comprehensive review. Meat Sci 2025; 219:109652. [PMID: 39265386 DOI: 10.1016/j.meatsci.2024.109652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
Tenderness is considered a crucial attribute of postmortem meat quality, directly influencing consumers' preferences and industrial economic benefits. The degradation of myofibrillar proteins by endogenous enzymes within muscle fibers is believed to be the most effective pathway for meat tenderization. After animals are slaughtered and exsanguinated, there is a significant accumulation of reactive oxygen species (ROS), and a dramatic depletion of adenosine triphosphate (ATP) in muscle, leading to inevitable cell death. Caspases are activated in postmortem muscle cells, which disrupt the cell structure and improve meat tenderness through protein hydrolysis. In this review, we systematically summarized the three primary types of cell death studied in postmortem muscle: apoptosis, autophagy and necrosis. Furthermore, we emphasized the molecular mechanisms of apoptosis and its corresponding apoptotic pathways (mitochondrial apoptosis, death receptors, and endoplasmic reticulum stress) that affect meat tenderness during muscle conversion to meat. Additionally, factors affecting apoptosis were comprehensively discussed, such as ROS, heat shock proteins, calcium (Ca2+)/calpains, and Bcl-2 family proteins. Finally, this comprehensive review of existing research reveals that apoptosis is mainly mediated by the mitochondrial pathway. This ultimately leads to myofibrillar proteins degradation through caspase activation, improving meat tenderness. This review summarizes the research progress on postmortem muscle apoptosis and its molecular mechanisms in meat tenderization. We hope this will enhance understanding of postmortem meat tenderness and provide a theoretical basis for meat tenderization techniques development in the future.
Collapse
Affiliation(s)
- Yingxin Zhao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Can Xiang
- Department of Flavor Chemistry, Institute of Food Science and Biotechnology, University of Hohenheim, Fruwirthstraße 12, 70599 Stuttgart, Germany
| | - Bimol C Roy
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Heather L Bruce
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Christophe Blecker
- Gembloux Agro-Bio Tech, Unit of Food Science and Formulation, University of Liège, Avenue de la Faculté d'Agronomie 2, Gembloux B-5030, Belgium
| | - Yanyan Zhang
- Department of Flavor Chemistry, Institute of Food Science and Biotechnology, University of Hohenheim, Fruwirthstraße 12, 70599 Stuttgart, Germany
| | - Chongxin Liu
- Institute of Food Science and Technology, Chinese Academy of Agriculture Sciences, Key Laboratory of Agro-Products Quality & Safety Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Dequan Zhang
- Institute of Food Science and Technology, Chinese Academy of Agriculture Sciences, Key Laboratory of Agro-Products Quality & Safety Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Li Chen
- Institute of Food Science and Technology, Chinese Academy of Agriculture Sciences, Key Laboratory of Agro-Products Quality & Safety Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing 100193, China.
| | - Caiyan Huang
- Institute of Food Science and Technology, Chinese Academy of Agriculture Sciences, Key Laboratory of Agro-Products Quality & Safety Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing 100193, China; Gembloux Agro-Bio Tech, Unit of Food Science and Formulation, University of Liège, Avenue de la Faculté d'Agronomie 2, Gembloux B-5030, Belgium.
| |
Collapse
|
2
|
Ahmed SAA, Ibrahim RE, Younis EM, Abdelwarith AA, Faroh KY, El Gamal SA, Badr S, Khamis T, Mansour AT, Davies SJ, ElHady M. Antagonistic Effect of Zinc Oxide Nanoparticles Dietary Supplementation Against Chronic Copper Waterborne Exposure on Growth, Behavioral, Biochemical, and Gene Expression Alterations of African Catfish, Clarias gariepinus (Burchell, 1822). Biol Trace Elem Res 2024; 202:5697-5713. [PMID: 38416342 DOI: 10.1007/s12011-024-04115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024]
Abstract
The harmful impact of waterborne copper (Cu) as a common abiotic stressor in aquatic environments has gained much more interest. The present study aimed to investigate the utilization of zinc oxide nanoparticles (ZnONPs) dietary supplementation to mitigate the chronic toxicity of Cu in African catfish (Clarias gariepinus). Two hundred and forty fish (92.94 ± 0.13 g) were assigned into six groups for 60 days. Control (C), ZnONPs20, and ZnONPs30 groups were fed on basal diets fortified with 0, 20, and 30 mg kg-1 ZnONPs without Cu exposure. Cu, Cu + ZnONPs20, and Cu + ZnONPs30 groups were exposed to Cu at a dose of 10 mg L-1 and fed on basal diets fortified with 0, 20, and 30 mg kg-1 ZnONPs, respectively. The results revealed that the Cu-exposed fish experienced abnormal clinical signs and behavioral changes. The growth indices and acetylcholine esterase activity were significantly decreased (P < 0.05) in the Cu group. Meanwhile, hepatorenal and serum stress indices (P < 0.05) were significantly elevated with chronic Cu exposure. In addition, a higher expression of stress (P < 0.05) (heat shock protein 60 and hypoxia-inducible factor-1 alpha) and apoptotic-related genes (C/EBP homologous protein, caspase-3, and Bcl-2 Associated X-protein) with down-regulation (P < 0.05) of the anti-apoptotic-related genes (B-cell lymphoma 2 and proliferating cell nuclear antigen) was noticed in the Cu-exposed fish. Histopathological alterations in the gills, liver, kidney, and spleen were markedly reported in the Cu-exposed group. The dietary supplementation with ZnONPs significantly alleviated the negative impacts of chronic waterborne-Cu exposure on growth performance, physiological changes, gene expression, and tissue architecture, especially at 30 mg kg-1 diet level. In particular, the inclusion of ZnONPs at the 30 mg kg-1 diet level produced better outcomes than the 20 mg kg-1 diet. Overall, ZnONPs could be added as a feed supplement in the C. gariepinus diet to boost the fish's health and productivity and alleviate the stress condition brought on by Cu exposure.
Collapse
Affiliation(s)
- Shaimaa A A Ahmed
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Rowida E Ibrahim
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt.
| | - Elsayed M Younis
- Department of Zoology, College of Science, King Saud University, PO Box 2455, 11451, Riyadh, Saudi Arabia
| | - Abdelwahab A Abdelwarith
- Department of Zoology, College of Science, King Saud University, PO Box 2455, 11451, Riyadh, Saudi Arabia
| | - Khaled Yehia Faroh
- Nanotechnology and Advanced Materials Central Lab, Agriculture Research Center (ARC), P.O. Box 12619, Giza, Egypt
| | - Samar A El Gamal
- Department of Fish Diseases, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Giza, Egypt
| | - Shereen Badr
- Department of Clinical Pathology, Mansoura Branch, Agriculture Research Center (ARC), Animal Health Research Institute (AHRI), Giza, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| | - Abdallah Tageldein Mansour
- Fish and Animal Production Department, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria, 21531, Egypt.
| | - Simon J Davies
- Aquaculture Nutrition Research Unit ANRU, Carna Research Station, Ryan Institute, College of Science and Engineering, University of Galway, Galway, H91V8Y1, Ireland
| | - Mohamed ElHady
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
3
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [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: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
4
|
Masaeli F, Omoomi S, Shafiee F. DNA fragmentation factor 40-based therapeutic approaches for cancer: a review article. Med Oncol 2024; 41:264. [PMID: 39397131 DOI: 10.1007/s12032-024-02511-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
DNA Fragmentation Factor (DFF) is a heterodimer protein involved in DNA fragmentation during apoptosis, which acts as a trigger downstream of caspase-3 activation. DFF40 catalytically active homo-oligomers break down chromosomal DNA. Previous scientific investigations have revealed a link between DFF40 expression changes and various cancers. DFF40 deletion or down-regulation has been observed in some cancers. Consequently, therapeutic strategies involving the DFF40 molecule compensating led to an increased rate of cancer cell apoptosis. In this review article, we aimed to introduce cancers with low expression of this protein first. The second part of this paper focuses on studies that utilized exogenous DFF40 protein produced by recombinant DNA technology and surveyed during in vitro and in vivo tests. Finally, compensation for diminished expression of the mentioned protein via gene therapy-based techniques to make up for this apoptotic molecule's low expression is the topic of the last part of this review article.
Collapse
Affiliation(s)
- Faezeh Masaeli
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran
| | - Saba Omoomi
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran
| | - Fatemeh Shafiee
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Hezar Jarib Ave., Isfahan, Iran.
- Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
5
|
La Marca JE, Kelly GL, Strasser A, Diepstraten ST. Don't fear the reaper: The role of regulated cell death in tumorigenesis and BH3-mimetics for cancer therapy. Dev Cell 2024; 59:2532-2548. [PMID: 39378839 DOI: 10.1016/j.devcel.2024.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/20/2024] [Accepted: 06/20/2024] [Indexed: 10/10/2024]
Abstract
From its earliest characterization, it has been recognized that there is a role for regulated (programmed) cell death in cancer. As our understanding of the different types of programmed cell death processes and their molecular control has advanced, so have the technologies that allow us to manipulate these processes to, for example, fight against cancer. In this review, we describe the roles of the different forms of regulated cell death in the development of cancer as well as their potential therapeutic exploitation. In that vein, we explore the development and use of BH3-mimetics, a unique class of drugs that can directly activate the apoptotic cell death machinery to treat cancer. Finally, we address key challenges that face the field to improve the use of these therapeutics and the efforts that are being undertaken to do so.
Collapse
Affiliation(s)
- John E La Marca
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Genome Engineering and Cancer Modelling Program, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC, Australia.
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Sarah T Diepstraten
- The Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
6
|
Wang WG, Jiang XF, Zhang C, Zhan XP, Cheng JG, Tao LM, Xu WP, Li Z, Zhang Y. Avermectin induced vascular damage in zebrafish larvae: association with mitochondria-mediated apoptosis and VEGF/Notch signaling pathway. JOURNAL OF HAZARDOUS MATERIALS 2024; 477:135376. [PMID: 39111175 DOI: 10.1016/j.jhazmat.2024.135376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/21/2024] [Accepted: 07/28/2024] [Indexed: 08/17/2024]
Abstract
Avermectin is a highly effective insecticide that has been widely used in agriculture since the 1990s. In recent years, the safety of avermectin for non-target organisms has received much attention. The vasculature is important organs in the body and participate in the composition of other organs. However, studies on the vascular safety of avermectin are lacking. The vasculature of zebrafish larvae is characterized by ease of observation and it is a commonly used model for vascular studies. Therefore, zebrafish larvae were used to explore the potential risk of avermectin on the vasculature. The results showed that avermectin induced vascular damage throughout the body of zebrafish larvae, including the head, eyes, intestine, somite, tail and other vasculature. The main forms of damage are reduction in vascular diameter, vascular area and vascular abundance. Meanwhile, avermectin induced a decrease in the number of endothelial cells and apoptosis within the vasculature. In addition, vascular damage may be related to impairment of mitochondrial function and mitochondria-mediated apoptosis. Finally, exploration of the molecular mechanisms revealed abnormal alterations in the expression of genes related to the VEGF/Notch signaling pathway. Therefore, the VEGF/Notch signaling pathway may be an important mechanism for avermectin-induced vascular damage in zebrafish larvae. This study demonstrates the vascular toxicity of avermectin in zebrafish larvae and reveals the possible molecular mechanism, which would hopefully draw more attention to the safety of avermectin in non-target organisms.
Collapse
Affiliation(s)
- Wei-Guo Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xu-Feng Jiang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, United States
| | - Xiu-Ping Zhan
- Shanghai Agricultural Technology Extension Center, Shanghai 201103, China
| | - Jia-Gao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Li-Ming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wen-Ping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
7
|
Kodra AL, Singh AS, de la Cova C, Ziosi M, Johnston LA. The Drosophila tumor necrosis factor Eiger promotes Myc supercompetition independent of canonical Jun N-terminal kinase signaling. Genetics 2024; 228:iyae107. [PMID: 38985651 PMCID: PMC11373512 DOI: 10.1093/genetics/iyae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/12/2024] Open
Abstract
Numerous factors have been implicated in the cell-cell interactions that lead to elimination of cells via cell competition, a context-dependent process of cell selection in somatic tissues that is based on comparisons of cellular fitness. Here, we use a series of genetic tests in Drosophila to explore the relative contribution of the pleiotropic cytokine tumor necrosis factor α (TNFα) in Myc-mediated cell competition (also known as Myc supercompetition or Myc cell competition). We find that the sole Drosophila TNF, Eiger (Egr), its receptor Grindelwald (Grnd/TNF receptor), and the adaptor proteins Traf4 and Traf6 are required to eliminate wild-type "loser" cells during Myc cell competition. Although typically the interaction between Egr and Grnd leads to cell death by activating the intracellular Jun N-terminal kinase (JNK) stress signaling pathway, our experiments reveal that many components of canonical JNK signaling are dispensable for cell death in Myc cell competition, including the JNKKK Tak1, the JNKK Hemipterous and the JNK Basket. Our results suggest that Egr/Grnd signaling participates in Myc cell competition but functions in a role that is largely independent of the JNK signaling pathway.
