1
|
Guilbaud E, Naulin F, Meziani L, Deutsch E, Galluzzi L. Impact of radiation therapy on the immunological tumor microenvironment. Cell Chem Biol 2025; 32:678-693. [PMID: 40280118 DOI: 10.1016/j.chembiol.2025.04.001] [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: 12/29/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
External beam radiation therapy (RT) is a cornerstone of modern cancer management, being utilized in both curative and palliative settings due to its safety, efficacy, and widespread availability. A primary biological effect of RT is DNA damage, which leads to significant cytostatic and cytotoxic effects. Importantly, malignant cells possess a limited capacity for DNA repair compared to normal cells, and when combined with irradiation techniques that minimize damage to healthy tissues, this creates an advantageous therapeutic window. However, the clinical effectiveness of RT also appears to involve both direct and indirect interactions between RT and non-transformed components of the tumoral ecosystem, particularly immune cells. In this review, we describe the molecular and cellular mechanisms by which irradiated cancer cells modify the immunological tumor microenvironment and how such changes ultimately impact tumor growth.
Collapse
Affiliation(s)
- Emma Guilbaud
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Flavie Naulin
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Lydia Meziani
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France
| | - Eric Deutsch
- Department of Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France; INSERM RAMO-IT U1030, Villejuif, France; Faculty of Medicine, University of Paris-Saclay, Le Kremlin, Bicêtre, France.
| | - Lorenzo Galluzzi
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Wu Q, Qi S, Kang Z, Bai X, Li Z, Cheng J, Dong X. PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target. J Inflamm Res 2025; 18:6245-6261. [PMID: 40386177 PMCID: PMC12085136 DOI: 10.2147/jir.s513367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.
Collapse
Affiliation(s)
- Qiqi Wu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhaofeng Kang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Cheng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
3
|
Tye H, Conos SA, Djajawi TM, Gottschalk TA, Abdoulkader N, Kong IY, Kammoun HL, Narayana VK, Kratina T, Speir M, Emery J, Simpson DS, Hall C, Vince AJ, Russo S, Crawley R, Rashidi M, Hildebrand JM, Murphy JM, Whitehead L, De Souza DP, Masters SL, Samson AL, Lalaoui N, Hawkins ED, Murphy AJ, Vince JE, Lawlor KE. Divergent roles of RIPK3 and MLKL in high-fat diet-induced obesity and MAFLD in mice. Life Sci Alliance 2025; 8:e202302446. [PMID: 39532538 PMCID: PMC11557689 DOI: 10.26508/lsa.202302446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death frequently occurs in the pathogenesis of obesity and metabolic dysfunction-associated fatty liver disease (MAFLD). However, the exact contribution of core cell death machinery to disease manifestations remains ill-defined. Here, we show via the direct comparison of mice genetically deficient in the essential necroptotic regulators, receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL), as well as mice lacking apoptotic caspase-8 in myeloid cells combined with RIPK3 loss, that RIPK3/caspase-8 signaling regulates macrophage inflammatory responses and drives adipose tissue inflammation and MAFLD upon high-fat diet feeding. In contrast, MLKL, divergent to RIPK3, contributes to both obesity and MAFLD in a manner largely independent of inflammation. We also uncover that MLKL regulates the expression of molecules involved in lipid uptake, transport, and metabolism, and congruent with this, we discover a shift in the hepatic lipidome upon MLKL deletion. Collectively, these findings highlight MLKL as an attractive therapeutic target to combat the growing obesity pandemic and metabolic disease.
