1
|
Zhou R, Zou X, Yu J, Wang Z, Lu W, Li X, Wei C, Li X, Wang F. KDELR3 and YOD1 proteins as critical endoplasmic reticulum stress mediators and potential therapeutic targets in diabetic foot ulcers: An integrated bioinformatics analysis. Int J Biol Macromol 2025:144095. [PMID: 40354856 DOI: 10.1016/j.ijbiomac.2025.144095] [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: 03/02/2025] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Diabetic foot ulcers (DFU) represent one of the most severe complications of diabetes mellitus and are closely associated with persistent hyperglycemia. Endoplasmic reticulum stress response proteins play critical roles in the development and progression of DFU, highlighting the urgent need for further research to identify novel biomarkers and therapeutic strategies. METHOD This study utilized DFU datasets from the GEO database and employed bioinformatics approaches to identify differentially expressed genes encoding endoplasmic reticulum stress (ERS) response proteins. Key regulatory proteins NCCRP1, KDELR3, BOK, and YOD1 were screened using WGCNA, machine learning algorithms, and molecular docking techniques, followed by an evaluation of their correlation with the immune microenvironment. Additionally, single-cell RNA sequencing and Mendelian randomization analysis were applied to investigate the structural and functional characteristics of these proteins in DFU pathogenesis. The expression levels of key protein biomarkers were validated using qRT-PCR. RESULT A total of 32 differentially expressed endoplasmic reticulum stress-related proteins associated with DFU were identified. Machine learning algorithms confirmed that NCCRP1, KDELR3, BOK, and YOD1 proteins demonstrated significant diagnostic potential as biomarkers. Immune analysis revealed associations between these stress-response proteins and immune cell infiltration, while molecular docking identified metronidazole as a promising therapeutic candidate targeting the KDELR3 and YOD1 protein structures. Experimental validation confirmed the differential expression of KDELR3 and YOD1 proteins in DFU tissues, and Mendelian randomization analysis suggested that BOK protein may be a potential causal factor in DFU development due to its structural interactions with ERS pathways. CONCLUSION Our study characterized specific ERS response proteins with significant diagnostic potential for DFU. These findings enhance the understanding of protein-mediated DFU pathogenesis and lay the foundation for improving diagnostic and therapeutic strategies targeting these biological macromolecules.
Collapse
Affiliation(s)
- Rongbin Zhou
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, Guangxi, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Urology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, China
| | - Xiaochong Zou
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi 530021, China
| | - Jiayin Yu
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi 530021, China
| | - Zuheng Wang
- Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi 530021, China
| | - Wenhao Lu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, Guangxi, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiao Li
- School of Life Sciences, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Chunmeng Wei
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi 530021, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Fubo Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, Guangxi, China; Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Urology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi 530021, China; School of Life Sciences, Guangxi Medical University, Nanning 530021, Guangxi, China; Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning 530021, Guangxi, China; School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
2
|
Hinkle JJ, Trychta KA, Wires ES, Osborn RM, Leach JR, Faraz ZF, Svarcbahs R, Richie CT, Dewhurst S, Harvey BK. Subcellular localization of SARS-CoV-2 E and 3a proteins along the secretory pathway. J Mol Histol 2025; 56:98. [PMID: 40025386 PMCID: PMC11872775 DOI: 10.1007/s10735-025-10375-w] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Abstract
SARS-CoV-2 E and 3a proteins are important for the assembly, budding, and release of viral particles. These two transmembrane proteins have been implicated in forming channels in the membrane that allow the transport of ions to favor viral replication. During an active infection, both proteins generally localize to the endoplasmic reticulum (ER), ER-Golgi intermediate compartment (ERGIC), and the Golgi where viral assembly occurs. The ER and Golgi are critical for the proper packaging and trafficking of cellular proteins along the secretory pathways which determine a protein's final destination inside or outside of the cell. The SARS-CoV-2 virus primarily infects epithelial cells that are highly secretory in nature such as those in the lung and gut. Here we quantified the distribution of SARS-CoV-2 E and 3a proteins along the secretory pathways in a human intestinal epithelial cell line. We used NaturePatternMatch to demonstrate that epitope-tagged E and 3a proteins expressed alone via transient transfection have a similar immunoreactivity pattern as E and 3a proteins expressed by wild-type viral infection. While E and 3a proteins localized with all selected cellular markers to varying degrees, 3a protein displayed a higher correlation coefficient with the Golgi, early/late endosome, lysosome, and plasma membrane when compared to E protein. This work is the first to provide quantification of the subcellular distribution of E and 3a proteins along the multiple components of the secretory pathway and serves as a basis to develop models for examining how E and 3a alter proteostasis within these structures and affect their function.
Collapse
Affiliation(s)
- Joshua J Hinkle
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| | - Kathleen A Trychta
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Emily S Wires
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Raven M Osborn
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Justin R Leach
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Zoha F Faraz
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Reinis Svarcbahs
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Stephen Dewhurst
- School of Medicine & Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, NIH, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Di Mattia M, Sallese M, Lopetuso LR. The interplay between gut microbiota and the unfolded protein response: Implications for intestinal homeostasis preservation and dysbiosis-related diseases. Microb Pathog 2025; 200:107279. [PMID: 39761770 DOI: 10.1016/j.micpath.2025.107279] [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: 03/08/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
The unfolded protein response (UPR) is a complex intracellular signal transduction system that orchestrates the cellular response during Endoplasmic Reticulum (ER) stress conditions to reestablish cellular proteostasis. If, on one side, prolonged ER stress conditions can lead to programmed cell death and autophagy as a cytoprotective mechanism, on the other, unresolved ER stress and improper UPR activation represent a perilous condition able to trigger or exacerbate inflammatory responses. Notably, intestinal and immune cells experience ER stress physiologically due to their high protein secretory rate. Indeed, there is evidence of UPR's involvement in both physiological and pathological intestinal conditions, while less is known about its bidirectional interaction with gut microbiota. However, gut microbes and their metabolites can influence ER stress and UPR pathways, and, in turn, ER stress conditions can shape gut microbiota composition, with important implications for overall intestinal health. Thus, targeting UPR components is an intriguing strategy for treating ER stress-linked dysbiosis and diseases, particularly intestinal inflammation.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
4
|
Omoluabi T, Hasan Z, Piche JE, Flynn ARS, Doré JJE, Walling SG, Weeks ACW, Benoukraf T, Yuan Q. Locus coeruleus vulnerability to tau hyperphosphorylation in a rat model. Aging Cell 2025; 24:e14405. [PMID: 39520141 PMCID: PMC11896524 DOI: 10.1111/acel.14405] [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: 08/02/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Post-mortem investigations indicate that the locus coeruleus (LC) is the initial site of hyperphosphorylated pretangle tau, a precursor to neurofibrillary tangles (NFTs) found in Alzheimer's disease (AD). The presence of pretangle tau and NFTs correlates with AD progression and symptomatology. LC neuron integrity and quantity are linked to cognitive performance, with degeneration strongly associated with AD. Despite their importance, the mechanisms of pretangle tau-induced LC degeneration are unclear. This study examined the transcriptomic and mitochondrial profiles of LC noradrenergic neurons after transduction with pseudophosphorylated human tau. Tau hyperphosphorylation increased the somatic expression of the L-type calcium channel (LTCC), impaired mitochondrial health, and led to deficits in spatial and olfactory learning. Sex-dependent alterations in gene expression were observed in rats transduced with pretangle tau. Chronic LTCC blockade prevented behavioral deficits and altered mitochondrial mRNA expression, suggesting a potential link between LTCC hyperactivity and mitochondrial dysfunction. Our research provides insights into the consequences of tau pathology in the originating structure of AD.
Collapse
Affiliation(s)
- Tamunotonye Omoluabi
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Zia Hasan
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Jessie E. Piche
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
- Department of Psychology, Faculty of Arts & ScienceNipissing UniversityNorth BayOntarioCanada
| | - Abeni R. S. Flynn
- Department of Psychology, Faculty of Arts & ScienceNipissing UniversityNorth BayOntarioCanada
| | - Jules J. E. Doré
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Susan G. Walling
- Department of Psychology, Faculty of ScienceMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Andrew C. W. Weeks
- Department of Psychology, Faculty of Arts & ScienceNipissing UniversityNorth BayOntarioCanada
| | - Touati Benoukraf
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| | - Qi Yuan
- Biomedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sNewfoundlandCanada
| |
Collapse
|
5
|
Wang W, Yun B, Xiang Z, Liu X, Yi C, Ouyang S, Zhang X, Xiong G, Zhuang Z, Wang C. A multi-omics analysis reveals KDELR1 promotes malignant progression and correlates with tumor microenvironment in head and neck squamous cell carcinoma. Cell Signal 2025; 127:111613. [PMID: 39875049 DOI: 10.1016/j.cellsig.2025.111613] [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: 07/23/2024] [Revised: 01/05/2025] [Accepted: 01/18/2025] [Indexed: 01/30/2025]
Abstract
KDELR1, a constituent of the KDEL endoplasmic reticulum protein retention receptors family, is implicated in immune responses and cancers progression. In this study, we delineate the clinicopathological significance and oncogenic role of KDELR1 in head and neck squamous cell carcinoma (HNSCC) through a comprehensive multi-omics approach. KDELR1 expression is correlated with tumor grade, tumor stage, lymph node metastasis, clinical stage and poor prognosis in HNSCC. Moreover, our results indicate a marked upregulation of KDELR1 expression in primary tumor tissues compared to normal and dysplasia tissues. Furthermore, increased KDELR1 expression is associated with tumor progression and indicates an unfavorable prognosis in HNSCC. Functional enrichment analysis highlights the involvement of KDELR1 in HNSCC malignant progression. Depletion of KDELR1 inhibits proliferation, stemness, migration and invasion in HNSCC cells. Bioinformatics analysis results indicate that KDELR1 promotes HNSCC progression with regulating wnt signaling pathway and CTNNB1 expression. Of note, KDELR1 is associated with HNSCC tumor microenvironment compositions, especially positively correlating with cancer associated fibroblasts and negatively correlating with CD8+ T cells and B cells infiltration. Collectively, these findings indicate that KDELR1 as an oncogene in driving progression and correlates with tumor microenvironment, suggesting its potential as a promising biomarker and a therapeutic target in HNSCC.
Collapse
Affiliation(s)
- Wenjin Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Bokai Yun
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhuqin Xiang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoyong Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Chen Yi
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Shengqi Ouyang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiliu Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Gan Xiong
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Zehang Zhuang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
| | - Cheng Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
6
|
Alexander C, Guo Z, Glover PB, Faulkner S, Pikramenou Z. Luminescent Lanthanides in Biorelated Applications: From Molecules to Nanoparticles and Diagnostic Probes to Therapeutics. Chem Rev 2025; 125:2269-2370. [PMID: 39960048 PMCID: PMC11869165 DOI: 10.1021/acs.chemrev.4c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Lanthanides are particularly effective in their clinical applications in magnetic resonance imaging and diagnostic assays. They have open-shell 4f electrons that give rise to characteristic narrow, line-like emission which is unique from other fluorescent probes in biological systems. Lanthanide luminescence signal offers selection of detection pathways based on the choice of the ion from the visible to the near-infrared with long luminescence lifetimes that lend themselves to time-resolved measurements for optical multiplexing detection schemes and novel bioimaging applications. The delivery of lanthanide agents in cells allows localized bioresponsive activity for novel therapies. Detection in the near-infrared region of the spectrum coupled with technological advances in microscopies opens new avenues for deep-tissue imaging and surgical interventions. This review focuses on the different ways in which lanthanide luminescence can be exploited in nucleic acid and enzyme detection, anion recognition, cellular imaging, tissue imaging, and photoinduced therapeutic applications. We have focused on the hierarchy of designs that include luminescent lanthanides as probes in biology considering coordination complexes, multimetallic lanthanide systems to metal-organic frameworks and nanoparticles highlighting the different strategies in downshifting, and upconversion revealing some of the opportunities and challenges that offer potential for further development in the field.
