1
|
Kochersberger A, Torkashvand MM, Lee D, Baskoylu S, Sengupta T, Koonce N, Emerson CE, Patel NV, Colón-Ramos D, Flavell S, Horvitz HR, Venkatachalam V, Hammarlund M. Programmed Cell Death Modifies Neural Circuits and Tunes Intrinsic Behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557249. [PMID: 37745399 PMCID: PMC10515839 DOI: 10.1101/2023.09.11.557249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Programmed cell death is a common feature of animal development. During development of the C. elegans hermaphrodite, programmed cell death (PCD) removes 131 cells from stereotyped positions in the cell lineage, mostly in neuronal lineages. Blocking cell death results in supernumerary "undead" neurons. We find that undead neurons can be wired into circuits, can display activity, and can modify specific behaviors. The two undead RIM-like neurons participate in the RIM-containing circuit that computes movement. The addition of these two extra neurons results in animals that initiate fewer reversals and lengthens the duration of those reversals that do occur. We describe additional behavioral alterations of cell-death mutants, including in turning angle and pharyngeal pumping. These findings reveal that, like too much PCD, too little PCD can modify nervous system function and animal behavior.
Collapse
Affiliation(s)
- Alison Kochersberger
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine; New Haven, CT 06536, USA
| | | | - Dongyeop Lee
- Howard Hughes Medical Institute, Department of Biology, MIT; Cambridge, MA 02139, USA
| | - Saba Baskoylu
- Picower Institute for Learning and Memory, MIT; Cambridge, MA 02139, USA
| | - Titas Sengupta
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Noelle Koonce
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Chloe E Emerson
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Nandan V Patel
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine; New Haven, CT 06536, USA
| | - Daniel Colón-Ramos
- Department of Neuroscience and Department of Cell Biology, Yale University School of Medicine; New Haven, CT 06536, USA
- MBL Fellows, Marine Biological Laboratory; Woods Hole, MA 02543, USA
- Instituto de Neurobiología, Recinto de Ciencias Médicas, Universidad de Puerto Rico; San Juan 00901, Puerto Rico
| | - Steven Flavell
- Picower Institute for Learning and Memory, MIT; Cambridge, MA 02139, USA
| | - H Robert Horvitz
- Howard Hughes Medical Institute, Department of Biology, MIT; Cambridge, MA 02139, USA
| | | | - Marc Hammarlund
- Department of Genetics and Department of Neuroscience, Yale University School of Medicine; New Haven, CT 06536, USA
| |
Collapse
|
2
|
Li Y, Tian L, Zhang Y, Shi Y. Structural insights into CED-3 activation. Life Sci Alliance 2023; 6:e202302056. [PMID: 37402593 PMCID: PMC10320015 DOI: 10.26508/lsa.202302056] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 07/06/2023] Open
Abstract
In Caenorhabditis elegans (C. elegans), onset of programmed cell death is marked with the activation of CED-3, a process that requires assembly of the CED-4 apoptosome. Activated CED-3 forms a holoenzyme with the CED-4 apoptosome to cleave a wide range of substrates, leading to irreversible cell death. Despite decades of investigations, the underlying mechanism of CED-4-facilitated CED-3 activation remains elusive. Here, we report cryo-EM structures of the CED-4 apoptosome and three distinct CED-4/CED-3 complexes that mimic different activation stages for CED-3. In addition to the previously reported octamer in crystal structures, CED-4, alone or in complex with CED-3, exists in multiple oligomeric states. Supported by biochemical analyses, we show that the conserved CARD-CARD interaction promotes CED-3 activation, and initiation of programmed cell death is regulated by the dynamic organization of the CED-4 apoptosome.
Collapse
Affiliation(s)
- Yini Li
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Lu Tian
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ying Zhang
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yigong Shi
- Beijing Frontier Research Center for Biological Structures, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Westlake Laboratory of Life Science and Biomedicine, Westlake Institute for Advanced Study, Hangzhou, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
3
|
Over Fifty Years of Life, Death, and Cannibalism: A Historical Recollection of Apoptosis and Autophagy. Int J Mol Sci 2021; 22:ijms222212466. [PMID: 34830349 PMCID: PMC8618802 DOI: 10.3390/ijms222212466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023] Open
Abstract
Research in biomedical sciences has changed dramatically over the past fifty years. There is no doubt that the discovery of apoptosis and autophagy as two highly synchronized and regulated mechanisms in cellular homeostasis are among the most important discoveries in these decades. Along with the advancement in molecular biology, identifying the genetic players in apoptosis and autophagy has shed light on our understanding of their function in physiological and pathological conditions. In this review, we first describe the history of key discoveries in apoptosis with a molecular insight and continue with apoptosis pathways and their regulation. We touch upon the role of apoptosis in human health and its malfunction in several diseases. We discuss the path to the morphological and molecular discovery of autophagy. Moreover, we dive deep into the precise regulation of autophagy and recent findings from basic research to clinical applications of autophagy modulation in human health and illnesses and the available therapies for many diseases caused by impaired autophagy. We conclude with the exciting crosstalk between apoptosis and autophagy, from the early discoveries to recent findings.
Collapse
|
4
|
Meier B, Volkova NV, Wang B, González-Huici V, Bertolini S, Campbell PJ, Gerstung M, Gartner A. C. elegans genome-wide analysis reveals DNA repair pathways that act cooperatively to preserve genome integrity upon ionizing radiation. PLoS One 2021; 16:e0258269. [PMID: 34614038 PMCID: PMC8494335 DOI: 10.1371/journal.pone.0258269] [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: 06/02/2021] [Accepted: 09/22/2021] [Indexed: 11/22/2022] Open
Abstract
Ionizing radiation (IR) is widely used in cancer therapy and accidental or environmental exposure is a major concern. However, little is known about the genome-wide effects IR exerts on germ cells and the relative contribution of DNA repair pathways for mending IR-induced lesions. Here, using C. elegans as a model system and using primary sequencing data from our recent high-level overview of the mutagenic consequences of 11 genotoxic agents, we investigate in detail the genome-wide mutagenic consequences of exposing wild-type and 43 DNA repair and damage response defective C. elegans strains to a Caesium (Cs-137) source, emitting γ-rays. Cs-137 radiation induced single nucleotide variants (SNVs) at a rate of ~1 base substitution per 3 Gy, affecting all nucleotides equally. In nucleotide excision repair mutants, this frequency increased 2-fold concurrently with increased dinucleotide substitutions. As observed for DNA damage induced by bulky DNA adducts, small deletions were increased in translesion polymerase mutants, while base changes decreased. Structural variants (SVs) were augmented with dose, but did not arise with significantly higher frequency in any DNA repair mutants tested. Moreover, 6% of all mutations occurred in clusters, but clustering was not significantly altered in any DNA repair mutant background. Our data is relevant for better understanding how DNA repair pathways modulate IR-induced lesions.
Collapse
Affiliation(s)
- Bettina Meier
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
| | - Nadezda V. Volkova
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom
| | - Bin Wang
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China
| | - Víctor González-Huici
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
| | - Simone Bertolini
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
| | - Peter J. Campbell
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Hinxton, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, United Kingdom
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Anton Gartner
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
5
|
Hounsell C, Fan Y. The Duality of Caspases in Cancer, as Told through the Fly. Int J Mol Sci 2021; 22:8927. [PMID: 34445633 PMCID: PMC8396359 DOI: 10.3390/ijms22168927] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022] Open
Abstract
Caspases, a family of cysteine-aspartic proteases, have an established role as critical components in the activation and initiation of apoptosis. Alongside this a variety of non-apoptotic caspase functions in proliferation, differentiation, cellular plasticity and cell migration have been reported. The activity level and context are important factors in determining caspase function. As a consequence of their critical role in apoptosis and beyond, caspases are uniquely situated to have pathological roles, including in cancer. Altered caspase function is a common trait in a variety of cancers, with apoptotic evasion defined as a "hallmark of cancer". However, the role that caspases play in cancer is much more complex, acting both to prevent and to promote tumourigenesis. This review focuses on the major findings in Drosophila on the dual role of caspases in tumourigenesis. This has major implications for cancer treatments, including chemotherapy and radiotherapy, with the activation of apoptosis being the end goal. However, such treatments may inadvertently have adverse effects on promoting tumour progression and acerbating the cancer. A comprehensive understanding of the dual role of caspases will aid in the development of successful cancer therapeutic approaches.
Collapse
Affiliation(s)
| | - Yun Fan
- School of Biosciences, University of Birmingham, Birmingham B15 2TT, UK;
| |
Collapse
|
6
|
Jia H, Xue S, Lei L, Fan M, Peng S, Li T, Nagarajan R, Carver B, Ma Z, Deng J, Yan L. A semi-dominant NLR allele causes whole-seedling necrosis in wheat. PLANT PHYSIOLOGY 2021; 186:483-496. [PMID: 33576803 PMCID: PMC8154059 DOI: 10.1093/plphys/kiab058] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/26/2021] [Indexed: 05/26/2023]
Abstract
Programmed cell death (PCD) and apoptosis have key functions in development and disease resistance in diverse organisms; however, the induction of necrosis remains poorly understood. Here, we identified a semi-dominant mutant allele that causes the necrotic death of the entire seedling (DES) of wheat (Triticum aestivum L.) in the absence of any pathogen or external stimulus. Positional cloning of the lethal allele mDES1 revealed that this premature death via necrosis was caused by a point mutation from Asp to Asn at amino acid 441 in a nucleotide-binding leucine-rich repeat protein containing nucleotide-binding domain and leucine-rich repeats. The overexpression of mDES1 triggered necrosis and PCD in transgenic plants. However, transgenic wheat harboring truncated wild-type DES1 proteins produced through gene editing that exhibited no significant developmental defects. The point mutation in mDES1 did not cause changes in this protein in the oligomeric state, but mDES1 failed to interact with replication protein A leading to abnormal mitotic cell division. DES1 is an ortholog of Sr35, which recognizes a Puccinia graminis f. sp. tritici stem rust disease effector in wheat, but mDES1 gained function as a direct inducer of plant death. These findings shed light on the intersection of necrosis, apoptosis, and autoimmunity in plants.