Collapse
Affiliation(s)
- Hazel Tye
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
| | - Stephanie A Conos
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Tirta M Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Timothy A Gottschalk
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Nasteho Abdoulkader
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
| | - Isabella Y Kong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | | | - Vinod K Narayana
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | | | - Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Jack Emery
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Daniel S Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Angelina J Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Sophia Russo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Rhiannan Crawley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Maryam Rashidi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Joanne M Hildebrand
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Seth L Masters
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Andre L Samson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Najoua Lalaoui
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Edwin D Hawkins
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | | | - James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- The Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
4
|
Bhardwaj M, Mazumder PM. The gut-liver axis: emerging mechanisms and therapeutic approaches for nonalcoholic fatty liver disease and type 2 diabetes mellitus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8421-8443. [PMID: 38861011 DOI: 10.1007/s00210-024-03204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), more appropriately known as metabolic (dysfunction) associated fatty liver disease (MAFLD), a prevalent condition in type 2 diabetes mellitus (T2DM) patients, is a complex condition involving hepatic lipid accumulation, inflammation, and liver fibrosis. The gut-liver axis is closely linked to metabolic dysfunction, insulin resistance, inflammation, and oxidative stress that are leading to the cooccurrence of MAFLD and T2DM cardiovascular diseases (CVDs). The purpose of this review is to raise awareness about the role of the gut-liver axis in the progression of MAFLD, T2DM and CVDs with a critical analysis of available treatment options for T2DM and MAFLD and their impact on cardiovascular health. This study analysed over 100 articles on this topic, using online searches and predefined keywords, to understand and summarise published research. Numerous studies have shown a strong correlation between gut dysfunction, particularly the gut microbiota and its metabolites, and the occurrence and progression of MAFLD and type 2 diabetes mellitus (T2DM). Herein, this article also examines the impact of the gut-liver axis on MAFLD, T2DM, and related complications, focusing on the role of gut microbiota dysbiosis in insulin resistance, T2DM and obesity-related cardiovascular complications. The study suggests potential treatment targets for MAFLD linked to T2DM, focusing on cardiovascular outcomes and the molecular mechanism of the gut-liver axis, as gut microbiota dysbiosis contributes to obesity-related metabolic abnormalities.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
5
|
Kasana S, Kumar S, Patel P, Kurmi BD, Jain S, Sahu S, Vaidya A. Caspase inhibitors: a review on recently patented compounds (2016-2023). Expert Opin Ther Pat 2024; 34:1047-1072. [PMID: 39206873 DOI: 10.1080/13543776.2024.2397732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Caspases are a family of protease enzymes that play a crucial role in apoptosis. Dysregulation of caspase activity has been implicated in various pathological conditions, making caspases an important focus of research in understanding cell death mechanisms and developing therapeutic strategies for diseases associated with abnormal apoptosis. AREAS COVERED It is a comprehensive review of caspase inhibitors that have been comprising recently granted patents from 2016 to 2023. It includes peptide and non-peptide caspase inhibitors with their application for different diseases. EXPERT OPINION This review categorizes and analyses recently patented caspase inhibitors on various diseases. Diseases linked to caspase dysregulation, including neurodegenerative disorders, and autoimmune conditions, are highlighted to accentuate the therapeutic relevance of the patented caspase inhibitors. This paper serves as a valuable resource for researchers, clinicians, and pharmaceutical developers seeking an up-to-date understanding of recently patented caspase inhibitors. The integration of recent patented compounds, structural insights, and mechanistic details provides a holistic view of the progress in caspase inhibitor research and its potential impact on addressing various diseases.
Collapse
Affiliation(s)
- Shivani Kasana
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Shivam Kumar
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, India
| | - Shweta Jain
- Sir Madanlal Institute of Pharmacy, Etawah, India
| | - Sanjeev Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ankur Vaidya
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Etawah, India
| |
Collapse
|
6
|
Nassauer L, Schott JW, Harre J, Warnecke A, Morgan M, Galla M, Schambach A. The caspase-inhibitor Emricasan efficiently counteracts cisplatin- and neomycin-induced cytotoxicity in cochlear cells. J Mol Med (Berl) 2024; 102:1163-1174. [PMID: 39110182 PMCID: PMC11358181 DOI: 10.1007/s00109-024-02472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/30/2024]
Abstract
Cisplatin is a chemotherapeutic agent widely used to treat solid tumors. However, it can also be highly ototoxic, resulting in high-frequency hearing loss. Cisplatin causes degeneration of hair cells (HCs) and spiral ganglion neurons (SGNs) in the inner ear, which are essential components of the hearing process and cannot be regenerated in mammals. As the affected cells primarily die by apoptosis, we tested several anti-apoptotic small molecules to protect these cells from drug-induced toxicity. We found that the general caspase inhibitor Emricasan could significantly counteract the toxic effects of cisplatin in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells, phoenix auditory cells, and primary SGNs. Importantly, the anti-cytotoxic effect in neuronal cells was even more pronounced than the effect of sodium thiosulfate (STS), which is currently the only approved prevention option for cisplatin-induced ototoxicity. Finally, we tested the protective effect of Emricasan treatment in the context of another ototoxic drug, i.e., the aminoglycoside antibiotic neomycin, and again found a significant increase in cell viability when the cultures were co-treated with Emricasan. These results suggest a promising strategy to prevent ototoxicity in patients by temporarily blocking the apoptotic pathway when applying cisplatin or aminoglycoside antibiotics. KEY MESSAGES: Anti-apoptotic small molecules can reduce cisplatin-induced toxicity. Emricasan can effectively exert its anti-apoptotic effect on cochlear cells. Strong protection from cisplatin- and neomycin-induced cytotoxicity with Emricasan. Sodium thiosulfate and Emricasan provide similar protective effects to cisplatin-treated cells. Emricasan is more potent than sodium thiosulfate in reducing neomycin-induced cytotoxicity.