Collapse
Affiliation(s)
- Albana L Kodra
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Aditi Sharma Singh
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Claire de la Cova
- Department of Biological Sciences, University of Wisconsin, Milwaukee, WI 53201, USA
| | | | - Laura A Johnston
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
8
|
Thuner J, Cognard J, Belot A. How to treat monogenic SLE? Best Pract Res Clin Rheumatol 2024; 38:101962. [PMID: 38876818 DOI: 10.1016/j.berh.2024.101962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024]
Abstract
Systemic lupus erythematosus is a rare and life-threatening autoimmune disease characterized by autoantibodies against double-stranded DNA, with an immunopathology that remains partially unclear. New insights into the disease have been provided by the discovery of key mutations leading to the development of monogenic SLE, occurring in the context of early-onset disease, syndromic lupus, or familial clustering. The increased frequency of discovering these mutations in recent years, thanks to the advent of genetic screening, has greatly enhanced our understanding of the immunopathogenesis of SLE. These monogenic defects include defective clearance of apoptotic bodies, abnormalities in nucleic acid sensing, activation of the type-I interferon pathway, and the breakdown of tolerance through B or T cell activation or lymphocyte proliferation due to anomalies in TLR signalling and/or NFκB pathway overactivation. The translation of genetic discoveries into therapeutic strategies is presented here, within the framework of personalized therapy.
Collapse
Affiliation(s)
- Jonathan Thuner
- Internal Medicine Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Jade Cognard
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France
| | - Alexandre Belot
- Pediatric Rheumatology, Nephrology, Dermatology Department, CMR RAISE, Women-Mother-Child Hospital, Hospices Civils de Lyon, Bron, France; CIRI, Centre International de Recherche en Infectiologie/International Center for Infectiology Research, Université de Lyon, INSERM, Institut National de La Santé Et de La Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Lyon, France; CNRS, Centre National de La Recherche Scientifique, UMR5308, Lyon, France.
| |
Collapse
|
9
|
Mertelsmann AM, Bowers SF, Wright D, Maganga JK, Mazigo HD, Ndhlovu LC, Changalucha JM, Downs JA. Effects of Schistosoma haematobium infection and treatment on the systemic and mucosal immune phenotype, gene expression and microbiome: A systematic review. PLoS Negl Trop Dis 2024; 18:e0012456. [PMID: 39250522 PMCID: PMC11412685 DOI: 10.1371/journal.pntd.0012456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/19/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Urogenital schistosomiasis caused by Schistosoma haematobium affects approximately 110 million people globally, with the majority of cases in low- and middle-income countries. Schistosome infections have been shown to impact the host immune system, gene expression, and microbiome composition. Studies have demonstrated variations in pathology between schistosome subspecies. In the case of S. haematobium, infection has been associated with HIV acquisition and bladder cancer. However, the underlying pathophysiology has been understudied compared to other schistosome species. This systematic review comprehensively investigates and assimilates the effects of S. haematobium infection on systemic and local host mucosal immunity, cellular gene expression and microbiome. METHODS We conducted a systematic review assessing the reported effects of S. haematobium infections and anthelmintic treatment on the immune system, gene expression and microbiome in humans and animal models. This review followed PRISMA guidelines and was registered prospectively in PROSPERO (CRD42022372607). Randomized clinical trials, cohort, cross-sectional, case-control, experimental ex vivo, and animal studies were included. Two reviewers performed screening independently. RESULTS We screened 3,177 studies and included 94. S. haematobium was reported to lead to: (i) a mixed immune response with a predominant type 2 immune phenotype, increased T and B regulatory cells, and select pro-inflammatory cytokines; (ii) distinct molecular alterations that would compromise epithelial integrity, such as increased metalloproteinase expression, and promote immunological changes and cellular transformation, specifically upregulation of genes p53 and Bcl-2; and (iii) microbiome dysbiosis in the urinary, intestinal, and genital tracts. CONCLUSION S. haematobium induces distinct alterations in the host's immune system, molecular profile, and microbiome. This leads to a diverse range of inflammatory and anti-inflammatory responses and impaired integrity of the local mucosal epithelial barrier, elevating the risks of secondary infections. Further, S. haematobium promotes cellular transformation with oncogenic potential and disrupts the microbiome, further influencing the immune system and genetic makeup. Understanding the pathophysiology of these interactions can improve outcomes for the sequelae of this devastating parasitic infection.
Collapse
Affiliation(s)
- Anna M Mertelsmann
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Sheridan F Bowers
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
| | - Drew Wright
- Samuel J. Wood Library & C.V. Starr Biomedical Information Center, Weill Cornell Medical College, New York, New York, United States of America
| | - Jane K Maganga
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Humphrey D Mazigo
- Department of Parasitology and Entomology, Catholic University of Health and Allied Sciences, Mwanza, Tanzania
| | - Lishomwa C Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - John M Changalucha
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
| | - Jennifer A Downs
- Center for Global Health, Weill Cornell Medicine, New York, New York, United States of America
- Mwanza Intervention Trials Unit/National Institute for Medical Research, Mwanza, Tanzania
- Weill Bugando School of Medicine, Mwanza, Tanzania
| |
Collapse
|
10
|
Chan ET, Kural C. Targeting endocytosis to sensitize cancer cells to programmed cell death. Biochem Soc Trans 2024; 52:1703-1713. [PMID: 39092762 PMCID: PMC11519968 DOI: 10.1042/bst20231332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024]
Abstract
Evading programmed cell death (PCD) is a hallmark of cancer that allows tumor cells to survive and proliferate unchecked. Endocytosis, the process by which cells internalize extracellular materials, has emerged as a key regulator of cell death pathways in cancer. Many tumor types exhibit dysregulated endocytic dynamics that fuel their metabolic demands, promote resistance to cytotoxic therapies, and facilitate immune evasion. This review examines the roles of endocytosis in apoptotic resistance and immune escape mechanisms utilized by cancer cells. We highlight how inhibiting endocytosis can sensitize malignant cells to therapeutic agents and restore susceptibility to PCD. Strategies to modulate endocytosis for enhanced cancer treatment are discussed, including targeting endocytic regulatory proteins, altering membrane biophysical properties, and inhibiting Rho-associated kinases. While promising, challenges remain regarding the specificity and selectivity of endocytosis-targeting agents. Nonetheless, harnessing endocytic pathways represents an attractive approach to overcome apoptotic resistance and could yield more effective therapies by rendering cancer cells vulnerable to PCD. Understanding the interplay between endocytosis and PCD regulation is crucial for developing novel anticancer strategies that selectively induce tumor cell death.
Collapse
Affiliation(s)
- Emily T. Chan
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
| | - Cömert Kural
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, U.S.A
- Department of Physics, The Ohio State University, Columbus, OH 43210, U.S.A
| |
Collapse
|
11
|
Feng Y, Liu Z, Han C, Chen J, Lin X, Du W, Zhang Y, Dong B, Zheng Y, Lu K, Liang Q. Ficus hirta Vahl. alleviate LPS induced apoptosis via down-regulating of miR-411 in orange-spotted grouper (Epinephelus coioides) spleen cell. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 157:105191. [PMID: 38705263 DOI: 10.1016/j.dci.2024.105191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Ficus hirta Vahl. (FhV) has been shown to have antimicrobial and antiviral efficacy. To further ascertain the pharmacological properties of FhV., and to search for alternatives to antibiotics. An in vitro experiment was carried out to evaluate what influence FhV. would have on LPS-induced apoptosis. In this study, Fas, an apoptosis receptor, was cloned, which included a 5'-UTR of 39 bp, an ORF of 951 bp, a protein of 316 amino acids, and a 3'-UTR of 845 bp. EcFas was most strongly expressed in the spleen tissue of orange-spotted groupers. In addition, the apoptosis of fish spleen cells induced by LPS was concentration-dependent. Interestingly, appropriate concentrations of FhV. alleviated LPS-induced apoptosis. Inhibition of miR-411 further decreased the inhibitory effect of Fas on apoptosis, which reduced Bcl-2 expression and mitochondrial membrane potential, enhanced the protein expression of Bax and Fas. More importantly, the FhV. could activate miR-411 to improve this effect. In addition, luciferase reporter assays showed that miR-411 binds to Fas 3'-UTR to inhibit Fas expression. These findings provide evidence that FhV. alleviates LPS-induced apoptosis by activating miR-411 to inhibit Fas expression and, therefore, provided possible strategies for bacterial infections in fish.
Collapse
Affiliation(s)
- YuXin Feng
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - ZhengXinYu Liu
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - CaoYuan Han
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - JiaQian Chen
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - XinHao Lin
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - WangHao Du
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - Yu Zhang
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - BeiBei Dong
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - YiKai Zheng
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China
| | - KeXiang Lu
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China.
| | - QingJian Liang
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang, 316022, China; College of Life Science, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
12
|
Masum AA, Aoki S, Rahman MM, Hisamatsu Y. Chemical synthetic approaches to mimic the TRAIL: promising cancer therapeutics. RSC Med Chem 2024; 15:d4md00183d. [PMID: 39246747 PMCID: PMC11376135 DOI: 10.1039/d4md00183d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Apoptosis is programmed cell death that eliminates undesired cells to maintain homeostasis in metazoan. Aberration of this process may lead to cancer genesis. The tumor necrosis factor related apoptosis inducing ligand (TRAIL) induces apoptosis in cancer cells after ligation with death receptors (DR4/DR5) while sparing most normal cells. Therefore, strategies to induce apoptosis in cancer cells by mimicking the TRAIL emerge as a promising therapeutic tool. Hence, approaches are taken to develop TRAIL/DR-based cancer therapeutics. The recombinant soluble TRAIL (rhTRAIL) and death receptor agonistic antibodies were produced and tested pre-clinically and clinically. Pre-clinical and clinical trial data demonstrate that these therapeutics are safe and relatively well tolerated. But some of these therapeutics failed to exert adequate efficacy in clinical settings. Besides these biotechnologically derived therapeutics, a few chemically synthesized therapeutics are reported. Some of these therapeutics exert considerable efficacy in vitro and in vivo. In this review, we will discuss chemically synthesized TRAIL/DR-based therapeutics, their chemical and biological behaviour, design concepts and strategies that may contribute to further improvement of TRAIL/DR-based therapeutics.
Collapse
Affiliation(s)
- Abdullah-Al Masum
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Science and Technology, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science 2641 Yamazaki, Noda-shi Chiba 278-8510 Japan
| | - Md Mahbubur Rahman
- Department of Pharmaceutical Sciences, North South University Bashundhara R/A Dhaka-1229 Bangladesh
| | - Yosuke Hisamatsu
- Graduate School of Pharmaceutical Sciences, Nagoya City University Mizuho-Ku Nagoya 467-8603 Japan
| |
Collapse
|
13
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [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: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
14
|
Ancer-Rodríguez J, Gopar-Cuevas Y, García-Aguilar K, Chávez-Briones MDL, Miranda-Maldonado I, Ancer-Arellano A, Ortega-Martínez M, Jaramillo-Rangel G. Cell Proliferation and Apoptosis-Key Players in the Lung Aging Process. Int J Mol Sci 2024; 25:7867. [PMID: 39063108 PMCID: PMC11276691 DOI: 10.3390/ijms25147867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, the global lifespan has increased, resulting in a higher proportion of the population over 65 years. Changes that occur in the lung during aging increase the risk of developing acute and chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. During normal tissue homeostasis, cell proliferation and apoptosis create a dynamic balance that constitutes the physiological cell turnover. In basal conditions, the lungs have a low rate of cell turnover compared to other organs. During aging, changes in the rate of cell turnover in the lung are observed. In this work, we review the literature that evaluates the role of molecules involved in cell proliferation and apoptosis in lung aging and in the development of age-related lung diseases. The list of molecules that regulate cell proliferation, apoptosis, or both processes in lung aging includes TNC, FOXM1, DNA-PKcs, MicroRNAs, BCL-W, BCL-XL, TCF21, p16, NOX4, NRF2, MDM4, RPIA, DHEA, and MMP28. However, despite the studies carried out to date, the complete signaling pathways that regulate cell turnover in lung aging are still unknown. More research is needed to understand the changes that lead to the development of age-related lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico; (J.A.-R.); (Y.G.-C.); (M.-d.-L.C.-B.); (I.M.-M.); (A.A.-A.); (M.O.-M.)
| |
Collapse
|
15
|
Shouman S, El-Kholy N, Hussien AE, El-Derby AM, Magdy S, Abou-Shanab AM, Elmehrath AO, Abdelwaly A, Helal M, El-Badri N. SARS-CoV-2-associated lymphopenia: possible mechanisms and the role of CD147. Cell Commun Signal 2024; 22:349. [PMID: 38965547 PMCID: PMC11223399 DOI: 10.1186/s12964-024-01718-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/15/2024] [Indexed: 07/06/2024] Open
Abstract
T lymphocytes play a primary role in the adaptive antiviral immunity. Both lymphocytosis and lymphopenia were found to be associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While lymphocytosis indicates an active anti-viral response, lymphopenia is a sign of poor prognosis. T-cells, in essence, rarely express ACE2 receptors, making the cause of cell depletion enigmatic. Moreover, emerging strains posed an immunological challenge, potentially alarming for the next pandemic. Herein, we review how possible indirect and direct key mechanisms could contribute to SARS-CoV-2-associated-lymphopenia. The fundamental mechanism is the inflammatory cytokine storm elicited by viral infection, which alters the host cell metabolism into a more acidic state. This "hyperlactic acidemia" together with the cytokine storm suppresses T-cell proliferation and triggers intrinsic/extrinsic apoptosis. SARS-CoV-2 infection also results in a shift from steady-state hematopoiesis to stress hematopoiesis. Even with low ACE2 expression, the presence of cholesterol-rich lipid rafts on activated T-cells may enhance viral entry and syncytia formation. Finally, direct viral infection of lymphocytes may indicate the participation of other receptors or auxiliary proteins on T-cells, that can work alone or in concert with other mechanisms. Therefore, we address the role of CD147-a novel route-for SARS-CoV-2 and its new variants. CD147 is not only expressed on T-cells, but it also interacts with other co-partners to orchestrate various biological processes. Given these features, CD147 is an appealing candidate for viral pathogenicity. Understanding the molecular and cellular mechanisms behind SARS-CoV-2-associated-lymphopenia will aid in the discovery of potential therapeutic targets to improve the resilience of our immune system against this rapidly evolving virus.