Collapse
Affiliation(s)
- Haiyan Jia
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Crop Genomics and Bioinformatics Center and National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Jiangsu, Nanjing 210095, China
| | - Shulin Xue
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Crop Genomics and Bioinformatics Center and National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Jiangsu, Nanjing 210095, China
| | - Lei Lei
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Min Fan
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Crop Genomics and Bioinformatics Center and National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Jiangsu, Nanjing 210095, China
| | - Shuxia Peng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Tian Li
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ragupathi Nagarajan
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Brett Carver
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Zhengqiang Ma
- Crop Genomics and Bioinformatics Center and National Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Jiangsu, Nanjing 210095, China
| | - Junpeng Deng
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Liuling Yan
- Department of Plant and Soil Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
7
|
Suehiro Y, Yoshina S, Motohashi T, Iwata S, Dejima K, Mitani S. Efficient collection of a large number of mutations by mutagenesis of DNA damage response defective animals. Sci Rep 2021; 11:7630. [PMID: 33828169 PMCID: PMC8027614 DOI: 10.1038/s41598-021-87226-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/24/2021] [Indexed: 02/01/2023] Open
Abstract
With the development of massive parallel sequencing technology, it has become easier to establish new model organisms that are ideally suited to the specific biological phenomena of interest. Considering the history of research using classical model organisms, we believe that the efficient construction and sharing of gene mutation libraries will facilitate the progress of studies using these new model organisms. Using C. elegans, we applied the TMP/UV mutagenesis method to animals lacking function in the DNA damage response genes atm-1 and xpc-1. This method produces genetic mutations three times more efficiently than mutagenesis of wild-type animals. Furthermore, we confirmed that the use of next-generation sequencing and the elimination of false positives through machine learning could automate the process of mutation identification with an accuracy of over 95%. Eventually, we sequenced the whole genomes of 488 strains and isolated 981 novel mutations generated by the present method; these strains have been made available to anyone who wants to use them. Since the targeted DNA damage response genes are well conserved and the mutagens used in this study are also effective in a variety of species, we believe that our method is generally applicable to a wide range of animal species.
Collapse
Affiliation(s)
- Yuji Suehiro
- Department of Physiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Sawako Yoshina
- Department of Physiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Tomoko Motohashi
- Department of Physiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Satoru Iwata
- Chubu University Center for Education in Laboratory Animal Research, Kasugai, Aichi, Japan
| | - Katsufumi Dejima
- Department of Physiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Shohei Mitani
- Department of Physiology, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan.
- Tokyo Women's Medical University Institute for Integrated Medical Sciences, Shinjuku, Tokyo, Japan.
| |
Collapse
|
8
|
Wang C, Xia C, Zhu Y, Zhang H. Innovative fluorescent probes for in vivo visualization of biomolecules in living Caenorhabditis elegans. Cytometry A 2021; 99:560-574. [PMID: 33638604 DOI: 10.1002/cyto.a.24325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022]
Abstract
Caenorhabditis elegans (C. elegans) as a well-established multicellular model organism has been widely used in the biological field for half a century. Its numerous advantages including small body size, rapid life cycle, high-reproductive rate, well-defined anatomy, and conserved genome, has made C. elegans one of the most successful multicellular model organisms. Discoveries obtained from the C. elegans model have made great contributions to research fields such as development, aging, biophysics, immunology, and neuroscience. Because of its transparent body and giant cell size, C. elegans is also an ideal subject for high resolution and high-throughput optical imaging and analysis. During the past decade, great advances have been made to develop biomolecule-targeting techniques for noninvasive optical imaging. These novel technologies expanded the toolbox for qualitative and quantitative analysis of biomolecules in C. elegans. In this review, we summarize recently developed fluorescent probes or labeling techniques for visualizing biomolecules at the cellular, subcellular or molecular scale by using C. elegans as the major model organism or designed specifically for the applications in C. elegans. Combining the technological advantages of the C. elegans model with the novel fluorescent labeling techniques will provide new horizons for high-efficiency quantitative optical analysis in live organisms.
Collapse
Affiliation(s)
- Chunxia Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Chujie Xia
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yi Zhu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Huimin Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
9
|
Quarleri J, Cevallos C, Delpino MV. Apoptosis in infectious diseases as a mechanism of immune evasion and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:1-24. [PMID: 33931136 DOI: 10.1016/bs.apcsb.2021.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In pluricellular organisms, apoptosis is indispensable for the development and homeostasis. During infection, apoptosis plays the main role in the elimination of infected cells. Infectious diseases control apoptosis, and this contributes to disease pathogenesis. Increased apoptosis may participate in two different ways. It can assist the dissemination of intracellular pathogens or induce immunosuppression to favor pathogen dissemination. In other conditions, apoptosis can benefit eradicate infectious agents from the host. Accordingly, bacteria, viruses, fungi, and parasites have developed strategies to inhibit host cell death by apoptosis to allow intracellular survival and persistence of the pathogen. The clarification of the intracellular signaling pathways, the receptors involved and the pathogen factors that interfere with apoptosis could disclose new therapeutic targets for blocking microbial actions on apoptotic pathways. In this review, we summarize the current knowledge on pathogen anti-apoptotic and apoptotic approaches and the mechanisms involving in disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Cintia Cevallos
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - María Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Gartner A, Engebrecht J. DNA repair, recombination, and damage signaling. Genetics 2021; 220:6522877. [PMID: 35137093 PMCID: PMC9097270 DOI: 10.1093/genetics/iyab178] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 01/09/2023] Open
Abstract
DNA must be accurately copied and propagated from one cell division to the next, and from one generation to the next. To ensure the faithful transmission of the genome, a plethora of distinct as well as overlapping DNA repair and recombination pathways have evolved. These pathways repair a large variety of lesions, including alterations to single nucleotides and DNA single and double-strand breaks, that are generated as a consequence of normal cellular function or by external DNA damaging agents. In addition to the proteins that mediate DNA repair, checkpoint pathways have also evolved to monitor the genome and coordinate the action of various repair pathways. Checkpoints facilitate repair by mediating a transient cell cycle arrest, or through initiation of cell suicide if DNA damage has overwhelmed repair capacity. In this chapter, we describe the attributes of Caenorhabditis elegans that facilitate analyses of DNA repair, recombination, and checkpoint signaling in the context of a whole animal. We review the current knowledge of C. elegans DNA repair, recombination, and DNA damage response pathways, and their role during development, growth, and in the germ line. We also discuss how the analysis of mutational signatures in C. elegans is helping to inform cancer mutational signatures in humans.
Collapse
Affiliation(s)
- Anton Gartner
- Department for Biological Sciences, IBS Center for Genomic Integrity, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea,Corresponding author: (A.G.); (J.E.)
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA,Corresponding author: (A.G.); (J.E.)
| |
Collapse
|
11
|
Abstract
Cell death is an important facet of animal development. In some developing tissues, death is the ultimate fate of over 80% of generated cells. Although recent studies have delineated a bewildering number of cell death mechanisms, most have only been observed in pathological contexts, and only a small number drive normal development. This Primer outlines the important roles, different types and molecular players regulating developmental cell death, and discusses recent findings with which the field currently grapples. We also clarify terminology, to distinguish between developmental cell death mechanisms, for which there is evidence for evolutionary selection, and cell death that follows genetic, chemical or physical injury. Finally, we suggest how advances in understanding developmental cell death may provide insights into the molecular basis of developmental abnormalities and pathological cell death in disease.
Collapse
Affiliation(s)
- Piya Ghose
- Department of Biology, The University of Texas at Arlington, 655 Mitchell St., Arlington, TX 76019, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
12
|
Activity-Dependent Regulation of the Proapoptotic BH3-Only Gene egl-1 in a Living Neuron Pair in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2019; 9:3703-3714. [PMID: 31519744 PMCID: PMC6829140 DOI: 10.1534/g3.119.400654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The BH3-only family of proteins is key for initiating apoptosis in a variety of contexts, and may also contribute to non-apoptotic cellular processes. Historically, the nematode Caenorhabditis elegans has provided a powerful system for studying and identifying conserved regulators of BH3-only proteins. In C. elegans, the BH3-only protein egl-1 is expressed during development to cell-autonomously trigger most developmental cell deaths. Here we provide evidence that egl-1 is also transcribed after development in the sensory neuron pair URX without inducing apoptosis. We used genetic screening and epistasis analysis to determine that its transcription is regulated in URX by neuronal activity and/or in parallel by orthologs of Protein Kinase G and the Salt-Inducible Kinase family. Because several BH3-only family proteins are also expressed in the adult nervous system of mammals, we suggest that studying egl-1 expression in URX may shed light on mechanisms that regulate conserved family members in higher organisms.
