1
|
Tian L, Piao S, Li X, Guo L, Huang L, Gao W. Functional Materials Targeted Regulation of Gasdermins: From Fundamentals to Functionalities and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500873. [PMID: 40273335 PMCID: PMC12021126 DOI: 10.1002/advs.202500873] [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: 01/14/2025] [Revised: 02/19/2025] [Indexed: 04/26/2025]
Abstract
Targeted regulation of pyroptosis to modulate the immune landscape has emerged as a novel design strategy for cancer immunotherapy and anti-inflammatory therapy. However, pyroptosis acts as a double-edged sword, making it important to optimize the design strategies of functional materials to appropriately activate pyroptosis for effective disease treatment. This paper summarizes and discusses the structure, pore formation, and molecular mechanisms of "executor" Gasdermins, as well as the events preceding and following these processes. Subsequently, the focus is on reviewing functional materials that directly regulate Gasdermin pore formation to target pyroptosis and those that indirectly regulate the events before and after Gasdermin pore formation to control pyroptosis activity. Finally, the advantages, disadvantages, and future prospects of designing such functional materials are provided, aiming to facilitate the precise design, pharmacological investigation, and clinical translation of pyroptosis-related functional materials.
Collapse
Affiliation(s)
- Luyao Tian
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Shuo Piao
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Xia Li
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Lanping Guo
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Luqi Huang
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Wenyuan Gao
- Key Laboratory of Pharmacology School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| |
Collapse
|
2
|
Kaczor S, Szewczyk-Roszczenko O, Pawlak D, Hermanowicz A, Hermanowicz JM. GSDM family and glioma. Biochim Biophys Acta Rev Cancer 2025; 1880:189283. [PMID: 39929378 DOI: 10.1016/j.bbcan.2025.189283] [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: 08/28/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Biomarkers play a central role in diagnosing, prognosis, and therapeutic management of gliomas, a diverse group of malignancies arising from glial cells in the brain and spinal cord. Among the various emerging biomarkers, the gasdermin protein family has attracted attention for its involvement in pyroptosis. Understanding the expression and function of GSDM in gliomas may provide new insights into tumor behavior and new avenues for therapeutic intervention. This review discusses the GSDM family's significance as a glioma biomarker, explores its dual role in tumor suppression, and highlights its potential utility in clinical practice as a novel target for glioma therapy.
Collapse
Affiliation(s)
- Szymon Kaczor
- Neurosurgery Department, Military Clinical Hospital in Elk, Kosciuszki 30, 19-300 Elk, Poland
| | - Olga Szewczyk-Roszczenko
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Adam Hermanowicz
- Department of Pediatric Surgery, Medical University of Bialystok, Waszyngtona 17, 15-274 Bialystok, Poland
| | | |
Collapse
|
3
|
Duan Y, Yao RQ, Ling H, Zheng LY, Fan Q, Li Q, Wang L, Zhou QY, Wu LM, Dai XG, Yao YM. Organellophagy regulates cell death:A potential therapeutic target for inflammatory diseases. J Adv Res 2025; 70:371-391. [PMID: 38740259 PMCID: PMC11976430 DOI: 10.1016/j.jare.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Dysregulated alterations in organelle structure and function have a significant connection with cell death, as well as the occurrence and development of inflammatory diseases. Maintaining cell viability and inhibiting the release of inflammatory cytokines are essential measures to treat inflammatory diseases. Recently, many studies have showed that autophagy selectively targets dysfunctional organelles, thereby sustaining the functional stability of organelles, alleviating the release of multiple cytokines, and maintaining organismal homeostasis. Organellophagy dysfunction is critically engaged in different kinds of cell death and inflammatory diseases. AIM OF REVIEW We summarized the current knowledge of organellophagy (e.g., mitophagy, reticulophagy, golgiphagy, lysophagy, pexophagy, nucleophagy, and ribophagy) and the underlying mechanisms by which organellophagy regulates cell death. KEY SCIENTIFIC CONCEPTS OF REVIEW We outlined the potential role of organellophagy in the modulation of cell fate during the inflammatory response to develop an intervention strategy for the organelle quality control in inflammatory diseases.
Collapse
Affiliation(s)
- Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou 423000, China; Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; Department of General Surgery, the First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| | - Hua Ling
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou 423000, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qi Fan
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qiong Li
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou 423000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Le-Min Wu
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou 423000, China
| | - Xin-Gui Dai
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou 423000, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
4
|
Liu Y, Wang D, Liu X, Yuan H, Liu D, Hu Y, Ning S. Biological and pharmacological roles of pyroptosis in pulmonary inflammation and fibrosis: recent advances and future directions. Cell Commun Signal 2024; 22:586. [PMID: 39639365 PMCID: PMC11619304 DOI: 10.1186/s12964-024-01966-3] [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: 09/08/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Pyroptosis, an inflammatory regulated cell death (RCD) mechanism, is characterized by cellular swelling, membrane rupture, and subsequent discharge of cellular contents, exerting robust proinflammatory effects. Recent studies have significantly advanced our understanding of pyroptosis, revealing that it can be triggered through inflammasome- and caspase-independent pathways, and interacts intricately with other RCD pathways (e.g., pyroptosis, necroptosis, ferroptosis, and cuproptosis). The pathogenesis of pulmonary fibrosis (PF), including idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases, involves a multifaceted interplay of factors such as pathogen infections, environmental pollutants, genetic variations, and immune dysfunction. This chronic and progressive interstitial lung disease is characterized by persistent inflammation, extracellular matrix (ECM) accumulation, and fibrotic alveolar wall thickening, which potentially contribute to deteriorated lung function. Despite recent advances in understanding pyroptosis, the mechanisms by which it regulates PF are not entirely elucidated, and effective strategies to improve clinical outcomes remain unclear. This review strives to deliver a comprehensive overview of the biological functions and molecular mechanisms of pyroptosis, exploring its roles in the pathogenesis of PF. Furthermore, it examines potential biomarkers and therapeutic agents for anti-fibrotic treatments.
Collapse
Affiliation(s)
- Ya Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Danxia Wang
- Department of Pharmacy, People's Hospital of Ningxiang City, Hunan University of Chinese Medicine, Changsha, 410600, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Haibin Yuan
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Dan Liu
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Yixiang Hu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
5
|
Santos TSD, Bordiga P, Avan P. Auditory changes in awake guinea pigs exposed to overcompressed music. Hear Res 2024; 453:109120. [PMID: 39306941 DOI: 10.1016/j.heares.2024.109120] [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: 04/23/2024] [Revised: 09/01/2024] [Accepted: 09/14/2024] [Indexed: 11/05/2024]
Abstract
Exposure to loud sound during leisure time is identified as a significant risk factor for hearing by health authorities worldwide. The current standard that defines unsafe exposure rests on the equal-energy hypothesis, according to which the maximum recommended exposure is a tradeoff between level and daily exposure duration, a satisfactory recipe except for strongly non-Gaussian intense sounds such as gunshots. Nowadays, sound broadcast by music and videoconference streaming services makes extensive use of numerical dynamic range compression. By filling in millisecond-long valleys in the signal to prevent competing noise from masking, it pulls sound-level statistics away from a Gaussian distribution, the framework where the equal-energy hypothesis emerged. Auditory effects of a single 4 hour exposure to the same music were compared in two samples of guinea pigs exposed either to its original or overcompressed version played at the same average level of 102 dBA allowed by French regulations. Apart from a temporary shift of otoacoustic emissions at the lowest two frequencies 2 and 3 kHz, music exposure had no detectable cochlear effect, as monitored at 1, 2 and 7 days post-exposure. Conversely, middle-ear muscle strength behaved differentially as the group exposed to original music had fully recovered one day after exposure whereas the group exposed to overcompressed music remained stuck to about 50% of baseline even after 7 days. Subsamples were then re-exposed to the same music as the first time and sacrificed for density measurements of inner-hair-cell synapses. No difference in synaptic density was found compared to unexposed controls with either type of music. The present results show that the same music piece, harmless when played in its original version, induces a protracted deficit of one auditory neural pathway when overcompressed at the same level. The induced disorder does not seem to involve inner-hair cell synapses.
Collapse
Affiliation(s)
- Thamara Suzi Dos Santos
- Laboratoire de Biophysique Neurosensorielle, Université Clermont Auvergne, 63001 Clermont-Ferrand, France; Association La Semaine du son, Paris, France; Speech-Language Pathology and Audiology Department, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pierrick Bordiga
- Laboratoire de Biophysique Neurosensorielle, Université Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Paul Avan
- Laboratoire de Biophysique Neurosensorielle, Université Clermont Auvergne, 63001 Clermont-Ferrand, France; Université Paris Cité, Institut Pasteur, AP-HP, Inserm, Fondation Pour l'Audition, Institut de l'Audition, IHU reConnect, F-75012 Paris, France.
| |
Collapse
|
6
|
Zheng Y, Zhou Z, Liu M, Chen Z. Targeting selective autophagy in CNS disorders by small-molecule compounds. Pharmacol Ther 2024; 263:108729. [PMID: 39401531 DOI: 10.1016/j.pharmthera.2024.108729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/27/2024]
Abstract
Autophagy functions as the primary cellular mechanism for clearing unwanted intracellular contents. Emerging evidence suggests that the selective elimination of intracellular organelles through autophagy, compared to the increased bulk autophagic flux, is crucial for the pathological progression of central nervous system (CNS) disorders. Notably, autophagic removal of mitochondria, known as mitophagy, is well-understood in an unhealthy brain. Accumulated data indicate that selective autophagy of other substrates, including protein aggregates, liposomes, and endoplasmic reticulum, plays distinctive roles in various pathological stages. Despite variations in substrates, the molecular mechanisms governing selective autophagy can be broadly categorized into two types: ubiquitin-dependent and -independent pathways, both of which can be subjected to regulation by small-molecule compounds. Notably, natural products provide the remarkable possibility for future structural optimization to regulate the highly selective autophagic clearance of diverse substrates. In this context, we emphasize the selectivity of autophagy in regulating CNS disorders and provide an overview of chemical compounds capable of modulating selective autophagy in these disorders, along with the underlying mechanisms. Further exploration of the functions of these compounds will in turn advance our understanding of autophagic contributions to brain disorders and illuminate precise therapeutic strategies for these diseases.