Collapse
Affiliation(s)
- Shaimaa Shouman
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Nada El-Kholy
- Department of Drug Discovery, H. Lee Moffit Cancer Center& Research Institute, Tampa, FL, 33612, USA
- Cancer Chemical Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Alaa E Hussien
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Azza M El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | - Ahmed M Abou-Shanab
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
| | | | - Ahmad Abdelwaly
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Institute for Computational Molecular Science, Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Mohamed Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12587, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12587, Egypt.
| |
Collapse
|
16
|
He W, Li ZQ, Gu HY, Pan QL, Lin FX. Targeted Therapy of Spinal Cord Injury: Inhibition of Apoptosis Is a Promising Therapeutic Strategy. Mol Neurobiol 2024; 61:4222-4239. [PMID: 38066400 DOI: 10.1007/s12035-023-03814-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/16/2023] [Indexed: 07/11/2024]
Abstract
Spinal cord injury (SCI) is a serious disabling central nervous system injury that can lead to motor, sensory, and autonomic dysfunction below the injury level. SCI can be divided into primary injury and secondary injury according to pathological process. Primary injury is mostly irreversible, while secondary injury is a dynamic regulatory process. Apoptosis is an important pathological event of secondary injury and has a significant effect on the recovery of nerve function after SCI. Nerve cell death can further aggravate the microenvironment of the injured site, leading to neurological dysfunction and thus affect the clinical outcome of patients. Therefore, apoptosis plays a crucial role in the pathological progression of secondary SCI, while inhibiting apoptosis may be a promising therapeutic strategy for SCI. This review will summarize and explore the factors that lead to cell death after SCI, the influence of cross talk between signaling pathways and pathways involved in apoptosis and discuss the influence of apoptosis on SCI, and the therapeutic significance of targeting apoptosis on SCI. This review helps us to understand the role of apoptosis in secondary SCI and provides a theoretical basis for the treatment of SCI based on apoptosis.
Collapse
Affiliation(s)
- Wei He
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Zhi-Qiang Li
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Qi-Lin Pan
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China.
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China.
| |
Collapse
|
17
|
Kural MH, Djakbarova U, Cakir B, Tanaka Y, Chan ET, Arteaga Muniz VI, Madraki Y, Qian H, Park J, Sewanan LR, Park IH, Niklason LE, Kural C. Mechano-inhibition of endocytosis sensitizes cancer cells to Fas-induced Apoptosis. Cell Death Dis 2024; 15:440. [PMID: 38909035 PMCID: PMC11193792 DOI: 10.1038/s41419-024-06822-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/24/2024]
Abstract
The transmembrane death receptor Fas transduces apoptotic signals upon binding its ligand, FasL. Although Fas is highly expressed in cancer cells, insufficient cell surface Fas expression desensitizes cancer cells to Fas-induced apoptosis. Here, we show that the increase in Fas microaggregate formation on the plasma membrane in response to the inhibition of endocytosis sensitizes cancer cells to Fas-induced apoptosis. We used a clinically accessible Rho-kinase inhibitor, fasudil, that reduces endocytosis dynamics by increasing plasma membrane tension. In combination with exogenous soluble FasL (sFasL), fasudil promoted cancer cell apoptosis, but this collaborative effect was substantially weaker in nonmalignant cells. The combination of sFasL and fasudil prevented glioblastoma cell growth in embryonic stem cell-derived brain organoids and induced tumor regression in a xenograft mouse model. Our results demonstrate that sFasL has strong potential for apoptosis-directed cancer therapy when Fas microaggregate formation is augmented by mechano-inhibition of endocytosis.
Collapse
Affiliation(s)
- Mehmet H Kural
- Department of Anesthesiology, School of Medicine, Yale University, New Haven, CT, 06519, USA.
- Humacyte Inc., Durham, NC, 27213, USA.
| | | | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06519, USA
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, QC, H1T 2M4, Canada
| | - Emily T Chan
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | | | - Yasaman Madraki
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Hong Qian
- Department of Anesthesiology, School of Medicine, Yale University, New Haven, CT, 06519, USA
- Humacyte Inc., Durham, NC, 27213, USA
| | - Jinkyu Park
- Yale Cardiovascular Research Center, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, 06519, USA
| | - Lorenzo R Sewanan
- Department of Internal Medicine, Columbia University, New York, NY, 10032, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Laura E Niklason
- Department of Anesthesiology, School of Medicine, Yale University, New Haven, CT, 06519, USA.
- Humacyte Inc., Durham, NC, 27213, USA.
| | - Comert Kural
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA.
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
18
|
Deng Z, Zhu H, Yuan Z, Zhang R, Wang Z, Li H, Yin L, Ruan X, Cheng Z, Li R, Peng H. Enhancing multiple myeloma staging: a novel cell death risk model approach. Clin Exp Med 2024; 24:95. [PMID: 38717497 PMCID: PMC11078818 DOI: 10.1007/s10238-024-01337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/21/2024] [Indexed: 05/12/2024]
Abstract
The prognostication of survival trajectories in multiple myeloma (MM) patients presents a substantial clinical challenge. Leveraging transcriptomic and clinical profiles from an expansive cohort of 2,088 MM patients, sourced from the Gene Expression Omnibus and The Cancer Genome Atlas repositories, we applied a sophisticated nested lasso regression technique to construct a prognostic model predicated on 28 gene pairings intrinsic to cell death pathways, thereby deriving a quantifiable risk stratification metric. Employing a threshold of 0.15, we dichotomized the MM samples into discrete high-risk and low-risk categories. Notably, the delineated high-risk cohort exhibited a statistically significant diminution in survival duration, a finding which consistently replicated across both training and external validation datasets. The prognostic acumen of our cell death signature was further corroborated by TIME ROC analyses, with the model demonstrating robust performance, evidenced by AUC metrics consistently surpassing the 0.6 benchmark across the evaluated arrays. Further analytical rigor was applied through multivariate COX regression analyses, which ratified the cell death risk model as an independent prognostic determinant. In an innovative stratagem, we amalgamated this risk stratification with the established International Staging System (ISS), culminating in the genesis of a novel, refined ISS categorization. This tripartite classification system was subjected to comparative analysis against extant prognostic models, whereupon it manifested superior predictive precision, as reflected by an elevated C-index. In summation, our endeavors have yielded a clinically viable gene pairing model predicated on cellular mortality, which, when synthesized with the ISS, engenders an augmented prognostic tool that exhibits pronounced predictive prowess in the context of multiple myeloma.
Collapse
Affiliation(s)
- Zeyu Deng
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Hongkai Zhu
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Zhaoshun Yuan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Rong Zhang
- National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Kashiwa, Japan
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Xueqin Ruan
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China.
| | - Ruijuan Li
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China.
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
- Institute of Hematology, Central South University, Changsha, Hunan, People's Republic of China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, People's Republic of China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
19
|
Jin Y, Miyama T, Brown A, Hayase T, Song X, Singh AK, Huang L, Flores II, McDaniel LK, Glover I, Halsey TM, Prasad R, Chapa V, Ahmed S, Zhang J, Rai K, Peterson CB, Lizee G, Karmouch J, Hayase E, Molldrem JJ, Chang CC, Tsai WB, Jenq RR. Tsyn-Seq: a T-cell Synapse-Based Antigen Identification Platform. Cancer Immunol Res 2024; 12:530-543. [PMID: 38363296 PMCID: PMC11065584 DOI: 10.1158/2326-6066.cir-23-0467] [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: 06/02/2023] [Revised: 11/02/2023] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
Tools for genome-wide rapid identification of peptide-major histocompatibility complex targets of T-cell receptors (TCR) are not yet universally available. We present a new antigen screening method, the T-synapse (Tsyn) reporter system, which includes antigen-presenting cells (APC) with a Fas-inducible NF-κB reporter and T cells with a nuclear factor of activated T cells (NFAT) reporter. To functionally screen for target antigens from a cDNA library, productively interacting T cell-APC aggregates were detected by dual-reporter activity and enriched by flow sorting followed by antigen identification quantified by deep sequencing (Tsyn-seq). When applied to a previously characterized TCR specific for the E7 antigen derived from human papillomavirus type 16 (HPV16), Tsyn-seq successfully enriched the correct cognate antigen from a cDNA library derived from an HPV16-positive cervical cancer cell line. Tsyn-seq provides a method for rapidly identifying antigens recognized by TCRs of interest from a tumor cDNA library. See related Spotlight by Makani and Joglekar, p. 515.
Collapse
Affiliation(s)
- Yimei Jin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Takahiko Miyama
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Alexandria Brown
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Tomo Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Xingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Anand K. Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Licai Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ivonne I. Flores
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Lauren K. McDaniel
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Israel Glover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Taylor M. Halsey
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Rishika Prasad
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Valerie Chapa
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Saira Ahmed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Christine B. Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Gregory Lizee
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jennifer Karmouch
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Eiko Hayase
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Jeffrey J. Molldrem
- Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Chia-Chi Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Wen-Bin Tsai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Robert R. Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
20
|
Abstract
Regulated cell death mediated by dedicated molecular machines, known as programmed cell death, plays important roles in health and disease. Apoptosis, necroptosis and pyroptosis are three such programmed cell death modalities. The caspase family of cysteine proteases serve as key regulators of programmed cell death. During apoptosis, a cascade of caspase activation mediates signal transduction and cellular destruction, whereas pyroptosis occurs when activated caspases cleave gasdermins, which can then form pores in the plasma membrane. Necroptosis, a form of caspase-independent programmed necrosis mediated by RIPK3 and MLKL, is inhibited by caspase-8-mediated cleavage of RIPK1. Disruption of cellular homeostatic mechanisms that are essential for cell survival, such as normal ionic and redox balance and lysosomal flux, can also induce cell death without invoking programmed cell death mechanisms. Excitotoxicity, ferroptosis and lysosomal cell death are examples of such cell death modes. In this Review, we provide an overview of the major cell death mechanisms, highlighting the latest insights into their complex regulation and execution, and their relevance to human diseases.
Collapse
Affiliation(s)
- Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Shanghai, China.
- Shanghai Key Laboratory of Aging Studies, Shanghai, China.
| | - Dimitry Ofengeim
- Sanofi, Rare and Neurological Diseases Research, Cambridge, MA, USA.
| |
Collapse
|
21
|
Schofield JH, Longo J, Sheldon RD, Albano E, Ellis AE, Hawk MA, Murphy S, Duong L, Rahmy S, Lu X, Jones RG, Schafer ZT. Acod1 expression in cancer cells promotes immune evasion through the generation of inhibitory peptides. Cell Rep 2024; 43:113984. [PMID: 38520689 PMCID: PMC11090053 DOI: 10.1016/j.celrep.2024.113984] [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: 09/27/2023] [Revised: 01/24/2024] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
Targeting programmed cell death protein 1 (PD-1) is an important component of many immune checkpoint blockade (ICB) therapeutic approaches. However, ICB is not an efficacious strategy in a variety of cancer types, in part due to immunosuppressive metabolites in the tumor microenvironment. Here, we find that αPD-1-resistant cancer cells produce abundant itaconate (ITA) due to enhanced levels of aconitate decarboxylase (Acod1). Acod1 has an important role in the resistance to αPD-1, as decreasing Acod1 levels in αPD-1-resistant cancer cells can sensitize tumors to αPD-1 therapy. Mechanistically, cancer cells with high Acod1 inhibit the proliferation of naive CD8+ T cells through the secretion of inhibitory factors. Surprisingly, inhibition of CD8+ T cell proliferation is not dependent on the secretion of ITA but is instead a consequence of the release of small inhibitory peptides. Our study suggests that strategies to counter the activity of Acod1 in cancer cells may sensitize tumors to ICB therapy.
