1
|
Fu JT, Liu J, Wu WB, Chen YT, Lu GD, Cao Q, Meng HB, Tong J, Zhu JH, Wang XJ, Liu Y, Zhuang C, Sheng C, Shen FM, Liu X, Wang H, Yu Y, Zhang Y, Liang HY, Zhang JB, Li DJ, Li X, Wang ZB, Wang P. Targeting EFHD2 inhibits interferon-γ signaling and ameliorates non-alcoholic steatohepatitis. J Hepatol 2024; 81:389-403. [PMID: 38670321 DOI: 10.1016/j.jhep.2024.04.009] [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] [Received: 09/18/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND & AIMS The precise pathomechanisms underlying the development of non-alcoholic steatohepatitis (NASH, also known as metabolic dysfunction-associated steatohepatitis [MASH]) remain incompletely understood. In this study, we investigated the potential role of EF-hand domain family member D2 (EFHD2), a novel molecule specific to immune cells, in the pathogenesis of NASH. METHODS Hepatic EFHD2 expression was characterized in patients with NASH and two diet-induced NASH mouse models. Single-cell RNA sequencing (scRNA-seq) and double-immunohistochemistry were employed to explore EFHD2 expression patterns in NASH livers. The effects of global and myeloid-specific EFHD2 deletion on NASH and NASH-related hepatocellular carcinoma were assessed. Molecular mechanisms underlying EFHD2 function were investigated, while chemical and genetic investigations were performed to assess its potential as a therapeutic target. RESULTS EFHD2 expression was significantly elevated in hepatic macrophages/monocytes in both patients with NASH and mice. Deletion of EFHD2, either globally or specifically in myeloid cells, improved hepatic steatosis, reduced immune cell infiltration, inhibited lipid peroxidation-induced ferroptosis, and attenuated fibrosis in NASH. Additionally, it hindered the development of NASH-related hepatocellular carcinoma. Specifically, deletion of myeloid EFHD2 prevented the replacement of TIM4+ resident Kupffer cells by infiltrated monocytes and reversed the decreases in patrolling monocytes and CD4+/CD8+ T cell ratio in NASH. Mechanistically, our investigation revealed that EFHD2 in myeloid cells interacts with cytosolic YWHAZ (14-3-3ζ), facilitating the translocation of IFNγR2 (interferon-γ receptor-2) onto the plasma membrane. This interaction mediates interferon-γ signaling, which triggers immune and inflammatory responses in macrophages during NASH. Finally, a novel stapled α-helical peptide targeting EFHD2 was shown to be effective in protecting against NASH pathology in mice. CONCLUSION Our study reveals a pivotal immunomodulatory and inflammatory role of EFHD2 in NASH, underscoring EFHD2 as a promising druggable target for NASH treatment. IMPACT AND IMPLICATIONS Non-alcoholic steatohepatitis (NASH) represents an advanced stage of non-alcoholic fatty liver disease (NAFLD); however, not all patients with NAFLD progress to NASH. A key challenge is identifying the factors that trigger inflammation, which propels the transition from simple fatty liver to NASH. Our research pinpointed EFHD2 as a pivotal driver of NASH, orchestrating the over-activation of interferon-γ signaling within the liver during NASH progression. A stapled peptide designed to target EFHD2 exhibited therapeutic promise in NASH mice. These findings support the potential of EFHD2 as a therapeutic target in NASH.
Collapse
Affiliation(s)
- Jiang-Tao Fu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jian Liu
- Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Wen-Bin Wu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Yi-Ting Chen
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Guo-Dong Lu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Qi Cao
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Hong-Bo Meng
- Department of General Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia-Hui Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xu-Jie Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Liu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunlin Zhuang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xingguang Liu
- Department of Pathogen Biology, Second Military Medical University, Shanghai, China
| | - Hua Wang
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, China
| | - Hai-Yan Liang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jia-Bao Zhang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiang Li
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China.
| | - Zhi-Bin Wang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai, China.