Collapse
Affiliation(s)
- Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Zhuchen Zhou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Mengting Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
7
|
Sun J, Leng J, Song L. The Evolution of NLR Inflammasome and Its Mediated Pyroptosis in Metazoa. Int J Mol Sci 2024; 25:11167. [PMID: 39456947 PMCID: PMC11508797 DOI: 10.3390/ijms252011167] [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: 10/05/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) inflammasomes are multiprotein signaling platforms that control the inflammatory response and coordinate antimicrobial defense. In the present study, the distribution of NLR, Caspase-1, and gasdermin (GSDM) homologues and their structural characteristics and evolutionary relationships were systematically analyzed in metazoa according to the genomes of species. In invertebrates, there were only NLRC and/or NLRD presented from sponge to amphioxus, and according to the evolutionary tree, NLR from sponge located in the most primitive position. Caspase-1 existed in some metazoan phyla (Brachiopoda, Ectoprocta, Arthropoda, Mollusca, Annelia, Nematoda, Platyelminthes, Coelenterate, and Porifera) and its activation sites were relatively conserved. The amino acid sequences and three-dimensional structures of N-terminal CARD/Death domain of NLR and Caspase-1 were similar in species from sponge to human. NLR and Caspase-1 co-existed in species of Brachiopoda, Mollusca, Annelia, Coelenterate, and Porifera. There was only GSDME or PJVK found in some phyla of invertebrates and their cleavage sites were conserved (DxxD). And it was predicted that the NLR inflammasome in inducing pyroptosis could occur in species of Brachiopoda, Mollusca, Annelia, and Coelenterate. These studies indicated that NLR inflammasome emerged early in sponges of metazoa, and NLR inflammasome in inducing pyroptosis first appeared in Coelenterate, suggesting that inflammasome and its mediated pyroptosis had existed in the early stage of metazoa, but they had been lost in many species during evolution.
Collapse
Affiliation(s)
- Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China;
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Jinyuan Leng
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China;
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China;
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian 116023, China
- Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai 519000, China
| |
Collapse
|
8
|
Vu JT, Tavasoli KU, Sheedy CJ, Chowdhury SP, Mandjikian L, Bacal J, Morrissey MA, Richardson CD, Gardner BM. A genome-wide screen links peroxisome regulation with Wnt signaling through RNF146 and TNKS/2. J Cell Biol 2024; 223:e202312069. [PMID: 38967608 PMCID: PMC11223164 DOI: 10.1083/jcb.202312069] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 07/06/2024] Open
Abstract
Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerases TNKS and TNKS2, which bind the peroxisomal membrane protein PEX14. We propose that RNF146 and TNKS/2 regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased TNKS/2 and RNF146-dependent degradation of non-peroxisomal substrates, including the β-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of β-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation but also a novel role in bridging peroxisome function with Wnt/β-catenin signaling during development.
Collapse
Affiliation(s)
- Jonathan T. Vu
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Katherine U. Tavasoli
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Connor J. Sheedy
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Soham P. Chowdhury
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Lori Mandjikian
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Julien Bacal
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Meghan A. Morrissey
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Chris D. Richardson
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Brooke M. Gardner
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
9
|
Salame M, Bonnet C, Singh-Estivalet A, Brahim SM, Roux S, Boussaty EC, Hadrami M, Hamed CT, Sidi AM, Veten F, Petit C, Houmeida A. Splice-altering variant of PJVK gene in a Mauritanian family with non-syndromic hearing impairment. J Appl Genet 2024:10.1007/s13353-024-00903-x. [PMID: 39230647 DOI: 10.1007/s13353-024-00903-x] [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: 02/14/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
PJVK gene was recently shown to create hypervulnerability to sound in humans and was the first human gene implicated in non-syndromic hearing impairment due to neural defect. Targeted next-generation sequencing of over 150 known deafness genes was performed in the proband. Sanger sequencing was used to validate the PJVK variant and confirm familial segregation of the disease. A minigene-based assay has been performed to assess the impact of the variant on splicing. We identified a novel c.550-6A > G acceptor splice-site variant in the PJVK gene in the homozygous state in a Mauritanian child with severe to profound congenital deafness. The substitution was located in intron 4. The effect of the variation was demonstrated by a minigene assay which showed that the variation, an insertion of an additional 5 bp, created a new splice site resulting in the appearance of a premature stop codon (p.Phe184Tyrfs*26) and likely a truncated protein. This result constitutes a new splice-site variant report in the PJVK gene leading to DFNB59 type associated with autosomal recessive non-syndromic hearing impairment (ARNSHI).
Collapse
Affiliation(s)
- Malak Salame
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania.
- Institut d'Hépato-Virologie, Nouakchott, Mauritania.
| | - Crystel Bonnet
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
| | | | - Selma Mohamed Brahim
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
- Centre National d'Oncologie (CNO), Unité de Recherche Et d'Enseignement, Nouakchott, Mauritania
| | - Solene Roux
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
| | - Ely Cheikh Boussaty
- Division of Otolaryngology, Department of Surgery, University of California, 9500 Gilman Drive, Mail Code 0666, La Jolla, San Diego, CA, 92093, USA
| | - Mouna Hadrami
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | | | - Abdellahi M'hamed Sidi
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | - Fatimetou Veten
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| | - Christine Petit
- Institut de L'Audition, Institut Pasteur, Inserm, Paris, France
- Collège de France, Paris, France
| | - Ahmed Houmeida
- Unité de Recherche Sur Les Biomarqueurs Dans La Population Mauritanienne, UN-FST, Nouakchott, Mauritania
| |
Collapse
|
10
|
He C, Gai H, Zhao W, Zhang H, Lai L, Ding C, Chen L, Ding J. Advances in the Study of Etiology and Molecular Mechanisms of Sensorineural Hearing Loss. Cell Biochem Biophys 2024; 82:1721-1734. [PMID: 38849694 DOI: 10.1007/s12013-024-01344-3] [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] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Sensorineural hearing loss (SNHL), a multifactorial progressive disorder, results from a complex interplay of genetic and environmental factors, with its underlying mechanisms remaining unclear. Several pathological factors are believed to contribute to SNHL, including genetic factors, ion homeostasis, cell apoptosis, immune inflammatory responses, oxidative stress, hormones, metabolic syndrome, human cytomegalovirus infection, mitochondrial damage, and impaired autophagy. These factors collectively interact and play significant roles in the onset and progression of SNHL. The present review offers a comprehensive overview of the various factors that contribute to SNHL, emphasizes recent developments in understanding its etiology, and explores relevant preventive and intervention measures.
Collapse
Affiliation(s)
- Cairong He
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Hongcun Gai
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Wen Zhao
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Haiqin Zhang
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Lin Lai
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Chenyu Ding
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Lin Chen
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Jie Ding
- Key laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
11
|
Chen KW, Broz P. Gasdermins as evolutionarily conserved executors of inflammation and cell death. Nat Cell Biol 2024; 26:1394-1406. [PMID: 39187689 DOI: 10.1038/s41556-024-01474-z] [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: 02/01/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024]
Abstract
The gasdermins are a family of pore-forming proteins that have recently emerged as executors of pyroptosis, a lytic form of cell death that is induced by the innate immune system to eradicate infected or malignant cells. Mammalian gasdermins comprise a cytotoxic N-terminal domain, a flexible linker and a C-terminal repressor domain. Proteolytic cleavage in the linker releases the cytotoxic domain, thereby allowing it to form β-barrel membrane pores. Formation of gasdermin pores in the plasma membrane eventually leads to a loss of the electrochemical gradient, cell death and membrane rupture. Here we review recent work that has expanded our understanding of gasdermin biology and function in mammals by revealing their activation mechanism, their regulation and their roles in autoimmunity, host defence and cancer. We further highlight fungal and bacterial gasdermin pore formation pointing to a conserved mechanism of cell death induction.
Collapse
Affiliation(s)
- Kaiwen W Chen
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
12
|
Jiao Y, Li W, Yang W, Wang M, Xing Y, Wang S. Icaritin Exerts Anti-Cancer Effects through Modulating Pyroptosis and Immune Activities in Hepatocellular Carcinoma. Biomedicines 2024; 12:1917. [PMID: 39200381 PMCID: PMC11351763 DOI: 10.3390/biomedicines12081917] [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: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 09/02/2024] Open
Abstract
Icaritin (ICT), a natural compound extracted from the dried leaves of the genus Epimedium, possesses antitumor and immunomodulatory properties. However, the mechanisms through which ICT modulates pyroptosis and immune response in hepatocellular carcinoma (HCC) remain unclear. This study demonstrated that ICT exhibits pyroptosis-inducing and anti-hepatocarcinoma effects. Specifically, the caspase1-GSDMD and caspase3-GSDME pathways were found to be involved in ICT-triggered pyroptosis. Furthermore, ICT promoted pyroptosis in co-cultivation of HepG2 cells and macrophages, regulating the release of inflammatory cytokines and the transformation of macrophages into a proinflammatory phenotype. In the Hepa1-6+Luc liver cancer model, ICT treatment significantly increased the expression of cleaved-caspase1, cleaved-caspase3, and granzyme B, modulated cytokine secretion, and stimulated CD8+ T cell infiltration, resulting in a reduction in tumor growth. In conclusion, the findings in this research suggested that ICT may modulate cell pyroptosis in HCC and subsequently regulate the immune microenvironment of the tumor. These observations may expand the understanding of the pharmacological mechanism of ICT, as well as the therapy of liver cancer.
Collapse
Affiliation(s)
- Yuanyuan Jiao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Poyanghu Road, Jinghai District, Tianjin 301617, China;
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Wenqian Li
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Daxue Road, Jinan 250355, China
| | - Wen Yang
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Mingyu Wang
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Yaling Xing
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| | - Shengqi Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Poyanghu Road, Jinghai District, Tianjin 301617, China;
- Bioinformatics Center of AMMS, Taiping Road, Haidian District, Beijing 100850, China; (W.L.); (W.Y.); (M.W.)
| |
Collapse
|
13
|
Long Y, Jia X, Chu L. Insight into the structure, function and the tumor suppression effect of gasdermin E. Biochem Pharmacol 2024; 226:116348. [PMID: 38852642 DOI: 10.1016/j.bcp.2024.116348] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/20/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Gasdermin E (GSDME), which is also known as DFNA5, was first identified as a deafness-related gene that is expressed in cochlear hair cells, and mutation of this gene causes autosomal dominant neurogenic hearing loss. Later studies revealed that GSDME is mostly expressed in the kidney, placenta, muscle and brain cells, but it is expressed at low levels in tumor cells. The GSDME gene encodes the GSDME protein, which is a member of the gasdermin (GSDM) family and has been shown to participate in the induction of apoptosis and pyroptosis. The current literature suggests that Caspase-3 and Granzyme B (Gzm B) can cleave GSDME to generate the active N-terminal fragment (GSDME-NT), which integrates with the cell membrane and forms pores in this membrane to induce pyroptosis. Furthermore, GSDME also forms pores in mitochondrial membranes to release apoptosis factors, such as cytochrome c (Cyt c) and high-temperature requirement protein A2 (HtrA2/Omi), and subsequently activates the intrinsic apoptosis pathway. In recent years, GSDME has been shown to exert tumor-suppressive effects, suggesting that it has potential therapeutic effects on tumors. In this review, we introduce the structure and function of GSDME and the mechanism by which it induces cell death, and we discuss its tumor suppressive effect.
