1
|
Mitra P, Deshmukh AS. Proteostasis is a key driver of the pathogenesis in Apicomplexa. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119824. [PMID: 39168412 DOI: 10.1016/j.bbamcr.2024.119824] [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: 01/31/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Proteostasis, including protein folding mediated by molecular chaperones, protein degradation, and stress response pathways in organelles like ER (unfolded protein response: UPR), are responsible for cellular protein quality control. This is essential for cell survival as it regulates and reprograms cellular processes. Here, we underscore the role of the proteostasis pathway in Apicomplexan parasites with respect to their well-characterized roles as well as potential roles in many parasite functions, including survival, multiplication, persistence, and emerging drug resistance. In addition to the diverse physiological importance of proteostasis in Apicomplexa, we assess the potential of the pathway's components as chemotherapeutic targets.
Collapse
Affiliation(s)
- Pallabi Mitra
- BRIC-Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| | | |
Collapse
|
2
|
Chen J, Zhang C, Yang Z, Wu W, Zou W, Xin Z, Zheng S, Liu R, Yang L, Peng H. Intestinal microbiota imbalance resulted by anti-Toxoplasma gondii immune responses aggravate gut and brain injury. Parasit Vectors 2024; 17:284. [PMID: 38956725 PMCID: PMC11221008 DOI: 10.1186/s13071-024-06349-8] [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: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Toxoplasma gondii infection affects a significant portion of the global population, leading to severe toxoplasmosis and, in immunocompromised patients, even death. During T. gondii infection, disruption of gut microbiota further exacerbates the damage to intestinal and brain barriers. Therefore, identifying imbalanced probiotics during infection and restoring their equilibrium can regulate the balance of gut microbiota metabolites, thereby alleviating tissue damage. METHODS Vimentin gene knockout (vim-/-) mice were employed as an immunocompromised model to evaluate the influence of host immune responses on gut microbiota balance during T. gondii infection. Behavioral experiments were performed to assess changes in cognitive levels and depressive tendencies between chronically infected vim-/- and wild-type (WT) mice. Fecal samples were subjected to 16S ribosomal RNA (rRNA) sequencing, and serum metabolites were analyzed to identify potential gut probiotics and their metabolites for the treatment of T. gondii infection. RESULTS Compared to the immunocompetent WT sv129 mice, the immunocompromised mice exhibited lower levels of neuronal apoptosis and fewer neurobehavioral abnormalities during chronic infection. 16S rRNA sequencing revealed a significant decrease in the abundance of probiotics, including several species of Lactobacillus, in WT mice. Restoring this balance through the administration of Lactobacillus murinus and Lactobacillus gasseri significantly suppressed the T. gondii burden in the intestine, liver, and brain. Moreover, transplantation of these two Lactobacillus spp. significantly improved intestinal barrier damage and alleviated inflammation and neuronal apoptosis in the central nervous system. Metabolite detection studies revealed that the levels of various Lactobacillus-related metabolites, including indole-3-lactic acid (ILA) in serum, decreased significantly after T. gondii infection. We confirmed that L. gasseri secreted much more ILA than L. murinus. Notably, ILA can activate the aromatic hydrocarbon receptor signaling pathway in intestinal epithelial cells, promoting the activation of CD8+ T cells and the secretion of interferon-gamma. CONCLUSION Our study revealed that host immune responses against T. gondii infection severely disrupted the balance of gut microbiota, resulting in intestinal and brain damage. Lactobacillus spp. play a crucial role in immune regulation, and the metabolite ILA is a promising therapeutic compound for efficient and safe treatment of T. gondii infection.
