1
|
Wibisono S, Wibisono P, Chen CH, Sun J, Liu Y. The Caenorhabditis elegans neuronal GPCR OCTR-1 modulates longevity responses to both warm and cold temperatures. iScience 2025; 28:112279. [PMID: 40264795 PMCID: PMC12013480 DOI: 10.1016/j.isci.2025.112279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 01/27/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Many animal species live longer in cold climates than in warm climates, which was traditionally explained using the rate of living theory, i.e., higher temperatures increase chemical reaction rates, thus speeding up the aging process. However, recent studies have identified specific molecules and cells that are involved in longevity responses to temperature, indicating that such responses are not simply thermodynamic but are regulated processes. Here, we report that Caenorhabditis elegans lacking the neuronal G protein-coupled receptor OCTR-1 have extended lifespans at a warm temperature but shortened lifespans at a cold temperature, demonstrating that OCTR-1 modulates temperature-induced longevity responses. These responses are regulated by the OCTR-1-expressing, chemosensory ASH neurons. Furthermore, the OCTR-1 pathway controls such responses to warm and cold temperatures by regulating the expressions of immune response genes and the intestinal transcriptional factor ELT-2, respectively. Overall, our study provides cellular and molecular insights into the relationship between temperature and longevity.
Collapse
Affiliation(s)
- Shawndra Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Chia-Hui Chen
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
- Genomics Core, Washington State University, Spokane, WA, USA
| |
Collapse
|
2
|
Wibisono P, Liu Y, Roberts KP, Baluya D, Sun J. Neuronal GPCR NMUR-1 regulates energy homeostasis in response to pathogen infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602733. [PMID: 39026696 PMCID: PMC11257582 DOI: 10.1101/2024.07.09.602733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
A key question in current immunology is how the innate immune system generates high levels of specificity. Our previous study in Caenorhabditis elegans revealed that NMUR-1, a neuronal G protein-coupled receptor homologous to mammalian receptors for the neuropeptide neuromedin U (NMU), regulates distinct innate immune responses to different bacterial pathogens. Here, by using quantitative proteomics and functional assays, we discovered that NMUR-1 regulates F1FO ATP synthase and ATP production in response to pathogen infection, and that such regulation contributes to NMUR-1-mediated specificity of innate immunity. We further demonstrated that ATP biosynthesis and its contribution to defense is neurally controlled by the NMUR-1 ligand CAPA-1 and its expressing neurons ASG. These findings indicate that NMUR-1 neural signaling regulates the specificity of innate immunity by controlling energy homeostasis as part of defense against pathogens. Our study provides mechanistic insights into the emerging roles of NMU signaling in immunity across animal phyla.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
- Genomics Core, Washington State University, Spokane, WA, USA
| | - Kenneth P Roberts
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Dodge Baluya
- Tissue Imaging, Metabolomics and Proteomics Laboratory, Washington State University, Pullman, WA, USA
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
3
|
Otarigho B, Butts AF, Aballay A. Neuronal NPR-15 modulates molecular and behavioral immune responses via the amphid sensory neuron-intestinal axis in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.27.550570. [PMID: 37546751 PMCID: PMC10402133 DOI: 10.1101/2023.07.27.550570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The survival of hosts during infections relies on their ability to mount effective molecular and behavioral immune responses. Despite extensive research on these defense strategies in various species, including the model organism Caenorhabditis elegans, the neural mechanisms underlying their interaction remain poorly understood. Previous studies have highlighted the role of neural G protein-coupled receptors (GPCRs) in regulating both immunity and pathogen avoidance, which is particularly dependent on aerotaxis. To address this knowledge gap, we conducted a screen of mutants in neuropeptide receptor family genes. We found that loss-of-function mutations in npr-15 activated immunity while suppressing pathogen avoidance behavior. Through further analysis, NPR-15 was found to regulate immunity by modulating the activity of key transcription factors, namely GATA/ELT-2 and TFEB/HLH-30. Surprisingly, the lack of pathogen avoidance of npr-15 mutant animals was not influenced by oxygen levels. Moreover, our studies revealed that the amphid sensory neuron ASJ is involved in mediating the immune and behavioral responses orchestrated by NPR-15. Additionally, NPR-15 was found to regulate avoidance behavior via the TRPM gene, GON-2, which may sense the intestinal distension caused by bacterial colonization to elicit pathogen avoidance. Our study contributes to a broader understanding of host defense strategies and mechanisms underlining the interaction between molecular and behavioral immune responses.
Collapse
Affiliation(s)
- Benson Otarigho
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anna Frances Butts
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alejandro Aballay
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, TX
| |
Collapse
|
4
|
Xiao Y, Liu F, Wu Q, Zhu X, Yu C, Jiang N, Li S, Liu Y. Dioscin Activates Endoplasmic Reticulum Unfolded Protein Response for Defense Against Pathogenic Bacteria in Caenorhabditis elegans via IRE-1/XBP-1 Pathway. J Infect Dis 2024; 229:237-244. [PMID: 37499184 DOI: 10.1093/infdis/jiad294] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023] Open
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved pathway that senses and responds to the accumulation of misfolded proteins in the endoplasmic reticulum (ER) lumen during bacterial infection. The IRE-1/XBP-1 pathway is a major branch of the UPRER that has been conserved from yeast to human. Dioscin, a steroidal saponin exhibits a broad spectrum of properties. However, whether dioscin influences the immune response and the underlying molecular mechanisms remain obscure. We find that dioscin increases resistance to Gram-negative pathogen Pseudomonas aeruginosa. Furthermore, dioscin also inhibits the growth of pathogenic bacteria. Meanwhile, dioscin enhances the resistance to pathogens by reducing bacterial burden in the intestine. Through genetic screening, we find that dioscin activates the UPRER to promote innate immunity via IRE-1/XBP-1 pathway. Intriguingly, dioscin requires the neural XBP-1 for immune response. Our findings suggest that dioscin may be a viable candidate for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Liu
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qinyi Wu
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xinting Zhu
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Changyan Yu
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nian Jiang
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Sanhua Li
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun Liu
- Guizhou Provincial College-Based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou, China
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- School of Forensic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
5
|
Wibisono P, Sun J. Pathogen infection induces specific transgenerational modifications to gene expression and fitness in Caenorhabditis elegans. Front Physiol 2023; 14:1225858. [PMID: 37811492 PMCID: PMC10556243 DOI: 10.3389/fphys.2023.1225858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
How pathogen infection in a parental generation affects response in future generations to the same pathogen via epigenetic modifications has been the topic of recent studies. These studies focused on changes attributed to transgenerational epigenetic inheritance and how these changes cause an observable difference in behavior or immune response in a population. However, we questioned if pathogen infection causes hidden epigenetic changes to fitness that are not observable at the population level. Using the nematode Caenorhabditis elegans as a model organism, we examined the generation-to-generation differences in survival of both an unexposed and primed lineage of animals against a human opportunistic pathogen Salmonella enterica. We discovered that training a lineage of C. elegans against a specific pathogen does not cause a significant change to overall survival, but rather narrows survival variability between generations. Quantification of gene expression revealed reduced variation of a specific member of the TFEB lipophagic pathway. We also provided the first report of a repeating pattern of survival times over the course of 12 generations in the control lineage of C. elegans. This repeating pattern indicates that the variability in survival between generations of the control lineage is not random but may be regulated by unknown mechanisms. Overall, our study indicates that pathogen infection can cause specific phenotypic changes due to epigenetic modifications, and a possible system of epigenetic regulation between generations.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| |
Collapse
|
6
|
Watt NT, McGrane A, Roberts LD. Linking the unfolded protein response to bioactive lipid metabolism and signalling in the cell non-autonomous extracellular communication of ER stress. Bioessays 2023; 45:e2300029. [PMID: 37183938 PMCID: PMC11475223 DOI: 10.1002/bies.202300029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/16/2023]
Abstract
The endoplasmic reticulum (ER) organelle is the key intracellular site of both protein and lipid biosynthesis. ER dysfunction, termed ER stress, can result in protein accretion within the ER and cell death; a pathophysiological process contributing to a range of metabolic diseases and cancers. ER stress leads to the activation of a protective signalling cascade termed the Unfolded Protein Response (UPR). However, chronic UPR activation can ultimately result in cellular apoptosis. Emerging evidence suggests that cells undergoing ER stress and UPR activation can release extracellular signals that can propagate UPR activation to target tissues in a cell non-autonomous signalling mechanism. Separately, studies have determined that the UPR plays a key regulatory role in the biosynthesis of bioactive signalling lipids including sphingolipids and ceramides. Here we weigh the evidence to combine these concepts and propose that during ER stress, UPR activation drives the biosynthesis of ceramide lipids, which are exported and function as cell non-autonomous signals to propagate UPR activation in target cells and tissues.
Collapse
Affiliation(s)
- Nicole T. Watt
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Anna McGrane
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| | - Lee D. Roberts
- Leeds Institute of Cardiovascular and Metabolic MedicineUniversity of LeedsLeedsUK
| |
Collapse
|
7
|
van Oosten-Hawle P. Exploiting inter-tissue stress signaling mechanisms to preserve organismal proteostasis during aging. Front Physiol 2023; 14:1228490. [PMID: 37469564 PMCID: PMC10352849 DOI: 10.3389/fphys.2023.1228490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023] Open
Abstract
Aging results in a decline of cellular proteostasis capacity which culminates in the accumulation of phototoxic material, causing the onset of age-related maladies and ultimately cell death. Mechanisms that regulate proteostasis such as cellular stress response pathways sense disturbances in the proteome. They are activated to increase the expression of protein quality control components that counteract cellular damage. Utilizing invertebrate model organisms such as Caenorhabditis elegans, it has become increasingly evident that the regulation of proteostasis and the activation of cellular stress responses is not a cell autonomous process. In animals, stress responses are orchestrated by signals coming from other tissues, including the nervous system, the intestine and the germline that have a profound impact on determining the aging process. Genetic pathways discovered in C. elegans that facilitate cell nonautonomous regulation of stress responses are providing an exciting feeding ground for new interventions. In this review I will discuss cell nonautonomous proteostasis mechanisms and their impact on aging as well as ongoing research and clinical trials that can increase organismal proteostasis to lengthen health- and lifespan.