Collapse
Affiliation(s)
- Yuge Long
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang, China
| | - Xiaoyuan Jia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, Zhejiang, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
14
|
Liu X, Lieberman J. Inflammasome-independent pyroptosis. Curr Opin Immunol 2024; 88:102432. [PMID: 38875738 DOI: 10.1016/j.coi.2024.102432] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024]
Abstract
Gasdermins are membrane pore-forming proteins that cause pyroptosis, an inflammatory cell death in which cells burst and release cytokines, chemokines, and other host alarm signals, such as ATP and HMGB1, which recruit and activate immune cells at sites of infection and danger. There are five gasdermins in humans - gasdermins A to E. Pyroptosis was first described in myeloid cells and mucosal epithelia, which express gasdermin D and activate it when cytosolic sensors of invasive infection or tissue damage assemble into large macromolecular structures, called inflammasomes. Inflammasomes recruit and activate inflammatory caspases (caspase 1, 4, 5, and 11), which cut gasdermin D to remove an inhibitory C-terminal domain, allowing the N-terminal domain to bind to membrane acidic lipids and oligomerize into pores. Recent studies have identified inflammasome-independent proteolytic pathways that activate gasdermin D and the other gasdermins. Here, we review inflammasome-independent pyroptosis pathways and what is known about their role in normal physiology and disease.
Collapse
Affiliation(s)
- Xing Liu
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Li Y, Hou Y, Sun Q, Zeng H, Meng F, Tian X, He Q, Shao F, Ding J. Cleavage-independent activation of ancient eukaryotic gasdermins and structural mechanisms. Science 2024; 384:adm9190. [PMID: 38662913 DOI: 10.1126/science.adm9190] [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: 11/13/2023] [Accepted: 03/28/2024] [Indexed: 05/18/2024]
Abstract
Gasdermins (GSDMs) are pore-forming proteins that execute pyroptosis for immune defense. GSDMs are two-domain proteins activated by proteolytic removal of the inhibitory domain. In this work, we report two types of cleavage-independent GSDM activation. First, TrichoGSDM, a pore-forming domain-only protein from the basal metazoan Trichoplax adhaerens, is a disulfides-linked autoinhibited dimer activated by reduction of the disulfides. The cryo-electron microscopy (cryo-EM) structure illustrates the assembly mechanism for the 44-mer TrichoGSDM pore. Second, RCD-1-1 and RCD-1-2, encoded by the polymorphic regulator of cell death-1 (rcd-1) gene in filamentous fungus Neurospora crassa, are also pore-forming domain-only GSDMs. RCD-1-1 and RCD-1-2, when encountering each other, form pores and cause pyroptosis, underlying allorecognition in Neurospora. The cryo-EM structure reveals a pore of 11 RCD-1-1/RCD-1-2 heterodimers and a heterodimerization-triggered pore assembly mechanism. This study shows mechanistic diversities in GSDM activation and indicates versatile functions of GSDMs.
Collapse
Affiliation(s)
- Yueyue Li
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yanjie Hou
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Sun
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Institute of Biological Sciences, Beijing, Beijing 102206, China
| | - Huan Zeng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Institute of Biological Sciences, Beijing, Beijing 102206, China
| | - Fanyi Meng
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiang Tian
- MOA Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qun He
- MOA Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Feng Shao
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Institute of Biological Sciences, Beijing, Beijing 102206, China
- Research Unit of Pyroptosis and Immunity, Chinese Academy of Medical Sciences and National Institute of Biological Sciences, Beijing 102206, China
- Changping Laboratory, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
- New Cornerstone Science Laboratory, Shenzhen 518054, China
| | - Jingjin Ding
- Key Laboratory of Biomacromolecules (CAS), National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- National Institute of Biological Sciences, Beijing, Beijing 102206, China
| |
Collapse
|
16
|
Yeo XY, Kwon S, Rinai KR, Lee S, Jung S, Park R. A Consolidated Understanding of the Contribution of Redox Dysregulation in the Development of Hearing Impairment. Antioxidants (Basel) 2024; 13:598. [PMID: 38790703 PMCID: PMC11118506 DOI: 10.3390/antiox13050598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The etiology of hearing impairment is multifactorial, with contributions from both genetic and environmental factors. Although genetic studies have yielded valuable insights into the development and function of the auditory system, the contribution of gene products and their interaction with alternate environmental factors for the maintenance and development of auditory function requires further elaboration. In this review, we provide an overview of the current knowledge on the role of redox dysregulation as the converging factor between genetic and environmental factor-dependent development of hearing loss, with a focus on understanding the interaction of oxidative stress with the physical components of the peripheral auditory system in auditory disfunction. The potential involvement of molecular factors linked to auditory function in driving redox imbalance is an important promoter of the development of hearing loss over time.
Collapse
Affiliation(s)
- Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Soohyun Kwon
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
- Department of BioNanotechnology, Gachon University, Seongnam 13120, Republic of Korea
| | - Kimberley R. Rinai
- Department of Life Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital and Medical School, Gwangju 61469, Republic of Korea;
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science & Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
17
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Billman ZP, Kovacs SB, Wei B, Kang K, Cissé OH, Miao EA. Caspase-1 activates gasdermin A in non-mammals. eLife 2024; 12:RP92362. [PMID: 38497531 PMCID: PMC10948149 DOI: 10.7554/elife.92362] [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] [Indexed: 03/19/2024] Open
Abstract
Gasdermins oligomerize to form pores in the cell membrane, causing regulated lytic cell death called pyroptosis. Mammals encode five gasdermins that can trigger pyroptosis: GSDMA, B, C, D, and E. Caspase and granzyme proteases cleave the linker regions of and activate GSDMB, C, D, and E, but no endogenous activation pathways are yet known for GSDMA. Here, we perform a comprehensive evolutionary analysis of the gasdermin family. A gene duplication of GSDMA in the common ancestor of caecilian amphibians, reptiles, and birds gave rise to GSDMA-D in mammals. Uniquely in our tree, amphibian, reptile, and bird GSDMA group in a separate clade than mammal GSDMA. Remarkably, GSDMA in numerous bird species contain caspase-1 cleavage sites like YVAD or FASD in the linker. We show that GSDMA from birds, amphibians, and reptiles are all cleaved by caspase-1. Thus, GSDMA was originally cleaved by the host-encoded protease caspase-1. In mammals the caspase-1 cleavage site in GSDMA is disrupted; instead, a new protein, GSDMD, is the target of caspase-1. Mammal caspase-1 uses exosite interactions with the GSDMD C-terminal domain to confer the specificity of this interaction, whereas we show that bird caspase-1 uses a stereotypical tetrapeptide sequence to confer specificity for bird GSDMA. Our results reveal an evolutionarily stable association between caspase-1 and the gasdermin family, albeit a shifting one. Caspase-1 repeatedly changes its target gasdermin over evolutionary time at speciation junctures, initially cleaving GSDME in fish, then GSDMA in amphibians/reptiles/birds, and finally GSDMD in mammals.
Collapse
Affiliation(s)
- Zachary Paul Billman
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Stephen Bela Kovacs
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Bo Wei
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Kidong Kang
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Ousmane H Cissé
- Critical Care Medicine Department, National Institutes of Health Clinical CenterBethesdaUnited States
| | - Edward A Miao
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| |
Collapse
|
19
|
Vu JT, Tavasoli KU, Mandjikian L, Sheedy CJ, Bacal J, Morrissey MA, Richardson CD, Gardner BM. A genome-wide screen links peroxisome regulation with Wnt signaling through RNF146 and tankyrase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578667. [PMID: 38352406 PMCID: PMC10862876 DOI: 10.1101/2024.02.02.578667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Peroxisomes are membrane-bound organelles harboring metabolic enzymes. In humans, peroxisomes are required for normal development, yet the genes regulating peroxisome function remain unclear. We performed a genome-wide CRISPRi screen to identify novel factors involved in peroxisomal homeostasis. We found that inhibition of RNF146, an E3 ligase activated by poly(ADP-ribose), reduced the import of proteins into peroxisomes. RNF146-mediated loss of peroxisome import depended on the stabilization and activity of the poly(ADP-ribose) polymerase tankyrase, which binds the peroxisomal membrane protein PEX14. We propose that RNF146 and tankyrase regulate peroxisome import efficiency by PARsylation of proteins at the peroxisome membrane. Interestingly, we found that the loss of peroxisomes increased tankyrase and RNF146-dependent degradation of non-peroxisomal substrates, including the beta-catenin destruction complex component AXIN1, which was sufficient to alter the amplitude of beta-catenin transcription. Together, these observations not only suggest previously undescribed roles for RNF146 in peroxisomal regulation, but also a novel role in bridging peroxisome function with Wnt/beta-catenin signaling during development.
