1
|
Chen Y, Wu Y, Tian X, Shao G, Lin Q, Sun A. Golgiphagy: a novel selective autophagy to the fore. Cell Biosci 2024; 14:130. [PMID: 39438975 PMCID: PMC11495120 DOI: 10.1186/s13578-024-01311-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
The Golgi apparatus is the central hub of the cellular endocrine pathway and plays a crucial role in processing, transporting, and sorting proteins and lipids. Simultaneously, it is a highly dynamic organelle susceptible to degradation or fragmentation under various physiological or pathological conditions, potentially contributing to the development of numerous human diseases. Autophagy serves as a vital pathway for eukaryotes to manage intracellular and extracellular stress and maintain homeostasis by targeting damaged or redundant organelles for removal. Recent research has revealed that autophagy mechanisms can specifically degrade Golgi components, known as Golgiphagy. This review summarizes recent findings on Golgiphagy while also addressing unanswered questions regarding its mechanisms and regulation, aiming to advance our understanding of the role of Golgiphagy in human disease.
Collapse
Affiliation(s)
- Yifei Chen
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yihui Wu
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xianyan Tian
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Genbao Shao
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiong Lin
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China.
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Aiqin Sun
- Institute of Urinary System Diseases, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang, 212002, China.
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
2
|
Sharma R, Dey Das K, Srinivasula SM. EGF-mediated Golgi dynamics and cell migration require CARP2. Cell Rep 2024; 43:114896. [PMID: 39441718 DOI: 10.1016/j.celrep.2024.114896] [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: 12/21/2023] [Revised: 05/21/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
In mammalian cells, the Golgi exists in ribbon architecture-individual stacks laterally linked to each other by tubular structures. Golgi architecture changes dynamically to cater to cellular needs. Loss of architecture is linked with pathological conditions like cancer and neurodegeneration. Not much is known about the regulators of Golgi dynamics. Here, we demonstrate that CARP2 (caspase-8- and caspase-10-associated RING-containing protein 2), an endosomal ubiquitin ligase and a known regulator of cell migration, modulates Golgi dynamics. Epidermal growth factor (EGF) treatment modestly increases CARP2 protein and disperses Golgi. An exogenous supply of CARP2 also leads to Golgi dispersal. Conversely, Golgi remains intact in CARP2 knockout (KO) cells upon EGF treatment. CARP2 variants defective in either endosomal association or ligase activity are unable to affect Golgi dispersal. Importantly, CARP2 targets Golgin45 for ubiquitination and degradation in EGF-stimulated cells. Collectively, our findings unravel the existence of crosstalk between endosomal ubiquitin signaling and Golgi dynamics.
Collapse
Affiliation(s)
- Rahul Sharma
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Srinivasa M Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India.
| |
Collapse
|
3
|
Mazuryk O, Gurgul I, Oszajca M, Polaczek J, Kieca K, Bieszczad-Żak E, Martyka T, Stochel G. Nitric Oxide Signaling and Sensing in Age-Related Diseases. Antioxidants (Basel) 2024; 13:1213. [PMID: 39456466 PMCID: PMC11504650 DOI: 10.3390/antiox13101213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Nitric oxide (NO) is a key signaling molecule involved in numerous physiological and pathological processes within the human body. This review specifically examines the involvement of NO in age-related diseases, focusing on the cardiovascular, nervous, and immune systems. The discussion delves into the mechanisms of NO signaling in these diseases, emphasizing the post-translational modifications of involved proteins, such as S-nitrosation and nitration. The review also covers the dual nature of NO, highlighting both its protective and harmful effects, determined by concentration, location, and timing. Additionally, potential therapies that modulate NO signaling, including the use of NO donors and nitric oxide synthases (NOSs) inhibitors in the treatment of cardiovascular, neurodegenerative, and oncological diseases, are analyzed. Particular attention is paid to the methods for the determination of NO and its derivatives in the context of illness diagnosis and monitoring. The review underscores the complexity and dual role of NO in maintaining cellular balance and suggests areas for future research in developing new therapeutic strategies.