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China; National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China.
| |
Collapse
|
2
|
Zhang W, Chen L, Lu X, Dong X, Feng M, Tu Y, Wang Z. EFHD2 regulates T cell receptor signaling and modulates T helper cell activation in early sepsis. Int Immunopharmacol 2024; 133:112087. [PMID: 38669951 DOI: 10.1016/j.intimp.2024.112087] [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: 02/17/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
EFHD2 (EF-hand domain family, member D2) has been identified as a calcium-binding protein with immunomodulatory effects. In this study, we characterized the phenotype of Efhd2-deficient mice in sepsis and examined the biological functions of EFHD2 in peripheral T cell activation and T helper (Th) cell differentiation. Increased levels of EFHD2 expression accompanied peripheral CD4+ T cell activation in the early stages of sepsis. Transcriptomic analysis indicated that immune response activation was impaired in Efhd2-deficient CD4+ T cells. Further, Efhd2-deficient CD4+ T cells isolated from the spleen of septic mice showed impaired T cell receptor (TCR)-induced Th differentiation, especially Th1 and Th17 differentiation. In vitro data also showed that Efhd2-deficient CD4+ T cells exhibit impaired Th1 and Th17 differentiation. In the CD4+ T cells and macrophages co-culture model for antigen presentation, the deficiency of Efhd2 in CD4+ T cells resulted in impaired formation of immunological synapses. In addition, Efhd2-deficient CD4+ T cells exhibited reduced levels of phospho-LCK and phospho-ZAP70, and downstream transcription factors including Nfat, Nfκb and Nur77 following TCR engagement. In summary, EFHD2 may promote TCR-mediated T cell activation subsequent Th1 and Th17 differentiation in the early stages of sepsis by regulating the intensity of TCR complex formation.
Collapse
Affiliation(s)
- Wenzhao Zhang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Linlin Chen
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Xin Lu
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Xiaohui Dong
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Meixia Feng
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Ye Tu
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Zhibin Wang
- Department of Critical Care Medicine, School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Soliman AS, Umstead A, Lamp J, Vega IE. EFhd2 co-aggregates with monomeric and filamentous tau in vitro. Front Neurosci 2024; 18:1373410. [PMID: 38765673 PMCID: PMC11100465 DOI: 10.3389/fnins.2024.1373410] [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: 01/19/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024] Open
Abstract
Tauopathies are characterized by the abnormal buildup of tau protein, with early oligomeric forms associated with neurodegeneration and the later neurofibrillary tangles possibly conferring neuroprotection. The molecular mechanisms governing the formation of these tau species are unclear. Lately, there has been an increased focus on examining the interactions between tau and other proteins, along with their influence on the aggregation of tau. Our previous work revealed EFhd2's association with pathological tau in animal models and tauopathy brains. Herein, we examined the impact of EFhd2 on monomeric and filamentous tau in vitro. The results demonstrated that EFhd2 incubation with monomeric full length human tau (hTau40) formed amorphous aggregates, where both EFhd2 and hTau40 colocalized. Moreover, EFhd2 is entangled with arachidonic acid (ARA)-induced filamentous hTau40. Furthermore, EFhd2-induced aggregation with monomeric and filamentous hTau40 is EFhd2 concentration dependent. Using sandwich ELISA assays, we assessed the reactivity of TOC1 and Alz50-two conformation-specific tau antibodies-to EFhd2-hTau40 aggregates (in absence and presence of ARA). No TOC1 signal was detected in EFhd2 aggregates with monomeric hTau40 whereas EFhd2 aggregates with hTau in the presence of ARA showed a higher signal compared to hTau40 filaments. In contrast, EFhd2 aggregates with both monomeric and filamentous hTau40 reduced Alz50 reactivity. Taken together, our results illustrate for the first time that EFhd2, a tau-associated protein, interacts with monomeric and filamentous hTau40 to form large aggregates that are starkly different from tau oligomers and filaments. Given these findings and previous research, we hypothesize that EFhd2 may play a role in the formation of tau aggregates. Nevertheless, further in vivo studies are imperative to test this hypothesis.