Collapse
Affiliation(s)
- Carlson Alexander
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
- Department
of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Zhilin Guo
- Department
of Materials Science and Engineering, Southern
University of Science and Technology, Shenzhen 518055, China
| | - Peter B. Glover
- Defence
Science and Technology Laboratory (DSTL), Porton Down, Salisbury SP4 0JQ, United
Kingdom
| | - Stephen Faulkner
- Chemistry
Research Laboratory, Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Zoe Pikramenou
- School
of Chemistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
7
|
Di Mattia M, Sallese M, Lopetuso LR. Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease. Acta Physiol (Oxf) 2025; 241:e14284. [PMID: 39822064 DOI: 10.1111/apha.14284] [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: 06/11/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Different physiological and pathological situations can produce alterations in the cell's endoplasmic reticulum (ER), leading to a condition known as ER stress, which can trigger an intricate intracellular signal transduction system known as the unfolded protein response (UPR). UPR is primarily tailored to restore proteostasis and ER equilibrium; otherwise, if ER stress persists, it can cause programmed cell death as a cytoprotective mechanism and drive inflammatory processes. Therefore, since intestinal cells strongly rely on UPR for their biological functions and unbalanced UPR has been linked to inflammatory, metabolic, and immune disorders, here we discussed the role of the UPR within the intestinal tract, focusing on the UPR contribution to inflammatory bowel disease development. Importantly, we also highlighted the promising potential of UPR components as therapeutic targets for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
8
|
Feng J, Zhao L, Fu L, Wang X, Ma D, Shang M, Xu B, Zhou J, Chen Z, Zhao H. KDELR3 overexpression as a novel prognostic and diagnostic biomarker in glioma: comprehensive bioinformatic analysis insights. Sci Rep 2024; 14:30783. [PMID: 39730475 DOI: 10.1038/s41598-024-80991-1] [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: 08/08/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
Glioma, the most prevalent primary intracranial malignancy among adults, is distinguished by its high morbidity and recurrence rates, posing a considerable threat to patients' quality of life and survival prospects. Consequently, the pursuit of efficacious molecular prognostic markers holds paramount importance. The exploration of the role of the KDELR3 kinase family in various neoplastic conditions constitutes an emerging area of research. However, the biological functions of KDELR3 and its prognostic implications in brain tumors remain largely undocumented. This study endeavored to ascertain the potential of KDELR3 as a prognostic indicator for glioma. We integrated a comprehensive dataset encompassing 1127 glioma samples, sourced from our cohort, The Cancer Genome Atlas (TCGA), and the Chinese Glioma Genome Atlas (CGGA), to delve into the expression patterns of KDELR3 in glioma and their associated implications. Notably, KDELR3 was markedly overexpressed in glioma and demonstrated a positive correlation with clinical progression. By utilizing Kaplan-Meier survival analysis and the Cox proportional hazards regression model, we evaluated the prognostic significance of KDELR3, revealing it as an independent predictor of adverse outcomes in glioma patients. Furthermore, gene set enrichment analysis unveiled potential signaling pathways associated with KDELR3 expression in glioma, primarily encompassing Cytokine-cytokine receptor interaction, extracellular matrix (ECM)-receptor interaction, and complement and coagulation cascades. In summation, our findings provide profound insights into the potential role of KDELR3 and its application as a novel and promising prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Jing Feng
- Department of Radiation Oncology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
- Department of Radiation Oncology, School of Medicine, Dongfang Hospital of Xiamen University, Xiamen University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
- Department of Radiation Oncology, Fujian University of Traditional Chinese Medicine, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Lin Zhao
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Liyuan Fu
- Department of Diagnostic Radiology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Xinpeng Wang
- Department of Radiation Oncology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Danyu Ma
- Department of Radiation Oncology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Mingchao Shang
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Baoqing Xu
- Department of Pathology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Jinping Zhou
- Department of Clinical Quality Management, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
| | - Zhonghua Chen
- Department of Radiation Oncology, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
- Department of Radiation Oncology, School of Medicine, Dongfang Hospital of Xiamen University, Xiamen University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
- Department of Radiation Oncology, Fujian University of Traditional Chinese Medicine, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
| | - Hu Zhao
- Department of General Surgery, Fuzong Clinical Medical College of Fujian Medical University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
- Department of General Surgery, School of Medicine, Dongfang Hospital of Xiamen University, Xiamen University, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
- Department of General Surgery, Fujian University of Traditional Chinese Medicine, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China.
| |
Collapse
|
9
|
Xie S, Yin M, Xiang M, Shao L, Zhang N, Shi L, Zhang J, Yu G. Lead (Pb) Induces Osteotoxicity Through the Activation of Mutually Reinforced ER Stress and ROS in MC3T3-E1 Cells. Biol Trace Elem Res 2024:10.1007/s12011-024-04427-7. [PMID: 39643796 DOI: 10.1007/s12011-024-04427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/17/2024] [Indexed: 12/09/2024]
Abstract
Lead (Pb) is the most common contaminant of heavy metals and is widely present in the environment. Destruction of bone structure, malformation of bone development, and loss of bone mass are important pathological features of lead-exposed individuals. However, the exact molecular mechanisms associated with lead exposure and osteogenic injury are still not fully understood. MC3T3-E1 mouse embryonic osteoblast is a cell line widely used in osteoblast cytology. It can differentiate into mature osteoblasts and express bone-specific genes in cell culture. The doses of 1, 2, and 4 mM Pb were adopted to study the toxicity of Pb on MC3T3-E1 proliferation and differentiation. In this study, the results show that Pb increases the expression of apoptosis-related proteins, including PARP1, cleaved caspase-3, Bax, and cleaved caspase-9. More importantly, Pb activated endoplasmic reticulum stress and oxidative stress, as evident by elevated PERK/ATF4/CHOP and ROS/NRF2 signaling pathway. Pb induced ROS production in MC3T3-E1 cells through endoplasmic reticulum stress and produced a lethal effect. NAC mitigated these effects. Endoplasmic reticulum stress inhibitor 4-PBA can block the ER stress pathway, reduce ROS production, and enhance cell viability. In addition, studies have shown that ERO1 activation in the ER stress pathway is responsible for inducing ROS production. ROS produced by the mitochondrial pathway also aggravates ER stress. This study suggests that Pb induces MC3T3-E1 cell apoptosis by inducing PERK-mediated ER stress and NRF2-mediated oxidative stress via mutual enhancement, which may be an important mechanism leading to skeletal toxicity.
Collapse
Affiliation(s)
- Siwen Xie
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Meng Yin
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengting Xiang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Litao Shao
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Nan Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Juan Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Gongchang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Shandong Mental Health Center, Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Lv H, Wang J, Wan Y, Zhou Y. Exploration of the Key Pathways and Genes Involved in Osteoarthritis Genesis: Evidence from Multiple Platforms and Real-World Validation. J Inflamm Res 2024; 17:10223-10237. [PMID: 39649419 PMCID: PMC11625429 DOI: 10.2147/jir.s488935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/14/2024] [Indexed: 12/10/2024] Open
Abstract
Background Osteoarthritis (OA), a degenerative and chronic joint disease, is essential for identifying novel biomarkers for the clinical diagnosis of OA. Methods We collected 35 OA patients and 32 healthy controls from four clinical cohorts and 8 real-world samples from our institute. The activation status of 7530 signalling pathways was calculated via the gene set enrichment analysis (GSEA) algorithm. Ten machine learning algorithms and 101 algorithm combinations were further applied to recognize the most diagnostic genes. KDELR3 was chosen for further validation via immunohistochemical staining to determine its diagnostic value in real-world samples. Results Sixteen pathways, namely, the cellular respiration chain, protein transport, lysosomal and endocytosis pathways, were activated in OA patients. A total of 101 types of algorithm combinations were considered for the diagnostic model, and 58 were successfully output. The two-step model of glmBoost plus RF had the highest average AUC value of 0.95 and was composed of LY86, SORL1, KDELR3, CSK, PTGS1, and PTGS2. Preferable consistency of the diagnostic mole and real conditions was observed in all four cohorts (GSE55235: Kappa=1.000, P<0.001; GSE55457: Kappa=0.700, P<0.001; GSE82107: Kappa=0.643, P=0.004; GSE1919: Kappa=1.000, P<0.001). KDELR3 was expressed at higher levels in OA patients than were the other genes, and with the help of immunohistochemistry (IHC), we confirmed that OA patients presented high levels of KDELR3 in synovial tissues. The infiltration of immunocytes, macrophages, and natural killer T cells was high in OA patients. KDELR3 might be involved in the activation and infiltration of effector memory CD4 T cells (Rpearson = 0.58, P < 0.001) and natural killer T cells (Rpearson = 0.53, P < 0.001). Conclusion We constructed and validated a six-gene diagnostic model for OA patients via machine learning, and KDELR3 emerged as a novel biomarker for OA.
Collapse
Affiliation(s)
- Hao Lv
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
| | - Jingkun Wang
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
| | - Yang Wan
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
- Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People’s Republic of China
| |
Collapse
|
11
|
Kong YX, Perdomo JS, Passam FH. A new track for KDELivery of designer cargo by platelets. J Thromb Haemost 2024; 22:3007-3009. [PMID: 39461809 DOI: 10.1016/j.jtha.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/29/2024]
Affiliation(s)
- Yvonne X Kong
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Jose S Perdomo
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Freda H Passam
- Haematology Research Group, Charles Perkins Centre, Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
12
|
Kong YX, Chiu J, Passam FH. "Sticki-ER": Functions of the Platelet Endoplasmic Reticulum. Antioxid Redox Signal 2024; 41:637-660. [PMID: 38284332 DOI: 10.1089/ars.2024.0566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Significance: The primary role of platelets is to generate a thrombus by platelet activation. Platelet activation relies on calcium mobilization from the endoplasmic reticulum (ER). ER resident proteins, which are externalized upon platelet activation, are essential for the function of platelet surface receptors and intercellular interactions. Recent Advances: The platelet ER is a conduit for changes in cellular function in response to the extracellular milieu. ER homeostasis is maintained by an appropriate redox balance, regulated calcium stores and normal protein folding. Alterations in ER function and ER stress results in ER proteins externalizing to the cell surface, including members of the protein disulfide isomerase family (PDIs) and chaperones. Critical Issues: The platelet ER is central to platelet function, but our understanding of its regulation is incomplete. Previous studies have focused on the function of PDIs in the extracellular space, and much less on their intracellular role. How platelets maintain ER homeostasis and how they direct ER chaperone proteins to facilitate intercellular signalling is unknown. Future Directions: An understanding of ER functions in the platelet is essential as these may determine critical platelet activities such as secretion and adhesion. Studies are necessary to understand the redox reactions of PDIs in the intracellular versus extracellular space, as these differentially affect platelet function. An unresolved question is how platelet ER proteins control calcium release. Regulation of protein folding in the platelet and downstream pathways of ER stress require further evaluation. Targeting the platelet ER may have therapeutic application in metabolic and neoplastic disease.
