1
|
Mebrahtom G, Hailay A, Aberhe W, Zereabruk K, Haile TG, Tadesse DB. Admission and outcomes of COVID-19 among chronic obstructive pulmonary diseases patients in Africa: protocol for a systematic review and meta-analysis. Int Health 2025; 17:245-250. [PMID: 39360405 PMCID: PMC12045080 DOI: 10.1093/inthealth/ihae062] [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: 05/20/2023] [Revised: 05/03/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
When the coronavirus case was originally reported in Wuhan, China, in December 2019, it quickly spread throughout the world and became a global public health problem. Evidence of the admission and outcomes of coronavirus disease among patients with chronic obstructive pulmonary disease (COPD) has not been reported in Africa. Consequently, this research protocol uses a systematic review and meta-analysis of the admission and outcomes of COVID-19 in patients with COPD in Africa. All observational studies published in the English language and reporting on the prevalence, admission and outcomes of COVID-19 among patients with COPD in Africa will be included. A search strategy will be implemented using electronic databases and the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocol recommendations. The findings of this review will be reported to health program designers, decision-makers and healthcare providers.
Collapse
Affiliation(s)
- Guesh Mebrahtom
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Abrha Hailay
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Woldu Aberhe
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Kidane Zereabruk
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Teklehaimanot Gereziher Haile
- Department of Maternity and Neonatal Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| | - Degena Bahrey Tadesse
- Department of Adult Health Nursing, School of Nursing, College of Health Science, Aksum University, Aksum, Ethiopia
| |
Collapse
|
2
|
Robinson KS, Sennhenn P, Yuan DS, Liu H, Taddei D, Qian Y, Luo W. TMBIM6/BI-1 is an intracellular environmental regulator that induces paraptosis in cancer via ROS and Calcium-activated ERAD II pathways. Oncogene 2025; 44:494-512. [PMID: 39609612 PMCID: PMC11832424 DOI: 10.1038/s41388-024-03222-x] [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: 06/15/2023] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024]
Abstract
Transmembrane B cell lymphoma 2-associated X protein inhibitor motif-containing (TMBIM) 6, also known as Bax Inhibitor-1 (BI-1), has been heavily researched for its cytoprotective functions. TMBIM6 functional diversity includes modulating cell survival, stress, metabolism, cytoskeletal dynamics, organelle function, regulating cytosolic acidification, calcium, and reactive oxygen species (ROS). Clinical research shows TMBIM6 plays a key role in many of the world's top diseases/injuries (i.e., Alzheimer's, Parkinson's, diabetes, obesity, brain injury, liver disease, heart disease, aging, etc.), including cancer, where TMBIM6 expression impacts patient survival, chemoresistance, cancer progression, and metastasis. We show TMBIM6 is activated by, and undergoes, different conformational changes that dictate its function following a significant change in the cell's IntraCellular Environment (ICE). TMBIM6 agonism, following ICE change, can help the cell overcome multiple stresses including toxin exposure, viral infection, wound healing, and excitotoxicity. However, in cancer cells TMBIM6 agonism results in rapid paraptotic induction irrespective of the cancer type, sub-type, genotype or phenotype. Furthermore, the level of TMBIM6 expression in cancer did not dictate the level of paraptotic induction; however, it did dictate the rate at which paraptosis occurred. TMBIM6 agonism did not induce paraptosis in cancer via canonical routes involving p38 MAPK, JNK, ERK, UPR, autophagy, proteasomes, or Caspase-9. Instead, TMBIM6 agonism in cancer upregulates cytosolic Ca2+ and ROS, activates lysosome biogenesis, and induces paraptosis via ERAD II mechanisms. In xenograft models, we show TMBIM6 agonism induces rapid cancer cell death with no toxicity, even at high doses of TMBIM6 agonist (>450 mg/kg). In summary, this study shows TMBIM6's functional diversity is only activated by severe ICE change in diseased/injured cells, highlighting its transformative potential as a therapeutic target across various diseases and injuries, including cancer.
Collapse
Affiliation(s)
| | | | | | - Hai Liu
- Viva Biotech, Shanghai, China
| | | | | | - Wei Luo
- MicroQuin, Cambridge, MA, USA
| |
Collapse
|
3
|
Qian S, Long Y, Tan G, Li X, Xiang B, Tao Y, Xie Z, Zhang X. Programmed cell death: molecular mechanisms, biological functions, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e70024. [PMID: 39619229 PMCID: PMC11604731 DOI: 10.1002/mco2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Programmed cell death represents a precisely regulated and active cellular demise, governed by a complex network of specific genes and proteins. The identification of multiple forms of programmed cell death has significantly advanced the understanding of its intricate mechanisms, as demonstrated in recent studies. A thorough grasp of these processes is essential across various biological disciplines and in the study of diseases. Nonetheless, despite notable progress, the exploration of the relationship between programmed cell death and disease, as well as its clinical application, are still in a nascent stage. Therefore, further exploration of programmed cell death and the development of corresponding therapeutic methods and strategies holds substantial potential. Our review provides a detailed examination of the primary mechanisms behind apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. Following this, the discussion delves into biological functions and diseases associated dysregulated programmed cell death. Finally, we highlight existing and potential therapeutic targets and strategies focused on cancers and neurodegenerative diseases. This review aims to summarize the latest insights on programmed cell death from mechanisms to diseases and provides a more reliable approach for clinical transformation.