Collapse
Affiliation(s)
- Jiating Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Chi Zhang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Zihan Yang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Weiling Wu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Weihao Zou
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Zixuan Xin
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Shuyu Zheng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Runchun Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Lili Yang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Diseases Research, School of Public Health, Key Laboratory of Infectious Diseases Research in South China (Southern Medical University), Ministry of Education, Southern Medical University, 1023-1063 South Shatai Rd, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
3
|
Hu Z, Wu T, Zhou Z, Zhang Y, Chen Q, Yao H, Ji M, Shen G, Dong C, Shi C, Huang Z, Jiang N, Han N, Tian X. Asiaticoside Attenuates Blood-Spinal Cord Barrier Disruption by Inhibiting Endoplasmic Reticulum Stress in Pericytes After Spinal Cord Injury. Mol Neurobiol 2024; 61:678-692. [PMID: 37653222 DOI: 10.1007/s12035-023-03605-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
The blood-spinal cord barrier (BSCB) plays a vital role in the recovery of spinal cord function after spinal cord injury (SCI). Pericytes, pluripotent members of the neurovascular unit (NVU), receive signals from neighboring cells and are critical for maintaining CNS function. Therapeutic targets for the BSCB include endothelial cells (ECs) and glial cells, but few drugs target pericytes. This study was designed to explore whether asiaticoside has a positively effect on pericytes and the integrity of the BSCB. In this study, we found that asiaticoside could inhibit the loss of junction proteins just 1 day after SCI in vivo, but our in vitro study showed no significant differences in the expression of endothelial junction proteins between the control and asiaticoside treatment groups. We also found that asiaticoside could inhibit endoplasmic reticulum (ER) stress and pericyte apoptosis, which might be associated with the inhibition of junction protein reduction in ECs. Thus, we investigated the interactions between pericytes and ECs. Our results showed that asiaticoside could decrease the release of matrix metalloproteinase (MMP)-9 in pericytes and therefore upregulate the expression of junction proteins in ECs. Furthermore, the protective effect of asiaticoside on pericytes is related to the inhibition of ER stress via the MAPK signaling pathway. Taken together, our results demonstrate that asiaticoside treatment inhibits BSCB disruption and enhances functional recovery after SCI.
Collapse
Affiliation(s)
- Zhenxin Hu
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Tingting Wu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ziheng Zhou
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yu Zhang
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315302, China
| | - Qiyue Chen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hanbing Yao
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Mengchu Ji
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ge Shen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chenling Dong
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chengge Shi
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhixian Huang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325035, China
| | - Nizhou Jiang
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Nan Han
- Department of Ultrasonography, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Xiliang Tian
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
4
|
Cudjoe O, Afful R, Hagan TA. Toxoplasma-host endoplasmic reticulum interaction: How T. gondii activates unfolded protein response and modulates immune response. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 6:100223. [PMID: 38352129 PMCID: PMC10861954 DOI: 10.1016/j.crmicr.2024.100223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Toxoplasma gondii is a neurotropic single-celled zoonotic parasite that can infect human beings and animals. Infection with T. gondii is usually asymptomatic in immune-competent individual, however, it can cause symptomatic and life-threatening conditions in immunocompromised individuals and in developing foetuses. Although the mechanisms that allow T. gondii to persist in host cells are poorly understood, studies in animal models have greatly improved our understanding of Toxoplasma-host cell interaction and how this interaction modulates parasite proliferation and development, host immune response and virulence of the parasite. T. gondii is capable of recruiting the host endoplasmic reticulum (ER), suggesting it may influence the host ER function. Herein, we provide an overview of T. gondii infection and the role of host ER during stressed conditions. Furthermore, we highlight studies that explore T. gondii's interaction with the host ER. We delve into how this interaction activates the unfolded protein response (UPR) and ER stress-mediated apoptosis. Additionally, we examine how T. gondii exploits these pathways to its advantage.
Collapse
Affiliation(s)
- Obed Cudjoe
- Department of Medical Laboratory Science, Klintaps College of Health and Allied Sciences, DTD TDC Plot 30A, Klagon, Tema, Ghana
- Department of Microbiology and Immunology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Ghana
| | - Roger Afful
- Department of Medical Laboratory Science, Klintaps College of Health and Allied Sciences, DTD TDC Plot 30A, Klagon, Tema, Ghana
| | - Tonny Abraham Hagan
- Department of Biomedical Engineering, School of Life Science and Technology, University of Electronic Science and Technology of China, China
| |
Collapse
|
5
|
Jung DM, Kim KK, Kim EM. Chloromethylisothiazolinone induces ER stress-induced stress granule formation in human keratinocytes. Anim Cells Syst (Seoul) 2023; 27:171-179. [PMID: 37636324 PMCID: PMC10448836 DOI: 10.1080/19768354.2023.2250852] [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: 03/13/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023] Open
Abstract
Chloromethylisothiazolinone (CMIT), a humidifier disinfectant, is known to be toxic to the respiratory system. While the toxic effect of CMIT on the lungs has been widely investigated, its effect on the skin is well unknown. In this study, we examined stress granule (SG) formation to investigate the cytotoxic effects of CMIT on human keratinocytes. We assessed the viability of the cells following CMIT exposure and performed immunofluorescence microscopy and immunoblot analyses to determine SG formation and downstream pathways. The IC50 values in human keratinocyte HaCaT cells after CMIT exposure for 1 and 24 h were 11 and 8 μg/mL, respectively, showing no significant difference. As determined using immunofluorescence microscopy, SG formation was effectively induced after CMIT exposure. Moreover, the phosphorylation of eukaryotic initiation factor-2α (eIF2α), a translation initiation factor, and protein kinase R-like endoplasmic reticulum (ER) kinase, which plays a role in the ER stress-mediated eIF2α phosphorylation, was confirmed by CMIT exposure. These results suggest that exposure to CMIT can have detrimental effects on the skin, even briefly, by inducing SG formation through ER stress in keratinocytes.