Collapse
Affiliation(s)
- James H Schofield
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Joseph Longo
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ryan D Sheldon
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Emma Albano
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Abigail E Ellis
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Mark A Hawk
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sean Murphy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Loan Duong
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sharif Rahmy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Xin Lu
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Zachary T Schafer
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
22
|
Wan W, Qian X, Zhou B, Gao J, Deng J, Zhao D. Integrative analysis and validation of necroptosis-related molecular signature for evaluating diagnosis and immune features in Rheumatoid arthritis. Int Immunopharmacol 2024; 131:111809. [PMID: 38484666 DOI: 10.1016/j.intimp.2024.111809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease that is characterized by persistent morning stiffness, joint pain, and swelling. However, there is a lack of reliable diagnostic markers and therapeutic targets that are both effective and trustworthy. METHODS In this study, gene expression profiles (GSE89408, GSE55235, GSE55457, and GSE77298) were obtained from the Gene Expression Omnibus database. Differentially expressed necroptosis-related genes were attained from intersection of necroptosis-related gene set, differentially expressed genes, and weighted gene co-expression network analysis. The LASSO, random forest, and SVM-RFE machine learning algorithms were utilized to further screen potential diagnostic genes for RA. Immune cell infiltration was analyzed using the CIBERSORT method. The expressions of diagnostic genes were validated through quantitative real-time PCR, western blotting, immunohistochemistry, and immunofluorescence staining in synovial tissues collected from three trauma controls and three RA patients. RESULTS Five core necroptosis-related genes (FAS, CYBB, TNFSF10, EIF2AK2, and BIRC2) were identified as potential biomarkers for RA. Two different necroptosis patterns based on these five genes were confirmed to significantly correlated with immune cells (especially macrophages). In vitro experiments showed significantly higher mRNA and protein expression levels of CYBB and EIF2AK2 in RA patients compared to normal controls, consistent with the bioinformatics analysis results. CONCLUSION Our study identified a novel necroptosis-related subtype and five diagnostic biomarkers of RA, revealed vital roles in the development and occurrence of RA, and offered potential targets for clinical diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Wei Wan
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Xinyu Qian
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Bole Zhou
- Department of Joint Bone Disease Surgery, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jie Gao
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jiewen Deng
- Department of Cardiovascular Diseases, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Dongbao Zhao
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
23
|
Li B, Zhang X, Zhang Q, Zheng T, Li Q, Yang S, Shao J, Guan W, Zhang S. Nutritional strategies to reduce intestinal cell apoptosis by alleviating oxidative stress. Nutr Rev 2024:nuae023. [PMID: 38626282 DOI: 10.1093/nutrit/nuae023] [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] [Indexed: 04/18/2024] Open
Abstract
The gut barrier is the first line of defense against harmful substances and pathogens in the intestinal tract. The balance of proliferation and apoptosis of intestinal epithelial cells (IECs) is crucial for maintaining the integrity of the intestinal mucosa and its function. However, oxidative stress and inflammation can cause DNA damage and abnormal apoptosis of the IECs, leading to the disruption of the intestinal epithelial barrier. This, in turn, can directly or indirectly cause various acute and chronic intestinal diseases. In recent years, there has been a growing understanding of the vital role of dietary ingredients in gut health. Studies have shown that certain amino acids, fibers, vitamins, and polyphenols in the diet can protect IECs from excessive apoptosis caused by oxidative stress, and limit intestinal inflammation. This review aims to describe the molecular mechanism of apoptosis and its relationship with intestinal function, and to discuss the modulation of IECs' physiological function, the intestinal epithelial barrier, and gut health by various nutrients. The findings of this review may provide a theoretical basis for the use of nutritional interventions in clinical intestinal disease research and animal production, ultimately leading to improved human and animal intestinal health.
Collapse
Affiliation(s)
- Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaoli Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiayuan Shao
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
24
|
Thome CD, Tausche P, Hohenberger K, Yang Z, Krammer S, Trufa DI, Sirbu H, Schmidt J, Finotto S. Short-chain fatty acids induced lung tumor cell death and increased peripheral blood CD4+ T cells in NSCLC and control patients ex vivo. Front Immunol 2024; 15:1328263. [PMID: 38650948 PMCID: PMC11033355 DOI: 10.3389/fimmu.2024.1328263] [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: 10/27/2023] [Accepted: 03/14/2024] [Indexed: 04/25/2024] Open
Abstract
Background Despite therapy advances, one of the leading causes of cancer deaths still remains lung cancer. To improve current treatments or prevent non-small cell lung cancer (NSCLC), the role of the nutrition in cancer onset and progression needs to be understood in more detail. While in colorectal cancer, the influence of local microbiota derived SCFAs have been well investigated, the influence of SCFA on lung cancer cells via peripheral blood immune system should be investigated more deeply. In this respect, nutrients absorbed via the gut might affect the tumor microenvironment (TME) and thus play an important role in tumor cell growth. Objective This study focuses on the impact of the short-chain fatty acid (SCFA) Sodium Butyrate (SB), on lung cancer cell survival. We previously described a pro-tumoral role of glucose on A549 lung adenocarcinoma cell line. In this study, we wanted to know if SB would counteract the effect of glucose and thus cultured A549 and H520 in vitro with and without SB in the presence or absence of glucose and investigated how the treatment with SB affects the survival of lung cancer cells and its influence on immune cells fighting against lung cancer. Methods In this study, we performed cell culture experiments with A549, H520 and NSCLC-patient-derived epithelial cells under different SB levels. To investigate the influence on the immune system, we performed in vitro culture of peripheral mononuclear blood cells (PBMC) from control, smoker and lung cancer patients with increasing SB concentrations. Results To investigate the effect of SB on lung tumor cells, we first analyzed the effect of 6 different concentrations of SB on A549 cells at 48 and 72 hours cell culture. Here we found that, SB treatment reduced lung cancer cell survival in a concentration dependent manner. We next focused our deeper analysis on the two concentrations, which caused the maximal reduction in cell survival. Here, we observed that SB led to cell cycle arrest and induced early apoptosis in A549 lung cancer cells. The expression of cell cycle regulatory proteins and A549 lung cancer stem cell markers (CD90) was induced. Additionally, this study explored the role of interferon-gamma (IFN-γ) and its receptor (IFN-γ-R1) in combination with SB treatment, revealing that, although IFN-γ-R1 expression was increased, IFN-γ did not affect the efficacy of SB in reducing tumor cell viability. Furthermore, we examined the effects of SB on immune cells, specifically CD8+ T cells and natural killer (NK) cells from healthy individuals, smokers, and NSCLC patients. SB treatment resulted in a decreased production of IFN-γ and granzyme B in CD8+ T cells and NK cells. Moreover, SB induced IFN-γ-R1 in NK cells and CD4+ T cells in the absence of glucose both in PBMCs from controls and NSCLC subjects. Conclusion Overall, this study highlights the potential of SB in inhibiting lung cancer cell growth, triggering apoptosis, inducing cell cycle arrest, and modulating immune responses by activating peripheral blood CD4+ T cells while selectively inducing IFN-γ-R1 in NK cells in peripheral blood and inhibiting peripheral blood CD8+ T cells and NK cells. Thus, understanding the mechanisms of action of SB in the TME and its influence on the immune system provide valuable insights of potentially considering SB as a candidate for adjunctive therapies in NSCLC.
Collapse
Affiliation(s)
- Carolin D. Thome
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Patrick Tausche
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Katja Hohenberger
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Zuqin Yang
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Susanne Krammer
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
| | - Denis I. Trufa
- Department of Thoracic Surgery, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Joachim Schmidt
- Department of Anesthesiology, University Medical School Hospital Erlangen (UKER), Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, University Medical School Hospital Erlangen (UKER) Friedrich-Alexander-University (FAU), Erlangen-Nürnberg, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|
25
|
Awasthi D, Sarode A. Neutrophils at the Crossroads: Unraveling the Multifaceted Role in the Tumor Microenvironment. Int J Mol Sci 2024; 25:2929. [PMID: 38474175 DOI: 10.3390/ijms25052929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Over the past decade, research has prominently established neutrophils as key contributors to the intricate landscape of tumor immune biology. As polymorphonuclear granulocytes within the innate immune system, neutrophils play a pivotal and abundant role, constituting approximately ∼70% of all peripheral leukocytes in humans and ∼10-20% in mice. This substantial presence positions them as the frontline defense against potential threats. Equipped with a diverse array of mechanisms, including reactive oxygen species (ROS) generation, degranulation, phagocytosis, and the formation of neutrophil extracellular traps (NETs), neutrophils undeniably serve as indispensable components of the innate immune system. While these innate functions enable neutrophils to interact with adaptive immune cells such as T, B, and NK cells, influencing their functions, they also engage in dynamic interactions with rapidly dividing tumor cells. Consequently, neutrophils are emerging as crucial regulators in both pro- and anti-tumor immunity. This comprehensive review delves into recent research to illuminate the multifaceted roles of neutrophils. It explores their diverse functions within the tumor microenvironment, shedding light on their heterogeneity and their impact on tumor recruitment, progression, and modulation. Additionally, the review underscores their potential anti-tumoral capabilities. Finally, it provides valuable insights into clinical therapies targeting neutrophils, presenting a promising approach to leveraging innate immunity for enhanced cancer treatment.
Collapse
Affiliation(s)
- Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Aditya Sarode
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
26
|
Soto M, Filbert EL, Yang H, Starzinski S, Starzinski A, Gin M, Chen B, Le P, Li T, Bol B, Cheung A, Zhang L, Hsu FJ, Ko A, Fong L, Keenan BP. Neoadjuvant CD40 Agonism Remodels the Tumor Immune Microenvironment in Locally Advanced Esophageal/Gastroesophageal Junction Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:200-212. [PMID: 38181044 PMCID: PMC10809910 DOI: 10.1158/2767-9764.crc-23-0550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
Sotigalimab is an agonistic anti-CD40 mAb that can modulate antitumor immune responses. In a phase II clinical trial of sotigalimab combined with neoadjuvant chemoradiation (CRT) in locally advanced esophageal/gastroesophageal junction (E/GEJ) cancer with the primary outcome of efficacy as measured by pathologic complete response (pCR) rate, the combination induced pCR in 38% of treated patients. We investigated the mechanism of action of sotigalimab in samples obtained from this clinical trial. Tumor biopsies and peripheral blood samples were collected at baseline, following an initial dose of sotigalimab, and at the time of surgery after CRT completion from six patients. High dimensional single-cell techniques were used, including combined single-cell RNA-sequencing and proteomics (CITEseq) and multiplexed ion beam imaging, to analyze immune responses. Sotigalimab dramatically remodeled the immune compartment in the periphery and within the tumor microenvironment (TME), increasing expression of molecules related to antigen processing and presentation and altering metabolic pathways in myeloid cells. Concomitant with these changes in myeloid cells, sotigalimab treatment primed new T cell clonotypes and increased the density and activation of T cells with enhanced cytotoxic function. Sotigalimab treatment also induced a decrease in the frequency of Tregs in the TME. These findings indicate that a single dose of sotigalimab leads to enhanced antigen presentation that can activate T cells and induce new T cell clones. This restructuring of the TME provides elements which are critical to the development of effective antitumor immune responses and improved clinical outcomes.