Collapse
Affiliation(s)
- Shen'er Qian
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Yao Long
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of PathologyXiangya Hospital, Central South UniversityChangshaHunanChina
| | - Guolin Tan
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| | - Xiaoguang Li
- Department of Otolaryngology Head and Neck SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
- Ear InstituteShanghai Jiao Tong University School of Medicine, Shanghai Key LabShanghaiChina
| | - Bo Xiang
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
- Furong LaboratoryCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Cancer Research InstituteSchool of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Zuozhong Xie
- Department of Otolaryngology Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Xiaowei Zhang
- Department of Otolaryngology Head and Neck SurgeryThe Third Xiangya Hospital, Central South UniversityChangshaHunanChina
| |
Collapse
|
4
|
Wallbank BA, Pardy RD, Brodsky IE, Hunter CA, Striepen B. Cryptosporidium impacts epithelial turnover and is resistant to induced death of the host cell. mBio 2024; 15:e0172024. [PMID: 38995074 PMCID: PMC11323733 DOI: 10.1128/mbio.01720-24] [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: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Infection with the apicomplexan parasite Cryptosporidium is a leading cause of diarrheal disease. Cryptosporidiosis is of particular importance in infants and shows a strong association with malnutrition, both as a risk factor and as a consequence. Cryptosporidium invades and replicates within the small intestine epithelial cells. This is a highly dynamic tissue that is developmentally stratified along the villus axis. New cells emerge from a stem cell niche in the crypt and differentiate into mature epithelial cells while moving toward the villus tip, where they are ultimately shed. Here, we studied the impact of Cryptosporidium infection on this dynamic architecture. Tracing DNA synthesis in pulse-chase experiments in vivo, we quantified the genesis and migration of epithelial cells along the villus. We found proliferation and epithelial migration to be elevated in response to Cryptosporidium infection. Infection also resulted in significant cell loss documented by imaging and molecular assays. Consistent with these observations, single-cell RNA sequencing of infected intestines showed a gain of young and a loss of mature cells. Interestingly, enhanced epithelial cell loss was not a function of enhanced apoptosis of infected cells. To the contrary, Cryptosporidium-infected cells were less likely to be apoptotic than bystanders, and experiments in tissue culture demonstrated that infection provided enhanced resistance to chemically induced apoptosis to the host but not bystander cells. Overall, this study suggests that Cryptosporidium may modulate cell apoptosis and documents pronounced changes in tissue homeostasis due to parasite infection, which may contribute to its long-term impact on the developmental and nutritional state of children. IMPORTANCE The intestine must balance its roles in digestion and nutrient absorption with the maintenance of an effective barrier to colonization and breach by numerous potential pathogens. An important component of this balance is its constant turnover, which is modulated by a gain of cells due to proliferation and loss due to death or extrusion. Here, we report that Cryptosporidium infection changes the dynamics of this process increasing both gain and loss of enterocytes speeding up the villus elevator. This leads to a much more immature epithelium and a reduction of the number of those cells typically found toward the villus apex best equipped to take up key nutrients including carbohydrates and lipids. These changes in the cellular architecture and physiology of the small intestine may be linked to the profound association between cryptosporidiosis and malnutrition.
Collapse
Affiliation(s)
- Bethan A. Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ryan D. Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Igor E. Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher A. Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Chen Y, Tian P, Li Y, Tang Z, Zhang H. Thiram exposure: Disruption of the blood-testis barrier and altered apoptosis-autophagy dynamics in testicular cells via the Bcl-2/Bax and mTOR/Atg5/p62 pathways in mice. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 203:106010. [PMID: 39084803 DOI: 10.1016/j.pestbp.2024.106010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Thiram, a prevalent dithiocarbamate insecticide in agriculture, is widely employed as a crop insecticide and preservative. Chronic exposure to thiram has been linked to various irreversible damages, including tibial cartilage dysplasia, erythrocytotoxicity, renal issues, and immune system compromise. Limited research exists on its effects on reproductive organs. This study investigated the reproductive toxicology in mouse testes exposure to varying concentrations (0, 30, 60, and 120 mg/kg) of thiram. Our study uncovered a series of adverse effects in mice subjected to thiram exposure, including emaciation, stunted growth, decreased water intake, and postponed testicular maturation. Biochemical analysis in thiram-exposed mice showed elevated levels of LDH and AST, while ALP, TG, ALT, and urea were decreased. Histologically, thiram disrupted the testis' microarchitecture and compromised its barrier function by widening the gap between spermatogenic cells and promoting fibrosis. The expression of pro-apoptotic genes (Bax, APAF1, Cytc, and Caspase-3) was downregulated, whereas Bcl-2 expression increased in thiram-treated mice compared to controls. Conversely, the expression of Atg5 was upregulated, and mTOR and p62 expression decreased, with a trend towards lower LC3b levels. Thiram also disrupted the blood-testis barrier, significantly reducing the mRNA expression of zona occludens-1 (ZO-1) and occludin. In conclusion, chronic exposure to high thiram concentrations (120 mg/kg) caused testicular tissue damage, affecting the blood-testis barrier and modulating apoptosis and autophagy through the Bcl-2/Bax and mTOR/Atg5/p62 pathways. This study contributes to understanding the molecular basis of thiram-induced reproductive toxicity and underscores the need for further research and precautions for those chronically exposed to thiram and its environmental residuals.