Collapse
Affiliation(s)
- Yvonne X Kong
- Haematology Research Group, Charles Perkins Centre; The University of Sydney, Camperdown, New South Wales, Australia
- Central Clinical School, Faculty of Medicine and Health; The University of Sydney, Camperdown, New South Wales, Australia
- Department of Haematology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Joyce Chiu
- ACRF Centenary Cancer Research Centre, The Centenary Institute; The University of Sydney, Camperdown, New South Wales, Australia
| | - Freda H Passam
- Haematology Research Group, Charles Perkins Centre; The University of Sydney, Camperdown, New South Wales, Australia
- Central Clinical School, Faculty of Medicine and Health; The University of Sydney, Camperdown, New South Wales, Australia
- Department of Haematology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
13
|
Wu X, Du F, Zhang A, Zhang G, Xu R, Du X. KDELR2 is necessary for chronic obstructive pulmonary disease airway Mucin5AC hypersecretion via an IRE1α/XBP-1s-dependent mechanism. J Cell Mol Med 2024; 28:e70125. [PMID: 39365189 PMCID: PMC11451269 DOI: 10.1111/jcmm.70125] [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: 11/02/2023] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
Airway mucus hypersecretion, a crucial pathological feature of chronic obstructive pulmonary disease (COPD), contributes to the initiation, progression, and exacerbation of this disease. As a macromolecular mucin, the secretory behaviour of Mucin5AC (MUC5AC) is highly dependent on a series of modifying and folding processes that occur in the endoplasmic reticulum (ER). In this study, we focused on the ER quality control protein KDEL receptor (KDELR) and demonstrated that KDELR2 and MUC5AC were colocalized in the airway epithelium of COPD patients and COPD model rats. In addition, knockdown of KDELR2 markedly reduced the expression of MUC5AC both in vivo and in vitro and knockdown of ATF6 further decreased the levels of KDELR2. Furthermore, pretreatment with 4μ8C, an IRE1α inhibitor, led to a partial reduction in the expression of KDELR2 and MUC5AC both in vivo and in vitro, which indicated the involvement of IRE1α/XBP-1s in the upstream signalling cascade. Our study revealed that KDELR2 plays a crucial role in airway MUC5AC hypersecretion in COPD, which might be dependent on ATF6 and IRE1α/XBP-1s upstream signalling.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Fawang Du
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Aijie Zhang
- Basic Laboratory, Key Laboratory of Metabolic DiseasesSuining Central HospitalSuiningChina
| | - Guoyue Zhang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Rui Xu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xianzhi Du
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
14
|
Dossat AM, Trychta KA, Glotfelty EJ, Hinkle JJ, Fortuno LV, Gore LN, Richie CT, Harvey BK. Excitotoxic glutamate levels cause the secretion of resident endoplasmic reticulum proteins. J Neurochem 2024; 168:2461-2478. [PMID: 38491746 PMCID: PMC11401966 DOI: 10.1111/jnc.16093] [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: 09/03/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/18/2024]
Abstract
Dysregulation of synaptic glutamate levels can lead to excitotoxicity such as that observed in stroke, traumatic brain injury, and epilepsy. The role of increased intracellular calcium (Ca2+) in the development of excitotoxicity is well established. However, less is known regarding the impact of glutamate on endoplasmic reticulum (ER)-Ca2+-mediated processes such as proteostasis. To investigate this, we expressed a secreted ER Ca2+ modulated protein (SERCaMP) in primary cortical neurons to monitor exodosis, a phenomenon whereby ER calcium depletion causes the secretion of ER-resident proteins that perform essential functions to the ER and the cell. Activation of glutamatergic receptors (GluRs) led to an increase in SERCaMP secretion indicating that normally ER-resident proteins are being secreted in a manner consistent with ER Ca2+ depletion. Antagonism of ER Ca2+ channels attenuated the effects of glutamate and GluR agonists on SERCaMP release. We also demonstrate that endogenous proteins containing an ER retention/retrieval sequence (ERS) are secreted in response to GluR activation supporting that neuronal activation by glutamate promotes ER exodosis. Ectopic expression of KDEL receptors attenuated the secretion of ERS-containing proteins caused by GluR agonists. Taken together, our data indicate that excessive GluR activation causes disruption of neuronal proteostasis by triggering the secretion of ER-resident proteins through ER Ca2+ depletion and describes a new facet of excitotoxicity.
Collapse
Affiliation(s)
- Amanda M. Dossat
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Kathleen A. Trychta
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Elliot J. Glotfelty
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Joshua J. Hinkle
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Lowella V. Fortuno
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Lana N. Gore
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Christopher T. Richie
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Brandon K. Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| |
Collapse
|
15
|
Chen P, Huang X, Li W, Wen W, Cao Y, Li J, Huang Y, Hu Y. Myeloid-derived growth factor in diseases: structure, function and mechanisms. Mol Med 2024; 30:103. [PMID: 39030488 PMCID: PMC11264862 DOI: 10.1186/s10020-024-00874-z] [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: 05/02/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Myeloid-derived growth factor (MYDGF) is a novel secreted protein with potent antiapoptotic and tissue-repairing properties that is present in nearly 140 human tissues and cell lines, with the highest abundance in the oral epithelium and skin. Initially, MYDGF was found in bone marrow-derived monocytes and macrophages for cardioprotection and repair after myocardial infarction. Subsequent studies have shown that MYDGF plays an important role in other cardiovascular diseases (e.g., atherosclerosis and heart failure), metabolic disorders, renal disease, autoimmune/inflammatory disorders, and cancers. Although the underlying mechanisms have not been fully explored, the role of MYDGF in health and disease may involve cell apoptosis and proliferation, tissue repair and regeneration, anti-inflammation, and glycolipid metabolism regulation. In this review, we summarize the current progress in understanding the role of MYDGF in health and disease, focusing on its structure, function and mechanisms. The graphical abstract shows the current role of MYDGF in different organs and diseases (Fig. 1).
Collapse
Affiliation(s)
- Peng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Weiwen Li
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Yue Cao
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Jiahuan Li
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, NSW2006, Australia.
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation Research, Guangzhou, 510000, China.
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
| |
Collapse
|
16
|
Wu X, Zhang G, Du X. Cigarette Smoke Extract Induces MUC5AC Expression Through the ROS/ IP3R/Ca 2+ Pathway in Calu-3 Cells. Int J Chron Obstruct Pulmon Dis 2024; 19:1635-1647. [PMID: 39045541 PMCID: PMC11264152 DOI: 10.2147/copd.s469866] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/29/2024] [Indexed: 07/25/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is caused by exposure to noxious external particles, air pollution, and the inhalation of cigarette smoke. Airway mucus hypersecretion particularly mucin5AC (MUC5AC), is a crucial pathological feature of COPD and is associated with its initiation and progression. In this study, we aimed to investigate the effects of cigarette smoke extract (CSE) on MUC5AC expression, particularly the mechanisms by which reactive oxygen species (ROS) induce MUC5AC expression. Methods The effects of CSE on the expression of MUC5AC and mucin5B (MUC5B) were investigated in vitro in Calu-3 cells. MUC5AC and MUC5B expression levels were measured using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), immunofluorescence staining, and enzyme-linked immunosorbent assay (ELISA). Total cellular levels of ROS and Ca2+ were determined using DCFH-DA and Fluo-4 AM. Subsequently, the expression levels of IP3R, IRE1α, p-IRE1α and XBP1s were measured by Western blotting. Gene silencing was achieved by using small-interfering RNAs. Results Our findings revealed that exposure to CSE increased MUC5AC levels and upregulated ROS, IP3R/Ca2+ and unfolded protein response (UPR)-associated factors. In addition, knockdown of IP3R using siRNA decreased CSE-induced Ca2+ production, UPR-associated factors, and MUC5AC expression. Furthermore, 10 mM N-acetyl-l-cysteine (NAC) treatment suppressed the effects of CSE, including ROS generation, IP3R/ Ca2+, UPR activation, and MUC5AC overexpression. Conclusion Our results suggest that ROS regulates CSE-induced UPR and MUC5AC overexpression through IP3R/ Ca2+ signaling. Additionally, we identified NAC as a promising therapeutic agent for mitigating CSE-induced MUC5AC overexpression.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Guoyue Zhang
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Xianzhi Du
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| |
Collapse
|
17
|
Kawanaka R, Jin H, Aoe T. Unraveling the Connection: Pain and Endoplasmic Reticulum Stress. Int J Mol Sci 2024; 25:4995. [PMID: 38732214 PMCID: PMC11084550 DOI: 10.3390/ijms25094995] [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: 04/03/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Pain is a complex and multifaceted experience. Recent research has increasingly focused on the role of endoplasmic reticulum (ER) stress in the induction and modulation of pain. The ER is an essential organelle for cells and plays a key role in protein folding and calcium dynamics. Various pathological conditions, such as ischemia, hypoxia, toxic substances, and increased protein production, may disturb protein folding, causing an increase in misfolding proteins in the ER. Such an overload of the folding process leads to ER stress and causes the unfolded protein response (UPR), which increases folding capacity in the ER. Uncompensated ER stress impairs intracellular signaling and cell function, resulting in various diseases, such as diabetes and degenerative neurological diseases. ER stress may be a critical universal mechanism underlying human diseases. Pain sensations involve the central as well as peripheral nervous systems. Several preclinical studies indicate that ER stress in the nervous system is enhanced in various painful states, especially in neuropathic pain conditions. The purpose of this narrative review is to uncover the intricate relationship between ER stress and pain, exploring molecular pathways, implications for various pain conditions, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Ryoko Kawanaka
- Department of Anesthesiology, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
| |
Collapse
|
18
|
Xu J, Song Y, Ding S, Duan W, Xiang G, Wang Z. Myeloid-derived growth factor and its effects on cardiovascular and metabolic diseases. Cytokine Growth Factor Rev 2024; 76:77-85. [PMID: 38185568 DOI: 10.1016/j.cytogfr.2023.12.005] [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: 12/04/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
Myeloid-derived growth factor (MYDGF) is a paracrine protein produced by bone marrow-derived monocytes and macrophages. Current research shows that it has protective effects on the cardiovascular system, such as repairing heart tissue after myocardial infarction, enhancing cardiomyocyte proliferation, improving cardiac regeneration after myocardial injury, regulating proliferation and survival of endothelial cells, reducing endothelial cell damage, resisting pressure overload-induced heart failure, as well as protecting against atherosclerosis. Furthermore, regarding the metabolic diseases, MYDGF has effects of improving type 2 diabetes mellitus, relieving non-alcoholic fatty liver disease, alleviating glomerular diseases, and resisting osteoporosis. Herein, we will discuss the biology of MYDGF and its effects on cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Jinling Xu
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Yanzhuo Song
- Nanchang University, Nanchang, Jiangxi 330031, China
| | - Sheng Ding
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Weizhe Duan
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China
| | - Guangda Xiang
- Department of Endocrinology, General Hospital of Central Theater Command, Wuluo Road 627, Wuhan, Hubei 430070, China.
| | - Zhongjing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, China.
| |
Collapse
|
19
|
Denecke J. Reply to: Does the KDEL receptor cycle between the Golgi and the ER? Nat Commun 2024; 15:2454. [PMID: 38509079 PMCID: PMC10954661 DOI: 10.1038/s41467-024-45850-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024] Open
Affiliation(s)
- Jurgen Denecke
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
20
|
Aniento F, Robinson DG. Does the KDEL receptor cycle between the Golgi and the ER? Nat Commun 2024; 15:2455. [PMID: 38509061 PMCID: PMC10954686 DOI: 10.1038/s41467-024-45849-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024] Open
Affiliation(s)
- Fernando Aniento
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universitat de València, 46100, Burjassot, Spain
| | - David G Robinson
- Centre for Organismal Studies, Univ. Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
21
|
Liu S, He K, Yang L, Xu F, Cui X, Qu L, Li X, Ren B. Endoplasmic reticulum stress regulators exhibit different prognostic, therapeutic and immune landscapes in pancreatic adenocarcinoma. J Cell Mol Med 2024; 28:e18092. [PMID: 38303549 PMCID: PMC10902308 DOI: 10.1111/jcmm.18092] [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: 05/31/2023] [Revised: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 02/03/2024] Open
Abstract
Endoplasmic reticulum stress (ERS) and unfolded protein response are the critical processes of tumour biology. However, the roles of ERS regulatory genes in pancreatic adenocarcinoma (PAAD) remain elusive. A novel ERS-related risk signature was constructed using the Lasso regression analysis. Its prognostic value, immune effect, metabolic influence, mutational feature and therapeutic correlation were comprehensively analysed through multiple bioinformatic approaches. The biofunctions of KDELR3 and YWHAZ in pancreatic cancer (PC) cells were also investigated through colony formation, Transwell assays, flow cytometric detection and a xenograft model. The upstream miRNA regulatory mechanism of KDELR3 was predicted and validated. ERS risk score was identified as an independent prognostic factor and could improve traditional prognostic model. Meanwhile, it was closely associated with metabolic reprogramming and tumour immune. High ERS risk enhanced glycolysis process and nucleotide metabolism, but was unfavourable for anti-tumour immune response. Moreover, ERS risk score could act as a potential biomarker for predicting the efficacy of ICBs. Overexpression of KDELR3 and YWHAZ stimulated the proliferation, migration and invasion of SW1990 and BxPC-3 cells. Silencing KDELR3 suppressed tumour growth in a xenograft model. miR-137 could weaken the malignant potentials of PC cells through inhibiting KDELR3 (5'-AGCAAUAA-3'). ERS risk score greatly contributed to PAAD clinical assessment. KDELR3 and YWHAZ possessed cancer-promoting capacities, showing promise as a novel treatment target.