Collapse
Affiliation(s)
- Maira Soto
- Pyxis Oncology, Inc., Boston, Massachusetts
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Erin L. Filbert
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Hai Yang
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Stephanie Starzinski
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alec Starzinski
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Marissa Gin
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Brandon Chen
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Phi Le
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Tony Li
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Brandon Bol
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alexander Cheung
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Li Zhang
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Frank J. Hsu
- Pyxis Oncology, Inc., Boston, Massachusetts
- Apexigen America, Inc, San Carlos, California (now a fully owned subsidiary of Pyxis Oncology, Inc.)
| | - Andrew Ko
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Lawrence Fong
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| | - Bridget P. Keenan
- Cancer Immunotherapy Program, University of California, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
- Division of Hematology/Oncology, University of California, San Francisco, California
| |
Collapse
|
27
|
Wang S, Cheng L. The role of apoptosis in spinal cord injury: a bibliometric analysis from 1994 to 2023. Front Cell Neurosci 2024; 17:1334092. [PMID: 38293650 PMCID: PMC10825042 DOI: 10.3389/fncel.2023.1334092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/29/2023] [Indexed: 02/01/2024] Open
Abstract
Background Apoptosis after spinal cord injury (SCI) plays a pivotal role in the secondary injury mechanisms, which cause the ultimate neurologic insults. A better understanding of the molecular and cellular basis of apoptosis in SCI allows for improved glial and neuronal survival via the administrations of anti-apoptotic biomarkers. The knowledge structure, development trends, and research hotspots of apoptosis and SCI have not yet been systematically investigated. Methods Articles and reviews on apoptosis and SCI, published from 1st January 1994 to 1st Oct 2023, were retrieved from the Web of Science™. Bibliometrix in R was used to evaluate annual publications, countries, affiliations, authors, sources, documents, key words, and hot topics. Results A total of 3,359 publications in accordance with the criterions were obtained, which exhibited an ascending trend in annual publications. The most productive countries were the USA and China. Journal of Neurotrauma was the most impactive journal; Wenzhou Medical University was the most prolific affiliation; Cuzzocrea S was the most productive and influential author. "Apoptosis," "spinal-cord-injury," "expression," "activation," and "functional recovery" were the most frequent key words. Additionally, "transplantation," "mesenchymal stemness-cells," "therapies," "activation," "regeneration," "repair," "autophagy," "exosomes," "nlrp3 inflammasome," "neuroinflammation," and "knockdown" were the latest emerging key words, which may inform the hottest themes. Conclusions Apoptosis after SCI may cause the ultimate neurological damages. Development of novel treatments for secondary SCI mainly depends on a better understanding of apoptosis-related mechanisms in molecular and cellular levels. Such therapeutic interventions involve the application of anti-apoptotic agents, free radical scavengers, as well as anti-inflammatory drugs, which can be targeted to inhibit core events in cellular and molecular injury cascades pathway.
Collapse
Affiliation(s)
- Siqiao Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai, China
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai, China
- Institute of Spinal and Spinal Cord Injury, Tongji University School of Medicine, Shanghai, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Li S, Tao G. Perish in the Attempt: Regulated Cell Death in Regenerative and Nonregenerative Tissue. Antioxid Redox Signal 2023; 39:1053-1069. [PMID: 37218435 PMCID: PMC10715443 DOI: 10.1089/ars.2022.0166] [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: 10/05/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Significance: A cell plays its roles throughout its life span, even during its demise. Regulated cell death (RCD) is one of the key topics in modern biomedical studies. It is considered the main approach for removing stressed and/or damaged cells. Research during the past two decades revealed more roles of RCD, such as coordinating tissue development and driving compensatory proliferation during tissue repair. Recent Advances: Compensatory proliferation, initially identified in primitive organisms during the regeneration of lost tissue, is an evolutionarily conserved process that also functions in mammals. Among various types of RCD, apoptosis is considered the top candidate to induce compensatory proliferation in damaged tissue. Critical Issues: The roles of apoptosis in the recovery of nonregenerative tissue are still vague. The roles of other types of RCD, such as necroptosis and ferroptosis, have not been well characterized in the context of tissue regeneration. Future Directions: In this review article, we attempt to summarize the recent insights on the role of RCD in tissue repair. We focus on apoptosis, with expansion to ferroptosis and necroptosis, in primitive organisms with significant regenerative capacity as well as common mammalian research models. After gathering hints from regenerative tissue, in the second half of the review, we take a notoriously nonregenerative tissue, the myocardium, as an example to discuss the role of RCD in terminally differentiated quiescent cells. Antioxid. Redox Signal. 39, 1053-1069.
Collapse
Affiliation(s)
- Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
29
|
Kuppusamy KM, Selvaraj S, Singaravelu P, John CM, Racheal K, Varghese K, Kaliyamoorthy D, Perumal E, Gunasekaran K. Anti-microbial and anti-cancer efficacy of acetone extract of Rosa chinensis against resistant strain and lung cancer cell line. BMC Complement Med Ther 2023; 23:406. [PMID: 37950173 PMCID: PMC10636979 DOI: 10.1186/s12906-023-04222-2] [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: 05/11/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Screening of herbal plants for various therapeutic properties is the hour as it shows promising activity. Scientific evidence of the pharmacological activity of the plant strengthens the traditional application of plants. METHODS Rose flowers (Rosa chinensis) were procured and grounded into a coarse powder. The DNA was isolated from rose flower and molecular identification was performed by rbcL-BF and rbcL-724R primers. Antibacterial activity was evaluated by using disc and agar diffusion methods and the anti-cancer effect of the rose flower extract (RE) was examined using MTT assay in lung cancer cell line. The mechanism of cell death induced by RE was qualitatively measured using Acridine orange/Ethidium bromide staining and Hoechst staining. GC-MS analysis was performed using GC-MS-5975C. RESULT The RE showed potent antimicrobial activity against various ATCC cultures. The rose extract strongly inhibits the growth of ESBL resistant organism along with inhibition of biofilm formation in the ESBL resistant organism. The extract caused apoptotic and necrotic cell death in lung cancer cells. GC-MS analysis demonstrated the presence of several biologically active compounds such as Clindamycin, Phytol, Octanoic acid, and Stigmasterol which might be the reason for the therapeutic properties of the plant. CONCLUSION This study shows the antimicrobial and biofilm inhibition activity against the clinical isolates of Klebsiella pneumonia. The study shows the cytotoxic and apoptotic activity in A549 cancer cell line. Thus, the plant may act as a potent antimicrobial drug against resistant strains.
Collapse
Affiliation(s)
- Kalaivani Madhavaram Kuppusamy
- Research Centre for Cellular Genomics and Cancer Research, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Sivakumar Selvaraj
- Molecular Biology section, Consultant Molecular Biologist, Medall Healthcare Private Limited, Chennai, India
| | - Pujithaa Singaravelu
- Research Centre for Cellular Genomics and Cancer Research, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Cordelia Mano John
- Department of Biomedical Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Kalaiselvan Racheal
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Keziaann Varghese
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Dinesh Kaliyamoorthy
- Department of Microbiology, Sree Balaji Medical College and Hospital, Chennai, 600044, India
| | - Elumalai Perumal
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| | - Krishnamoorthy Gunasekaran
- Department of Medical Biochemistry, College of Health Sciences, Dambi Dollo University, KelamWelega Zone, Dembidolo, P.O. Box: 360, Oromia Region, Ethiopia.
| |
Collapse
|
30
|
Sun Y, Yao H, Yu X, Chen Y, Chai C. Adult's Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis: A case report. Clin Case Rep 2023; 11:e8117. [PMID: 37927984 PMCID: PMC10622403 DOI: 10.1002/ccr3.8117] [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: 09/02/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023] Open
Abstract
Adult's Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis is a rare and life-threatening condition characterized by atypical initial symptoms and rapid disease progression. To facilitate early diagnosis and prompt treatment, it is imperative to implement early multidisciplinary intervention and prioritize pathogen detection, as these measures significantly contribute to enhancing patient prognosis.
Collapse
Affiliation(s)
- Yi Sun
- Department of General SurgeryThe People's Hospital of SNDSuzhouChina
| | - Han Yao
- Department of General SurgeryThe People's Hospital of SNDSuzhouChina
| | - Xiaofeng Yu
- Department of General SurgeryThe People's Hospital of SNDSuzhouChina
| | - Yunzhao Chen
- Department of PathologyThe People's Hospital of SNDSuzhouChina
| | - Chen Chai
- Department of General SurgeryThe People's Hospital of SNDSuzhouChina
| |
Collapse
|
31
|
Alsalloum A, Alrhmoun S, Shevchenko J, Fisher M, Philippova J, Perik-Zavodskii R, Perik-Zavodskaia O, Lopatnikova J, Kurilin V, Volynets M, Akahori Y, Shiku H, Silkov A, Sennikov S. TCR-Engineered Lymphocytes Targeting NY-ESO-1: In Vitro Assessment of Cytotoxicity against Tumors. Biomedicines 2023; 11:2805. [PMID: 37893178 PMCID: PMC10604587 DOI: 10.3390/biomedicines11102805] [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: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Adoptive T-cell therapies tailored for the treatment of solid tumors encounter intricate challenges, necessitating the meticulous selection of specific target antigens and the engineering of highly specific T-cell receptors (TCRs). This study delves into the cytotoxicity and functional characteristics of in vitro-cultured T-lymphocytes, equipped with a TCR designed to precisely target the cancer-testis antigen NY-ESO-1. Flow cytometry analysis unveiled a notable increase in the population of cells expressing activation markers upon encountering the NY-ESO-1-positive tumor cell line, SK-Mel-37. Employing the NanoString platform, immune transcriptome profiling revealed the upregulation of genes enriched in Gene Ontology Biological Processes associated with the IFN-γ signaling pathway, regulation of T-cell activation, and proliferation. Furthermore, the modified T cells exhibited robust cytotoxicity in an antigen-dependent manner, as confirmed by the LDH assay results. Multiplex immunoassays, including LEGENDplex™, additionally demonstrated the elevated production of cytotoxicity-associated cytokines driven by granzymes and soluble Fas ligand (sFasL). Our findings underscore the specific targeting potential of engineered TCR T cells against NY-ESO-1-positive tumors. Further comprehensive in vivo investigations are essential to thoroughly validate these results and effectively harness the intrinsic potential of genetically engineered T cells for combating cancer.
Collapse
Affiliation(s)
- Alaa Alsalloum
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Saleh Alrhmoun
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Julia Shevchenko
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Marina Fisher
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Julia Philippova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Roman Perik-Zavodskii
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Olga Perik-Zavodskaia
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Julia Lopatnikova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Vasily Kurilin
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Marina Volynets
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Yasushi Akahori
- Department of Personalized Cancer Immunotherapy, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan;
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Graduate School of Medicine, Mie University, Tsu 514-8507, Japan;
| | - Alexander Silkov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
| | - Sergey Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution Research Institute of Fundamental and Clinical Immunology, Novosibirsk 630099, Russia; (A.A.); (S.A.); (J.S.); (M.F.); (J.P.); (R.P.-Z.); (O.P.-Z.); (J.L.); (V.K.); (M.V.); (A.S.)
- Department of Immunology, V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Novosibirsk 630090, Russia
| |
Collapse
|
32
|
Sikora JP, Karawani J, Sobczak J. Neutrophils and the Systemic Inflammatory Response Syndrome (SIRS). Int J Mol Sci 2023; 24:13469. [PMID: 37686271 PMCID: PMC10488036 DOI: 10.3390/ijms241713469] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
We are not entirely able to understand, assess, and modulate the functioning of the immune system in clinical situations that lead to a systemic inflammatory response. In the search for diagnostic and treatment strategies (which are still far from perfect), it became very important to study the pathogenesis and participation of endogenous inflammation mediators. This study attempts to more precisely establish the role of neutrophils in individual phenomena occurring during an inflammatory and anti-inflammatory reaction, taking into account their cidal, immunoregulatory, and reparative abilities. Pro- and anticoagulatory properties of endothelium in systemic inflammatory response syndrome (SIRS) are emphasised, along with the resulting clinical implications (the application of immunotherapy using mesenchymal stem/stromal cells (MSCs) or IL-6 antagonists in sepsis and COVID-19 treatment, among others). Special attention is paid to reactive oxygen species (ROS), produced by neutrophils activated during "respiratory burst" in the course of SIRS; the protective and pathogenic role of these endogenous mediators is highlighted. Moreover, clinically useful biomarkers of SIRS (neutrophil extracellular traps, cell-free DNA, DAMP, TREMs, NGAL, miRNA, selected cytokines, ROS, and recognised markers of endothelial damage from the group of adhesins by means of immunohistochemical techniques) related to the neutrophils are presented, and their role in the diagnosing and forecasting of sepsis, burn disease, and COVID-19 is emphasised. Finally, examples of immunomodulation of sepsis and antioxidative thermal injury therapy are presented.