Collapse
Affiliation(s)
- Yongjian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Peipei Tian
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
6
|
Yan P, Liu J, Huang Y, Li Y, Yu J, Xia J, Liu M, Bai R, Wang N, Guo L, Liu G, Yang X, Zeng J, He B. Lotus leaf extract can attenuate salpingitis in laying hens by inhibiting apoptosis. Poult Sci 2023; 102:102865. [PMID: 37499615 PMCID: PMC10413199 DOI: 10.1016/j.psj.2023.102865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 07/29/2023] Open
Abstract
This study aimed to determine whether the lotus leaf extract (LLE) had the effect of treating salpingitis in laying hens. First, the salpingitis model was established by the method of bacterial infection. Differential genes between salpingitis and healthy laying hens were identified by transcriptome sequencing, and GO and KEGG enrichment analyses were performed. Groups of treatment of antibiotics and LLE were established to verify the feasibility of the lotus leaf extract in treating salpingitis. Furthermore, the active component and pharmacological effects of LLE were identified using the UPLC-Q-TOF-MS and network pharmacology technique. At last, the mechanism of LLE treating salpingitis was further evaluated by DF-1 cells infected with bacteria. The results showed that LLE significantly reduced the levels of TLR4 and IFN-γ (P < 0.05), accelerated the levels of IgA and IgG (P < 0.05), regulated the levels of SOD and MDA (P < 0.05) in laying hens with salpingitis. A total of 1,874 differential genes were obtained according to the transcriptome sequencing. It was revealed a significant role in cell cycle and apoptosis by enrichment analysis. In addition, among the 28 components identified by UPLC-Q-TOF-MS, 20 components acted on 58 genes, including CDK1, BIRC5, and CA2 for treating salpingitis. After bacterial infection, cells were damaged and unable to complete the normal progression of the cell cycle, leading to cell cycle arrest and further apoptosis formation. However, with the intervention of LLE, bacterial infection was resisted. The cells proliferation was extensively restored, and the expression of NO was increased. The addition of LLE significantly decreased cell apoptosis. The G1 phase increased, the S phase and the G2 phase decreased in the model group; after the intervention of LLE, the G1 phase gradually returned to the average level, and G2 and S phases increased. The mRNA expression levels of BIRC5, CDK1, and CA2 were consistent with the predicted results in network pharmacology. At the same time, the mRNA expression levels of Caspase-3 and Caspase-7 were reduced after added with LLE. The mRNA expression levels of TNF-α, TRADD, FADD, Caspase-8, Caspase-10, and Caspase-9 (P < 0.05), which would inhibit death receptor activation and decrease the apoptotic cascade, were upregulated after bacterial infection. However, the results in LLE groups were downregulated (P < 0.05). Meanwhile, the mRNA expression levels of BCL-2 in LLE groups were increased significantly compared with it in model group (P < 0.05). Notably, LLE administration inhibited apoptosis and regulated the cell cycle distribution in the salpingitis induced by bacterial infection. These results indicated that the LLE attenuated bacterial-induced salpingitis by modulating apoptosis and immune function in laying hens.
Collapse
Affiliation(s)
- Pupu Yan
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Jiali Liu
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Yongxi Huang
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Yana Li
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Jie Yu
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Jinjin Xia
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Man Liu
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Ruonan Bai
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Ning Wang
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Liwei Guo
- School of Animal Science, Yangtze University, Jingzhou 434020, China.
| | - Guoping Liu
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Xiaolin Yang
- School of Animal Science, Yangtze University, Jingzhou 434020, China
| | - Jianguo Zeng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Bin He
- Animal and Veterinary Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| |
Collapse
|
7
|
Vinkler M, Fiddaman SR, Těšický M, O'Connor EA, Savage AE, Lenz TL, Smith AL, Kaufman J, Bolnick DI, Davies CS, Dedić N, Flies AS, Samblás MMG, Henschen AE, Novák K, Palomar G, Raven N, Samaké K, Slade J, Veetil NK, Voukali E, Höglund J, Richardson DS, Westerdahl H. Understanding the evolution of immune genes in jawed vertebrates. J Evol Biol 2023; 36:847-873. [PMID: 37255207 PMCID: PMC10247546 DOI: 10.1111/jeb.14181] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023]
Abstract
Driven by co-evolution with pathogens, host immunity continuously adapts to optimize defence against pathogens within a given environment. Recent advances in genetics, genomics and transcriptomics have enabled a more detailed investigation into how immunogenetic variation shapes the diversity of immune responses seen across domestic and wild animal species. However, a deeper understanding of the diverse molecular mechanisms that shape immunity within and among species is still needed to gain insight into-and generate evolutionary hypotheses on-the ultimate drivers of immunological differences. Here, we discuss current advances in our understanding of molecular evolution underpinning jawed vertebrate immunity. First, we introduce the immunome concept, a framework for characterizing genes involved in immune defence from a comparative perspective, then we outline how immune genes of interest can be identified. Second, we focus on how different selection modes are observed acting across groups of immune genes and propose hypotheses to explain these differences. We then provide an overview of the approaches used so far to study the evolutionary heterogeneity of immune genes on macro and microevolutionary scales. Finally, we discuss some of the current evidence as to how specific pathogens affect the evolution of different groups of immune genes. This review results from the collective discussion on the current key challenges in evolutionary immunology conducted at the ESEB 2021 Online Satellite Symposium: Molecular evolution of the vertebrate immune system, from the lab to natural populations.
Collapse
Affiliation(s)
- Michal Vinkler
- Department of ZoologyFaculty of ScienceCharles UniversityPragueCzech Republic
| | | | - Martin Těšický
- Department of ZoologyFaculty of ScienceCharles UniversityPragueCzech Republic
| | | | - Anna E. Savage
- Department of BiologyUniversity of Central FloridaFloridaOrlandoUSA
| | - Tobias L. Lenz
- Research Unit for Evolutionary ImmunogenomicsDepartment of BiologyUniversity of HamburgHamburgGermany
| | | | - Jim Kaufman
- Institute for Immunology and Infection ResearchUniversity of EdinburghEdinburghUK
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - Daniel I. Bolnick
- Department of Ecology and Evolutionary BiologyUniversity of ConnecticutStorrsConnecticutUSA
| | | | - Neira Dedić
- Department of Botany and ZoologyMasaryk UniversityBrnoCzech Republic
| | - Andrew S. Flies
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - M. Mercedes Gómez Samblás
- Department of ZoologyFaculty of ScienceCharles UniversityPragueCzech Republic
- Department of ParasitologyUniversity of GranadaGranadaSpain
| | | | - Karel Novák
- Department of Genetics and BreedingInstitute of Animal SciencePragueUhříněvesCzech Republic
| | - Gemma Palomar
- Faculty of BiologyInstitute of Environmental SciencesJagiellonian UniversityKrakówPoland
| | - Nynke Raven
- Department of ScienceEngineering and Build EnvironmentDeakin UniversityVictoriaWaurn PondsAustralia
| | - Kalifa Samaké
- Department of Genetics and MicrobiologyFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Joel Slade
- Department of BiologyCalifornia State UniversityFresnoCaliforniaUSA
| | | | - Eleni Voukali
- Department of ZoologyFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Jacob Höglund
- Department of Ecology and GeneticsUppsala UniversitetUppsalaSweden
| | | | | |
Collapse
|
8
|
Yuk JM, Park EJ, Kim IS, Jo EK. Itaconate family-based host-directed therapeutics for infections. Front Immunol 2023; 14:1203756. [PMID: 37261340 PMCID: PMC10228716 DOI: 10.3389/fimmu.2023.1203756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Itaconate is a crucial anti-infective and anti-inflammatory immunometabolite that accumulates upon disruption of the Krebs cycle in effector macrophages undergoing inflammatory stress. Esterified derivatives of itaconate (4-octyl itaconate and dimethyl itaconate) and its isomers (mesaconate and citraconate) are promising candidate drugs for inflammation and infection. Several itaconate family members participate in host defense, immune and metabolic modulation, and amelioration of infection, although opposite effects have also been reported. However, the precise mechanisms by which itaconate and its family members exert its effects are not fully understood. In addition, contradictory results in different experimental settings and a lack of clinical data make it difficult to draw definitive conclusions about the therapeutic potential of itaconate. Here we review how the immune response gene 1-itaconate pathway is activated during infection and its role in host defense and pathogenesis in a context-dependent manner. Certain pathogens can use itaconate to establish infections. Finally, we briefly discuss the major mechanisms by which itaconate family members exert antimicrobial effects. To thoroughly comprehend how itaconate exerts its anti-inflammatory and antimicrobial effects, additional research on the actual mechanism of action is necessary. This review examines the current state of itaconate research in infection and identifies the key challenges and opportunities for future research in this field.
Collapse
Affiliation(s)
- Jae-Min Yuk
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Infection Biology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - In Soo Kim
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Infection Control Convergence Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Maekawa T, Kashkar H, Coll NS. Dying in self-defence: a comparative overview of immunogenic cell death signalling in animals and plants. Cell Death Differ 2023; 30:258-268. [PMID: 36195671 PMCID: PMC9950082 DOI: 10.1038/s41418-022-01060-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022] Open
Abstract
Host organisms utilise a range of genetically encoded cell death programmes in response to pathogen challenge. Host cell death can restrict pathogen proliferation by depleting their replicative niche and at the same time dying cells can alert neighbouring cells to prepare environmental conditions favouring future pathogen attacks. As expected, many pathogenic microbes have strategies to subvert host cell death to promote their virulence. The structural and lifestyle differences between animals and plants have been anticipated to shape very different host defence mechanisms. However, an emerging body of evidence indicates that several components of the host-pathogen interaction machinery are shared between the two major branches of eukaryotic life. Many proteins involved in cell death execution or cell death-associated immunity in plants and animals exert direct effects on endomembrane and loss of membrane integrity has been proposed to explain the potential immunogenicity of dying cells. In this review we aim to provide a comparative view on how cell death processes are linked to anti-microbial defence mechanisms in plants and animals and how pathogens interfere with these cell death programmes. In comparison to the several well-defined cell death programmes in animals, immunogenic cell death in plant defence is broadly defined as the hypersensitive response. Our comparative overview may help discerning whether specific types of immunogenic cell death exist in plants, and correspondingly, it may provide new hints for previously undiscovered cell death mechanism in animals.
Collapse
Affiliation(s)
- Takaki Maekawa
- Department of Biology, Institute for Plant Sciences, University of Cologne, 50674, Cologne, Germany.
- CEPLAS Cluster of Excellence on Plant Sciences at the University of Cologne, Cologne, Germany.
| | - Hamid Kashkar
- Faculty of Medicine and University Hospital of Cologne, Institute for Molecular Immunology, University of Cologne, 50931, Cologne, Germany.
- Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.
| | - Núria S Coll
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, 08193, Bellaterra, Spain.
- Consejo Superior de Investigaciones Científicas (CSIC), 08001, Barcelona, Spain.
| |
Collapse
|
10
|
Souza IDP, Amoras EDSG, de Sousa FDM, de Sousa PVNR, Lima SS, Cayres-Vallinoto IMV, Ishak R, Vallinoto ACR, Queiroz MAF. Plasma Levels of sFas-sFasL and FASL Gene Expression Are Associated with Tuberculosis. Biomolecules 2022; 13:biom13010080. [PMID: 36671466 PMCID: PMC9855614 DOI: 10.3390/biom13010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/03/2023] Open
Abstract
Apoptosis of macrophages infected by Mycobacterium tuberculosis via Fas-FasL is an important immune mechanism against infection. This study investigated the association of tuberculosis (TB) with the presence of the polymorphisms FAS -670A/G and FASL -124A/G, the levels of sFas and sFasL, and the gene expression of FASL and cytokines. Samples of 200 individuals diagnosed with TB and 200 healthy controls were evaluated. Real-time PCR (genotyping and gene expression) and ELISA (dosages of sFas, sFasL, IFN-γ, and IL-10) tests were performed. There was no association of FAS -670A/G and FASL -124A/G polymorphisms with TB. The TB group exhibited high plasma levels of sFas and reduced plasma levels of sFasL (p < 0.05). The correlation analysis between these markers revealed a positive correlation between the levels of sFas and sFasL, sFasL and FASL expression, and between sFas and FASL expression (p < 0.05). In the TB group, there was a positive correlation between FASL expression and IFN-γ levels and higher levels of IL-10 compared to IFN-γ (p < 0.05). High levels of sFas and reduced levels of sFasL and FASL expression may contribute to the inhibition of apoptosis in infected cells and represent a possible bacterial resistance resource to maintain the infection.
Collapse
Affiliation(s)
- Iury de Paula Souza
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
| | - Francisca Dayse Martins de Sousa
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
| | | | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
| | | | - Ricardo Ishak
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
| | | | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66075-110, Brazil
- Correspondence:
| |
Collapse
|
11
|
Abstract
Blocking host cell death is an important virulence strategy employed by many bacterial pathogens. We recently reported that Shigella flexneri inhibits host pyroptosis by delivering a type III secretion system (T3SS) effector OspC3 that catalyzes a novel arginine ADP-riboxanation modification on caspase-4/11. Here, we investigated the OspC3 homologue CopC from Chromobacterium violaceum, an opportunistic but sometimes deadly bacterial pathogen. CopC bears the same arginine ADP-riboxanase activity as OspC3, but with a different substrate specificity. Through proteomic analysis, we first identified host calmodulin (CaM) as a binding partner of CopC. The analyses additionally revealed that CopC preferably modifies apoptotic caspases including caspase-7, -8 and -9. This results in suppression of both extrinsic and intrinsic apoptosis programs in C. violaceum-infected cells. Biochemical reconstitution showed that CopC requires binding to CaM, specifically in the calcium-free state, to achieve efficient ADP-riboxanation of the caspases. We determined crystal structure of the CaM-CopC-CASP7 ternary complex, which illustrates the caspase recognition mechanism and a unique CaM-binding mode in CopC. Structure-directed mutagenesis validated the functional significance of CaM binding for stimulating CopC modification of its caspase substrates. CopC adopts an ADP-ribosyltransferase-like fold with a unique His-Phe-Glu catalytic triad, featuring two acidic residues critical for site-specific arginine ADP-riboxanation. Our study expands and deepens our understanding of the OspC family of ADP-riboxanase effectors.
Collapse
|
12
|
Riera Romo M. Cell death as part of innate immunity: Cause or consequence? Immunology 2021; 163:399-415. [PMID: 33682112 DOI: 10.1111/imm.13325] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Regulated or programmed cell death plays a critical role in the development and tissue organization and function. In addition, it is intrinsically connected with immunity and host defence. An increasing cellular and molecular findings cause a change in the concept of cell death, revealing an expanding network of regulated cell death modalities and their biochemical programmes. Likewise, recent evidences demonstrate the interconnection between cell death pathways and how they are involved in different immune mechanisms. This work provides an overview of the main cell death programmes and their implication in innate immunity not only as an immunogenic/inflammatory process, but also as an active defence strategy during immune response and at the same time as a regulatory mechanism.
Collapse
Affiliation(s)
- Mario Riera Romo
- Radiology Department, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Friedrich A, Beare PA, Schulze-Luehrmann J, Cordsmeier A, Pazen T, Sonnewald S, Lührmann A. The Coxiella burnetii effector protein CaeB modulates endoplasmatic reticulum (ER) stress signalling and is required for efficient replication in Galleria mellonella. Cell Microbiol 2021; 23:e13305. [PMID: 33355405 DOI: 10.1111/cmi.13305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 01/02/2023]
Abstract
The obligate intracellular pathogen Coxiella burnetii is the causative agent of the zoonosis Q fever. C. burnetii infection can have severe outcomes due to the development of chronic infection. To establish and maintain an infection, C. burnetii depends on a functional type IVB secretion system (T4BSS) and, thus, on the translocation of effector proteins into the host cell. Here, we showed that the C. burnetii T4BSS effector protein CaeB targets the conserved endoplasmatic reticulum (ER) stress sensor IRE1 during ER stress in mammalian and plant cells. CaeB-induced upregulation of IRE1 RNase activity was essential for CaeB-mediated inhibition of ER stress-induced cell death. Our data reveal a novel role for CaeB in ER stress signalling modulation and demonstrate that CaeB is involved in pathogenicity in vivo. Furthermore, we provide evidence that C. burnetii infection leads to modulation of the ER stress sensors IRE1 and PERK, but not ATF6 during ER stress. While the upregulation of the RNase activity of IRE1 during ER stress depends on CaeB, modulation of PERK is CaeB independent, suggesting that C. burnetii encodes several factors influencing ER stress during infection.