Collapse
Affiliation(s)
- Larissa Nassauer
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625, Hannover, Germany
| | - Juliane W Schott
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625, Hannover, Germany
| | - Jennifer Harre
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625, Hannover, Germany
- Cluster of Excellence "Hearing4all", Hannover Medical School, 30625, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625, Hannover, Germany
- Cluster of Excellence "Hearing4all", Hannover Medical School, 30625, Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625, Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625, Hannover, Germany.
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625, Hannover, Germany.
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Fuster-Martínez I, Calatayud S. The current landscape of antifibrotic therapy across different organs: A systematic approach. Pharmacol Res 2024; 205:107245. [PMID: 38821150 DOI: 10.1016/j.phrs.2024.107245] [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: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Fibrosis is a common pathological process that can affect virtually all the organs, but there are hardly any effective therapeutic options. This has led to an intense search for antifibrotic therapies over the last decades, with a great number of clinical assays currently underway. We have systematically reviewed all current and recently finished clinical trials involved in the development of new antifibrotic drugs, and the preclinical studies analyzing the relevance of each of these pharmacological strategies in fibrotic processes affecting tissues beyond those being clinically studied. We analyze and discuss this information with the aim of determining the most promising options and the feasibility of extending their therapeutic value as antifibrotic agents to other fibrotic conditions.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia 46020, Spain.
| | - Sara Calatayud
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; CIBERehd (Centro de Investigación Biomédica en Red - Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
8
|
Prado-Acosta M, Jeong S, Utrero-Rico A, Goncharov T, Webster JD, Holler E, Morales G, Dellepiane S, Levine JE, Rothenberg ME, Vucic D, Ferrara JLM. Inhibition of RIP1 improves immune reconstitution and reduces GVHD mortality while preserving graft-versus-leukemia effects. Sci Transl Med 2023; 15:eadf8366. [PMID: 38117900 PMCID: PMC11157567 DOI: 10.1126/scitranslmed.adf8366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Graft-versus-host disease (GVHD) remains the major cause of morbidity and nonrelapse mortality (NRM) after hematopoietic cell transplantation (HCT). Inflammatory cytokines mediate damage to key GVHD targets such as intestinal stem cells (ISCs) and also activate receptor interacting protein kinase 1 (RIP1; RIPK1), a critical regulator of apoptosis and necroptosis. We therefore investigated the role of RIP1 in acute GVHD using samples from HCT patients, modeling GVHD damage in vitro with both human and mouse gastrointestinal (GI) organoids, and blocking RIP1 activation in vivo using several well-characterized mouse HCT models. Increased phospho-RIP1 expression in GI biopsies from patients with acute GVHD correlated with tissue damage and predicted NRM. Both the genetic inactivation of RIP1 and the RIP1 inhibitor GNE684 prevented GVHD-induced apoptosis of ISCs in vivo and in vitro. Daily administration of GNE684 for 14 days reduced inflammatory infiltrates in three GVHD target organs (intestine, liver, and spleen) in mice. Unexpectedly, GNE684 administration also reversed the marked loss of regulatory T cells in the intestines and liver during GVHD and reduced splenic T cell exhaustion, thus improving immune reconstitution. Pharmacological and genetic inhibition of RIP1 improved long-term survival without compromising the graft-versus-leukemia (GVL) effect in lymphocytic and myeloid leukemia mouse models. Thus, RIP1inhibition may represent a nonimmunosuppressive treatment for GVHD.