Collapse
Affiliation(s)
- Janusz P. Sikora
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
| | - Jakub Karawani
- Faculty of Medicine, Lazarski University, ul. Świeradowska 43, 02-662 Warsaw, Poland;
| | - Jarosław Sobczak
- Department of Paediatric Emergency Medicine, 2nd Chair of Paediatrics, Central Clinical Hospital, Medical University of Łódź, ul. Sporna 36/50, 91-738 Łódź, Poland;
- Department of Management and Logistics in Healthcare, Medical University of Łódź, ul. Lindleya 6, 90-131 Łódź, Poland
| |
Collapse
|
33
|
Grisanti LA. TRAIL and its receptors in cardiac diseases. Front Physiol 2023; 14:1256852. [PMID: 37621762 PMCID: PMC10445540 DOI: 10.3389/fphys.2023.1256852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Loss of cardiomyocytes that occurs during many types of damage to the heart such as ischemic injury and stress caused by pressure overload, diminishes cardiac function due to their limited regenerative capacity and promotes remodeling, which further damages the heart. Cardiomyocyte death occurs through two primary mechanisms, necrosis and apoptosis. Apoptosis is a highly regulated form of cell death that can occur through intrinsic (mitochondrial) or extrinsic (receptor mediated) pathways. Extrinsic apoptosis occurs through a subset of Tumor Necrosis Receptor (TNF) family receptors termed "Death Receptors." While some ligands for death receptors have been extensively studied in the heart, such as TNF-α, others have been virtually unstudied. One poorly characterized cardiac TNF related ligand is TNF-Related Apoptosis Inducing Ligand (TRAIL). TRAIL binds to two apoptosis-inducing receptors, Death Receptor (DR) 4 and DR5. There are also three decoy TRAIL receptors, Decoy Receptor (DcR) 1, DcR2 and osteoprotegerin (OPG). While TRAIL has been extensively studied in the cancer field due to its ability to selectively induce apoptosis in transformed cell types, emerging clinical evidence points towards a role for TRAIL and its receptors in cardiac pathology. This article will highlight our current understanding of TRAIL and its receptors in normal and pathological conditions in the heart.
Collapse
Affiliation(s)
- Laurel A. Grisanti
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
34
|
Gu C, Fan X, Yu W. Functional Diversity of Mammalian Small Heat Shock Proteins: A Review. Cells 2023; 12:1947. [PMID: 37566026 PMCID: PMC10417760 DOI: 10.3390/cells12151947] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
The small heat shock proteins (sHSPs), whose molecular weight ranges from 12∼43 kDa, are members of the heat shock protein (HSP) family that are widely found in all organisms. As intracellular stress resistance molecules, sHSPs play an important role in maintaining the homeostasis of the intracellular environment under various stressful conditions. A total of 10 sHSPs have been identified in mammals, sharing conserved α-crystal domains combined with variable N-terminal and C-terminal regions. Unlike large-molecular-weight HSP, sHSPs prevent substrate protein aggregation through an ATP-independent mechanism. In addition to chaperone activity, sHSPs were also shown to suppress apoptosis, ferroptosis, and senescence, promote autophagy, regulate cytoskeletal dynamics, maintain membrane stability, control the direction of cellular differentiation, modulate angiogenesis, and spermatogenesis, as well as attenuate the inflammatory response and reduce oxidative damage. Phosphorylation is the most significant post-translational modification of sHSPs and is usually an indicator of their activation. Furthermore, abnormalities in sHSPs often lead to aggregation of substrate proteins and dysfunction of client proteins, resulting in disease. This paper reviews the various biological functions of sHSPs in mammals, emphasizing the roles of different sHSPs in specific cellular activities. In addition, we discuss the effect of phosphorylation on the function of sHSPs and the association between sHSPs and disease.
Collapse
Affiliation(s)
- Chaoguang Gu
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Xiasha High-Tech Zone No.2 Road, Hangzhou 310018, China;
| | - Xinyi Fan
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S1A1, Canada;
| | - Wei Yu
- Institute of Biochemistry, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Xiasha High-Tech Zone No.2 Road, Hangzhou 310018, China;
| |
Collapse
|
35
|
Ren M, Yao B, Han B, Li C. Nuclear Imaging of CAR T Immunotherapy to Solid Tumors: In Terms of Biodistribution, Viability, and Cytotoxic Effect. Adv Biol (Weinh) 2023; 7:e2200293. [PMID: 36642820 DOI: 10.1002/adbi.202200293] [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: 10/29/2022] [Revised: 12/25/2022] [Indexed: 01/17/2023]
Abstract
Immunotherapy has become a mainstay of cancer therapy. Since chimeric antigen receptor (CAR) T immunotherapy achieves unprecedented success in curing hematological malignancies, the possibility of it revolutionizing the paradigm of solid tumors has aroused increasing attention. However, the restricted accessibility to tumor parenchyma, the immunosuppressive tumor microenvironment, and antigen heterogeneity of solid tumors make it difficult to replicate its success. Therefore, dynamic evaluation of CAR T cells' tumor accessibility, intratumoral viability, and anti-tumor cytotoxicity is necessary to facilitate its translation to solid tumors. Besides, real-timely imaging above events in vivo can help evaluate therapeutic responses and optimize CAR T immunotherapy for solid tumors. Nuclear imaging, including positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging, is frequently applied for evaluating adoptive cell therapies owing to its excellent sensitivity, high tissue penetration, and great translation potential. In addition, quantitative analysis can be performed in dynamic and noninvasive patterns. This review focuses on recent advances in PET/SPECT technologies and imaging probes in monitoring CAR T cells' migration, viability, and cytotoxicity to solid tumors post-administration. Prospects of what should be done in the next stage to promote CAR T therapy's application in solid tumors are also discussed.
Collapse
Affiliation(s)
- Mingliang Ren
- Minhang Hospital and Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, School of Pharmacy, Fudan University, Zhangheng Road 826, 201203, Shanghai, China
| | - Bolin Yao
- Minhang Hospital and Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, School of Pharmacy, Fudan University, Zhangheng Road 826, 201203, Shanghai, China
| | - Bing Han
- Minhang Hospital, Fudan University, Shanghai, China
| | - Cong Li
- Minhang Hospital and Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, School of Pharmacy, Fudan University, Zhangheng Road 826, 201203, Shanghai, China
| |
Collapse
|
36
|
Wang Y, Wu W, Gong J. Live or death in cells: from micronutrition metabolism to cell fate. Front Cell Dev Biol 2023; 11:1185989. [PMID: 37250891 PMCID: PMC10213646 DOI: 10.3389/fcell.2023.1185989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Micronutrients and cell death have a strong relationship and both are essential for human to maintain good body health. Dysregulation of any micronutrients causes metabolic or chronic diseases, including obesity, cardiometabolic condition, neurodegeneration, and cancer. The nematode Caenorhabditis elegans is an ideal genetic organism for researching the mechanisms of micronutrients in metabolism, healthspan, and lifespan. For example, C. elegans is a haem auxotroph, and the research of this special haem trafficking pathway contributes important reference to mammal study. Also, C. elegans characteristics including anatomy simply, clear cell lineage, well-defined genetics, and easily differentiated cell forms make it a powerful tool for studying the mechanisms of cell death including apoptosis, necrosis, autophagy, and ferroptosis. Here, we describe the understanding of micronutrient metabolism currently and also sort out the fundamental mechanisms of different kinds of cell death. A thorough understanding of these physiological processes not only builds a foundation for developing better treatments for various micronutrient disorders but also provides key insights into human health and aging.
Collapse
Affiliation(s)
- Yuting Wang
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianke Gong
- Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
37
|
Chen Y, Liu X, Yan D, Xu J, Luan S, Xiao C, Huang Q. Exposure to emamectin benzoate confers cytotoxic effects on human molt-4 T-cells and possible ameliorative role of vitamin E and dithiothreitol. Drug Chem Toxicol 2023; 46:413-422. [PMID: 35266429 DOI: 10.1080/01480545.2022.2044350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Emamectin benzoate (EMB) is an avermectin insecticide that is extensively used for pest control, but there are few reports concerning its cytotoxic effects on human lymphocytes. In the current study, the hematotoxicity of EMB was evaluated in Molt-4 T-cells, a human T-lymphoblastic cell line with high motility, and the role of vitamin E (VitE) and dithiothreitol (DTT) in attenuating EMB cytotoxicity was characterized. Exposure of Molt-4 cells to EMB decreased cell viability and proliferation, induced a loss of cell clusters, and significantly increased membrane collapse and chromatin condensation. Moreover, EMB significantly increased cell death and suppressed transglutaminase activity. EMB treatment modulated the NF-κB signaling pathway, decreased the expression of p105, p50, and p65/RelA in cytosolic and nuclear fractions, and increased nuclear IκBα expression. EMB increased oxidative stress, as demonstrated by a significant increase in the levels of reactive oxygen species (ROS). Treatment with non-cytotoxic concentrations of VitE or DTT ameliorated the hematotoxicity induced by pretreatment with EMB, increased Molt-4 cell viability, raised the IC50 values of EMB, limited intracellular ROS generation, and mitigated EMB-mediated effects on NF-κB signaling. The results indicate the potential cytotoxicity of EMB on human lymphocytes, and demonstrate that VitE and DTT treatment can reduce the cytotoxic effects of EMB.
Collapse
Affiliation(s)
- Yongjun Chen
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xuefeng Liu
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Dongmei Yan
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jialin Xu
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shaorong Luan
- School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Ciying Xiao
- School of Biochemical Engineering, East China University of Science and Technology, Shanghai, China
| | - Qingchun Huang
- Shanghai Key Lab of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
38
|
Haluck-Kangas A, Fink M, Bartom ET, Peter ME. CD95/Fas ligand mRNA is toxic to cells through more than one mechanism. MOLECULAR BIOMEDICINE 2023; 4:11. [PMID: 37059938 PMCID: PMC10105004 DOI: 10.1186/s43556-023-00119-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/03/2023] [Indexed: 04/16/2023] Open
Abstract
CD95/Fas ligand (CD95L) induces apoptosis through protein binding to the CD95 receptor. However, CD95L mRNA also induces toxicity in the absence of CD95 through induction of DISE (Death Induced by Survival Gene Elimination), a form of cell death mediated by RNA interference (RNAi). We now report that CD95L mRNA processing generates a short (s)RNA nearly identical to shL3, a commercial CD95L-targeting shRNA that led to the discovery of DISE. Neither of the miRNA biogenesis proteins Drosha nor Dicer are required for this processing. Interestingly, CD95L toxicity depends on the core component of the RISC, Ago2, in some cell lines, but not in others. In the HCT116 colon cancer cell line, Ago 1-4 appear to function redundantly in RNAi. In fact, Ago 1/2/3 knockout cells retain sensitivity to CD95L mRNA toxicity. Toxicity was only blocked by mutation of all in-frame start codons in the CD95L ORF. Dying cells exhibited an enrichment of RISC bound (R)-sRNAs with toxic 6mer seed sequences, while expression of the non-toxic CD95L mutant enriched for loading of R-sRNAs with nontoxic 6mer seeds. However, CD95L is not the only source of these R-sRNAs. We find that CD95L mRNA may induce DISE directly and indirectly, and that alternate mechanisms may underlie CD95L mRNA processing and toxicity.
Collapse
Affiliation(s)
- Ashley Haluck-Kangas
- Department of Medicine/Division Hematology/Oncology, Feinberg School of Medicine, Chicago, IL, USA
| | - Madelaine Fink
- Department of Medicine/Division Hematology/Oncology, Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth T Bartom
- Department of Biochemistry and Molecular Genetics, Chicago, IL, USA
- Department of Preventive Medicine/Division of Biostatistics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marcus E Peter
- Department of Medicine/Division Hematology/Oncology, Feinberg School of Medicine, Chicago, IL, USA.
- Department of Biochemistry and Molecular Genetics, Chicago, IL, USA.
| |
Collapse
|
39
|
Panwar D, Rawal L, Ali S. The potential role of the KFG and KITLG proteins in preventing granulosa cell apoptosis in Bubalus bubalis. J Genet Eng Biotechnol 2023; 21:39. [PMID: 37000378 PMCID: PMC10066048 DOI: 10.1186/s43141-023-00480-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 02/09/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND The dynamics of mammalian follicular development and atresia is an intricate process involving the cell-cell communication mediated by secreted ovarian factors. These interactions are critical for oocyte development and regulation of follicular atresia which in part are mediated by keratinocyte growth factor (KGF) and kit ligand (KITLG), but their roles in the regulation of apoptosis in buffalo granulosa cells have not yet been defined. During mammalian follicular development, granulosa cell apoptosis triggers the atresia so ~ 1% follicles reach the ovulation stage. In the present study, we used buffalo granulosa cells to examine the effects of KGF and KITLG in apoptosis regulation and investigated potential mechanism on Fas-FasL and Bcl-2 signaling pathways. RESULT Isolated buffalo granulosa cells were cultured with KGF and KITLG proteins using different doses (0, 10, 20, and 50 ng/ml) independently or in combination. Expression analysis for both anti-apoptotic (Bcl-2, Bcl-xL, and cFLIP) and pro-apoptotic (Bax, Fas, and FasL) genes at transcriptional levels were carried out by real-time PCR. Upon treatments, expression levels of anti-apoptotic genes were significantly upregulated in a dose-dependent manner, showing an upregulation at 50 ng/ml (independently), and at 10 ng/ml in combination. Additionally, upregulation of growth-promoting factors, bFGF, and α-Inhibin was also observed. CONCLUSIONS Our findings suggest the potential roles of KGF and KITLG in determining granulosa cell growth and regulating apoptosis.