Collapse
Affiliation(s)
- Shanshan Liu
- Department of Rheumatology and ImmunologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Kaini He
- Department of GastroenterologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Longbao Yang
- Department of GastroenterologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Fangshi Xu
- Department of MedicineXi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xiaoguang Cui
- Department of Rheumatology and ImmunologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Li Qu
- Department of Rheumatology and ImmunologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Xueyi Li
- Department of Rheumatology and ImmunologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Bin‐cheng Ren
- Department of Rheumatology and ImmunologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| |
Collapse
|
22
|
Zoine JT, Immadisetty K, Ibanez-Vega J, Moore SE, Nevitt C, Thanekar U, Tian L, Karouni A, Chockley PJ, Arthur B, Sheppard H, Klco JM, Langfitt DM, Krenciute G, Gottschalk S, Babu MM, Velasquez MP. Peptide-scFv antigen recognition domains effectively confer CAR T cell multiantigen specificity. Cell Rep Med 2024; 5:101422. [PMID: 38350450 PMCID: PMC10897625 DOI: 10.1016/j.xcrm.2024.101422] [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: 04/13/2023] [Revised: 10/06/2023] [Accepted: 01/19/2024] [Indexed: 02/15/2024]
Abstract
The emergence of immune escape is a significant roadblock to developing effective chimeric antigen receptor (CAR) T cell therapies against hematological malignancies, including acute myeloid leukemia (AML). Here, we demonstrate feasibility of targeting two antigens simultaneously by combining a GRP78-specific peptide antigen recognition domain with a CD123-specific scFv to generate a peptide-scFv bispecific antigen recognition domain (78.123). To achieve this, we test linkers with varying length and flexibility and perform immunophenotypic and functional characterization. We demonstrate that bispecific CAR T cells successfully recognize and kill tumor cells that express GRP78, CD123, or both antigens and have improved antitumor activity compared to their monospecific counterparts when both antigens are expressed. Protein structure prediction suggests that linker length and compactness influence the functionality of the generated bispecific CARs. Thus, we present a bispecific CAR design strategy to prevent immune escape in AML that can be extended to other peptide-scFv combinations.
Collapse
Affiliation(s)
- Jaquelyn T Zoine
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kalyan Immadisetty
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jorge Ibanez-Vega
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sarah E Moore
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Chris Nevitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Unmesha Thanekar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Liqing Tian
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Karouni
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter J Chockley
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bright Arthur
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Deanna M Langfitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Madan Babu
- Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
23
|
Jia J, Zhu L, Yue X, Tang S, Jing S, Tan C, Du Y, Gao J, Lee I, Qian Y. Crosstalk between KDEL receptor and EGF receptor mediates cell proliferation and migration via STAT3 signaling. Cell Commun Signal 2024; 22:140. [PMID: 38378560 PMCID: PMC10880305 DOI: 10.1186/s12964-024-01517-w] [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: 09/05/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Hostile microenvironment of cancer cells provoke a stressful condition for endoplasmic reticulum (ER) and stimulate the expression and secretion of ER chaperones, leading to tumorigenic effects. However, the molecular mechanism underlying these effects is largely unknown. In this study, we reveal that the last four residues of ER chaperones, which are recognized by KDEL receptor (KDELR), is required for cell proliferation and migration induced by secreted chaperones. By combining proximity-based mass spectrometry analysis, split venus imaging and membrane yeast two hybrid assay, we present that EGF receptor (EGFR) may be a co-receptor for KDELR on the surface. Prior to ligand addition, KDELR spontaneously oligomerizes and constantly undergoes recycling near the plasma membrane. Upon KDEL ligand binding, the interactions of KDELR with itself and with EGFR increase rapidly, leading to augmented internalization of KDELR and tyrosine phosphorylation in the C-terminus of EGFR. STAT3, which binds the phosphorylated tyrosine motif on EGFR, is subsequently activated by EGFR and mediates cell growth and migration. Taken together, our results suggest that KDELR serves as a bona fide cell surface receptor for secreted ER chaperones and transactivates EGFR-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
- Present address: Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| |
Collapse
|
24
|
Wu X, Zheng L, Reboll MR, Hyde LF, Mass E, Niessen HW, Kosanke M, Pich A, Giannitsis E, Tillmanns J, Bauersachs J, Heineke J, Wang Y, Korf-Klingebiel M, Polten F, Wollert KC. Cysteine-rich with EGF-like domains 2 (CRELD2) is an endoplasmic reticulum stress-inducible angiogenic growth factor promoting ischemic heart repair. NATURE CARDIOVASCULAR RESEARCH 2024; 3:186-202. [PMID: 39196188 PMCID: PMC11358006 DOI: 10.1038/s44161-023-00411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/07/2023] [Indexed: 08/29/2024]
Abstract
Tissue repair after myocardial infarction (MI) is guided by autocrine and paracrine-acting proteins. Deciphering these signals and their upstream triggers is essential when considering infarct healing as a therapeutic target. Here we perform a bioinformatic secretome analysis in mouse cardiac endothelial cells and identify cysteine-rich with EGF-like domains 2 (CRELD2), an endoplasmic reticulum stress-inducible protein with poorly characterized function. CRELD2 was abundantly expressed and secreted in the heart after MI in mice and patients. Creld2-deficient mice and wild-type mice treated with a CRELD2-neutralizing antibody showed impaired de novo microvessel formation in the infarct border zone and developed severe postinfarction heart failure. CRELD2 protein therapy, conversely, improved heart function after MI. Exposing human coronary artery endothelial cells to recombinant CRELD2 induced angiogenesis, associated with a distinct phosphoproteome signature. These findings identify CRELD2 as an angiogenic growth factor and unravel a link between endoplasmic reticulum stress and ischemic tissue repair.
Collapse
Affiliation(s)
- Xuekun Wu
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Stanford University School of Medicine, Stanford, CA, USA
| | - Linqun Zheng
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiology, Shanghai General Hospital, Shanghai, China
| | - Marc R Reboll
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Lillian F Hyde
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Hans W Niessen
- Department of Pathology and Department of Cardiac Surgery, Institute for Cardiovascular Research, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Andreas Pich
- Core Unit Proteomics and Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | | | - Jochen Tillmanns
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Joerg Heineke
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Physiology, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yong Wang
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Mortimer Korf-Klingebiel
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Felix Polten
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Kai C Wollert
- Division of Molecular and Translational Cardiology, Hans Borst Center for Heart and Stem Cell Research, Hannover Medical School, Hannover, Germany.
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
25
|
Grossmann D, Malburg N, Glaß H, Weeren V, Sondermann V, Pfeiffer JF, Petters J, Lukas J, Seibler P, Klein C, Grünewald A, Hermann A. Mitochondria-Endoplasmic Reticulum Contact Sites Dynamics and Calcium Homeostasis Are Differentially Disrupted in PINK1-PD or PRKN-PD Neurons. Mov Disord 2023; 38:1822-1836. [PMID: 37449534 DOI: 10.1002/mds.29525] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND It is generally believed that the pathogenesis of PINK1/parkin-related Parkinson's disease (PD) is due to a disturbance in mitochondrial quality control. However, recent studies have found that PINK1 and Parkin play a significant role in mitochondrial calcium homeostasis and are involved in the regulation of mitochondria-endoplasmic reticulum contact sites (MERCSs). OBJECTIVE The aim of our study was to perform an in-depth analysis of the role of MERCSs and impaired calcium homeostasis in PINK1/Parkin-linked PD. METHODS In our study, we used induced pluripotent stem cell-derived dopaminergic neurons from patients with PD with loss-of-function mutations in PINK1 or PRKN. We employed a split-GFP-based contact site sensor in combination with the calcium-sensitive dye Rhod-2 AM and applied Airyscan live-cell super-resolution microscopy to determine how MERCSs are involved in the regulation of mitochondrial calcium homeostasis. RESULTS Our results showed that thapsigargin-induced calcium stress leads to an increase of the abundance of narrow MERCSs in wild-type neurons. Intriguingly, calcium levels at the MERCSs remained stable, whereas the increased net calcium influx resulted in elevated mitochondrial calcium levels. However, PINK1-PD or PRKN-PD neurons showed an increased abundance of MERCSs at baseline, accompanied by an inability to further increase MERCSs upon thapsigargin-induced calcium stress. Consequently, calcium distribution at MERCSs and within mitochondria was disrupted. CONCLUSIONS Our results demonstrated how the endoplasmic reticulum and mitochondria work together to cope with calcium stress in wild-type neurons. In addition, our results suggests that PRKN deficiency affects the dynamics and composition of MERCSs differently from PINK1 deficiency, resulting in differentially affected calcium homeostasis. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Dajana Grossmann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Nina Malburg
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Hannes Glaß
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Veronika Weeren
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Verena Sondermann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Julia F Pfeiffer
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Janine Petters
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Jan Lukas
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University Medical Center Rostock, University of Rostock, Rostock, Germany
| | - Philip Seibler
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Anne Grünewald
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht Kossel," Department of Neurology, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University Medical Center Rostock, University of Rostock, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Rostock/Greifswald, Rostock, Germany
| |
Collapse
|
26
|
Parkkinen I, Their A, Asghar MY, Sree S, Jokitalo E, Airavaara M. Pharmacological Regulation of Endoplasmic Reticulum Structure and Calcium Dynamics: Importance for Neurodegenerative Diseases. Pharmacol Rev 2023; 75:959-978. [PMID: 37127349 DOI: 10.1124/pharmrev.122.000701] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell, composed of a continuous network of sheets and tubules, and is involved in protein, calcium (Ca2+), and lipid homeostasis. In neurons, the ER extends throughout the cell, both somal and axodendritic compartments, and is highly important for neuronal functions. A third of the proteome of a cell, secreted and membrane-bound proteins, are processed within the ER lumen and most of these proteins are vital for neuronal activity. The brain itself is high in lipid content, and many structural lipids are produced, in part, by the ER. Cholesterol and steroid synthesis are strictly regulated in the ER of the blood-brain barrier protected brain cells. The high Ca2+ level in the ER lumen and low cytosolic concentration is needed for Ca2+-based intracellular signaling, for synaptic signaling and Ca2+ waves, and for preparing proteins for correct folding in the presence of high Ca2+ concentrations to cope with the high concentrations of extracellular milieu. Particularly, ER Ca2+ is controlled in axodendritic areas for proper neurito- and synaptogenesis and synaptic plasticity and remodeling. In this review, we cover the physiologic functions of the neuronal ER and discuss it in context of common neurodegenerative diseases, focusing on pharmacological regulation of ER Ca2+ Furthermore, we postulate that heterogeneity of the ER, its protein folding capacity, and ensuring Ca2+ regulation are crucial factors for the aging and selective vulnerability of neurons in various neurodegenerative diseases. SIGNIFICANCE STATEMENT: Endoplasmic reticulum (ER) Ca2+ regulators are promising therapeutic targets for degenerative diseases for which efficacious drug therapies do not exist. The use of pharmacological probes targeting maintenance and restoration of ER Ca2+ can provide restoration of protein homeostasis (e.g., folding of complex plasma membrane signaling receptors) and slow down the degeneration process of neurons.