Collapse
Affiliation(s)
- Mariano Prado-Acosta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Seihwan Jeong
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alberto Utrero-Rico
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Joshua D. Webster
- Department of Pathology, Genentech, South San Francisco, CA 94080, USA
| | - Ernst Holler
- Department of Hematology and Oncology, University of Regensburg, Regensburg 93042, Germany
| | - George Morales
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergio Dellepiane
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John E. Levine
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Domagoj Vucic
- Immunology Discovery, Genentech, South San Francisco, CA 94080, USA
| | - James L. M. Ferrara
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
9
|
Qi Z, Zhu L, Wang K, Wang N. PANoptosis: Emerging mechanisms and disease implications. Life Sci 2023; 333:122158. [PMID: 37806654 DOI: 10.1016/j.lfs.2023.122158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
PANoptosis, a unique new form of programmed cell death (PCD), is characterized by pyroptosis, apoptosis, and necroptosis, but it cannot be explained by pyroptosis, apoptosis or necroptosis alone. Assembly of the PANoptosome complex is a key feature of PANoptosis. To date, four kinds of PANoptosomes with distinct sensors and regulators have been defined, namely Z-DNA binding protein 1 (ZBP1) PANoptosome, absent in melanoma 2 (AIM2) PANoptosome, receptor-interacting protein kinase 1 (RIPK1) PANoptosome, and nucleotide-binding leucine-rich repeat-containing receptor 12 (NLRP12). Each PANoptosome contains three components: sensors for pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), adaptors as connected bridges, and catalytic effectors or executioners. Mechanistically, different PAMPs or DAMPs are recognized by the sensors in a context-dependent manner, which initiates PANoptosome assembly through adaptors, and ultimately engages synchronous activation of pyroptosis, apoptosis, and necroptosis via different catalytic effectors. Resultantly, PANoptosis is emerged as a prospective and promising therapeutic target for various diseases. This review covers the accumulating evidence about the roles and mechanisms of PANoptosis in innate immunity and discusses the attractive prospect of manipulating PANoptosis as a new treatment for diseases.
Collapse
Affiliation(s)
- Zehong Qi
- Department of Pathophysiology, Key Laboratory of Sepsis Translational Medicine of Hunan, School of Basic Medical Science, Central South University, 410008 Changsha, Hunan, China
| | - Lili Zhu
- Department of Pathophysiology, Key Laboratory of Sepsis Translational Medicine of Hunan, School of Basic Medical Science, Central South University, 410008 Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, Key Laboratory of Sepsis Translational Medicine of Hunan, School of Basic Medical Science, Central South University, 410008 Changsha, Hunan, China.
| | - Nian Wang
- Department of Pathophysiology, Key Laboratory of Sepsis Translational Medicine of Hunan, School of Basic Medical Science, Central South University, 410008 Changsha, Hunan, China.
| |
Collapse
|
10
|
Zhang X, Zhang H, Zhang J, Yang M, Zhu M, Yin Y, Fan X, Yu F. The paradoxical role of radiation-induced cGAS-STING signalling network in tumour immunity. Immunology 2023; 168:375-388. [PMID: 36217274 DOI: 10.1111/imm.13592] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 10/06/2022] [Indexed: 11/27/2022] Open
Abstract
The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is an essential component of the innate immune system and is central to the identification of abnormal DNA leakage caused by ionising radiation (IR) damage. Cell-intrinsic cGAS-STING initiation has been revealed to have tremendous potential for facilitating interferon synthesis and T-cell priming. Targeting the cGAS-STING axis has been proposed as a strategy to improve radiosensitivity or enhance immunosurveillance. However, due to the complex biology of the irradiated tumour microenvironment and the extensive involvement of the cGAS-STING pathway in various physiological and pathological processes, many defects in this strategy limit the therapeutic effect. Here, we outline the molecular mechanisms by which IR activates the cGAS-STING pathway and analyse the dichotomous roles of the cGAS-STING pathway in modulating cancer immunity after radiotherapy (RT). Then, based on the crosstalk between the cGAS-STING pathway and other signalling events induced by IR, such as necroptosis, autophagy and other cellular effects, we discuss the immunomodulatory actions of the broad cGAS-STING signalling network in RT and their potential therapeutic applications. Finally, recent advances in combination therapeutic strategies targeting cGAS-STING in RT are explored.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Han Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Jiajia Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Mengqin Zhu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Yuzhen Yin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Xin Fan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Abstract
Numerous mitochondrial constituents and metabolic products can function as damage-associated molecular patterns (DAMPs) and promote inflammation when released into the cytosol or extracellular milieu. Several safeguards are normally in place to prevent mitochondria from eliciting detrimental inflammatory reactions, including the autophagic disposal of permeabilized mitochondria. However, when the homeostatic capacity of such systems is exceeded or when such systems are defective, inflammatory reactions elicited by mitochondria can become pathogenic and contribute to the aetiology of human disorders linked to autoreactivity. In addition, inefficient inflammatory pathways induced by mitochondrial DAMPs can be pathogenic as they enable the establishment or progression of infectious and neoplastic disorders. Here we discuss the molecular mechanisms through which mitochondria control inflammatory responses, the cellular pathways that are in place to control mitochondria-driven inflammation and the pathological consequences of dysregulated inflammatory reactions elicited by mitochondrial DAMPs.
Collapse
Affiliation(s)
- Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|