Collapse
Affiliation(s)
- Jonathan T Vu
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Katherine U Tavasoli
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Lori Mandjikian
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Connor J Sheedy
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Julien Bacal
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Meghan A Morrissey
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Chris D Richardson
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Brooke M Gardner
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
20
|
Raas Q, Wood A, Stevenson TJ, Swartwood S, Liu S, Kannan RM, Kannan S, Bonkowsky JL. Generation and characterization of a zebrafish gain-of-function ACOX1 Mitchell disease model. Front Pediatr 2024; 12:1326886. [PMID: 38357503 PMCID: PMC10864527 DOI: 10.3389/fped.2024.1326886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 02/16/2024] Open
Abstract
Background Mitchell syndrome is a rare, neurodegenerative disease caused by an ACOX1 gain-of-function mutation (c.710A>G; p.N237S), with fewer than 20 reported cases. Affected patients present with leukodystrophy, seizures, and hearing loss. ACOX1 serves as the rate-limiting enzyme in peroxisomal beta-oxidation of very long-chain fatty acids. The N237S substitution has been shown to stabilize the active ACOX1 dimer, resulting in dysregulated enzymatic activity, increased oxidative stress, and glial damage. Mitchell syndrome lacks a vertebrate model, limiting insights into the pathophysiology of ACOX1-driven white matter damage and neuroinflammatory insults. Methods We report a patient presenting with rapidly progressive white matter damage and neurological decline, who was eventually diagnosed with an ACOX1 N237S mutation through whole genome sequencing. We developed a zebrafish model of Mitchell syndrome using transient ubiquitous overexpression of the human ACOX1 N237S variant tagged with GFP. We assayed zebrafish behavior, oligodendrocyte numbers, expression of white matter and inflammatory transcripts, and analysis of peroxisome counts. Results The patient experienced progressive leukodystrophy and died 2 years after presentation. The transgenic zebrafish showed a decreased swimming ability, which was restored with the reactive microglia-targeted antioxidant dendrimer-N-acetyl-cysteine conjugate. The mutants showed no effect on oligodendrocyte counts but did display activation of the integrated stress response (ISR). Using a novel SKL-targeted mCherry reporter, we found that mutants had reduced density of peroxisomes. Conclusions We developed a vertebrate (zebrafish) model of Mitchell syndrome using transient ubiquitous overexpression of the human ACOX1 N237S variant. The transgenic mutants exhibited motor impairment and showed signs of activated ISR, but interestingly, there were no changes in oligodendrocyte counts. However, the mutants exhibited a deficiency in the number of peroxisomes, suggesting a possible shared mechanism with the Zellweger spectrum disorders.
Collapse
Affiliation(s)
- Quentin Raas
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, Paris, France
| | - Austin Wood
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Tamara J. Stevenson
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Shanna Swartwood
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Suzanne Liu
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Rangaramanujam M. Kannan
- Department of Ophthalmology, Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sujatha Kannan
- Department of Ophthalmology, Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joshua L. Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
- Center for Personalized Medicine, Primary Children’s Hospital, Salt Lake City, UT, United States
| |
Collapse
|
21
|
Billman ZP, Kovacs SB, Wei B, Kang K, Cissé OH, Miao EA. Caspase-1 activates gasdermin A in non-mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.28.559989. [PMID: 37987010 PMCID: PMC10659411 DOI: 10.1101/2023.09.28.559989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Gasdermins oligomerize to form pores in the cell membrane, causing regulated lytic cell death called pyroptosis. Mammals encode five gasdermins that can trigger pyroptosis: GSDMA, B, C, D, and E. Caspase and granzyme proteases cleave the linker regions of and activate GSDMB, C, D, and E, but no endogenous activation pathways are yet known for GSDMA. Here, we perform a comprehensive evolutionary analysis of the gasdermin family. A gene duplication of GSDMA in the common ancestor of caecilian amphibians, reptiles and birds gave rise to GSDMA-D in mammals. Uniquely in our tree, amphibian, reptile and bird GSDMA group in a separate clade than mammal GSDMA. Remarkably, GSDMA in numerous bird species contain caspase-1 cleavage sites like YVAD or FASD in the linker. We show that GSDMA from birds, amphibians, and reptiles are all cleaved by caspase-1. Thus, GSDMA was originally cleaved by the host-encoded protease caspase-1. In mammals the caspase-1 cleavage site in GSDMA is disrupted; instead, a new protein, GSDMD, is the target of caspase-1. Mammal caspase-1 uses exosite interactions with the GSDMD C-terminal domain to confer the specificity of this interaction, whereas we show that bird caspase-1 uses a stereotypical tetrapeptide sequence to confer specificity for bird GSDMA. Our results reveal an evolutionarily stable association between caspase-1 and the gasdermin family, albeit a shifting one. Caspase-1 repeatedly changes its target gasdermin over evolutionary time at speciation junctures, initially cleaving GSDME in fish, then GSDMA in amphibians/reptiles/birds, and finally GSDMD in mammals.
Collapse
Affiliation(s)
- Zachary P Billman
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
- Department of Microbiology and Immunology; Chapel Hill, NC, USA
| | - Stephen B Kovacs
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
- Department of Microbiology and Immunology; Chapel Hill, NC, USA
| | - Bo Wei
- Duke University School of Medicine
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| | - Kidong Kang
- Duke University School of Medicine
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| | - Ousmane H Cissé
- National Institutes of Health
- Critical Care Medicine Department; Bethesda, MD, USA
| | - Edward A Miao
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| |
Collapse
|
22
|
Qiu Y, Wang H, Pan H, Ding X, Guan J, Zhuang Q, Wu K, Lei Z, Cai H, Dong Y, Zhou H, Lin A, Wang Q, Yan Q. NADH improves AIF dimerization and inhibits apoptosis in iPSCs-derived neurons from patients with auditory neuropathy spectrum disorder. Hear Res 2024; 441:108919. [PMID: 38043402 DOI: 10.1016/j.heares.2023.108919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
Auditory neuropathy spectrum disorder (ANSD) is a hearing impairment involving disruptions to inner hair cells (IHCs), ribbon synapses, spiral ganglion neurons (SGNs), and/or the auditory nerve itself. The outcomes of cochlear implants (CI) for ANSD are variable and dependent on the location of lesion sites. Discovering a potential therapeutic agent for ANSD remains an urgent requirement. Here, 293T stable transfection cell lines and patient induced pluripotent stem cells (iPSCs)-derived auditory neurons carrying the apoptosis inducing factor (AIF) p.R422Q variant were used to pursue a therapeutic regent for ANSD. Nicotinamide adenine dinucleotide (NADH) is a main electron donor in the electron transport chain (ETC). In 293T stable transfection cells with the p.R422Q variant, NADH treatment improved AIF dimerization, rescued mitochondrial dysfunctions, and decreased cell apoptosis. The effects of NADH were further confirmed in patient iPSCs-derived neurons. The relative level of AIF dimers was increased to 150.7 % (P = 0.026) from 59.2 % in patient-neurons upon NADH treatment. Such increased AIF dimerization promoted the mitochondrial import of coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4), which further restored mitochondrial functions. Similarly, the content of mitochondrial calcium (mCa2+) was downregulated from 136.7 % to 102.3 % (P = 0.0024) in patient-neurons upon NADH treatment. Such decreased mCa2+ levels inhibited calpain activity, ultimately reducing the percentage of apoptotic cells from 30.5 % to 21.1 % (P = 0.021). We also compared the therapeutic effects of gene correction and NADH treatment on hereditary ANSD. NADH treatment had comparable restorative effects on functions of ANSD patient-specific cells to that of gene correction. Our findings offer evidence of the molecular mechanisms of ANSD and introduce NADH as a potential therapeutic agent for ANSD therapy.
Collapse
Affiliation(s)
- Yue Qiu
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; Institute of Brain Science, Wannan Medical College, Wuhu, Anhui 241000, China
| | - Hongyang Wang
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Huaye Pan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xue Ding
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jing Guan
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Qianqian Zhuang
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kaiwen Wu
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Zhaoying Lei
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huajian Cai
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yufei Dong
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hui Zhou
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Aifu Lin
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiuju Wang
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Qingfeng Yan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
23
|
Angosto-Bazarra D, Guijarro A, Pelegrín P. Evolution of the gasdermin family and pyroptosis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105060. [PMID: 37734430 DOI: 10.1016/j.dci.2023.105060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/11/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023]
Abstract
Gasdermins have been identified as playing a prominent role in the innate immune response as the executors of a specific type of cell death called pyroptosis. Specific proteolytic cleavage of gasdermins generates an N-terminal that oligomerizes and forms pores in the cell membrane. Although pyroptosis has been widely described in mammals, the importance of gasdermins and gasdermin-like proteins in inducing cell death in other vertebrates, in invertebrates and in other taxa including fungi and bacteria is still being determined. Mammalian, fungal and bacterial gasdermins have in common the fact that they go through the same stages (such as proteolytic activation) when inducing membrane rupture, which suggests that pyroptosis is as an ancient mechanism. In this review, we summarize the evolution and function of the gasdermin and gasdermin-like proteins in animals, fungi and bacteria.
Collapse
Affiliation(s)
- Diego Angosto-Bazarra
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain.
| | - Adriana Guijarro
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain
| | - Pablo Pelegrín
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, 30120, Murcia, Spain; Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120 Murcia, Spain.
| |
Collapse
|
24
|
Feng Y, Hu S, Zhao S, Chen M. Recent advances in genetic etiology of non-syndromic deafness in children. Front Neurosci 2023; 17:1282663. [PMID: 37928735 PMCID: PMC10620706 DOI: 10.3389/fnins.2023.1282663] [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: 08/24/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
Congenital auditory impairment is a prevalent anomaly observed in approximately 2-3 per 1,000 infants. The consequences associated with hearing loss among children encompass the decline of verbal communication, linguistic skills, educational progress, social integration, cognitive aptitude, and overall well-being. Approaches to reversing or preventing genetic hearing loss are limited. Patients with mild and moderate hearing loss can only use hearing aids, while those with severe hearing loss can only acquire speech and language through cochlear implants. Both environmental and genetic factors contribute to the occurrence of congenital hearing loss, and advancements in our understanding of the pathophysiology and molecular mechanisms underlying hearing loss, coupled with recent progress in genetic testing techniques, will facilitate the development of innovative approaches for treatment and screening. In this paper, the latest research progress in genetic etiology of non-syndromic deafness in children with the highest incidence is summarized in order to provide help for personalized diagnosis and treatment of deafness in children.
Collapse
|
25
|
Weindel CG, Ellzey LM, Martinez EL, Watson RO, Patrick KL. Gasdermins gone wild: new roles for GSDMs in regulating cellular homeostasis. Trends Cell Biol 2023; 33:773-787. [PMID: 37062616 PMCID: PMC10611448 DOI: 10.1016/j.tcb.2023.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/13/2023] [Accepted: 02/24/2023] [Indexed: 04/18/2023]
Abstract
Since their discovery, members of the gasdermin (GSDM) family of proteins have been firmly established as executors of pyroptosis, with the N-terminal fragment of most GSDMs capable of forming pores in the plasma membrane. More recent findings suggest that some GSDMs can drive additional cell death pathways, such as apoptosis and necroptosis, through mechanisms independent of plasma membrane perforation. There is also emerging evidence that by associating with cellular compartments such as mitochondria, peroxisomes, endosomes, and the nucleus, GSDMs regulate cell death-independent aspects of cellular homeostasis. Here, we review the diversity of GSDM function across several cell types and explore how various cellular stresses can promote relocalization - and thus refunctionalization - of GSDMs.