Collapse
Affiliation(s)
- Ilmari Parkkinen
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Anna Their
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Muhammad Yasir Asghar
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Sreesha Sree
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Zhang C, Xiao J, Fa L, Jiang F, Jiang H, Zhou L, Xu Z. Identification of co-expressed gene networks promoting CD8 + T cell infiltration and having prognostic value in uveal melanoma. BMC Ophthalmol 2023; 23:354. [PMID: 37563735 PMCID: PMC10416479 DOI: 10.1186/s12886-023-03098-7] [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: 12/05/2022] [Accepted: 08/01/2023] [Indexed: 08/12/2023] Open
Abstract
Current immunotherapies are unsatisfactory against uveal melanoma (UM); however, elevated CD8+ T cell infiltration level indicates poor prognosis in UM. Here, we aimed to identify co-expressed gene networks promoting CD8+ T cell infiltration in UM and created a prognostic hazard model based on the identified hub genes. Raw data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Stromal-immune comprehensive score (ESTIMATE) was used to evaluate the immune-infiltration landscape of the tumor microenvironment. Single-Sample Gene Set Enrichment Analysis (ssGSEA) and Weighted Correlation Network Analysis (WGCNA) were used to quantify CD8+ T cell infiltration level and identify hub genes. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to analyze the biological processes. Least absolute shrinkage and selection operator (LASSO) Cox regression were used to establish a prognostic model, which was further validated. Finally, pan-cancer analysis evaluated these genes to be associated with CD8+ T cell infiltration in other tumors. In conclusion, the proposed four-gene (PTPN12, IDH2, P2RX4, and KDELR2) prognostic hazard model had satisfactory prognostic ability. These hub genes may promote CD8+ T cell infiltration in UM through antigen presentation, and CD8+ T cell possibly function as Treg, resulting in poor prognosis. These findings might facilitate the development of novel immunotherapies.
Collapse
Affiliation(s)
- Chun Zhang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Jing Xiao
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Luzhong Fa
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Fanwen Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Hui Jiang
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Lin Zhou
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China
| | - Zhuping Xu
- Department of ophthalmology, West China Hospital, Sichuan University, Sichuan Province, 610041, Chengdu, China.
| |
Collapse
|
28
|
Verjan Garcia N, Hong KU, Matoba N. The Unfolded Protein Response and Its Implications for Novel Therapeutic Strategies in Inflammatory Bowel Disease. Biomedicines 2023; 11:2066. [PMID: 37509705 PMCID: PMC10377089 DOI: 10.3390/biomedicines11072066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The endoplasmic reticulum (ER) is a multifunctional organelle playing a vital role in maintaining cell homeostasis, and disruptions to its functions can have detrimental effects on cells. Dysregulated ER stress and the unfolded protein response (UPR) have been linked to various human diseases. For example, ER stress and the activation of the UPR signaling pathways in intestinal epithelial cells can either exacerbate or alleviate the severity of inflammatory bowel disease (IBD), contingent on the degree and conditions of activation. Our recent studies have shown that EPICERTIN, a recombinant variant of the cholera toxin B subunit containing an ER retention motif, can induce a protective UPR in colon epithelial cells, subsequently promoting epithelial restitution and mucosal healing in IBD models. These findings support the idea that compounds modulating UPR may be promising pharmaceutical candidates for the treatment of the disease. In this review, we summarize our current understanding of the ER stress and UPR in IBD, focusing on their roles in maintaining cell homeostasis, dysregulation, and disease pathogenesis. Additionally, we discuss therapeutic strategies that promote the cytoprotection of colon epithelial cells and reduce inflammation via pharmacological manipulation of the UPR.
Collapse
Affiliation(s)
- Noel Verjan Garcia
- UofL Health-Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kyung U Hong
- UofL Health-Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Nobuyuki Matoba
- UofL Health-Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
29
|
Svarcbahs R, Blossom SM, Baffoe-Bonnie HS, Trychta KA, Greer LK, Pickel J, Henderson MJ, Harvey BK. A transgenic mouse line for assaying tissue-specific changes in endoplasmic reticulum proteostasis. Transgenic Res 2023; 32:209-221. [PMID: 37133648 PMCID: PMC10195735 DOI: 10.1007/s11248-023-00349-7] [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: 12/19/2022] [Accepted: 04/17/2023] [Indexed: 05/04/2023]
Abstract
Maintenance of calcium homeostasis is important for proper endoplasmic reticulum (ER) function. When cellular stress conditions deplete the high concentration of calcium in the ER, ER-resident proteins are secreted into the extracellular space in a process called exodosis. Monitoring exodosis provides insight into changes in ER homeostasis and proteostasis resulting from cellular stress associated with ER calcium dysregulation. To monitor cell-type specific exodosis in the intact animal, we created a transgenic mouse line with a Gaussia luciferase (GLuc)-based, secreted ER calcium-modulated protein, SERCaMP, preceded by a LoxP-STOP-LoxP (LSL) sequence. The Cre-dependent LSL-SERCaMP mice were crossed with albumin (Alb)-Cre and dopamine transporter (DAT)-Cre mouse lines. GLuc-SERCaMP expression was characterized in mouse organs and extracellular fluids, and the secretion of GLuc-SERCaMP in response to cellular stress was monitored following pharmacological depletion of ER calcium. In LSL-SERCaMP × Alb-Cre mice, robust GLuc activity was observed only in the liver and blood, whereas in LSL-SERCaMP × DAT-Cre mice, GLuc activity was seen in midbrain dopaminergic neurons and tissue samples innervated by dopaminergic projections. After calcium depletion, we saw increased GLuc signal in the plasma and cerebrospinal fluid collected from the Alb-Cre and DAT-Cre crosses, respectively. This mouse model can be used to investigate the secretion of ER-resident proteins from specific cell and tissue types during disease pathogenesis and may aid in the identification of therapeutics and biomarkers of disease.
Collapse
Affiliation(s)
- Reinis Svarcbahs
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sarah M Blossom
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Helena S Baffoe-Bonnie
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kathleen A Trychta
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lacey K Greer
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - James Pickel
- Transgenic Technology Core, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Mark J Henderson
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Brandon K Harvey
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
30
|
Wu KJ, Hung TW, Wang YS, Chen YH, Bae EK, Yu SJ. Prosaposin PS18 reduces dopaminergic neurodegeneration in a 6-hydroxydopamine rat model of Parkinson's disease. Sci Rep 2023; 13:8148. [PMID: 37208379 DOI: 10.1038/s41598-023-35274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
Saposin and its precursor prosaposin are endogenous proteins with neurotrophic and anti-apoptotic properties. Prosaposin or its analog prosaposin-derived 18-mer peptide (PS18) reduced neuronal damage in hippocampus and apoptosis in stroke brain. Its role in Parkinson's disease (PD) has not been well characterized. This study aimed to examine the physiological role of PS18 in 6-hydroxydopamine (6-OHDA) cellular and animal models of PD. We found that PS18 significantly antagonized 6-OHDA -mediated dopaminergic neuronal loss and TUNEL in rat primary dopaminergic neuronal culture. In SH-SY5Y cells overexpressing the secreted ER calcium-monitoring proteins, we found that PS18 significantly reduced thapsigargin and 6-OHDA-mediated ER stress. The expression of prosaposin and the protective effect of PS18 were next examined in hemiparkinsonian rats. 6-OHDA was unilaterally administered to striatum. The expression of prosaposin was transiently upregulated in striatum on D3 (day 3) after lesioning and returned below the basal level on D29. The 6-OHDA-lesioned rats developed bradykinesia and an increase in methamphetamine-mediated rotation, which was antagonized by PS18. Brain tissues were collected for Western blot, immunohistochemistry, and qRTPCR analysis. Tyrosine hydroxylase immunoreactivity was significantly reduced while the expressions of PERK, ATF6, CHOP, and BiP were upregulated in the lesioned nigra; these responses were significantly antagonized by PS18. Taken together, our data support that PS18 is neuroprotective in cellular and animal models of PD. The mechanisms of protection may involve anti-ER stress.
Collapse
Affiliation(s)
- Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yun-Hsiang Chen
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan.
| |
Collapse
|
31
|
Scheid JF, Eraslan B, Hudak A, Brown EM, Sergio D, Delorey TM, Phillips D, Lefkovith A, Jess AT, Duck LW, Elson CO, Vlamakis H, Plichta DR, Deguine J, Ananthakrishnan AN, Graham DB, Regev A, Xavier RJ. Remodeling of colon plasma cell repertoire within ulcerative colitis patients. J Exp Med 2023; 220:e20220538. [PMID: 36752797 PMCID: PMC9949229 DOI: 10.1084/jem.20220538] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 10/03/2022] [Accepted: 01/11/2023] [Indexed: 02/09/2023] Open
Abstract
Plasma cells (PCs) constitute a significant fraction of colonic mucosal cells and contribute to inflammatory infiltrates in ulcerative colitis (UC). While gut PCs secrete bacteria-targeting IgA antibodies, their role in UC pathogenesis is unknown. We performed single-cell V(D)J- and RNA-seq on sorted B cells from the colon of healthy individuals and patients with UC. A large fraction of B cell clones is shared between different colon regions, but inflammation in UC broadly disrupts this landscape, causing transcriptomic changes characterized by an increase in the unfolded protein response (UPR) and antigen presentation genes, clonal expansion, and isotype skewing from IgA1 and IgA2 to IgG1. We also directly expressed and assessed the specificity of 152 mAbs from expanded PC clones. These mAbs show low polyreactivity and autoreactivity and instead target both shared bacterial antigens and specific bacterial strains. Altogether, our results characterize the microbiome-specific colon PC response and how its disruption might contribute to inflammation in UC.
Collapse
Affiliation(s)
- Johannes F. Scheid
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Basak Eraslan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew Hudak
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric M. Brown
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dallis Sergio
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni M. Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Alison T. Jess
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lennard W. Duck
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles O. Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Ashwin N. Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Daniel B. Graham
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
32
|
RNAseq Analysis of FABP4 Knockout Mouse Hippocampal Transcriptome Suggests a Role for WNT/β-Catenin in Preventing Obesity-Induced Cognitive Impairment. Int J Mol Sci 2023; 24:ijms24043381. [PMID: 36834799 PMCID: PMC9961923 DOI: 10.3390/ijms24043381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Microglial fatty-acid binding protein 4 (FABP4) is a regulator of neuroinflammation. We hypothesized that the link between lipid metabolism and inflammation indicates a role for FABP4 in regulating high fat diet (HFD)-induced cognitive decline. We have previously shown that obese FABP4 knockout mice exhibit decreased neuroinflammation and cognitive decline. FABP4 knockout and wild type mice were fed 60% HFD for 12 weeks starting at 15 weeks old. Hippocampal tissue was dissected and RNA-seq was performed to measure differentially expressed transcripts. Reactome molecular pathway analysis was utilized to examine differentially expressed pathways. Results showed that HFD-fed FABP4 knockout mice have a hippocampal transcriptome consistent with neuroprotection, including associations with decreased proinflammatory signaling, ER stress, apoptosis, and cognitive decline. This is accompanied by an increase in transcripts upregulating neurogenesis, synaptic plasticity, long-term potentiation, and spatial working memory. Pathway analysis revealed that mice lacking FABP4 had changes in metabolic function that support reduction in oxidative stress and inflammation, and improved energy homeostasis and cognitive function. Analysis suggested a role for WNT/β-Catenin signaling in the protection against insulin resistance, alleviating neuroinflammation and cognitive decline. Collectively, our work shows that FABP4 represents a potential target in alleviating HFD-induced neuroinflammation and cognitive decline and suggests a role for WNT/β-Catenin in this protection.
