1
|
Choi YJ, Nam YA, Hyun JY, Yu J, Mun Y, Yun SH, Lee W, Park CJ, Han BW, Lee BH. Impaired chaperone-mediated autophagy leads to abnormal SORT1 (sortilin 1) turnover and CES1-dependent triglyceride hydrolysis. Autophagy 2025; 21:827-839. [PMID: 39611307 PMCID: PMC11925108 DOI: 10.1080/15548627.2024.2435234] [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: 01/22/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
SORT1 (sortilin 1), a member of the the Vps10 (vacuolar protein sorting 10) family, is involved in hepatic lipid metabolism by regulating very low-density lipoprotein (VLDL) secretion and facilitating the lysosomal degradation of CES1 (carboxylesterase 1), crucial for triglyceride (TG) breakdown in the liver. This study explores whether SORT1 is targeted for degradation by chaperone-mediated autophagy (CMA), a selective protein degradation pathway that directs proteins containing KFERQ-like motifs to lysosomes via LAMP2A (lysosomal-associated membrane protein 2A). Silencing LAMP2A or HSPA8/Hsc70 with siRNA increased cytosolic SORT1 protein levels. Leupeptin treatment induced lysosomal accumulation of SORT1, unaffected by siLAMP2A co-treatment, indicating CMA-dependent degradation. Human SORT1 contains five KFERQ-like motifs (658VVTKQ662, 730VREVK734, 733VKDLK737, 734KDLKK738, and 735DLKKK739), crucial for HSPA8 recognition; mutating any single amino acid within these motifs decreased HSPA8 binding. Furthermore, compromised CMA activity resulted in elevated SORT1-mediated degradation of CES1, contributing to increased lipid accumulation in hepatocytes. Consistent with in vitro findings, LAMP2A knockdown in mice exacerbated high-fructose diet-induced fatty liver, marked by increased SORT1 and decreased CES1 levels. Conversely, LAMP2A overexpression promoted SORT1 degradation and CES1D accumulation, counteracting fasting-induced CES1D suppression through CMA activation. Our findings reveal that SORT1 is a substrate of CMA, highlighting its crucial role in directing CES1 to lysosomes. Consequently, disrupting CMA-mediated SORT1 degradation significantly affects CES1-dependent TG hydrolysis, thereby affecting hepatic lipid homeostasis.Abbreviations: APOB: apolipoprotein B; CES1: carboxylesterase 1; CMA: chaperone-mediated autophagy; HSPA8/Hsc70: heat shock protein family A (Hsp70) member 8; LAMP2A: lysosomal associated membrane protein 2A; LDL-C: low-density lipoprotein-cholesterol; PLIN: perilipin; SORT1: sortilin 1; TG: triglyceride; VLDL: very low-density lipoprotein; Vps10: vacuolar protein sorting 10.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Gyeongsangbuk-do, Republic of Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Ah Nam
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Ye Hyun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jihyeon Yu
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yewon Mun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Ho Yun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wonseok Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheon Jun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byung Woo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Cornelis MC, Fazlollahi A, Bennett DA, Schneider JA, Ayton S. Genetic Markers of Postmortem Brain Iron. J Neurochem 2025; 169:e16309. [PMID: 39918201 PMCID: PMC11804167 DOI: 10.1111/jnc.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025]
Abstract
Brain iron (Fe) dyshomeostasis is implicated in neurodegenerative diseases. Genome-wide association studies (GWAS) have identified plausible loci correlated with peripheral levels of Fe. Systemic organs and the brain share several Fe regulatory proteins but there likely exist different homeostatic pathways. We performed the first GWAS of inductively coupled plasma mass spectrometry measures of postmortem brain Fe from 635 Rush Memory and Aging Project (MAP) participants. Sixteen single nucleotide polymorphisms (SNPs) associated with Fe in at least one of four brain regions were measured (p < 5 × 10-8). Promising SNPs (p < 5 × 10-6) were followed up for replication in published GWAS of blood, spleen, and brain imaging Fe traits and mapped to candidate genes for targeted cortical transcriptomic and epigenetic analysis of postmortem Fe in MAP. Results for SNPs previously associated with other Fe traits were also examined. Ninety-eight SNPs associated with postmortem brain Fe were at least nominally (p < 0.05) associated with one or more related Fe traits. Most novel loci identified had no direct links to Fe regulatory pathways but rather endoplasmic reticulum-Golgi trafficking (SORL1, SORCS2, MARCH1, CLTC), heparan sulfate (HS3ST4, HS3ST1), and coenzyme A (SLC5A6, PANK3); supported by nearest gene function and omic analyses. We replicated (p < 0.05) several previously published Fe loci mapping to candidate genes in cellular and systemic Fe regulation. Finally, novel loci (BMAL, COQ5, SLC25A11) and replication of prior loci (PINK1, PPIF, LONP1) lend support to the role of circadian rhythms and mitochondria function in Fe regulation more generally. In summary, we provide support for novel loci linked to pathways that may have greater relevance to brain Fe accumulation; some of which are implicated in neurodegeneration. However, replication of a subset of prior loci for blood Fe suggests that genetic determinants or biological pathways underlying Fe accumulation in the brain are not completely distinct from those of Fe circulating in the periphery.
Collapse
Affiliation(s)
- Marilyn C. Cornelis
- Department of Preventive MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Amir Fazlollahi
- Department of Radiology, Royal Melbourne HospitalUniversity of MelbourneMelbourneVictoriaAustralia
- Queensland Brain InstituteThe University of QueenslandBrisbaneQueenslandAustralia
| | | | | | - Scott Ayton
- The Florey Institute of Neuroscience and Mental HealthMelbourneVictoriaAustralia
- Florey Department of Neuroscience and Mental HealthThe University of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Huo Y, Zhao D, Zhu X, Zheng N, Yang D, Meng J, Chen Y, Zhang YW. RPS23RG1 inhibits SORT1-mediated lysosomal degradation of MDGA2 to protect against autism. Theranostics 2025; 15:1338-1352. [PMID: 39816685 PMCID: PMC11729561 DOI: 10.7150/thno.100451] [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: 07/03/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025] Open
Abstract
Rationale: Mutations in the synaptic protein MAM domain containing glycosylphosphatidylinositol anchor 2 (MDGA2) have been associated with autism spectrum disorder (ASD). Therefore, elucidating the regulatory mechanisms of MDGA2 can help develop effective treatments for ASD. Methods: Liquid chromatography-tandem mass spectrometry was carried out to identify proteins interacting with the extracellular domain of RPS23RG1 and with MDGA2, followed by co-immunoprecipitation assays to confirm protein-protein interactions. RPS23RG1 and SORT1 levels were downregulated by siRNAs to study their effects on MDGA2 degradation, with additional applications of immunoblotting and immunostaining assays. Lysosome isolation was performed to determine the lysosomal degradation of MDGA2 further. Rps23rg1 knockout mice and Mdga2 +/- mice were subjected to various behavioral tests to study their ASD-like phenotypes. AAVs expressing MDGA2 were delivered in Rps23rg1 knockout mice, and RPS23RG1-derived peptide was delivered in Mdga2 +/- mice to study their rescuing effects. Results: We found that both RPS23RG1 and SORT1 interacted with MDGA2. MDGA2 was primarily degraded through the SORT1-mediated lysosomal degradation pathway. RPS23RG1 competed with SORT1 for MDGA2 binding to inhibit MDGA2 degradation. Furthermore, we showed that Rps23rg1 knockout mice exhibited decreased MDGA2 levels and ASD-like behaviors, whereas restoration of MDGA2 levels attenuated social defects in Rps23rg1 KO mice. Moreover, we identified a crucial region of RPS23RG1 for MDGA2 interaction and found that a peptide derived from this region not only bound MDGA2 and promoted MDGA2 levels, but also rescued social defects in Mdga2 +/- mice. Conclusion: Our findings highlight a crucial role of RPS23RG1 in antagonizing SORT1-mediated lysosomal degradation of MDGA2 and suggest a potential for targeting the RPS23RG1-MDGA2 axis to treat ASD with MDGA2 deficiency.
Collapse
Affiliation(s)
- Yuanhui Huo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Dongdong Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiang Zhu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Naizhen Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Dingting Yang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Jian Meng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yiqing Chen
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| | - Yun-wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Li YJ, Fang CB, Wang SS, Chen XQ, Li Y, Liu Q, Qi YK, Du SS. Design and synthesis of TH19P01-Camptothecin based hybrid peptides inducing effective anticancer responses on sortilin positive cancer cells. Bioorg Med Chem 2024; 111:117869. [PMID: 39126834 DOI: 10.1016/j.bmc.2024.117869] [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: 06/09/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Recently, the sortilin receptor (SORT1) was found to be preferentially over-expressed on the surface of many cancer cells, which makes SORT1 a novel anticancer target. The SORT1 binding proprietary peptide TH19P01 could achieve the SORT1-mediated cancer cell binding and subsequent internalization. Inspired by the peptide-drug conjugate (PDC) strategy, the TH19P01-camptothecin (CPT) conjugates were designed, efficiently synthesized, and evaluated for their anticancer potential in this study. The water solubility, in vitro anticancer activity, time-kill kinetics, cellular uptake, anti-migration activity, and hemolysis effects were systematically estimated. Besides, in order to monitor the release of CPT from conjugates in real-time, the CPT/Dnp-based "turn on" hybrid peptide was designed, which indicted that CPT could be sustainably released from the hybrid peptide in both human serum and cancer cellular environments. Strikingly, compared with free CPT, the water solubility, cellular uptake, and selectivity towards cancer cells of hybrid peptide LYJ-2 have all been significantly enhanced. Moreover, unlike free CPT or TH19P01, LYJ-2 exhibited selective anti-proliferative and anti-migration effects against SORT1-positive MDA-MB-231 cells. Collectively, this study not only established efficient strategies to improve the solubility and anticancer potential of chemotherapeutic agent CPT, but also provided important references for the future development of TH19P01 based PDCs targeting SORT1.
Collapse
Affiliation(s)
- Ya-Jie Li
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Chang-Bo Fang
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Shu-Shu Wang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266555, China
| | - Xin-Qi Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yantao Li
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qing Liu
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yun-Kun Qi
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China.
| | - Shan-Shan Du
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
5
|
Georgoula M, Ntavaroukas P, Androutsopoulou A, Xiromerisiou G, Kalala F, Speletas M, Asprodini E, Vasilaki A, Papoutsopoulou S. Sortilin Expression Levels and Peripheral Immunity: A Potential Biomarker for Segregation between Parkinson's Disease Patients and Healthy Controls. Int J Mol Sci 2024; 25:1791. [PMID: 38339069 PMCID: PMC10855941 DOI: 10.3390/ijms25031791] [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: 12/09/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Parkinson's disease (PD) is characterized by substantial phenotypic heterogeneity that limits the disease prognosis and patient's counseling, and complicates the design of further clinical trials. There is an unmet need for the development and validation of biomarkers for the prediction of the disease course. In this study, we utilized flow cytometry and in vitro approaches on peripheral blood cells and isolated peripheral blood mononuclear cell (PBMC)-derived macrophages to characterize specific innate immune populations in PD patients versus healthy donors. We found a significantly lower percentage of B lymphocytes and monocyte populations in PD patients. Monocytes in PD patients were characterized by a higher CD40 expression and on-surface expression of the type I membrane glycoprotein sortilin, which showed a trend of negative correlation with the age of the patients. These results were further investigated in vitro on PBMC-derived macrophages, which, in PD patients, showed higher sortilin expression levels compared to cells from healthy donors. The treatment of PD-derived macrophages with oxLDL led to higher foam cell formation compared to healthy donors. In conclusion, our results support the hypothesis that surface sortilin expression levels on human peripheral monocytes may potentially be utilized as a marker of Parkinson's disease and may segregate the sporadic versus the genetically induced forms of the disease.
Collapse
Affiliation(s)
- Maria Georgoula
- Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (M.G.); (P.N.); (A.A.)
| | - Panagiotis Ntavaroukas
- Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (M.G.); (P.N.); (A.A.)
| | - Anastasia Androutsopoulou
- Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (M.G.); (P.N.); (A.A.)
| | | | - Fani Kalala
- Laboratory of of Immunology & Histocompatibility, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (F.K.); (M.S.)
| | - Matthaios Speletas
- Laboratory of of Immunology & Histocompatibility, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (F.K.); (M.S.)
| | - Eftihia Asprodini
- Laboratory of Clinical Pharmacology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
| | - Anna Vasilaki
- Laboratory of Pharmacology, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece;
| | - Stamatia Papoutsopoulou
- Department of Biochemistry & Biotechnology, University of Thessaly, 41500 Larissa, Greece; (M.G.); (P.N.); (A.A.)
| |
Collapse
|
6
|
Zhuang W, Zhang W, Xie L, Wang L, Li Y, Wang Z, Zhang A, Qiu H, Feng J, Zhang B, Hu Y. Generation and Characterization of SORT1-Targeted Antibody-Drug Conjugate for the Treatment of SORT1-Positive Breast Tumor. Int J Mol Sci 2023; 24:17631. [PMID: 38139459 PMCID: PMC10743877 DOI: 10.3390/ijms242417631] [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: 11/08/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have greatly improved the outcomes of advanced breast tumors. However, the treatment of breast tumors with existing ADCs is still hindered by many issues, such as tumor antigen heterogeneity and drug resistance. Therefore, ADCs against new targets would provide options for the treatment of these challenges. Sortilin-1 (SORT1) may be a promising target for ADC as it is upregulated in breast cancer. To evaluate the possibility of SORT1 as an ADC target, a humanized antibody_8D302 with high affinity against SORT1 was generated. Additionally, 8D302 was conjugated with MMAE and DXd to generate two ADCs_8D302-MMAE and 8D302-DXd, respectively. Both 8D302-MMAE and 8D302-DXd showed effective cytotoxicity against SORT1 positive breast tumor cell lines and induced bystander killing. Consequently, 8D302-MMAE showed relatively better anti-tumor activity than 8D302-DXd both in vitro and in vivo, but 8D302-DXd had superior safety profile and pharmacokinetics profile over 8D302-MMAE. Furthermore, SORT1 induced faster internalization and lysosomal trafficking of antibodies and had a higher turnover compared with HER2. Also, 8D302-DXd exhibited superior cell cytotoxicity and tumor suppression over trastuzumab-DXd, a HER2-targeted ADC. We hypothesize that the high turnover of SORT1 enables SORT1-targeted ADC to be a powerful agent for the treatment of SORT1-positive breast tumor.
Collapse
Affiliation(s)
- Weiliang Zhuang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Wei Zhang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Liping Xie
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Lei Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
| | - Yuan Li
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Ziyu Wang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Ao Zhang
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Haitao Qiu
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Jun Feng
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| | - Baohong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; (W.Z.); (L.W.)
| | - Youjia Hu
- China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China; (W.Z.); (L.X.); (Y.L.); (Z.W.); (A.Z.); (H.Q.); (J.F.)
| |
Collapse
|
7
|
Berger K, Persson E, Gregersson P, Ruiz-Martínez S, Jonasson E, Ståhlberg A, Rhost S, Landberg G. Interleukin-6 Induces Stem Cell Propagation through Liaison with the Sortilin-Progranulin Axis in Breast Cancer. Cancers (Basel) 2023; 15:5757. [PMID: 38136303 PMCID: PMC10741783 DOI: 10.3390/cancers15245757] [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: 08/11/2023] [Revised: 10/25/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Unraveling the complex network between cancer cells and their tumor microenvironment is of clinical importance, as it might allow for the identification of new targets for cancer treatment. Cytokines and growth factors secreted by various cell types present in the tumor microenvironment have the potential to affect the challenging subpopulation of cancer stem cells showing treatment-resistant properties as well as aggressive features. By using various model systems, we investigated how the breast cancer stem cell-initiating growth factor progranulin influenced the secretion of cancer-associated proteins. In monolayer cultures, progranulin induced secretion of several inflammatory-related cytokines, such as interleukin (IL)-6 and -8, in a sortilin-dependent manner. Further, IL-6 increased the cancer stem fraction similarly to progranulin in the breast cancer cell lines MCF7 and MDA-MB-231 monitored by the surrogate mammosphere-forming assay. In a cohort of 63 patient-derived scaffold cultures cultured with breast cancer cells, we observed significant correlations between IL-6 and progranulin secretion, clearly validating the association between IL-6 and progranulin also in human-based microenvironments. In conclusion, the interplay between progranulin and IL-6 highlights a dual breast cancer stem cell-promoting function via sortilin, further supporting sortilin as a highly relevant therapeutic target for aggressive breast cancer.
Collapse
Affiliation(s)
- Karoline Berger
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Emma Persson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Pernilla Gregersson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Santiago Ruiz-Martínez
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Emma Jonasson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
- Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, 41390 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 41346 Gothenburg, Sweden
| | - Sara Rhost
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| | - Göran Landberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (K.B.); (E.P.); (P.G.); (S.R.-M.); (E.J.); (A.S.); (S.R.)
| |
Collapse
|
8
|
Overby M, Serrano-Rodriguez A, Dadras S, Christiansen AK, Ozcelik G, Lichtenthaler SF, Weick JP, Müller HK. Neuron-specific gene NSG1 binds to and positively regulates sortilin ectodomain shedding via a metalloproteinase-dependent mechanism. J Biol Chem 2023; 299:105446. [PMID: 37949230 PMCID: PMC10704435 DOI: 10.1016/j.jbc.2023.105446] [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: 05/30/2023] [Revised: 10/15/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
Increasing evidence suggests that aberrant regulation of sortilin ectodomain shedding can contribute to amyloid-β pathology and frontotemporal dementia, although the mechanism by which this occurs has not been elucidated. Here, we probed for novel binding partners of sortilin using multiple and complementary approaches and identified two proteins of the neuron-specific gene (NSG) family, NSG1 and NSG2, that physically interact and colocalize with sortilin. We show both NSG1 and NSG2 induce subcellular redistribution of sortilin to NSG1- and NSG2-enriched compartments. However, using cell surface biotinylation, we found only NSG1 reduced sortilin cell surface expression, which caused significant reductions in uptake of progranulin, a molecular determinant for frontotemporal dementia. In contrast, we demonstrate NSG2 has no effect on sortilin cell surface abundance or progranulin uptake, suggesting specificity for NSG1 in the regulation of sortilin cell surface expression. Using metalloproteinase inhibitors and A disintegrin and metalloproteinase 10 KO cells, we further show that NSG1-dependent reduction of cell surface sortilin occurred via proteolytic processing by A disintegrin and metalloproteinase 10 with a concomitant increase in shedding of sortilin ectodomain to the extracellular space. This represents a novel regulatory mechanism for sortilin ectodomain shedding that is regulated in a neuron-specific manner. Furthermore, this finding has implications for the development of strategies for brain-specific regulation of sortilin and possibly sortilin-driven pathologies.
Collapse
Affiliation(s)
- Malene Overby
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Antonio Serrano-Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Somayeh Dadras
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Ann Kathrine Christiansen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gözde Ozcelik
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jason Porter Weick
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
9
|
Zhou L, He Y, Deng Y, Li X, Wang W, Chen J. Ciclopirox mitigates inflammatory response in LPS-induced septic shock via inactivation of SORT1-mediated wnt/β-Catenin signaling pathway. Immunopharmacol Immunotoxicol 2023; 45:701-708. [PMID: 37606515 DOI: 10.1080/08923973.2023.2231628] [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: 10/28/2022] [Accepted: 06/23/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVE Septic shock, the most severe stage of sepsis, is a deadly inflammatory disorder with high mortality. Ciclopirox (CPX) is a broad-spectrum antimycotic agent which also exerts anti-inflammatory effects in human diseases. However, whether CPX can relieve inflammatory response in LPS-induced septic shock remains unclear. MATERIALS AND METHODS Male C57BL/6 mice LPS were injected intraperitoneally with LPS to simulate septic shock in vivo. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were subject to LPS treatment to simulate septic shock in vitro. ELISA was applied to detect the level of pro-inflammatory cytokines. Cell viability was assessed by CCK-8 assay. Protein levels was detected by western blotting. RESULTS CPX enhanced the survival rate and attenuated inflammation in mice with LPS-induced septic shock. Similarly, CPX dose-dependently mitigated LPS-induced inflammation in BMDMs. It was also found that Sortilin 1 (SORT1) was upregulated in both in vivo and in vitro models of LPS-induced septic shock. In addition, SORT1 overexpression counteracted the alleviative effects of CPX on the inflammation response of LPS-challenged BMDMs by activating the Wnt/β-Catenin signaling. Furthermore, BML-284 (a Wnt/β-Catenin agonist) treatment also abrogated CPX-mediated moderation of LPS-triggered inflammatory reaction in BMDMs. CONCLUSIONS In sum, we found that CPX protected against LPS-induced septic shock by mitigating inflammation via SORT1-mediated Wnt/β-Catenin signaling pathway.
Collapse
Affiliation(s)
- Liangliang Zhou
- Department of Emergency Intensive Care Medicine and Department of Emergency Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Yingfeng He
- Department of Emergency Intensive Care Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Yijun Deng
- Department of Emergency Intensive Care Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Xinxin Li
- Department of Emergency Intensive Care Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Wei Wang
- Department of Emergency Intensive Care Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| | - Jianjun Chen
- Department of Emergency Intensive Care Medicine and Department of Emergency Medicine, The Fourth Affiliated Hospital of Nantong University/The First People's Hospital of Yancheng, Yancheng, Jiangsu, China
| |
Collapse
|
10
|
Thomasen PB, Salasova A, Kjaer-Sorensen K, Woloszczuková L, Lavický J, Login H, Tranberg-Jensen J, Almeida S, Beel S, Kavková M, Qvist P, Kjolby M, Ovesen PL, Nolte S, Vestergaard B, Udrea AC, Nejsum LN, Chao MV, Van Damme P, Krivanek J, Dasen J, Oxvig C, Nykjaer A. SorCS2 binds progranulin to regulate motor neuron development. Cell Rep 2023; 42:113333. [PMID: 37897724 DOI: 10.1016/j.celrep.2023.113333] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/25/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Motor neuron (MN) development and nerve regeneration requires orchestrated action of a vast number of molecules. Here, we identify SorCS2 as a progranulin (PGRN) receptor that is required for MN diversification and axon outgrowth in zebrafish and mice. In zebrafish, SorCS2 knockdown also affects neuromuscular junction morphology and fish motility. In mice, SorCS2 and PGRN are co-expressed by newborn MNs from embryonic day 9.5 until adulthood. Using cell-fate tracing and nerve segmentation, we find that SorCS2 deficiency perturbs cell-fate decisions of brachial MNs accompanied by innervation deficits of posterior nerves. Additionally, adult SorCS2 knockout mice display slower motor nerve regeneration. Interestingly, primitive macrophages express high levels of PGRN, and their interaction with SorCS2-positive motor axon is required during axon pathfinding. We further show that SorCS2 binds PGRN to control its secretion, signaling, and conversion into granulins. We propose that PGRN-SorCS2 signaling controls MN development and regeneration in vertebrates.
Collapse
Affiliation(s)
- Pernille Bogetofte Thomasen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lucie Woloszczuková
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Josef Lavický
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Hande Login
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jeppe Tranberg-Jensen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sergio Almeida
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sander Beel
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Michaela Kavková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mads Kjolby
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Peter Lund Ovesen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Stella Nolte
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Benedicte Vestergaard
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Andreea-Cornelia Udrea
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Moses V Chao
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Philip Van Damme
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Jeremy Dasen
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Anders Nykjaer
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
11
|
Klein M, Failla AV, Hermey G. Internally tagged Vps10p-domain receptors reveal uptake of the neurotrophin BDNF. J Biol Chem 2023; 299:105216. [PMID: 37660918 PMCID: PMC10540051 DOI: 10.1016/j.jbc.2023.105216] [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: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
The Vps10p-domain (Vps10p-D) receptor family consists of Sortilin, SorLA, SorCS1, SorCS2, and SorCS3. They mediate internalization and intracellular sorting of specific cargo in various cell types, but underlying molecular determinants are incompletely understood. Deciphering the dynamic intracellular itineraries of Vps10p-D receptors is crucial for understanding their role in physiological and cytopathological processes. However, studying their spatial and temporal dynamics by live imaging has been challenging so far, as terminal tagging with fluorophores presumably impedes several of their protein interactions and thus functions. Here, we addressed the lack of appropriate tools and developed functional versions of all family members internally tagged in their ectodomains. We predict folding of the newly designed receptors by bioinformatics and show their exit from the endoplasmic reticulum. We examined their subcellular localization in immortalized cells and primary cultured neurons by immunocytochemistry and live imaging. This was, as far as known, identical to that of wt counterparts. We observed homodimerization of fluorophore-tagged SorCS2 by coimmunoprecipitation and fluorescence lifetime imaging, suggesting functional leucine-rich domains. Through ligand uptake experiments, live imaging and fluorescence lifetime imaging, we show for the first time that all Vps10p-D receptors interact with the neurotrophin brain-derived neurotrophic factor and mediate its uptake, indicating functionality of the Vps10p-Ds. In summary, we developed versions of all Vps10p-D receptors, with internal fluorophore tags that preserve several functions of the cytoplasmic and extracellular domains. These newly developed fluorophore-tagged receptors are likely to serve as powerful functional tools for accurate live studies of the individual cellular functions of Vps10p-D receptors.
Collapse
Affiliation(s)
- Marcel Klein
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | - Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
12
|
Purrahman D, Shojaeian A, Poniatowski ŁA, Piechowski-Jóźwiak B, Mahmoudian-Sani MR. The Role of Progranulin (PGRN) in the Pathogenesis of Ischemic Stroke. Cell Mol Neurobiol 2023; 43:3435-3447. [PMID: 37561339 PMCID: PMC11410000 DOI: 10.1007/s10571-023-01396-8] [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: 12/11/2022] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Stroke is a life-threatening medical condition and is a leading cause of disability. Cerebral ischemia is characterized by a distinct inflammatory response starting with the production of various cytokines and other inflammation-related agents. Progranulin (PGRN), a multifunctional protein, is critical in diverse physiological reactions, such as cell proliferation, inflammation, wound healing, and nervous system development. A mature PGRN is anti-inflammatory, while granulin, its derivative, conversely induces pro-inflammatory cytokine expression. PGRN is significantly involved in the brain tissue and its damage, for example, improving mood and cognitive disorders caused by cerebral ischemia. It may also have protective effects against nerve and spinal cord injuries by inhibiting neuroinflammatory response and apoptosis or it may be related to the proliferation, accumulation, differentiation, and activation of microglia. PGRN is a neurotrophic factor in the central nervous system. It may increase post-stroke neurogenesis of the subventricular zone (SVZ), which is particularly important in improving long-term brain function following cerebral ischemia. The neurogenesis enhanced via PGRN in the ischemic brain SVZ may be attributed to the induction of PI3K/AKT and MAPK/ERK signaling routes. PGRN can also promote the proliferation of neural stem/progenitor cells through PI3K/AKT signaling pathway. PGRN increases hippocampal neurogenesis, reducing anxiety and impaired spatial learning post-cerebral ischemia. PGRN alleviates cerebral ischemia/reperfusion injury by reducing endoplasmic reticulum stress and suppressing the NF-κB signaling pathway. PGRN can be introduced as a potent neuroprotective agent capable of improving post-ischemia neuronal actions, mainly by reducing and elevating the inflammatory and anti-inflammatory cytokines. Expression, storage, cleavage, and function of progranulin (PGRN) in the pathogenesis of ischemic stroke.
Collapse
Affiliation(s)
- Daryush Purrahman
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Łukasz A Poniatowski
- Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Salvador-Allende-Straße 30, 17036, Neubrandenburg, Germany
| | - Bartłomiej Piechowski-Jóźwiak
- Neurological Institute, Cleveland Clinic Abu Dhabi, 59 Hamouda Bin Ali Al Dhaheri Street, Jazeerat Al Maryah, PO Box 112412, Abu Dhabi, United Arab Emirates
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
13
|
Febbraro F, Andersen HHB, Kitt MM, Willnow TE. Spatially and temporally distinct patterns of expression for VPS10P domain receptors in human cerebral organoids. Front Cell Dev Biol 2023; 11:1229584. [PMID: 37842085 PMCID: PMC10570844 DOI: 10.3389/fcell.2023.1229584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023] Open
Abstract
Vacuolar protein sorting 10 protein (VPS10P) domain receptors are a unique class of intracellular sorting receptors that emerge as major risk factors associated with psychiatric and neurodegenerative diseases, including bipolar disorders, autism, schizophrenia, as well as Alzheimer's disease and frontotemporal dementia. Yet, the lack of suitable experimental models to study receptor functions in the human brain has hampered elucidation of receptor actions in brain disease. Here, we have adapted protocols using human cerebral organoids to the detailed characterization of VPS10P domain receptor expression during neural development and differentiation, including single-cell RNA sequencing. Our studies uncovered spatial and temporal patterns of expression unique to individual receptor species in the human brain. While SORL1 expression is abundant in stem cells and SORCS1 peaks in neural progenitors at onset of neurogenesis, SORT1 and SORCS2 show increasing expression with maturation of neuronal and non-neuronal cell types, arguing for distinct functions in development versus the adult brain. In neurons, subcellular localization also distinguishes between types of receptor species, either mainly localized to the cell soma (SORL1 and SORT1) or also to neuronal projections (SORCS1 and SORCS2), suggesting divergent functions in protein sorting between Golgi and the endo-lysosomal system or along axonal and dendritic tracks. Taken together, our findings provide an important resource on temporal, spatial, and subcellular patterns of VPS10P domain receptor expression in cerebral organoids for further elucidation of receptor (dys) functions causative of behavioral and cognitive defects of the human brain.
Collapse
Affiliation(s)
- Fabia Febbraro
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Meagan M. Kitt
- Max Delbrueck Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas E. Willnow
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Max Delbrueck Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
14
|
Omagari D, Toriumi T, Tsuda H, Hayatsu M, Watanabe K, Mizutami Y, Honda M, Mikami Y. Inductive effect of SORT1 on odontoblastic differentiation of human dental pulp-derived stem cells. Differentiation 2023; 133:88-97. [PMID: 37579565 DOI: 10.1016/j.diff.2023.08.001] [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: 01/12/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023]
Abstract
This study investigated the expression of sortilin 1 (SORT1) in cultured human dental pulp-derived stem cells (hDPSCs) and its role in their odontoblastic differentiation. Permanent teeth were extracted from five patients, and the dental pulp was harvested for explant culture. Fluorescence-activated cell sorting was used to analyze the outgrowth of adherent cells and cells that had migrated from the tissue margin. SORT1 expression was detected in hDPSCs simultaneously expressing the mesenchymal stem cell markers CD44 and CD90. The odontoblastic differentiation potential of SORT1-positive hDPSCs was examined via staining for alkaline phosphatase (ALP), an early odontoblastic differentiation marker. ALP staining was more intense in SORT1-positive than in SORT1-negative hDPSCs. Consistently, the expression of mRNA encoding SORT1 and p75NTR, a binding partner of SORT1, increased in SORT1-positive hDPSCs during odontoblastic differentiation. In addition, pro-nerve growth factor (NGF), a ligand for SORT1-p75NTR co-receptor, promoted ALP expression in SORT1-positive hDPSCs, and the interaction between SORT1 and p75NTR was detected using a coimmunoprecipitation assay. The function of SORT1 in odontoblastic differentiation was examined via RNA interference using shRNA targeting SORT1. ALP staining intensity in SORT1/shRNA-transfected cells was markedly lower than in control/shRNA-transfected cells. SORT1 knockdown decreased JUN phosphorylation and recruitment of phosphorylated JUN to the ALP promoter. Collectively, these results indicate that SORT1 is involved in the odontoblastic differentiation of hDPSCs through the JUN N-terminal kinases (JNK)/JUN signaling pathway and that the binding of SORT1 and p75NTR plays an important role in this process.
Collapse
Affiliation(s)
- Daisuke Omagari
- Department of Pathology, Tsurumi University School of Dental Medicine, Kanagawa, Japan
| | - Taku Toriumi
- Department of Physical Therapy, Faculty of Rehabilitation, Kyushu Nutrition Welfare University, Fukuoka, Japan; Department of Anatomy, The Nippon Dental University School of Life Dentistry at Niigata, Niigata, Japan
| | - Hiromasa Tsuda
- Department of Biochemistry, Nihon University School of Dentistry, Tokyo, Japan
| | - Manabu Hayatsu
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Keisuke Watanabe
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yusuke Mizutami
- Office of Institutional Research, Hokkaido University, Hokkaido, Japan
| | - Masaki Honda
- Department of Oral Anatomy, Aichi Gakuin University School of Dentistry, Aichi, Japan
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| |
Collapse
|
15
|
Richter E, Lohmann CH, Dell’Accio F, Goettsch C, Bertrand J. Sortilin Is Upregulated in Osteoarthritis-Dependent Cartilage Calcification and Associated with Cellular Senescence. Int J Mol Sci 2023; 24:12343. [PMID: 37569721 PMCID: PMC10418692 DOI: 10.3390/ijms241512343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by articular cartilage calcification, loss of articular cartilage, bone changes, pain, and disability. Cartilage calcification is one hallmark of OA and is predominantly caused by basic calcium crystals formed due to an imbalance of the pyrophosphate pathway. Sortilin is a transmembrane protein that contributes to vascular calcification in atherosclerosis by externalizing alkaline phosphatase (ALP)-containing vesicles. Calcification in atherosclerosis and osteoarthritis has been associated with cellular senescence. The aim of this study was to investigate the potential role of sortilin and senescence in osteoarthritis-dependent cartilage calcification. Osteoarthritic cartilage from human knee joints was collected after joint replacement, and samples were analyzed by immunohistochemistry and quantitative RT-PCR analysis. Human chondrocytes were treated with osteogenic medium for up to 21 days to induce calcification. Western blots for sortilin and ALP, as well as an ALP activity assay, were performed. Human chondrocytes were treated with mitomycin C to induce senescence, and sortilin expression was quantified at the protein and gene levels. Sections of knee joints from a murine model of osteoarthritis were stained for sortilin and p16 and analyzed by immunohistochemistry. Treatment of wild-type chondrocytes using an osteogenic medium similar to human chondrocytes was performed. Osteoarthritic cartilage from mouse and human knee joints showed an increased number of sortilin and p16-positive chondrocytes compared to healthy cartilage. This observation was corroborated by increased gene expression of sortilin and p16 in mild and moderate osteoarthritic cartilage samples. To investigate the mechanism of sortilin regulation, human chondrocytes were treated with osteogenic medium to induce calcification. Sortilin protein levels and expression were increased after 7 days of stimulation, whereas ALP levels and activity were upregulated after 21 days of stimulation. Similar observations were made in a murine osteoarthritis model. Mechanistically, senescent chondrocytes induced by mitomycin C showed an upregulation of sortilin and ALP gene expression compared to non-senescent chondrocytes. Our data indicate that sortilin and ALP are upregulated during cartilage calcification, which is associated with chondrocyte senescence and thus might contribute to the pathogenesis of osteoarthritis. Cellular senescence seems to induce sortilin expression.
Collapse
Affiliation(s)
- Elisabeth Richter
- Department of Orthopaedic Surgery, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany; (E.R.); (C.H.L.)
| | - Christoph H. Lohmann
- Department of Orthopaedic Surgery, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany; (E.R.); (C.H.L.)
| | - Francesco Dell’Accio
- William Harvey Research Institute, Queen Mary University London, London EC1M 6BQ, UK;
| | - Claudia Goettsch
- Department of Internal Medicine I-Cardiology, RWTH Aachen University, 52062 Aachen, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany; (E.R.); (C.H.L.)
| |
Collapse
|
16
|
Keller MP, Hudkins KL, Shalev A, Bhatnagar S, Kebede MA, Merrins MJ, Davis DB, Alpers CE, Kimple ME, Attie AD. What the BTBR/J mouse has taught us about diabetes and diabetic complications. iScience 2023; 26:107036. [PMID: 37360692 PMCID: PMC10285641 DOI: 10.1016/j.isci.2023.107036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Human and mouse genetics have delivered numerous diabetogenic loci, but it is mainly through the use of animal models that the pathophysiological basis for their contribution to diabetes has been investigated. More than 20 years ago, we serendipidously identified a mouse strain that could serve as a model of obesity-prone type 2 diabetes, the BTBR (Black and Tan Brachyury) mouse (BTBR T+ Itpr3tf/J, 2018) carrying the Lepob mutation. We went on to discover that the BTBR-Lepob mouse is an excellent model of diabetic nephropathy and is now widely used by nephrologists in academia and the pharmaceutical industry. In this review, we describe the motivation for developing this animal model, the many genes identified and the insights about diabetes and diabetes complications derived from >100 studies conducted in this remarkable animal model.
Collapse
Affiliation(s)
- Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Anath Shalev
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Melkam A. Kebede
- School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, Sydney, NSW 2006, Australia
| | - Matthew J. Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Charles E. Alpers
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Michelle E. Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
17
|
Lee AK, Yi N, Khaled H, Feller B, Takahashi H. SorCS1 inhibits amyloid-β binding to neurexin and rescues amyloid-β-induced synaptic pathology. Life Sci Alliance 2023; 6:e202201681. [PMID: 36697254 PMCID: PMC9880023 DOI: 10.26508/lsa.202201681] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
Amyloid-β oligomers (AβOs), toxic peptide aggregates found in Alzheimer's disease, cause synapse pathology. AβOs interact with neurexins (NRXs), key synaptic organizers, and this interaction dampens normal trafficking and function of NRXs. Axonal trafficking of NRX is in part regulated by its interaction with SorCS1, a protein sorting receptor, but the impact of SorCS1 regulation of NRXs in Aβ pathology was previously unstudied. Here, we show competition between the SorCS1 ectodomain and AβOs for β-NRX binding and rescue effects of the SorCS1b isoform on AβO-induced synaptic pathology. Like AβOs, the SorCS1 ectodomain binds to NRX1β through the histidine-rich domain of NRX1β, and the SorCS1 ectodomain and AβOs compete for NRX1β binding. In cultured hippocampal neurons, SorCS1b colocalizes with NRX1β on the axon surface, and axonal expression of SorCS1b rescues AβO-induced impairment of NRX-mediated presynaptic organization and presynaptic vesicle recycling and AβO-induced structural defects in excitatory synapses. Thus, our data suggest a role for SorCS1 in the rescue of AβO-induced NRX dysfunction and synaptic pathology, providing the basis for a novel potential therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Alfred Kihoon Lee
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Nayoung Yi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Husam Khaled
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Benjamin Feller
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
| | - Hideto Takahashi
- Synapse Development and Plasticity Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
- Department of Medicine, Université de Montréal, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| |
Collapse
|
18
|
Kamran M, Laighneach A, Bibi F, Donohoe G, Ahmed N, Rehman AU, Morris DW. Independent Associated SNPs at SORCS3 and Its Protein Interactors for Multiple Brain-Related Disorders and Traits. Genes (Basel) 2023; 14:482. [PMID: 36833409 PMCID: PMC9956385 DOI: 10.3390/genes14020482] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/26/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Sortilin-related vacuolar protein sorting 10 (VPS10) domain containing receptor 3 (SORCS3) is a neuron-specific transmembrane protein involved in the trafficking of proteins between intracellular vesicles and the plasma membrane. Genetic variation at SORCS3 is associated with multiple neuropsychiatric disorders and behavioural phenotypes. Here, we undertake a systematic search of published genome-wide association studies to identify and catalogue associations between SORCS3 and brain-related disorders and traits. We also generate a SORCS3 gene-set based on protein-protein interactions and investigate the contribution of this gene-set to the heritability of these phenotypes and its overlap with synaptic biology. Analysis of association signals at SORSC3 showed individual SNPs to be associated with multiple neuropsychiatric and neurodevelopmental brain-related disorders and traits that have an impact on the experience of feeling, emotion or mood or cognitive function, while multiple LD-independent SNPs were associated with the same phenotypes. Across these SNPs, alleles associated with the more favourable outcomes for each phenotype (e.g., decreased risk of neuropsychiatric illness) were associated with increased expression of the SORCS3 gene. The SORCS3 gene-set was enriched for heritability contributing to schizophrenia (SCZ), bipolar disorder (BPD), intelligence (IQ) and education attainment (EA). Eleven genes from the SORCS3 gene-set were associated with more than one of these phenotypes at the genome-wide level, with RBFOX1 associated with SCZ, IQ and EA. Functional annotation revealed that the SORCS3 gene-set is enriched for multiple ontologies related to the structure and function of synapses. Overall, we find many independent association signals at SORCS3 with brain-related disorders and traits, with the effect possibly mediated by reduced gene expression, resulting in a negative impact on synaptic function.
Collapse
Affiliation(s)
- Muhammad Kamran
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Farhana Bibi
- Department of Biosciences, Grand Asian University, Sialkot 51040, Pakistan
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| | - Naveed Ahmed
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Asim Ur Rehman
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics (NICOG), School of Biological and Chemical Sciences and School of Psychology, University of Galway, H91 CF50 Galway, Ireland
| |
Collapse
|
19
|
Saeedi-Boroujeni A, Purrahman D, Shojaeian A, Poniatowski ŁA, Rafiee F, Mahmoudian-Sani MR. Progranulin (PGRN) as a regulator of inflammation and a critical factor in the immunopathogenesis of cardiovascular diseases. J Inflamm (Lond) 2023; 20:1. [PMID: 36658641 PMCID: PMC9851114 DOI: 10.1186/s12950-023-00327-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Immune dysregulation has been identified as a critical cause of the most common types of cardiovascular diseases (CVDs). Notably, the innate and adaptive immune responses under physiological conditions are typically regulated with high sensitivity to avoid the exacerbation of inflammation, but any dysregulation can probably be associated with CVDs. In this respect, progranulin (PGRN) serves as one of the main components of the regulation of inflammatory processes, which significantly contributes to the immunopathogenesis of such disorders. PGRN has been introduced among the secreted growth factors as one related to wound healing, inflammation, and human embryonic development, as well as a wide variety of autoimmune diseases. The relationship between the serum PGRN and TNF-α ratio with the spontaneous bacterial peritonitis constitute one of the independent predictors of these conditions. The full-length PGRN can thus effectively reduce the calcification of valve interstitial cells, and the granulin precursor (GRN), among the degradation products of PGRN, can be beneficial. Moreover, it was observed that, PGRN protects the heart against ischemia-reperfusion injury. Above all, PGRN also provides protection in the initial phase following myocardial ischemia-reperfusion injury. The protective impact of PGRN on this may be associated with the early activation of the PI3K/Akt signaling pathway. PGRN also acts as a protective factor in hyperhomocysteinemia, probably by down-regulating the wingless-related integration site Wnt/β-catenin signaling pathway. Many studies have further demonstrated that SARS-CoV-2 (COVID-19) has dramatically increased the risks of CVDs due to inflammation, so PGRN has drawn much more attention among scholars. Lysosomes play a pivotal role in the inflammation process, and PGRN is one of the key regulators in their functioning, which contributes to the immunomodulatory mechanism in the pathogenesis of CVDs. Therefore, investigation of PGRN actions can help find new prospects in the treatment of CVDs. This review aims to summarize the role of PGRN in the immunopathogenesis of CVD, with an emphasis on its treatment.
Collapse
Affiliation(s)
- Ali Saeedi-Boroujeni
- Department of Microbiology, School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Daryush Purrahman
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Shojaeian
- grid.411950.80000 0004 0611 9280Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Łukasz A. Poniatowski
- grid.491786.50000 0001 0211 9062Department of Neurosurgery, Dietrich-Bonhoeffer-Klinikum, Neubrandenburg, Germany
| | - Fatemeh Rafiee
- grid.469309.10000 0004 0612 8427Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Science, Zanjan, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- grid.411230.50000 0000 9296 6873Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran ,grid.411230.50000 0000 9296 6873Clinical Research Development Unit, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
20
|
Gospodinova KO, Olsen D, Kaas M, Anderson SM, Phillips J, Walker RM, Bermingham ML, Payne AL, Giannopoulos P, Pandya D, Spires-Jones TL, Abbott CM, Porteous DJ, Glerup S, Evans KL. Loss of SORCS2 is Associated with Neuronal DNA Double-Strand Breaks. Cell Mol Neurobiol 2023; 43:237-249. [PMID: 34741697 PMCID: PMC9813074 DOI: 10.1007/s10571-021-01163-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/29/2021] [Indexed: 01/09/2023]
Abstract
SORCS2 is one of five proteins that constitute the Vps10p-domain receptor family. Members of this family play important roles in cellular processes linked to neuronal survival, differentiation and function. Genetic and functional studies implicate SORCS2 in cognitive function, as well as in neurodegenerative and psychiatric disorders. DNA damage and DNA repair deficits are linked to ageing and neurodegeneration, and transient neuronal DNA double-strand breaks (DSBs) also occur as a result of neuronal activity. Here, we report a novel role for SORCS2 in DSB formation. We show that SorCS2 loss is associated with elevated DSB levels in the mouse dentate gyrus and that knocking out SORCS2 in a human neuronal cell line increased Topoisomerase IIβ-dependent DSB formation and reduced neuronal viability. Neuronal stimulation had no impact on levels of DNA breaks in vitro, suggesting that the observed differences may not be the result of aberrant neuronal activity in these cells. Our findings are consistent with studies linking the VPS10 receptors and DNA damage to neurodegenerative conditions.
Collapse
Affiliation(s)
- Katerina O. Gospodinova
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Ditte Olsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Susan M. Anderson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Jonathan Phillips
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Rosie M. Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK ,Present Address: University of Edinburgh, Chancellor’s Building, 49, Edinburgh, EH16 4SB UK
| | - Mairead L. Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Abigail L. Payne
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Panagiotis Giannopoulos
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Divya Pandya
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Tara L. Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Catherine M. Abbott
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| |
Collapse
|
21
|
Knockdown of sortilin improves the neurological injury and regional cerebral blood flow in rats after subarachnoid hemorrhage. Neuroreport 2022; 33:697-704. [DOI: 10.1097/wnr.0000000000001833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Ertaş S, Ünver G, Gonca Okumuş Z, Yılmaz S, Dağ İ, Turhan U. Maternal serum Sortilin-1 level as a potential biomarker for intrahepatic cholestasis of pregnancy. Gynecol Endocrinol 2022; 38:935-938. [PMID: 36068972 DOI: 10.1080/09513590.2022.2119957] [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: 01/18/2023] Open
Abstract
Objective: Intrahepatic cholestasis of pregnancy (ICP) is the most common liver disease related to pregnancy in women. Sortilin-1 is a sorting receptor belonging to the vacuolar protein sorting 10 (Vps10p) domain family, and recent studies have shown that Sortilin-1 has a distinct role in the pathogenesis of biliary fibrosis and cirrhosis. We aimed to evaluate maternal serum Sortilin-1 level as a potential biomarker in pregnant women with intrahepatic cholestasis.Materials and methods: A prospective observational cohort study was conducted. We enrolled 80 pregnant women, 49 with the diagnosis of intrahepatic cholestasis of pregnancy and 31 healthy controls. Then, we measured maternal serum Sortilin-1 levels using an enzyme-linked immunosorbent assay method and compared them between groups.Results: The mean Sortilin-1 level in the ICP group was higher than control group (3.3 ± 1.7 ng/mL vs. 2.0 ± 0.6 ng/mL, respectively, p < .001). The receiver operating characteristic curve (ROC) analysis based on maternal serum Sortilin-1 levels to predict the presence of ICP was 85.3% controls [area under the curve (AUC), 0.853; 95% CI, 0.738-0.938, p < .001]. The optimal cutoff value of Sortilin-1 was 2.24 ng/mL (71.4% sensitivity and 74.2% specificity) to detect intrahepatic cholestasis of pregnancy.Conclusion: Elevated maternal serum Sortilin-1 levels are associated with ICP and can be used as a disease biomarker. Sortilin-1 levels can be combined with total bile acids, transaminases, and blood coagulation profile in the follow-up of ICP.
Collapse
Affiliation(s)
- Sinem Ertaş
- VKV American Hospital, Women's Health Center, İstanbul, Turkey
| | - Gökhan Ünver
- Samsun Training and Research Hospital, Samsun, Turkey
| | | | | | | | | |
Collapse
|
23
|
Mitok KA, Keller MP, Attie AD. Sorting through the extensive and confusing roles of sortilin in metabolic disease. J Lipid Res 2022; 63:100243. [PMID: 35724703 PMCID: PMC9356209 DOI: 10.1016/j.jlr.2022.100243] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023] Open
Abstract
Sortilin is a post-Golgi trafficking receptor homologous to the yeast vacuolar protein sorting receptor 10 (VPS10). The VPS10 motif on sortilin is a 10-bladed β-propeller structure capable of binding more than 50 proteins, covering a wide range of biological functions including lipid and lipoprotein metabolism, neuronal growth and death, inflammation, and lysosomal degradation. Sortilin has a complex cellular trafficking itinerary, where it functions as a receptor in the trans-Golgi network, endosomes, secretory vesicles, multivesicular bodies, and at the cell surface. In addition, sortilin is associated with hypercholesterolemia, Alzheimer's disease, prion diseases, Parkinson's disease, and inflammation syndromes. The 1p13.3 locus containing SORT1, the gene encoding sortilin, carries the strongest association with LDL-C of all loci in human genome-wide association studies. However, the mechanism by which sortilin influences LDL-C is unclear. Here, we review the role sortilin plays in cardiovascular and metabolic diseases and describe in detail the large and often contradictory literature on the role of sortilin in the regulation of LDL-C levels.
Collapse
Affiliation(s)
- Kelly A Mitok
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
24
|
Dong F, Wu C, Jiang W, Zhai M, Li H, Zhai L, Zhang X. Cryo-EM structure studies of the human VPS10 domain-containing receptor SorCS3. Biochem Biophys Res Commun 2022; 624:89-94. [DOI: 10.1016/j.bbrc.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
|
25
|
Cryo-EM structures reveal distinct apo conformations of sortilin-related receptor SORLA. Biochem Biophys Res Commun 2022; 600:75-79. [DOI: 10.1016/j.bbrc.2022.01.108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 01/26/2022] [Indexed: 01/12/2023]
|
26
|
Binkle L, Klein M, Borgmeyer U, Kuhl D, Hermey G. The adaptor protein PICK1 targets the sorting receptor SorLA. Mol Brain 2022; 15:18. [PMID: 35183222 PMCID: PMC8858569 DOI: 10.1186/s13041-022-00903-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/07/2022] [Indexed: 11/26/2022] Open
Abstract
SorLA is a member of the Vps10p-domain (Vps10p-D) receptor family of type-I transmembrane proteins conveying neuronal endosomal sorting. The extracellular/luminal moiety of SorLA has a unique mosaic domain composition and interacts with a large number of different and partially unrelated ligands, including the amyloid precursor protein as well as amyloid-β. Several studies support a strong association of SorLA with sporadic and familial forms of Alzheimer’s disease (AD). Although SorLA seems to be an important factor in AD, the large number of different ligands suggests a role as a neuronal multifunctional receptor with additional intracellular sorting capacities. Therefore, understanding the determinants of SorLA’s subcellular targeting might be pertinent for understanding neuronal endosomal sorting mechanisms in general. A number of cytosolic adaptor proteins have already been demonstrated to determine intracellular trafficking of SorLA. Most of these adaptors and several ligands of the extracellular/luminal moiety are shared with the Vps10p-D receptor Sortilin. Although SorLA and Sortilin show both a predominant intracellular and endosomal localization, they are targeted to different endosomal compartments. Thus, independent adaptor proteins may convey their differential endosomal targeting. Here, we hypothesized that Sortilin and SorLA interact with the cytosolic adaptors PSD95 and PICK1 which have been shown to bind the Vps10p-D receptor SorCS3. We observed only an interaction for SorLA and PICK1 in mammalian-two-hybrid, pull-down and cellular recruitment experiments. We demonstrate by mutational analysis that the C-terminal minimal PDZ domain binding motif VIA of SorLA mediates the interaction. Moreover, we show co-localization of SorLA and PICK1 at vesicular structures in primary neurons. Although the physiological role of the interaction between PICK1 and SorLA remains unsolved, our study suggests that PICK1 partakes in regulating SorLA’s intracellular itinerary.
Collapse
|
27
|
Bellomo TR, Bone WP, Chen BY, Gawronski KAB, Zhang D, Park J, Levin M, Tsao N, Klarin D, Lynch J, Assimes TL, Gaziano JM, Wilson PW, Cho K, Vujkovic M, O’Donnell CJ, Chang KM, Tsao PS, Rader DJ, Ritchie MD, Damrauer SM, Voight BF. Multi-Trait Genome-Wide Association Study of Atherosclerosis Detects Novel Pleiotropic Loci. Front Genet 2022; 12:787545. [PMID: 35186008 PMCID: PMC8847690 DOI: 10.3389/fgene.2021.787545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Although affecting different arterial territories, the related atherosclerotic vascular diseases coronary artery disease (CAD) and peripheral artery disease (PAD) share similar risk factors and have shared pathobiology. To identify novel pleiotropic loci associated with atherosclerosis, we performed a joint analysis of their shared genetic architecture, along with that of common risk factors. Using summary statistics from genome-wide association studies of nine known atherosclerotic (CAD, PAD) and atherosclerosis risk factors (body mass index, smoking initiation, type 2 diabetes, low density lipoprotein, high density lipoprotein, total cholesterol, and triglycerides), we perform 15 separate multi-trait genetic association scans which resulted in 25 novel pleiotropic loci not yet reported as genome-wide significant for their respective traits. Colocalization with single-tissue eQTLs identified candidate causal genes at 14 of the detected signals. Notably, the signal between PAD and LDL-C at the PCSK6 locus affects PCSK6 splicing in human liver tissue and induced pluripotent derived hepatocyte-like cells. These results show that joint analysis of related atherosclerotic disease traits and their risk factors allowed identification of unified biology that may offer the opportunity for therapeutic manipulation. The signal at PCSK6 represent possible shared causal biology where existing inhibitors may be able to be leveraged for novel therapies.
Collapse
Affiliation(s)
- Tiffany R. Bellomo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - William P. Bone
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Brian Y. Chen
- School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, United States
| | | | - David Zhang
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
| | - Joseph Park
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael Levin
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Noah Tsao
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
| | - Derek Klarin
- VA Boston Healthcare System, Boston, MA, United States
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, United States
- Division of Vascular Surgery and Endovascular Therapy, University of Florida School of Medicine, Gainesville, FL, United States
- Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Julie Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, United States
- University of Massachusetts College of Nursing and Health Sciences, Boston, MA, United States
| | - Themistocles L. Assimes
- VA Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - J. Michael Gaziano
- VA Boston Healthcare System, Boston, MA, United States
- Massachusetts Veterans Epidemiology Research and Information Center, Veterans Affairs Boston Healthcare System, Boston, MA, United States
- Department of Medicine, Brigham Women’s Hospital, Boston, MA, United States
| | - Peter W. Wilson
- Atlanta VA Medical Center, Decatur, GA, United States
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Kelly Cho
- VA Boston Healthcare System, Boston, MA, United States
- Department of Medicine, Brigham Women’s Hospital, Boston, MA, United States
| | - Marijana Vujkovic
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher J. O’Donnell
- VA Boston Healthcare System, Boston, MA, United States
- Department of Medicine, Brigham Women’s Hospital, Boston, MA, United States
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Philip S. Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, United States
- Department of Medicine, Stanford University, Stanford, CA, United States
| | - Daniel J. Rader
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, United States
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Benjamin F. Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
28
|
Xu J, Shen CJ, Ooi JD, Tang YS, Xiao Z, Yuan QJ, Zhong Y, Zhou QL. Serum Sortilin Is Associated with Coronary Artery Calcification and Cardiovascular and Cerebrovascular Events in Maintenance Hemodialysis Patients. KIDNEY DISEASES (BASEL, SWITZERLAND) 2021; 7:503-513. [PMID: 34901196 PMCID: PMC8613630 DOI: 10.1159/000517304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/16/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To analyze the role of serum sortilin in coronary artery calcification (CAC) and cardiovascular and cerebrovascular events (CCE) in maintenance hemodialysis (MHD) patients. METHODS One hundred eleven patients with MHD ≥3 months were included in this study. The general data, clinical features, hematological data, and medication history of the patients were recorded. Eighty-five cases were examined by vascular color Doppler ultrasound, cardiac color Doppler ultrasound, lateral lumbar radiography, and coronary artery calcification score. The patients were followed up for a median time of 45 months. The primary endpoint was CCE or death from a vascular event, and the role of sortilin in this process was analyzed. RESULTS Among 85 MHD patients, 51 cases (60.00%) had different degrees of CAC. There were significant differences in diabetes, dialysis time, serum phosphorus, calcium-phosphorus product, medical history of phosphate binders, sortilin, and carotid artery plaque between 4 different degrees of calcification groups (p < 0.05). Logistic regression analysis showed that diabetes (OR = 5.475; 95% CI: 1.794-16.71, p = 0.003), calcium-phosphorus product (OR = 2.953; 95% CI: 1.198-7.279, p = 0.019), and sortilin (OR = 1.475 per 100 pg/mL; 95% CI: 1.170-1.858, p = 0.001) were independent risk factors for CAC. During the follow-up, 28 cases of 111 patients (25.23%) suffered from CCE. There were significant differences in CCE between mild, moderate, and severe CAC groups and noncalcification groups (p < 0.05). Cox regression analysis showed that diabetes mellitus (HR 3.424; 95% CI: 1.348-8.701, p = 0.010), CAC (HR 5.210; 95% CI: 1.093-24.83, p = 0.038), and serum sortilin (HR = 8.588; 95% CI: 1.919-38.43, p = 0.005) were independent risk factors for CCE. Besides, we proposed a cutoff value of 418 pg/mL for serum sortilin level, which was able to predict the occurrence of CCE with 75.0% sensitivity and 71.9% specificity. The area under the curve was 0.778 (95% CI: 0.673-0.883). CONCLUSION Sortilin is newly found to be independently associated with CAC and CCE in MHD patients.
Collapse
Affiliation(s)
- Jie Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Chan-Juan Shen
- Department of Hematology, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, China
| | - Joshua D. Ooi
- Centre for Inflammatory Diseases, Monash University, Clayton, Victoria, Australia
| | - Yang-Shuo Tang
- Department of Ultrasonography, Xiangya Hospital, Central South University, Changsha, China
| | - Zhou Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiong-Jing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Zhong
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiao-Ling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Retromer dependent changes in cellular homeostasis and Parkinson's disease. Essays Biochem 2021; 65:987-998. [PMID: 34528672 PMCID: PMC8709886 DOI: 10.1042/ebc20210023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/18/2022]
Abstract
To date, mechanistic treatments targeting the initial cause of Parkinson's disease (PD) are limited due to the underlying biological cause(s) been unclear. Endosomes and their associated cellular homeostasis processes have emerged to have a significant role in the pathophysiology associated with PD. Several variants within retromer complex have been identified and characterised within familial PD patients. The retromer complex represents a key sorting platform within the endosomal system that regulates cargo sorting that maintains cellular homeostasis. In this review, we summarise the current understandings of how PD-associated retromer variants disrupt cellular trafficking and how the retromer complex can interact with other PD-associated genes to contribute to the disease progression.
Collapse
|
30
|
A Particular SORL1 Micro-haplotype May Prevent Severe Liver Disease in a French Cohort of Alpha 1-Antitrypsin-deficient Children. J Pediatr Gastroenterol Nutr 2021; 73:e68-e72. [PMID: 33720088 DOI: 10.1097/mpg.0000000000003125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The presence of modifier genes is now well recognized in severe liver disease outcome associated with alpha-1-antitrypsin deficiency (A1ATD) but their identification remains to be fully elucidated. To address this goal, we performed a candidate gene study with the SORL1 gene, already identified as risk gene in early-onset Alzheimer Disease families. A particular SORL1 micro-haplotype constituted with 3 SNPs (wild-type form TTG) was genotyped on 86 ZZ A1ATD children issued from 66 families. Interestingly, the mutated forms of this micro-haplotype (CAT most of the time) were associated with lower occurrence of severe liver disease and in cellulo studies showed that SORL1 influences Z-A1ATD cellular toxicity and biogenesis. These data suggest that the mutated CAT form of SORL1 micro-haplotype may partly prevent from severe liver disease in A1ATD children. Overall, these findings support a replication study on an independent cohort and additional in cellulo studies to confirm these promising results.
Collapse
|
31
|
Zhao Y, Tang F, Lee D, Xiong WC. Expression of Low Level of VPS35-mCherry Fusion Protein Diminishes Vps35 Depletion Induced Neuron Terminal Differentiation Deficits and Neurodegenerative Pathology, and Prevents Neonatal Death. Int J Mol Sci 2021; 22:8394. [PMID: 34445101 PMCID: PMC8395035 DOI: 10.3390/ijms22168394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
Vps35 (vacuolar protein sorting 35) is a key component of retromer that consists of Vps35, Vps26, and Vps29 trimers, and sortin nexin dimers. Dysfunctional Vps35/retromer is believed to be a risk factor for development of various neurodegenerative diseases. Vps35Neurod6 mice, which selectively knock out Vps35 in Neurod6-Cre+ pyramidal neurons, exhibit age-dependent impairments in terminal differentiation of dendrites and axons of cortical and hippocampal neurons, neuro-degenerative pathology (i.e., increases in P62 and Tdp43 (TAR DNA-binding protein 43) proteins, cell death, and reactive gliosis), and neonatal death. The relationships among these phenotypes and the underlying mechanisms remain largely unclear. Here, we provide evidence that expression of low level of VPS35-mCherry fusion protein in Vps35Neurod6 mice could diminish the phenotypes in an age-dependent manner. Specifically, we have generated a conditional transgenic mouse line, LSL-Vps35-mCherry, which expresses VPS35-mCherry fusion protein in a Cre-dependent manner. Crossing LSL-Vps35-mCherry with Vps35Neurod6 to obtain TgVPS35-mCherry, Vps35Neurod6 mice prevent the neonatal death and diminish the dendritic morphogenesis deficit and gliosis at the neonatal, but not the adult age. Further studies revealed that the Vps35-mCherry transgene expression was low, and the level of Vps35 mRNA comprised only ~5-7% of the Vps35 mRNA of control mice. Such low level of VPS35-mCherry could restore the amount of other retromer components (Vps26a and Vps29) at the neonatal age (P14). Importantly, the neurodegenerative pathology presented in the survived adult TgVps35-mCherry; Vps35Neurod6 mice. These results demonstrate the sufficiency of low level of VPS35-mCherry fusion protein to diminish the phenotypes in Vps35Neurod6 mice at the neonatal age, verifying a key role of neuronal Vps35 in stabilizing retromer complex proteins, and supporting the view for Vps35 as a potential therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (Y.Z.); (D.L.)
| | - Fulei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Daehoon Lee
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (Y.Z.); (D.L.)
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (Y.Z.); (D.L.)
| |
Collapse
|
32
|
Wang Y, Wang J, Zuo YC, Jiang J, Tu T, Yan XX, Liu F. Elevation of CSF Sortilin Following Subarachnoid Hemorrhage in Patients and Experimental Model Rats. Neuroscience 2021; 470:23-36. [PMID: 34273414 DOI: 10.1016/j.neuroscience.2021.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 10/20/2022]
Abstract
Subarachnoid hemorrhage (SAH) can cause acute neuronal injury and chronic neurocognitive deficits; biomarkers reflecting its associated neuronal injury are of potential prognostic value. Sortilin, a member of the vacuolar protein sorting 10p (Vps10p) family, is enriched in neurons and is likely involved in neurodegenerative diseases. Here, we explored sortilin in the cerebrospinal fluid (CSF) as a potential biomarker for early neuronal injury after SAH. Sortilin levels in the CSF of SAH patients (n = 11) and controls (n = 6) were analyzed by immunoblot. SAH rats surviving 3-72 h (h) were evaluated neurologically, with their brain and CSF samples examined histologically and biochemically. Sortilin protein ~100 kDa was detected in the CSF from SAH patients only, with its levels correlated to Hunt-Hess scale. Rats in the SAH groups showed poorer Garcia score and beam balancing capability than sham controls. Sortilin ~100 kDa was detectable in the CSF of the SAH, but not sham, animals. Levels of sortilin ~100 kDa and fragments ~40 kDa in cortical lysates were elevated in the SAH relative to control rats. Levels of cortical glial fibrillary acidic protein (GFAP) were also elevated in the SAH rats. In immunohistochemistry, the pattern of sortilin labeling in the brain was largely comparable between the SAH and control rats, whereas an increased astrocytic GFAP immunolabeling was evident in the former. Together, these results suggest that SAH can cause an early and remarkable rise of sortilin products in CSF, likely reflecting neuronal change. Sortilin could be further explored as a potential biomarker in some brain disorders.
Collapse
Affiliation(s)
- Yiping Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Jikai Wang
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Yu-Chun Zuo
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, Hunan 410008, China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Tian Tu
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, Hunan 410013, China.
| | - Fei Liu
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong 519000, China.
| |
Collapse
|
33
|
Qiu G, Zhu FQ, Xu C. Identification of two pathogenic mutations in SORL1 in early-onset Alzheimer's disease. J Clin Neurosci 2021; 89:243-248. [PMID: 34119275 DOI: 10.1016/j.jocn.2021.05.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 10/21/2022]
Abstract
The sortilin-related receptor 1 (SORL1) gene has been the subject of many studies focusing on frequent polymorphisms, which is associated with increased risk for Alzheimer's Disease (AD). By whole-exome sequencing (WES), we identified two pathogenic missense mutations c.579C > G (p.F193L) and c.1397A > G (p.N466S) in SORL1. The two mutations were located in the same protein domain, and the two unrelated probands both had an onset of memory problems at less than 65 years of age, but their clinical manifestations and cranial imaging are different. The protein structure and function affected by these mutations were predicted using bioinformatics analysis, which suggested they were pathogenic. 3D protein structural analysis revealed that these amino acid substitutions might result in instability of protein structure and adverse intramolecular interactions. These findings suggest that both F193L and N466S should be thought as potential causative mutations in early-onset Alzheimer's disease (EOAD) patients. Further functional studies are warranted to evaluate their roles in the pathogenesis of AD.
Collapse
Affiliation(s)
- Guozhen Qiu
- The Third Affiliated Hospital of Shenzhen University, Cognitive Impairment Ward of Neurology Department, Shenzhen, Guangdong Province, China.
| | - Fei-Qi Zhu
- The Third Affiliated Hospital of Shenzhen University, Cognitive Impairment Ward of Neurology Department, Shenzhen, Guangdong Province, China
| | - Chunyan Xu
- The Third Affiliated Hospital of Shenzhen University, Cognitive Impairment Ward of Neurology Department, Shenzhen, Guangdong Province, China
| |
Collapse
|
34
|
Herrera GA, del Pozo-Yauner L, Teng J, Zeng C, Shen X, Moriyama T, Ramirez Alcantara V, Liu B, Turbat-Herrera EA. Glomerulopathic Light Chain-Mesangial Cell Interactions: Sortilin-Related Receptor (SORL1) and Signaling. Kidney Int Rep 2021; 6:1379-1396. [PMID: 34013116 PMCID: PMC8116754 DOI: 10.1016/j.ekir.2021.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/08/2021] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Deciphering the intricacies of the interactions of glomerulopathic Ig light chains with mesangial cells is key to delineate signaling events responsible for the mesangial pathologic alterations that ensue. METHODS Human mesangial cells, caveolin 1 (CAV1), wild type (WT) ,and knockout (KO), were incubated with glomerulopathic light chains purified from the urine of patients with light chain-associated (AL) amyloidosis or light chain deposition disease. Associated signaling events induced by surface interactions of glomerulopathic light chains with caveolins and other membrane proteins, as well as the effect of epigallocatechin-3-gallate (EGCG) on the capacity of mesangial cells to intracellularly process AL light chains were investigated using a variety of techniques, including chemical crosslinking with mass spectroscopy, immunofluorescence, and ultrastructural immunolabeling. RESULTS Crosslinking experiments provide evidence suggesting that sortilin-related receptor (SORL1), a transmembrane sorting receptor that regulates cellular trafficking of proteins, is a component of the receptor on mesangial cells for glomerulopathic light chains. Colocalization of glomerulopathic light chains with SORL1 in caveolae and also in lysosomes when light chain internalization occurred, was documented using double immunofluorescence and immunogold labeling ultrastructural techniques. It was found that EGCG directly blocks c-Fos cytoplasmic to nuclei signal translocation after interactions of AL light chains with mesangial cells, resulting in a decrease in amyloid formation. CONCLUSION Our findings document for the first time a role for SORL1 linked to glomerular pathology and signaling events that take place when certain monoclonal light chains interact with mesangial cells. This finding may lead to novel therapies for treating renal injury caused by glomerulopathic light chains.
Collapse
Affiliation(s)
- Guillermo A. Herrera
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
- Correspondence: Guillermo A. Herrera, Department of Pathology, University of South Alabama, College of Medicine, 2451 USA Medical Center Drive, Mobile, Alabama 36617, USA.
| | - Luis del Pozo-Yauner
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Jiamin Teng
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Chun Zeng
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Xinggui Shen
- Louisiana State University, Health Sciences Center, Shreveport, Louisiana, USA
| | - Takahito Moriyama
- Department of Medicine, Kidney Center, Tokyo Women’s Medical University, Tokyo, Japan
| | | | - Bing Liu
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Elba A. Turbat-Herrera
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
- Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
35
|
Yang J, Ma Q, Dincheva I, Giza J, Jing D, Marinic T, Milner TA, Rajadhyaksha A, Lee FS, Hempstead BL. SorCS2 is required for social memory and trafficking of the NMDA receptor. Mol Psychiatry 2021; 26:927-940. [PMID: 31988435 DOI: 10.1038/s41380-020-0650-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/21/2019] [Accepted: 12/02/2019] [Indexed: 11/09/2022]
Abstract
Social memory processing requires functional CA2 neurons, however the specific mechanisms that regulate their activity are poorly understood. Here, we document that SorCS2, a member of the family of the Vps10 family of sorting receptors, is highly expressed in pyramidal neurons of CA2, as well as ventral CA1, a circuit implicated in social memory. SorCS2 specifically localizes to the postsynaptic density and endosomes within dendritic spines of CA2 neurons. We have discovered that SorCS2 is a selective regulator of NMDA receptor surface trafficking in hippocampal neurons, without altering AMPA receptor trafficking. In addition, SorCS2 regulates dendritic spine density in CA2 neurons where SorCS2 expression is enriched, but not in dorsal CA1 neurons, which normally express very low levels of this protein. To specifically test the role of SorCS2 in behavior, we generated a novel SorCS2-deficient mouse, and identify a significant social memory deficit, with no change in sociability, olfaction, anxiety, or several hippocampal-dependent behaviors. Mutations in sorCS2 have been associated with bipolar disease, schizophrenia, and attention deficient-hyperactivity disorder, and abnormalities in social memory are core components of these neuropsychiatric conditions. Thus, our findings provide a new mechanism for social memory formation, through regulating synaptic receptor trafficking in pyramidal neurons by SorCS2.
Collapse
Affiliation(s)
- Jianmin Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China. .,Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Qian Ma
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Iva Dincheva
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Joanna Giza
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA.,Department of Science, Borough of Manhattan Community College, The City University of New York, 199 Chambers Street N699J, New York, NY, 10007, USA
| | - Deqiang Jing
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Tina Marinic
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.,Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | | | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Barbara L Hempstead
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
36
|
Ghaemimanesh F, Mehravar M, Milani S, Poursani EM, Saliminejad K. The multifaceted role of sortilin/neurotensin receptor 3 in human cancer development. J Cell Physiol 2021; 236:6271-6281. [PMID: 33634506 DOI: 10.1002/jcp.30344] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/20/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022]
Abstract
Sortilin (also known as neurotensin receptor 3) is a multitasking protein implicated in numerous pathophysiological processes, including cancer development, cardiovascular impairment, Alzheimer-type dementia, and depression. Although the definitive role of sortilin in human solid and hematological malignancies has been evidenced, few articles reviewed the task. The aim of the current review is to unravel the mechanisms by which sortilin controls oncogenicity and cancer progression; and also to summarize and discuss the original data obtained from international research laboratories on this topic. Questions on how sortilin is involving in the impairment of cell junctions, in exosomes composition and release, as well as in the regulation of epidermal growth factor receptor trafficking are also responded. In addition, we provide a special focus on the regulatory role of sortilin in signal transduction by either neurotrophins or neurotensin in normal and malignant cells. The relevance of sortilin with normal and cancer stem cells is also discussed. The last section provides a general overview of sortilin applications as a diagnostic and prognostic biomarker in the context of cancer detection. Finally, we comment on the future research aspects in which the field of cancer diagnosis, prognosis, and therapy might be developed.
Collapse
Affiliation(s)
- Fatemeh Ghaemimanesh
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Majid Mehravar
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Saeideh Milani
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ensieh M Poursani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kioomars Saliminejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
37
|
Miyakawa S, Sakuma H, Warude D, Asanuma S, Arimura N, Yoshihara T, Tavares D, Hata A, Ida K, Hori Y, Okuzono Y, Yamamoto S, Iida K, Shimizu H, Kondo S, Sato S. Anti-sortilin1 Antibody Up-Regulates Progranulin via Sortilin1 Down-Regulation. Front Neurosci 2020; 14:586107. [PMID: 33384578 PMCID: PMC7770147 DOI: 10.3389/fnins.2020.586107] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Progranulin (PGRN) haploinsufficiency associated with loss-of-function mutations in the granulin gene causes frontotemporal dementia (FTD). This suggests that increasing PGRN levels could have promising therapeutic implications for patients carrying GRN mutations. In this study, we explored the therapeutic potential of sortilin1 (SORT1), a clearance receptor of PGRN, by generating and characterizing monoclonal antibodies against SORT1. Anti-SORT1 monoclonal antibodies were generated by immunizing Sort1 knockout mice with SORT1 protein. The antibodies were classified into 7 epitope bins based on their competitive binding to the SORT1 protein and further defined by epitope bin-dependent characteristics, including SORT1-PGRN blocking, SORT1 down-regulation, and binding to human and mouse SORT1. We identified a positive correlation between PGRN up-regulation and SORT1 down-regulation. Furthermore, we also characterized K1-67 antibody via SORT1 down-regulation and binding to mouse SORT1 in vivo and confirmed that K1-67 significantly up-regulated PGRN levels in plasma and brain interstitial fluid of mice. These data indicate that SORT1 down-regulation is a key mechanism in increasing PGRN levels via anti-SORT1 antibodies and suggest that SORT1 is a potential target to correct PGRN reduction, such as that in patients with FTD caused by GRN mutation.
Collapse
Affiliation(s)
- Shuuichi Miyakawa
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroyuki Sakuma
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Dnyaneshwar Warude
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Satomi Asanuma
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Naoto Arimura
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoki Yoshihara
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Daniel Tavares
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Akito Hata
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Koh Ida
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Yuri Hori
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuumi Okuzono
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Koichi Iida
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hisao Shimizu
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shinichi Kondo
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shuji Sato
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
38
|
VPS10P Domain Receptors: Sorting Out Brain Health and Disease. Trends Neurosci 2020; 43:870-885. [DOI: 10.1016/j.tins.2020.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
|
39
|
Wu Y, Cao H, Baranova A, Huang H, Li S, Cai L, Rao S, Dai M, Xie M, Dou Y, Hao Q, Zhu L, Zhang X, Yao Y, Zhang F, Xu M, Wang Q. Multi-trait analysis for genome-wide association study of five psychiatric disorders. Transl Psychiatry 2020; 10:209. [PMID: 32606422 PMCID: PMC7326916 DOI: 10.1038/s41398-020-00902-6] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 06/06/2020] [Accepted: 06/16/2020] [Indexed: 02/05/2023] Open
Abstract
We conducted a cross-trait meta-analysis of genome-wide association study on schizophrenia (SCZ) (n = 65,967), bipolar disorder (BD) (n = 41,653), autism spectrum disorder (ASD) (n = 46,350), attention deficit hyperactivity disorder (ADHD) (n = 55,374), and depression (DEP) (n = 688,809). After the meta-analysis, the number of genomic loci increased from 14 to 19 in ADHD, from 3 to 10 in ASD, from 45 to 57 in DEP, from 8 to 54 in BD, and from 64 to 87 in SCZ. We observed significant enrichment of overlapping genes among different disorders and identified a panel of cross-disorder genes. A total of seven genes were found being commonly associated with four out of five psychiatric conditions, namely GABBR1, GLT8D1, HIST1H1B, HIST1H2BN, HIST1H4L, KCNB1, and DCC. The SORCS3 gene was highlighted due to the fact that it was involved in all the five conditions of study. Analysis of correlations unveiled the existence of two clusters of related psychiatric conditions, SCZ and BD that were separate from the other three traits, and formed another group. Our results may provide a new insight for genetic basis of the five psychiatric disorders.
Collapse
Affiliation(s)
- Yulu Wu
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hongbao Cao
- Department of Psychiatry, First Clinical Medical College/First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University (GMU), Fairfax, VA, USA
- Research Centre for Medical Genetics, Moscow, Russia
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sheng Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, 1954 Huashan Road, Xuhui, 200030, Shanghai, China
| | - Lei Cai
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, 1954 Huashan Road, Xuhui, 200030, Shanghai, China
| | - Shuquan Rao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Minhan Dai
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Min Xie
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yikai Dou
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qinjian Hao
- The Center of Gerontology and Geriatrics, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ling Zhu
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiangrong Zhang
- Department of Geriatric Psychiatry, Nanjing Brain Hospital, Affiliated to Nanjing Medical University, Nanjing, China
| | - Yin Yao
- Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, 264 Guangzhou Road, Nanjing, China.
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiaotong University, 1954 Huashan Road, Xuhui, 200030, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 South Wanping Road, Xuhui, 200030, Shanghai, China.
| | - Qiang Wang
- Mental Health Center and Psychiatric Laboratory, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
- West China Brain Research Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
40
|
Monti G, Jensen ML, Mehmedbasic A, Jørgensen MM, Holm IE, Barkholt P, Zole E, Vægter CB, Vorum H, Nyengaard JR, Andersen OM. SORLA Expression in Synaptic Plexiform Layers of Mouse Retina. Mol Neurobiol 2020; 57:3106-3117. [PMID: 32472518 DOI: 10.1007/s12035-020-01946-x] [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: 12/18/2019] [Accepted: 05/13/2020] [Indexed: 11/25/2022]
Abstract
Sorting protein-related receptor containing LDLR class A repeats (SORLA; also known as LR11) exerts intraneuronal trafficking functions in the central nervous system. Recently, involvement of SORLA in retinogenesis was proposed, but no studies have examined yet in detail the expression pattern of this sorting receptor in the retina. Here, we provide a spatio-temporal characterization of SORL1 mRNA and its translational product SORLA in the postnatal mouse retina. Using stereological analysis, we confirmed previous studies showing that receptor depletion in knockout mice significantly reduces the number of cells in the inner nuclear layer (INL), suggesting that functional SORLA expression is essential for the development of this retinal strata. qPCR and Western blot analyses showed that SORL1/SORLA expression peaks at postnatal day 15, just after eye opening. Interestingly, we found that transcripts are somatically located in several neuronal populations residing in the INL and the ganglion cell layer, whereas SORLA protein is also present in the synaptic plexiform layers. In line with receptor expression in dendritic terminals, we found delayed stratification of the inner plexiform layer in knockout mice, indicating an involvement of SORLA in neuronal connectivity. Altogether, these data suggest a novel role of SORLA in synaptogenesis. Receptor dysfunctions may be implicated in morphological and functional impairments of retinal inner layer formation associated with eye disorders.
Collapse
Affiliation(s)
- Giulia Monti
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Marianne L Jensen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Arnela Mehmedbasic
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Margarita Melnikova Jørgensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Ida E Holm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Pernille Barkholt
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Egija Zole
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Christian B Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark
| | - Henrik Vorum
- Department of Ophthalmology, Aalborg University Hospital, Hobrovej 18-22, DK-9000, Aalborg, Denmark
| | - Jens R Nyengaard
- Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Olav M Andersen
- Danish Research Institute of Translational Neuroscience (DANDRITE) Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
41
|
Konijnenberg E, Tijms BM, Gobom J, Dobricic V, Bos I, Vos S, Tsolaki M, Verhey F, Popp J, Martinez-Lage P, Vandenberghe R, Lleó A, Frölich L, Lovestone S, Streffer J, Bertram L, Blennow K, Teunissen CE, Veerhuis R, Smit AB, Scheltens P, Zetterberg H, Visser PJ. APOE ε4 genotype-dependent cerebrospinal fluid proteomic signatures in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2020; 12:65. [PMID: 32460813 PMCID: PMC7254647 DOI: 10.1186/s13195-020-00628-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/06/2020] [Indexed: 02/04/2023]
Abstract
Background Aggregation of amyloid β into plaques in the brain is one of the earliest pathological events in Alzheimer’s disease (AD). The exact pathophysiology leading to dementia is still uncertain, but the apolipoprotein E (APOE) ε4 genotype plays a major role. We aimed to identify the molecular pathways associated with amyloid β aggregation using cerebrospinal fluid (CSF) proteomics and to study the potential modifying effects of APOE ε4 genotype. Methods We tested 243 proteins and protein fragments in CSF comparing 193 subjects with AD across the cognitive spectrum (65% APOE ε4 carriers, average age 75 ± 7 years) against 60 controls with normal CSF amyloid β, normal cognition, and no APOE ε4 allele (average age 75 ± 6 years). Results One hundred twenty-nine proteins (53%) were associated with aggregated amyloid β. APOE ε4 carriers with AD showed altered concentrations of proteins involved in the complement pathway and glycolysis when cognition was normal and lower concentrations of proteins involved in synapse structure and function when cognitive impairment was moderately severe. APOE ε4 non-carriers with AD showed lower expression of proteins involved in synapse structure and function when cognition was normal and lower concentrations of proteins that were associated with complement and other inflammatory processes when cognitive impairment was mild. Repeating analyses for 114 proteins that were available in an independent EMIF-AD MBD dataset (n = 275) showed that 80% of the proteins showed group differences in a similar direction, but overall, 28% effects reached statistical significance (ranging between 6 and 87% depending on the disease stage and genotype), suggesting variable reproducibility. Conclusions These results imply that AD pathophysiology depends on APOE genotype and that treatment for AD may need to be tailored according to APOE genotype and severity of the cognitive impairment.
Collapse
Affiliation(s)
- Elles Konijnenberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
| | - Johan Gobom
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
| | - Isabelle Bos
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Stephanie Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Thessaloniki, Macedonia, Greece
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Pablo Martinez-Lage
- Department of Neurology, Center for Research and Advanced Therapies, CITA-Alzheimer Foundation, San Sebastian, Spain
| | | | - Alberto Lleó
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, University of Heidelberg, Mannheim, Germany
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, UK.,Janssen R&D, Beerse, Belgium
| | - Johannes Streffer
- Early Clinical Neurology, UCB Biopharma SPRL, Braine-l'Alleud, Belgium.,Present Address: Janssen R&D, LLC, Beerse, Belgium
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany.,School of Public Health, Imperial College London, London, UK.,Department of Psychology, University of Oslo, Oslo, Norway
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Robert Veerhuis
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.,Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute, London, UK
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.,Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands.,Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Instutet, Stockholm, Sweden
| |
Collapse
|
42
|
Chen P, Xu L, Zhang J, Cai X, Yang Y, Yu J, Qiu J, Ge J, Yu K, Zhuang J. Up-Regulation of SorCS1, an Important Sorting Receptor, in the Retina of a Form-Deprivation Rat Model. Cell Mol Neurobiol 2020; 40:395-405. [PMID: 31605284 PMCID: PMC11449034 DOI: 10.1007/s10571-019-00740-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
Visually guided regulation is a sophisticated and active process, whereby sensory input helps to shape ocular development. Here, we sought to investigate the potential involvement of SorCS1, an important protein in synaptic transmission in neuron, in retinal development. A form-deprivation (FD) rat model was established. Ocular variations induced by FD were examined, including changes to eye axial length and retinal thickness. Scotopic electroretinogram (ERG) was used to examine retinal function. RD-PCR assays were screened for differentially expressed genes in FD rat eyes. Immunofluorescence staining identified the expression pattern and localization of SorCS1 in rat retina, with or without FD treatment. Additionally, primary retinal neural cells were cultured and incubated with or without a light-dark cycle, and western blot and real-time PCR assays were used to examine the expression of SorCS1. Retinal neural cells were treated with recombinant SorCS1 (h-SorCS1) coated with beads in serum-free conditions to test for effects on cellular physiology and expression of neurotransmitters involved in visual development. To monitor cell viability, a CCK8 assay was employed. Our data demonstrated that FD led to ocular axial elongation and retinal thinning. ERG tests showed FD impaired electrophysiological function in rat. An age-related expression pattern of SorCS1 was observed in the rat retina, and SorCS1 was significantly up-regulated in the FD rat retina. In addition, in vitro evidence suggested a strong correlation between light exposure and SorCS1 expression. Furthermore, treatment of retinal neural cells with h-SorCS1-beads promoted cell viability, neurite outgrowth, and up-regulation of inhibitory neurotransmitter expression, which implies that over-expression of SorCS1 may cause abnormal retinal development. Our findings suggest that SorCS1 is involved in the physiological processes of light/visually guided ocular growth.
Collapse
Affiliation(s)
- Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Lijun Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiaoxiao Cai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jingzhi Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
43
|
El Bitar F, Qadi N, Al Rajeh S, Majrashi A, Abdulaziz S, Majrashi N, Al Inizi M, Taher A, Al Tassan N. Genetic Study of Alzheimer's Disease in Saudi Population. J Alzheimers Dis 2020; 67:231-242. [PMID: 30636737 DOI: 10.3233/jad-180415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurological disorder associated with mental decline and dementia. Several studies focused on investigating the molecular basis of the disease that led to the identification of several causative genes and risk associated alleles. Replication of these studies and findings from different populations is very important. OBJECTIVE Molecular assessment of a cohort of 117 familial and sporadic AD cases from Saudi Arabia. METHODS Comprehensive screening for point mutations was carried out by direct sequencing of coding regions in the three known AD causative genes: PSEN1, PSEN2, APP, as well as the AD associated gene SORL1. All patients were also genotyped for APOE alleles. In silico 3D protein structure analysis was performed for two novel SORL1 variants. RESULTS We identified a total of eight potential pathogenic missense variants in all studied genes. Five of these variants were not previously reported including four in SORL1 (p.Val297Met, p.Arg1084Cys, p.Asp1100Asn, and p.Pro1213Ser) and one in APP (p.Glu380Lys). The frequency of APOE-ɛ4 allele was 21.37% of total investigated cases. In silico 3D protein structure analysis of two SORL1 novel missense variants (p.Pro1213Ser and p.Arg1084Cys) suggested that these variants may affect the folding of the proteins and disturb their structure. CONCLUSIONS Our comprehensive analysis of the open reading frame of the known genes have identified potential pathogenic rare variants in 18/117 cases. We found that point mutations in AD main genes (PSEN1, PSEN2, and APP) were underrepresented in our cohort of patients. Our results confirm involvement of SORL1 in familial and sporadic AD cases.