Collapse
|
13
|
Wang G, Sun L, Reina CP, Song I, Gabel CV, Driscoll M. CED-4 CARD domain residues can modulate non-apoptotic neuronal regeneration functions independently from apoptosis. Sci Rep 2019; 9:13315. [PMID: 31527664 PMCID: PMC6746752 DOI: 10.1038/s41598-019-49633-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 06/20/2019] [Indexed: 12/31/2022] Open
Abstract
A major challenge in regenerative medicine is the repair of injured neurons. Regeneration of laser-cut C. elegans neurons requires early action of core apoptosis activator CED-4/Apaf1 and CED-3/caspase. While testing models for CED-4 as a candidate calcium-sensitive activator of repair, we unexpectedly discovered that amino acid substitutions affecting alpha-helix-6 within the CED-4 caspase recruitment domain (CARD) confer a CED-4 gain-of-function (gf) activity that increases axonal regrowth without disrupting CED-4 apoptosis activity. The in vivo caspase reporter CA-GFP reveals a rapid localized increase in caspase activity upon axotomy, which is absent in ced-4 and ced-3 loss-of-function mutants but present in the ced-4(gf) mutant. The ced-3 loss-of-function mutation can significantly suppress the axonal regrowth of the ced-4(gf) mutant, indicating that CED-4(gf) regeneration depends on CED-3 caspase. Thus, we identified a subdomain within the CED-4 CARD that regulates the dynamic and controlled caspase activity required for efficient regeneration.
Collapse
Affiliation(s)
- Guoqiang Wang
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Lin Sun
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Christopher P Reina
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Isaac Song
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Christopher V Gabel
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
14
|
Cullin-4B E3 ubiquitin ligase mediates Apaf-1 ubiquitination to regulate caspase-9 activity. PLoS One 2019; 14:e0219782. [PMID: 31329620 PMCID: PMC6645535 DOI: 10.1371/journal.pone.0219782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/01/2019] [Indexed: 11/25/2022] Open
Abstract
Apoptotic protease-activating factor 1 (Apaf-1) is a component of apoptosome, which regulates caspase-9 activity. In addition to apoptosis, Apaf-1 plays critical roles in the intra-S-phase checkpoint; therefore, impaired expression of Apaf-1 has been demonstrated in chemotherapy-resistant malignant melanoma and nuclear translocation of Apaf-1 has represented a favorable prognosis of patients with non-small cell lung cancer. In contrast, increased levels of Apaf-1 protein are observed in the brain in Huntington’s disease. The regulation of Apaf-1 protein is not yet fully understood. In this study, we show that etoposide triggers the interaction of Apaf-1 with Cullin-4B, resulting in enhanced Apaf-1 ubiquitination. Ubiquitinated Apaf-1, which was degraded in healthy cells, binds p62 and forms aggregates in the cytosol. This complex of ubiquitinated Apaf-1 and p62 induces caspase-9 activation following MG132 treatment of HEK293T cells that stably express bcl-xl. These results show that ubiquitinated Apaf-1 may activate caspase-9 under conditions of proteasome impairment.
Collapse
|
15
|
Profile of Junying Yuan. Proc Natl Acad Sci U S A 2019; 116:11564-11566. [DOI: 10.1073/pnas.1906915116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
16
|
Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019; 29:347-364. [PMID: 30948788 PMCID: PMC6796845 DOI: 10.1038/s41422-019-0164-5] [Citation(s) in RCA: 1451] [Impact Index Per Article: 290.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
Cells may die from accidental cell death (ACD) or regulated cell death (RCD). ACD is a biologically uncontrolled process, whereas RCD involves tightly structured signaling cascades and molecularly defined effector mechanisms. A growing number of novel non-apoptotic forms of RCD have been identified and are increasingly being implicated in various human pathologies. Here, we critically review the current state of the art regarding non-apoptotic types of RCD, including necroptosis, pyroptosis, ferroptosis, entotic cell death, netotic cell death, parthanatos, lysosome-dependent cell death, autophagy-dependent cell death, alkaliptosis and oxeiptosis. The in-depth comprehension of each of these lethal subroutines and their intercellular consequences may uncover novel therapeutic targets for the avoidance of pathogenic cell loss.
Collapse
Affiliation(s)
- Daolin Tang
- The Third Affiliated Hospital, Protein Modification and Degradation Lab, School of Basic Medical Sciences, Guangzhou Medical University, 510510, Guangzhou, Guangdong, China.
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tom Vanden Berghe
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, 9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Laboratory of Pathophysiology, Faculty of Biomedical Sciences, University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, 9052, Ghent, Belgium
- Department for Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Methusalem program, Ghent University, 9000, Ghent, Belgium
| | - Guido Kroemer
- Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France.
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, 75006, Paris, France.
- Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.
- Université Pierre et Marie Curie, 75006, Paris, France.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800, Villejuif, France.
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015, Paris, France.
- Department of Women's and Children's Health, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
17
|
Harders RH, Morthorst TH, Lande AD, Hesselager MO, Mandrup OA, Bendixen E, Stensballe A, Olsen A. Dynein links engulfment and execution of apoptosis via CED-4/Apaf1 in C. elegans. Cell Death Dis 2018; 9:1012. [PMID: 30262881 PMCID: PMC6160458 DOI: 10.1038/s41419-018-1067-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/21/2018] [Accepted: 09/10/2018] [Indexed: 12/14/2022]
Abstract
Apoptosis ensures removal of damaged cells and helps shape organs during development by removing excessive cells. To prevent the intracellular content of the apoptotic cells causing damage to surrounding cells, apoptotic cells are quickly cleared by engulfment. Tight regulation of apoptosis and engulfment is needed to prevent several pathologies such as cancer, neurodegenerative and autoimmune diseases. There is increasing evidence that the engulfment machinery can regulate the execution of apoptosis. However, the underlying molecular mechanisms are poorly understood. We show that dynein mediates cell non-autonomous cross-talk between the engulfment and apoptotic programs in the Caenorhabditis elegans germline. Dynein is an ATP-powered microtubule-based molecular motor, built from several subunits. Dynein has many diverse functions including transport of cargo around the cell. We show that both dynein light chain 1 (DLC-1) and dynein heavy chain 1 (DHC-1) localize to the nuclear membrane inside apoptotic germ cells in C. elegans. Strikingly, lack of either DLC-1 or DHC-1 at the nuclear membrane inhibits physiological apoptosis specifically in mutants defective in engulfment. This suggests that a cell fate determining dialogue takes place between engulfing somatic sheath cells and apoptotic germ cells. The underlying mechanism involves the core apoptotic protein CED-4/Apaf1, as we find that DLC-1 and the engulfment protein CED-6/GULP are required for the localization of CED-4 to the nuclear membrane of germ cells. A better understanding of the communication between the engulfment machinery and the apoptotic program is essential for identifying novel therapeutic targets in diseases caused by inappropriate engulfment or apoptosis.
Collapse
Affiliation(s)
- Rikke Hindsgaul Harders
- Department of Chemistry and Biosciences, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark
| | - Tine Hørning Morthorst
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus C, DK-8000, Denmark
| | - Anna Dippel Lande
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus C, DK-8000, Denmark
| | | | - Ole Aalund Mandrup
- Department of Engineering, Aarhus University, Gustav Wieds Vej 10C, Aarhus C, DK-8000, Denmark
| | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus C, DK-8000, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 7E, Aalborg, DK-9220, Denmark
| | - Anders Olsen
- Department of Chemistry and Biosciences, Aalborg University, Fredrik Bajers Vej 7H, Aalborg, DK-9220, Denmark.
| |
Collapse
|
18
|
Endoribonuclease ENDU-2 regulates multiple traits including cold tolerance via cell autonomous and nonautonomous controls in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2018; 115:8823-8828. [PMID: 30104389 DOI: 10.1073/pnas.1808634115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Environmental temperature acclimation is essential to animal survival, yet thermoregulation mechanisms remain poorly understood. We demonstrate cold tolerance in Caenorhabditis elegans as regulated by paired ADL chemosensory neurons via Ca2+-dependent endoribonuclease (EndoU) ENDU-2. Loss of ENDU-2 function results in life span, brood size, and synaptic remodeling abnormalities in addition to enhanced cold tolerance. Enzymatic ENDU-2 defects localized in the ADL and certain muscle cells led to increased cold tolerance in endu-2 mutants. Ca2+ imaging revealed ADL neurons were responsive to temperature stimuli through transient receptor potential (TRP) channels, concluding that ADL function requires ENDU-2 action in both cell-autonomous and cell-nonautonomous mechanisms. ENDU-2 is involved in caspase expression, which is central to cold tolerance and synaptic remodeling in dorsal nerve cord. We therefore conclude that ENDU-2 regulates cell type-dependent, cell-autonomous, and cell-nonautonomous cold tolerance.