Collapse
Affiliation(s)
- Olga Mazuryk
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Ilona Gurgul
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Maria Oszajca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Justyna Polaczek
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| | - Konrad Kieca
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ewelina Bieszczad-Żak
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Tobiasz Martyka
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
- Doctoral School of Science and Life Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Grażyna Stochel
- Faculty of Chemistry, Jagiellonian University, 30-387 Krakow, Poland; (O.M.); (I.G.); (J.P.); (K.K.); (E.B.-Ż.); (T.M.)
| |
Collapse
|
4
|
Wang J, Fourriere L, Gleeson PA. Advances in the cell biology of the trafficking and processing of amyloid precursor protein: impact of familial Alzheimer's disease mutations. Biochem J 2024; 481:1297-1325. [PMID: 39302110 PMCID: PMC11555708 DOI: 10.1042/bcj20240056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
The production of neurotoxic amyloid-β peptides (Aβ) is central to the initiation and progression of Alzheimer's disease (AD) and involves sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. APP and the secretases are transmembrane proteins and their co-localisation in the same membrane-bound sub-compartment is necessary for APP cleavage. The intracellular trafficking of APP and the β-secretase, BACE1, is critical in regulating APP processing and Aβ production and has been studied in several cellular systems. Here, we summarise the intracellular distribution and transport of APP and its secretases, and the intracellular location for APP cleavage in non-polarised cells and neuronal models. In addition, we review recent advances on the potential impact of familial AD mutations on APP trafficking and processing. This is critical information in understanding the molecular mechanisms of AD progression and in supporting the development of novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Jingqi Wang
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Lou Fourriere
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A. Gleeson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
5
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kwon YJ, Kang HC. Selective delivery of imaging probes and therapeutics to the endoplasmic reticulum or Golgi apparatus: Current strategies and beyond. Adv Drug Deliv Rev 2024; 212:115386. [PMID: 38971180 DOI: 10.1016/j.addr.2024.115386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
To maximize therapeutic effects and minimize unwanted effects, the interest in drug targeting to the endoplasmic reticulum (ER) or Golgi apparatus (GA) has been recently growing because two organelles are distributing hubs of cellular building/signaling components (e.g., proteins, lipids, Ca2+) to other organelles and the plasma membrane. Their structural or functional damages induce organelle stress (i.e., ER or GA stress), and their aggravation is strongly related to diseases (e.g., cancers, liver diseases, brain diseases). Many efforts have been developed to image (patho)physiological functions (e.g., oxidative stress, protein/lipid-related processing) and characteristics (e.g., pH, temperature, biothiols, reactive oxygen species) in the target organelles and to deliver drugs for organelle disruption using organelle-targeting moieties. Therefore, this review will overview the structure, (patho)physiological functions/characteristics, and related diseases of the organelles of interest. Future direction on ER or GA targeting will be discussed by understanding current strategies and investigations on targeting, imaging/sensing, and therapeutic systems.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Kang Moo Huh
- Departments of Polymer Science and Engineering & Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea.
| |
Collapse
|
6
|
Liu M, Duan Y, Dong J, Zhang K, Jin X, Gao M, Jia H, Chen J, Liu M, Wei M, Zhong X. Early signs of neurodegenerative diseases: Possible mechanisms and targets for Golgi stress. Biomed Pharmacother 2024; 175:116646. [PMID: 38692058 DOI: 10.1016/j.biopha.2024.116646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
The Golgi apparatus plays a crucial role in mediating the modification, transport, and sorting of intracellular proteins and lipids. The morphological changes occurring in the Golgi apparatus are exceptionally important for maintaining its function. When exposed to external pressure or environmental stimulation, the Golgi apparatus undergoes adaptive changes in both structure and function, which are known as Golgi stress. Although certain signal pathway responses or post-translational modifications have been observed following Golgi stress, further research is needed to comprehensively summarize and understand the related mechanisms. Currently, there is evidence linking Golgi stress to neurodegenerative diseases; however, the role of Golgi stress in the progression of neurodegenerative diseases such as Alzheimer's disease remains largely unexplored. This review focuses on the structural and functional alterations of the Golgi apparatus during stress, elucidating potential mechanisms underlying the involvement of Golgi stress in regulating immunity, autophagy, and metabolic processes. Additionally, it highlights the pivotal role of Golgi stress as an early signaling event implicated in the pathogenesis and progression of neurodegenerative diseases. Furthermore, this study summarizes prospective targets that can be therapeutically exploited to mitigate neurodegenerative diseases by targeting Golgi stress. These findings provide a theoretical foundation for identifying novel breakthroughs in preventing and treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shayang, Liaoning 110005, China
| | - Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Kaisong Zhang
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, Liaoning 110167, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
7
|
Xing Y, Jian Y, Zhao X, Zhang Y, Zhang Z, Zhang X, Zhang X. Morphological determination of localization and function of Golgi proteins. BIOPHYSICS REPORTS 2024; 10:121-132. [PMID: 38774352 PMCID: PMC11103716 DOI: 10.52601/bpr.2024.240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 05/24/2024] Open
Abstract
In animal cells, the Golgi apparatus serves as the central hub of the endomembrane secretory pathway. It is responsible for the processing, modification, and sorting of proteins and lipids. The unique stacking and ribbon-like architecture of the Golgi apparatus forms the foundation for its precise functionality. Under cellular stress or pathological conditions, the structure of the Golgi and its important glycosylation modification function may change. It is crucial to employ suitable methodologies to study the structure and function of the Golgi apparatus, particularly when assessing the involvement of a target protein in Golgi regulation. This article provides a comprehensive overview of the diverse microscopy techniques used to determine the specific location of the target protein within the Golgi apparatus. Additionally, it outlines methods for assessing changes in the Golgi structure and its glycosylation modification function following the knockout of the target gene.