Collapse
Affiliation(s)
- Ahlam S. Soliman
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Andrew Umstead
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Integrated Mass Spectrometry Unit, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Jared Lamp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
| | - Irving E. Vega
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Neuroscience Program, Michigan State University, East Lansing, MI, United States
- Integrated Mass Spectrometry Unit, College of Human Medicine, Michigan State University, Grand Rapids, MI, United States
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Michigan Alzheimer's Disease Research Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
4
|
Thylur Puttalingaiah R. Role of Swiprosin-1/EFHD2 as a biomarker in the development of chronic diseases. Life Sci 2022; 297:120462. [PMID: 35276221 DOI: 10.1016/j.lfs.2022.120462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
Swiprosin-1 or EFHD2, is a Ca2+ binding actin protein and its expression has been shown to be distinct in various cell types. The expression of swiprosin-1 is upregulated during the activation of immune cells, epithelial and endothelial cells. The expression of swiprosin-1 is regulated by diverse signaling pathways that are contingent upon the specific type of cells. The aim of this review is to summarize and provide an overview of the role of swiprosin-1 in pathophysiological conditions of cancers, cardiovascular diseases, diabetic nephropathy, neuropsychiatric diseases, and in the process of inflammation, immune response, and inflammatory diseases. Novel approaches for the targeting of swiprosin-1 as a biomarker in the early detection and prevention of various development of chronic diseases are also explored.
Collapse
Affiliation(s)
- Ramesh Thylur Puttalingaiah
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, 1700 Tulane Avenue, Room 945-B1, New Orleans, LA 70112, USA..
| |
Collapse
|
5
|
Soliman AS, Umstead A, Grabinski T, Kanaan NM, Lee A, Ryan J, Lamp J, Vega IE. EFhd2 brain interactome reveals its association with different cellular and molecular processes. J Neurochem 2021; 159:992-1007. [PMID: 34543436 PMCID: PMC9552186 DOI: 10.1111/jnc.15517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/11/2021] [Accepted: 09/09/2021] [Indexed: 01/06/2023]
Abstract
EFhd2 is a conserved calcium-binding protein that is highly expressed in the central nervous system. We have shown that EFhd2 interacts with tau protein, a key pathological hallmark in Alzheimer's disease and related dementias. However, EFhd2's physiological and pathological functions in the brain are still poorly understood. To gain insights into its physiological function, we identified proteins that co-immunoprecipitated with EFhd2 from mouse forebrain and hindbrain, using tandem mass spectrometry (MS). In addition, quantitative mass spectrometry was used to detect protein abundance changes due to the deletion of the Efhd2 gene in mouse forebrain and hindbrain regions. Our data show that mouse EFhd2 is associated with cytoskeleton components, vesicle trafficking modulators, cellular stress response-regulating proteins, and metabolic proteins. Moreover, proteins associated with the cytoskeleton, vesicular transport, calcium signaling, stress response, and metabolic pathways showed differential abundance in Efhd2(-/-) mice. This study presents, for the first time, an EFhd2 brain interactome that it is associated with different cellular and molecular processes. These findings will help prioritize further studies to investigate the mechanisms by which EFhd2 modulates these processes in physiological and pathological conditions of the nervous system.