Collapse
|
19
|
Dorstyn L, Akey CW, Kumar S. New insights into apoptosome structure and function. Cell Death Differ 2018; 25:1194-1208. [PMID: 29765111 PMCID: PMC6030056 DOI: 10.1038/s41418-017-0025-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023] Open
Abstract
The apoptosome is a platform that activates apical procaspases in response to intrinsic cell death signals. Biochemical and structural studies in the past two decades have extended our understanding of apoptosome composition and structure, while illuminating the requirements for initiator procaspase activation. A number of studies have now provided high-resolution structures for apoptosomes from C. elegans (CED-4), D. melanogaster (Dark), and H. sapiens (Apaf-1), which define critical protein interfaces, including intra and interdomain interactions. This work also reveals interactions of apoptosomes with their respective initiator caspases, CED-3, Dronc and procaspase-9. Structures of the human apoptosome have defined the requirements for cytochrome c binding, which triggers the conversion of inactive Apaf-1 molecules to an extended, assembly competent state. While recent data have provided a detailed understanding of apoptosome formation and procaspase activation, they also highlight important evolutionary differences with functional implications for caspase activation. Comparison of the CARD/CARD disks and apoptosomes formed by CED-4, Dark and Apaf-1. Cartoons of the active states of the CARD-CARD disks, illustrating the two CED-4 CARD tetrameric ring layers (CED4a and CED4b; top row) and the binding of 8 Dronc CARDs and between 3-4 pc-9 CARDs, to the Dark and Apaf-1 CARD disk respectively (middle and lower rows). Ribbon diagrams of the active CED-4, Dark and Apaf-1 apoptosomes are shown (right column).
Collapse
Affiliation(s)
- Loretta Dorstyn
- Center for Cancer Biology, University of South Australia and SA Pathology, Frome Road, Adelaide, SA, 5001, Australia.
| | - Christopher W Akey
- Department of Physiology and Biophysics, Boston University School of Medicine, 700 Albany Street, Boston, MA, 02118, USA
| | - Sharad Kumar
- Center for Cancer Biology, University of South Australia and SA Pathology, Frome Road, Adelaide, SA, 5001, Australia.
| |
Collapse
|
20
|
Cao R, Wang Q, Yang D, Liu Y, Ran W, Qu Y, Wu H, Cong M, Li F, Ji C, Zhao J. CO 2-induced ocean acidification impairs the immune function of the Pacific oyster against Vibrio splendidus challenge: An integrated study from a cellular and proteomic perspective. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 625:1574-1583. [PMID: 29996454 DOI: 10.1016/j.scitotenv.2018.01.056] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/02/2018] [Accepted: 01/07/2018] [Indexed: 06/08/2023]
Abstract
Ocean acidification (OA) and pathogenic diseases pose a considerable threat to key species of marine ecosystem. However, few studies have investigated the combined impact of reduced seawater pH and pathogen challenge on the immune responses of marine invertebrates. In this study, Pacific oysters, Crassostrea gigas, were exposed to OA (~2000 ppm) for 28 days and then challenged with Vibrio splendidus for another 72 h. Hemocyte parameters showed that V. splendidus infection exacerbated the impaired oyster immune responses under OA exposure. An iTRAQ-based quantitative proteomic analysis revealed that C. gigas responded differently to OA stress and V. splendidus challenge, alone or in combination. Generally, OA appears to act via a generalized stress response by causing oxidative stress, which could lead to cellular injury and cause disruption to the cytoskeleton, protein turnover, immune responses and energy metabolism. V. splendidus challenge in oysters could suppress the immune system directly and lead to a disturbed cytoskeleton structure, increased protein turnover and energy metabolism suppression, without causing oxidative stress. The combined OA- and V. splendidus-treated oysters ultimately presented a similar, but stronger proteomic response pattern compared with OA treatment alone. Overall, the impaired oyster immune functions caused by OA exposure may have increased the risk of V. splendidus infection. These results have important implications for the impact of OA on disease outbreaks in marine invertebrates, which would have significant economic and ecological repercussions.
Collapse
Affiliation(s)
- Ruiwen Cao
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qing Wang
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China
| | - Dinglong Yang
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China
| | - Yongliang Liu
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China
| | - Wen Ran
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yi Qu
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Huifeng Wu
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, PR China
| | - Ming Cong
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, PR China
| | - Fei Li
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, PR China
| | - Chenglong Ji
- Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong 264003, PR China
| | - Jianmin Zhao
- Muping Coastal Environmental Research Station, Yantai Institute of Coastal Zone Research, Yantai, Shandong 264117, PR China.
| |
Collapse
|
21
|
Kumar S, Calianese D, Birge RB. Efferocytosis of dying cells differentially modulate immunological outcomes in tumor microenvironment. Immunol Rev 2018; 280:149-164. [PMID: 29027226 DOI: 10.1111/imr.12587] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Programmed cell death (apoptosis) is an integral part of tissue homeostasis in complex organisms, allowing for tissue turnover, repair, and renewal while simultaneously inhibiting the release of self antigens and danger signals from apoptotic cell-derived constituents that can result in immune activation, inflammation, and autoimmunity. Unlike cells in culture, the physiological fate of cells that die by apoptosis in vivo is their rapid recognition and engulfment by phagocytic cells (a process called efferocytosis). To this end, apoptotic cells express specific eat-me signals, such as externalized phosphatidylserine (PS), that are recognized in a specific context by receptors to initiate signaling pathways for engulfment. The importance of carefully regulated recognition and clearance pathways is evident in the spectrum of inflammatory and autoimmune disorders caused by defects in PS receptors and signaling molecules. However, in recent years, several additional cell death pathways have emerged, including immunogenic cell death, necroptosis, pyroptosis, and netosis that interweave different cell death pathways with distinct innate and adaptive responses from classical apoptosis that can shape long-term host immunity. In this review, we discuss the role of different cell death pathways in terms of their immune potential outcomes specifically resulting in specific cell corpse/phagocyte interactions (phagocytic synapses) that impinge on host immunity, with a main emphasis on tolerance and cancer immunotherapy.
Collapse
Affiliation(s)
- Sushil Kumar
- New Jersey Medical School, Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, USA
| | - David Calianese
- New Jersey Medical School, Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, USA
| | - Raymond B Birge
- New Jersey Medical School, Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, Newark, NJ, USA
| |
Collapse
|
22
|
King SD, Gray CF, Song L, Nechushtai R, Gumienny TL, Mittler R, Padilla PA. The cisd gene family regulates physiological germline apoptosis through ced-13 and the canonical cell death pathway in Caenorhabditis elegans. Cell Death Differ 2018; 26:162-178. [PMID: 29666474 PMCID: PMC6294797 DOI: 10.1038/s41418-018-0108-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/13/2018] [Accepted: 03/05/2018] [Indexed: 11/09/2022] Open
Abstract
Programmed cell death, which occurs through a conserved core molecular pathway, is important for fundamental developmental and homeostatic processes. The human iron-sulfur binding protein NAF-1/CISD2 binds to Bcl-2 and its disruption in cells leads to an increase in apoptosis. Other members of the CDGSH iron sulfur domain (CISD) family include mitoNEET/CISD1 and Miner2/CISD3. In humans, mutations in CISD2 result in Wolfram syndrome 2, a disease in which the patients display juvenile diabetes, neuropsychiatric disorders and defective platelet aggregation. The C. elegans genome contains three previously uncharacterized cisd genes that code for CISD-1, which has homology to mitoNEET/CISD1 and NAF-1/CISD2, and CISD-3.1 and CISD-3.2, both of which have homology to Miner2/CISD3. Disrupting the function of the cisd genes resulted in various germline abnormalities including distal tip cell migration defects and a significant increase in the number of cell corpses within the adult germline. This increased germ cell death is blocked by a gain-of-function mutation of the Bcl-2 homolog CED-9 and requires functional caspase CED-3 and the APAF-1 homolog CED-4. Furthermore, the increased germ cell death is facilitated by the pro-apoptotic, CED-9-binding protein CED-13, but not the related EGL-1 protein. This work is significant because it places the CISD family members as regulators of physiological germline programmed cell death acting through CED-13 and the core apoptotic machinery.
Collapse
Affiliation(s)
- Skylar D King
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Chipo F Gray
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Luhua Song
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Rachel Nechushtai
- Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 91904, Israel
| | - Tina L Gumienny
- Department of Biology, Texas Woman's University, Denton, TX, 76204, USA
| | - Ron Mittler
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Pamela A Padilla
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA.
| |
Collapse
|
23
|
Viewing BCL2 and cell death control from an evolutionary perspective. Cell Death Differ 2017; 25:13-20. [PMID: 29099481 DOI: 10.1038/cdd.2017.145] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/14/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022] Open
Abstract
The last 30 years of studying BCL2 have brought cell death research into the molecular era, and revealed its relevance to human pathophysiology. Most, if not all metazoans use an evolutionarily conserved process for cellular self destruction that is controlled and implemented by proteins related to BCL2. We propose the anti-apoptotic BCL2-like and pro-apoptotic BH3-only members of the family arose through duplication and modification of genes for the pro-apoptotic multi-BH domain family members, such as BAX and BAK1. In that way, a cell suicide process that initially evolved as a mechanism for defense against intracellular parasites was then also used in multicellular organisms for morphogenesis and to maintain the correct number of cells in adults by balancing cell production by mitosis.
Collapse
|
24
|
Sung M, Kawasaki I, Shim YH. Depletion of cdc-25.3, a Caenorhabditis elegans orthologue of cdc25, increases physiological germline apoptosis. FEBS Lett 2017. [PMID: 28627101 DOI: 10.1002/1873-3468.12717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In Caenorhabditis elegans hermaphrodites, physiological germline apoptosis is higher in cdc-25.3 mutants than in wild-type. The elevated germline apoptosis in cdc-25.3 mutants seems to be induced by accumulation of double-stranded DNA breaks (DSBs). Both DNA damage and synapsis checkpoint genes are required to increase the germline apoptosis. Notably, the number of germ cells that lose P-granule components, PGL-1 and PGL-3, increase in cdc-25.3 mutants, and the increase in germline apoptosis requires the activity of SIR-2.1, a Sirtuin orthologue. These results suggest that elevation of germline apoptosis in cdc-25.3 mutants is induced by accumulation of DSBs, leading to a loss of PGL-1 and PGL-3 in germ cells, which promotes cytoplasmic translocation of SIR-2.1, and finally activates the core apoptotic machinery.