Collapse
|
8
|
Wang AJ, Wibisono P, Geppert BM, Liu Y. Using single-worm RNA sequencing to study C. elegans responses to pathogen infection. BMC Genomics 2022; 23:653. [PMID: 36104659 PMCID: PMC9472404 DOI: 10.1186/s12864-022-08878-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background Caenorhabditis elegans is an excellent research model whose populations have been used in many studies to address various biological questions. Although worm-to-worm phenotypic variations in isogenic populations have been persistently observed, they are not well understood and are often ignored or averaged out in studies, masking the impacts of such variations on data collection and interpretation. Single-worm RNA sequencing that profiles the transcriptomes of individual animals has the power to examine differences between individuals in a worm population, but this approach has been understudied. The integrity of the starting RNA, the quality of the library and sequence data, as well as the transcriptome-profiling effectiveness of single-worm RNA-seq remain unclear. Therefore, more studies are needed to improve this technique and its application in research. Results In this study, we aimed to develop a single-worm RNA-seq method that includes five steps: worm lysis and RNA extraction, cDNA synthesis, library preparation, sequencing, and sequence data analysis. We found that the mechanical lysis of worms using a Qiagen TissueLyser maintained RNA integrity and determined that the quality of our single-worm libraries was comparable to that of standard RNA-seq libraries based on assessments of a variety of parameters. Furthermore, analysis of pathogen infection-induced gene expression using single-worm RNA-seq identified a core set of genes and biological processes relating to the immune response and metabolism affected by infection. These results demonstrate the effectiveness of our single-worm RNA-seq method in transcriptome profiling and its usefulness in addressing biological questions. Conclusions We have developed a single-worm RNA-seq method to effectively profile gene expression in individual C. elegans and have applied this method to study C. elegans responses to pathogen infection. Key aspects of our single-worm RNA-seq libraries were comparable to those of standard RNA-seq libraries. The single-worm method captured the core set of, but not all, infection-affected genes and biological processes revealed by the standard method, indicating that there was gene regulation that is not shared by all individuals in a population. Our study suggests that combining single-worm and standard RNA-seq approaches will allow for detecting and distinguishing shared and individual-specific gene activities in isogenic populations. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08878-x.
Collapse
|
9
|
Ferreira JV, da Rosa Soares A, Pereira P. Cell Non-autonomous Proteostasis Regulation in Aging and Disease. Front Neurosci 2022; 16:878296. [PMID: 35757551 PMCID: PMC9220288 DOI: 10.3389/fnins.2022.878296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Aging is a risk factor for a number of diseases, being the more notorious ones perhaps neurodegenerative diseases such as Alzheimer's and Parkinson's. These and other age-related pathologies are often associated with accumulation of proteotoxic material inside cells, as well as with the accumulation of protein deposits extracellularly. It is widely accepted that this accumulation of toxic proteins trails a progressive decline in the mechanisms that regulate protein homeostasis, or proteostasis, during aging. However, despite significant efforts, the progress in terms of novel or improved therapies targeting accumulation of proteotoxic material has been rather limited. For example, clinical trials for new drugs aimed at treating Alzheimer's disease, by preventing accumulation of toxic proteins, have notoriously failed. On the other hand, it is becoming increasingly apparent that regulation of proteostasis is not a cell autonomous process. In fact, cells rely on complex transcellular networks to maintain tissue and organ homeostasis involving endocrine and paracrine signaling pathways. In this review we will discuss the impact of cell non-autonomous proteostasis mechanisms and their impact in aging and disease. We will focus on how transcellular proteostasis networks can shed new light into stablished paradigms about the aging of organisms.
Collapse
Affiliation(s)
- Joao Vasco Ferreira
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana da Rosa Soares
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Paulo Pereira
- Proteostasis and Intercellular Communication Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
Sang Y, Ren J, Aballay A. The transcription factor HLH-26 controls probiotic-mediated protection against intestinal infection through up-regulation of the Wnt/BAR-1 pathway. PLoS Biol 2022; 20:e3001581. [PMID: 35263319 PMCID: PMC8936500 DOI: 10.1371/journal.pbio.3001581] [Citation(s) in RCA: 7] [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: 07/17/2021] [Revised: 03/21/2022] [Accepted: 02/23/2022] [Indexed: 12/31/2022] Open
Abstract
Probiotics play a critical role in the control of host intestinal microbial balance, protecting the host from gastrointestinal pathogens, modulating the host immune response, and decreasing host susceptibility to infection. To understand the mechanism underlying the protective effect of probiotics against infections through immune regulation, we examined protection against Salmonella enterica infection following exposure to nonpathogenic Enterococcus faecium in the nematode Caenorhabditis elegans. We found that the transcription factor HLH-26, a REF-1 family member of basic helix-loop-helix transcription factors, was required in the intestine for E. faecium-mediated protection of C. elegans against a lethal S. enterica infection. In addition, we uncovered that defense response genes controlled by the canonical Wnt/BAR-1 pathway were activated upon exposure to E. faecium in an HLH-26-dependent manner. Our findings highlight a role for REF-1/HLH-26 in the control of the Wnt/BAR-1 pathway in probiotic-mediated protection against gut infection.
Collapse
Affiliation(s)
- Yu Sang
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jie Ren
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Alejandro Aballay
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
11
|
Wibisono P, Wibisono S, Watteyne J, Chen CH, Sellegounder D, Beets I, Liu Y, Sun J. Neuronal GPCR NMUR-1 regulates distinct immune responses to different pathogens. Cell Rep 2022; 38:110321. [PMID: 35139379 PMCID: PMC8869846 DOI: 10.1016/j.celrep.2022.110321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/21/2021] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
A key question in current immunology is how the innate immune system generates high levels of specificity. Using the Caenorhabditis elegans model system, we demonstrate that functional loss of NMUR-1, a neuronal G-protein-coupled receptor homologous to mammalian receptors for the neuropeptide neuromedin U, has diverse effects on C. elegans innate immunity against various bacterial pathogens. Transcriptomic analyses and functional assays reveal that NMUR-1 modulates C. elegans transcription activity by regulating the expression of transcription factors involved in binding to RNA polymerase II regulatory regions, which, in turn, controls the expression of distinct immune genes in response to different pathogens. These results uncover a molecular basis for the specificity of C. elegans innate immunity. Given the evolutionary conservation of NMUR-1 signaling in immune regulation across multicellular organisms, our study could provide mechanistic insights into understanding the specificity of innate immunity in other animals, including mammals.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Shawndra Wibisono
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Jan Watteyne
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Chia-Hui Chen
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Durai Sellegounder
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Isabel Beets
- Department of Biology, KU Leuven, Leuven, Belgium
| | - Yiyong Liu
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA; Genomics Core, Washington State University, Spokane, WA, USA.
| | - Jingru Sun
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA.
| |
Collapse
|
12
|
The regulation of animal behavior by cellular stress responses. Exp Cell Res 2021; 405:112720. [PMID: 34217715 PMCID: PMC8363813 DOI: 10.1016/j.yexcr.2021.112720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/18/2021] [Accepted: 06/27/2021] [Indexed: 01/18/2023]
Abstract
Cellular stress responses exist to detect the effects of stress on cells, and to activate protective mechanisms that promote resilience. As well as acting at the cellular level, stress response pathways can also regulate whole organism responses to stress. One way in which animals facilitate their survival in stressful environments is through behavioral adaptation; this review considers the evidence that activation of cellular stress responses plays an important role in mediating the changes to behavior that promote organismal survival upon stress.
Collapse
|
13
|
Wibisono P, Sun J. Neuro-immune communication in C. elegans defense against pathogen infection. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:60-65. [PMID: 34368754 PMCID: PMC8344176 DOI: 10.1016/j.crimmu.2021.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The innate immune system is a complex collection of physical barriers and physiological defense responses to internal and external environmental assaults. Recent studies in the model organism Caenorhabditis elegans have highlighted how the nervous system interacts with the innate immune system to generate coordinated protective responses. Indeed, studies on neuro-immune interaction pathways have provided mechanistic insights into the roles of neuro-immune communication in modulating both immune and behavioral responses to pathogen attacks. The nervous system releases a variety of neurotransmitters, peptides, and hormones that regulate the innate immune response, while the innate immune system also relays information to the nervous system to affect learning and behavioral responses. Although these interactions still need further investigation, the knowledge that we have gained thus far has improved our understanding of how separate biological systems can act collectively for the survival and well-being of an organism. Here, we review recent studies on neuro-immune communication related to the survival and defense of C. elegans against pathogens. Neuro-immune communication is essential for generating coordinated defense against pathogen infection to improve host survival. Neuro-immune communication modulates both immune and behavioral responses to pathogen attacks. C.elegans is an excellent model system for studying neuro-immune communication.
Collapse
Affiliation(s)
- Phillip Wibisono
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Jingru Sun
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
14
|
Abstract
Although Caenorhabditis elegans has been used as a model host for studying host-pathogen interactions for more than 20 years, the mechanisms by which it identifies pathogens are not well understood. This is largely due to its lack of most known pattern recognition receptors (PRRs) that recognize pathogen-derived molecules. Recent behavioral research in C. elegans indicates that its nervous system plays a major role in microbe sensing. With the increasing integration of neurobiology in immunological research, future studies may find that neuronal detection of pathogens is an integral part of C. elegans-pathogen interactions. Similar to that of mammals, the C. elegans nervous system regulates its immune system to maintain immunological homeostasis. Studies in the nematode have revealed unprecedented details regarding the molecules, cells, and signaling pathways involved in neural regulation of immunity. Notably, some of the studies indicate that some neuroimmune regulatory circuits need not be "activated" by pathogen infection because they are tonically active and that there could be a predetermined set point for internal immunity, around which the nervous system adjusts immune responses to internal or external environmental changes. Here, we review recent progress on the roles of the C. elegans nervous system in pathogen detection and immune regulation. Because of its advantageous characteristics, we expect that the C. elegans model will be critical for deciphering complex neuroimmune signaling mechanisms that integrate and process multiple sensory cues.
Collapse
Affiliation(s)
- Yiyong Liu
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
- Genomics Core, Washington State University, Spokane, Washington, USA
| | - Jingru Sun
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| |
Collapse
|
15
|
Joshi KK, Matlack TL, Pyonteck S, Vora M, Menzel R, Rongo C. Biogenic amine neurotransmitters promote eicosanoid production and protein homeostasis. EMBO Rep 2021; 22:e51063. [PMID: 33470040 PMCID: PMC7926251 DOI: 10.15252/embr.202051063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022] Open
Abstract
Metazoans use protein homeostasis (proteostasis) pathways to respond to adverse physiological conditions, changing environment, and aging. The nervous system regulates proteostasis in different tissues, but the mechanism is not understood. Here, we show that Caenorhabditis elegans employs biogenic amine neurotransmitters to regulate ubiquitin proteasome system (UPS) proteostasis in epithelia. Mutants for biogenic amine synthesis show decreased poly-ubiquitination and turnover of a GFP-based UPS substrate. Using RNA-seq and mass spectrometry, we found that biogenic amines promote eicosanoid production from poly-unsaturated fats (PUFAs) by regulating expression of cytochrome P450 monooxygenases. Mutants for one of these P450s share the same UPS phenotype observed in biogenic amine mutants. The production of n-6 eicosanoids is required for UPS substrate turnover, whereas accumulation of n-6 eicosanoids accelerates turnover. Our results suggest that sensory neurons secrete biogenic amines to modulate lipid signaling, which in turn activates stress response pathways to maintain UPS proteostasis.
Collapse
Affiliation(s)
- Kishore K Joshi
- Department of GeneticsThe Waksman InstituteRutgers The State University of New JerseyPiscatawayNJUSA
| | - Tarmie L Matlack
- Department of GeneticsThe Waksman InstituteRutgers The State University of New JerseyPiscatawayNJUSA
| | - Stephanie Pyonteck
- Department of GeneticsThe Waksman InstituteRutgers The State University of New JerseyPiscatawayNJUSA
| | - Mehul Vora
- Department of GeneticsThe Waksman InstituteRutgers The State University of New JerseyPiscatawayNJUSA
| | - Ralph Menzel
- Institute of Biology and EcologyHumboldt University BerlinBerlinGermany
| | - Christopher Rongo
- Department of GeneticsThe Waksman InstituteRutgers The State University of New JerseyPiscatawayNJUSA
| |
Collapse
|
16
|
Lee SK. Endoplasmic Reticulum Homeostasis and Stress Responses in Caenorhabditis elegans. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:279-303. [PMID: 34050871 DOI: 10.1007/978-3-030-67696-4_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The unfolded protein response (UPR) is an evolutionarily conserved adaptive regulatory pathway that alleviates protein-folding defects in the endoplasmic reticulum (ER). Physiological demands, environmental perturbations and pathological conditions can cause accumulation of unfolded proteins in the ER and the stress signal is transmitted to the nucleus to turn on a series of genes to respond the challenge. In metazoan, the UPR pathways consisted of IRE1/XBP1, PEK-1 and ATF6, which function in parallel and downstream transcriptional activation triggers the proteostasis networks consisting of molecular chaperones, protein degradation machinery and other stress response pathways ((Labbadia J, Morimoto RI, F1000Prime Rep 6:7, 2014); (Shen X, Ellis RE, Lee K, Annu Rev Biochem 28:893-903, 2014)). The integrated responses act on to resolve the ER stress by increasing protein folding capacity, attenuating ER-loading translation, activating ER-associated proteasomal degradation (ERAD), and regulating IRE1-dependent decay of mRNA (RIDD). Therefore, the effective UPR to internal and external causes is linked to the multiple pathophysiological conditions such as aging, immunity, and neurodegenerative diseases. Recent development in the research of the UPR includes cell-nonautonomous features of the UPR, interplay between the UPR and other stress response pathways, unconventional UPR inducers, and noncanonical UPR independent of the three major branches, originated from multiple cellular and molecular machineries in addition to ER. Caenorhabditis elegans model system has critically contributed to these unprecedented aspects of the ER UPR and broadens the possible therapeutic targets to treat the ER-stress associated human disorders and time-dependent physiological deterioration of aging.
Collapse
Affiliation(s)
- Sun-Kyung Lee
- Department of Life Science, Research Institute for Natural Sciences, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
17
|
Özbey NP, Imanikia S, Krueger C, Hardege I, Morud J, Sheng M, Schafer WR, Casanueva MO, Taylor RC. Tyramine Acts Downstream of Neuronal XBP-1s to Coordinate Inter-tissue UPR ER Activation and Behavior in C. elegans. Dev Cell 2020; 55:754-770.e6. [PMID: 33232669 PMCID: PMC7758879 DOI: 10.1016/j.devcel.2020.10.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/17/2020] [Accepted: 10/30/2020] [Indexed: 12/28/2022]
Abstract
In C. elegans, expression of the UPRER transcription factor xbp-1s in neurons cell non-autonomously activates the UPRER in the intestine, leading to enhanced proteostasis and lifespan. To better understand this signaling pathway, we isolated neurons from animals expressing neuronal xbp-1s for transcriptomic analysis, revealing a striking remodeling of transcripts involved in neuronal signaling. We then identified signaling molecules required for cell non-autonomous intestinal UPRER activation, including the biogenic amine tyramine. Expression of xbp-1s in just two pairs of neurons that synthesize tyramine, the RIM and RIC interneurons, induced intestinal UPRER activation and extended longevity, and exposure to stress led to splicing and activation of xbp-1 in these neurons. In addition, we found that neuronal xbp-1s modulates feeding behavior and reproduction, dependent upon tyramine synthesis. XBP-1s therefore remodels neuronal signaling to coordinately modulate intestinal physiology and stress-responsive behavior, functioning as a global regulator of organismal responses to stress.
Collapse
Affiliation(s)
- Neşem P Özbey
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Soudabeh Imanikia
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Christel Krueger
- Epigenetics Programme, The Babraham Institute, Babraham CB22 3AT, UK
| | - Iris Hardege
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Julia Morud
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Ming Sheng
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Rebecca C Taylor
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
18
|
Taylor RC, Hetz C. Mastering organismal aging through the endoplasmic reticulum proteostasis network. Aging Cell 2020; 19:e13265. [PMID: 33128506 PMCID: PMC7681052 DOI: 10.1111/acel.13265] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/14/2022] Open
Abstract
The aging process is characterized by a progressive decline in the function of most tissues, representing the main risk factor in the development of a variety of human diseases. Studies in multiple animal models have demonstrated that interventions that improve the capacity to maintain endoplasmic reticulum (ER) proteostasis prolong life and healthspan. ER stress is monitored by the unfolded protein response (UPR), a signaling pathway that mediates adaptive processes to restore proteostasis or the elimination of damaged cells by apoptosis. Here, we discuss recent advances in understanding the significance of the UPR to aging and its implications for the maintenance of cell physiology of various cell types and organs. The possible benefits of targeting the UPR to extend healthspan and reduce the risk of developing age-related diseases are also discussed.
Collapse
Affiliation(s)
| | - Claudio Hetz
- Center for GeroscienceBrain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Buck Institute for Research on AgingNovatoCAUSA
| |
Collapse
|
19
|
Prahlad V. The discovery and consequences of the central role of the nervous system in the control of protein homeostasis. J Neurogenet 2020; 34:489-499. [PMID: 32527175 PMCID: PMC7736053 DOI: 10.1080/01677063.2020.1771333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/14/2020] [Indexed: 12/30/2022]
Abstract
Organisms function despite wide fluctuations in their environment through the maintenance of homeostasis. At the cellular level, the maintenance of proteins as functional entities at target expression levels is called protein homeostasis (or proteostasis). Cells implement proteostasis through universal and conserved quality control mechanisms that surveil and monitor protein conformation. Recent studies that exploit the powerful ability to genetically manipulate specific neurons in C. elegans have shown that cells within this metazoan lose their autonomy over this fundamental survival mechanism. These studies have uncovered novel roles for the nervous system in controlling how and when cells activate their protein quality control mechanisms. Here we discuss the conceptual underpinnings, experimental evidence and the possible consequences of such a control mechanism. PRELUDE: Whether the detailed examination of parts of the nervous system and their selective perturbation is sufficient to reconstruct how the brain generates behavior, mental disease, music and religion remains an open question. Yet, Sydney Brenner's development of C. elegans as an experimental organism and his faith in the bold reductionist approach that 'the understanding of wild-type behavior comes best after the discovery and analysis of mutations that alter it', has led to discoveries of unexpected roles for neurons in the biology of organisms.
Collapse
Affiliation(s)
- Veena Prahlad
- Department of Biology, Aging Mind and Brain Initiative, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
20
|
Metcalf MG, Higuchi-Sanabria R, Garcia G, Tsui CK, Dillin A. Beyond the cell factory: Homeostatic regulation of and by the UPR ER. SCIENCE ADVANCES 2020; 6:eabb9614. [PMID: 32832649 PMCID: PMC7439504 DOI: 10.1126/sciadv.abb9614] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/02/2020] [Indexed: 05/02/2023]
Abstract
The endoplasmic reticulum (ER) is commonly referred to as the factory of the cell, as it is responsible for a large amount of protein and lipid synthesis. As a membrane-bound organelle, the ER has a distinct environment that is ideal for its functions in synthesizing these primary cellular components. Many different quality control machineries exist to maintain ER stability under the stresses associated with synthesizing, folding, and modifying complex proteins and lipids. The best understood of these mechanisms is the unfolded protein response of the ER (UPRER), in which transmembrane proteins serve as sensors, which trigger a coordinated transcriptional response of genes dedicated for mitigating the stress. As the name suggests, the UPRER is most well described as a functional response to protein misfolding stress. Here, we focus on recent findings and emerging themes in additional roles of the UPRER outside of protein homeostasis, including lipid homeostasis, autophagy, apoptosis, and immunity.
Collapse
|
21
|
Batista A, Rodvold JJ, Xian S, Searles SC, Lew A, Iwawaki T, Almanza G, Waller TC, Lin J, Jepsen K, Carter H, Zanetti M. IRE1α regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol 2020; 18:e3000687. [PMID: 32520957 PMCID: PMC7307794 DOI: 10.1371/journal.pbio.3000687] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/22/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
In the tumor microenvironment, local immune dysregulation is driven in part by macrophages and dendritic cells that are polarized to a mixed proinflammatory/immune-suppressive phenotype. The unfolded protein response (UPR) is emerging as the possible origin of these events. Here we report that the inositol-requiring enzyme 1 (IRE1α) branch of the UPR is directly involved in the polarization of macrophages in vitro and in vivo, including the up-regulation of interleukin 6 (IL-6), IL-23, Arginase1, as well as surface expression of CD86 and programmed death ligand 1 (PD-L1). Macrophages in which the IRE1α/X-box binding protein 1 (Xbp1) axis is blocked pharmacologically or deleted genetically have significantly reduced polarization and CD86 and PD-L1 expression, which was induced independent of IFNγ signaling, suggesting a novel mechanism in PD-L1 regulation in macrophages. Mice with IRE1α- but not Xbp1-deficient macrophages showed greater survival than controls when implanted with B16.F10 melanoma cells. Remarkably, we found a significant association between the IRE1α gene signature and CD274 gene expression in tumor-infiltrating macrophages in humans. RNA sequencing (RNASeq) analysis showed that bone marrow-derived macrophages with IRE1α deletion lose the integrity of the gene connectivity characteristic of regulated IRE1α-dependent decay (RIDD) and the ability to activate CD274 gene expression. Thus, the IRE1α/Xbp1 axis drives the polarization of macrophages in the tumor microenvironment initiating a complex immune dysregulation leading to failure of local immune surveillance.
Collapse
Affiliation(s)
- Alyssa Batista
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Jeffrey J. Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Su Xian
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Stephen C. Searles
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Alyssa Lew
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Takao Iwawaki
- Laboratory for Cell Recovery Mechanisms, Brain Science Institute, RIKEN, Hirosawa, Japan
| | - Gonzalo Almanza
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - T. Cameron Waller
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Jonathan Lin
- Department of Pathology, Stanford University, Palo Alto, California, United States of America
| | - Kristen Jepsen
- IGM Genomics Center, University of California, San Diego, La Jolla, California, United States of America
| | - Hannah Carter
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
22
|
Zhang Z, Bai M, Barbosa GO, Chen A, Wei Y, Luo S, Wang X, Wang B, Tsukui T, Li H, Sheppard D, Kornberg TB, Ma DK. Broadly conserved roles of TMEM131 family proteins in intracellular collagen assembly and secretory cargo trafficking. SCIENCE ADVANCES 2020; 6:eaay7667. [PMID: 32095531 PMCID: PMC7015688 DOI: 10.1126/sciadv.aay7667] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
Collagen is the most abundant protein in animals. Its dysregulation contributes to aging and many human disorders, including pathological tissue fibrosis in major organs. How premature collagen proteins in the endoplasmic reticulum (ER) assemble and route for secretion remains molecularly undefined. From an RNA interference screen, we identified an uncharacterized Caenorhabditis elegans gene tmem-131, deficiency of which impairs collagen production and activates ER stress response. We find that amino termini of human TMEM131 contain bacterial PapD chaperone-like domains, which recruit premature collagen monomers for proper assembly and secretion. Carboxy termini of TMEM131 interact with TRAPPC8, a component of the TRAPP tethering complex, to drive collagen cargo trafficking from ER to the Golgi. We provide evidence that previously undescribed roles of TMEM131 in collagen recruitment and secretion are evolutionarily conserved in C. elegans, Drosophila, and humans.
Collapse
Affiliation(s)
- Zhe Zhang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Meirong Bai
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Guilherme Oliveira Barbosa
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Andrew Chen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuehua Wei
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shuo Luo
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xin Wang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bingying Wang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tatsuya Tsukui
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dean Sheppard
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Thomas B. Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dengke K. Ma
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
23
|
O'Brien D, Jones LM, Good S, Miles J, Vijayabaskar MS, Aston R, Smith CE, Westhead DR, van Oosten-Hawle P. A PQM-1-Mediated Response Triggers Transcellular Chaperone Signaling and Regulates Organismal Proteostasis. Cell Rep 2019; 23:3905-3919. [PMID: 29949773 PMCID: PMC6045774 DOI: 10.1016/j.celrep.2018.05.093] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/04/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
In metazoans, tissues experiencing proteotoxic stress induce "transcellular chaperone signaling" (TCS) that activates molecular chaperones, such as hsp-90, in distal tissues. How this form of inter-tissue communication is mediated to upregulate systemic chaperone expression and whether it can be utilized to protect against protein misfolding diseases remain open questions. Using C. elegans, we identified key components of a systemic stress signaling pathway that links the innate immune response with proteostasis maintenance. We show that mild perturbation of proteostasis in the neurons or the intestine activates TCS via the GATA zinc-finger transcription factor PQM-1. PQM-1 coordinates neuron-activated TCS via the innate immunity-associated transmembrane protein CLEC-41, whereas intestine-activated TCS depends on the aspartic protease ASP-12. Both TCS pathways can induce hsp-90 in muscle cells and facilitate amelioration of Aβ3-42-associated toxicity. This may have powerful implications for the treatment of diseases related to proteostasis dysfunction.
Collapse
Affiliation(s)
- Daniel O'Brien
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Laura M Jones
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Sarah Good
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Jo Miles
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - M S Vijayabaskar
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Rebecca Aston
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Catrin E Smith
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - David R Westhead
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
24
|
Singh J, Aballay A. Neural control of behavioral and molecular defenses in C. elegans. Curr Opin Neurobiol 2019; 62:34-40. [PMID: 31812835 DOI: 10.1016/j.conb.2019.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/14/2019] [Indexed: 01/22/2023]
Abstract
The nervous and immune systems use bi-directional communication to control host responses against microbial pathogens. Recent studies at the interface of the two systems have highlighted important roles of the nervous system in the regulation of both microbicidal pathways and pathogen avoidance behaviors. Studies on the neural circuits in the simple model host Caenorhabditis elegans have significantly improved our understanding of the roles of conserved neural mechanisms in controlling innate immunity. Moreover, behavioral studies have advanced our understanding of how the nervous system may sense potential pathogens and consequently elicit pathogen avoidance, reducing the risk of infection. In this review, we discuss the neural circuits that regulate both behavioral immunity and molecular immunity in C. elegans.
Collapse
Affiliation(s)
- Jogender Singh
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alejandro Aballay
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
25
|
Ascaroside Pheromones: Chemical Biology and Pleiotropic Neuronal Functions. Int J Mol Sci 2019; 20:ijms20163898. [PMID: 31405082 PMCID: PMC6719183 DOI: 10.3390/ijms20163898] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 07/26/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022] Open
Abstract
Pheromones are neuronal signals that stimulate conspecific individuals to react to environmental stressors or stimuli. Research on the ascaroside (ascr) pheromones in Caenorhabditis elegans and other nematodes has made great progress since ascr#1 was first isolated and biochemically defined in 2005. In this review, we highlight the current research on the structural diversity, biosynthesis, and pleiotropic neuronal functions of ascr pheromones and their implications in animal physiology. Experimental evidence suggests that ascr biosynthesis starts with conjugation of ascarylose to very long-chain fatty acids that are then processed via peroxisomal β-oxidation to yield diverse ascr pheromones. We also discuss the concentration and stage-dependent pleiotropic neuronal functions of ascr pheromones. These functions include dauer induction, lifespan extension, repulsion, aggregation, mating, foraging and detoxification, among others. These roles are carried out in coordination with three G protein-coupled receptors that function as putative pheromone receptors: SRBC-64/66, SRG-36/37, and DAF-37/38. Pheromone sensing is transmitted in sensory neurons via DAF-16-regulated glutamatergic neurotransmitters. Neuronal peroxisomal fatty acid β-oxidation has important cell-autonomous functions in the regulation of neuroendocrine signaling, including neuroprotection. In the future, translation of our knowledge of nematode ascr pheromones to higher animals might be beneficial, as ascr#1 has some anti-inflammatory effects in mice. To this end, we propose the establishment of pheromics (pheromone omics) as a new subset of integrated disciplinary research area within chemical ecology for system-wide investigation of animal pheromones.
Collapse
|
26
|
Miles J, Scherz-Shouval R, van Oosten-Hawle P. Expanding the Organismal Proteostasis Network: Linking Systemic Stress Signaling with the Innate Immune Response. Trends Biochem Sci 2019; 44:927-942. [PMID: 31303384 DOI: 10.1016/j.tibs.2019.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022]
Abstract
Stress response pathways regulate proteostasis and mitigate macromolecular damage to promote long-term cellular health. Intercellular signaling is an essential layer of systemic proteostasis in an organism and is facilitated via transcellular signaling molecules that orchestrate the activation of stress responses across tissues and organs. Accumulating evidence indicates that components of the immune response act as signaling factors that regulate the cell-non-autonomous proteostasis network. Here, we review emergent advances in our understanding of cell-non-autonomous regulators of proteostasis networks in multicellular settings, from the model organism, Caenorhabditis elegans, to humans. We further discuss how innate immune responses can be players of the organismal proteostasis network and discuss how both are linked in cancer.
Collapse
Affiliation(s)
- Jay Miles
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Ruth Scherz-Shouval
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Patricija van Oosten-Hawle
- School of Molecular and Cell Biology and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
27
|
Sepahi A, Kraus A, Casadei E, Johnston CA, Galindo-Villegas J, Kelly C, García-Moreno D, Muñoz P, Mulero V, Huertas M, Salinas I. Olfactory sensory neurons mediate ultrarapid antiviral immune responses in a TrkA-dependent manner. Proc Natl Acad Sci U S A 2019; 116:12428-12436. [PMID: 31160464 DOI: 10.1101/464214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
The nervous system regulates host immunity in complex ways. Vertebrate olfactory sensory neurons (OSNs) are located in direct contact with pathogens; however, OSNs' ability to detect danger and initiate immune responses is unclear. We report that nasal delivery of rhabdoviruses induces apoptosis in crypt OSNs via the interaction of the OSN TrkA receptor with the viral glycoprotein in teleost fish. This signal results in electrical activation of neurons and very rapid proinflammatory responses in the olfactory organ (OO), but dampened inflammation in the olfactory bulb (OB). CD8α+ cells infiltrate the OO within minutes of nasal viral delivery, and TrkA blocking, but not caspase-3 blocking, abrogates this response. Infiltrating CD8α+ cells were TCRαβ T cells with a nonconventional phenotype that originated from the microvasculature surrounding the OB and not the periphery. Nasal delivery of viral glycoprotein (G protein) recapitulated the immune responses observed with the whole virus, and antibody blocking of viral G protein abrogated these responses. Ablation of crypt neurons in zebrafish resulted in increased susceptibility to rhabdoviruses. These results indicate a function for OSNs as a first layer of pathogen detection in vertebrates and as orchestrators of nasal-CNS antiviral immune responses.
Collapse
Affiliation(s)
- Ali Sepahi
- Center of Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM 87131
| | - Aurora Kraus
- Center of Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM 87131
| | - Elisa Casadei
- Center of Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM 87131
| | | | - Jorge Galindo-Villegas
- Department of Cell Biology and Histology, Faculty of Biology, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain
- Faculty of Biosciences and Aquaculture, Nord University, 8049 Bodø, Norway
| | - Cecelia Kelly
- Center of Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM 87131
| | - Diana García-Moreno
- Department of Cell Biology and Histology, Faculty of Biology, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain
| | - Pilar Muñoz
- Department of Faculty of Veterinary, University of Murcia, 30100 Murcia, Spain
| | - Victoriano Mulero
- Department of Cell Biology and Histology, Faculty of Biology, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Campus Universitario de Espinardo, University of Murcia, 30100 Murcia, Spain
| | - Mar Huertas
- Department of Biology, Texas State University, San Marcos, TX 78666
| | - Irene Salinas
- Center of Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM 87131;
| |
Collapse
|
28
|
Olfactory sensory neurons mediate ultrarapid antiviral immune responses in a TrkA-dependent manner. Proc Natl Acad Sci U S A 2019; 116:12428-12436. [PMID: 31160464 DOI: 10.1073/pnas.1900083116] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The nervous system regulates host immunity in complex ways. Vertebrate olfactory sensory neurons (OSNs) are located in direct contact with pathogens; however, OSNs' ability to detect danger and initiate immune responses is unclear. We report that nasal delivery of rhabdoviruses induces apoptosis in crypt OSNs via the interaction of the OSN TrkA receptor with the viral glycoprotein in teleost fish. This signal results in electrical activation of neurons and very rapid proinflammatory responses in the olfactory organ (OO), but dampened inflammation in the olfactory bulb (OB). CD8α+ cells infiltrate the OO within minutes of nasal viral delivery, and TrkA blocking, but not caspase-3 blocking, abrogates this response. Infiltrating CD8α+ cells were TCRαβ T cells with a nonconventional phenotype that originated from the microvasculature surrounding the OB and not the periphery. Nasal delivery of viral glycoprotein (G protein) recapitulated the immune responses observed with the whole virus, and antibody blocking of viral G protein abrogated these responses. Ablation of crypt neurons in zebrafish resulted in increased susceptibility to rhabdoviruses. These results indicate a function for OSNs as a first layer of pathogen detection in vertebrates and as orchestrators of nasal-CNS antiviral immune responses.
Collapse
|
29
|
Koelle MR. Neurotransmitter signaling through heterotrimeric G proteins: insights from studies in C. elegans. WORMBOOK : THE ONLINE REVIEW OF C. ELEGANS BIOLOGY 2018; 2018:1-52. [PMID: 26937633 PMCID: PMC5010795 DOI: 10.1895/wormbook.1.75.2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Neurotransmitters signal via G protein coupled receptors (GPCRs) to modulate activity of neurons and muscles. C. elegans has ∼150 G protein coupled neuropeptide receptor homologs and 28 additional GPCRs for small-molecule neurotransmitters. Genetic studies in C. elegans demonstrate that neurotransmitters diffuse far from their release sites to activate GPCRs on distant cells. Individual receptor types are expressed on limited numbers of cells and thus can provide very specific regulation of an individual neural circuit and behavior. G protein coupled neurotransmitter receptors signal principally via the three types of heterotrimeric G proteins defined by the G alpha subunits Gαo, Gαq, and Gαs. Each of these G alpha proteins is found in all neurons plus some muscles. Gαo and Gαq signaling inhibit and activate neurotransmitter release, respectively. Gαs signaling, like Gαq signaling, promotes neurotransmitter release. Many details of the signaling mechanisms downstream of Gαq and Gαs have been delineated and are consistent with those of their mammalian orthologs. The details of the signaling mechanism downstream of Gαo remain a mystery. Forward genetic screens in C. elegans have identified new molecular components of neural G protein signaling mechanisms, including Regulators of G protein Signaling (RGS proteins) that inhibit signaling, a new Gαq effector (the Trio RhoGEF domain), and the RIC-8 protein that is required for neuronal Gα signaling. A model is presented in which G proteins sum up the variety of neuromodulator signals that impinge on a neuron to calculate its appropriate output level.
Collapse
Affiliation(s)
- Michael R Koelle
- Department of Molecular Biophysics & Biochemistry, Yale University, New Haven CT 06520 USA
| |
Collapse
|
30
|
Groenendyk J, Paskevicius T, Urra H, Viricel C, Wang K, Barakat K, Hetz C, Kurgan L, Agellon LB, Michalak M. Cyclosporine A binding to COX-2 reveals a novel signaling pathway that activates the IRE1α unfolded protein response sensor. Sci Rep 2018; 8:16678. [PMID: 30420769 PMCID: PMC6232179 DOI: 10.1038/s41598-018-34891-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/26/2018] [Indexed: 12/26/2022] Open
Abstract
Cyclosporine, a widely used immunosuppressant in organ transplantation and in treatment of various autoimmune diseases, activates the unfolded protein response (UPR), an ER stress coping response. In this study we discovered a new and unanticipated cyclosporine-dependent signaling pathway, with cyclosporine triggering direct activation of the UPR. COX-2 binds to and activates IRE1α, leading to IRE1α splicing of XBP1 mRNA. Molecular interaction and modeling analyses identified a novel interaction site for cyclosporine with COX-2 which caused enhancement of COX-2 enzymatic activity required for activation of the IRE1α branch of the UPR. Cyclosporine-dependent activation of COX-2 and IRE1α in mice indicated that cyclosporine-COX-2-IRE1α signaling pathway was functional in vivo. These findings identify COX-2 as a new IRE1α binding partner and regulator of the IRE1α branch of the UPR pathway, and establishes the mechanism underlying cytotoxicity associated with chronic cyclosporine exposure.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada
| | - Tautvydas Paskevicius
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada
| | - Hery Urra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Clement Viricel
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada
| | - Kui Wang
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, People's Republic of China
| | - Khaled Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism (GERO), University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA.,The Buck Institute for Research in Aging, Novato, CA, 94945, USA
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, 23284, USA
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, H9X 3V9, Canada.
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2S7, Canada.
| |
Collapse
|
31
|
Wong MY, Chen K, Antonopoulos A, Kasper BT, Dewal MB, Taylor RJ, Whittaker CA, Hein PP, Dell A, Genereux JC, Haslam SM, Mahal LK, Shoulders MD. XBP1s activation can globally remodel N-glycan structure distribution patterns. Proc Natl Acad Sci U S A 2018; 115:E10089-E10098. [PMID: 30305426 PMCID: PMC6205500 DOI: 10.1073/pnas.1805425115] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Classically, the unfolded protein response (UPR) safeguards secretory pathway proteostasis. The most ancient arm of the UPR, the IRE1-activated spliced X-box binding protein 1 (XBP1s)-mediated response, has roles in secretory pathway maturation beyond resolving proteostatic stress. Understanding the consequences of XBP1s activation for cellular processes is critical for elucidating mechanistic connections between XBP1s and development, immunity, and disease. Here, we show that a key functional output of XBP1s activation is a cell type-dependent shift in the distribution of N-glycan structures on endogenous membrane and secreted proteomes. For example, XBP1s activity decreased levels of sialylation and bisecting GlcNAc in the HEK293 membrane proteome and secretome, while substantially increasing the population of oligomannose N-glycans only in the secretome. In HeLa cell membranes, stress-independent XBP1s activation increased the population of high-mannose and tetraantennary N-glycans, and also enhanced core fucosylation. mRNA profiling experiments suggest that XBP1s-mediated remodeling of the N-glycome is, at least in part, a consequence of coordinated transcriptional resculpting of N-glycan maturation pathways by XBP1s. The discovery of XBP1s-induced N-glycan structural remodeling on a glycome-wide scale suggests that XBP1s can act as a master regulator of N-glycan maturation. Moreover, because the sugars on cell-surface proteins or on proteins secreted from an XBP1s-activated cell can be molecularly distinct from those of an unactivated cell, these findings reveal a potential new mechanism for translating intracellular stress signaling into altered interactions with the extracellular environment.
Collapse
Affiliation(s)
- Madeline Y Wong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Kenny Chen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | - Brian T Kasper
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, NY 10003
| | - Mahender B Dewal
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Rebecca J Taylor
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Charles A Whittaker
- Barbara K. Ostrom (1978) Bioinformatics and Computing Facility, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Pyae P Hein
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Joseph C Genereux
- Department of Chemistry, University of California, Riverside, CA 92521
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom;
| | - Lara K Mahal
- Biomedical Chemistry Institute, Department of Chemistry, New York University, New York, NY 10003;
| | - Matthew D Shoulders
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139;
| |
Collapse
|
32
|
Abstract
Insufficient or excessive immune responses to pathogen infection are major causes of disease. Increasing evidence indicates that the nervous system regulates the immune system to help maintain immunological homeostasis. However, the precise mechanisms of this regulation are largely unknown. Here we show the existence of an octopaminergic immunoinhibitory pathway in Caenorhabditis elegans. Our study results indicate that this pathway is tonically active under normal conditions to maintain immunological homeostasis or suppress unwanted innate immune responses but downregulated upon pathogen infection to allow enhanced innate immunity. As excessive innate immune responses have been linked to human health conditions such as Crohn's disease, rheumatoid arthritis, atherosclerosis, diabetes, and Alzheimer's disease, elucidating octopaminergic neural regulation of innate immunity could be helpful in the development of new treatments for innate immune diseases. Upon pathogen infection, the nervous system regulates innate immunity to confer coordinated protection to the host. However, the precise mechanisms of such regulation remain unclear. Previous studies have demonstrated that OCTR-1, a putative G protein-coupled receptor for catecholamine, functions in the sensory neurons designated “ASH” to suppress innate immune responses in Caenorhabditis elegans. It is unknown what molecules act as OCTR-1 ligands in the neural immune regulatory circuit. Here we identify neurotransmitter octopamine (OA) as an endogenous ligand for OCTR-1 in immune regulation and show that the OA-producing RIC neurons function in the OCTR-1 neural circuit to suppress innate immunity. RIC neurons are deactivated in the presence of pathogens but transiently activated by nonpathogenic bacteria. Our data support a model whereby an octopaminergic immunoinhibitory pathway is tonically active under normal conditions to maintain immunological homeostasis or suppress unwanted innate immune responses but downregulated upon pathogen infection to allow enhanced innate immunity. As excessive innate immune responses have been linked to a myriad of human health concerns, our study could potentially benefit the development of more-effective treatments for innate immune disorders.
Collapse
|
33
|
Imanikia S, Sheng M, Taylor RC. Cell Non-autonomous UPR ER Signaling. Curr Top Microbiol Immunol 2018; 414:27-43. [PMID: 28879522 DOI: 10.1007/82_2017_38] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The UPRER is an important regulator of secretory pathway homeostasis, and plays roles in many physiological processes. Its broad range of targets and ability to modulate secretion and membrane trafficking make it perfectly positioned to influence intercellular communication, enabling the UPRER to coordinate physiological processes between cells and tissues. Recent evidence suggests that the activation of the UPRER can itself be communicated between cells. This cell non-autonomous route to UPRER activation occurs in multiple species, and enables organism-wide responses to stress that involve processes as diverse as immunity, metabolism, aging and reproduction. It may also play roles in disease progression, making the pathways that mediate cell non-autonomous UPRER signaling a potential source of novel future therapeutics.
Collapse
Affiliation(s)
| | - Ming Sheng
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | | |
Collapse
|
34
|
Chapple RH, Hu T, Tseng YJ, Liu L, Kitano A, Luu V, Hoegenauer KA, Iwawaki T, Li Q, Nakada D. ERα promotes murine hematopoietic regeneration through the Ire1α-mediated unfolded protein response. eLife 2018; 7:31159. [PMID: 29451493 PMCID: PMC5829925 DOI: 10.7554/elife.31159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 02/15/2018] [Indexed: 01/08/2023] Open
Abstract
Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.
Collapse
Affiliation(s)
- Richard H Chapple
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Tianyuan Hu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| | - Lu Liu
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Ayumi Kitano
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Victor Luu
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Kevin A Hoegenauer
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Qing Li
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, United States
| | - Daisuke Nakada
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, United States.,Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, United States
| |
Collapse
|
35
|
Higuchi-Sanabria R, Frankino PA, Paul JW, Tronnes SU, Dillin A. A Futile Battle? Protein Quality Control and the Stress of Aging. Dev Cell 2018; 44:139-163. [PMID: 29401418 PMCID: PMC5896312 DOI: 10.1016/j.devcel.2017.12.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/30/2017] [Accepted: 12/20/2017] [Indexed: 12/15/2022]
Abstract
There exists a phenomenon in aging research whereby early life stress can have positive impacts on longevity. The mechanisms underlying these observations suggest a robust, long-lasting induction of cellular defense mechanisms. These include the various unfolded protein responses of the endoplasmic reticulum (ER), cytosol, and mitochondria. Indeed, ectopic induction of these pathways, in the absence of stress, is sufficient to increase lifespan in organisms as diverse as yeast, worms, and flies. Here, we provide an overview of the protein quality control mechanisms that operate in the cytosol, mitochondria, and ER and discuss how they affect cellular health and viability during stress and aging.
Collapse
Affiliation(s)
- Ryo Higuchi-Sanabria
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Phillip Andrew Frankino
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph West Paul
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sarah Uhlein Tronnes
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andrew Dillin
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; The Glenn Center for Aging Research, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
36
|
Cao X, Kajino-Sakamoto R, Doss A, Aballay A. Distinct Roles of Sensory Neurons in Mediating Pathogen Avoidance and Neuropeptide-Dependent Immune Regulation. Cell Rep 2017; 21:1442-1451. [PMID: 29117551 PMCID: PMC5726787 DOI: 10.1016/j.celrep.2017.10.050] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/19/2017] [Accepted: 10/13/2017] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence implies an extensive and universal interaction between the immune system and the nervous system. Previous studies showed that OCTR-1, a neuronal G-protein-coupled receptor (GPCR) analogous to human norepinephrine receptors, functions in sensory neurons to control the gene expression of both microbial killing pathways and the unfolded protein response (UPR) in Caenorhabditis elegans. Here, we found that OCTR-1-expressing neurons, ASH, are involved in controlling innate immune pathways. In contrast, another group of OCTR-1-expressing neurons, ASI, was shown to promote pathogen avoidance behavior. We also identified neuropeptide NLP-20 and AIA interneurons, which are responsible for the integration of conflicting cues and behaviors, as downstream components of the ASH/ASI neural circuit. These findings provide insights into a neuronal network involved in regulating pathogen defense mechanisms in C. elegans and might have broad implications for the strategies utilized by metazoans to balance the energy-costly immune activation and behavioral response.
Collapse
Affiliation(s)
- Xiou Cao
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Rie Kajino-Sakamoto
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Division of Molecular Pathology, Aichi Cancer Center Research Institute, Nagoya, Aichi 464-8681, Japan
| | - Argenia Doss
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alejandro Aballay
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
37
|
Martínez G, Duran‐Aniotz C, Cabral‐Miranda F, Vivar JP, Hetz C. Endoplasmic reticulum proteostasis impairment in aging. Aging Cell 2017; 16:615-623. [PMID: 28436203 PMCID: PMC5506418 DOI: 10.1111/acel.12599] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbed neuronal proteostasis is a salient feature shared by both aging and protein misfolding disorders. The proteostasis network controls the health of the proteome by integrating pathways involved in protein synthesis, folding, trafficking, secretion, and their degradation. A reduction in the buffering capacity of the proteostasis network during aging may increase the risk to undergo neurodegeneration by enhancing the accumulation of misfolded proteins. As almost one-third of the proteome is synthetized at the endoplasmic reticulum (ER), maintenance of its proper function is fundamental to sustain neuronal function. In fact, ER stress is a common feature of most neurodegenerative diseases. The unfolded protein response (UPR) operates as central player to maintain ER homeostasis or the induction of cell death of chronically damaged cells. Here, we discuss recent evidence placing ER stress as a driver of brain aging, and the emerging impact of neuronal UPR in controlling global proteostasis at the whole organismal level. Finally, we discuss possible therapeutic interventions to improve proteostasis and prevent pathological brain aging.
Collapse
Affiliation(s)
- Gabriela Martínez
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Center for Integrative BiologyUniversidad MayorSantiagoChile
| | - Claudia Duran‐Aniotz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Felipe Cabral‐Miranda
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de JaneiroRio de JaneiroBrasil
| | - Juan P. Vivar
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Claudio Hetz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Buck Institute for Research on AgingNovatoCA94945USA
- Department of Immunology and Infectious diseasesHarvard School of Public HealthBostonMA02115USA
| |
Collapse
|
38
|
Kyriakakis E, Charmpilas N, Tavernarakis N. Differential adiponectin signalling couples ER stress with lipid metabolism to modulate ageing in C. elegans. Sci Rep 2017; 7:5115. [PMID: 28698593 PMCID: PMC5505976 DOI: 10.1038/s41598-017-05276-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 05/26/2017] [Indexed: 12/21/2022] Open
Abstract
The metabolic and endocrine functions of adipose tissue and the ability of organisms to cope with cellular stress have a direct impact on physiological ageing and the aetiology of various diseases such as obesity-related pathologies and cancer. The endocrine effects of adipose tissue are mediated by secreted adipokines, which modulate metabolic processes and influence related maladies. Although a plethora of molecules and signaling pathways associate ageing with proteotoxic stress and cellular metabolism, our understanding of how these pathways interconnect to coordinate organismal physiology remains limited. We dissected the mechanisms linking adiponectin signalling pathways and endoplasmic reticulum (ER) proteotoxic stress responses that individually or synergistically affect longevity in C. elegans. Animals deficient for the adiponectin receptor PAQR-1 respond to ER stress, by rapidly activating the canonical ER unfolded protein response (UPRER) pathway, which is primed in these animals under physiological conditions by specific stress defence transcription factors. PAQR-1 loss enhances survival and promotes longevity under ER stress and reduced insulin/IGF-1 signalling. PAQR-1 engages UPRER, autophagy and lipase activity to modulate lipid metabolism during ageing. Our findings demonstrate that moderating adiponectin receptor -1 activity extends lifespan under stress, and directly implicate adiponectin signalling as a coupler between proteostasis and lipid metabolism during ageing.
Collapse
Affiliation(s)
- Emmanouil Kyriakakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece
| | - Nikolaos Charmpilas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece
- Department of Biology, University of Crete, Heraklion 70013, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas, Nikolaou Plastira 100, Heraklion 70013, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion 71110, Crete, Greece.
| |
Collapse
|
39
|
Regulation of cell-non-autonomous proteostasis in metazoans. Essays Biochem 2017; 60:133-142. [PMID: 27744329 PMCID: PMC5065704 DOI: 10.1042/ebc20160006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 04/28/2016] [Indexed: 12/24/2022]
Abstract
Cells have developed robust adaptation mechanisms to survive environmental conditions that challenge the integrity of their proteome and ensure cellular viability. These are stress signalling pathways that integrate extracellular signals with the ability to detect and efficiently respond to protein-folding perturbations within the cell. Within the context of an organism, the cell-autonomous effects of these signalling mechanisms are superimposed by cell-non-autonomous stress signalling pathways that allow co-ordination of stress responses across tissues. These transcellular stress signalling pathways orchestrate and maintain the cellular proteome at an organismal level. This article focuses on mechanisms in both invertebrate and vertebrate organisms that activate stress responses in a cell-non-autonomous manner. We discuss emerging insights and provide specific examples on how components of the cell-non-autonomous proteostasis network are used in cancer and protein-folding diseases to drive disease progression across tissues.
Collapse
|
40
|
Endoplasmic Reticulum Stress Caused by Lipoprotein Accumulation Suppresses Immunity against Bacterial Pathogens and Contributes to Immunosenescence. mBio 2017; 8:mBio.00778-17. [PMID: 28559483 PMCID: PMC5449662 DOI: 10.1128/mbio.00778-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The unfolded protein response (UPR) is a stress response pathway that is activated upon increased unfolded and/or misfolded proteins in the endoplasmic reticulum (ER), and enhanced ER stress response prolongs life span and improves immunity. However, the mechanism by which ER stress affects immunity remains poorly understood. Using the nematode Caenorhabditis elegans, we show that mutations in the lipoproteins vitellogenins, which are homologs of human apolipoprotein B-100, resulted in upregulation of the UPR. Lipoprotein accumulation in the intestine adversely affects the immune response and the life span of the organism, suggesting that it could be a contributing factor to immunosenescence. We show that lipoprotein accumulation inhibited the expression of several immune genes encoding proteins secreted by the intestinal cells in an IRE-1-independent manner. Our studies provide a mechanistic explanation for adverse effects caused by protein aggregation and ER stress on immunity and highlight the role of an IRE-1-independent pathway in the suppression of the expression of genes encoding secreted proteins. Increased accumulation of unfolded and/or misfolded proteins in the endoplasmic reticulum (ER) leads to enhanced ER stress. However, the mechanism(s) by which ER stress affects immunity remain understudied. Using the nematode C. elegans, we showed that mutations in lipoproteins lead to their accumulation in the intestine, causing ER stress and adversely affecting the life span of the organisms and their resistance to pathogen infection. Our results indicate that the ER stress caused by lipoprotein accumulation significantly reduced the levels of expression of genes encoding secreted immune effectors, contributing to immunosenescence. It is known that ER stress may suppress gene expression via IRE-1, which is a sensor of ER stress. The novel mechanism uncovered in our study is IRE-1 independent, which highlights the role of a novel process by which ER stress suppresses innate immunity.
Collapse
|
41
|
Liu Y, Sun J. G protein-coupled receptors mediate neural regulation of innate immune responses in caenorhabditis elegans. RECEPTORS & CLINICAL INVESTIGATION 2017; 4:e1543. [PMID: 30386810 PMCID: PMC6206870 DOI: 10.14800/rci.1543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
G protein-coupled receptors (GPCRs) are a large family of transmembrane proteins that perceive many extracellular signals and transduce them into cellular physiological responses. GPCRs regulate immunity in both vertebrates and invertebrates. However, the mechanisms responsible for such regulation are not fully understood. Recent research using the genetically tractable model organism Caenorhabditis elegans has led to the identification of specific GPCRs, neurotransmitters, neurons and non-neural cells in the regulation of innate immunity. Several neural circuits have been demonstrated to function in GPCR-dependent immuno-regulatory pathways. Besides being essential in neural-immune interactions, GPCRs also regulate innate immune response in non-neural tissues cell-autonomously through mechanisms independent of neural circuits. Here we review GPCR-mediated neural control of innate immunity in C. elegans and briefly discuss GPCR-dependent immune regulation via non-neural mechanisms.
Collapse
Affiliation(s)
- Yiyong Liu
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, 99202, USA
| | - Jingru Sun
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, 99202, USA
| |
Collapse
|
42
|
Gupta A, Singh V. GPCR Signaling in C. elegans and Its Implications in Immune Response. Adv Immunol 2017; 136:203-226. [DOI: 10.1016/bs.ai.2017.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
43
|
Liu Y, Sellegounder D, Sun J. Neuronal GPCR OCTR-1 regulates innate immunity by controlling protein synthesis in Caenorhabditis elegans. Sci Rep 2016; 6:36832. [PMID: 27833098 PMCID: PMC5104976 DOI: 10.1038/srep36832] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/18/2016] [Indexed: 01/09/2023] Open
Abstract
Upon pathogen infection, microbial killing pathways and cellular stress pathways are rapidly activated by the host innate immune system. These pathways must be tightly regulated because insufficient or excessive immune responses have deleterious consequences. Increasing evidence indicates that the nervous system regulates the immune system to confer coordinated protection to the host. However, the precise mechanisms of neural-immune communication remain unclear. Previously we have demonstrated that OCTR-1, a neuronal G protein-coupled receptor, functions in the sensory neurons ASH and ASI to suppress innate immune responses in non-neural tissues against Pseudomonas aeruginosa in Caenorhabditis elegans. In the current study, by using a mass spectrometry-based quantitative proteomics approach, we discovered that OCTR-1 regulates innate immunity by suppressing translation and the unfolded protein response (UPR) pathways at the protein level. Functional assays revealed that OCTR-1 inhibits specific protein synthesis factors such as ribosomal protein RPS-1 and translation initiation factor EIF-3.J to reduce infection-triggered protein synthesis and UPR. Translational inhibition by chemicals abolishes the OCTR-1-controlled innate immune responses, indicating that activation of the OCTR-1 pathway is dependent on translation upregulation such as that induced by pathogen infection. Because OCTR-1 downregulates protein translation activities, the OCTR-1 pathway could function to suppress excessive responses to infection or to restore protein homeostasis after infection.
Collapse
Affiliation(s)
- Yiyong Liu
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| | - Durai Sellegounder
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| | - Jingru Sun
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| |
Collapse
|
44
|
Martínez G, Duran-Aniotz C, Cabral-Miranda F, Hetz C. Commentary: XBP-1 Is a Cell-Nonautonomous Regulator of Stress Resistance and Longevity. Front Aging Neurosci 2016; 8:182. [PMID: 27534903 PMCID: PMC4971125 DOI: 10.3389/fnagi.2016.00182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/14/2016] [Indexed: 11/17/2022] Open
Affiliation(s)
- Gabriela Martínez
- Center for Geroscience, Brain Health and MetabolismSantiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of ChileSantiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of ChileSantiago, Chile; Center for Integrative Biology, Universidad MayorSantiago, Chile
| | - Claudia Duran-Aniotz
- Center for Geroscience, Brain Health and MetabolismSantiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of ChileSantiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| | - Felipe Cabral-Miranda
- Center for Geroscience, Brain Health and MetabolismSantiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of ChileSantiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| | - Claudio Hetz
- Center for Geroscience, Brain Health and MetabolismSantiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of ChileSantiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of ChileSantiago, Chile; Buck Institute for Research on AgingNovato, CA, USA; Department of Immunology and Infectious diseases, Harvard School of Public HealthBoston, MA, USA
| |
Collapse
|
45
|
Abstract
Amyotrophic lateral sclerosis (ALS) is proving intractable. Difficulties in pre-clinical studies contribute in small measure to this futility, but the chief reason for failure is an inadequate understanding of disease pathogenesis. Many acquired and inherited processes have been advanced as potential causes of ALS but, while they may predispose to disease, it seems increasingly likely that none leads directly to ALS. Rather, two recent overlapping considerations, both involving aberrant protein homeostasis, may provide a better explanation for a common disease phenotype and a common terminal pathogenesis. If so, therapeutic approaches will need to be altered and carefully nuanced, since protein homeostasis is essential and highly conserved. Nonetheless, these considerations provide new optimism in a difficult disease which has hitherto defied treatment.
Collapse
|
46
|
Joshi KK, Matlack TL, Rongo C. Dopamine signaling promotes the xenobiotic stress response and protein homeostasis. EMBO J 2016; 35:1885-901. [PMID: 27261197 DOI: 10.15252/embj.201592524] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 05/03/2016] [Indexed: 01/11/2023] Open
Abstract
Multicellular organisms encounter environmental conditions that adversely affect protein homeostasis (proteostasis), including extreme temperatures, toxins, and pathogens. It is unclear how they use sensory signaling to detect adverse conditions and then activate stress response pathways so as to offset potential damage. Here, we show that dopaminergic mechanosensory neurons in C. elegans release the neurohormone dopamine to promote proteostasis in epithelia. Signaling through the DA receptor DOP-1 activates the expression of xenobiotic stress response genes involved in pathogenic resistance and toxin removal, and these genes are required for the removal of unstable proteins in epithelia. Exposure to a bacterial pathogen (Pseudomonas aeruginosa) results in elevated removal of unstable proteins in epithelia, and this enhancement requires DA signaling. In the absence of DA signaling, nematodes show increased sensitivity to pathogenic bacteria and heat-shock stress. Our results suggest that dopaminergic sensory neurons, in addition to slowing down locomotion upon sensing a potential bacterial feeding source, also signal to frontline epithelia to activate the xenobiotic stress response so as to maintain proteostasis and prepare for possible infection.
Collapse
Affiliation(s)
- Kishore K Joshi
- Department of Genetics, The Waksman Institute Rutgers The State University of New Jersey, Piscataway, NJ, USA
| | - Tarmie L Matlack
- Department of Genetics, The Waksman Institute Rutgers The State University of New Jersey, Piscataway, NJ, USA
| | - Christopher Rongo
- Department of Genetics, The Waksman Institute Rutgers The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
47
|
Wu Q, Cao X, Yan D, Wang D, Aballay A. Genetic Screen Reveals Link between the Maternal Effect Sterile Gene mes-1 and Pseudomonas aeruginosa-induced Neurodegeneration in Caenorhabditis elegans. J Biol Chem 2015; 290:29231-9. [PMID: 26475858 DOI: 10.1074/jbc.m115.674259] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence indicates that immune responses to microbial infections may contribute to neurodegenerative diseases. Here, we show that Pseudomonas aeruginosa infection of Caenorhabditis elegans causes a number of neural changes that are hallmarks of neurodegeneration. Using an unbiased genetic screen to identify genes involved in the control of P. aeruginosa-induced neurodegeneration, we identified mes-1, which encodes a receptor tyrosine kinase-like protein that is required for unequal cell divisions in the early embryonic germ line. We showed that sterile but not fertile mes-1 animals were resistant to neurodegeneration induced by P. aeruginosa infection. Similar results were observed using animals carrying a mutation in the maternal effect gene pgl-1, which is required for postembryonic germ line development, and the germ line-deficient strains glp-1 and glp-4. Additional studies indicated that the FOXO transcription factor DAF-16 is required for resistance to P. aeruginosa-induced neurodegeneration in germ line-deficient strains. Thus, our results demonstrate that P. aeruginosa infection results in neurodegeneration phenotypes in C. elegans that are controlled by the germ line in a cell-nonautonomous manner.
Collapse
Affiliation(s)
- Qiuli Wu
- From the Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 27710 and the Medical School of Southeast University, Nanjing 210009, China
| | - Xiou Cao
- From the Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 27710 and
| | - Dong Yan
- From the Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 27710 and
| | - Dayong Wang
- the Medical School of Southeast University, Nanjing 210009, China
| | - Alejandro Aballay
- From the Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina 27710 and
| |
Collapse
|
48
|
The evolving paradigm of cell-nonautonomous UPR-based regulation of immunity by cancer cells. Oncogene 2015; 35:269-78. [PMID: 25893303 DOI: 10.1038/onc.2015.108] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 12/19/2022]
Abstract
The endoplasmic reticulum (ER) stress response/unfolded protein response (UPR) has been thought to influence tumorigenesis mainly through cell-intrinsic, pro-survival effects. In recent years, however, new evidence has emerged showing that the UPR is also the source of cell-extrinsic effects, particularly directed at those immune cells within the tumor microenvironment. Here we will review and discuss this new body of information with focus on the role of cell-extrinsic effects on innate and adaptive immunity, suggesting that the transmission of ER stress from cancer cells to myeloid cells in particular is an expedient used by cancer cells to control the immune microenvironment, which acquires pro-inflammatory as well as immune-suppressive characteristics. These new findings can now be seen in the broader context of similar phenomena described in Caenorhabditis elegans, and an analogy with quorum sensing and 'community effects' in prokaryotes and eukaryotes can be drawn, arguing that a cell-nonautonomous UPR-based regulation of heterologous cells may be phylogenetically conserved. Finally, we will discuss the role of aneuploidy as an inducer of proteotoxic stress and potential initiator of cell-nonautonomous UPR-based regulation. In presenting these new views, we wish to bring attention to the cell-extrinsic regulation of tumor growth, including tumor UPR-based cell-nonautonomous signaling as a mechanism of maintaining tumor heterogeneity and resistance to therapy, and suggest therapeutically targeting such mechanisms within the tumor microenvironment.
Collapse
|
49
|
Abstract
In eukaryotic cells, protein folding and modification in the endoplasmic reticulum (ER) is highly sensitive to disturbances of homeostasis. The accumulation of unfolded and misfolded proteins in the ER lumen, termed ER stress, activates intracellular signaling pathways to resolve the protein-folding defect. This unfolded protein response (UPR) increases the capacity of ER protein folding, reduces global protein synthesis, and activates ER-associated protein degradation. If ER stress is too severe or chronic, or the UPR is compromised and not able to restore ER protein-folding homeostasis, numerous apoptotic signaling pathways are activated. Preclinical and clinical studies in the past decade indicate that ER stress and the UPR have a significant impact on the pathogenesis of inflammatory bowel disease. Paneth and goblet cells, 2 epithelial cell populations in the gut, rely on a robust ER function for protein folding and secretion. Several immune cells are orchestrated by ER stress and the UPR for differentiation, activation, migration, and survival. In addition, a variety of exogenous and endogenous molecules in the intestinal lumen affect ER function, making ER stress and the UPR relevant cellular signals in intestinal homeostasis. Recent studies demonstrated that unresolved ER stress and/or dysregulated UPR may cause inflammatory bowel disease by inducing epithelial cell death, impairing mucosal barrier function, and activating proinflammatory response in the gut. With our increased understanding of ER stress in inflammatory bowel disease pathogenesis, it is now possible to develop novel therapies to improve ER protein-folding homeostasis and target-specific UPR pathways in cells residing in the intestinal mucosa.
Collapse
|
50
|
Schinzel R, Dillin A. Endocrine aspects of organelle stress—cell non-autonomous signaling of mitochondria and the ER. Curr Opin Cell Biol 2015; 33:102-10. [PMID: 25677685 DOI: 10.1016/j.ceb.2015.01.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 01/23/2015] [Indexed: 12/12/2022]
Abstract
Organisms have to cope with an unpredictable and dynamic environment. It is crucial for any living being to respond to these changes by buffering the effects on cellular homeostasis. Failure to appropriately respond to stress can have severe consequences for health and survival. Eukaryotic cells possess several organelle-specific stress responses to cope with this challenge. Besides their central role in stress resistance, these pathways have also been shown to be important in the regulation of proteome maintenance, development and longevity. Intriguingly, many of these effects seem to be controlled by only a subset of cells implying a systemic regulation in a cell non-autonomous manner. The understanding of the nature of this stress communication across tissues, its mechanisms and impact, will be paramount in understanding disease etiology and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Robert Schinzel
- Department of Molecular and Cell Biology, The University of California, Berkeley, Li Ka Shing Center for Biomedical and Health Sciences, USA
| | - Andrew Dillin
- Department of Molecular and Cell Biology, The University of California, Berkeley, Li Ka Shing Center for Biomedical and Health Sciences, USA; Howard Hughes Medical Institute, The University of California, Berkeley, USA.
| |
Collapse
|