Collapse
Affiliation(s)
- Ahlam S Soliman
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Andrew Umstead
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA
| | - Tessa Grabinski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA
| | - Andy Lee
- NeuroInitiatives LLC, Jacksonville, Florida, USA
| | - John Ryan
- NeuroInitiatives LLC, Jacksonville, Florida, USA
| | - Jared Lamp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA
| | - Irving E Vega
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA.,Neuroscience Program, Michigan State University, Grand Rapids, Michigan, USA.,Integrated Mass Spectrometry Unit, College of Human Medicine, Grand Rapids, Michigan, USA.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, Michigan, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
6
|
Umstead A, Soliman AS, Lamp J, Vega IE. Validation of recombinant human protein purified from bacteria: An important step to increase scientific rigor. Anal Biochem 2020; 611:113999. [DOI: 10.1016/j.ab.2020.113999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/17/2020] [Indexed: 11/29/2022]
|
7
|
Gao M, Zeng K, Li Y, Liu YP, Xia X, Xu FL, Yao J, Wang BJ. Association between EFHD2 gene polymorphisms and schizophrenia among the Han population in northern China. J Int Med Res 2020; 48:300060520932801. [PMID: 32567430 PMCID: PMC7309398 DOI: 10.1177/0300060520932801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Objective Schizophrenia is a severe neurodevelopmental disorder with a complex genetic and environmental etiology. The gene encoding EF-hand domain-containing protein D2 (EFHD2) may be a genetic risk locus for schizophrenia. Methods We genotyped four EFHD2 single-nucleotide polymorphisms (281 schizophrenia cases [SCZ], 321 controls) from northern Chinese Han individuals using Sanger sequencing and polymerase chain reaction-restriction fragment length polymorphism analysis. Differences existed in genotype, allele, and haplotype frequency distributions between SCZ and control groups. Results The rs2473357 genotype and allele frequency distributions differed between SCZ and controls; however, this difference disappeared after Bonferroni correction. Differences in rs2473357 genotype and allele frequency distributions between SCZ and controls were more pronounced in men than in women. The G allele increased schizophrenia risk (odds ratio = 1.807, 95% confidence interval = 1.164–2.803). Among six haplotypes (G–, A–, G-insC, A-C, G-C, and G-T), the G– haplotype frequency distribution differed between SCZ and controls in women; the A-C and G-C haplotype frequency distributions differed between SCZ and controls in men. Conclusions EFHD2 may be involved in schizophrenia. Sex differences in EFHD2 genotype and allele frequency distributions existed among schizophrenia patients. Further research is needed to determine the role of EFHD2 in schizophrenia.
Collapse
Affiliation(s)
- Meng Gao
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Kuo Zeng
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Ya Li
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Yong-Ping Liu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Xi Xia
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Feng-Ling Xu
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, China
| | - Bao-Jie Wang
- School of Forensic Medicine, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Vega IE, Umstead A, Kanaan NM. EFhd2 Affects Tau Liquid-Liquid Phase Separation. Front Neurosci 2019; 13:845. [PMID: 31456657 PMCID: PMC6700279 DOI: 10.3389/fnins.2019.00845] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 07/30/2019] [Indexed: 12/17/2022] Open
Abstract
The transition of tau proteins from its soluble physiological conformation to the pathological aggregate forms found in Alzheimer's disease and related dementias, is poorly understood. Therefore, understanding the process that modulates the formation of toxic tau oligomers and their conversion to putative neuroprotective neurofibrillary tangles will lead to better therapeutic strategies. We previously identified that EFhd2 is associated with aggregated tau species in AD brains and the coiled-coil domain in EFhd2 mediates the interaction with tau. To further characterize the association between EFhd2 and tau, we examined whether EFhd2 could affect the liquid-liquid phase separation properties of tau under molecular crowding conditions. We demonstrate that EFhd2 alters tau liquid phase behavior in a calcium and coiled-coil domain dependent manner. Co-incubation of EFhd2 and tau in the absence of calcium leads to the formation of solid-like structures containing both proteins, while in the presence of calcium these two proteins phase separate together into liquid droplets. EFhd2's coiled-coil domain is necessary to alter tau's liquid phase separation, indicating that protein-protein interaction is required. The results demonstrate that EFhd2 affects the liquid-liquid phase separation of tau proteins in vitro, suggesting that EFhd2 modulates the structural dynamics of tau proteins.
Collapse
Affiliation(s)
- Irving E Vega
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, Grand Rapids, MI, United States.,Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Andrew Umstead
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Grand Rapids, MI, United States
| | - Nicholas M Kanaan
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
9
|
Peled M, Dragovich MA, Adam K, Strazza M, Tocheva AS, Vega IE, Mor A. EF Hand Domain Family Member D2 Is Required for T Cell Cytotoxicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2824-2831. [PMID: 30275048 PMCID: PMC6200634 DOI: 10.4049/jimmunol.1800839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 12/29/2022]
Abstract
Programmed cell death 1 (PD-1) is a major coinhibitory receptor and a member of the immunological synapse (IS). To uncover proteins that regulate PD-1 recruitment to the IS, we searched for cytoskeleton-related proteins that also interact with PD-1 using affinity purification mass spectrometry. Among these proteins, EF hand domain family member D2 (EFHD2), a calcium binding adaptor protein, was functionally and mechanistically analyzed for its contribution to PD-1 signaling. EFHD2 was required for PD-1 to inhibit cytokine secretion, proliferation, and adhesion of human T cells. Interestingly, EFHD2 was also required for human T cell-mediated cytotoxicity and for mounting an antitumor immune response in a syngeneic murine tumor model. Mechanistically, EFHD2 contributed to IS stability, lytic vesicles trafficking, and granzyme B secretion. Altogether, EFHD2 is an important regulator of T cell cytotoxicity and further studies should evaluate its role in T cell-mediated inflammation.