Collapse
Affiliation(s)
- Chi G Weindel
- Texas A&M University School of Medicine, Bryan, TX, USA
| | - Lily M Ellzey
- Texas A&M University School of Medicine, Bryan, TX, USA
| | | | | | | |
Collapse
|
26
|
Huston HC, Anderson MJ, Fink SL. Pyroptosis and the cellular consequences of gasdermin pores. Semin Immunol 2023; 69:101803. [PMID: 37437353 PMCID: PMC10530493 DOI: 10.1016/j.smim.2023.101803] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
The family of gasdermin proteins plays a key role in the host response against external and internal pathogenic signals by mediating the form of inflammatory regulated cell death known as pyroptosis. One of the most well-studied gasdermins within innate immunity is gasdermin D, which is cleaved, oligomerizes, and forms plasma membrane pores. Gasdermin D pores lead to a number of downstream cellular consequences including plasma membrane rupture, or cell lysis. In this review, we describe mechanisms of activation for each of the gasdermins, their cell type specificity and some disease associations. We then discuss downstream consequences of gasdermin pore formation, including cellular mechanisms of membrane repair. Finally, we present some important next steps to better understand pyroptosis and the cellular consequences of gasdermin pore formation.
Collapse
Affiliation(s)
- Hanna C Huston
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Marisa J Anderson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
27
|
Abstract
Orchestration of protein production and degradation and the regulation of protein lifetimes play a central role in many basic biological processes. Nearly all mammalian proteins are replenished by protein turnover in waves of synthesis and degradation. Protein lifetimes in vivo are typically measured in days, but a small number of extremely long-lived proteins (ELLPs) persist for months or even years. ELLPs are rare in all tissues but are enriched in tissues containing terminally differentiated post-mitotic cells and extracellular matrix. Consistently, emerging evidence suggests that the cochlea may be particularly enriched in ELLPs. Damage to ELLPs in specialized cell types, such as crystallin in the lens cells of the eye, causes organ failure such as cataracts. Similarly, damage to cochlear ELLPs is likely to occur with many insults, including acoustic overstimulation, drugs, anoxia, and antibiotics, and may play an underappreciated role in hearing loss. Furthermore, hampered protein degradation may contribute to acquired hearing loss. In this review, I highlight our knowledge of the lifetimes of cochlear proteins with an emphasis on ELLPs and the potential contribution that impaired cochlear protein degradation has on acquired hearing loss and the emerging relevance of ELLPs.
Collapse
Affiliation(s)
- Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
28
|
Allali-Boumara I, Marrero AD, Quesada AR, Martínez-Poveda B, Medina MÁ. Pyroptosis Modulators: New Insights of Gasdermins in Health and Disease. Antioxidants (Basel) 2023; 12:1551. [PMID: 37627547 PMCID: PMC10451529 DOI: 10.3390/antiox12081551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Pyroptosis is an inflammation-dependent type of cell death that has been in the spotlight for the scientific community in the last few years. Crucial players in the process of pyroptosis are the members of the gasdermin family of proteins, which have been parallelly studied. Upon induction of pyroptosis, gasdermins suffer from structural changes leading to the formation of pores in the membrane that subsequently cause the release of pro-inflammatory contents. Recently, it has been discovered that oxidation plays a key role in the activation of certain gasdermins. Here, we review the current knowledge on pyroptosis and human gasdermins, focusing on the description of the different members of the family, their molecular structures, and their influence on health and disease directly or non-directly related to inflammation. Noteworthy, we have focused on the existing understanding of the role of this family of proteins in cancer, which could translate into novel promising strategies aimed at benefiting human health. In conclusion, the modulation of pyroptosis and gasdermins by natural and synthetic compounds through different mechanisms, including modification of the redox state of cells, has been proven effective and sets precedents for future therapeutic strategies.
Collapse
Affiliation(s)
- Imane Allali-Boumara
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.A.-B.); (A.D.M.); (A.R.Q.); (B.M.-P.)
| | - Ana Dácil Marrero
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.A.-B.); (A.D.M.); (A.R.Q.); (B.M.-P.)
- Instituto de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Ana R. Quesada
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.A.-B.); (A.D.M.); (A.R.Q.); (B.M.-P.)
- Instituto de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Beatriz Martínez-Poveda
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.A.-B.); (A.D.M.); (A.R.Q.); (B.M.-P.)
- Instituto de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Miguel Ángel Medina
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.A.-B.); (A.D.M.); (A.R.Q.); (B.M.-P.)
- Instituto de Investigación Biomédica y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
29
|
Privitera G, Rana N, Armuzzi A, Pizarro TT. The gasdermin protein family: emerging roles in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol 2023; 20:366-387. [PMID: 36781958 PMCID: PMC10238632 DOI: 10.1038/s41575-023-00743-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/15/2023]
Abstract
Since the identification and characterization of gasdermin (GSDM) D as the main effector of inflammatory regulated cell death (or pyroptosis), literature on the GSDM family of pore-forming proteins is rapidly expanding, revealing novel mechanisms regulating their expression and functions that go beyond pyroptosis. Indeed, a growing body of evidence corroborates the importance of GSDMs within the gastrointestinal system, underscoring their critical contributions to the pathophysiology of gastrointestinal cancers, enteric infections and gut mucosal inflammation, such as inflammatory bowel disease. However, with this increase in knowledge, several important and controversial issues have arisen regarding basic GSDM biology and its role(s) during health and disease states. These include critical questions centred around GSDM-dependent lytic versus non-lytic functions, the biological activities of cleaved versus full-length proteins, the differential roles of GSDM-expressing mucosal immune versus epithelial cells, and whether GSDMs promote pathogenic or protective effects during specific disease settings. This Review provides a comprehensive summary and interpretation of the current literature on GSDM biology, specifically focusing on the gastrointestinal tract, highlighting the main controversial issues and their clinical implications, and addressing future areas of research to unravel the specific role(s) of this intriguing, yet enigmatic, family of proteins.
Collapse
Affiliation(s)
- Giuseppe Privitera
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Nitish Rana
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
30
|
Li H, Lismont C, Costa CF, Hussein MAF, Baes M, Fransen M. Enhanced Levels of Peroxisome-Derived H2O2 Do Not Induce Pexophagy but Impair Autophagic Flux in HEK-293 and HeLa Cells. Antioxidants (Basel) 2023; 12:antiox12030613. [PMID: 36978861 PMCID: PMC10045779 DOI: 10.3390/antiox12030613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Peroxisomes are functionally specialized organelles that harbor multiple hydrogen peroxide (H2O2)-producing and -degrading enzymes. Given that this oxidant functions as a major redox signaling agent, peroxisomes have the intrinsic ability to mediate and modulate H2O2-driven processes, including autophagy. However, it remains unclear whether changes in peroxisomal H2O2 (po-H2O2) emission impact the autophagic process and to which extent peroxisomes with a disturbed H2O2 metabolism are selectively eliminated through a process called “pexophagy”. To address these issues, we generated and validated HEK-293 and HeLa pexophagy reporter cell lines in which the production of po-H2O2 can be modulated. We demonstrate that (i) po-H2O2 can oxidatively modify multiple selective autophagy receptors and core autophagy proteins, (ii) neither modest nor robust levels of po-H2O2 emission act as a prime determinant of pexophagy, and (iii) high levels of po-H2O2 impair autophagic flux by oxidative inhibition of enzymes involved in LC3II formation. Unexpectedly, our analyses also revealed that the autophagy receptor optineurin can be recruited to peroxisomes, thereby triggering pexophagy. In summary, these findings lend support to the idea that, during cellular and organismal aging, peroxisomes with enhanced H2O2 release can escape pexophagy and downregulate autophagic activity, thereby perpetuating the accumulation of damaged and toxic cellular debris.
Collapse
Affiliation(s)
- Hongli Li
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Celien Lismont
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Cláudio F. Costa
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Mohamed A. F. Hussein
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Asyut 71515, Egypt
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Marc Fransen
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-330114
| |
Collapse
|
31
|
Abstract
Pyroptosis is a form of regulated cell death that is mediated by the membrane-targeting, pore-forming gasdermin family of proteins. Pyroptosis was initially described as a caspase 1- and inflammasome-dependent cell death pathway typified by the loss of membrane integrity and the secretion of cytokines such as IL-1β. However, gasdermins are now recognized as the principal effectors of this form of regulated cell death; activated gasdermins insert into cell membranes, where they form pores that result in the secretion of cytokines, alarmins and damage-associated molecular patterns and cause cell membrane rupture. It is now evident that gasdermins can be activated by inflammasome- and caspase-independent mechanisms in multiple cell types and that crosstalk occurs between pyroptosis and other cell death pathways. Although they are important for host antimicrobial defence, a growing body of evidence supports the notion that pyroptosis and gasdermins have pathological roles in cancer and several non-microbial diseases involving the gut, liver and skin. The well-documented roles of inflammasome activity and apoptosis pathways in kidney diseases suggests that gasdermins and pyroptosis may also be involved to some extent. However, despite some evidence for involvement of pyroptosis in the context of acute kidney injury and chronic kidney disease, our understanding of gasdermin biology and pyroptosis in the kidney remains limited.