Collapse
Affiliation(s)
- Anja Friedrich
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.,Lehrstuhl für Biochemie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Paul A Beare
- Coxiella Pathogenesis Section, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Jan Schulze-Luehrmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arne Cordsmeier
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Pazen
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sophia Sonnewald
- Lehrstuhl für Biochemie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Lührmann
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
14
|
Camilli G, Blagojevic M, Naglik JR, Richardson JP. Programmed Cell Death: Central Player in Fungal Infections. Trends Cell Biol 2020; 31:179-196. [PMID: 33293167 DOI: 10.1016/j.tcb.2020.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/08/2020] [Accepted: 11/11/2020] [Indexed: 12/26/2022]
Abstract
Fungal diseases contribute significantly to morbidity and mortality in humans. Although recent research has improved our understanding of the complex and dynamic interplay that occurs between pathogenic fungi and the human host, much remains to be elucidated concerning the molecular mechanisms that drive fungal pathogenicity and host responses to fungal infections. In recent times, there has been a significant increase in studies investigating the immunological functions of microbial-induced host cell death. In addition, pathogens use many strategies to manipulate host cell death pathways to facilitate their survival and dissemination. This review will focus on the mechanisms of host programmed cell death that occur during opportunistic fungal infections, and explore how cell death pathways may affect immunity towards pathogenic fungi.
Collapse
Affiliation(s)
- Giorgio Camilli
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK.
| | - Mariana Blagojevic
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| |
Collapse
|
15
|
Ashida H, Sasakawa C, Suzuki T. A unique bacterial tactic to circumvent the cell death crosstalk induced by blockade of caspase-8. EMBO J 2020; 39:e104469. [PMID: 32657447 PMCID: PMC7459423 DOI: 10.15252/embj.2020104469] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022] Open
Abstract
Upon invasive bacterial infection of colonic epithelium, host cells induce several types of cell death to eliminate pathogens. For instance, necroptosis is a RIPK‐dependent lytic cell death that serves as a backup system to fully eliminate intracellular pathogens when apoptosis is inhibited; this phenomenon has been termed “cell death crosstalk”. To maintain their replicative niche and multiply within cells, some enteric pathogens prevent epithelial cell death by delivering effectors via the type III secretion system. In this study, we found that Shigella hijacks host cell death crosstalk via a dual mechanism: inhibition of apoptosis by the OspC1 effector and inhibition of necroptosis by the OspD3 effector. Upon infection by Shigella, host cells recognize blockade of caspase‐8 apoptosis signaling by OspC1 effector as a key danger signal and trigger necroptosis as a backup form of host defense. To counteract this backup defense, Shigella delivers the OspD3 effector, a protease, to degrade RIPK1 and RIPK3, preventing necroptosis. We believe that blockade of host cell death crosstalk by Shigella is a unique intracellular survival tactic for prolonging the bacterium's replicative niche.
Collapse
Affiliation(s)
- Hiroshi Ashida
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Medical Mycology Research Center, Chiba University, Chiba, Japan
| | - Chihiro Sasakawa
- Medical Mycology Research Center, Chiba University, Chiba, Japan.,Nippon Institute for Biological Science, Tokyo, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Infection and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
16
|
León DL, Matthey P, Fellay I, Blanchard M, Martinvalet D, Mantel PY, Filgueira L, Walch M. Granzyme B Attenuates Bacterial Virulence by Targeting Secreted Factors. iScience 2020; 23:100932. [PMID: 32151975 PMCID: PMC7063247 DOI: 10.1016/j.isci.2020.100932] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/23/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Pathogenic bacteria secrete virulence factors that interact with the human host to establish infections. The human immune system evolved multiple mechanisms to fight bacterial invaders, including immune proteases that were demonstrated to contribute crucially to antibacterial defense. Here we show that granzyme B degrades multiple secreted virulence mediators from Listeria monocytogenes, Salmonella typhimurium, and Mycobacteria tuberculosis. Pathogenic bacteria, when infected in the presence of granzyme B or granzyme-secreting killer cells, fail to grow in human macrophages and epithelial cells owing to their crippled virulence. A granzyme B-uncleavable mutant form of the major Listeria virulence factor, listeriolysin O, rescued the virulence defect in response to granzyme treatment. Hence, we link the degradation of a single factor with the observed decrease in virulent bacteria growth. Overall, we reveal here an innate immune barrier function of granzyme B by disrupting bacterial virulence to facilitate bacteria clearance by bystander immune and non-immune cells.
Collapse
Affiliation(s)
- Diego López León
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Patricia Matthey
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Isabelle Fellay
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Marianne Blanchard
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Denis Martinvalet
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35121 Padova, Italy
| | - Pierre-Yves Mantel
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Luis Filgueira
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland
| | - Michael Walch
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy Unit, University of Fribourg, PER03.14, Route Albert Gockel 1, 1700 Fribourg, Switzerland.
| |
Collapse
|
17
|
Tsuchiya K. Inflammasome‐associated cell death: Pyroptosis, apoptosis, and physiological implications. Microbiol Immunol 2020; 64:252-269. [DOI: 10.1111/1348-0421.12771] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research InstituteKanazawa UniversityKanazawa Japan
- Institute for Frontier Science Initiative (InFiniti)Kanazawa UniversityKanazawa Japan
| |
Collapse
|
18
|
Mahib MR, Hosojima S, Kushiyama H, Kinoshita T, Shiroishi T, Suda T, Tsuchiya K. Caspase-7 mediates caspase-1-induced apoptosis independently of Bid. Microbiol Immunol 2019; 64:143-152. [PMID: 31687791 DOI: 10.1111/1348-0421.12756] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/16/2019] [Accepted: 11/01/2019] [Indexed: 01/26/2023]
Abstract
Inflammasomes are innate immune mechanisms that activate caspase-1 in response to a variety of stimuli, including Salmonella infection. Active caspase-1 has a potential to induce two different types of cell death, depending on the expression of the pyroptosis mediator gasdermin D (GSDMD); following caspase-1 activation, GSDMD-sufficient and GSDMD-null/low cells undergo pyroptosis and apoptosis, respectively. Although Bid, a caspase-1 substrate, plays a critical role in caspase-1 induction of apoptosis in GSDMD-null/low cells, an additional mechanism that mediates this cell death independently of Bid has also been suggested. This study investigated the Bid-independent pathway of caspase-1-induced apoptosis. Caspase-1 has been reported to process caspase-6 and caspase-7. Silencing of caspase-7, but not caspase-6, significantly reduced the activation of caspase-3 induced by caspase-1, which was activated by chemical dimerization, in GSDMD/Bid-deficient cells. CRISPR/Cas9-mediated depletion of caspase-7 had the same effect on the caspase-3 activation. Moreover, in the absence of GSDMD and Bid, caspase-7 depletion reduced apoptosis induced by caspase-1 activation. Caspase-7 was activated following caspase-1 activation independently of caspase-3, suggesting that caspase-7 acts downstream of caspase-1 and upstream of caspase-3. Salmonella induced the activation of caspase-3 in GSDMD-deficient macrophages, which relied partly on Bid and largely on caspase-1. The caspase-3 activation and apoptotic morphological changes seen in Salmonella-infected GSDMD/Bid-deficient macrophages were attenuated by caspase-7 knockdown. These results suggest that in addition to Bid, caspase-7 can also mediate caspase-1-induced apoptosis and provide mechanistic insights into inflammasome-associated cell death that is one major effector mechanism of inflammasomes.
Collapse
Affiliation(s)
- Mamunur Rashid Mahib
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Shoko Hosojima
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroko Kushiyama
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Takeshi Kinoshita
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | - Takashi Suda
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.,Institute for Frontier Science Initiative (InFiniti), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
19
|
Frank AC. Molecular host mimicry and manipulation in bacterial symbionts. FEMS Microbiol Lett 2019; 366:5342066. [PMID: 30877310 DOI: 10.1093/femsle/fnz038] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/18/2019] [Indexed: 12/17/2022] Open
Abstract
It is common among intracellular bacterial pathogens to use eukaryotic-like proteins that mimic and manipulate host cellular processes to promote colonization and intracellular survival. Eukaryotic-like proteins are bacterial proteins with domains that are rare in bacteria, and known to function in the context of a eukaryotic cell. Such proteins can originate through horizontal gene transfer from eukaryotes or, in the case of simple repeat proteins, through convergent evolution. Recent studies of microbiomes associated with several eukaryotic hosts suggest that similar molecular strategies are deployed by cooperative bacteria that interact closely with eukaryotic cells. Some mimics, like ankyrin repeats, leucine rich repeats and tetratricopeptide repeats are shared across diverse symbiotic systems ranging from amoebae to plants, and may have originated early, or evolved independently in multiple systems. Others, like plant-mimicking domains in members of the plant microbiome are likely to be more recent innovations resulting from horizontal gene transfer from the host, or from microbial eukaryotes occupying the same host. Host protein mimics have only been described in a limited set of symbiotic systems, but are likely to be more widespread. Systematic searches for eukaryote-like proteins in symbiont genomes could lead to the discovery of novel mechanisms underlying host-symbiont interactions.
Collapse
Affiliation(s)
- A Carolin Frank
- Life and Environmental Sciences, 5200 North Lake Rd, University of California Merced, Merced, CA 95343, USA.,Sierra Nevada Research Institute, School of Natural Sciences, 5200 North Lake Rd, University of California Merced, Merced, CA 95343, USA
| |
Collapse
|
20
|
Lim MCC, Maubach G, Naumann M. NF-κB-regulated ubiquitin-editing enzyme A20 paves the way for infection persistency. Cell Cycle 2018; 17:3-4. [PMID: 29099284 DOI: 10.1080/15384101.2017.1387435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Michelle C C Lim
- a Institute of Experimental Internal Medicine , Otto von Guericke University , Magdeburg , Germany
| | - Gunter Maubach
- a Institute of Experimental Internal Medicine , Otto von Guericke University , Magdeburg , Germany
| | - Michael Naumann
- a Institute of Experimental Internal Medicine , Otto von Guericke University , Magdeburg , Germany
| |
Collapse
|
21
|
Wang J, Ge P, Qiang L, Tian F, Zhao D, Chai Q, Zhu M, Zhou R, Meng G, Iwakura Y, Gao GF, Liu CH. The mycobacterial phosphatase PtpA regulates the expression of host genes and promotes cell proliferation. Nat Commun 2017; 8:244. [PMID: 28811474 PMCID: PMC5557760 DOI: 10.1038/s41467-017-00279-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/16/2017] [Indexed: 01/05/2023] Open
Abstract
Mycobacterium tuberculosis PtpA is a secreted effector protein that dephosphorylates several proteins in the host cell cytoplasm, such as p-JNK, p-p38, and p-VPS33B, leading to suppression of host innate immunity. Here we show that, in addition, PtpA enters the nucleus of host cells and regulates the expression of host genes, some of which are known to be involved in host innate immunity or in cell proliferation and migration (such as GADD45A). PtpA can bind directly to the promoter region of GADD45A in vitro. Both phosphatase activity and DNA-binding ability of PtpA are important in suppressing host innate immune responses. Furthermore, PtpA-expressing Mycobacterium bovis BCG promotes proliferation and migration of human lung adenoma A549 cells in vitro and in a mouse xenograft model. Further research is needed to test whether mycobacteria, via PtpA, might affect cell proliferation or migration in humans. Mycobacterium tuberculosis secretes a protein, PtpA, that dephosphorylates proteins in the host cell cytoplasm, weakening immune responses. Here, the authors show that PtpA also enters the nucleus, affects the expression of several host genes, and promotes proliferation and migration of a cancer cell line.
Collapse
Affiliation(s)
- Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pupu Ge
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Lihua Qiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute of Health Sciences, Anhui University, Hefei, 230601, China
| | - Feng Tian
- Center for Bioinformatics, Peking University, Beijing, 100871, China
| | - Dongdong Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Mingzhao Zhu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Rongbin Zhou
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, Chinese Academy of Sciences Center for Excellence in Molecular Cell Sciences, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China
| | - Guangxun Meng
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, 278-0022, Japan
| | - George Fu Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China. .,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
22
|
Mahon RN, Hafner R. Applying Precision Medicine and Immunotherapy Advances from Oncology to Host-Directed Therapies for Infectious Diseases. Front Immunol 2017; 8:688. [PMID: 28706516 PMCID: PMC5489679 DOI: 10.3389/fimmu.2017.00688] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/29/2017] [Indexed: 12/11/2022] Open
Abstract
To meet the challenges of increasing antimicrobial resistance, the infectious disease community needs innovative therapeutics. Precision medicine and immunotherapies are transforming cancer therapeutics by targeting the regulatory signaling pathways that are involved not only in malignancies but also in the metabolic and immunologic function of the tumor microenvironment. Infectious diseases target many of the same regulatory pathways as they modulate host metabolic functions for their own nutritional requirements and to impede host immunity. These similarities and the advances made in precision medicine and immuno-oncology that are relevant for the current development of host-directed therapies (HDTs) to treat infectious diseases are discussed. To harness this potential, improvements in drug screening methods and development of assays that utilize the research tools including high throughput multiplexes already developed by oncology are essential. A multidisciplinary approach that brings together immunologists, infectious disease specialists, and oncologists will be necessary to fully develop the potential of HDTs.
Collapse
Affiliation(s)
- Robert N Mahon
- Division of AIDS, Columbus Technologies, Inc., Contractor to National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Richard Hafner
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
23
|
Augenstreich J, Arbues A, Simeone R, Haanappel E, Wegener A, Sayes F, Le Chevalier F, Chalut C, Malaga W, Guilhot C, Brosch R, Astarie-Dequeker C. ESX-1 and phthiocerol dimycocerosates of Mycobacterium tuberculosis act in concert to cause phagosomal rupture and host cell apoptosis. Cell Microbiol 2017; 19. [PMID: 28095608 DOI: 10.1111/cmi.12726] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/12/2017] [Accepted: 01/14/2017] [Indexed: 12/20/2022]
Abstract
Although phthiocerol dimycocerosates (DIM) are major virulence factors of Mycobacterium tuberculosis (Mtb), the causative agent of human tuberculosis, little is known about their mechanism of action. Localized in the outer membrane of mycobacterial pathogens, DIM are predicted to interact with host cell membranes. Interaction with eukaryotic membranes is a property shared with another virulence factor of Mtb, the early secretory antigenic target EsxA (also known as ESAT-6). This small protein, which is secreted by the type VII secretion system ESX-1 (T7SS/ESX-1), is involved in phagosomal rupture and cell death induced by virulent mycobacteria inside host phagocytes. In this work, by the use of several knock-out or knock-in mutants of Mtb or Mycobacterium bovis BCG strains and different cell biological assays, we present conclusive evidence that ESX-1 and DIM act in concert to induce phagosomal membrane damage and rupture in infected macrophages, ultimately leading to host cell apoptosis. These results identify an as yet unknown function for DIM in the infection process and open up a new research field for the study of the interaction of lipid and protein virulence factors of Mtb.
Collapse
Affiliation(s)
- Jacques Augenstreich
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Ainhoa Arbues
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Roxane Simeone
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
| | - Evert Haanappel
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Alice Wegener
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Fadel Sayes
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
| | - Fabien Le Chevalier
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
| | - Christian Chalut
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Wladimir Malaga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Christophe Guilhot
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, Institut Pasteur, Paris, France
| | - Catherine Astarie-Dequeker
- Institut de Pharmacologie et de Biologie Structurale (IPBS), CNRS-Université de Toulouse (UPS), Toulouse, France
| |
Collapse
|
24
|
Friedrich A, Pechstein J, Berens C, Lührmann A. Modulation of host cell apoptotic pathways by intracellular pathogens. Curr Opin Microbiol 2017; 35:88-99. [DOI: 10.1016/j.mib.2017.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/03/2016] [Accepted: 03/01/2017] [Indexed: 12/13/2022]
|
25
|
Jorgensen I, Rayamajhi M, Miao EA. Programmed cell death as a defence against infection. Nat Rev Immunol 2017; 17:151-164. [PMID: 28138137 DOI: 10.1038/nri.2016.147] [Citation(s) in RCA: 719] [Impact Index Per Article: 89.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Eukaryotic cells can die from physical trauma, which results in necrosis. Alternatively, they can die through programmed cell death upon the stimulation of specific signalling pathways. In this Review, we discuss the role of different cell death pathways in innate immune defence against bacterial and viral infection: apoptosis, necroptosis, pyroptosis and NETosis. We describe the interactions that interweave different programmed cell death pathways, which create complex signalling networks that cross-guard each other in the evolutionary 'arms race' with pathogens. Finally, we describe how the resulting cell corpses - apoptotic bodies, pore-induced intracellular traps (PITs) and neutrophil extracellular traps (NETs) - promote the clearance of infection.
Collapse
Affiliation(s)
- Ine Jorgensen
- Department of Immunology, Oslo University Hospital, Sognsvannsveien 20, Rikshospitalet 0372, Oslo, Norway
| | - Manira Rayamajhi
- Camargo Pharmaceutical Services, 2505 Meridian Parkway, Suite 175, Durham, North Carolina 27713, USA
| | - Edward A Miao
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, and Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|