Collapse
Affiliation(s)
- Michael Peled
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
- Pulmonary Department, The Chaim Sheba Medical Center, Ramat Gan 52620, Israel
| | - Matthew A Dragovich
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Kieran Adam
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Marianne Strazza
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Anna S Tocheva
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503; and
| | - Adam Mor
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016;
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
10
|
Regensburger M, Prots I, Reimer D, Brachs S, Loskarn S, Lie DC, Mielenz D, Winner B. Impact of Swiprosin-1/Efhd2 on Adult Hippocampal Neurogenesis. Stem Cell Reports 2018; 10:347-355. [PMID: 29337116 PMCID: PMC5830914 DOI: 10.1016/j.stemcr.2017.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022] Open
Abstract
Swiprosin-1/Efhd2 (Efhd2) is highly expressed in the CNS during development and in the adult. EFHD2 is regulated by Ca2+ binding, stabilizes F-actin, and promotes neurite extension. Previous studies indicated a dysregulation of EFHD2 in human Alzheimer's disease brains. We hypothesized a detrimental effect of genetic ablation of Efhd2 on hippocampal integrity and specifically investigated adult hippocampal neurogenesis. Efhd2 was expressed throughout adult neuronal development and in mature neurons. We observed a severe reduction of the survival of adult newborn neurons in Efhd2 knockouts, starting at the early neuroblast stage. Spine formation and dendrite growth of newborn neurons were compromised in full Efhd2 knockouts, but not upon cell-autonomous Efhd2 deletion. Together with our finding of severe hippocampal tauopathy in Efhd2 knockout mice, these data connect Efhd2 to impaired synaptic plasticity as present in Alzheimer's disease and identify a role of Efhd2 in neuronal survival and synaptic integration in the adult hippocampus. Efhd2 is expressed in the dentate gyrus and its loss reduces adult neurogenesis Reduced neurite complexity and spine density in new neurons of Efhd2 knockout mice Role of cell-extrinsic EFHD2 for dendrite morphology of adult newborn neurons Increased levels of pathological TAU in the hippocampus of Efhd2 knockout mice
Collapse
Affiliation(s)
- Martin Regensburger
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany; Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany; IZKF Junior Research Group III and BMBF Research Group Neuroscience, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Iryna Prots
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany; IZKF Junior Research Group III and BMBF Research Group Neuroscience, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Dorothea Reimer
- Department of Molecular Immunology, Department of Internal Medicine III, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany
| | - Sebastian Brachs
- Department of Molecular Immunology, Department of Internal Medicine III, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany
| | - Sandra Loskarn
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany; Department of Neurology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany; IZKF Junior Research Group III and BMBF Research Group Neuroscience, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Dieter Chichung Lie
- Emil-Fischer Centre, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Dirk Mielenz
- Department of Molecular Immunology, Department of Internal Medicine III, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany.
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Glueckstrasse 6, Erlangen 91054, Germany; IZKF Junior Research Group III and BMBF Research Group Neuroscience, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen 91054, Germany.