Collapse
Affiliation(s)
- Yusheng Xing
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yannan Jian
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaodan Zhao
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yue Zhang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenqian Zhang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xing Zhang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaoyan Zhang
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
8
|
Read CB, Ali AN, Stephenson DJ, Macknight HP, Maus KD, Cockburn CL, Kim M, Xie X, Carlyon JA, Chalfant CE. Ceramide-1-phosphate is a regulator of Golgi structure and is co-opted by the obligate intracellular bacterial pathogen Anaplasma phagocytophilum. mBio 2024; 15:e0029924. [PMID: 38415594 PMCID: PMC11005342 DOI: 10.1128/mbio.00299-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Many intracellular pathogens structurally disrupt the Golgi apparatus as an evolutionarily conserved promicrobial strategy. Yet, the host factors and signaling processes involved are often poorly understood, particularly for Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We found that A. phagocytophilum elevated cellular levels of the bioactive sphingolipid, ceramide-1-phosphate (C1P), to promote Golgi fragmentation that enables bacterial proliferation, conversion from its non-infectious to infectious form, and productive infection. A. phagocytophilum poorly infected mice deficient in ceramide kinase, the Golgi-localized enzyme responsible for C1P biosynthesis. C1P regulated Golgi morphology via activation of a PKCα/Cdc42/JNK signaling axis that culminates in phosphorylation of Golgi structural proteins, GRASP55 and GRASP65. siRNA-mediated depletion of Cdc42 blocked A. phagocytophilum from altering Golgi morphology, which impaired anterograde trafficking of trans-Golgi vesicles into and maturation of the pathogen-occupied vacuole. Cells overexpressing phosphorylation-resistant versions of GRASP55 and GRASP65 presented with suppressed C1P- and A. phagocytophilum-induced Golgi fragmentation and poorly supported infection by the bacterium. By studying A. phagocytophilum, we identify C1P as a regulator of Golgi structure and a host factor that is relevant to disease progression associated with Golgi fragmentation.IMPORTANCECeramide-1-phosphate (C1P), a bioactive sphingolipid that regulates diverse processes vital to mammalian physiology, is linked to disease states such as cancer, inflammation, and wound healing. By studying the obligate intracellular bacterium Anaplasma phagocytophilum, we discovered that C1P is a major regulator of Golgi morphology. A. phagocytophilum elevated C1P levels to induce signaling events that promote Golgi fragmentation and increase vesicular traffic into the pathogen-occupied vacuole that the bacterium parasitizes. As several intracellular microbial pathogens destabilize the Golgi to drive their infection cycles and changes in Golgi morphology is also linked to cancer and neurodegenerative disorder progression, this study identifies C1P as a potential broad-spectrum therapeutic target for infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Curtis B. Read
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Anika N. Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Daniel J. Stephenson
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - H. Patrick Macknight
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth D. Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Chelsea L. Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Xiujie Xie
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Charles E. Chalfant
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, Virginia, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, Virginia, USA
| |
Collapse
|
9
|
Mayer J, Boeck D, Werner M, Frankenhauser D, Geley S, Farhan H, Shimozawa M, Nilsson P. Inhibition of Autophagy Alters Intracellular Transport of APP Resulting in Increased APP Processing. Traffic 2024; 25:e12934. [PMID: 38613404 DOI: 10.1111/tra.12934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 03/03/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
Alzheimer's disease (AD) pathology is characterized by amyloid beta (Aβ) plaques and dysfunctional autophagy. Aβ is generated by sequential proteolytic cleavage of amyloid precursor protein (APP), and the site of intracellular APP processing is highly debated, which may include autophagosomes. Here, we investigated the involvement of autophagy, including the role of ATG9 in APP intracellular trafficking and processing by applying the RUSH system, which allows studying the transport of fluorescently labeled mCherry-APP-EGFP in a systematic way, starting from the endoplasmic reticulum. HeLa cells, expressing the RUSH mCherry-APP-EGFP system, were investigated by live cell imaging, immunofluorescence, and Western blot. We found that mCherry-APP-EGFP passed through the Golgi faster in ATG9 knockout cells. Furthermore, ATG9 deletion shifted mCherry-APP-EGFP from early endosomes and lysosomes toward the plasma membrane concomitant with reduced endocytosis. Importantly, this alteration in mCherry-APP-EGFP transport resulted in increased secreted mCherry-soluble APP and C-terminal fragment-EGFP. These effects were also phenocopied by pharmacological inhibition of ULK1, indicating that autophagy is regulating the intracellular trafficking and processing of APP. These findings contribute to the understanding of the role of autophagy in APP metabolism and could potentially have implications for new therapeutic approaches for AD.