Collapse
|
32
|
Ivanov AI, Rana N, Privitera G, Pizarro TT. The enigmatic roles of epithelial gasdermin B: Recent discoveries and controversies. Trends Cell Biol 2023; 33:48-59. [PMID: 35821185 PMCID: PMC9789163 DOI: 10.1016/j.tcb.2022.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 01/06/2023]
Abstract
Gasdermin B (GSDMB) belongs to a family of structurally related proteins [(i.e., gasdermins (GSDMs)]. It distinguishes itself from other members by the lack of autoinhibition but clear bioactivity of its full-length form, its preference to bind to phosphatidylinositol phosphates and sulfatides, and the ability to promote both lytic and nonlytic cellular functions. It is the only gasdermin that lacks a mouse ortholog, making in vivo mechanistic studies challenging to perform. GSDMB is abundantly expressed in epithelial cells lining organs that directly interface with the external environment, such as the gastrointestinal tract, with emerging evidence supporting its role in enteric infections, inflammatory bowel disease (IBD), and colorectal cancer. This review discusses the unique features of GSDMB among other gasdermin family members and controversies surrounding GSDMB-dependent mammalian inflammatory cell death (i.e., pyroptosis), including recent discoveries revealing both lytic and nonlytic functions of epithelial-derived GSDMB, particularly during gut health and disease.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Inflammation and Immunity, Learner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Nitish Rana
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giuseppe Privitera
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
33
|
Li Y, Li S, Wu L, Wu T, Li M, Du D, Chen Y, Wang C, Li X, Zhang S, Zhao Z, Zheng L, Chen M, Li M, Li T, Shi X, Qiao Y. Sestrin 2 Deficiency Exacerbates Noise-Induced Cochlear Injury Through Inhibiting ULK1/Parkin-Mediated Mitophagy. Antioxid Redox Signal 2023; 38:115-136. [PMID: 35708118 PMCID: PMC9885551 DOI: 10.1089/ars.2021.0283] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/03/2023]
Abstract
Aims: Noise damage to auditory hair cells is associated with oxidative stress and mitochondrial dysfunction. This study aimed to investigate the possible effect of sestrin 2 (SESN2), an endogenous antioxidant protein, on noise-induced hearing loss (NIHL) and the underlying mechanisms. Results: We identified SESN2 as a protective factor against oxidative stress in NIHL through activation of Parkin-mediated mitophagy. Consistently, SESN2 expression was increased and mitophagy was induced during the early stage after a temporary threshold shift due to noise exposure or hydrogen peroxide(H2O2) stimulation; conversely, SESN2 deficiency blocked mitophagy and exacerbated acoustic trauma. Mechanistically, SESN2 interacted with Unc-51-like protein kinase 1(ULK1), promoting ULK1 protein-level stabilization by interfering with its proteasomal degradation. This stabilization is essential for mitophagy initiation, since restoring ULK1 expression in SESN2-silenced cells rescued mitophagy defects. Innovation and Conclusion: Our results provide novel insights regarding SESN2 as a therapeutic target against noise-induced cochlear injury, possibly through improved mitophagy. Antioxid. Redox Signal. 38, 115-136.
Collapse
Affiliation(s)
- Yalan Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Shengsheng Li
- Department of Neurosurgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, P.R. China
| | - Liyuan Wu
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Tingting Wu
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Mengxiao Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Deliang Du
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Yalin Chen
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Caiji Wang
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Xuanyi Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| | - Shili Zhang
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Zeqi Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| | - Liting Zheng
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Mengbing Chen
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Menghua Li
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Ting Li
- School of Life Sciences, Xuzhou Medical University, Xuzhou, P.R. China
| | - Xi Shi
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
| | - Yuehua Qiao
- Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, P.R. China
- The Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, P.R. China
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, P.R. China
| |
Collapse
|
34
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Li P, Li S, Wang L, Li H, Wang Y, Liu H, Wang X, Zhu X, Liu Z, Ye F, Zhang Y. Mitochondrial dysfunction in hearing loss: Oxidative stress, autophagy and NLRP3 inflammasome. Front Cell Dev Biol 2023; 11:1119773. [PMID: 36891515 PMCID: PMC9986271 DOI: 10.3389/fcell.2023.1119773] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Sensorineural deafness becomes an inevitable worldwide healthy problem, yet the current curative therapy is limited. Emerging evidences demonstrate mitochondrial dysfunction plays a vital role of in the pathogenesis of deafness. Reactive oxygen species (ROS)-induced mitochondrial dysfunction combined with NLRP3 inflammasome activation is involved in cochlear damage. Autophagy not only clears up undesired proteins and damaged mitochondria (mitophagy), but also eliminate excessive ROS. Appropriate enhancement of autophagy can reduce oxidative stress, inhibit cell apoptosis, and protect auditory cells. In addition, we further discuss the interplays linking ROS generation, NLRP3 inflammasome activation, and autophagy underlying the pathogenesis of deafness, including ototoxic drugs-, noise- and aging-related hearing loss.
Collapse
Affiliation(s)
- Peipei Li
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Research Center for Kidney Disease, Zhengzhou, China
| | - Shen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Le Wang
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongmin Li
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Wang
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongbing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaodan Zhu
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Province Research Center for Kidney Disease, Zhengzhou, China
| | - Fanglei Ye
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Zhang
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Pore-forming proteins as drivers of membrane permeabilization in cell death pathways. Nat Rev Mol Cell Biol 2022; 24:312-333. [PMID: 36543934 DOI: 10.1038/s41580-022-00564-w] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 12/24/2022]
Abstract
Regulated cell death (RCD) relies on activation and recruitment of pore-forming proteins (PFPs) that function as executioners of specific cell death pathways: apoptosis regulator BAX (BAX), BCL-2 homologous antagonist/killer (BAK) and BCL-2-related ovarian killer protein (BOK) for apoptosis, gasdermins (GSDMs) for pyroptosis and mixed lineage kinase domain-like protein (MLKL) for necroptosis. Inactive precursors of PFPs are converted into pore-forming entities through activation, membrane recruitment, membrane insertion and oligomerization. These mechanisms involve protein-protein and protein-lipid interactions, proteolytic processing and phosphorylation. In this Review, we discuss the structural rearrangements incurred by RCD-related PFPs and describe the mechanisms that manifest conversion from autoinhibited to membrane-embedded molecular states. We further discuss the formation and maturation of membrane pores formed by BAX/BAK/BOK, GSDMs and MLKL, leading to diverse pore architectures. Lastly, we highlight commonalities and differences of PFP mechanisms involving BAX/BAK/BOK, GSDMs and MLKL and conclude with a discussion on how, in a population of challenged cells, the coexistence of cell death modalities may have profound physiological and pathophysiological implications.
Collapse
|
37
|
Sun Y, Zou S, He Z, Chen X. The role of autophagy and ferroptosis in sensorineural hearing loss. Front Neurosci 2022; 16:1068611. [PMID: 36578828 PMCID: PMC9791179 DOI: 10.3389/fnins.2022.1068611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Hearing loss has become a common sensory defect in humans. Because of the limited regenerative ability of mammalian cochlear hair cells (HCs), HC damage (caused by ototoxic drugs, aging, and noise) is the main risk factor of hearing loss. However, how HCs can be protected from these risk factors remains to be investigated. Autophagy is a process by which damaged cytoplasmic components are sequestered into lysosomes for degradation. Ferroptosis is a novel form of non-apoptotic regulated cell death involving intracellular iron overloading and iron-dependent lipid peroxide accumulation. Recent studies have confirmed that autophagy is associated with ferroptosis, and their crosstalk may be the potential therapeutic target for hearing loss. In this review, we provide an overview of the mechanisms of ferroptosis and autophagy as well as their relationship with HC damage, which may provide insights for a new future in the protection of HCs.
Collapse
|
38
|
Mauriac SA, Peineau T, Zuberi A, Lutz C, Géléoc GSG. Loss of Pex1 in Inner Ear Hair Cells Contributes to Cochlear Synaptopathy and Hearing Loss. Cells 2022; 11:cells11243982. [PMID: 36552747 PMCID: PMC9777190 DOI: 10.3390/cells11243982] [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: 11/13/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Peroxisome Biogenesis Disorders (PBD) and Zellweger syndrome spectrum disorders (ZSD) are rare genetic multisystem disorders that include hearing impairment and are associated with defects in peroxisome assembly, function, or both. Mutations in 13 peroxin (PEX) genes have been found to cause PBD-ZSD with ~70% of patients harboring mutations in PEX1. Limited research has focused on the impact of peroxisomal disorders on auditory function. As sensory hair cells are particularly vulnerable to metabolic changes, we hypothesize that mutations in PEX1 lead to oxidative stress affecting hair cells of the inner ear, subsequently resulting in hair cell degeneration and hearing loss. Global deletion of the Pex1 gene is neonatal lethal in mice, impairing any postnatal studies. To overcome this limitation, we created conditional knockout mice (cKO) using Gfi1Creor VGlut3Cre expressing mice crossed to floxed Pex1 mice to allow for selective deletion of Pex1 in the hair cells of the inner ear. We find that Pex1 excision in inner hair cells (IHCs) leads to progressive hearing loss associated with significant decrease in auditory brainstem responses (ABR), specifically ABR wave I amplitude, indicative of synaptic defects. Analysis of IHC synapses in cKO mice reveals a decrease in ribbon synapse volume and functional alterations in exocytosis. Concomitantly, we observe a decrease in peroxisomal number, indicative of oxidative stress imbalance. Taken together, these results suggest a critical function of Pex1 in development and maturation of IHC-spiral ganglion synapses and auditory function.
Collapse
Affiliation(s)
- Stephanie A. Mauriac
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA 02115, USA
- Kirby Neurobiology Center, Harvard Medical School, Boston, MA 02115, USA
| | - Thibault Peineau
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA 02115, USA
- Kirby Neurobiology Center, Harvard Medical School, Boston, MA 02115, USA
| | - Aamir Zuberi
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Technology Evaluation and Development Research Laboratory, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Cathleen Lutz
- Rare Disease Translational Center, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Gwénaëlle S. G. Géléoc
- Department of Otolaryngology, Boston Children’s Hospital, Boston, MA 02115, USA
- Kirby Neurobiology Center, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: ; Tel.: +1-617-919-4061
| |
Collapse
|
39
|
Kochaj RM, Martelletti E, Ingham NJ, Buniello A, Sousa BC, Wakelam MJO, Lopez-Clavijo AF, Steel KP. The Effect of a Pex3 Mutation on Hearing and Lipid Content of the Inner Ear. Cells 2022; 11:cells11203206. [PMID: 36291074 PMCID: PMC9600510 DOI: 10.3390/cells11203206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Peroxisome biogenesis disorders (due to PEX gene mutations) are associated with symptoms that range in severity and can lead to early childhood death, but a common feature is hearing impairment. In this study, mice carrying Pex3 mutations were found to show normal auditory development followed by an early-onset progressive increase in auditory response thresholds. The only structural defect detected in the cochlea at four weeks old was the disruption of synapses below inner hair cells. A conditional approach was used to establish that Pex3 expression is required locally within the cochlea for normal hearing, rather than hearing loss being due to systemic effects. A lipidomics analysis of the inner ear revealed a local reduction in plasmalogens in the Pex3 mouse mutants, comparable to the systemic plasmalogen reduction reported in human peroxisome biogenesis disorders. Thus, mice with Pex3 mutations may be a useful tool to understand the physiological basis of peroxisome biogenesis disorders.