| |
Collapse
|
11
|
Physiological and pathophysiological functions of Swiprosin-1/EFhd2 in the nervous system. Biochem J 2017; 473:2429-37. [PMID: 27515255 DOI: 10.1042/bcj20160168] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022]
Abstract
Synaptic dysfunction and dysregulation of Ca(2+) are linked to neurodegenerative processes and behavioural disorders. Our understanding of the causes and factors involved in behavioural disorders and neurodegeneration, especially Alzheimer's disease (AD), a tau-related disease, is on the one hand limited and on the other hand controversial. Here, we review recent data about the links between the Ca(2+)-binding EF-hand-containing cytoskeletal protein Swiprosin-1/EFhd2 and neurodegeneration. Specifically, we summarize the functional biochemical data obtained in vitro with the use of recombinant EFhd2 protein, and integrated them with in vivo data in order to interpret the emerging role of EFhd2 in synaptic plasticity and in the pathophysiology of neurodegenerative disorders, particularly involving the tauopathies. We also discuss its functions in actin remodelling through cofilin and small GTPases, thereby linking EFhd2, synapses and the actin cytoskeleton. Expression data and functional experiments in mice and in humans have led to the hypothesis that down-regulation of EFhd2, especially in the cortex, is involved in dementia.
Collapse
|
12
|
Park KR, An JY, Kang JY, Lee JG, Lee Y, Mun SA, Jun CD, Song WK, Eom SH. Structural mechanism underlying regulation of human EFhd2/Swiprosin-1 actin-bundling activity by Ser183 phosphorylation. Biochem Biophys Res Commun 2017; 483:442-448. [DOI: 10.1016/j.bbrc.2016.12.124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022]
|
13
|
Vega IE. EFhd2, a Protein Linked to Alzheimer's Disease and Other Neurological Disorders. Front Neurosci 2016; 10:150. [PMID: 27064956 PMCID: PMC4814571 DOI: 10.3389/fnins.2016.00150] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/21/2016] [Indexed: 01/20/2023] Open
Abstract
EFhd2 is a conserved calcium binding protein linked to different neurological disorders and types of cancer. Although, EFhd2 is more abundant in neurons, it is also found in other cell types. The physiological function of this novel protein is still unclear, but it has been shown in vitro to play a role in calcium signaling, apoptosis, actin cytoskeleton, and regulation of synapse formation. Recently, EFhd2 was shown to promote cell motility by modulating the activity of Rac1, Cdc42, and RhoA. Although, EFhd2's role in promoting cell invasion and metastasis is of great interest in cancer biology, this review focusses on the evidence that links EFhd2 to Alzheimer's disease (AD) and other neurological disorders. Altered expression of EFhd2 has been documented in AD, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, and schizophrenia, indicating that Efhd2 gene expression is regulated in response to neuropathological processes. However, the specific role that EFhd2 plays in the pathophysiology of neurological disorders is still poorly understood. Recent studies demonstrated that EFhd2 has structural characteristics similar to amyloid proteins found in neurological disorders. Moreover, EFhd2 co-aggregates and interacts with known neuropathological proteins, such as tau, C9orf72, and Lrrk2. These results suggest that EFhd2 may play an important role in the pathophysiology of neurodegenerative diseases. Therefore, the understanding of EFhd2's role in health and disease could lead to decipher molecular mechanisms that become activated in response to neuronal stress and degeneration.
Collapse
Affiliation(s)
- Irving E Vega
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University Grand Rapids, MI, USA
| |
Collapse
|
14
|
The calcium-binding protein EFhd2 modulates synapse formation in vitro and is linked to human dementia. J Neuropathol Exp Neurol 2015; 73:1166-82. [PMID: 25383639 PMCID: PMC4238966 DOI: 10.1097/nen.0000000000000138] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
EFhd2 is a calcium-binding adaptor protein that has been found to be associated with pathologically aggregated tau in the brain in Alzheimer disease and in a mouse model of frontotemporal dementia. EFhd2 has cell type-specific functions, including the modulation of intracellular calcium responses, actin dynamics, and microtubule transport. Here we report that EFhd2 protein and mRNA levels are reduced in human frontal cortex tissue affected by different types of dementia with and without tau pathology. We show that EFhd2 is mainly a neuronal protein in the brain and is abundant in the forebrain. Using short hairpin RNA-mediated knockdown of EFhd2 expression in cultured cortical neurons, we demonstrate that loss of EFhd2 affects the number of synapses developed in vitro whereas it does not alter neurite outgrowth per se. Our data suggest that EFhd2 is involved in the control of synapse development and maintenance through means other than affecting neurite development. The changes in expression levels observed in human dementias might, therefore, play a significant role in disease onset and progression of dementia, which is characterized by the loss of synapses.
Collapse
|