Collapse
Affiliation(s)
- Minhee Sung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Ichiro Kawasaki
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| | - Yhong-Hee Shim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Korea
| |
Collapse
|
25
|
Abstract
In 1882, Elie Metchnikoff identified myeloid-like cells from starfish larvae responding to the invasion by a foreign body (rose thorn). This marked the origins for the study of innate immunity, and an appreciation that cellular immunity was well established even in these "primitive" organisms. This chapter focuses on these myeloid cells as well as the newest members of this family, the dendritic cells, and explores their evolutionary origins. Our goal is to provide evolutionary context for the development of the multilayered immune system of mammals, where myeloid cells now serve as central effectors of innate immunity and regulators of adaptive immunity. Overall, we find that core contributions of myeloid cells to the regulation of inflammation are based on mechanisms that have been honed over hundreds of millions of years of evolution. Using phagocytosis as a platform, we show how fairly simple beginnings have offered a robust foundation onto which additional control features have been integrated, resulting in central regulatory nodes that now manage multifactorial aspects of homeostasis and immunity.
Collapse
|
26
|
Programmed Cell Death During Caenorhabditis elegans Development. Genetics 2017; 203:1533-62. [PMID: 27516615 DOI: 10.1534/genetics.115.186247] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/22/2016] [Indexed: 12/21/2022] Open
Abstract
Programmed cell death is an integral component of Caenorhabditis elegans development. Genetic and reverse genetic studies in C. elegans have led to the identification of many genes and conserved cell death pathways that are important for the specification of which cells should live or die, the activation of the suicide program, and the dismantling and removal of dying cells. Molecular, cell biological, and biochemical studies have revealed the underlying mechanisms that control these three phases of programmed cell death. In particular, the interplay of transcriptional regulatory cascades and networks involving multiple transcriptional regulators is crucial in activating the expression of the key death-inducing gene egl-1 and, in some cases, the ced-3 gene in cells destined to die. A protein interaction cascade involving EGL-1, CED-9, CED-4, and CED-3 results in the activation of the key cell death protease CED-3, which is tightly controlled by multiple positive and negative regulators. The activation of the CED-3 caspase then initiates the cell disassembly process by cleaving and activating or inactivating crucial CED-3 substrates; leading to activation of multiple cell death execution events, including nuclear DNA fragmentation, mitochondrial elimination, phosphatidylserine externalization, inactivation of survival signals, and clearance of apoptotic cells. Further studies of programmed cell death in C. elegans will continue to advance our understanding of how programmed cell death is regulated, activated, and executed in general.
Collapse
|
27
|
Regulation of CED-3 caspase localization and activation by C. elegans nuclear-membrane protein NPP-14. Nat Struct Mol Biol 2016; 23:958-964. [PMID: 27723735 DOI: 10.1038/nsmb.3308] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 09/16/2016] [Indexed: 12/18/2022]
Abstract
Caspases are cysteine proteases with critical roles in apoptosis. The Caenorhabditis elegans caspase CED-3 is activated by autocatalytic cleavage, a process enhanced by CED-4. Here we report that the CED-3 zymogen localizes to the perinuclear region in C. elegans germ cells and that CED-3 autocatalytic cleavage is held in check by C. elegans nuclei and activated by CED-4. The nuclear-pore protein NPP-14 interacts with the CED-3 zymogen prodomain, colocalizes with CED-3 in vivo and inhibits CED-3 autoactivation in vitro. Several missense mutations in the CED-3 prodomain result in stronger association with NPP-14 and decreased CED-3 activation by CED-4 in the presence of nuclei or NPP-14, thus leading to cell-death defects. Those same mutations enhance autocatalytic cleavage of CED-3 in vitro and increase apoptosis in vivo in the absence of npp-14. Our results reveal a critical role of nuclei and nuclear-membrane proteins in regulating the activation and localization of CED-3.
Collapse
|
28
|
Al-Amin M, Min H, Shim YH, Kawasaki I. Somatically expressed germ-granule components, PGL-1 and PGL-3, repress programmed cell death in C. elegans. Sci Rep 2016; 6:33884. [PMID: 27650246 PMCID: PMC5030653 DOI: 10.1038/srep33884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/05/2016] [Indexed: 01/15/2023] Open
Abstract
We previously reported that germline apoptosis in C. elegans increased by loss of PGL-1 and PGL-3, members of a family of constitutive germ-granule components, from germ cells in adult hermaphrodite gonads. In this study, we found that somatic apoptosis was reduced in synthetic multivulva class B (synMuv B) mutants due to ectopic expression of PGL-1 and PGL-3 in the soma. In synMuv B-mutant somatic cells, CED-4 expression level was reduced due to ectopic expression of PGL-1. Furthermore, in contrast to wild type, somatic apoptosis in synMuv B mutants increased following DNA damage in a SIR-2.1-dependent manner. Intriguingly, somatic apoptosis was repressed not only in synMuv B mutants but also by ectopically expressing pgl-1 and/or pgl-3 transgenes in wild-type somatic cells. Our study demonstrates that germ-granule components, PGL-1 and PGL-3, can serve as negative regulators of apoptosis not only in the germline but also in the soma in C. elegans.
Collapse
Affiliation(s)
- Mohammad Al-Amin
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Hyemin Min
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Yhong-Hee Shim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Ichiro Kawasaki
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
29
|
Wang X, Yang C. Programmed cell death and clearance of cell corpses in Caenorhabditis elegans. Cell Mol Life Sci 2016; 73:2221-36. [PMID: 27048817 PMCID: PMC11108496 DOI: 10.1007/s00018-016-2196-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 01/01/2023]
Abstract
Programmed cell death is critical to the development of diverse animal species from C. elegans to humans. In C. elegans, the cell death program has three genetically distinguishable phases. During the cell suicide phase, the core cell death machinery is activated through a protein interaction cascade. This activates the caspase CED-3, which promotes numerous pro-apoptotic activities including DNA degradation and exposure of the phosphatidylserine "eat me" signal on the cell corpse surface. Specification of the cell death fate involves transcriptional activation of the cell death initiator EGL-1 or the caspase CED-3 by coordinated actions of specific transcription factors in distinct cell types. In the cell corpse clearance stage, recognition of cell corpses by phagocytes triggers several signaling pathways to induce phagocytosis of apoptotic cell corpses. Cell corpse-enclosing phagosomes ultimately fuse with lysosomes for digestion of phagosomal contents. This article summarizes our current knowledge about programmed cell death and clearance of cell corpses in C. elegans.
Collapse
Affiliation(s)
- Xiaochen Wang
- National Institute of Biological Sciences, No. 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
| | - Chonglin Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
30
|
|
31
|
Wang MX, Itoh M, Li S, Hida Y, Ohta K, Hayakawa M, Nishida E, Ueda M, Islam S, Tana, Nakagawa T. CED-4 is an mRNA-binding protein that delivers ced-3 mRNA to ribosomes. Biochem Biophys Res Commun 2015; 470:48-53. [PMID: 26740177 DOI: 10.1016/j.bbrc.2015.12.102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/22/2015] [Indexed: 10/22/2022]
Abstract
Cell death abnormal (ced)-3 and ced-4 genes regulate apoptosis to maintain tissue homeostasis in Caenorhabditis elegans. Apoptosome formation and CED-4 translocation drive CED-3 activation. However, the precise role of CED-4 translocation is not yet fully understood. In this study, using a combination of immunoprecipitation and reverse transcription-polymerase chain reaction methods in cells and a glutathione-S-transferase pull down assay in a cell-free system, we show that CED-4 binds ced-3 mRNA. In the presence of ced-3 mRNA, CED-4 protein is enriched in the microsomal fraction and interacts with ribosomal protein L10a in mammalian cells, increasing the levels of CED-3. These results suggest that CED-4 forms a complex with ced-3 mRNA and delivers it to ribosomes for translation.
Collapse
Affiliation(s)
- Miao-Xing Wang
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Masanori Itoh
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Shimo Li
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Yoko Hida
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Kazunori Ohta
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Miki Hayakawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Emika Nishida
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Masashi Ueda
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Saiful Islam
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Tana
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Toshiyuki Nakagawa
- Department of Neurobiology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan.
| |
Collapse
|
32
|
Leung A, Hua K, Ramachandran P, Hingwing K, Wu M, Koh PL, Hawkins N. C. elegans HAM-1 functions in the nucleus to regulate asymmetric neuroblast division. Dev Biol 2015; 410:56-69. [PMID: 26703426 DOI: 10.1016/j.ydbio.2015.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 01/19/2023]
Abstract
All 302 neurons in the C. elegans hermaphrodite arise through asymmetric division of neuroblasts. During embryogenesis, the C. elegans ham-1 gene is required for several asymmetric neuroblast divisions in lineages that generate both neural and apoptotic cells. By antibody staining, endogenous HAM-1 is found exclusively at the cell cortex in many cells during embryogenesis and is asymmetrically localized in dividing cells. Here we show that in transgenic embryos expressing a functional GFP::HAM-1 fusion protein, GFP expression is also detected in the nucleus, in addition to the cell cortex. Consistent with the nuclear localization is the presence of a putative DNA binding winged-helix domain within the N-terminus of HAM-1. Through a deletion analysis we determined that the C-terminus of the protein is required for nuclear localization and we identified two nuclear localization sequences (NLSs). A subcellular fractionation experiment from wild type embryos, followed by Western blotting, revealed that endogenous HAM-1 is primarily found in the nucleus. Our analysis also showed that the N-terminus is necessary for cortical localization. While ham-1 function is essential for asymmetric division in the lineage that generates the PLM mechanosensory neuron, we showed that cortical localization may not required. Thus, our results suggest that there is a nuclear function for HAM-1 in regulating asymmetric neuroblast division and that the requirement for cortical localization may be lineage dependent.