Collapse
Affiliation(s)
- Deepak Panwar
- Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
- Present Address: National Reference Laboratory, Dr. Lal Pathlabs, B7 Road, Block E, Rohini Sector 18, New Delhi, 110085, India
| | - Leena Rawal
- Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
- Present Address: National Reference Laboratory, Dr. Lal Pathlabs, B7 Road, Block E, Rohini Sector 18, New Delhi, 110085, India
| | - Sher Ali
- Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India.
- Present Address: Department of Personalized Medicine, VC Office, Era University, Lucknow, 226003, India.
| |
Collapse
|
40
|
Ding Y, Peng Y, Wu H, Huang Y, Sheng K, Li C, Chu M, Ji W, Guo X. The protective roles of liraglutide on Kawasaki disease via AMPK/mTOR/NF-κB pathway. Int Immunopharmacol 2023; 117:110028. [PMID: 36934674 DOI: 10.1016/j.intimp.2023.110028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/19/2023]
Abstract
Kawasaki disease (KD) is an acute febrile rash illness among children of unknown etiology, with coronary artery injury. The main purpose of this study was to investigate the protective effects of liraglutide on KD, and elucidate the underlying mechanisms. The candida albicans water-soluble fraction (CAWS)-induced coronary arteritis of mouse KD model in vivo and tumor necrosis factor α (TNF-α) induced endothelial cell injury of human umbilical vein endothelial cell (HUVEC) model in vitro were used to explore the anti-inflammation and anti-apoptosis effects of liraglutide on KD. In vivo results showed that liraglutide could significantly alleviate the coronary artery injury of KD mice, as evidenced by the reduction of inflammatory infiltration around the coronary arteries, downregulation of inflammatory cytokines and chemokines expressions, and decrease of TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling) positive cell rates. The results in vitro also displayed that liraglutide could markedly relieve the inflammatory of TNF-α induced HUVECs through downregulating the expressions of inflammatory and chemokine indicators as well as inhibit TNF-α induced HUVEC apoptosis by the less ratio of apoptotic cells, the more loss of mitochondrial membrane potential (△Ψm), the lower level of intracellular reactive oxygen species (ROS), and the more ratio of BCL-2/BAX. Further in vivo and in vitro studies demonstrated that liraglutide could rescue endothelial cell injury through AMPK/mTOR/NF-κB pathway. In conclusion, liraglutide could play protective roles on KD through inhibiting endothelial cell inflammation and apoptosis via the activation of AMPK/mTOR/NF-κB pathway.
Collapse
Affiliation(s)
- Yinjuan Ding
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongmiao Peng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huilan Wu
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Huang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Sheng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chao Li
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Maoping Chu
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Weiping Ji
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of General Surgery, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Children Genitourinary Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
41
|
Leak L, Dixon SJ. Surveying the landscape of emerging and understudied cell death mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119432. [PMID: 36690038 PMCID: PMC9969746 DOI: 10.1016/j.bbamcr.2023.119432] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/09/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
Cell death can be a highly regulated process. A large and growing number of mammalian cell death mechanisms have been described over the past few decades. Major pathways with established roles in normal or disease biology include apoptosis, necroptosis, pyroptosis and ferroptosis. However, additional non-apoptotic cell death mechanisms with unique morphological, genetic, and biochemical features have also been described. These mechanisms may play highly specialized physiological roles or only become activated in response to specific lethal stimuli or conditions. Understanding the nature of these emerging and understudied mechanisms may provide new insight into cell death biology and suggest new treatments for diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Logan Leak
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
42
|
Apaza Alccayhuaman KA, Heimel P, Lee JS, Tangl S, Kuchler U, Marchesan J, Panahipour L, Lettner S, Matalová E, Gruber R. FasL is a catabolic factor in alveolar bone homeostasis. J Clin Periodontol 2023; 50:396-405. [PMID: 36384160 PMCID: PMC10946845 DOI: 10.1111/jcpe.13750] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/20/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022]
Abstract
AIM Fas ligand (FasL) belongs to the tumour necrosis factor superfamily regulating bone turnover, inflammation, and apoptosis. The appendicular and axial skeleton phenotype of mature Faslgld mice has been reported. The impact of FasL on the alveolar bone providing support for the teeth at mature stages under healthy and induced inflammatory conditions remains unknown. MATERIALS AND METHODS We performed a phenotypical analysis of mice carrying the homozygous Faslgld mutation and wild-type (WT) mice (C57BL/6) under healthy conditions and upon ligature-induced periodontitis. After 12 days, micro-computed tomography analysis revealed the distance between the cement enamel junction and the alveolar bone crest. Additional structural parameters, such as the bone volume fraction (BV/TV) and the periodontal ligament space volume, were measured. Histological analyses were performed to visualize the catabolic changes at the defect site. RESULTS Healthy Faslgld mice were found to have more periodontal bone than their WT littermates. Faslgld had no significant effect on inflammatory osteolysis compared to WT controls with ligatures. Histology revealed eroded surfaces at the root and in the inter-proximal bone in both strains. CONCLUSIONS Our findings suggest that FasL is a catabolic factor in alveolar bone homeostasis but it does not affect the inflammatory osteolysis.
Collapse
Affiliation(s)
- Karol Alí Apaza Alccayhuaman
- Department of Oral BiologyUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Karl Donath Laboratory for Hard Tissue and Biomaterial ResearchUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Patrick Heimel
- Karl Donath Laboratory for Hard Tissue and Biomaterial ResearchUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Department for BioimagingLudwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVAViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Jung Seok Lee
- Department of Oral BiologyUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Department of PeriodontologyResearch Institute for Periodontal Regeneration, College of Dentistry, Yonsei UniversitySeoulRepublic of Korea
| | - Stefan Tangl
- Karl Donath Laboratory for Hard Tissue and Biomaterial ResearchUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Ulrike Kuchler
- Department of Oral SurgeryUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Julie Marchesan
- Division of Comprehensive Oral HealthAdams School of Dentistry, University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Layla Panahipour
- Department of Oral BiologyUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
| | - Stefan Lettner
- Karl Donath Laboratory for Hard Tissue and Biomaterial ResearchUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Eva Matalová
- Laboratory of Odontogenesis and OsteogenesisInstitute of Animal Physiology and Genetics, Czech Academy of SciencesBrnoCzech Republic
| | - Reinhard Gruber
- Department of Oral BiologyUniversity Clinic of Dentistry, Medical University of ViennaViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Department of PeriodontologySchool of Dental Medicine, University of BernBernSwitzerland
| |
Collapse
|
43
|
Liu Z, Wang J, Dai F, Zhang D, Li W. DUSP1 mediates BCG induced apoptosis and inflammatory response in THP-1 cells via MAPKs/NF-κB signaling pathway. Sci Rep 2023; 13:2606. [PMID: 36788275 PMCID: PMC9926451 DOI: 10.1038/s41598-023-29900-6] [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: 10/25/2022] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Tuberculosis (TB) is a zoonotic infectious disease caused by Mycobacterium tuberculosis (Mtb). Apoptosis and necrosis caused by the interaction between the host and the pathogen, as well as the host's inflammatory response, play an important role in the pathogenesis of TB. Dual-specificity phosphatase 1 (DUSP1) plays a vital role in regulating the host immune responses. However, the role of DUSP1 in the regulation of THP-1 macrophage apoptosis induced by attenuated Mycobacterium bovis Bacillus Calmette-Guérin (BCG) infection remains unclear. In the present study, we report that infection with BCG significantly induces macrophage apoptosis and induces the production of DUSP1, TNF-α and IL-1β. DUSP1 knockdown significantly inhibited BCG-induced macrophage apoptosis and activation of MAPKs/NF-κB signaling pathway. In addition, DUSP1 knockdown suppressed BCG-induced inflammation in vivo. Taken together, this study demonstrates that DUSP1, as a regulator of MAPKs/NF-κB signaling pathway, plays a novel role in BCG-induced macrophage apoptosis and inflammatory response.
Collapse
Affiliation(s)
- Zhanyou Liu
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Yinchuan, 750021, Ningxia, China
- School of Life Sciences, Ningxia University, 539 W. Helanshan Road, Yinchuan, 750021, Ningxia, China
| | - Jianhong Wang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Yinchuan, 750021, Ningxia, China
- School of Life Sciences, Ningxia University, 539 W. Helanshan Road, Yinchuan, 750021, Ningxia, China
| | - Fan Dai
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Yinchuan, 750021, Ningxia, China
- School of Life Sciences, Ningxia University, 539 W. Helanshan Road, Yinchuan, 750021, Ningxia, China
| | - Dongtao Zhang
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Yinchuan, 750021, Ningxia, China
- School of Life Sciences, Ningxia University, 539 W. Helanshan Road, Yinchuan, 750021, Ningxia, China
| | - Wu Li
- Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Yinchuan, 750021, Ningxia, China.
- School of Life Sciences, Ningxia University, 539 W. Helanshan Road, Yinchuan, 750021, Ningxia, China.
| |
Collapse
|
44
|
Qian R, Chen H, Lin H, Jiang Y, He P, Ding Y, Wu H, Peng Y, Wang L, Chen C, Wang D, Ji W, Guo X, Shan X. The protective roles of allicin on type 1 diabetes mellitus through AMPK/mTOR mediated autophagy pathway. Front Pharmacol 2023; 14:1108730. [PMID: 36817124 PMCID: PMC9937553 DOI: 10.3389/fphar.2023.1108730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Background: Type 1 diabetes mellitus (T1DM) is one of the most common endocrine and metabolic diseases in children. Pancreatic β cells are thought to be critical cells involved in the progression of T1DM, and their injury would directly lead to impaired insulin secretion. Purpose: To investigate the protective effects of allicin on pancreatic β cell injury and elucidate the underlying mechanism. Methods: The streptozotocin (STZ)-induced mouse T1DM model in vivo and STZ-induced pancreatic β cell Min6 model in vitro were used to explore the effects of allicin on T1DM. The experiments include fasting blood glucose test, oral glucose tolerance detection, HE staining, immunohistochemistry, immunofluorescence, TUNEL staining, western blot, real-time quantitative PCR (RT-qPCR), and flow cytometry. Results: Allicin could significantly decrease blood glucose level, improve islet structure and insulin expression, and inhibit apoptosis to reduce STZ-induced pancreatic β cell injury and loss through activating AMPK/mTOR mediated autophagy pathway. Conclusion: Allicin treatment significantly reduced STZ-induced T1DM progression, suggesting that allicin may be a potential therapy option for T1DM patients.
Collapse
Affiliation(s)
- Rengcheng Qian
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huihui Chen
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongzhou Lin
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yalan Jiang
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingping He
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yinjuan Ding
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huilan Wu
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongmiao Peng
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingfei Wang
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congde Chen
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Children Genitourinary Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dexuan Wang
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Children Genitourinary Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiping Ji
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Weiping Ji, ; Xiaoling Guo, ; Xiaoou Shan,
| | - Xiaoling Guo
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Children Genitourinary Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Basic Medical Research Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Weiping Ji, ; Xiaoling Guo, ; Xiaoou Shan,
| | - Xiaoou Shan
- Department of Pediatrics, The Second Schoozl of Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Children Genitourinary Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Structural Malformations in Children of Zhejiang Province, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Weiping Ji, ; Xiaoling Guo, ; Xiaoou Shan,
| |
Collapse
|
45
|
de Paula JJ, Paiva RERP, Souza-Silva NG, Rosa DV, Duran FLDS, Coimbra RS, Costa DDS, Dutenhefner PR, Oliveira HSD, Camargos ST, Vasconcelos HMM, de Oliveira Carvalho N, da Silva JB, Silveira MB, Malamut C, Oliveira DM, Molinari LC, de Oliveira DB, Januário JN, Silva LC, De Marco LA, Queiroz DMDM, Meira W, Busatto G, Miranda DM, Romano-Silva MA. Selective visuoconstructional impairment following mild COVID-19 with inflammatory and neuroimaging correlation findings. Mol Psychiatry 2023; 28:553-563. [PMID: 35701598 PMCID: PMC9196149 DOI: 10.1038/s41380-022-01632-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 01/07/2023]
Abstract
People recovered from COVID-19 may still present complications including respiratory and neurological sequelae. In other viral infections, cognitive impairment occurs due to brain damage or dysfunction caused by vascular lesions and inflammatory processes. Persistent cognitive impairment compromises daily activities and psychosocial adaptation. Some level of neurological and psychiatric consequences were expected and described in severe cases of COVID-19. However, it is debatable whether neuropsychiatric complications are related to COVID-19 or to unfoldings from a severe infection. Nevertheless, the majority of cases recorded worldwide were mild to moderate self-limited illness in non-hospitalized people. Thus, it is important to understand what are the implications of mild COVID-19, which is the largest and understudied pool of COVID-19 cases. We aimed to investigate adults at least four months after recovering from mild COVID-19, which were assessed by neuropsychological, ocular and neurological tests, immune markers assay, and by structural MRI and 18FDG-PET neuroimaging to shed light on putative brain changes and clinical correlations. In approximately one-quarter of mild-COVID-19 individuals, we detected a specific visuoconstructive deficit, which was associated with changes in molecular and structural brain imaging, and correlated with upregulation of peripheral immune markers. Our findings provide evidence of neuroinflammatory burden causing cognitive deficit, in an already large and growing fraction of the world population. While living with a multitude of mild COVID-19 cases, action is required for a more comprehensive assessment and follow-up of the cognitive impairment, allowing to better understand symptom persistence and the necessity of rehabilitation of the affected individuals.