Collapse
|
33
|
Song N, Song Y, Hu B, Liu X, Yu X, Zhou H, Long J, Yu Z. Persistent Endoplasmic Reticulum Stress Stimulated by Peptide Assemblies for Sensitizing Cancer Chemotherapy. Adv Healthc Mater 2023; 12:e2202039. [PMID: 36353887 DOI: 10.1002/adhm.202202039] [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: 08/12/2022] [Revised: 10/31/2022] [Indexed: 11/11/2022]
Abstract
Pharmacological targeting of endoplasmic reticulum (ER) stress represents one of important methods for disease therapy, which, however, is significantly suppressed by the ER homeostatic processe. Herein, a proof-of-concept strategy is reported for persistent stimulation of ER stress via preventing ER stress adaptation by utilizing multifunctional peptide assemblies. The strategy is established via creation of peptide assemblies with ER-targeting and chaperone glucose-regulated protein 78 (GRP78)-inhibiting functions. The peptides assemblies form well-defined nanofibers that are retrieved by ER organelles in human cervical cancer cell. The underlying mechanism studies unravel that the ER-accumulated peptide assemblies simultaneously stimulate ER stress and inhibit GRP78 refolding activity and thereby promoting endogenous protein aggregation. Combining the internalized peptide assemblies with the induced protein aggregates leads to the persistent stimulation of ER stress. The persistent ER stress induced by the peptide assemblies bestows their application in sensitizing cancer chemotherapy. Both in vitro and in vivo results confirm the enhanced cytotoxicity of drug toyocamycin against HeLa cells by peptide assemblies, thus efficiently inhibiting in vivo tumor growth. The strategy reported here discloses the fundamental keys for efficient promotion of ER stress, thus providing the guidance for development of ER-targeting-assisted cancer chemotherapy in the future.
Collapse
Affiliation(s)
- Na Song
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yanqiu Song
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Binbin Hu
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xin Liu
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xiunan Yu
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Hao Zhou
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiafu Long
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Zhilin Yu
- Ministry of Education Key Laboratory of Functional Polymer Materials, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry, College of Chemistry, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| |
Collapse
|
34
|
E96V Mutation in the Kdelr3 Gene Is Associated with Type 2 Diabetes Susceptibility in Obese NZO Mice. Int J Mol Sci 2023; 24:ijms24010845. [PMID: 36614300 PMCID: PMC9820861 DOI: 10.3390/ijms24010845] [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: 11/29/2022] [Revised: 12/16/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Type 2 diabetes (T2D) represents a multifactorial metabolic disease with a strong genetic predisposition. Despite elaborate efforts in identifying the genetic variants determining individual susceptibility towards T2D, the majority of genetic factors driving disease development remain poorly understood. With the aim to identify novel T2D risk genes we previously generated an N2 outcross population using the two inbred mouse strains New Zealand obese (NZO) and C3HeB/FeJ (C3H). A linkage study performed in this population led to the identification of the novel T2D-associated quantitative trait locus (QTL) Nbg15 (NZO blood glucose on chromosome 15, Logarithm of odds (LOD) 6.6). In this study we used a combined approach of positional cloning, gene expression analyses and in silico predictions of DNA polymorphism on gene/protein function to dissect the genetic variants linking Nbg15 to the development of T2D. Moreover, we have generated congenic strains that associated the distal sublocus of Nbg15 to mechanisms altering pancreatic beta cell function. In this sublocus, Cbx6, Fam135b and Kdelr3 were nominated as potential causative genes associated with the Nbg15 driven effects. Moreover, a putative mutation in the Kdelr3 gene from NZO was identified, negatively influencing adaptive responses associated with pancreatic beta cell death and induction of endoplasmic reticulum stress. Importantly, knockdown of Kdelr3 in cultured Min6 beta cells altered insulin granules maturation and pro-insulin levels, pointing towards a crucial role of this gene in islets function and T2D susceptibility.
Collapse
|
35
|
Endoplasmic Reticulum Stress-Regulated Chaperones as a Serum Biomarker Panel for Parkinson's Disease. Mol Neurobiol 2023; 60:1476-1485. [PMID: 36478320 PMCID: PMC9899193 DOI: 10.1007/s12035-022-03139-0] [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: 08/03/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022]
Abstract
Examination of post-mortem brain tissues has previously revealed a strong association between Parkinson's disease (PD) pathophysiology and endoplasmic reticulum (ER) stress. Evidence in the literature regarding the circulation of ER stress-regulated factors released from neurons provides a rationale for investigating ER stress biomarkers in the blood to aid diagnosis of PD. The levels of ER stress-regulated proteins in serum collected from 29 PD patients and 24 non-PD controls were measured using enzyme-linked immunosorbent assays. A panel of four biomarkers, protein disulfide-isomerase A1, protein disulfide-isomerase A3, mesencephalic astrocyte-derived neurotrophic factor, and clusterin, together with age and gender had higher ability (area under the curve 0.64, sensitivity 66%, specificity 57%) and net benefit to discriminate PD patients from the non-PD group compared with other analyzed models. Addition of oligomeric and total α-synuclein to the model did not improve the diagnostic power of the biomarker panel. We provide evidence that ER stress-regulated proteins merit further investigation for their potential as diagnostic biomarkers of PD.
Collapse
|
36
|
Chen S, Hao X, Chen G, Liu G, Yuan X, Shen P, Guo D. Effects of mesencephalic astrocyte-derived neurotrophic factor on sepsis-associated acute kidney injury. World J Emerg Med 2023; 14:386-392. [PMID: 37908790 PMCID: PMC10613790 DOI: 10.5847/wjem.j.1920-8642.2023.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/20/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND To determine the protective role of mesencephalic astrocyte-derived neurotrophic factor (MANF) in regulating sepsis-associated acute kidney injury (S-AKI). METHODS A total of 96 mice were randomly divided into the control group, control+MANF group, S-AKI group, and S-AKI+MANF group. The S-AKI model was established by injecting lipopolysaccharide (LPS) at 10 mg/kg intraperitoneally. MANF (200 μg/kg) was administered to the control+MANF and S-AKI+MANF groups. An equal dose of normal saline was administered daily intraperitoneally in the control and S-AKI groups. Serum and kidney tissue samples were obtained for biochemical analysis. Western blotting was used to detect the protein expression of MANF in the kidney, and enzyme-linked immunosorbent assay (ELISA) was used to determine expression of MANF in the serum, pro-inflammatory cytokines (tumor necrosis factor-α [TNF-α] and interleukin-6 [IL-6]). Serum creatinine (SCr), and blood urea nitrogen (BUN) were examined using an automatic biochemical analyzer. In addition, the kidney tissue was observed for pathological changes by hematoxylin-eosin staining. The comparison between two groups was performed by unpaired Student's t-test, and statistics among multiple groups were carried out using Tukey's post hoc test following one-way analysis of variance (ANOVA). A P-value <0.05 was considered statistically significant. RESULTS At the early stage of S-AKI, MANF in the kidney tissue was up-regulated, but with the development of the disease, it was down-regulated. Renal function was worsened in the S-AKI group, and TNF-α and IL-6 were elevated. The administration of MANF significantly alleviated the elevated levels of SCr and BUN and inhibited the expression of TNF-α and IL-6 in the kidney. The pathological changes were more extensive in the S-AKI group than in the S-AKI+MANF group. CONCLUSION MANF treatment may significantly alleviate renal injury, reduce the inflammatory response, and alleviate or reverse kidney tissue damage. MANF may have a protective effect on S-AKI, suggesting a potential treatment for S-AKI.
Collapse
Affiliation(s)
- Saifeng Chen
- Postgraduate Training Base at Shanghai Gongli Hospital, Ningxia Medical College, Shanghai 200135, China
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| | - Xuewei Hao
- Postgraduate Training Base at Shanghai Gongli Hospital, Ningxia Medical College, Shanghai 200135, China
| | - Guo Chen
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| | - Guorong Liu
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| | - Xiaoyan Yuan
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| | - Peiling Shen
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| | - Dongfeng Guo
- Postgraduate Training Base at Shanghai Gongli Hospital, Ningxia Medical College, Shanghai 200135, China
- Department of Emergency Medicine, Shanghai Gongli Hospital, Shanghai 200135, China
| |
Collapse
|
37
|
Li J, Wang G, Xv X, Li Z, Shen Y, Zhang C, Zhang X. Identification of immune-associated genes in diagnosing osteoarthritis with metabolic syndrome by integrated bioinformatics analysis and machine learning. Front Immunol 2023; 14:1134412. [PMID: 37138862 PMCID: PMC10150333 DOI: 10.3389/fimmu.2023.1134412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
Background In the pathogenesis of osteoarthritis (OA) and metabolic syndrome (MetS), the immune system plays a particularly important role. The purpose of this study was to find key diagnostic candidate genes in OA patients who also had metabolic syndrome. Methods We searched the Gene Expression Omnibus (GEO) database for three OA and one MetS dataset. Limma, weighted gene co-expression network analysis (WGCNA), and machine learning algorithms were used to identify and analyze the immune genes associated with OA and MetS. They were evaluated using nomograms and receiver operating characteristic (ROC) curves, and finally, immune cells dysregulated in OA were investigated using immune infiltration analysis. Results After Limma analysis, the integrated OA dataset yielded 2263 DEGs, and the MetS dataset yielded the most relevant module containing 691 genes after WGCNA, with a total of 82 intersections between the two. The immune-related genes were mostly enriched in the enrichment analysis, and the immune infiltration analysis revealed an imbalance in multiple immune cells. Further machine learning screening yielded eight core genes that were evaluated by nomogram and diagnostic value and found to have a high diagnostic value (area under the curve from 0.82 to 0.96). Conclusion Eight immune-related core genes were identified (FZD7, IRAK3, KDELR3, PHC2, RHOB, RNF170, SOX13, and ZKSCAN4), and a nomogram for the diagnosis of OA and MetS was established. This research could lead to the identification of potential peripheral blood diagnostic candidate genes for MetS patients who also suffer from OA.
Collapse
Affiliation(s)
- Junchen Li
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Genghong Wang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xilin Xv
- The Third Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhigang Li
- The Second Department of Orthopedics and Traumatology, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yiwei Shen
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Cheng Zhang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaofeng Zhang
- Teaching and Research Section of Orthopedics and Traumatology, Heilongjiang University of Chinese Medicine, Harbin, China
- The Bone Injury Teaching Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Xiaofeng Zhang,
| |
Collapse
|
38
|
Griffin H, Sullivan SC, Barger SW, Phelan KD, Baldini G. Liraglutide Counteracts Endoplasmic Reticulum Stress in Palmitate-Treated Hypothalamic Neurons without Restoring Mitochondrial Homeostasis. Int J Mol Sci 2022; 24:ijms24010629. [PMID: 36614074 PMCID: PMC9820707 DOI: 10.3390/ijms24010629] [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: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
One feature of high-fat diet-induced neurodegeneration in the hypothalamus is an increased level of palmitate, which is associated with endoplasmic reticulum (ER) stress, loss of CoxIV, mitochondrial fragmentation, and decreased abundance of MC4R. To determine whether antidiabetic drugs protect against ER and/or mitochondrial dysfunction by lipid stress, hypothalamic neurons derived from pre-adult mice and neuronal Neuro2A cells were exposed to elevated palmitate. In the hypothalamic neurons, palmitate exposure increased expression of ER resident proteins, including that of SERCA2, indicating ER stress. Liraglutide reverted such altered ER proteostasis, while metformin only normalized SERCA2 expression. In Neuro2A cells liraglutide, but not metformin, also blunted dilation of the ER induced by palmitate treatment, and enhanced abundance and expression of MC4R at the cell surface. Thus, liraglutide counteracts, more effectively than metformin, altered ER proteostasis, morphology, and folding capacity in neurons exposed to fat. In palmitate-treated hypothalamic neurons, mitochondrial fragmentation took place together with loss of CoxIV and decreased mitochondrial membrane potential (MMP). Metformin, but not liraglutide, reverted mitochondrial fragmentation, and both liraglutide and metformin did not protect against either loss of CoxIV abundance or MMP. Thus, ER recovery from lipid stress can take place in hypothalamic neurons in the absence of recovered mitochondrial homeostasis.