Collapse
Affiliation(s)
- Fadia El Bitar
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Najeeb Qadi
- Department of Neurosciences, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Saad Al Rajeh
- Al Habib Medical Center, Riyadh, Kingdom of Saudi Arabia
| | - Amna Majrashi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Sara Abdulaziz
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Nada Majrashi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Maznah Al Inizi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Asma Taher
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Nada Al Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
44
|
Hermey G, Hoffmeister-Ullerich SA, Merz B, Groß D, Kuhl D, Kins S. Amyloidosis causes downregulation of SorLA, SorCS1 and SorCS3 expression in mice. Biol Chem 2020; 400:1181-1189. [PMID: 31095505 DOI: 10.1515/hsz-2019-0146] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/03/2019] [Indexed: 12/30/2022]
Abstract
Accumulation of β-amyloid peptide (Aβ) is regarded as a primary cause of Alzheimer's disease (AD). Aβ is derived by sequential cleavage of the amyloid precursor protein (APP). Alterations in the subcellular targeting of APP are thought to affect the degree of Aβ production. Sorting receptors, such as SorLA, convey subcellular targeting of APP. Dysfunction of SorLA, and likely of the related receptors SorCS1 and SorCS3, cause AD. Nevertheless, disease progression could also provoke altered expression of the receptors. Here, we assessed if Aβ plaque formation promotes altered expression of SorLA, SorCS1 and SorCS3. We analyzed transcript levels during aging and after amyloidosis in brain areas characterized by early amyloid plaque formation in an AD mouse model (APPPS1) and wild types. We observed stable expression levels during aging (1-12 months). After plaque formation, SorCS1 and SorLA expression were markedly reduced in the frontal cerebral cortex and to a minor extent in the hippocampus, whereas SorCS3 expression was solely reduced in the frontal cerebral cortex. Our results indicate that disease progression, associated with Aβ accumulation, can negatively regulate expression of the receptors.
Collapse
Affiliation(s)
- Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Sabine A Hoffmeister-Ullerich
- Bioanalytics, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Barbara Merz
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Dagmar Groß
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Dietmar Kuhl
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D-20251 Hamburg, Germany
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of Kaiserslautern, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| |
Collapse
|
45
|
Su X, Peng D. New insight into sortilin in controlling lipid metabolism and the risk of atherogenesis. Biol Rev Camb Philos Soc 2020; 95:232-243. [PMID: 31625271 DOI: 10.1111/brv.12561] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Xin Su
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Daoquan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
46
|
PAK Kinases Target Sortilin and Modulate Its Sorting. Mol Cell Biol 2020; 40:MCB.00411-19. [PMID: 31767632 PMCID: PMC6965034 DOI: 10.1128/mcb.00411-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022] Open
Abstract
The multifunctional type 1 receptor sortilin is involved in endocytosis and intracellular transport of ligands. The short intracellular domain of sortilin binds several cytoplasmic adaptor proteins (e.g., the AP-1 complex and GGA1 to -3), most of which target two well-defined motifs: a C-terminal acidic cluster dileucine motif and a YXXΦ motif in the proximal third of the domain. Both motifs contribute to endocytosis as well as Golgi-endosome trafficking of sortilin. The multifunctional type 1 receptor sortilin is involved in endocytosis and intracellular transport of ligands. The short intracellular domain of sortilin binds several cytoplasmic adaptor proteins (e.g., the AP-1 complex and GGA1 to -3), most of which target two well-defined motifs: a C-terminal acidic cluster dileucine motif and a YXXΦ motif in the proximal third of the domain. Both motifs contribute to endocytosis as well as Golgi-endosome trafficking of sortilin. The C-terminal acidic cluster harbors a serine residue, which is subject to phosphorylation by casein kinase. Phosphorylation of this serine residue is known to modulate adaptor binding to sortilin. Here, we show that the cytoplasmic domain of sortilin also engages Rac-p21-activated kinases 1 to 3 (PAK1-3) via a binding segment that includes a tyrosine-based motif, also encompassing a serine residue. We further demonstrate that PAK1-3 specifically phosphorylate this serine residue and that this phosphorylation alters the affinity for AP-1 binding and consequently changes the intracellular localization of sortilin as a result of modulated trafficking. Our findings suggest that trafficking of ligands bound to sortilin is in part regulated by group A PAK kinases, which are downstream effectors of Rho GTPases and are known to affect a variety of processes by remodeling the cytoskeleton and by promoting gene transcription and cell survival.
Collapse
|
47
|
Gu XH, Li H, Zhang L, He T, Chai X, Wei H, Gao DS. Differential expression of glial cell line-derived neurotrophic factor splice variants in the mouse brain. Neural Regen Res 2020; 15:270-276. [PMID: 31552899 PMCID: PMC6905338 DOI: 10.4103/1673-5374.265561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) plays a critical role in neuronal survival and function. GDNF has two major splice variants in the brain, α-pro-GDNF and β-pro-GDNF, and both isoforms have strong neuroprotective effects on dopamine neurons. However, the expression of the GDNF splice variants in dopaminergic neurons in the brain remains unclear. Therefore, in this study, we investigated the mRNA and protein expression of α- and β-pro-GDNF in the mouse brain by real-time quantitative polymerase chain reaction, using splice variant-specific primers, and western blot analysis. At the mRNA level, β-pro-GDNF expression was significantly greater than that of α-pro-GDNF in the mouse brain. In contrast, at the protein level, α-pro-GDNF expression was markedly greater than that of β-pro-GDNF. To clarify the mechanism underlying this inverse relationship in mRNA and protein expression levels of the GDNF splice variants, we analyzed the expression of sorting protein-related receptor with A-type repeats (SorLA) by real-time quantitative polymerase chain reaction. At the mRNA level, SorLA was positively associated with β-pro-GDNF expression, but not with α-pro-GDNF expression. This suggests that the differential expression of α- and β-pro-GDNF in the mouse brain is related to SorLA expression. As a sorting protein, SorLA could contribute to the inverse relationship among the mRNA and protein levels of the GDNF isoforms. This study was approved by the Animal Ethics Committee of Xuzhou Medical University, China on July 14, 2016.
Collapse
Affiliation(s)
- Xiao-He Gu
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Heng Li
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Lin Zhang
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Tao He
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xiang Chai
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - He Wei
- Department of Neurosurgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Dian-Shuai Gao
- Department of Anatomy and Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
48
|
Demir İ, Yildirim Akan O, Guler A, Bozkaya G, Aslanipour B, Calan M. Relation of Decreased Circulating Sortilin Levels With Unfavorable Metabolic Profiles in Subjects With Newly Diagnosed Type 2 Diabetes Mellitus. Am J Med Sci 2019; 359:8-16. [PMID: 31902442 DOI: 10.1016/j.amjms.2019.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/01/2019] [Accepted: 10/15/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Sortilin, a pluripotent peptide hormone, plays a role in glucose and lipid metabolism. A link between sortilin and insulin sensitivity has been implicated. However, the clinical implications of this link remain elusive. Our aims were to investigate whether sortilin levels were altered in subjects with newly diagnosed type 2 diabetes mellitus (nT2DM) compared with subjects with normal glucose tolerance (NGT) and to determine whether a link exist between sortilin levels and metabolic parameters. MATERIALS AND METHODS A total of 150 subjects including 75 nT2DM patients and 75 subjects with NGT who were matched in age, body mass index, and sex were enrolled into this case-control study. The circulating levels of sortilin were measured using enzyme-linked immunosorbent assay. A 2-hour 75-g oral glucose tolerance test was used for diagnosis of T2DM. Metabolic parameters of enrolled subjects were also determined. RESULTS The circulating levels of sortilin were found to be significantly lower in subjects with nT2DM than in controls (138.44 ± 38.39 vs. 184.93 ± 49.67 pg/mL, P < 0.001). Sortilin levels showed a negative correlation with insulin resistance and unfavorable lipid profiles, while they were positively correlated with high-density lipoprotein cholesterol in subjects with nT2DM. Linear regression analysis showed an independent and inverse link between sortilin and insulin resistance and unfavorable lipid profiles. Moreover, logistic regression analysis revealed that the subjects with the lowest sortilin levels had an increased risk of nT2DM compared with those subjects with the highest sortilin levels. CONCLUSIONS Decreased circulating levels of sortilin were associated with unfavorable metabolic profiles in subjects with nT2DM.
Collapse
Affiliation(s)
- İsmail Demir
- Division of Endocrinology and Metabolism, Department of Internal Medicine
| | | | | | - Giray Bozkaya
- Department of Clinical Biochemistry, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Behnaz Aslanipour
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Izmir, Tukey
| | - Mehmet Calan
- Division of Endocrinology and Metabolism, Department of Internal Medicine.
| |
Collapse
|
49
|
Sun S, Yang J, Xie W, Peng T, Lv Y. Complicated trafficking behaviors involved in paradoxical regulation of sortilin in lipid metabolism. J Cell Physiol 2019; 235:3258-3269. [PMID: 31608989 DOI: 10.1002/jcp.29292] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/30/2019] [Indexed: 11/06/2022]
Abstract
This review aims to summarize and discuss the most recent advances in our understanding of the underlying mechanisms of the paradoxical effects of sortilin on lipid metabolism. The vacuolar protein sorting 10 protein (Vps10p) domain in the sortilin protein is responsible for substrate binding. Its cytoplasmic tail interacts with adaptor molecules, and modifications can determine whether sortilin trafficking occurs via the anterograde or retrograde pathway. The complicated trafficking behaviors likely contribute to the paradoxical roles of sortilin in lipid metabolism. The anterograde pathway of sortilin trafficking in hepatocytes, enterocytes, and peripheral cells likely causes an increase in plasma lipid levels, while the retrograde pathway leads to the opposite effect. Hepatocyte sortilin functions via the anterograde or retrograde pathway in a complicated and paradoxical manner to regulate apoB-containing lipoprotein metabolism. Clarifying the regulatory mechanisms underlying the trafficking behaviors of sortilin is necessary and may lead to artificial sortilin intervention as a potential therapeutic strategy for lipid disorder diseases. Conclusively, the paradoxical regulation of sortilin in lipid metabolism is likely due to its complicated trafficking behaviors.
Collapse
Affiliation(s)
- Sha Sun
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang City, China
| | - Jing Yang
- Clinical Medical Research Institute of the First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang City, China
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang City, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang City, China
| | - Yuncheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang City, China
| |
Collapse
|
50
|
Ribeiro LF, Verpoort B, Nys J, Vennekens KM, Wierda KD, de Wit J. SorCS1-mediated sorting in dendrites maintains neurexin axonal surface polarization required for synaptic function. PLoS Biol 2019; 17:e3000466. [PMID: 31658245 PMCID: PMC6837583 DOI: 10.1371/journal.pbio.3000466] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/07/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
The pre- and postsynaptic membranes comprising the synaptic junction differ in protein composition. The membrane trafficking mechanisms by which neurons control surface polarization of synaptic receptors remain poorly understood. The sorting receptor Sortilin-related CNS expressed 1 (SorCS1) is a critical regulator of trafficking of neuronal receptors, including the presynaptic adhesion molecule neurexin (Nrxn), an essential synaptic organizer. Here, we show that SorCS1 maintains a balance between axonal and dendritic Nrxn surface levels in the same neuron. Newly synthesized Nrxn1α traffics to the dendritic surface, where it is endocytosed. Endosomal SorCS1 interacts with the Rab11 GTPase effector Rab11 family-interacting protein 5 (Rab11FIP5)/Rab11 interacting protein (Rip11) to facilitate the transition of internalized Nrxn1α from early to recycling endosomes and bias Nrxn1α surface polarization towards the axon. In the absence of SorCS1, Nrxn1α accumulates in early endosomes and mispolarizes to the dendritic surface, impairing presynaptic differentiation and function. Thus, SorCS1-mediated sorting in dendritic endosomes controls Nrxn axonal surface polarization required for proper synapse development and function.
Collapse
Affiliation(s)
- Luís F. Ribeiro
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| | - Ben Verpoort
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| | - Julie Nys
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| | - Kristel M. Vennekens
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| | - Keimpe D. Wierda
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat, Leuven, Belgium
- KU Leuven, Department of Neurosciences, Leuven Brain Institute, Herestraat, Leuven, Belgium
| |
Collapse
|