Collapse
Affiliation(s)
- Da-Min Jung
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Kee K. Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Mi Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| |
Collapse
|
6
|
Liu G, Guan Y, Liu Y, Wang Y, Zhang J, Liu Y, Liu X. Saikosaponin D Inducing Apoptosis and Autophagy through the Activation of Endoplasmic Reticulum Stress in Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1-12. [PMID: 36467888 PMCID: PMC9715330 DOI: 10.1155/2022/5489553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
Saikosaponin D (SSD), a saponin derivative, is extracted from Bupleurum falcatum. It exhibits an inhibitory effect on a number of tumor cells and is relatively safe when used at therapeutic doses. However, its effects on glioblastoma multiforme (GBM) have not been fully explored. This study is aimed at investigating the cytotoxic effects of SSD in GBM cell lines. SSD induces apoptosis and autophagy by activating endoplasmic reticulum (ER) stress in GBM cells. GBM cell proliferation activity and morphology were observed using the Cell Counting Kit-8 assay and hematoxylin and eosin staining. Hoechst 33258 fluorescence staining and flow cytometry were performed to assess apoptosis. Western blotting and immunocytochemical staining were used to detect protein expression and distribution. SSD significantly inhibited the proliferation of RG-2, U87-MG, and U251 cells in a dose-dependent manner, and the proportion of apoptotic cells increased significantly. Additionally, the expressions of ER-, apoptosis-, and autophagy-related proteins were significantly upregulated and distributed in the cytoplasm and nucleus. Therefore, SSD may be considered a novel treatment option for GBM. This study demonstrated the anti-GBM effect of SSD from the perspectives of cell apoptosis and autophagy.
Collapse
Affiliation(s)
- Guimei Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yuehong Guan
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yongxian Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yaping Wang
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Jing Zhang
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yusi Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Xiaobin Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| |
Collapse
|
7
|
Wu XJ, Gao J, Mu BJ, Yu LM, Wang ZR, Zheng WB, Gao WW, Zhu XQ, Liu Q. Transcriptomic analysis of LMH cells in response to the overexpression of a protein of Eimeria tenella encoded by the locus ETH_00028350. Front Vet Sci 2022; 9:1053701. [DOI: 10.3389/fvets.2022.1053701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
A protein of Eimeria tenella (encoded by the locus ETH_00028350) homologous to Toxoplasma gondii dense granule protein 9, designated as EtHGRA9 hereafter, was reported to be expressed in all life cycle stages of E. tenella. However, no data are currently available regarding its functional properties. In the present study, a recombinant vector harboring a 741 bp gene segment encoding the mature form of EtHGRA9 was constructed and transfected into leghorn male hepatoma (LMH) cells. Then, transcriptomic analysis of the transfected LMH cells was carried out by using a high-throughput RNA-seq technology. The LMH cells overexpressing EtHGRA9 was validated by means of Western blotting as well as indirect immunofluorescence staining. The results demonstrated that the expression of 547 genes (275 upregulated genes and 272 downregulated genes) was altered by EtHGRA9. The quantitative real-time polymerase chain reaction (qRT-PCR) validation of the ten genes with differential expression between the two groups was consistent with the transcriptome analysis. According to pathway enrichment analysis for the obtained differentially expressed genes, seven pathways were significantly affected by EtHGRA9, such as cytokine-cytokine receptor interaction, MAPK signaling pathway, and protein processing in endoplasmic reticulum. Our data reveal several possible roles of EtHGRA9 in immune or inflammatory responses, which paves the way for a better understanding of the molecular interplay between E. tenella and its host.
Collapse
|