Collapse
Affiliation(s)
- Rafael M. Kochaj
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Elisa Martelletti
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Neil J. Ingham
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Annalisa Buniello
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Bebiana C. Sousa
- Lipidomics Facility, The BBSRC Babraham Institute, Cambridge CB22 3AT, UK
| | | | | | - Karen P. Steel
- Wolfson Centre for Age-Related Diseases, King’s College London, Guy’s Campus, London SE1 1UL, UK
- Correspondence:
| |
Collapse
|
40
|
Wan H, Zhang Y, Hua Q. Cellular autophagy, the compelling roles in hearing function and dysfunction. Front Cell Neurosci 2022; 16:966202. [PMID: 36246522 PMCID: PMC9561951 DOI: 10.3389/fncel.2022.966202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
Abstract
Sensorineural hearing loss (SNHL) is currently a major health issue. As one of the most common neurodegenerative diseases, SNHL is associated with the degradation of hair cells (HCs), spiral ganglion neurons (SGNs), the stria vascularis, supporting cells and central auditory system cells. Autophagy is a highly integrated cellular system that eliminates impaired components and replenishes energy to benefit cellular homeostasis. Etiological links between autophagy alterations and neurodegenerative diseases, such as SNHL, have been established. The hearing pathway is complex and depends on the comprehensive functions of many types of tissues and cells in auditory system. In this review, we discuss the roles of autophagy in promoting and inhibiting hearing, paying particular attention to specific cells in the auditory system, as discerned through research. Hence, our review provides enlightening ideas for the role of autophagy in hearing development and impairment.
Collapse
Affiliation(s)
- Huanzhi Wan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyuan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yuanyuan Zhang,
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Qingquan Hua,
| |
Collapse
|
41
|
Gámez-Chiachio M, Molina-Crespo Á, Ramos-Nebot C, Martinez-Val J, Martinez L, Gassner K, Llobet FJ, Soriano M, Hernandez A, Cordani M, Bernadó-Morales C, Diaz E, Rojo-Sebastian A, Triviño JC, Sanchez L, Rodríguez-Barrueco R, Arribas J, Llobet-Navás D, Sarrió D, Moreno-Bueno G. Gasdermin B over-expression modulates HER2-targeted therapy resistance by inducing protective autophagy through Rab7 activation. J Exp Clin Cancer Res 2022; 41:285. [PMID: 36163066 PMCID: PMC9511784 DOI: 10.1186/s13046-022-02497-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/19/2022] [Indexed: 11/10/2022] Open
Abstract
Background Gasdermin B (GSDMB) over-expression promotes poor prognosis and aggressive behavior in HER2 breast cancer by increasing resistance to therapy. Decoding the molecular mechanism of GSDMB-mediated drug resistance is crucial to identify novel effective targeted treatments for HER2/GSDMB aggressive tumors. Methods Different in vitro approaches (immunoblot, qRT-PCR, flow cytometry, proteomic analysis, immunoprecipitation, and confocal/electron microscopy) were performed in HER2 breast and gastroesophageal carcinoma cell models. Results were then validated using in vivo preclinical animal models and analyzing human breast and gastric cancer samples. Results GSDMB up-regulation renders HER2 cancer cells more resistant to anti-HER2 agents by promoting protective autophagy. Accordingly, the combination of lapatinib with the autophagy inhibitor chloroquine increases the therapeutic response of GSDMB-positive cancers in vitro and in zebrafish and mice tumor xenograft in vivo models. Mechanistically, GSDMB N-terminal domain interacts with the key components of the autophagy machinery LC3B and Rab7, facilitating the Rab7 activation during pro-survival autophagy in response to anti-HER2 therapies. Finally, we validated these results in clinical samples where GSDMB/Rab7/LC3B co-expression associates significantly with relapse in HER2 breast and gastric cancers. Conclusion Our findings uncover for the first time a functional link between GSDMB over-expression and protective autophagy in response to HER2-targeted therapies. GSDMB behaves like an autophagy adaptor and plays a pivotal role in modulating autophagosome maturation through Rab7 activation. Finally, our results provide a new and accessible therapeutic approach for HER2/GSDMB + cancers with adverse clinical outcome. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02497-w.
Collapse
|
42
|
Yuan Z, Jiang S, Qin K, Sun L. New insights into the evolutionary dynamic and lineage divergence of gasdermin E in metazoa. Front Cell Dev Biol 2022; 10:952015. [PMID: 35938154 PMCID: PMC9355259 DOI: 10.3389/fcell.2022.952015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Gasdermin (GSDM) is a family of pore-forming proteins that induce pyroptosis. To date, the origin and evolution of GSDM in Metazoa remain elusive. Here, we found that GSDM emerged early in Placozoa but is absent in a large number of invertebrates. In the lower vertebrate, fish, three types of GSDME, i.e., GSDMEa, GSDMEb, and a previously unreported type (designated GSDMEc), were idenitied. Evolutionarily, the three GSDMEs are distinctly separated: GSDMEa is closely related to tetrapod GSDME; GSDMEb exists exclusively in fish; GSDMEc forms the lineage root of tetrapod GSDMA/B/C/D. GSDMEc shares conserved genomic features with and is probably the prototype of GSDMA, which we found existing in all tetrapod classes. GSDMEc displays fast evolutionary dynamics, likely as a result of genomic transposition. A cross-metazoan analysis of GSDME revealed that GSDMEa shares a conserved caspase recognition motif with the GSDME of tetrapods and cnidarians, whereas GSDMEb has a unique caspase recognition motif similar to that of mammalian GSDMD, and GSDMEc exhibits no apparent caspase recognition motif. Through functional test, four highly conserved residues in vertebrate GSDME proved to be essential to auto-inhibition. Together our results provide new insights into the origin, evolution, and function of metazoan GSDMs.
Collapse
Affiliation(s)
- Zihao Yuan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shuai Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- *Correspondence: Shuai Jiang, ; Li Sun,
| | - Kunpeng Qin
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Shuai Jiang, ; Li Sun,
| |
Collapse
|
43
|
Magnani L, Colantuoni M, Mortellaro A. Gasdermins: New Therapeutic Targets in Host Defense, Inflammatory Diseases, and Cancer. Front Immunol 2022; 13:898298. [PMID: 35844522 PMCID: PMC9285118 DOI: 10.3389/fimmu.2022.898298] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022] Open
Abstract
Gasdermins (GSDMs) are a class of pore-forming proteins related to pyroptosis, a programmed cell death pathway that is induced by a range of inflammatory stimuli. Small-scale GSDM activation and pore formation allow the passive release of cytokines, such as IL-1β and IL-18, and alarmins, but, whenever numerous GSDM pores are assembled, osmotic lysis and cell death occur. Such GSDM-mediated pyroptosis promotes pathogen clearance and can help restore homeostasis, but recent studies have revealed that dysregulated pyroptosis is at the root of many inflammation-mediated disease conditions. Moreover, new homeostatic functions for gasdermins are beginning to be revealed. Here, we review the newly discovered mechanisms of GSDM activation and their prominent roles in host defense and human diseases associated with chronic inflammation. We also highlight the potential of targeting GSDMs as a new therapeutic approach to combat chronic inflammatory diseases and cancer and how we might overcome the current obstacles to realize this potential.
Collapse
|
44
|
Luong AM, Koestel J, Bhati KK, Batoko H. Cargo receptors and adaptors for selective autophagy in plant cells. FEBS Lett 2022; 596:2104-2132. [PMID: 35638898 DOI: 10.1002/1873-3468.14412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/08/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Abstract
Plant selective (macro)autophagy is a highly regulated process whereby eukaryotic cells spatiotemporally degrade some of their constituents that have become superfluous or harmful. The identification and characterization of the factors determining this selectivity make it possible to integrate selective (macro)autophagy into plant cell physiology and homeostasis. The specific cargo receptors and/or scaffold proteins involved in this pathway are generally not structurally conserved, as are the biochemical mechanisms underlying recognition and integration of a given cargo into the autophagosome in different cell types. This review discusses the few specific cargo receptors described in plant cells to highlight key features of selective autophagy in the plant kingdom and its integration with plant physiology, so as to identify evolutionary convergence and knowledge gaps to be filled by future research.
Collapse
Affiliation(s)
- Ai My Luong
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Jérôme Koestel
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Kaushal Kumar Bhati
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| | - Henri Batoko
- Louvain Institute of Biomolecular Science and Technology, University of Louvain Croix du Sud 4, L7.07.14, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
45
|
Chen X, Tian PC, Wang K, Wang M, Wang K. Pyroptosis: Role and Mechanisms in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:897815. [PMID: 35647057 PMCID: PMC9130572 DOI: 10.3389/fcvm.2022.897815] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/04/2022] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular disease (CVD) is a common disease that poses a huge threat to human health. Irreversible cardiac damage due to cardiomyocyte death and lack of regenerative capacity under stressful conditions, ultimately leading to impaired cardiac function, is the leading cause of death worldwide. The regulation of cardiomyocyte death plays a crucial role in CVD. Previous studies have shown that the modes of cardiomyocyte death include apoptosis and necrosis. However, another new form of death, pyroptosis, plays an important role in CVD pathogenesis. Pyroptosis induces the amplification of inflammatory response, increases myocardial infarct size, and accelerates the occurrence of cardiovascular disease, and the control of cardiomyocyte pyroptosis holds great promise for the treatment of cardiovascular disease. In this paper, we summarized the characteristics, occurrence and regulation mechanism of pyroptosis are reviewed, and also discussed its role and mechanisms in CVD, such as atherosclerosis (AS), myocardial infarction (MI), arrhythmia and cardiac hypertrophy.
Collapse
Affiliation(s)
- Xinzhe Chen
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Peng-Chao Tian
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, National Center for Cardiovascular Diseases, Peking Union Medical College, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Kai Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Man Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
46
|
Ryder CB, Kondolf HC, O’Keefe ME, Zhou B, Abbott DW. Chemical Modulation of Gasdermin-Mediated Pyroptosis and Therapeutic Potential. J Mol Biol 2022; 434:167183. [PMID: 34358546 PMCID: PMC8810912 DOI: 10.1016/j.jmb.2021.167183] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/24/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022]
Abstract
Pyroptosis, a lytic form of programmed cell death, both stimulates effective immune responses and causes tissue damage. Gasdermin (GSDM) proteins are a family of pore-forming executors of pyroptosis. While the most-studied member, GSDMD, exerts critical functions in inflammasome biology, emerging evidence demonstrates potential broad relevance for GSDM-mediated pyroptosis across diverse pathologies. In this review, we describe GSDM biology, outline conditions where inflammasomes and GSDM-mediated pyroptosis represent rational therapeutic targets, and delineate strategies to manipulate these central immunologic processes for the treatment of human disease.
Collapse
Affiliation(s)
- Christopher B. Ryder
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106,Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA 44106
| | - Hannah C. Kondolf
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Meghan E. O’Keefe
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Bowen Zhou
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106
| | - Derek W. Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA 44106,Corresponding author: ()
| |
Collapse
|
47
|
Domínguez-Ruiz M, Rodríguez-Ballesteros M, Gandía M, Gómez-Rosas E, Villamar M, Scimemi P, Mancini P, Rendtorff ND, Moreno-Pelayo MA, Tranebjaerg L, Medà C, Santarelli R, del Castillo I. Novel Pathogenic Variants in PJVK, the Gene Encoding Pejvakin, in Subjects with Autosomal Recessive Non-Syndromic Hearing Impairment and Auditory Neuropathy Spectrum Disorder. Genes (Basel) 2022; 13:149. [PMID: 35052489 PMCID: PMC8775161 DOI: 10.3390/genes13010149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
Pathogenic variants in the PJVK gene cause the DFNB59 type of autosomal recessive non-syndromic hearing impairment (AR-NSHI). Phenotypes are not homogeneous, as a few subjects show auditory neuropathy spectrum disorder (ANSD), while others show cochlear hearing loss. The numbers of reported cases and pathogenic variants are still small to establish accurate genotype-phenotype correlations. We investigated a cohort of 77 Spanish familial cases of AR-NSHI, in whom DFNB1 had been excluded, and a cohort of 84 simplex cases with isolated ANSD in whom OTOF variants had been excluded. All seven exons and exon-intron boundaries of the PJVK gene were sequenced. We report three novel DFNB59 cases, one from the AR-NSHI cohort and two from the ANSD cohort, with stable, severe to profound NSHI. Two of the subjects received unilateral cochlear implantation, with apparent good outcomes. Our study expands the spectrum of PJVK mutations, as we report four novel pathogenic variants: p.Leu224Arg, p.His294Ilefs*43, p.His294Asp and p.Phe317Serfs*20. We review the reported cases of DFNB59, summarize the clinical features of this rare subtype of AR-NSHI and discuss the involvement of PJVK in ANSD.
Collapse
Affiliation(s)
- María Domínguez-Ruiz
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Montserrat Rodríguez-Ballesteros
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Marta Gandía
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Elena Gómez-Rosas
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Manuela Villamar
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Pietro Scimemi
- Department of Neurosciences, University of Padua, 35121 Padua, Italy; (P.S.); (R.S.)
- Audiology Service, Santi Giovanni e Paolo Hospital, 30122 Venice, Italy
| | - Patrizia Mancini
- Department of Sense Organs, University La Sapienza, 00162 Rome, Italy;
| | - Nanna D. Rendtorff
- Department of Clinical Genetics, University Hospital, Copenhagen/The Kennedy Centre, DK-2600 Glostrup, Denmark; (N.D.R.); (L.T.)
| | - Miguel A. Moreno-Pelayo
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| | - Lisbeth Tranebjaerg
- Department of Clinical Genetics, University Hospital, Copenhagen/The Kennedy Centre, DK-2600 Glostrup, Denmark; (N.D.R.); (L.T.)
- Department of Clinical Medicine, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Carme Medà
- Unidad de Prevención de Enfermedades del Oído, Conselleria de Salut, Illes Balears, 07120 Palma de Mallorca, Spain;
| | - Rosamaria Santarelli
- Department of Neurosciences, University of Padua, 35121 Padua, Italy; (P.S.); (R.S.)
- Audiology Service, Santi Giovanni e Paolo Hospital, 30122 Venice, Italy
| | - Ignacio del Castillo
- Servicio de Genética, Hospital Universitario Ramón y Cajal, IRYCIS, 28034 Madrid, Spain; (M.D.-R.); (M.R.-B.); (M.G.); (E.G.-R.); (M.V.); (M.A.M.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28034 Madrid, Spain
| |
Collapse
|
48
|
Angosto-Bazarra D, Alarcón-Vila C, Hurtado-Navarro L, Baños MC, Rivers-Auty J, Pelegrín P. Evolutionary analyses of the gasdermin family suggest conserved roles in infection response despite loss of pore-forming functionality. BMC Biol 2022; 20:9. [PMID: 34996441 PMCID: PMC8742441 DOI: 10.1186/s12915-021-01220-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gasdermins are ancient (>500million-years-ago) proteins, constituting a family of pore-forming proteins that allow the release of intracellular content including proinflammatory cytokines. Despite their importance in the immune response, and although gasdermin and gasdermin-like genes have been identified across a wide range of animal and non-animal species, there is limited information about the evolutionary history of the gasdermin family, and their functional roles after infection. In this study, we assess the lytic functions of different gasdermins across Metazoa species, and use a mouse model of sepsis to evaluate the expression of the different gasdermins during infection. RESULTS We show that the majority of gasdermin family members from distantly related animal clades are pore-forming, in line with the function of the ancestral proto-gasdermin and gasdermin-like proteins of Bacteria. We demonstrate the first expansion of this family occurred through a duplication of the ancestral gasdermin gene which formed gasdermin E and pejvakin prior to the divergence of cartilaginous fish and bony fish ~475 mya. We show that pejvakin from cartilaginous fish and mammals lost the pore-forming functionality and thus its role in cell lysis. We describe that the pore-forming gasdermin A formed ~320 mya as a duplication of gasdermin E prior to the divergence of the Sauropsida clade (the ancestral lineage of reptiles, turtles, and birds) and the Synapsid clade (the ancestral lineage of mammals). We then demonstrate that the gasdermin A gene duplicated to form the rest of the gasdermin family including gasdermins B, C, and D: pore-forming proteins that present a high variation of the exons in the linker sequence, which in turn allows for diverse activation pathways. Finally, we describe expression of murine gasdermin family members in different tissues in a mouse sepsis model, indicating function during infection response. CONCLUSIONS In this study we explored the evolutionary history of the gasdermin proteins in animals and demonstrated that the pore-formation functionality has been conserved from the ancient proto-gasdermin protein. We also showed that one gasdermin family member, pejvakin, lost its pore-forming functionality, but that all gasdermin family members, including pejvakin, likely retained a role in inflammation and the physiological response to infection.
Collapse
Affiliation(s)
- Diego Angosto-Bazarra
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista s/n. 30120 El Palmar, Murcia, Spain
| | - Cristina Alarcón-Vila
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista s/n. 30120 El Palmar, Murcia, Spain
| | - Laura Hurtado-Navarro
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista s/n. 30120 El Palmar, Murcia, Spain
| | - María C Baños
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista s/n. 30120 El Palmar, Murcia, Spain
| | - Jack Rivers-Auty
- Tasmanian School of Medicine, University of Tasmania, Tasmania, Australia
| | - Pablo Pelegrín
- Línea de Inflamación Molecular, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista s/n. 30120 El Palmar, Murcia, Spain. .,Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120, Murcia, Spain.
| |
Collapse
|
49
|
Li H, Lismont C, Revenco I, Hussein MAF, Costa CF, Fransen M. The Peroxisome-Autophagy Redox Connection: A Double-Edged Sword? Front Cell Dev Biol 2021; 9:814047. [PMID: 34977048 PMCID: PMC8717923 DOI: 10.3389/fcell.2021.814047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 01/18/2023] Open
Abstract
Peroxisomes harbor numerous enzymes that can produce or degrade hydrogen peroxide (H2O2). Depending on its local concentration and environment, this oxidant can function as a redox signaling molecule or cause stochastic oxidative damage. Currently, it is well-accepted that dysfunctional peroxisomes are selectively removed by the autophagy-lysosome pathway. This process, known as "pexophagy," may serve a protective role in curbing peroxisome-derived oxidative stress. Peroxisomes also have the intrinsic ability to mediate and modulate H2O2-driven processes, including (selective) autophagy. However, the molecular mechanisms underlying these phenomena are multifaceted and have only recently begun to receive the attention they deserve. This review provides a comprehensive overview of what is known about the bidirectional relationship between peroxisomal H2O2 metabolism and (selective) autophagy. After introducing the general concepts of (selective) autophagy, we critically examine the emerging roles of H2O2 as one of the key modulators of the lysosome-dependent catabolic program. In addition, we explore possible relationships among peroxisome functioning, cellular H2O2 levels, and autophagic signaling in health and disease. Finally, we highlight the most important challenges that need to be tackled to understand how alterations in peroxisomal H2O2 metabolism contribute to autophagy-related disorders.
Collapse
Affiliation(s)
- Hongli Li
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Celien Lismont
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Iulia Revenco
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Mohamed A. F. Hussein
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Department of Biochemistry, Faculty of Pharmacy, Assiut University, Asyut, Egypt
| | - Cláudio F. Costa
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Marc Fransen
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Zou J, Zheng Y, Huang Y, Tang D, Kang R, Chen R. The Versatile Gasdermin Family: Their Function and Roles in Diseases. Front Immunol 2021; 12:751533. [PMID: 34858408 PMCID: PMC8632255 DOI: 10.3389/fimmu.2021.751533] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
The gasdermin (GSDM) family, a novel group of structure-related proteins, consists of GSDMA, GSDMB, GSDMC, GSDMD, GSDME/DNFA5, and PVJK/GSDMF. GSDMs possess a C-terminal repressor domain, cytotoxic N-terminal domain, and flexible linker domain (except for GSDMF). The GSDM-NT domain can be cleaved and released to form large oligomeric pores in the membrane that facilitate pyroptosis. The emerging roles of GSDMs include the regulation of various physiological and pathological processes, such as cell differentiation, coagulation, inflammation, and tumorigenesis. Here, we introduce the basic structure, activation, and expression patterns of GSDMs, summarize their biological and pathological functions, and explore their regulatory mechanisms in health and disease. This review provides a reference for the development of GSDM-targeted drugs to treat various inflammatory and tissue damage-related conditions.
Collapse
Affiliation(s)
- Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yixiang Zheng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|