Collapse
Affiliation(s)
- Johanna Mayer
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
| | - Dominik Boeck
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
- Institute of Molecular Neurogenetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michelle Werner
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
| | | | - Stephan Geley
- Institute of Pathophysiology, Innsbruck Medical University, Innsbruck, Austria
| | - Hesso Farhan
- Institute of Pathophysiology, Innsbruck Medical University, Innsbruck, Austria
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
10
|
Benvenuto G, Leone S, Astoricchio E, Bormke S, Jasek S, D'Aniello E, Kittelmann M, McDonald K, Hartenstein V, Baena V, Escrivà H, Bertrand S, Schierwater B, Burkhardt P, Ruiz-Trillo I, Jékely G, Ullrich-Lüter J, Lüter C, D'Aniello S, Arnone MI, Ferraro F. Evolution of the ribbon-like organization of the Golgi apparatus in animal cells. Cell Rep 2024; 43:113791. [PMID: 38428420 DOI: 10.1016/j.celrep.2024.113791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/31/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024] Open
Abstract
The "ribbon," a structural arrangement in which Golgi stacks connect to each other, is considered to be restricted to vertebrate cells. Although ribbon disruption is linked to various human pathologies, its functional role in cellular processes remains unclear. In this study, we investigate the evolutionary origin of the Golgi ribbon. We observe a ribbon-like architecture in the cells of several metazoan taxa suggesting its early emergence in animal evolution predating the appearance of vertebrates. Supported by AlphaFold2 modeling, we propose that the evolution of Golgi reassembly and stacking protein (GRASP) binding by golgin tethers may have driven the joining of Golgi stacks resulting in the ribbon-like configuration. Additionally, we find that Golgi ribbon assembly is a shared developmental feature of deuterostomes, implying a role in embryogenesis. Overall, our study points to the functional significance of the Golgi ribbon beyond vertebrates and underscores the need for further investigations to unravel its elusive biological roles.
Collapse
Affiliation(s)
- Giovanna Benvenuto
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | - Serena Leone
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | - Emanuele Astoricchio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | | | - Sanja Jasek
- Living Systems Institute, University of Exeter, Exeter, UK; Heidelberg University, Centre for Organismal Studies (COS), Heidelberg, Germany
| | - Enrico D'Aniello
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | - Maike Kittelmann
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Kent McDonald
- Electron Microscope Lab, University of California Berkeley, Berkeley, CA, USA
| | - Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Valentina Baena
- Department of Cell Biology, UConn Health, Farmington, CT, USA
| | - Héctor Escrivà
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-Mer, France
| | - Stephanie Bertrand
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-Mer, France
| | - Bernd Schierwater
- Institute of Ecology and Evolution, Hannover University of Veterinary Medicine Foundation, Hannover, Germany
| | | | - Iñaki Ruiz-Trillo
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Passeig Marítim de la Barceloneta, Barcelona, Spain; ICREA, Barcelona, Spain
| | - Gáspár Jékely
- Living Systems Institute, University of Exeter, Exeter, UK; Heidelberg University, Centre for Organismal Studies (COS), Heidelberg, Germany
| | | | | | - Salvatore D'Aniello
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | - Maria Ina Arnone
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy
| | - Francesco Ferraro
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn (SZN), Naples, Italy.
| |
Collapse
|
11
|
Wiśniewska K, