1
|
Su L, Hu P, Luo X, Ding H, Zhang R, Qian Y, Qi S, Tian X, Ling W. Development and Application of a BODIPY Carbazole Derivative Probe for Lysosomal Imaging: Insights into Lysosomal Dynamics and Dysfunction in Inflammation-Related Diseases. ACS APPLIED MATERIALS & INTERFACES 2025; 17:607-616. [PMID: 39688339 DOI: 10.1021/acsami.4c17607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Inflammation is crucial in neurodegenerative and chronic diseases, including Alzheimer disease (AD) and liver fibrosis. To gain a deeper understanding of lysosomal functions in cellular physiology and disease mechanisms, we developed a carbazole-based BODIPY lysosomal probe, designated LysoI. This probe specifically targets lysosomes within 15 min and exhibits a Stokes shift of approximately 180 nm, enabling continuous incubation for up to 5 h without the need for washing steps. Interestingly, LysoI remained effective for long-term imaging, even up to 24 h poststaining. Despite varying pH values and conditions, such as autophagy, apoptosis, and inflammation, it consistently provides excellent lysosomal imaging. Notably, inflammation disrupts lysosomal morphology and motility, as evidenced by an increased size, a decrease in number, and a reduction in movement speed, as observed with LysoI. Furthermore, lysosomal rupture and impaired clearance may exacerbate inflammation and contribute to cellular apoptosis. These findings suggest that lysosomal dysfunction is closely associated with disease progression; therefore, protection and repair targeting lysosomes may offer promising strategies for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Liping Su
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Panyi Hu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, People's Republic of China
| | - Xinmei Luo
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Haitao Ding
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Rundong Zhang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Yeben Qian
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, People's Republic of China
| | - Shiqian Qi
- Department of Urology, Institute of Urology, State Key Laboratory of Biotherapy, West China Hospital, College of Life Sciences, Sichuan University, and National Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Xiaohe Tian
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| | - Wenwu Ling
- Department of Medical Ultrasound, State Key Laboratory of Biotherapy, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital of Sichuan University, Sichuan University, Chengdu 610000, Sichuan, People's Republic of China
| |
Collapse
|
2
|
Gabaldón C, Karakuzu O, Garsin DA. SKN-1 activation during infection of Caenorhabditis elegans requires CDC-48 and endoplasmic reticulum proteostasis. Genetics 2024; 228:iyae131. [PMID: 39166513 PMCID: PMC11538416 DOI: 10.1093/genetics/iyae131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
During challenge of Caenorhabditis elegans with human bacterial pathogens such as Pseudomonas aeruginosa and Enterococcus faecalis, the elicited host response can be damaging if not properly controlled. The activation of Nrf (nuclear factor erythroid-related factor)/CNC (Cap-n-collar) transcriptional regulators modulates the response by upregulating genes that neutralize damaging molecules and promote repair processes. Activation of the C. elegans Nrf ortholog, SKN-1, is tightly controlled by a myriad of regulatory mechanisms, but a central feature is an activating phosphorylation accomplished by the p38 mitogen-activated kinase (MAPK) cascade. In this work, loss of CDC-48, an AAA+ ATPase, was observed to severely compromise SKN-1 activation on pathogen and we sought to understand the mechanism. CDC-48 is part of the endoplasmic reticulum (ER)-associated degradation (ERAD) complex where it functions as a remodeling chaperone enabling the translocation of proteins from the ER to the cytoplasm for degradation by the proteosome. Interestingly, one of the proteins retrotranslocated by ERAD, a process necessary for its activation, is SKN-1A, the ER isoform of SKN-1. However, we discovered that SKN-1A is not activated by pathogen exposure in marked contrast to the cytoplasmic-associated isoform SKN-1C. Rather, loss of CDC-48 blocks the antioxidant response normally orchestrated by SKN-1C by strongly inducing the unfolded protein response (UPRER). The data are consistent with the model of these 2 pathways being mutually inhibitory and support the emerging paradigm in the field of coordinated cooperation between different stress responses.
Collapse
Affiliation(s)
- Carolaing Gabaldón
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ozgur Karakuzu
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Danielle A Garsin
- Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
3
|
Emser J, Wernet N, Hetzer B, Wohlmann E, Fischer R. The cysteine-rich virulence factor NipA of Arthrobotrys flagrans interferes with cuticle integrity of Caenorhabditis elegans. Nat Commun 2024; 15:5795. [PMID: 38987250 PMCID: PMC11237121 DOI: 10.1038/s41467-024-50096-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Animals protect themself from microbial attacks by robust skins or a cuticle as in Caenorhabditis elegans. Nematode-trapping fungi, like Arthrobotrys flagrans, overcome the cuticle barrier and colonize the nematode body. While lytic enzymes are important for infection, small-secreted proteins (SSPs) without enzymatic activity, emerge as crucial virulence factors. Here, we characterized NipA (nematode induced protein) which A. flagrans secretes at the penetration site. In the absence of NipA, A. flagrans required more time to penetrate C. elegans. Heterologous expression of the fungal protein in the epidermis of C. elegans led to blister formation. NipA contains 13 cysteines, 12 of which are likely to form disulfide bridges, and the remaining cysteine was crucial for blister formation. We hypothesize that NipA interferes with cuticle integrity to facilitate fungal entry. Genome-wide expression analyses of C. elegans expressing NipA revealed mis-regulation of genes associated with extracellular matrix (ECM) maintenance and innate immunity.
Collapse
Affiliation(s)
- Jennifer Emser
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Nicole Wernet
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Birgit Hetzer
- Max Rubner-Institut (MRI) - Federal Research Institute of Nutrition and Food, Haid-und-Neu-Strasse 9, Karlsruhe, 76131, Germany
| | - Elke Wohlmann
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany
| | - Reinhard Fischer
- Institute for Applied Biosciences. Department of Microbiology, Karlsruhe Institute of Technology (KIT) - South Campus, Fritz-Haber-Weg 4, Karlsruhe, 76131, Germany.
| |
Collapse
|
4
|
Pu X, Qi B. Lysosomal dysfunction by inactivation of V-ATPase drives innate immune response in C. elegans. Cell Rep 2024; 43:114138. [PMID: 38678555 DOI: 10.1016/j.celrep.2024.114138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/10/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024] Open
Abstract
Pathogens target vacuolar ATPase (V-ATPase) to inhibit lysosomal acidification or lysosomal fusion, causing lysosomal dysfunction. However, it remains unknown whether cells can detect dysfunctional lysosomes and initiate an immune response. In this study, we discover that dysfunction of lysosomes caused by inactivation of V-ATPase enhances innate immunity against bacterial infections. We find that lysosomal V-ATPase interacts with DVE-1, whose nuclear localization serves as a proxy for the induction of mitochondrial unfolded protein response (UPRmt). The inactivation of V-ATPase promotes the nuclear localization of DVE-1, activating UPRmt and inducing downstream immune response genes. Furthermore, pathogen resistance conferred by inactivation of V-ATPase requires dve-1 and its downstream immune effectors. Interestingly, animals grow slower after vha RNAi, suggesting that the vha-RNAi-induced immune response costs the most energy through activation of DVE-1, which trades off with growth. This study reveals how dysfunctional lysosomes can trigger an immune response, emphasizing the importance of conserving energy during immune defense.
Collapse
Affiliation(s)
- Xuepiao Pu
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Bin Qi
- Southwest United Graduate School, Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory of Conservation and Utilization of Bio-resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China.
| |
Collapse
|
5
|
Xiao Y, Zhou H, Cui Y, Zhu X, Li S, Yu C, Jiang N, Liu L, Liu F. Schisandrin A enhances pathogens resistance by targeting a conserved p38 MAPK pathway. Int Immunopharmacol 2024; 128:111472. [PMID: 38176342 DOI: 10.1016/j.intimp.2023.111472] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Schizandrin A (SA), also known as deoxyschizandrin, is one of the most biologically active lignans isolated from the traditional Chinese medicine Fructus schisandrae chinensis. Schisandrin A has proven benefits for anti-cancer, anti-inflammation, hepatoprotection, anti-oxidation, neuroprotection, anti-diabetes. But the influence of Schisandrin A to the innate immune response and its molecular mechanisms remain obscure. In this study, we found that Schisandrin A increased resistance to not only the Gram-negative pathogens Pseudomonas aeruginosa and Salmonella enterica but also the Gram-positive pathogen Listeria monocytogenes. Meanwhile, Schisandrin A protected the animals from the infection by enhancing the tolerance to the pathogens infection rather than by reducing the bacterial burden. Through the screening of the conserved immune pathways in Caenorhabditis elegans, we found that Schisandrin A enhanced innate immunity via p38 MAPK pathway. Furthermore, Schisandrin A increased the expression of antibacterial peptide genes, such as K08D8.5, lys-2, F35E12.5, T24B8.5, and C32H11.12 by activation PMK-1/p38 MAPK. Importantly, Schisandrin A-treated mice also enhanced resistance to P. aeruginosa PA14 infection and significantly increased the levels of active PMK-1. Thus, promoted PMK-1/p38 MAPK-mediated innate immunity by Schisandrin A is conserved from worms to mammals. Our work provides a conserved mechanism by which Schisandrin A enhances innate immune response and boosts its therapeutic application in the treatment of infectious diseases.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Hanlin Zhou
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Yingwen Cui
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Xinting Zhu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Nian Jiang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; Institute of Life Sciences, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Liu Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, Guizhou 563000, China; College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
6
|
VenkataKrishna LM, Balasubramaniam B, Sushmitha TJ, Ravichandiran V, Balamurugan K. Cronobacter sakazakii infection implicates multifaceted neuro-immune regulatory pathways of Caenorhabditis elegans. Mol Omics 2024; 20:48-63. [PMID: 37818754 DOI: 10.1039/d3mo00167a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
The neural pathways of Caenorhabditis elegans play a crucial role in regulating host immunity and inflammation during pathogenic infections. To understand the major neuro-immune signaling pathways, this study aimed to identify the key regulatory proteins in the host C. elegans during C. sakazakii infection. We used high-throughput label-free quantitative proteomics and identified 69 differentially expressed proteins. KEGG analysis revealed that C. sakazakii elicited host immune signaling cascades primarily including mTOR signaling, axon regeneration, metabolic pathways (let-363 and acox-1.4), calcium signaling (mlck-1), and longevity regulating pathways (ddl-2), respectively. The abrogation in functional loss of mTOR-associated players deciphered that C. sakazakii infection negatively regulated the lifespan of mutant worms (akt-1, let-363 and dlk-1), including physiological aberrations, such as reduced pharyngeal pumping and egg production. Additionally, the candidate pathway proteins were validated by transcriptional profiling of their corresponding genes. Furthermore, immunoblotting showed the downregulation of mTORC2/SGK-1 during the later hours of pathogen exposure. Overall, our findings profoundly provide an understanding of the specificity of proteome imbalance in affecting neuro-immune regulations during C. sakazakii infection.
Collapse
Affiliation(s)
| | | | - T J Sushmitha
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India.
| | - V Ravichandiran
- National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, India
| | | |
Collapse
|
7
|
Zhang Z, Gaetjens TK, Ou J, Zhou Q, Yu Y, Mallory DP, Abel SM, Yu Y. Propulsive cell entry diverts pathogens from immune degradation by remodeling the phagocytic synapse. Proc Natl Acad Sci U S A 2023; 120:e2306788120. [PMID: 38032935 PMCID: PMC10710034 DOI: 10.1073/pnas.2306788120] [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: 05/01/2023] [Accepted: 10/05/2023] [Indexed: 12/02/2023] Open
Abstract
Phagocytosis is a critical immune function for infection control and tissue homeostasis. During phagocytosis, pathogens are internalized and degraded in phagolysosomes. For pathogens that evade immune degradation, the prevailing view is that virulence factors are required to disrupt the biogenesis of phagolysosomes. In contrast, we present here that physical forces from motile pathogens during cell entry divert them away from the canonical degradative pathway. This altered fate begins with the force-induced remodeling of the phagocytic synapse formation. We used the parasite Toxoplasma gondii as a model because live Toxoplasma actively invades host cells using gliding motility. To differentiate the effects of physical forces from virulence factors in phagocytosis, we employed magnetic forces to induce propulsive entry of inactivated Toxoplasma into macrophages. Experiments and computer simulations show that large propulsive forces hinder productive activation of receptors by preventing their spatial segregation from phosphatases at the phagocytic synapse. Consequently, the inactivated parasites are engulfed into vacuoles that fail to mature into degradative units, similar to the live motile parasite's intracellular pathway. Using yeast cells and opsonized beads, we confirmed that this mechanism is general, not specific to the parasite used. These results reveal new aspects of immune evasion by demonstrating how physical forces during active cell entry, independent of virulence factors, enable pathogens to circumvent phagolysosomal degradation.
Collapse
Affiliation(s)
- Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Thomas K. Gaetjens
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN37996
| | - Jin Ou
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Qiong Zhou
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - D. Paul Mallory
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Steven M. Abel
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN37996
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| |
Collapse
|
8
|
Li K, Wei X, Li K, Zhang Q, Zhang J, Wang D, Yang J. Dietary restriction to optimize T cell immunity is an ancient survival strategy conserved in vertebrate evolution. Cell Mol Life Sci 2023; 80:219. [PMID: 37470873 PMCID: PMC11071854 DOI: 10.1007/s00018-023-04865-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Recent advances highlight a key role of transient fasting in optimizing immunity of human and mouse. However, it remains unknown whether this strategy is independently acquired by mammals during evolution or instead represents gradually evolved functions common to vertebrates. Using a tilapia model, we report that T cells are the main executors of the response of the immune system to fasting and that dietary restriction bidirectionally modulates T cell immunity. Long-term fasting impaired T cell immunity by inducing intense autophagy, apoptosis, and aberrant inflammation. However, transient dietary restriction triggered moderate autophagy to optimize T cell response by maintaining homeostasis, alleviating inflammation and tissue damage, as well as enhancing T cell activation, proliferation and function. Furthermore, AMPK is the central hub linking fasting and autophagy-controlled T cell immunity in tilapia. Our findings demonstrate that dietary restriction to optimize immunity is an ancient strategy conserved in vertebrate evolution, providing novel perspectives for understanding the adaptive evolution of T cell response.
Collapse
Affiliation(s)
- Kunming Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Qian Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ding Wang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
9
|
Rezaeianaran F, Gijs MAM. Difference in Intestine Content of Caenorhabditis elegans When Fed on Non-Pathogenic or Pathogenic Bacteria. MICROMACHINES 2023; 14:1386. [PMID: 37512697 PMCID: PMC10384281 DOI: 10.3390/mi14071386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
We investigated the bacterial food digestion and accumulation in wild-type adult Caenorhabditis elegans (C. elegans) worms that have fed on either non-pathogenic RFP-expressing Escherichia coli (E. coli) OP50 or pathogenic-RFP-expressing Pseudomonas aeruginosa (P. aeruginosa) PAO1 during the first 4 days of adulthood. Once the worms had completed their planned feeding cycles, they were loaded on microfluidic chips, where they were fixed to allow high-resolution z-stack fluorescence imaging of their intestines utilizing a Spinning Disk Confocal Microscope (SDCM) equipped with a high-resolution oil-immersion objective (60×). IMARIS software was used to visualize and analyze the obtained images, resulting in the production of three-dimensional constructs of the intestinal bacterial load. We discovered two distinct patterns for the bacteria-derived fluorescence signal in the intestine: (i) individual fluorescent spots, originating from intact bacteria, were present in the fluorescent E. coli-OP50-fed worms, and (ii) individual fluorescent spots (originating from intact bacteria) were dispersed in large regions of diffuse fluorescence (RDF), originating from disrupted bacteria, in fluorescent P. aeruginosa-PAO1-fed worms. We performed a semi-automated single-worm-resolution quantitative analysis of the intestinal bacterial load, which showed that the intestinal bacterial load generally increases with age of the worms, but more rapidly for the fluorescent P. aeruginosa-PAO1-fed worms.
Collapse
Affiliation(s)
- Farzad Rezaeianaran
- Laboratory of Microsystems, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Martin A M Gijs
- Laboratory of Microsystems, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
10
|
Huang XW, Lu S, Pan W, Zhong MZ, Chai JW, Liu YH, Zeng K, Xi LY. Autophagy benefits the in vitro and in vivo clearance of Talaromyces marneffei. Microb Pathog 2023; 180:106146. [PMID: 37150309 DOI: 10.1016/j.micpath.2023.106146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023]
Abstract
Talaromycosis, namely Talaromyces marneffei infection, is increasing gradually and has a high mortality rate even under antifungal therapy. Although autophagy acts differently on different pathogens, it is a promising therapeutic strategy. However, information on autophagy in macrophages and animals upon infection by T. marneffei is still limited. Therefore, several models were employed here to investigate the role of autophagy in host defense against T. marneffei, including RAW264.7 macrophages as in vitro models, different types of Caenorhabditis elegans and BALB/c mice as in vivo models. We applied the clinical T. marneffei isolate SUMS0152 in this study. T. marneffei-infected macrophages exhibit increased formation of autophagosomes. Further, macrophage autophagy promoted by rapamycin or Earle's balanced salt solution (EBSS) inhibited the viability of intracellular T. marneffei. In vivo, compared with uninfected Caenorhabditis elegans, the wild-type nematodes upregulated the expression of the autophagy-related gene lgg-1 and atg-18, and nematodes carrying GFP reporter were induced to form autophagosomes (GFP::LGG-1) after T. marneffei infection. Furthermore, the knockdown of lgg-1 significantly reduced the survival rate of T. marneffei-infected nematodes. Likewise, the autophagy activator rapamycin reduced the fungal burden and suppressed lung inflammation in a mouse model of infection. In conclusion, autophagy is essential for host defense against T. marneffei in vitro and in vivo. Therefore, autophagy may be an attractive target for developing new therapeutics to treat talaromycosis.
Collapse
Affiliation(s)
- Xiao-Wen Huang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sha Lu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 West Yanjiang Rd., Guangzhou, 510120, China
| | - Wen Pan
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Mei-Zhen Zhong
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jin-Wei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ying-Hui Liu
- Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Li-Yan Xi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 West Yanjiang Rd., Guangzhou, 510120, China; Dermatology Department, Dermatology Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Zhang Z, Gaetjens TK, Yu Y, Paul Mallory D, Abel SM, Yu Y. Propulsive cell entry diverts pathogens from immune degradation by remodeling the phagocytic synapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538287. [PMID: 37162866 PMCID: PMC10168248 DOI: 10.1101/2023.04.25.538287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Phagocytosis is a critical immune function for infection control and tissue homeostasis. This process is typically described as non-moving pathogens being internalized and degraded in phagolysosomes. For pathogens that evade immune degradation, the prevailing view is that virulence factors that biochemically disrupt the biogenesis of phagoslysosomes are required. In contrast, here we report that physical forces exerted by pathogens during cell entry divert them away from the canonical phagolysosomal degradation pathway, and this altered intracellular fate is determined at the time of phagocytic synapse formation. We used the eukaryotic parasite Toxoplasma gondii as a model because live Toxoplasma uses gliding motility to actively invade into host cells. To differentiate the effect of physical forces from that of virulence factors in phagocytosis, we developed a strategy that used magnetic forces to induce propulsive entry of inactivated Toxoplasma into macrophage cells. Experiments and computer simulations collectively reveal that large propulsive forces suppress productive activation of receptors by hindering their spatial segregation from phosphatases at the phagocytic synapse. Consequently, the inactivated parasites, instead of being degraded in phagolysosomes, are engulfed into vacuoles that fail to mature into degradative units, following an intracellular pathway strikingly similar to that of the live motile parasite. Using opsonized beads, we further confirmed that this mechanism is general, not specific to the parasite used. These results reveal previously unknown aspects of immune evasion by demonstrating how physical forces exerted during active cell entry, independent of virulence factors, can help pathogens circumvent phagolysosomal degradation.
Collapse
Affiliation(s)
- Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - Thomas K. Gaetjens
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996
| | - Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - D. Paul Mallory
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - Steven M. Abel
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| |
Collapse
|
12
|
Profile of Prof. Ke-Qin Zhang. SCIENCE CHINA. LIFE SCIENCES 2023; 66:436-438. [PMID: 36680677 DOI: 10.1007/s11427-022-2247-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
13
|
Dou T, Chen J, Wang R, Pu X, Wu H, Zhao Y. Complementary protective effects of autophagy and oxidative response against graphene oxide toxicity in Caenorhabditis elegans. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 248:114289. [PMID: 36379072 DOI: 10.1016/j.ecoenv.2022.114289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
Graphene oxide (GO) exposure may cause damage to C. elegans. However, the role of autophagy and its interactive effect with oxidative response in GO toxicity still remain largely unclear. In the present study, we investigated the protective role of autophagy against GO and its association with oxidative response using C. elegans as an in vivo system. Results indicated that GO exposure induced autophagy in a dose dependent manner in C. elegans. Autophagy inhibitor 3-methyladenine (3-MA) and silencing autophagy genes lgg-1, bec-1 and unc-51 exacerbated the toxicity of GO whereas autophagy activator rapamycin alleviated it. In addition, the antioxidant N-Acetyl-L-cysteine (NAC) effectively suppressed the toxicity of GO with increased resistance to oxidative stress. Worms with RNAi-induced antioxidative genes sod-1, sod-2, sod-3 and sod-4 knockdown were more sensitive to GO. 3-MA increased the expression of superoxide dismutase SOD-3 under GO exposure conditions and exacerbated the toxicity of GO under the anti-oxidation inaction condition by sod-3 RNAi. In contrast, NAC reduced autophagy levels in GO exposed nematodes and increased tolerance to GO in autophagy-defective worms. These results suggested that autophagy and antioxidative response provide complementary protection against GO in C. elegans.
Collapse
Affiliation(s)
- Tingting Dou
- School of Public Health, Bengbu Medical College, Bengbu, People's Republic of China
| | - Jingya Chen
- School of Public Health, Bengbu Medical College, Bengbu, People's Republic of China
| | - Rui Wang
- School of Public Health, Bengbu Medical College, Bengbu, People's Republic of China
| | - Xiaoxiao Pu
- School of Public Health, Bengbu Medical College, Bengbu, People's Republic of China
| | - Huazhang Wu
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, People's Republic of China.
| | - Yunli Zhao
- School of Public Health, Bengbu Medical College, Bengbu, People's Republic of China; Key Laboratory of Environmental Medicine Engineering in Ministry of Education, Medical School, Southeast University, Nanjing, People's Republic of China.
| |
Collapse
|
14
|
Bhat CG, Budhwar R, Godwin J, Dillman AR, Rao U, Somvanshi VS. RNA-Sequencing of Heterorhabditis nematodes to identify factors involved in symbiosis with Photorhabdus bacteria. BMC Genomics 2022; 23:741. [PMCID: PMC9639317 DOI: 10.1186/s12864-022-08952-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022] Open
Abstract
Background Nematodes are a major group of soil inhabiting organisms. Heterorhabditis nematodes are insect-pathogenic nematodes and live in a close symbiotic association with Photorhabdus bacteria. Heterorhabditis-Photorhabdus pair offers a powerful and genetically tractable model to study animal-microbe symbiosis. It is possible to generate symbiont bacteria free (axenic) stages in Heterorhabditis. Here, we compared the transcriptome of symbiotic early-adult stage Heterorhabditis nematodes with axenic early-adult nematodes to determine the nematode genes and pathways involved in symbiosis with Photorhabdus bacteria. Results A de-novo reference transcriptome assembly of 95.7 Mb was created for H. bacteriophora by using all the reads. The assembly contained 46,599 transcripts with N50 value of 2,681 bp and the average transcript length was 2,054 bp. The differentially expressed transcripts were identified by mapping reads from symbiotic and axenic nematodes to the reference assembly. A total of 754 differentially expressed transcripts were identified in symbiotic nematodes as compared to the axenic nematodes. The ribosomal pathway was identified as the most affected among the differentially expressed transcripts. Additionally, 12,151 transcripts were unique to symbiotic nematodes. Endocytosis, cAMP signalling and focal adhesion were the top three enriched pathways in symbiotic nematodes, while a large number of transcripts coding for various responses against bacteria, such as bacterial recognition, canonical immune signalling pathways, and antimicrobial effectors could also be identified. Conclusions The symbiotic Heterorhabditis nematodes respond to the presence of symbiotic bacteria by expressing various transcripts involved in a multi-layered immune response which might represent non-systemic and evolved localized responses to maintain mutualistic bacteria at non-threatening levels. Subject to further functional validation of the identified transcripts, our findings suggest that Heterorhabditis nematode immune system plays a critical role in maintenance of symbiosis with Photorhabdus bacteria. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08952-4.
Collapse
Affiliation(s)
- Chaitra G. Bhat
- grid.418196.30000 0001 2172 0814Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, Delhi, 110012 India
| | - Roli Budhwar
- Bionivid Technology Private Limited, 209, 4th Cross Rd., B. Channasandra, Kasturi Nagar, Bengaluru, Karnataka 560043 India
| | - Jeffrey Godwin
- Bionivid Technology Private Limited, 209, 4th Cross Rd., B. Channasandra, Kasturi Nagar, Bengaluru, Karnataka 560043 India
| | - Adler R. Dillman
- grid.266097.c0000 0001 2222 1582Department of Nematology, University of California, Riverside, 92521 USA
| | - Uma Rao
- grid.418196.30000 0001 2172 0814Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, Delhi, 110012 India
| | - Vishal S. Somvanshi
- grid.418196.30000 0001 2172 0814Division of Nematology, ICAR-Indian Agricultural Research Institute, New Delhi, Delhi, 110012 India
| |
Collapse
|
15
|
The p38 MAPK/PMK-1 Pathway Is Required for Resistance to Nocardia farcinica Infection in Caenorhabditis elegance. Pathogens 2022; 11:pathogens11101071. [PMID: 36297128 PMCID: PMC9609018 DOI: 10.3390/pathogens11101071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/02/2022] Open
Abstract
Nocardia farcinica is an opportunistic pathogen that causes nocardiosis primarily in patients with compromised immune systems. In this study, we used the genetically tractable organism Caenorhabditis elegans as a model to study the innate immune responses to N. farcinica infection. We found that unlike other pathogenic bacteria such as Pseudomonas aeruginosa and Staphylococcus aureus, N. farcinica failed to kill adult worms. In another words, adult worms exposed to N. farcinica exhibited a normal lifespan, compared with those fed the standard laboratory food bacterium Escherichia coli OP50. Interestingly, deletion of three core genes (pmk-1, nsy-1 and sek-1) in the p38 MAPK/PMK-1 pathway reduced the survival of worm exposure to N. farcinica, highlighting a crucial role of this pathway for C. elegans in resistance to N. farcinica. Furthermore, our results revealed that N. farcinica exposure up-regulated the level of PMK-1 phosphorylation. The activation of PMK-1 promoted nuclear translocation of a transcription factor SKN-1/Nrf2, which in turn mediated N. farcinica infection resistance in C. elegans. Our results provide an excellent example that the integrity of immune system is key aspect for counteract with pathogenesis of N. farcinica.
Collapse
|
16
|
Reza RN, Serra ND, Detwiler AC, Hanna-Rose W, Crook M. Noncanonical necrosis in 2 different cell types in a Caenorhabditis elegans NAD+ salvage pathway mutant. G3 (BETHESDA, MD.) 2022; 12:jkac033. [PMID: 35143646 PMCID: PMC8982427 DOI: 10.1093/g3journal/jkac033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 01/27/2022] [Indexed: 11/17/2022]
Abstract
Necrosis was once described as a chaotic unregulated response to cellular insult. We now know that necrosis is controlled by multiple pathways in response to many different cellular conditions. In our pnc-1 NAD+ salvage deficient Caenorhabditis elegans model excess nicotinamide induces excitotoxic death in uterine-vulval uv1 cells and OLQ mechanosensory neurons. We sought to characterize necrosis in our pnc-1 model in the context of well-characterized necrosis, apoptosis, and autophagy pathways in C. elegans. We confirmed that calpain and aspartic proteases were required for uv1 necrosis, but changes in intracellular calcium levels and autophagy were not, suggesting that uv1 necrosis occurs by a pathway that diverges from mec-4d-induced touch cell necrosis downstream of effector aspartic proteases. OLQ necrosis does not require changes in intracellular calcium, the function of calpain or aspartic proteases, or autophagy. Instead, OLQ survival requires the function of calreticulin and calnexin, pro-apoptotic ced-4 (Apaf1), and genes involved in both autophagy and axon guidance. In addition, the partially OLQ-dependent gentle nose touch response decreased significantly in pnc-1 animals on poor quality food, further suggesting that uv1 and OLQ necrosis differ downstream of their common trigger. Together these results show that, although phenotypically very similar, uv1, OLQ, and touch cell necrosis are very different at the molecular level.
Collapse
Affiliation(s)
- Rifath N Reza
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Nicholas D Serra
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ariana C Detwiler
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA
| | - Wendy Hanna-Rose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Matt Crook
- Department of Life Sciences, Texas A&M University-San Antonio, San Antonio, TX 78224, USA
| |
Collapse
|
17
|
Subsequent infection differentially affects the proteome of Caenorhabditis elegans by abrogating the intestinal cell proliferation. Microb Pathog 2021; 162:105350. [PMID: 34952153 DOI: 10.1016/j.micpath.2021.105350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
With a wide range of bacterial infections growing, it has become a big challenge to the research field to combat the newly emerging diseases. Immuno-compromised patients are vulnerable to opportunistic infections. P. mirabilis, an opportunistic pathogen infects the nematode when the immune system is compromised. In the present study, the C. elegans was pre-exposed to S. aureus for a short term, and then consecutively infected with P. mirabilis. The primary infection caused by S. aureus makes the immune system of C. elegans vulnerable making it easy for P. mirabilis to colonize efficiently during subsequent exposure, thereby stimulating the immune system of the nematode. In this study, the C. elegans exposed to the pathogens (S. aureus 4 h/P. mirabilis 40 h and S. aureus 8 h/P. mirabilis 60 h time points) showed a substantial difference in the banding patterns of SDS-PAGE gel, when compared to their respective OP50 fed controls. 2-DE identified a total of 235 proteins from all the time points which had >2 fold regulation. The regulated protein spots were identified by MALDI-ToF/ToF analysis and one common protein CDC-25.1 was found to be regulated in all the comparative time points. CDC-25.1 seemed to down regulate during subsequent infection and up regulate in single infection. The transcriptomic regulation of cdc-25.1 also reflects the protein regulation. In addition to it, survival assay in cdc-25.1 mutant nematodes confirm the susceptibility of host during subsequent infection.
Collapse
|
18
|
Lee S, Zhang Z, Yu Y. Real‐Time Simultaneous Imaging of Acidification and Proteolysis in Single Phagosomes Using Bifunctional Janus‐Particle Probes. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202111094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Seonik Lee
- Department of Chemistry Indiana University—Bloomington Bloomington IN 47405 USA
| | - Zihan Zhang
- Department of Chemistry Indiana University—Bloomington Bloomington IN 47405 USA
| | - Yan Yu
- Department of Chemistry Indiana University—Bloomington Bloomington IN 47405 USA
| |
Collapse
|
19
|
Lee S, Zhang Z, Yu Y. Real-Time Simultaneous Imaging of Acidification and Proteolysis in Single Phagosomes Using Bifunctional Janus-Particle Probes. Angew Chem Int Ed Engl 2021; 60:26734-26739. [PMID: 34624158 PMCID: PMC8648996 DOI: 10.1002/anie.202111094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Indexed: 12/15/2022]
Abstract
The digestion of pathogens inside phagosomes by immune cells occurs through a sequence of reactions including acidification and proteolysis, but how the reactions are orchestrated in the right order is unclear due to a lack of methods to simultaneously measure more than one reaction in phagosomes. Here we report a bifunctional Janus-particle probe to simultaneously monitor acidification and proteolysis in single phagosomes in live cells. Each probe consists of a pH reporter and a proteolysis reporter that are spatially separated but function concurrently. Using the Janus probes, we found the acidic pH needed to initiate and maintain proteolysis, revealing the mechanism for the sequential occurrence of both reactions during pathogen digestion. We showed how bacterium-derived lipopolysaccharides alter the acidification and proteolysis in phagosomes. This study showcases Janus-particle probes as a generally applicable tool for monitoring multiple reactions in intracellular vesicles.
Collapse
Affiliation(s)
- Seonik Lee
- Department of Chemistry, Indiana University-Bloomington, Bloomington, IN, 47405, USA
| | - Zihan Zhang
- Department of Chemistry, Indiana University-Bloomington, Bloomington, IN, 47405, USA
| | - Yan Yu
- Department of Chemistry, Indiana University-Bloomington, Bloomington, IN, 47405, USA
| |
Collapse
|
20
|
Garcia-Sanchez JA, Ewbank JJ, Visvikis O. Ubiquitin-related processes and innate immunity in C. elegans. Cell Mol Life Sci 2021; 78:4305-4333. [PMID: 33630111 PMCID: PMC11072174 DOI: 10.1007/s00018-021-03787-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Innate immunity is an evolutionary ancient defence strategy that serves to eliminate infectious agents while maintaining host health. It involves a complex network of sensors, signaling proteins and immune effectors that detect the danger, then relay and execute the immune programme. Post-translational modifications relying on conserved ubiquitin and ubiquitin-like proteins are an integral part of the system. Studies using invertebrate models of infection, such as the nematode Caenorhabditis elegans, have greatly contributed to our understanding of how ubiquitin-related processes act in immune sensing, regulate immune signaling pathways, and participate to host defence responses. This review highlights the interest of working with a genetically tractable model organism and illustrates how C. elegans has been used to identify ubiquitin-dependent immune mechanisms, discover novel ubiquitin-based resistance strategies that mediate pathogen clearance, and unravel the role of ubiquitin-related processes in tolerance, preserving host fitness during pathogen attack. Special emphasis is placed on processes that are conserved in mammals.
Collapse
Affiliation(s)
- Juan A Garcia-Sanchez
- INSERM, C3M, Côte D'Azur University, Nice, France
- INSERM, CNRS, CIML, Turing Centre for Living Systems, Aix-Marseille University, Marseille, France
| | - Jonathan J Ewbank
- INSERM, CNRS, CIML, Turing Centre for Living Systems, Aix-Marseille University, Marseille, France.
| | | |
Collapse
|
21
|
Ma YC, Dai LL, Qiu BB, Zhou Y, Zhao YQ, Ran Y, Zhang KQ, Zou CG. TOR functions as a molecular switch connecting an iron cue with host innate defense against bacterial infection. PLoS Genet 2021; 17:e1009383. [PMID: 33657091 PMCID: PMC7928448 DOI: 10.1371/journal.pgen.1009383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 01/26/2021] [Indexed: 01/22/2023] Open
Abstract
As both host and pathogen require iron for survival, iron is an important regulator of host-pathogen interactions. However, the molecular mechanism by which how the availability of iron modulates host innate immunity against bacterial infections remains largely unknown. Using the metazoan Caenorhabditis elegans as a model, we demonstrate that infection with a pathogenic bacterium Salmonella enterica serovar Typhimurium induces autophagy by inactivating the target of rapamycin (TOR). Although the transcripts of ftn-1 and ftn-2 encoding two H-ferritin subunits are upregulated upon S. Typhimurium infection, the ferritin protein is kept at a low level due to its degradation mediated by autophagy. Autophagy, but not ferritin, is required for defense against S. Typhimurium infection under normal circumstances. Increased abundance of iron suppresses autophagy by activating TOR, leading to an increase in the ferritin protein level. Iron sequestration, but not autophagy, becomes pivotal to protect the host from S. Typhimurium infection in the presence of exogenous iron. Our results show that TOR acts as a regulator linking iron availability with host defense against bacterial infection. Iron, an essential nutrient for both hosts and pathogens, can impact host-pathogen interactions. Thus, depending on availability of iron, hosts may use distinct strategies to defend against bacterial infections. Using the model organism Caenorhabditis elegans, we show that autophagy is activated and required for resistance to S. Typhimurium infection under normal conditions. In the presence of exogenous iron, autophagy is suppressed, leading to an increase in the protein level of ferritin, which in turn protects worms against S. Typhimurium infection. Finally, our data demonstrate that TOR is a molecular switch for turning autophagy and ferritin on and off. Thus, our study reveals a previously unknown role of TOR in modulating the transition between autophagy and iron sequestration in response to a bacterial infection via sensing an iron cue.
Collapse
Affiliation(s)
- Yi-Cheng Ma
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Li-Li Dai
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- School of Agronomy and Life Sciences, Kunming University, Kunming, Yunnan, China
| | - Bei-Bei Qiu
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ying Zhou
- College of Chemical Science and Technology, Yunnan University, Kunming, Yunnan, China
| | - Yu-Qiang Zhao
- College of Chemical Science and Technology, Yunnan University, Kunming, Yunnan, China
| | - Yu Ran
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ke-Qin Zhang
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- * E-mail: (K-QZ); (C-GZ)
| | - Cheng-Gang Zou
- State Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
- * E-mail: (K-QZ); (C-GZ)
| |
Collapse
|
22
|
Yang Y, Huang X, Yuan W, Xiang Y, Guo X, Wei W, Soberón M, Bravo A, Liu K. Bacillus thuringiensis cry toxin triggers autophagy activity that may enhance cell death. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2021; 171:104728. [PMID: 33357550 DOI: 10.1016/j.pestbp.2020.104728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/09/2020] [Accepted: 10/10/2020] [Indexed: 06/12/2023]
Abstract
Although it is well known that Bacillus thuringiensis Cry toxins kill insect pest by disrupting midgut cells of susceptible larvae through their pore formation activity, it is not clear what intracellular events are triggered after pore formation on the cell membrane of the target cells. Here we analyzed the role of Cry toxins on autophagy activation using several cell lines as models as well as in Helicoverpa armigera larvae. The selected insect cell lines (Hi5, Sl-HP and Sf9) were susceptible to activated Cry1Ca toxin, but only Sl-HP cells were also susceptible to activated Cry1Ac toxin. In contrast, the mammalian cell line 293 T was not susceptible to Cry1Ac or to Cry1Ca. Results show that Cry toxins induced autophagy only in the susceptible cell lines as shown by the analysis of the changes in the ratio of Atg8-PE to Atg8 and by formation of autophagosome dots containing Atg8-PE. The Cry1Ac enhanced autophagy in the midgut tissue of H. armigera larvae. Silencing expression of specific genes by RNAi assays confirmed that the autophagy induced by activated Cry toxins was dependent on AMPK and JNK pathways. Finally, inhibition of autophagy in the cell lines by specific inhibitors or RNAi assays resulted in delayed cell death triggered by Cry toxins, suggesting that the increased autophagy activity observed after toxin intoxication may contribute to cell death.
Collapse
Affiliation(s)
- Yongbo Yang
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Xiaoying Huang
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Wanli Yuan
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Yang Xiang
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Xueqin Guo
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Wei Wei
- School of Life Sciences, Central China Normal University, Wuhan 430070, China
| | - Mario Soberón
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apdo. Postal 510-3, Cuernavaca 62250, Morelos, Mexico
| | - Alejandra Bravo
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Apdo. Postal 510-3, Cuernavaca 62250, Morelos, Mexico
| | - Kaiyu Liu
- School of Life Sciences, Central China Normal University, Wuhan 430070, China,.
| |
Collapse
|
23
|
Goswamy D, Irazoqui JE. A unifying hypothesis on the central role of reactive oxygen species in bacterial pathogenesis and host defense in C. elegans. Curr Opin Immunol 2020; 68:9-20. [PMID: 32898751 DOI: 10.1016/j.coi.2020.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 01/06/2023]
Abstract
During intestinal infection, microbes induce ROS by various mechanisms in C. elegans. ROS can have beneficial roles, acting as antimicrobials and as signaling molecules that activate cytoprotective pathways. Failure to maintain appropriate levels of ROS causes oxidative stress and cellular damage. This review uses the Damage Response Framework to interpret several recent observations on the relationships between infection, host response, and host damage, with a focus on mechanisms mediated by ROS. We propose a unifying hypothesis that ROS drive a collapse in proteostasis in infected C. elegans, which results in death during unresolved infection. Because the signaling pathways highlighted here are conserved in mammals, the mentioned and future studies can provide new tools of hypothesis generation in human health and disease.
Collapse
Affiliation(s)
- Debanjan Goswamy
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, United States; Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Javier E Irazoqui
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, United States; Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, MA 01605, United States.
| |
Collapse
|
24
|
Zárate-Potes A, Yang W, Pees B, Schalkowski R, Segler P, Andresen B, Haase D, Nakad R, Rosenstiel P, Tetreau G, Colletier JP, Schulenburg H, Dierking K. The C. elegans GATA transcription factor elt-2 mediates distinct transcriptional responses and opposite infection outcomes towards different Bacillus thuringiensis strains. PLoS Pathog 2020; 16:e1008826. [PMID: 32970778 PMCID: PMC7513999 DOI: 10.1371/journal.ppat.1008826] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
The nematode Caenorhabditis elegans has been extensively used as a model for the study of innate immune responses against bacterial pathogens. While it is well established that the worm mounts distinct transcriptional responses to different bacterial species, it is still unclear in how far it can fine-tune its response to different strains of a single pathogen species, especially if the strains vary in virulence and infection dynamics. To rectify this knowledge gap, we systematically analyzed the C. elegans response to two strains of Bacillus thuringiensis (Bt), MYBt18247 (Bt247) and MYBt18679 (Bt679), which produce different pore forming toxins (PFTs) and vary in infection dynamics. We combined host transcriptomics with cytopathological characterizations and identified both a common and also a differentiated response to the two strains, the latter comprising almost 10% of the infection responsive genes. Functional genetic analyses revealed that the AP-1 component gene jun-1 mediates the common response to both Bt strains. In contrast, the strain-specific response is mediated by the C. elegans GATA transcription factor ELT-2, a homolog of Drosophila SERPENT and vertebrate GATA4-6, and a known master regulator of intestinal responses in the nematode. elt-2 RNAi knockdown decreased resistance to Bt679, but remarkably, increased survival on Bt247. The elt-2 silencing-mediated increase in survival was characterized by reduced intestinal tissue damage despite a high pathogen burden and might thus involve increased tolerance. Additional functional genetic analyses confirmed the involvement of distinct signaling pathways in the C. elegans defense response: the p38-MAPK pathway acts either directly with or in parallel to elt-2 in mediating resistance to Bt679 infection but is not required for protection against Bt247. Our results further suggest that the elt-2 silencing-mediated increase in survival on Bt247 is multifactorial, influenced by the nuclear hormone receptors NHR-99 and NHR-193, and may further involve lipid metabolism and detoxification. Our study highlights that the nematode C. elegans with its comparatively simple immune defense system is capable of generating a differentiated response to distinct strains of the same pathogen species. Importantly, our study provides a molecular insight into the diversity of biological processes that are influenced by a single master regulator and jointly determine host survival after pathogen infection.
Collapse
Affiliation(s)
- Alejandra Zárate-Potes
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Wentao Yang
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Barbara Pees
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Rebecca Schalkowski
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Philipp Segler
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Bentje Andresen
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Daniela Haase
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Rania Nakad
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute for Clinical Molecular Biology (IKMB), Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Guillaume Tetreau
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | | | - Hinrich Schulenburg
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Max Planck Institute for Evolutionary Biology, Ploen, Germany
| | - Katja Dierking
- Department of Evolutionary Ecology and Genetics, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
25
|
YAP in epithelium senses gut barrier loss to deploy defenses against pathogens. PLoS Pathog 2020; 16:e1008766. [PMID: 32857822 PMCID: PMC7454999 DOI: 10.1371/journal.ppat.1008766] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/01/2020] [Indexed: 12/24/2022] Open
Abstract
Pathogens commonly disrupt the intestinal epithelial barrier; however, how the epithelial immune system senses the loss of intestinal barrier as a danger signal to activate self-defense is unclear. Through an unbiased approach in the model nematode Caenorhabditis elegans, we found that the EGL-44/TEAD transcription factor and its transcriptional activator YAP-1/YAP (Yes-associated protein) were activated when the intestinal barrier was disrupted by infections with the pathogenic bacterium Pseudomonas aeruginosa PA14. Gene Ontology enrichment analysis of the genes containing the TEAD-binding sites revealed that “innate immune response” and “defense response to Gram-negative bacterium” were two top significantly overrepresented terms. Genetic inactivation of yap-1 and egl-44 significantly reduced the survival rate and promoted bacterial accumulation in worms after bacterial infections. Furthermore, we found that disturbance of the E-cadherin-based adherens junction triggered the nuclear translocation and activation of YAP-1/YAP in the gut of worms. Although YAP is a major downstream effector of the Hippo signaling, our study revealed that the activation of YAP-1/YAP was independent of the Hippo pathway during disruption of intestinal barrier. After screening 10 serine/threonine phosphatases, we identified that PP2A phosphatase was involved in the activation of YAP-1/YAP after intestinal barrier loss induced by bacterial infections. Additionally, our study demonstrated that the function of YAP was evolutionarily conserved in mice. Our study highlights how the intestinal epithelium recognizes the loss of the epithelial barrier as a danger signal to deploy defenses against pathogens, uncovering an immune surveillance program in the intestinal epithelium. The intestinal epithelial barrier is an important line of defense against pathogenic bacteria infecting the intestine. Persistent bacterial infections can cause disruption of the intestinal barrier; however, how the epithelia immune system recognizes the loss of intestinal barrier as a danger signal to activate self-defense against pathogens is unclear. Using the nematode Caenorhabditis elegans as a model animal, we show that the EGL-44/TEAD transcription factor and its transcriptional activator YAP-1/YAP (Yes-associated protein) are activated when the intestinal barrier is disrupted by bacterial infections. Gene Ontology enrichment reveals that EGL-44/TEAD orchestrates a complex host response composed of innate immune response and defense response to Gram-negative bacteria. Furthermore, our data demonstrate that YAP-1/YAP and EGL-44/TEAD are required for resistance to infections with pathogenic bacteria when the intestinal barrier is disrupted in worms and mice. Our study reveals a novel strategy for the intestinal epithelium to sense danger through its internal architecture and initiate innate immunity.
Collapse
|
26
|
Wong SQ, Kumar AV, Mills J, Lapierre LR. C. elegans to model autophagy-related human disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:325-373. [PMID: 32620247 DOI: 10.1016/bs.pmbts.2020.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autophagy is a highly conserved degradation process that clears damaged intracellular macromolecules and organelles in order to maintain cellular health. Dysfunctional autophagy is fundamentally linked to the development of various human disorders and pathologies. The use of the nematode Caenorhabditis elegans as a model system to study autophagy has improved our understanding of its regulation and function in organismal physiology. Here, we review the genetic, functional, and regulatory conservation of the autophagy pathway in C. elegans and we describe tools to quantify and study the autophagy process in this incredibly useful model organism. We further discuss how these nematodes have been modified to model autophagy-related human diseases and underscore the important insights obtained from such models. Altogether, we highlight the strengths of C. elegans as an exceptional tool to understand the genetic and molecular foundations underlying autophagy-related human diseases.
Collapse
Affiliation(s)
- Shi Quan Wong
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Anita V Kumar
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Joslyn Mills
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Louis R Lapierre
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States.
| |
Collapse
|
27
|
Kim JY, Le TAN, Lee SY, Song DG, Hong SC, Cha KH, Lee JW, Pan CH, Kang K. 3,3'-Diindolylmethane Improves Intestinal Permeability Dysfunction in Cultured Human Intestinal Cells and the Model Animal Caenorhabditis elegans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9277-9285. [PMID: 31353906 DOI: 10.1021/acs.jafc.9b03039] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
3,3'-Diindolylmethane (DIM), a digestive metabolite originating from cruciferous vegetables, has dietary potential for the treatment of various human intestinal diseases. Although intestinal permeability dysfunction is closely related to the initiation and progression of human intestinal inflammatory diseases (IBDs), the effect of DIM on intestinal permeability is unclear. We evaluated the effect of DIM on the intestinal permeability of human intestinal cell monolayers and the animal model Caenorhabditis elegans, which were treated with IL-1β and Pseudomonas aeruginosa, respectively, to mimic IBD conditions. DIM substantially restored the intestinal permeability of differentiated Caco-2 cells by enhancing the expression of tight junction proteins (including occludin and ZO-1). Compared to the IL-1β single treatment (551.0 ± 49.0 Ω·cm2), DIM (10 μM) significantly increased the transepithelial electrical resistance (TEER) of Caco-2 cell monolayers (919.0 ± 66.4 Ω·cm2, p < 0.001). DIM also ameliorated the impaired intestinal permeability and extended the lifespan of C. elegans fed P. aeruginosa. The mean lifespan of DIM-treated worms (10.8 ± 1.3 days) was higher than that of control-treated worms (9.7 ± 1.1 days, p < 0.01). Thus, DIM is a potential nutraceutical candidate for the treatment of leaky gut syndrome by improving intestinal permeability.
Collapse
Affiliation(s)
- Joo Yeon Kim
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Tram Anh Ngoc Le
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - So Young Lee
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Dae-Geun Song
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Sung-Chul Hong
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
| | - Jae Wook Lee
- Natural Products Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| | - Kyungsu Kang
- Natural Product Informatics Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School , Korea University of Science and Technology (UST) , Seoul 02792 , Republic of Korea
| |
Collapse
|
28
|
Williams AB, Heider F, Messling JE, Rieckher M, Bloch W, Schumacher B. Restoration of Proteostasis in the Endoplasmic Reticulum Reverses an Inflammation-Like Response to Cytoplasmic DNA in Caenorhabditis elegans. Genetics 2019; 212:1259-1278. [PMID: 31248887 PMCID: PMC6707470 DOI: 10.1534/genetics.119.302422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/24/2019] [Indexed: 12/17/2022] Open
Abstract
Innate immune responses protect organisms against various insults, but may lead to tissue damage when aberrantly activated. In higher organisms, cytoplasmic DNA can trigger inflammatory responses that can lead to tissue degeneration. Simpler metazoan models could shed new mechanistic light on how inflammatory responses to cytoplasmic DNA lead to pathologies. Here, we show that in a DNase II-defective Caenorhabditis elegans strain, persistent cytoplasmic DNA leads to systemic tissue degeneration and loss of tissue functionality due to impaired proteostasis. These pathological outcomes can be therapeutically alleviated by restoring protein homeostasis, either via ectopic induction of the ER unfolded protein response or N-acetylglucosamine treatment. Our results establish C. elegans as an ancestral metazoan model for studying the outcomes of inflammation-like conditions caused by persistent cytoplasmic DNA and provide insight into potential therapies for human conditions involving chronic inflammation.
Collapse
Affiliation(s)
- Ashley B Williams
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931, Germany
| | - Felix Heider
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931, Germany
| | - Jan-Erik Messling
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931, Germany
| | - Matthias Rieckher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, German Sports University, 50933 Cologne, Germany
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University of Cologne, 50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931, Germany
| |
Collapse
|
29
|
Balla KM, Lažetić V, Troemel ER. Natural variation in the roles of C. elegans autophagy components during microsporidia infection. PLoS One 2019; 14:e0216011. [PMID: 31013330 PMCID: PMC6478341 DOI: 10.1371/journal.pone.0216011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/14/2022] Open
Abstract
Natural genetic variation can determine the outcome of an infection, and often reflects the co-evolutionary battle between hosts and pathogens. We previously found that a natural variant of the nematode Caenorhabditis elegans from Hawaii (HW) has increased resistance against natural microsporidian pathogens in the Nematocida genus, when compared to the standard laboratory strain of N2. In particular, HW animals can clear infection, while N2 animals cannot. In addition, HW animals have lower levels of initial colonization of Nematocida inside intestinal cells, compared to N2. Here we investigate how this natural variation in resistance relates to autophagy. We found that there is much better targeting of autophagy-related machinery to parasites under conditions where they are cleared. In particular, ubiquitin targeting to Nematocida cells correlates very well with their subsequent clearance in terms of timing, host strain and age, as well as species of Nematocida. Furthermore, clearance correlates with targeting of the LGG-2/LC3 autophagy protein to parasite cells, with HW animals having much more efficient targeting of LGG-2 to parasite cells than N2 animals. Surprisingly, however, we found that LGG-2 is not required to clear infection. Instead, we found that LGG-2/LC3 regulates Nematocida colonization inside intestinal cells. Interestingly, LGG-2/LC3 regulates intracellular colonization only in the HW strain, and not in N2. Altogether these results demonstrate that there is natural genetic variation in an LGG-2-dependent process that regulates microsporidia colonization inside intestinal cells, although not microsporidia clearance.
Collapse
Affiliation(s)
- Keir M. Balla
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Vladimir Lažetić
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| | - Emily R. Troemel
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
30
|
Wang Q, Zhou Y, Fu R, Zhu Y, Song B, Zhong Y, Wu S, Shi Y, Wu Y, Su Y, Zhang H, He Y. Distinct autophagy-inducing abilities of similar-sized nanoparticles in cell culture and live C. elegans. NANOSCALE 2018; 10:23059-23069. [PMID: 30511716 DOI: 10.1039/c8nr05851b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nanomaterial-induced autophagy has raised increasing concerns. A variety of nanomaterials, conventional or recently emerged, have the capability of inducing autophagy. As a consequence, it is becoming a popular belief that induction of autophagy is a common response of cells upon exposure to nanoscale materials. In order to clarify whether the "nanoscale" size is the determining factor for the nanomaterials to induce autophagy, we utilized in vitro cultured cells and an in vivo Caenorhabditis elegans (C. elegans) model to systemically investigate the autophagy-inducing ability of nanomaterials. We selected four types of representative nanomaterials with similar sizes, namely silicon nanoparticles (SiNPs), CdTe quantum dots (QDs), carbon dots (CDs) and gold nanoparticles (AuNPs). We demonstrated that, unlike most other nanomaterials tested, no autophagosome formation was detected in cultured cells or in live C. elegans with SiNP treatment. The expression of autophagy-related genes and the lipidation of LGG-1/LC3 in cells and C. elegans also remained unchanged after the treatment of SiNPs. In addition, the ability of the nanomaterials to induce autophagy appeared to correlate with those to incur subcellular organelle damage. Together, our studies demonstrate that SiNPs do not induce autophagy in vitro or in vivo in the selected model organisms and cell lines, thus clarifying that the "nanoscale" size is not the determining factor for the nanomaterials to induce autophagy. The results also suggest that the autophagy-inducing ability of most nanomaterials could be merely a reflection of their detrimental effect on cellular structures.
Collapse
Affiliation(s)
- Qin Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou, Jiangsu 215123, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Khan F, Jain S, Oloketuyi SF. Bacteria and bacterial products: Foe and friends to Caenorhabditis elegans. Microbiol Res 2018; 215:102-113. [DOI: 10.1016/j.micres.2018.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/11/2018] [Accepted: 06/24/2018] [Indexed: 02/07/2023]
|
32
|
Kuo CJ, Hansen M, Troemel E. Autophagy and innate immunity: Insights from invertebrate model organisms. Autophagy 2018; 14:233-242. [PMID: 29130360 DOI: 10.1080/15548627.2017.1389824] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy/autophagy is a fundamental intracellular degradation process with multiple roles in immunity, including direct elimination of intracellular microorganisms via 'xenophagy.' In this review, we summarize studies from the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans that highlight the roles of autophagy in innate immune responses to viral, bacterial, and fungal pathogens. Research from these genetically tractable invertebrates has uncovered several conserved immunological paradigms, such as direct targeting of intracellular pathogens by xenophagy and regulation of autophagy by pattern recognition receptors in D. melanogaster. Although C. elegans has no known pattern recognition receptors, this organism has been particularly useful in understanding many aspects of innate immunity. Indeed, work in C. elegans was the first to show xenophagic targeting of microsporidia, a fungal pathogen that infects all animals, and to identify TFEB/HLH-30, a helix-loop-helix transcription factor, as an evolutionarily conserved regulator of autophagy gene expression and host tolerance. Studies in C. elegans have also highlighted the more recently appreciated relationship between autophagy and tolerance to extracellular pathogens. Studies of simple, short-lived invertebrates such as flies and worms will continue to provide valuable insights into the molecular mechanisms by which autophagy and immunity pathways intersect and their contribution to organismal survival. Abbreviations Atg autophagy related BECN1 Beclin 1 CALCOCO2 calcium binding and coiled-coil domain 2 Cry5B crystal toxin 5B Daf abnormal dauer formation DKF-1 D kinase family-1 EPG-7 Ectopic P Granules-7 FuDR fluorodeoxyuridine GFP green fluorescent protein HLH-30 Helix Loop Helix-30 Imd immune deficiency ins-18 INSulin related-18; LET-363, LEThal-363 lgg-1 LC3, GABARAP and GATE-16 family-1 MAPK mitogen-activated protein kinase MATH the meprin and TRAF homology MTOR mechanistic target of rapamycin NBR1 neighbor of BRCA1 gene 1 NFKB nuclear factor of kappa light polypeptide gene enhancer in B cells NOD nucleotide-binding oligomerization domain containing OPTN optineurin PAMPs pathogen-associated molecular patterns Park2 Parkinson disease (autosomal recessive, juvenile) 2, parkin pdr-1 Parkinson disease related PFTs pore-forming toxins PGRP peptidoglycan-recognition proteins PIK3C3 phosphatidylinositol 3- kinase catalytic subunit type 3 pink-1 PINK (PTEN-I induced kinase) homolog PRKD protein kinase D; PLC, phospholipase C PRKN parkin RBR E3 ubiquitin protein ligase PRRs pattern-recognition receptors PtdIns3P phosphatidylinositol-3-phosphate rab-5 RAB family-5 RB1CC1 RB1-inducible coiled-coil 1 RNAi RNA interference sqst SeQueSTosome related SQSTM1 sequestosome 1 TBK1 TANK-binding kinase 1 TFEB transcription factor EB TGFB/TGF-β transforming growth factor beta TLRs toll-like receptors unc-51 UNCoordinated-51 VPS vacuolar protein sorting; VSV, vesicular stomatitis virus VSV-G VSV surface glycoprotein G Wipi2 WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Cheng-Ju Kuo
- a Division of Biological Sciences , University of California , San Diego, La Jolla , CA , USA.,b Institute of Basic Medical Sciences , College of Medicine , National Cheng Kung University , Tainan , Taiwan
| | - Malene Hansen
- c Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging and Regeneration , La Jolla , CA , USA
| | - Emily Troemel
- a Division of Biological Sciences , University of California , San Diego, La Jolla , CA , USA
| |
Collapse
|
33
|
Balbi T, Cortese K, Ciacci C, Bellese G, Vezzulli L, Pruzzo C, Canesi L. Autophagic processes in Mytilus galloprovincialis hemocytes: Effects of Vibrio tapetis. FISH & SHELLFISH IMMUNOLOGY 2018; 73:66-74. [PMID: 29208501 DOI: 10.1016/j.fsi.2017.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 06/07/2023]
Abstract
Autophagy is a highly conserved and regulated catabolic process involved in maintaining cell homeostasis in response to different stressors. The autophagic machinery is also used as an innate immune mechanism against microbial infection. In invertebrates, that lack acquired immunity, autophagy may thus play a key role in the protection against potential pathogens. In aquatic molluscs, evidence has been provided for induction of autophagy by starvation and different environmental stressors; however, no information is available on autophagic pathways in the immune cells, the hemocytes. In this work, the autophagic processes were investigated in the hemocytes of the marine bivalve, the mussel Mytilus galloprovincialis. The effects of classical inducers/inhibitors of mammalian autophagy were first tested. Rapamycin induced a decrease in lysosomal membrane stability-LMS that was prevented by the autophagy inhibitor Wortmannin. Increased MDC fluorescence and expression of LC3-II were also observed. Moreover, responses to in vitro challenge with the bivalve pathogen Vibrio tapetis were evaluated. Mussel hemocytes were unable to activate the immune response towards V. tapetis; however, bacterial challenge induced a moderate decrease in LMS, corresponding to lysosomal activation but no cytotoxicity; the effect was prevented by Wortmannin. TEM observations showed that V. tapetis resulted in rapid formation of autophagosomes and autolysosomes. Accordingly, increased LC3-II expression, decreased levels of phosphorylated mTor and of p62 were observed. The results represent the first evidence for autophagic processes in bivalve hemocytes in response to bacterial challenge, and underline the protective role of autophagy towards potential pathogenic vibrios.
Collapse
Affiliation(s)
- Teresa Balbi
- Dept. of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Italy
| | - Katia Cortese
- Dept. of Experimental Medicine (DIMES), University of Genoa, Italy
| | - Caterina Ciacci
- Dept. of Biomolecular Sciences (DISBM), University of Urbino, Italy
| | - Grazia Bellese
- Dept. of Experimental Medicine (DIMES), University of Genoa, Italy
| | - Luigi Vezzulli
- Dept. of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Italy
| | - Carla Pruzzo
- Dept. of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Italy
| | - Laura Canesi
- Dept. of Earth, Environment and Life Sciences (DISTAV), University of Genoa, Italy.
| |
Collapse
|
34
|
Dierking K, Yang W, Schulenburg H. Antimicrobial effectors in the nematode Caenorhabditis elegans: an outgroup to the Arthropoda. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0299. [PMID: 27160601 DOI: 10.1098/rstb.2015.0299] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2016] [Indexed: 12/14/2022] Open
Abstract
Nematodes and arthropods likely form the taxon Ecdysozoa. Information on antimicrobial effectors from the model nematode Caenorhabditis elegans may thus shed light on the evolutionary origin of these defences in arthropods. This nematode species possesses an extensive armory of putative antimicrobial effector proteins, such as lysozymes, caenopores (or saposin-like proteins), defensin-like peptides, caenacins and neuropeptide-like proteins, in addition to the production of reactive oxygen species and autophagy. As C. elegans is a bacterivore that lives in microbe-rich environments, some of its effector peptides and proteins likely function in both digestion of bacterial food and pathogen elimination. In this review, we provide an overview of C. elegans immune effector proteins and mechanisms. We summarize the experimental evidence of their antimicrobial function and involvement in the response to pathogen infection. We further evaluate the microbe-induced expression of effector genes using WormExp, a recently established database for C. elegans gene expression analysis. We emphasize the need for further analysis at the protein level to demonstrate an antimicrobial activity of these molecules both in vitro and in vivoThis article is part of the themed issue 'Evolutionary ecology of arthropod antimicrobial peptides'.
Collapse
Affiliation(s)
- Katja Dierking
- Department of Evolutionary Ecology and Genetics, University of Kiel, Kiel 24098, Germany
| | - Wentao Yang
- Department of Evolutionary Ecology and Genetics, University of Kiel, Kiel 24098, Germany
| | - Hinrich Schulenburg
- Department of Evolutionary Ecology and Genetics, University of Kiel, Kiel 24098, Germany
| |
Collapse
|
35
|
Kamaladevi A, Balamurugan K. Global Proteomics Revealed Klebsiella pneumoniae Induced Autophagy and Oxidative Stress in Caenorhabditis elegans by Inhibiting PI3K/AKT/mTOR Pathway during Infection. Front Cell Infect Microbiol 2017; 7:393. [PMID: 28932706 PMCID: PMC5592217 DOI: 10.3389/fcimb.2017.00393] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/22/2017] [Indexed: 01/29/2023] Open
Abstract
The enterobacterium, Klebsiella pneumoniae invades the intestinal epithelium of humans by interfering with multiple host cell response. To uncover a system-level overview of host response during infection, we analyzed the global dynamics of protein profiling in Caenorhabditis elegans using quantitative proteomics approach. Comparison of protein samples of nematodes exposed to K. pneumoniae for 12, 24, and 36 h by 2DE revealed several changes in host proteome. A total of 266 host-encoded proteins were identified by 2DE MALDI-MS/MS and LC-MS/MS and the interacting partners of the identified proteins were predicted by STRING 10.0 analysis. In order to understand the interacting partners of regulatory proteins with similar or close pI ranges, a liquid IEF was performed and the isolated fractions containing proteins were identified by LC-MS/MS. Functional bioinformatics analysis on identified proteins deciphered that they were mostly related to the metabolism, dauer formation, apoptosis, endocytosis, signal transduction, translation, developmental, and reproduction process. Gene enrichment analysis suggested that the metabolic process as the most overrepresented pathway regulated against K. pneumoniae infection. The dauer-like formation in infected C. elegans along with intestinal atrophy and ROS during the physiological analysis indicated that the regulation of metabolic pathway is probably through the involvement of mTOR. Immunoblot analysis supported the above notion that the K. pneumoniae infection induced protein mis-folding in host by involving PI3Kinase/AKT-1/mTOR mediated pathway. Furthermore, the susceptibility of pdi-2, akt-1, and mTOR C. elegans mutants confirmed the role and involvement of PI3K/AKT/mTOR pathway in mediating protein mis-folding which appear to be translating the vulnerability of host defense toward K. pneumoniae infection.
Collapse
|
36
|
Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, Cuervo AM, Debnath J, Deretic V, Dikic I, Eskelinen EL, Fimia GM, Fulda S, Gewirtz DA, Green DR, Hansen M, Harper JW, Jäättelä M, Johansen T, Juhasz G, Kimmelman AC, Kraft C, Ktistakis NT, Kumar S, Levine B, Lopez-Otin C, Madeo F, Martens S, Martinez J, Melendez A, Mizushima N, Münz C, Murphy LO, Penninger JM, Piacentini M, Reggiori F, Rubinsztein DC, Ryan KM, Santambrogio L, Scorrano L, Simon AK, Simon HU, Simonsen A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Kroemer G. Molecular definitions of autophagy and related processes. EMBO J 2017; 36:1811-1836. [PMID: 28596378 PMCID: PMC5494474 DOI: 10.15252/embj.201796697] [Citation(s) in RCA: 1176] [Impact Index Per Article: 147.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/15/2022] Open
Abstract
Over the past two decades, the molecular machinery that underlies autophagic responses has been characterized with ever increasing precision in multiple model organisms. Moreover, it has become clear that autophagy and autophagy-related processes have profound implications for human pathophysiology. However, considerable confusion persists about the use of appropriate terms to indicate specific types of autophagy and some components of the autophagy machinery, which may have detrimental effects on the expansion of the field. Driven by the overt recognition of such a potential obstacle, a panel of leading experts in the field attempts here to define several autophagy-related terms based on specific biochemical features. The ultimate objective of this collaborative exchange is to formulate recommendations that facilitate the dissemination of knowledge within and outside the field of autophagy research.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Université Paris Descartes/Paris V, Paris, France
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - José Manuel Bravo-San Pedro
- Université Paris Descartes/Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Francesco Cecconi
- Department of Biology, University of Tor Vergata, Rome, Italy
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pediatric Hematology and Oncology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Augustine M Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Charleen T Chu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrice Codogno
- Université Paris Descartes/Paris V, Paris, France
- Institut Necker-Enfants Malades (INEM), Paris, France
- INSERM, U1151, Paris, France
- CNRS, UMR8253, Paris, France
| | - Maria Isabel Colombo
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología (IHEM)-CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jayanta Debnath
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ivan Dikic
- Institute of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt Main, Germany
- Department of Immunology and Medical Genetics, University of Split School of Medicine, Split, Croatia
| | | | - Gian Maria Fimia
- National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David A Gewirtz
- Department of Pharmacology and Toxicology and Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Gabor Juhasz
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
- Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Alec C Kimmelman
- Department of Radiation Oncology, Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY, USA
| | - Claudine Kraft
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
| | | | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Beth Levine
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute (HHMI), Dallas, TX, USA
| | - Carlos Lopez-Otin
- Department de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación en Red de Cáncer, Oviedo, Spain
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Sascha Martens
- Max F. Perutz Laboratories, Department of Biochemistry and Cell Biology, Vienna Biocenter, University of Vienna, Vienna, Austria
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Alicia Melendez
- Department of Biology, Queens College, Queens, NY, USA
- Graduate Center, City University of New York, New York, NY, USA
| | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zurich, Switzerland
| | - Leon O Murphy
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Campus Vienna BioCentre, Vienna, Austria
| | - Mauro Piacentini
- Department of Biology, University of Tor Vergata, Rome, Italy
- National Institute for Infectious Diseases "L. Spallanzani" IRCCS, Rome, Italy
| | - Fulvio Reggiori
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences Osaka University, Osaka, Japan
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qing Zhong
- Center for Autophagy Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| |
Collapse
|
37
|
Fu Q, Chen K, Zhu Q, Wang W, Huang F, Miao L, Wu X. β-catenin promotes intracellular bacterial killing via suppression of Pseudomonas aeruginosa-triggered macrophage autophagy. J Int Med Res 2017; 45:556-569. [PMID: 28415949 PMCID: PMC5536651 DOI: 10.1177/0300060517692147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Objective To investigate β-catenin-mediated bacterial elimination during Pseudomonas aeruginosa infection of macrophage-like RAW264.7 cells. Methods Cell viability and catenin beta 1 (CTNNB1) expression in RAW264.7 cells following P. aeruginosa infection versus uninfected cells, were detected by cell counting kit-8 assay and β-catenin Western blots. RAW264.7 cells with CTNNB1 overexpression were established with β-catenin lentivirus using flow cytometry and clonogenic limiting dilution assays. Bacterial killing was measured by plate counts; phagocytosis and nitric oxide (NO) were measured by flow cytometry; and reactive oxygen species (ROS) were measured using Griess reaction. Autophagy was determined by microtubule-associated protein 1 light chain 3 alpha-phosphatidylethanolamine conjugate (LC3-II) protein levels and formation of LC3 puncta, using Western blot and immunofluorescence staining. Results Following P. aeruginosa infection, RAW264.7 cell β-catenin levels were reduced in a time- and multiplicity of infection-dependent manner. CTNNB1 overexpression was associated with increased P. aeruginosa elimination, but had no effect on RAW264.7 cell phagocytosis, ROS and NO. CTNNB1 overexpression reduced LC3-II levels and formation of LC3 puncta, suggesting autophagy inhibition. Rapamycin/starvation-induced autophagy resulted in reduced bacterial killing following P. aeruginosa infection. Conclusion β-catenin may promote bacterial killing via suppression of P. aeruginosa-induced macrophage autophagy.
Collapse
Affiliation(s)
- Qiang Fu
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| | - Kang Chen
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| | - Qian Zhu
- 2 Institute of Quality Standard and Testing Technology for Agro-product, Shandong Academy of Agricultural Science, Jinan, China
| | - Weijia Wang
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| | - Fuda Huang
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| | - Lishao Miao
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| | - Xinger Wu
- 1 Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-Sen University, Zhongshan, China
| |
Collapse
|
38
|
Research progress of hydroxychloroquine and autophagy inhibitors on cancer. Cancer Chemother Pharmacol 2016; 79:287-294. [PMID: 27889812 DOI: 10.1007/s00280-016-3197-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/11/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Hydroxychloroquine (HCQ), the analog of chloroquine, augments the effect of chemotherapies and radiotherapy on various tumors identified in the current clinical trials. Meanwhile, the toxicity of HCQ retinopathy raises concern worldwide. Thus, the potent autophagy inhibitors are urgently needed. METHODS A systematic review was related to 'hydroxychloroquine' or 'chloroquine' with 'clinical trials,' 'retinopathy' and 'new autophagy inhibitors.' This led to many cross-references involving HCQ, and these data have been incorporated into the following study. RESULTS Many preclinical studies indicate that the combination of HCQ with chemotherapies or radiotherapies may enhance the effect of anticancer, providing base for launching cancer clinical trials involving HCQ. The new and more sensitive diagnostic techniques report a prevalence of HCQ retinopathy up to 7.5%. Lys05, SAR405, verteporfin, VATG-027, mefloquine and spautin-1 may be potent autophagy inhibitors. CONCLUSION Additional mechanistic studies of HCQ in preclinical models are still required in order to answer these questions whether HCQ actually inhibits autophagy in non-selective tumors and whether the extent of inhibition would be sufficient to alter chemotherapy or radiotherapy sensitivity.
Collapse
|
39
|
Marudhupandiyan S, Balamurugan K. Intrinsic JNK-MAPK pathway involvement requires daf-16-mediated immune response during Shigella flexneri infection in C. elegans. Immunol Res 2016; 65:609-621. [DOI: 10.1007/s12026-016-8879-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
40
|
Fan TF, Bu LL, Wang WM, Ma SR, Liu JF, Deng WW, Mao L, Yu GT, Huang CF, Liu B, Zhang WF, Sun ZJ. Tumor growth suppression by inhibiting both autophagy and STAT3 signaling in HNSCC. Oncotarget 2016; 6:43581-93. [PMID: 26561201 PMCID: PMC4791252 DOI: 10.18632/oncotarget.6294] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
Autophagy is considered as a double-edged sword. It can prolong the survival of cancer cells and enhance its resistance to apoptosis, and paradoxically, defective autophagy has been linked to increased tumorigenesis, but the mechanism behind this phenomenon is unclear. In this study, we demonstrated that decreased phosphorylation of signal transducer and activator of transcription 3 (p-STAT3) was correlated with increased autophagy through the Akt/mTOR and Erk signaling pathways in human head and neck squamous cell carcinoma (HNSCC). We also showed that blockage of STAT3 by NSC74859 could markedly induce apoptotic cell death and autophagy. Meanwhile, increased autophagy inhibited apoptosis. The pharmacological or genetic inhibition of autophagy and STAT3 further sensitized HNSCC cells to apoptosis. Furthermore, evidence from xenograft model proved that suppressed STAT3 activity combined with inhibition of autophagy promoted tumor regression better than either treatment alone. Taken together, this present study demonstrated that autophagy alleviates apoptotic cell death in HNSCC, and combination of inhibition of STAT3 by NSC74859 and autophagy might be a promising new therapeutic strategy for HNSCC.
Collapse
Affiliation(s)
- Teng-Fei Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Wei-Ming Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Si-Rui Ma
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Jian-Feng Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Wei-Wei Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Liang Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Guang-Tao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Cong-Fa Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wen-Feng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
41
|
Martin C, Espaillat MP, Santiago-Schwarz F. IL-10 restricts dendritic cell (DC) growth at the monocyte-to-monocyte-derived DC interface by disrupting anti-apoptotic and cytoprotective autophagic molecular machinery. Immunol Res 2016; 63:131-43. [PMID: 26395023 DOI: 10.1007/s12026-015-8700-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An evolving premise is that cytoprotective autophagy responses are essential to monocyte-macrophage differentiation. Whether autophagy functions similarly during the monocyte-to-dendritic cell (DC) transition is unclear. IL-10, which induces apoptosis in maturing human DCs, has been shown to inhibit starvation-induced autophagy in murine macrophage cell lines. Based on the strict requirement that Bcl-2-mediated anti-apoptotic processes are implemented during the monocyte-to-DC transition, we hypothesized that cytoprotective autophagy responses also operate at the monocyte-DC interface and that IL-10 inhibits both anti-apoptotic and cytoprotective autophagy responses at this critical juncture. In support of our premise, we show that levels of anti-apoptotic Bcl-2 and autophagy-associated LC3 and Beclin-1 proteins are coincidentally upregulated during the monocyte-to-DC transition. Autophagy was substantiated by increased autophagosome visualization after bafilomycin treatment. Moreover, the autophagy inhibitor 3-MA restricted DC differentiation by prompting apoptosis. IL-10 implemented apoptosis that was coincidentally associated with reduced levels of Bcl-2 and widespread disruption of the autophagic flux. During peak apoptosis, IL-10 produced the death of newly committed DCs. However, cells surviving the IL-10 apoptotic schedule were highly phagocytic macrophage-like cells displaying reduced capacity to stimulate allogeneic naïve T cells in a mixed leukocyte reaction, increased levels of LC3, and mature autophagosomes. Thus, IL-10's negative control of DC-driven adaptive immunity at the monocyte-DC interface includes disruption of coordinately regulated molecular networks involved in pro-survival autophagy and anti-apoptotic responses.
Collapse
Affiliation(s)
- Carla Martin
- Biology Department, Farmingdale State College, State University of New York, Farmingdale, NY, 11735, USA
| | - Mel Pilar Espaillat
- Molecular Genetics and Microbiology and Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY, 11794, USA
| | - Frances Santiago-Schwarz
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
42
|
Role of autophagy in cellular response to infection with Orf virus Jilin isolate. Vet Microbiol 2016; 193:22-7. [PMID: 27599926 DOI: 10.1016/j.vetmic.2016.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 07/26/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022]
Abstract
Autophagy is a conserved catabolic process of the cell, which has been described to be involved in the development of various viral diseases. However, the role of autophagy in Orf virus (ORFV) replication remains unknown. In this study, we provide the first evidence that ORFV infection triggered autophagy in primary ovine fetal turbinate cells (OFTu) based on the appearance of abundant double- and single-membrane vesicles, the accumulation of LC3 fluorescent puncta, the enhancement of LC3-I/-II conversion, and autophagic flux. Moreover, modulation of ORFV-induced autophagy by rapamycin (RAPA), Earle's balanced salts solution (EBSS), chloroquine (CQ) or 3-methyladenime (3-MA) does not affect virus production. In conclusion, these results suggest that autophagy can be induced in host cells by ORFV infection, but which maybe not essential for ORFV replication.
Collapse
|
43
|
Abstract
Aging is a universal phenomenon in metazoans, characterized by a general decline of the organism physiology associated with an increased risk of mortality and morbidity. Aging of an organism correlates with a decline in function of its cells, as shown for muscle, immune, and neuronal cells. As the DNA content of most cells within an organism remains largely identical throughout the life span, age-associated transcriptional changes must be achieved by epigenetic mechanisms. However, how aging may impact on the epigenetic state of cells is only beginning to be understood. In light of a growing number of studies demonstrating that noncoding RNAs can provide molecular signals that regulate expression of protein-coding genes and define epigenetic states of cells, we hypothesize that noncoding RNAs could play a direct role in inducing age-associated profiles of gene expression. In this context, the role of long noncoding RNAs (lncRNAs) as regulators of gene expression might be important for the overall transcriptional landscape observed in aged human cells. The possible functions of lncRNAs and other noncoding RNAs, and their roles in the regulation of aging-related cellular pathways will be analyzed.
Collapse
|
44
|
Prithika U, Deepa V, Balamurugan K. External induction of heat shock stimulates the immune response and longevity of Caenorhabditis elegans towards pathogen exposure. Innate Immun 2016; 22:466-78. [PMID: 27317398 DOI: 10.1177/1753425916654557] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022] Open
Abstract
Heat shock proteins (HSPs) are highly chaperonic molecules that give immediate response during any stress, tissue damage or bacterial infections. In the present study, the role of HSPs upon bacterial encounter is studied by applying external heat induction to live Caenorhabditis elegans Heat shock was observed to increase the life span of wild type C. elegans upon pathogenic encounter, indicating a role of HSPs in bacterial infection and immunity. Similar increase in resistance towards pathogenesis observed in long-lived C. elegans daf-2 mutants and the increase in the lifespan indicated a role for the insulin/IGF-1 signaling (IIS) pathway in HSP-mediated pathogenic resistance. The microscopic observation of C. elegans after external heat induction and sequential exposure of pathogens indicated reduction of egg viability. Results of Real-time PCR and immunoblotting analysis of candidate genes revealed that heat shock and IIS pathways collaborate in the observed pathogenic resistance and further suggested SGK-1 to be the possible factor linking both these pathways. In addition, survival assays carried out using mutants equips us with supporting evidence that HSP and HSF-1 are necessary for the accelerated lifespan of C. elegans Our findings thus confirm that crosstalk between HSPs and SGK-1 influences C. elegans longevity.
Collapse
Affiliation(s)
- Udayakumar Prithika
- Department of Biotechnology, Science Campus, Alagappa University, Tamil Nadu, India
| | - Veerappan Deepa
- Department of Biotechnology, Science Campus, Alagappa University, Tamil Nadu, India
| | | |
Collapse
|
45
|
Krokowski S, Mostowy S. Interactions between Shigella flexneri and the Autophagy Machinery. Front Cell Infect Microbiol 2016; 6:17. [PMID: 26904515 PMCID: PMC4748040 DOI: 10.3389/fcimb.2016.00017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/25/2016] [Indexed: 11/13/2022] Open
Abstract
Autophagy, an intracellular degradation process, is increasingly recognized as having important roles in host defense. Interactions between Shigella flexneri and the autophagy machinery were first discovered in 2005. Since then, work has shown that multiple autophagy pathways are triggered by S. flexneri, and autophagic responses can have different roles during Shigella infection. Here, we review the interactions between S. flexneri and the autophagy machinery, highlighting that studies using Shigella can reveal the breadth of autophagic responses available to the host.
Collapse
Affiliation(s)
- Sina Krokowski
- Department of Medicine, MRC Centre of Molecular Bacteriology and Infection, Imperial College London London, UK
| | - Serge Mostowy
- Department of Medicine, MRC Centre of Molecular Bacteriology and Infection, Imperial College London London, UK
| |
Collapse
|
46
|
Bacillus thuringiensis Crystal Protein Cry6Aa Triggers Caenorhabditis elegans Necrosis Pathway Mediated by Aspartic Protease (ASP-1). PLoS Pathog 2016; 12:e1005389. [PMID: 26795495 PMCID: PMC4721865 DOI: 10.1371/journal.ppat.1005389] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/18/2015] [Indexed: 02/07/2023] Open
Abstract
Cell death plays an important role in host-pathogen interactions. Crystal proteins (toxins) are essential components of Bacillus thuringiensis (Bt) biological pesticides because of their specific toxicity against insects and nematodes. However, the mode of action by which crystal toxins to induce cell death is not completely understood. Here we show that crystal toxin triggers cell death by necrosis signaling pathway using crystal toxin Cry6Aa-Caenorhabditis elegans toxin-host interaction system, which involves an increase in concentrations of cytoplasmic calcium, lysosomal lyses, uptake of propidium iodide, and burst of death fluorescence. We find that a deficiency in the necrosis pathway confers tolerance to Cry6Aa toxin. Intriguingly, the necrosis pathway is specifically triggered by Cry6Aa, not by Cry5Ba, whose amino acid sequence is different from that of Cry6Aa. Furthermore, Cry6Aa-induced necrosis pathway requires aspartic protease (ASP-1). In addition, ASP-1 protects Cry6Aa from over-degradation in C. elegans. This is the first demonstration that deficiency in necrosis pathway confers tolerance to Bt crystal protein, and that Cry6A triggers necrosis represents a newly added necrosis paradigm in the C. elegans. Understanding this model could lead to new strategies for nematode control. Necrosis contributes to many devastating pathological conditions, such as neurodegenerative diseases and microbial pathogenesis. Bacillus thuringiensis crystal proteins are effective biopesticides. Our study reveals that B. thuringiensis Cry6Aa protein triggers the necrosis pathway using Caenorhabditis elegans as a model. We show that aspartic protease ASP-1 is required for Cry6Aa protein-induced necrosis, whereas intrinsic insults induce necrosis mediated by ASP-3 and ASP-4. Our findings contribute to the understanding of the mechanism of Bt crystal protein action and host-pathogen interactions. Because necrosis mechanisms are conserved from nematodes to humans, the fact that necrosis can be induced by Cry6Aa provides a model system for studying necrosis mechanisms in human diseases.
Collapse
|
47
|
MacInnes AW. The role of the ribosome in the regulation of longevity and lifespan extension. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 7:198-212. [PMID: 26732699 DOI: 10.1002/wrna.1325] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/14/2015] [Accepted: 11/17/2015] [Indexed: 12/11/2022]
Abstract
The most energy-consuming process that a cell must undertake to stay viable is the continuous biogenesis of ribosomes for the translation of RNA into protein. Given the inextricable links between energy consumption and cellular lifespan, it is not surprising that mutations and environmental cues that reduce ribosome biogenesis result in an extension of eukaryotic lifespan. This review goes into detail describing recent discoveries of different and often unexpected elements that play a role in the regulation of longevity by virtue of their ribosome biogenesis functions. These roles include controlling the transcription and processing of ribosomal RNA (rRNA), the translation of ribosomal protein (RP) genes, and the number of ribosomes overall. Together these findings suggest that a fundamental mechanism across eukaryotic species for extending lifespan is to slow down or halt the expenditure of cellular energy that is normally absorbed by the manufacturing and assembly of new ribosomes.
Collapse
|
48
|
Agarwal S, Kim H, Chan RB, Agarwal S, Williamson R, Cho W, Paolo GD, Paolo GD, Satchell KJF. Autophagy and endosomal trafficking inhibition by Vibrio cholerae MARTX toxin phosphatidylinositol-3-phosphate-specific phospholipase A1 activity. Nat Commun 2015; 6:8745. [PMID: 26498860 PMCID: PMC4640098 DOI: 10.1038/ncomms9745] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/25/2015] [Indexed: 12/16/2022] Open
Abstract
Vibrio cholerae, responsible for acute gastroenteritis secretes a large multifunctional-autoprocessing repeat-in-toxin (MARTX) toxin linked to evasion of host immune system, facilitating colonization of small intestine. Unlike other effector domains of the multifunctional toxin that target cytoskeleton, the function of alpha-beta hydrolase (ABH) remained elusive. This study demonstrates that ABH is an esterase/lipase with catalytic Ser–His–Asp triad. ABH binds with high affinity to phosphatidylinositol-3-phosphate (PtdIns3P) and cleaves the fatty acid in PtdIns3P at the sn1 position in vitro making it the first PtdIns3P-specific phospholipase A1 (PLA1). Expression of ABH in vivo reduces intracellular PtdIns3P levels and its PtdIns3P-specific PLA1 activity blocks endosomal and autophagic pathways. In accordance with recent studies acknowledging the potential of extracellular pathogens to evade or exploit autophagy to prevent their clearance and facilitate survival, this is the first report highlighting the role of ABH in inhibiting autophagy and endosomal trafficking induced by extracellular V. cholerae. The MARTX toxin of V. cholerae processes itself to deliver three distinct effector domains to the cytosol. Here the authors show that the third effector domain is a phosphatidylinositol-3-phosphate (PtdIns3P)-specific phospholipase that inhibits autophagy in target cells.
Collapse
Affiliation(s)
- Shivani Agarwal
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Hyunjin Kim
- Department of Chemistry, University of Illinois at Chicago, 845 West Taylor Street, Chicago, Illinois 60607, USA
| | - Robin B Chan
- Department of Pathology and Cell Biology, 630 West 168th Street, Columbia University, New York, New York 10032, USA
| | - Shivangi Agarwal
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| | - Rebecca Williamson
- Department of Pathology and Cell Biology, 630 West 168th Street, Columbia University, New York, New York 10032, USA
| | - Wonhwa Cho
- Department of Chemistry, University of Illinois at Chicago, 845 West Taylor Street, Chicago, Illinois 60607, USA
| | - Gilbert Di Paolo
- Department of Pathology and Cell Biology, 630 West 168th Street, Columbia University, New York, New York 10032, USA
| | - Gilbert D Paolo
- Department of Pathology and Cell Biology, 630 West 168th Street, Columbia University, New York, New York 10032, USA
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, Illinois 60611, USA
| |
Collapse
|
49
|
Zhou Y, Wang Q, Song B, Wu S, Su Y, Zhang H, He Y. A real-time documentation and mechanistic investigation of quantum dots-induced autophagy in live Caenorhabditis elegans. Biomaterials 2015; 72:38-48. [PMID: 26342559 DOI: 10.1016/j.biomaterials.2015.08.044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/25/2015] [Indexed: 12/31/2022]
Abstract
Autophagy is a highly important intracellular process for the degradation of endogenous or foreign contents in the cytoplasm. Though nanomaterials-induced autophagy has been extensively studied, real-time information about the autophagic process induced by nanomaterials in live organisms remains unknown. Here by using Caenorhabditis elegans as the model organism and fluorescent semiconductor quantum dots (QDs) as a representative nanomaterial, we systematically investigated the phenomenon of QDs-induced autophagy in live organisms. Our results demonstrated that the internalized QDs trigger a complete autophagic process in C. elegans intestinal cells. Further investigations revealed that this QD-induced autophagy in C. elegans is neither a response to released heavy metal ions by the QDs, nor an attempt to engulf exogenous QD materials, but a defensive strategy of the organism to clear and recycle damaged endosomes. Of particular significance, for the first time, we presented real-time tracking of autophagosomes formation in live organisms, providing detailed temporal-spatial information of this process. This study may help us better understand the relationship between nanomaterials and autophagy in vivo, and provide invaluable information for safety evaluation and bio-application of nanomaterials.
Collapse
Affiliation(s)
- Yanfeng Zhou
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China; Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China
| | - Qin Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China; Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China
| | - Bin Song
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Sicong Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Yuanyuan Su
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China
| | - Huimin Zhang
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China.
| | - Yao He
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, China.
| |
Collapse
|
50
|
Emerging strategies to effectively target autophagy in cancer. Oncogene 2015; 35:1-11. [PMID: 25893285 DOI: 10.1038/onc.2015.99] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 02/07/2023]
Abstract
Autophagy serves a dichotomous role in cancer and recent advances have helped delineate the appropriate settings where inhibiting or promoting autophagy may confer therapeutic efficacy in patients. Our evolving understanding of the molecular machinery responsible for the tightly controlled regulation of this homeostatic mechanism has begun to bear fruit in the way of autophagy-oriented clinical trials and promising lead compounds to modulate autophagy for therapeutic benefit. In this manuscript we review the recent preclinical and clinical therapeutic strategies that involve autophagy modulation in cancer.
Collapse
|