Collapse
Affiliation(s)
- Haven Griffin
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sarah C. Sullivan
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Steven W. Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin D. Phelan
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
39
|
Zandi Shafagh R, Youhanna S, Keulen J, Shen JX, Taebnia N, Preiss LC, Klein K, Büttner FA, Bergqvist M, van der Wijngaart W, Lauschke VM. Bioengineered Pancreas-Liver Crosstalk in a Microfluidic Coculture Chip Identifies Human Metabolic Response Signatures in Prediabetic Hyperglycemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203368. [PMID: 36285680 PMCID: PMC9731722 DOI: 10.1002/advs.202203368] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/05/2022] [Indexed: 05/19/2023]
Abstract
Aberrant glucose homeostasis is the most common metabolic disturbance affecting one in ten adults worldwide. Prediabetic hyperglycemia due to dysfunctional interactions between different human tissues, including pancreas and liver, constitutes the largest risk factor for the development of type 2 diabetes. However, this early stage of metabolic disease has received relatively little attention. Microphysiological tissue models that emulate tissue crosstalk offer emerging opportunities to study metabolic interactions. Here, a novel modular multitissue organ-on-a-chip device is presented that allows for integrated and reciprocal communication between different 3D primary human tissue cultures. Precisely controlled heterologous perfusion of each tissue chamber is achieved through a microfluidic single "synthetic heart" pneumatic actuation unit connected to multiple tissue chambers via specific configuration of microchannel resistances. On-chip coculture experiments of organotypic primary human liver spheroids and intact primary human islets demonstrate insulin secretion and hepatic insulin response dynamics at physiological timescales upon glucose challenge. Integration of transcriptomic analyses with promoter motif activity data of 503 transcription factors reveals tissue-specific interacting molecular networks that underlie β-cell stress in prediabetic hyperglycemia. Interestingly, liver and islet cultures show surprising counter-regulation of transcriptional programs, emphasizing the power of microphysiological coculture to elucidate the systems biology of metabolic crosstalk.
Collapse
Affiliation(s)
- Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | - Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Jibbe Keulen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Nayere Taebnia
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
| | - Lena C Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Department of Drug Metabolism and Pharmacokinetics (DMPK), The Healthcare Business of Merck KGaA, 64293, Darmstadt, Germany
| | - Kathrin Klein
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Florian A Büttner
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| | - Mikael Bergqvist
- Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden
| | | | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, 17711, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376, Stuttgart, Germany
- University of Tuebingen, 72074, Tuebingen, Germany
| |
Collapse
|
40
|
Peng H, Hu B, Xie LQ, Su T, Li CJ, Liu Y, Yang M, Xiao Y, Feng X, Zhou R, Guo Q, Zhou HY, Huang Y, Jiang TJ, Luo XH. A mechanosensitive lipolytic factor in the bone marrow promotes osteogenesis and lymphopoiesis. Cell Metab 2022; 34:1168-1182.e6. [PMID: 35705079 DOI: 10.1016/j.cmet.2022.05.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022]
Abstract
Exercise can prevent osteoporosis and improve immune function, but the mechanism remains unclear. Here, we show that exercise promotes reticulocalbin-2 secretion from the bone marrow macrophages to initiate bone marrow fat lipolysis. Given the crucial role of lipolysis in exercise-stimulated osteogenesis and lymphopoiesis, these findings suggest that reticulocalbin-2 is a pivotal regulator of a local adipose-osteogenic/immune axis. Mechanistically, reticulocalbin-2 binds to a functional receptor complex, which is composed of neuronilin-2 and integrin beta-1, to activate a cAMP-PKA signaling pathway that mobilizes bone marrow fat via lipolysis to fuel the differentiation and function of mesenchymal and hematopoietic stem cells. Notably, the administration of recombinant reticulocalbin-2 in tail-suspended and old mice remarkably decreases bone marrow fat accumulation and promotes osteogenesis and lymphopoiesis. These findings identify reticulocalbin-2 as a novel mechanosensitive lipolytic factor in maintaining energy homeostasis in bone resident cells, and it provides a promising target for skeletal and immune health.
Collapse
Affiliation(s)
- Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ling-Qi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Tie-Jian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan 410008, China.
| |
Collapse
|
41
|
Greer LK, Meilleur KG, Harvey BK, Wires ES. Identification of ER/SR resident proteins as biomarkers for ER/SR calcium depletion in skeletal muscle cells. Orphanet J Rare Dis 2022; 17:225. [PMID: 35698232 PMCID: PMC9195201 DOI: 10.1186/s13023-022-02368-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aberrations to endoplasmic/sarcoplasmic reticulum (ER/SR) calcium concentration can result in the departure of endogenous proteins in a phenomenon termed exodosis. Redistribution of the ER/SR proteome can have deleterious effects to cell function and cell viability, often contributing to disease pathogenesis. Many proteins prone to exodosis reside in the ER/SR via an ER retention/retrieval sequence (ERS) and are involved in protein folding, protein modification, and protein trafficking. While the consequences of their extracellular presence have yet to be fully delineated, the proteins that have undergone exodosis may be useful for biomarker development. Skeletal muscle cells rely upon tightly coordinated ER/SR calcium release for muscle contractions, and perturbations to calcium homeostasis can result in myopathies. Ryanodine receptor type-1 (RYR1) is a calcium release channel located in the SR. Mutations to the RYR1 gene can compromise calcium homeostasis leading to a vast range of clinical phenotypes encompassing hypotonia, myalgia, respiratory insufficiency, ophthalmoplegia, fatigue and malignant hyperthermia (MH). There are currently no FDA approved treatments for RYR1-related myopathies (RYR1-RM). RESULTS Here we examine the exodosis profile of skeletal muscle cells following ER/SR calcium depletion. Proteomic analysis identified 4,465 extracellular proteins following ER/SR calcium depletion with 1,280 proteins significantly different than vehicle. A total of 54 ERS proteins were identified and 33 ERS proteins significantly increased following ER/SR calcium depletion. Specifically, ERS protein, mesencephalic astrocyte-derived neurotrophic factor (MANF), was elevated following calcium depletion, making it a potential biomarker candidate for human samples. Despite no significant elevation of MANF in plasma levels among healthy volunteers and RYR1-RM individuals, MANF plasma levels positively correlated with age in RYR1-RM individuals, presenting a potential biomarker of disease progression. Selenoprotein N (SEPN1) was also detected only in extracellular samples following ER/SR calcium depletion. This protein is integral to calcium handling and SEPN1 variants have a causal role in SEPN1-related myopathies (SEPN1-RM). Extracellular presence of ER/SR membrane proteins may provide new insight into proteomic alterations extending beyond ERS proteins. Pre-treatment of skeletal muscle cells with bromocriptine, an FDA approved drug recently found to have anti-exodosis effects, curbed exodosis of ER/SR resident proteins. CONCLUSION Changes to the extracellular content caused by intracellular calcium dysregulation presents an opportunity for biomarker development and drug discovery.
Collapse
Affiliation(s)
- Lacey K Greer
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | | | - Brandon K Harvey
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| | - Emily S Wires
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
42
|
Fels JA, Casalena G, Konrad C, Holmes HE, Dellinger RW, Manfredi G. Gene expression profiles in sporadic ALS fibroblasts define disease subtypes and the metabolic effects of the investigational drug EH301. Hum Mol Genet 2022; 31:3458-3477. [PMID: 35652455 DOI: 10.1093/hmg/ddac118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/04/2022] [Accepted: 05/17/2022] [Indexed: 01/18/2023] Open
Abstract
Metabolic alterations shared between the nervous system and skin fibroblasts have emerged in ALS. Recently, we found that a subgroup of sporadic ALS (sALS) fibroblasts (sALS1) is characterized by metabolic profiles distinct from other sALS cases (sALS2) and controls, suggesting that metabolic therapies could be effective in sALS. The metabolic modulators nicotinamide riboside and pterostilbene (EH301) are under clinical development for the treatment of ALS. Here, we studied the transcriptome and metabolome of sALS cells to understand the molecular bases of sALS metabotypes and the impact of EH301. Metabolomics and transcriptomics were investigated at baseline and after EH301 treatment. Moreover, weighted gene co-expression network analysis (WGCNA) was used to investigate the association of metabolic and clinical features. We found that the sALS1 transcriptome is distinct from sALS2 and that EH301 modifies gene expression differently in sALS1, sALS2, and controls. Furthermore, EH301 had strong protective effects against metabolic stress, an effect linked to anti-inflammatory and antioxidant pathways. WGCNA revealed that ALS functional rating scale and metabotypes are associated with gene modules enriched for cell cycle, immunity, autophagy, and metabolism genes, which are modified by EH301. Meta-analysis of publicly available transcriptomics data from induced motor neurons by Answer ALS confirmed functional associations of genes correlated with disease traits. A subset of genes differentially expressed in sALS fibroblasts was used in a machine learning model to predict disease progression. In conclusion, multi-omics analyses highlighted differential metabolic and transcriptomic profiles in patient-derived fibroblast sALS, which translate into differential responses to the investigational drug EH301.
Collapse
Affiliation(s)
- Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065.,Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Ave, New York, NY 10065
| | - Gabriella Casalena
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Csaba Konrad
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | | | | | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| |
Collapse
|
43
|
KDEL Receptors: Pathophysiological Functions, Therapeutic Options, and Biotechnological Opportunities. Biomedicines 2022; 10:biomedicines10061234. [PMID: 35740256 PMCID: PMC9220330 DOI: 10.3390/biomedicines10061234] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
KDEL receptors (KDELRs) are ubiquitous seven-transmembrane domain proteins encoded by three mammalian genes. They bind to and retro-transport endoplasmic reticulum (ER)-resident proteins with a C-terminal Lys-Asp-Glu-Leu (KDEL) sequence or variants thereof. In doing this, KDELR participates in the ER quality control of newly synthesized proteins and the unfolded protein response. The binding of KDEL proteins to KDELR initiates signaling cascades involving three alpha subunits of heterotrimeric G proteins, Src family kinases, protein kinases A (PKAs), and mitogen-activated protein kinases (MAPKs). These signaling pathways coordinate membrane trafficking flows between secretory compartments and control the degradation of the extracellular matrix (ECM), an important step in cancer progression. Considering the basic cellular functions performed by KDELRs, their association with various diseases is not surprising. KDELR mutants unable to bind the collagen-specific chaperon heat-shock protein 47 (HSP47) cause the osteogenesis imperfecta. Moreover, the overexpression of KDELRs appears to be linked to neurodegenerative diseases that share pathological ER-stress and activation of the unfolded protein response (UPR). Even immune function requires a functional KDELR1, as its mutants reduce the number of T lymphocytes and impair antiviral immunity. Several studies have also brought to light the exploitation of the shuttle activity of KDELR during the intoxication and maturation/exit of viral particles. Based on the above, KDELRs can be considered potential targets for the development of novel therapeutic strategies for a variety of diseases involving proteostasis disruption, cancer progression, and infectious disease. However, no drugs targeting KDELR functions are available to date; rather, KDELR has been leveraged to deliver drugs efficiently into cells or improve antigen presentation.
Collapse
|
44
|
Abstract
Constitutive vesicle trafficking is the default pathway used by all cells for movement of intracellular cargoes between subcellular compartments and in and out of the cell. Classically, constitutive trafficking was thought to be continuous and unregulated, in contrast to regulated secretion, wherein vesicles are stored intracellularly until undergoing synchronous membrane fusion following a Ca2+ signal. However, as shown in the literature reviewed here, many continuous trafficking steps can be up- or down-regulated by Ca2+, including several steps associated with human pathologies. Notably, we describe a series of Ca2+ pumps, channels, Ca2+-binding effector proteins, and their trafficking machinery targets that together regulate the flux of cargo in response to genetic alterations as well as baseline and agonist-dependent Ca2+ signals. Here, we review the most recent advances, organized by organellar location, that establish the importance of these components in trafficking steps. Ultimately, we conclude that Ca2+ regulates an expanding series of distinct mechanistic steps. Furthermore, the involvement of Ca2+ in trafficking is complex. For example, in some cases, the same Ca2+ effectors regulate surprisingly distinct trafficking steps, or even the same trafficking step with opposing influences, through binding to different target proteins.
Collapse
Affiliation(s)
- John Sargeant
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Jesse C Hay
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| |
Collapse
|
45
|
Caffeine and MDMA (Ecstasy) Exacerbate ER Stress Triggered by Hyperthermia. Int J Mol Sci 2022; 23:ijms23041974. [PMID: 35216090 PMCID: PMC8880705 DOI: 10.3390/ijms23041974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Drugs of abuse can cause local and systemic hyperthermia, a known trigger of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). Another trigger of ER stress and UPR is ER calcium depletion, which causes ER exodosis, the secretion of ER-resident proteins. In rodent models, club drugs such as 3,4-methylenedioxymethamphetamine (MDMA, ‘ecstasy’) can create hyperthermic conditions in the brain and cause toxicity that is affected by the environmental temperature and the presence of other drugs, such as caffeine. In human studies, MDMA stimulated an acute, dose-dependent increase in core body temperature, but an examination of caffeine and MDMA in combination remains a topic for clinical research. Here we examine the secretion of ER-resident proteins and activation of the UPR under combined exposure to MDMA and caffeine in a cellular model of hyperthermia. We show that hyperthermia triggers the secretion of normally ER-resident proteins, and that this aberrant protein secretion is potentiated by the presence of MDMA, caffeine, or a combination of the two drugs. Hyperthermia activates the UPR but the addition of MDMA or caffeine does not alter the canonical UPR gene expression despite the drug effects on ER exodosis of UPR-related proteins. One exception was increased BiP/GRP78 mRNA levels in MDMA-treated cells exposed to hyperthermia. These findings suggest that club drug use under hyperthermic conditions exacerbates disruption of ER proteostasis, contributing to cellular toxicity.
Collapse
|
46
|
Hebbar N, Epperly R, Vaidya A, Thanekar U, Moore SE, Umeda M, Ma J, Patil SL, Langfitt D, Huang S, Cheng C, Klco JM, Gottschalk S, Velasquez MP. CAR T cells redirected to cell surface GRP78 display robust anti-acute myeloid leukemia activity and do not target hematopoietic progenitor cells. Nat Commun 2022; 13:587. [PMID: 35102167 PMCID: PMC8803836 DOI: 10.1038/s41467-022-28243-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Developing CAR T cells for acute myeloid leukemia (AML) has been hampered by a paucity of targets that are expressed on AML blasts and not on hematopoietic progenitor cells (HPCs). Here we demonstrate that GRP78 is expressed on the cell surface of primary AML blasts but not HPCs. To target GRP78, we generate T cell expressing a GRP78-specific peptide-based CAR, which show evidence of minimal fratricide post activation/transduction and antigen-dependent T cell differentiation. GRP78-CAR T cells recognize and kill GRP78-positive AML cells without toxicity to HPCs. In vivo, GRP78-CAR T cells have significant anti-AML activity. To prevent antigen-dependent T cell differentiation, we block CAR signaling and GRP78 cell surface expression post activation by using dasatinib during GRP78-CAR T cell manufacturing. This significantly improves their effector function in vitro and in vivo. Thus, targeting cell surface GRP78-positive AML with CAR T cells is feasible, and warrants further active exploration.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cell Survival/drug effects
- Cytokines/metabolism
- Cytotoxicity, Immunologic/drug effects
- Dasatinib/pharmacology
- Endoplasmic Reticulum Chaperone BiP/immunology
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/immunology
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Chimeric Antigen/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Nikhil Hebbar
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Rebecca Epperly
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Abishek Vaidya
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Unmesha Thanekar
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sarah E Moore
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Masayuki Umeda
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jing Ma
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sagar L Patil
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Deanna Langfitt
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sujuan Huang
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Stephen Gottschalk
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - M Paulina Velasquez
- Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
47
|
Jha V, Kumari T, Manickam V, Assar Z, Olson KL, Min JK, Cho J. ERO1-PDI Redox Signaling in Health and Disease. Antioxid Redox Signal 2021; 35:1093-1115. [PMID: 34074138 PMCID: PMC8817699 DOI: 10.1089/ars.2021.0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Significance: Protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductase 1 (ERO1) are crucial for oxidative protein folding in the endoplasmic reticulum (ER). These enzymes are frequently overexpressed and secreted, and they contribute to the pathology of neurodegenerative, cardiovascular, and metabolic diseases. Recent Advances: Tissue-specific knockout mouse models and pharmacologic inhibitors have been developed to advance our understanding of the cell-specific functions of PDI and ERO1. In addition to their roles in protecting cells from the unfolded protein response and oxidative stress, recent studies have revealed that PDI and ERO1 also function outside of the cells. Critical Issues: Despite the well-known contributions of PDI and ERO1 to specific disease pathology, the detailed molecular and cellular mechanisms underlying these activities remain to be elucidated. Further, although PDI and ERO1 inhibitors have been identified, the results from previous studies require careful evaluation, as many of these agents are not selective and may have significant cytotoxicity. Future Directions: The functions of PDI and ERO1 in the ER have been extensively studied. Additional studies will be required to define their functions outside the ER.
Collapse
Affiliation(s)
- Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zahra Assar
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Kirk L Olson
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
48
|
Samy A, Yamano-Adachi N, Koga Y, Omasa T. Secretion of a low-molecular-weight species of endogenous GRP94 devoid of the KDEL motif during endoplasmic reticulum stress in Chinese hamster ovary cells. Traffic 2021; 22:425-438. [PMID: 34536241 PMCID: PMC9293085 DOI: 10.1111/tra.12818] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 01/04/2023]
Abstract
GRP94 (glucose‐regulated protein 94) is a well‐studied chaperone with a lysine, aspartic acid, glutamic acid and leucine (KDEL) motif at its C‐terminal, which is responsible for GRP94 localization in the endoplasmic reticulum (ER). GRP94 is upregulated during ER stress to help fold unfolded proteins or direct proteins to ER‐associated degradation. In a previous study, engineered GRP94 without the KDEL motif stimulated a powerful immune response in vaccine cells. In this report, we show that endogenous GRP94 is naturally secreted into the medium in a truncated form that lacks the KDEL motif in Chinese hamster ovary cells. The secretion of the truncated form of GRP94 was stimulated by the induction of ER stress. These truncations prevent GRP94 recognition by KDEL receptors and retention inside the cell. This study sheds light on a potential trafficking phenomenon during the unfolded protein response that may help understand the functional role of GRP94 as a trafficking molecule.
Collapse
Affiliation(s)
- Andrew Samy
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan
| | - Noriko Yamano-Adachi
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan.,Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Yuichi Koga
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan.,Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Takeshi Omasa
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Japan.,Industrial Biotechnology Initiative Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| |
Collapse
|
49
|
McCaughey J, Stevenson NL, Mantell JM, Neal CR, Paterson A, Heesom K, Stephens DJ. A general role for TANGO1, encoded by MIA3, in secretory pathway organization and function. J Cell Sci 2021; 134:jcs259075. [PMID: 34350936 PMCID: PMC8524724 DOI: 10.1242/jcs.259075] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 11/25/2022] Open
Abstract
Complex machinery is required to drive secretory cargo export from the endoplasmic reticulum (ER), which is an essential process in eukaryotic cells. In vertebrates, the MIA3 gene encodes two major forms of transport and Golgi organization protein 1 (TANGO1S and TANGO1L), which have previously been implicated in selective trafficking of procollagen. Using genome engineering of human cells, light microscopy, secretion assays, genomics and proteomics, we show that disruption of the longer form, TANGO1L, results in relatively minor defects in secretory pathway organization and function, including having limited impacts on procollagen secretion. In contrast, loss of both long and short forms results in major defects in cell organization and secretion. These include a failure to maintain the localization of ERGIC53 (also known as LMAN1) and SURF4 to the ER-Golgi intermediate compartment and dramatic changes to the ultrastructure of the ER-Golgi interface. Disruption of TANGO1 causes significant changes in early secretory pathway gene and protein expression, and impairs secretion not only of large proteins, but of all types of secretory cargo, including small soluble proteins. Our data support a general role for MIA3/TANGO1 in maintaining secretory pathway structure and function in vertebrate cells.
Collapse
Affiliation(s)
- Janine McCaughey
- Cell Biology Laboratories,
School of Biochemistry, Faculty of Life Sciences, University Walk,
University of Bristol, Bristol, BS8 1TD,
UK
| | - Nicola L. Stevenson
- Cell Biology Laboratories,
School of Biochemistry, Faculty of Life Sciences, University Walk,
University of Bristol, Bristol, BS8 1TD,
UK
| | - Judith M. Mantell
- Wolfson Bioimaging Facility, Faculty of Life
Sciences, University Walk, University of
Bristol, Bristol, BS8 1TD,
UK
| | - Chris R. Neal
- Wolfson Bioimaging Facility, Faculty of Life
Sciences, University Walk, University of
Bristol, Bristol, BS8 1TD,
UK
| | | | - Kate Heesom
- Proteomics Facility, Faculty of Life
Sciences, University Walk, University of
Bristol, Bristol, BS8 1TD,
UK
| | - David J. Stephens
- Cell Biology Laboratories,
School of Biochemistry, Faculty of Life Sciences, University Walk,
University of Bristol, Bristol, BS8 1TD,
UK
| |
Collapse
|
50
|
Albert K, Raymundo DP, Panhelainen A, Eesmaa A, Shvachiy L, Araújo GR, Chmielarz P, Yan X, Singh A, Cordeiro Y, Palhano FL, Foguel D, Luk KC, Domanskyi A, Voutilainen MH, Huttunen HJ, Outeiro TF, Saarma M, Almeida MS, Airavaara M. Cerebral dopamine neurotrophic factor reduces α-synuclein aggregation and propagation and alleviates behavioral alterations in vivo. Mol Ther 2021; 29:2821-2840. [PMID: 33940158 PMCID: PMC8417450 DOI: 10.1016/j.ymthe.2021.04.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/11/2021] [Accepted: 04/27/2021] [Indexed: 01/16/2023] Open
Abstract
A molecular hallmark in Parkinson's disease (PD) pathogenesis are α-synuclein aggregates. Cerebral dopamine neurotrophic factor (CDNF) is an atypical growth factor that is mostly resident in the endoplasmic reticulum but exerts its effects both intracellularly and extracellularly. One of the beneficial effects of CDNF can be protecting neurons from the toxic effects of α-synuclein. Here, we investigated the effects of CDNF on α-synuclein aggregation in vitro and in vivo. We found that CDNF directly interacts with α-synuclein with a KD = 23 ± 6 nM and reduces its auto-association. Using nuclear magnetic resonance (NMR) spectroscopy, we identified interaction sites on the CDNF protein. Remarkably, CDNF reduces the neuronal internalization of α-synuclein fibrils and induces the formation of insoluble phosphorylated α-synuclein inclusions. Intra-striatal CDNF administration alleviates motor deficits in rodents challenged with α-synuclein fibrils, though it did not reduce the number of phosphorylated α-synuclein inclusions in the substantia nigra. CDNF's beneficial effects on rodent behavior appear not to be related to the number of inclusions formed in the current context, and further study of its effects on the aggregation mechanism in vivo are needed. Nonetheless, the interaction of CDNF with α-synuclein, modifying its aggregation, spreading, and associated behavioral alterations, provides novel insights into the potential of CDNF as a therapeutic strategy in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Katrina Albert
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Diana P Raymundo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Anne Panhelainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ave Eesmaa
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Liana Shvachiy
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina, Universidade de Lisboa, Av Prof Egas Moniz, 1649-028 Lisbon, Portugal
| | - Gabriela R Araújo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Piotr Chmielarz
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow 31-343, Poland
| | - Xu Yan
- Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Aastha Singh
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Yraima Cordeiro
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Fernando L Palhano
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Debora Foguel
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil
| | - Kelvin C Luk
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Merja H Voutilainen
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Henri J Huttunen
- Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Herantis Pharma Plc, 20520 Espoo, Finland
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073 Göttingen, Germany; Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany; Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Marcius S Almeida
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil; Protein Advanced Biochemistry, CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21.941-902, Brazil.
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Neuroscience Center, HiLIFE, University of Helsinki, 00014 Helsinki, Finland; Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|