Collapse
Affiliation(s)
- Amy Leung
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Khang Hua
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | | | - Kyla Hingwing
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Maria Wu
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Pei Luan Koh
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Nancy Hawkins
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
33
|
Chakraborty S, Lambie EJ, Bindu S, Mikeladze-Dvali T, Conradt B. Engulfment pathways promote programmed cell death by enhancing the unequal segregation of apoptotic potential. Nat Commun 2015; 6:10126. [PMID: 26657541 PMCID: PMC4682117 DOI: 10.1038/ncomms10126] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 11/04/2015] [Indexed: 12/14/2022] Open
Abstract
Components of the conserved engulfment pathways promote programmed cell death in Caenorhabditis elegans (C. elegans) through an unknown mechanism. Here we report that the phagocytic receptor CED-1 mEGF10 is required for the formation of a dorsal–ventral gradient of CED-3 caspase activity within the mother of a cell programmed to die and an increase in the level of CED-3 protein within its dying daughter. Furthermore, CED-1 becomes enriched on plasma membrane regions of neighbouring cells that appose the dorsal side of the mother, which later forms the dying daughter. Therefore, we propose that components of the engulfment pathways promote programmed cell death by enhancing the polar localization of apoptotic factors in mothers of cells programmed to die and the unequal segregation of apoptotic potential into dying and surviving daughters. Our findings reveal a novel function of the engulfment pathways and provide a better understanding of how apoptosis is initiated during C. elegans development. Programed cell death occurs in a stereotypic fashion during C. elegans development, and it is thought that engulfment promotes programmed cell death. Here the authors present evidence that a signaling function of the conserved engulfment pathways, not the process of engulfment itself, promotes apoptotic cell death.
Collapse
Affiliation(s)
- Sayantan Chakraborty
- Department of Biology II, Ludwig-Maximilians-University, Munich, Center for Integrated Protein Science Munich-CIPSM, LMU Biocenter, Planegg-Martinsried 82152, Germany
| | - Eric J Lambie
- Department of Biology II, Ludwig-Maximilians-University, Munich, Center for Integrated Protein Science Munich-CIPSM, LMU Biocenter, Planegg-Martinsried 82152, Germany
| | - Samik Bindu
- Department of Biology II, Ludwig-Maximilians-University, Munich, Center for Integrated Protein Science Munich-CIPSM, LMU Biocenter, Planegg-Martinsried 82152, Germany.,Department of Surgery Cardiac &Thoracic Surgery The University of Chicago Biological Sciences, 5841 S. Maryland Ave., Chicago, Illinosis 60637, USA
| | - Tamara Mikeladze-Dvali
- Department of Biology II, Ludwig-Maximilians-University, Munich, Center for Integrated Protein Science Munich-CIPSM, LMU Biocenter, Planegg-Martinsried 82152, Germany
| | - Barbara Conradt
- Department of Biology II, Ludwig-Maximilians-University, Munich, Center for Integrated Protein Science Munich-CIPSM, LMU Biocenter, Planegg-Martinsried 82152, Germany
| |
Collapse
|
34
|
Zhou M, Li Y, Hu Q, Bai XC, Huang W, Yan C, Scheres SHW, Shi Y. Atomic structure of the apoptosome: mechanism of cytochrome c- and dATP-mediated activation of Apaf-1. Genes Dev 2015; 29:2349-61. [PMID: 26543158 PMCID: PMC4691890 DOI: 10.1101/gad.272278.115] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022]
Abstract
In this study, Zhou et al. report the first atomic structure of the mammalian apoptosome, determined at 3.8 Å resolution by cryo-electron microscopy. These findings provide novel insight into how CytC relieves the autoinhibition of Apaf-1 and how dATP triggers Apaf-1 oligomerization. The apoptotic protease-activating factor 1 (Apaf-1) controls the onset of many known forms of intrinsic apoptosis in mammals. Apaf-1 exists in normal cells as an autoinhibited monomer. Upon binding to cytochrome c and dATP, Apaf-1 oligomerizes into a heptameric complex known as the apoptosome, which recruits and activates cell-killing caspases. Here we present an atomic structure of an intact mammalian apoptosome at 3.8 Å resolution, determined by single-particle, cryo-electron microscopy (cryo-EM). Structural analysis, together with structure-guided biochemical characterization, uncovered how cytochrome c releases the autoinhibition of Apaf-1 through specific interactions with the WD40 repeats. Structural comparison with autoinhibited Apaf-1 revealed how dATP binding triggers a set of conformational changes that results in the formation of the apoptosome. Together, these results constitute the molecular mechanism of cytochrome c- and dATP-mediated activation of Apaf-1.
Collapse
Affiliation(s)
- Mengying Zhou
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yini Li
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qi Hu
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiao-Chen Bai
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Weiyun Huang
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chuangye Yan
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Sjors H W Scheres
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, United Kingdom
| | - Yigong Shi
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
35
|
Identifying Regulators of Morphogenesis Common to Vertebrate Neural Tube Closure and Caenorhabditis elegans Gastrulation. Genetics 2015; 202:123-39. [PMID: 26434722 DOI: 10.1534/genetics.115.183137] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
Neural tube defects including spina bifida are common and severe congenital disorders. In mice, mutations in more than 200 genes can result in neural tube defects. We hypothesized that this large gene set might include genes whose homologs contribute to morphogenesis in diverse animals. To test this hypothesis, we screened a set of Caenorhabditis elegans homologs for roles in gastrulation, a topologically similar process to vertebrate neural tube closure. Both C. elegans gastrulation and vertebrate neural tube closure involve the internalization of surface cells, requiring tissue-specific gene regulation, actomyosin-driven apical constriction, and establishment and maintenance of adhesions between specific cells. Our screen identified several neural tube defect gene homologs that are required for gastrulation in C. elegans, including the transcription factor sptf-3. Disruption of sptf-3 in C. elegans reduced the expression of early endodermally expressed genes as well as genes expressed in other early cell lineages, establishing sptf-3 as a key contributor to multiple well-studied C. elegans cell fate specification pathways. We also identified members of the actin regulatory WAVE complex (wve-1, gex-2, gex-3, abi-1, and nuo-3a). Disruption of WAVE complex members reduced the narrowing of endodermal cells' apical surfaces. Although WAVE complex members are expressed broadly in C. elegans, we found that expression of a vertebrate WAVE complex member, nckap1, is enriched in the developing neural tube of Xenopus. We show that nckap1 contributes to neural tube closure in Xenopus. This work identifies in vivo roles for homologs of mammalian neural tube defect genes in two manipulable genetic model systems.
Collapse
|
36
|
Abstract
Cell death is a common and important feature of animal development, and cell death defects underlie many human disease states. The nematode Caenorhabditis elegans has proven fertile ground for uncovering molecular and cellular processes controlling programmed cell death. A core pathway consisting of the conserved proteins EGL-1/BH3-only, CED-9/BCL2, CED-4/APAF1, and CED-3/caspase promotes most cell death in the nematode, and a conserved set of proteins ensures the engulfment and degradation of dying cells. Multiple regulatory pathways control cell death onset in C. elegans, and many reveal similarities with tumor formation pathways in mammals, supporting the idea that cell death plays key roles in malignant progression. Nonetheless, a number of observations suggest that our understanding of developmental cell death in C. elegans is incomplete. The interaction between dying and engulfing cells seems to be more complex than originally appreciated, and it appears that key aspects of cell death initiation are not fully understood. It has also become apparent that the conserved apoptotic pathway is dispensable for the demise of the C. elegans linker cell, leading to the discovery of a previously unexplored gene program promoting cell death. Here, we review studies that formed the foundation of cell death research in C. elegans and describe new observations that expand, and in some cases remodel, this edifice. We raise the possibility that, in some cells, more than one death program may be needed to ensure cell death fidelity.
Collapse
Affiliation(s)
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, USA.
| |
Collapse
|
37
|
Gruber J, Chen CB, Fong S, Ng LF, Teo E, Halliwell B. Caenorhabditis elegans: What We Can and Cannot Learn from Aging Worms. Antioxid Redox Signal 2015; 23:256-79. [PMID: 25544992 DOI: 10.1089/ars.2014.6210] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE The nematode Caenorhabditis elegans is a widely used model organism for research into aging. However, nematodes diverged from other animals between 600 and 1300 million years ago. Beyond the intuitive impression that some aspects of aging appear to be universal, is there evidence that insights into the aging process of nematodes may be applicable to humans? RECENT ADVANCES There have been a number of results in nematodes that appear to contradict long-held beliefs about mechanisms and causes of aging. For example, ablation of several key antioxidant systems has often failed to result in lifespan shortening in C. elegans. CRITICAL ISSUES While it is clear that some central signaling pathways controlling lifespan are broadly conserved across large evolutionary distances, it is less clear to what extent downstream molecular mechanisms of aging are conserved. In this review we discuss the biology of C. elegans and mammals in the context of aging and age-dependent diseases. We consider evidence from studies that attempt to investigate basic, possibly conserved mechanisms of aging especially in the context of the free radical theory of aging. Practical points, such as the need for blinding of lifespan studies and for appropriate biomarkers, are also considered. FUTURE DIRECTIONS As data on the aging process(es) in different organisms increase, it is becoming increasingly clear that there are both conserved (public) and private aspects to aging. It is important to explore the dividing lines between these two aspects and to be aware of the large gray areas in-between.
Collapse
Affiliation(s)
- Jan Gruber
- 1 Department of Biochemistry, National University of Singapore , Singapore, Singapore .,2 Yale-NUS College , Singapore, Singapore
| | - Ce-Belle Chen
- 3 Department of Physics, National University of Singapore , Singapore, Singapore
| | - Sheng Fong
- 4 Duke-NUS Graduate Medical School , Singapore, Singapore
| | - Li Fang Ng
- 1 Department of Biochemistry, National University of Singapore , Singapore, Singapore
| | - Emelyne Teo
- 1 Department of Biochemistry, National University of Singapore , Singapore, Singapore
| | - Barry Halliwell
- 1 Department of Biochemistry, National University of Singapore , Singapore, Singapore
| |
Collapse
|
38
|
Meng L, Mulcahy B, Cook SJ, Neubauer M, Wan A, Jin Y, Yan D. The Cell Death Pathway Regulates Synapse Elimination through Cleavage of Gelsolin in Caenorhabditis elegans Neurons. Cell Rep 2015; 11:1737-48. [PMID: 26074078 DOI: 10.1016/j.celrep.2015.05.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 05/01/2015] [Accepted: 05/15/2015] [Indexed: 11/17/2022] Open
Abstract
Synapse elimination occurs in development, plasticity, and disease. Although the importance of synapse elimination has been documented in many studies, the molecular mechanisms underlying this process are unclear. Here, using the development of C. elegans RME neurons as a model, we have uncovered a function for the apoptosis pathway in synapse elimination. We find that the conserved apoptotic cell death (CED) pathway and axonal mitochondria are required for the elimination of transiently formed clusters of presynaptic components in RME neurons. This function of the CED pathway involves the activation of the actin-filament-severing protein, GSNL-1. Furthermore, we show that caspase CED-3 cleaves GSNL-1 at a conserved C-terminal region and that the cleaved active form of GSNL-1 promotes its actin-severing ability. Our data suggest that activation of the CED pathway contributes to selective elimination of synapses through disassembly of the actin filament network.
Collapse
Affiliation(s)
- Lingfeng Meng
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Research Drive, Durham, NC 27710, USA
| | - Ben Mulcahy
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON M5G 1X5, Canada
| | - Steven J Cook
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marianna Neubauer
- Department of Physics and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Airong Wan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Research Drive, Durham, NC 27710, USA
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Dong Yan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Research Drive, Durham, NC 27710, USA; Department of Neurobiology and Duke Institute for Brain Sciences, Duke University Medical Center, Research Drive, Durham, NC 27710, USA.
| |
Collapse
|
39
|
Abstract
A little over 50 years ago, Sydney Brenner had the foresight to develop the nematode (round worm) Caenorhabditis elegans as a genetic model for understanding questions of developmental biology and neurobiology. Over time, research on C. elegans has expanded to explore a wealth of diverse areas in modern biology including studies of the basic functions and interactions of eukaryotic cells, host-parasite interactions, and evolution. C. elegans has also become an important organism in which to study processes that go awry in human diseases. This primer introduces the organism and the many features that make it an outstanding experimental system, including its small size, rapid life cycle, transparency, and well-annotated genome. We survey the basic anatomical features, common technical approaches, and important discoveries in C. elegans research. Key to studying C. elegans has been the ability to address biological problems genetically, using both forward and reverse genetics, both at the level of the entire organism and at the level of the single, identified cell. These possibilities make C. elegans useful not only in research laboratories, but also in the classroom where it can be used to excite students who actually can see what is happening inside live cells and tissues.
Collapse
Affiliation(s)
- Ann K Corsi
- Biology Department, The Catholic University of America, Washington, DC 20064
| | - Bruce Wightman
- Biology Department, Muhlenberg College, Allentown, Pennsylvania 18104
| | - Martin Chalfie
- Department of Biological Sciences, Columbia University, New York, New York 10027
| |
Collapse
|
40
|
Wang J, Chitturi J, Ge Q, Laskova V, Wang W, Li X, Ding M, Zhen M, Huang X. The C. elegans COE transcription factor UNC-3 activates lineage-specific apoptosis and affects neurite growth in the RID lineage. Development 2015; 142:1447-57. [PMID: 25790851 DOI: 10.1242/dev.119479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/17/2015] [Indexed: 12/23/2022]
Abstract
Mechanisms that regulate apoptosis in a temporal and lineage-specific manner remain poorly understood. The COE (Collier/Olf/EBF) transcription factors have been implicated in the development of many cell types, including neurons. Here, we show that the sole Caenorhabditis elegans COE protein, UNC-3, together with a histone acetyltransferase, CBP-1/P300, specifies lineage-specific apoptosis and certain aspects of neurite trajectory. During embryogenesis, the RID progenitor cell gives rise to the RID neuron and RID sister cell; the latter undergoes apoptosis shortly after cell division upon expression of the pro-apoptotic gene egl-1. We observe UNC-3 expression in the RID progenitor, and the absence of UNC-3 results in the failure of the RID lineage to express a Pegl-1::GFP reporter and in the survival of the RID sister cell. Lastly, UNC-3 interacts with CBP-1, and cbp-1 mutants exhibit a similar RID phenotype to unc-3. Thus, in addition to playing a role in neuronal terminal differentiation, UNC-3 is a cell lineage-specific regulator of apoptosis.
Collapse
Affiliation(s)
- Jinbo Wang
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jyothsna Chitturi
- Lunenfeld and Tanebaum Research Institute, University of Toronto, Toronto, Ontario, Canada M5G 1X5
| | - Qinglan Ge
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Valeriya Laskova
- Lunenfeld and Tanebaum Research Institute, University of Toronto, Toronto, Ontario, Canada M5G 1X5
| | - Wei Wang
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xia Li
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Ding
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Mei Zhen
- Lunenfeld and Tanebaum Research Institute, University of Toronto, Toronto, Ontario, Canada M5G 1X5
| | - Xun Huang
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
41
|
Teng X, Hardwick JM. Cell death in genome evolution. Semin Cell Dev Biol 2015; 39:3-11. [PMID: 25725369 PMCID: PMC4410082 DOI: 10.1016/j.semcdb.2015.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 12/23/2022]
Abstract
Inappropriate survival of abnormal cells underlies tumorigenesis. Most discoveries about programmed cell death have come from studying model organisms. Revisiting the experimental contexts that inspired these discoveries helps explain confounding biases that inevitably accompany such discoveries. Amending early biases has added a newcomer to the collection of cell death models. Analysis of gene-dependent death in yeast revealed the surprising influence of single gene mutations on subsequent eukaryotic genome evolution. Similar events may influence the selection for mutations during early tumorigenesis. The possibility that any early random mutation might drive the selection for a cancer driver mutation is conceivable but difficult to demonstrate. This was tested in yeast, revealing that mutation of almost any gene appears to specify the selection for a new second mutation. Some human tumors contain pairs of mutant genes homologous to co-occurring mutant genes in yeast. Here we consider how yeast again provide novel insights into tumorigenesis.
Collapse
Affiliation(s)
- Xinchen Teng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, PR China; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - J Marie Hardwick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
Guanine nucleotide exchange factor OSG-1 confers functional aging via dysregulated Rho signaling in Caenorhabditis elegans neurons. Genetics 2014; 199:487-96. [PMID: 25527286 DOI: 10.1534/genetics.114.173500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Rho signaling regulates a variety of biological processes, but whether it is implicated in aging remains an open question. Here we show that a guanine nucleotide exchange factor of the Dbl family, OSG-1, confers functional aging by dysregulating Rho GTPases activities in C. elegans. Thus, gene reporter analysis revealed widespread OSG-1 expression in muscle and neurons. Loss of OSG-1 gene function was not associated with developmental defects. In contrast, suppression of OSG-1 lessened loss of function (chemotaxis) in ASE sensory neurons subjected to conditions of oxidative stress generated during natural aging, by oxidative challenges, or by genetic mutations. RNAi analysis showed that OSG-1 was specific toward activation of RHO-1 GTPase signaling. RNAi further implicated actin-binding proteins ARX-3 and ARX-5, thus the actin cytoskeleton, as one of the targets of OSG-1/RHO-1 signaling. Taken together these data suggest that OSG-1 is recruited under conditions of oxidative stress, a hallmark of aging, and contributes to promote loss of neuronal function by affecting the actin cytoskeleton via altered RHO-1 activity.
Collapse
|
43
|
Levi-Ferber M, Salzberg Y, Safra M, Haviv-Chesner A, Bülow HE, Henis-Korenblit S. It's all in your mind: determining germ cell fate by neuronal IRE-1 in C. elegans. PLoS Genet 2014; 10:e1004747. [PMID: 25340700 PMCID: PMC4207656 DOI: 10.1371/journal.pgen.1004747] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 09/11/2014] [Indexed: 01/26/2023] Open
Abstract
The C. elegans germline is pluripotent and mitotic, similar to self-renewing mammalian tissues. Apoptosis is triggered as part of the normal oogenesis program, and is increased in response to various stresses. Here, we examined the effect of endoplasmic reticulum (ER) stress on apoptosis in the C. elegans germline. We demonstrate that pharmacological or genetic induction of ER stress enhances germline apoptosis. This process is mediated by the ER stress response sensor IRE-1, but is independent of its canonical downstream target XBP-1. We further demonstrate that ire-1-dependent apoptosis in the germline requires both CEP-1/p53 and the same canonical apoptotic genes as DNA damage-induced germline apoptosis. Strikingly, we find that activation of ire-1, specifically in the ASI neurons, but not in germ cells, is sufficient to induce apoptosis in the germline. This implies that ER stress related germline apoptosis can be determined at the organism level, and is a result of active IRE-1 signaling in neurons. Altogether, our findings uncover ire-1 as a novel cell non-autonomous regulator of germ cell apoptosis, linking ER homeostasis in sensory neurons and germ cell fate.
Collapse
Affiliation(s)
- Mor Levi-Ferber
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Yehuda Salzberg
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, New York, United States of America
| | - Modi Safra
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Anat Haviv-Chesner
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, New York, United States of America
| | - Sivan Henis-Korenblit
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| |
Collapse
|
44
|
Lant B, Derry WB. Analysis of apoptosis in Caenorhabditis elegans. Cold Spring Harb Protoc 2014; 2014:2014/5/pdb.top070458. [PMID: 24786497 DOI: 10.1101/pdb.top070458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The nematode worm Caenorhabditis elegans has provided researchers with a wealth of information on the molecular mechanisms controlling programmed cell death (apoptosis). Its genetic tractability, optical clarity, and relatively short lifespan are key advantages for rapid assessment of apoptosis in vivo. The use of forward and reverse genetics methodology, coupled with in vivo imaging, has provided deep insights into how a multicellular organism orchestrates the self-destruction of specific cells during development and in response to exogenous stresses. Strains of C. elegans carrying mutations in the core elements of the apoptotic pathway, or in tissue-specific regulators of apoptosis, can be used for genetic analyses to reveal conserved mechanisms by which apoptosis is regulated in the somatic and reproductive (germline) tissue. Here we present an introduction to the study of apoptosis in C. elegans, including current techniques for visualization, analysis, and screening.
Collapse
Affiliation(s)
- Benjamin Lant
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | | |
Collapse
|
45
|
Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol 2014; 15:49-63. [PMID: 24355989 DOI: 10.1038/nrm3722] [Citation(s) in RCA: 2214] [Impact Index Per Article: 221.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The BCL-2 protein family determines the commitment of cells to apoptosis, an ancient cell suicide programme that is essential for development, tissue homeostasis and immunity. Too little apoptosis can promote cancer and autoimmune diseases; too much apoptosis can augment ischaemic conditions and drive neurodegeneration. We discuss the biochemical, structural and genetic studies that have clarified how the interplay between members of the BCL-2 family on mitochondria sets the apoptotic threshold. These mechanistic insights into the functions of the BCL-2 family are illuminating the physiological control of apoptosis, the pathological consequences of its dysregulation and the promising search for novel cancer therapies that target the BCL-2 family.
Collapse
|
46
|
Chai J, Shi Y. Apoptosome and inflammasome: conserved machineries for caspase activation. Natl Sci Rev 2014. [DOI: 10.1093/nsr/nwt025] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Apoptosome and inflammasome are multimeric protein complexes that mediate the activation of specific caspases at the onset of apoptosis and inflammation. The central component of apoptosome or inflammasome is a tripartite scaffold protein, exemplified by Apaf-1 and NLRC4, which contains an amino-terminal homotypic interaction motif, a central nucleotide-binding oligomerization domain and a carboxyl-terminal ligand-sensing domain. In the absence of death cue or an inflammatory signal, Apaf-1 or NLRC4 exists in an auto-inhibited, monomeric state, which is stabilized by adenosine diphosphate (ADP). Binding to an apoptosis- or inflammation-inducing ligand, together with replacement of ADP by adenosine triphosphate (ATP), results in the formation of a multimeric apoptosome or inflammasome. The assembled apoptosome and inflammasome serve as dedicated machineries to facilitate the activation of specific caspases. In this review, we describe the structure and functional mechanisms of mammalian inflammasome and apoptosomes from three representative organisms. Emphasis is placed on the molecular mechanism of caspase activation and the shared features of apoptosomes and inflammasomes.
Collapse
Affiliation(s)
- Jijie Chai
- Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yigong Shi
- Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
47
|
Concepts of tissue injury and cell death in inflammation: a historical perspective. Nat Rev Immunol 2013; 14:51-9. [PMID: 24336099 DOI: 10.1038/nri3561] [Citation(s) in RCA: 265] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Emerging evidence indicates that the molecular mechanisms of cell death have regulatory roles in inflammation and that the molecular changes that are associated with different forms of cell death affect the course of inflammation in different ways. In this Timeline article, we discuss how our understanding of the mechanisms and functional roles of tissue injury and cell death in inflammation has evolved on the basis of almost two centuries of study. We describe how such ideas have led to our current models of cell death and inflammation, and we highlight the remaining gaps in our knowledge of the subject.
Collapse
|
48
|
Denton D, Aung-Htut MT, Kumar S. Developmentally programmed cell death in Drosophila. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3499-3506. [DOI: 10.1016/j.bbamcr.2013.06.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/16/2013] [Indexed: 12/24/2022]
|
49
|
Huang W, Jiang T, Choi W, Qi S, Pang Y, Hu Q, Xu Y, Gong X, Jeffrey PD, Wang J, Shi Y. Mechanistic insights into CED-4-mediated activation of CED-3. Genes Dev 2013; 27:2039-48. [PMID: 24065769 PMCID: PMC3792479 DOI: 10.1101/gad.224428.113] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Programmed cell death in C. elegans requires caspase CED-3 activation. The CED-4 apoptosome facilitates CED-3 activation, yet how CED-4 recognizes CED-3 is unknown. Huang et al. now show that CED-3 directly binds CED-4 and plays a pivotal role in forming an active CED-4–CED-3 holoenzyme. Structural and biochemical analyses suggest a model whereby two CED-3 molecules are forced into the CED-4 apoptosome, which then undergoes dimerization and autocatalytic maturation. This work provides a major revision of the prevailing model for initiator caspase activation. Programmed cell death in Caenorhabditis elegans requires activation of the caspase CED-3, which strictly depends on CED-4. CED-4 forms an octameric apoptosome, which binds the CED-3 zymogen and facilitates its autocatalytic maturation. Despite recent advances, major questions remain unanswered. Importantly, how CED-4 recognizes CED-3 and how such binding facilitates CED-3 activation remain completely unknown. Here we demonstrate that the L2′ loop of CED-3 directly binds CED-4 and plays a major role in the formation of an active CED-4–CED-3 holoenzyme. The crystal structure of the CED-4 apoptosome bound to the L2′ loop fragment of CED-3, determined at 3.2 Å resolution, reveals specific interactions between a stretch of five hydrophobic amino acids from CED-3 and a shallow surface pocket within the hutch of the funnel-shaped CED-4 apoptosome. Structure-guided biochemical analysis confirms the functional importance of the observed CED-4–CED-3 interface. Structural analysis together with published evidence strongly suggest a working model in which two molecules of CED-3 zymogen, through specific recognition, are forced into the hutch of the CED-4 apoptosome, consequently undergoing dimerization and autocatalytic maturation. The mechanism of CED-3 activation represents a major revision of the prevailing model for initiator caspase activation.
Collapse
Affiliation(s)
- Weijiao Huang
- Ministry of Education Protein Science Laboratory, Center for Structural Biology, School of Life Sciences
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Reina CP, Driscoll M, Gabel CV. Neuronal repair: Apoptotic proteins make good. WORM 2013; 2:e22285. [PMID: 24058867 PMCID: PMC3704441 DOI: 10.4161/worm.22285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 09/18/2012] [Indexed: 11/25/2022]
Abstract
The potential of the central nervous system (CNS) to regenerate is regulated by a complex interaction of neuronal intrinsic and extrinsic factors that remain poorly understood. Significant research has been dedicated to identifying these factors to facilitate design of therapies that will treat the functional impairment associated with CNS injuries. Over the last decade, the development of in vivo laser severing of single axons in C. elegans has established an invaluable model for the genetic identification of novel regeneration factors. In a recent study we report the unexpected identification of the core apoptotic proteins CED-4/Apaf-1 and the executioner caspase CED-3 as important factors that promote early events in regeneration in C. elegans. Other upstream regulators of apoptosis do not influence regeneration, indicating the existence of a novel mechanism for activation of CED-4 and CED-3 in neuronal repair. CED-4 and CED-3 function downstream of injury-induced calcium transients and appear to act through the conserved DLK-1 pathway to promote regeneration. We propose a working model for calcium-dependent localized activation of CED-4 and CED-3 caspase and discuss questions raised including mechanisms for spatially regulating activated CED-3 and the possible substrates that it might cleave to initiate regeneration.
Collapse
Affiliation(s)
- Christopher P Reina
- Department of Molecular Biology and Biochemistry; Rutgers University; Piscataway, NJ USA
| | | | | |
Collapse
|