Collapse
Affiliation(s)
- Jonas Jardim de Paula
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
- Departamento de Saúde Mental, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Rachel E R P Paiva
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
| | - Nathália Gualberto Souza-Silva
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
| | - Daniela Valadão Rosa
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
| | | | - Roney Santos Coimbra
- Neurogenômica / Imunopatologia. Instituto René Rachou, Fiocruz, Belo Horizonte-MG, Brazil
| | - Danielle de Souza Costa
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
| | - Pedro Robles Dutenhefner
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
- Departamento de Computação Científica, ICEX, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Henrique Soares Dutra Oliveira
- Departamento de Clínica Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Sarah Teixeira Camargos
- Departamento de Clínica Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Herika Martins Mendes Vasconcelos
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
| | - Nara de Oliveira Carvalho
- Núcleo de Ações e Pesquisa em Apoio Diagnóstico (NUPAD), Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | | | | | - Carlos Malamut
- UPPR, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte-MG, Brazil
| | - Derick Matheus Oliveira
- Departamento de Computação Científica, ICEX, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Luiz Carlos Molinari
- Departamento de Saúde Mental, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Danilo Bretas de Oliveira
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - José Nélio Januário
- Departamento de Clínica Médica, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
- Núcleo de Ações e Pesquisa em Apoio Diagnóstico (NUPAD), Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | | | - Luiz Armando De Marco
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
- Departamento de Cirurgia, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | | | - Wagner Meira
- Departamento de Psiquiatria, Faculdade de Medicina da USP, São Paulo-SP, Brazil
- Centro de Inovação em Inteligência Artificial para a Saúde (CIIAS-Saúde), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Geraldo Busatto
- Departamento de Psiquiatria, Faculdade de Medicina da USP, São Paulo-SP, Brazil
| | - Débora Marques Miranda
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil
- Centro de Inovação em Inteligência Artificial para a Saúde (CIIAS-Saúde), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
- Departamento de Pediatria, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil
| | - Marco Aurélio Romano-Silva
- Centro de Tecnologia em Medicina Molecular (CTMM), Universidade Federal de Minas Gerais (UFMG), Av Alfredo Balena 190, Belo Horizonte-MG, Brazil.
- Departamento de Saúde Mental, Faculdade de Medicina da Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil.
- Centro de Inovação em Inteligência Artificial para a Saúde (CIIAS-Saúde), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte-MG, Brazil.
| |
Collapse
|
46
|
Recent development of multi-targeted inhibitors of human topoisomerase II enzyme as potent cancer therapeutics. Int J Biol Macromol 2023; 226:473-484. [PMID: 36495993 DOI: 10.1016/j.ijbiomac.2022.12.013] [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: 08/30/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Multi-target therapies have been considered one of the viable options to overcome the challenges to eradicate intrinsic and acquired drug-resistant cancer cells. While to increase the efficacy of therapeutics, the use of a single drug against multiple structurally similar sites, which noncommittedly modulate several vital cellular pathways proposed as a potential alternative to a 'single drug single target'. Besides, it reduces the usage of a number of drugs and their side effects. Topoisomerase II enzyme plays a very significant role in DNA replication and thus served as an important target for numerous anti-cancer agents. However, in spite of promising clinical results, in several cases, it was found that cancer cells have developed resistance against the anti-cancer agents targeting this enzyme. Therefore, multi-target therapies have been proposed as an alternative to overcome different drug resistance mechanisms while topoisomerases II are a primary target site. In this review, we have tried to discuss the characteristics of the binding cavity available for interactions of drugs, and potent inhibitors concurrently modulate the functions of topoisomerases II as well as other structurally related target sites. Additionally, the mechanism of drug resistance by considering molecular and cellular insights by including various types of cancers.
Collapse
|
47
|
Franzoni G, Pedrera M, Sánchez-Cordón PJ. African Swine Fever Virus Infection and Cytokine Response In Vivo: An Update. Viruses 2023; 15:233. [PMID: 36680273 PMCID: PMC9864779 DOI: 10.3390/v15010233] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023] Open
Abstract
African swine fever (ASF) is a hemorrhagic viral disease of domestic pigs and wild suids (all Sus scrofa) caused by the ASF virus (ASFV). The disease is spreading worldwide without control, threatening pig production due to the absence of licensed vaccine or commercially available treatments. A thorough understanding of the immunopathogenic mechanisms behind ASFV infection is required to better fight the disease. Cytokines are small, non-structural proteins, which play a crucial role in many aspects of the immune responses to viruses, including ASFV. Infection with virulent ASFV isolates often results in exacerbated immune responses, with increased levels of serum pro-inflammatory interleukins (IL-1α, IL-1β, IL-6), TNF and chemokines (CCL2, CCL5, CXCL10). Increased levels of IL-1, IL-6 and TNF are often detected in several tissues during acute ASFV infections and associated with lymphoid depletion, hemorrhages and oedemas. IL-1Ra is frequently released during ASFV infection to block further IL-1 activity, with its implication in ASFV immunopathology having been suggested. Increased levels of IFN-α and of the anti-inflammatory IL-10 seem to be negatively correlated with animal survival, whereas some correlation between virus-specific IFN-γ-producing cells and protection has been suggested in different studies where different vaccine candidates were tested, although future works should elucidate whether IFN-γ release by specific cell types is related to protection or disease development.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, 07100 Sassari, Italy
| | - Miriam Pedrera
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos, 28130 Madrid, Spain
| | - Pedro J. Sánchez-Cordón
- Centro de Investigación en Sanidad Animal (CISA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Consejo Superior de Investigaciones Científicas (CSIC), Valdeolmos, 28130 Madrid, Spain
| |
Collapse
|
48
|
Inoue T, Matsuda K, Matsusaka K, Nakajima M, Takeno Y, Miyazaki T, Shintaku T, Yoda N, Saito T, Ikeda E, Mano Y, Shinohara K, Rahmutulla B, Fukuyo M, Kita K, Nemoto T, Kaneda A. Anti-proliferating and apoptosis-inducing activity of chemical compound FTI-6D in association with p53 in human cancer cell lines. Chem Biol Interact 2023; 369:110257. [PMID: 36375514 DOI: 10.1016/j.cbi.2022.110257] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Compounds with 3,4-fused tricyclic indole (FTI) frameworks are attractive scaffolds for drug discovery. We synthesized FTI-6D, a compound with this framework, which was cytotoxic in several human cancer cell lines. FTI-6D induced apoptosis via activation of the p53 downstream mitochondria-related apoptotic pathway, characterized by an increased ratio of pro-apoptotic Bcl-2 family members to anti-apoptotic members. This change was followed by caspase-9 and caspase-3 cleavage and activation in two cancer cell lines, RKO and AGS. The anti-proliferating effect of FTI-6D was remarkably detected in eight cancer cells with wild-type TP53 (TP53_wt), including RKO and AGS, but not in seven cancer cells with mutated TP53 (TP53_mut). Additionally, p53 protein levels increased after FTI-6D treatment in TP53_wt cancer cells, and the cytotoxic effect of FTI-6D was decreased by TP53 knockdown. Accordingly, the expression of p53 downstream genes involved in apoptotic signaling pathways, such as BBC3 and TP53INP1, and those involved in cell growth inhibition, such as CDKN1A, was upregulated in TP53_wt cancer cells. These results suggest that the anti-proliferative and apoptosis-inducing activities of FTI-6D rely on p53 and the corresponding signaling processes. This study demonstrated that FTI-6D shows anti-cancer activity against TP53_wt cancer cells. FTI-6D may have potential as a prototype compound for a new drug to utilize a functional p53 pathway in TP53_wt cancer cells.
Collapse
Affiliation(s)
- Takato Inoue
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kazuaki Matsuda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Keisuke Matsusaka
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masaya Nakajima
- Department of Pharmaceutical Chemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8670, Japan
| | - Yukari Takeno
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Toko Miyazaki
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takahiko Shintaku
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Natsumi Yoda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Takahiko Saito
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Eriko Ikeda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Yasunobu Mano
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kenichi Shinohara
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Kazuko Kita
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Tetsuhiro Nemoto
- Department of Pharmaceutical Chemistry, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, 260-8670, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan.
| |
Collapse
|
49
|
İYİBOZKURT A, ÇAKMAKOĞLU B, ERTUGRUL B, İPLİK ES. NF-қB and COX-2 Relation Between Endometrial Cancer and the Clinicopathological Parameters. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2022. [DOI: 10.33808/clinexphealthsci.1049382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Objective: Our study examines nuclear factor kappa B (NF-қB) and cyclooxygenase-2 (COX-2) polymorphisms in the most common gynecological cancer type, endometrial cancer, and the relationship between disease parameters and these polymorphisms.
Methods: In our patient group; while 109 endometrial cancer patients were examined and treated in the Department of Gynecology and Obstetrics, Istanbul Medical Faculty, and 106 healthy women without the disease were included in the control group. DNA of blood samples taken from all groups were isolated; COX-2 765C> G and COX-2 1195A> G polymorphisms were studied with NF-қB-94 ins / delATTG. Genotypes analyzed using the PCR-based restriction fragment length polymorphisms (RFLP) method were investigated in terms of the relationship between endometrial cancer susceptibility and endometrial cancer disease parameters. Results in SPSS 17 program; Student’s t-tests were analyzed
using Anova, Fisher’s exact, and Chi-square tests.
Results: NF-қB D + and DD genotype, COX-2 765 G + and GG genotype, and COX-2 1195 AA genotype were found to be significantly more common in the endometrial cancer group compared to the control group (p
Collapse
Affiliation(s)
| | - Bedia ÇAKMAKOĞLU
- İSTANBUL ÜNİVERSİTESİ, AZİZ SANCAR DENEYSEL TIP ARAŞTIRMA ENSTİTÜSÜ
| | - Baris ERTUGRUL
- ISTANBUL UNIVERSITY, AZIZ SANCAR INSTITUTE OF EXPERIMENTAL MEDICINE
| | | |
Collapse
|
50
|
Niu J, Meng F, Hao Q, Zong C, Fu J, Xue H, Tian M, Yu X. Ratiometric and Discriminative Visualization of Autophagy and Apoptosis with a Single Fluorescent Probe Based on the Aggregation/Monomer Principle. Anal Chem 2022; 94:17885-17894. [PMID: 36516436 DOI: 10.1021/acs.analchem.2c03815] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autophagy and apoptosis play a central role in maintaining homeostasis in mammals. Therefore, discriminative visualization of the two cellular processes is an important and challenging task. However, fluorescent probes enabling ratiometric visualization of both autophagy and apoptosis with different sets of fluorescence signals have not been developed yet. In this work, we constructed a versatile single fluorescent probe (NKLR) based on the aggregation/monomer principle for the ratiometric and discriminative visualization of autophagy and apoptosis. NKLR can simultaneously perform two-color imaging of RNA (deep red channel) and lysosomes (yellow channel) in aggregation and monomer states, respectively. During autophagy, NKLR migrated from cytoplasmic RNA and nuclear RNA to lysosomes, showing enhanced yellow emission and sharply decreased deep red fluorescence. Moreover, this migration process was reversible upon the recovery of autophagy. Comparatively, during apoptosis, NKLR immigrated from lysosomes to RNA, and the yellow emission decreased and even disappeared, while the fluorescence of the deep red channel slightly increased. Overall, autophagy and apoptosis could be discriminatively visualized via the fluorescence intensity ratios of the two channels. Meanwhile, the cells in three different states (healthy, autophagic, apoptotic) could be distinguished by three point-to-point fluorescence images via the localization and emission color of NKLR. Therefore, the probe NKLR can serve as a desirable molecular tool to reveal the in-depth relation between autophagy and apoptosis and facilitate the study on the two cellular processes.
Collapse
Affiliation(s)
- Jie Niu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Fangfang Meng
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Qiuhua Hao
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Chong Zong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Jinyu Fu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| | - Haiyan Xue
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, P. R. China
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, P. R. China
| | - Xiaoqiang Yu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, Shandong 250100, P. R. China
| |
Collapse
|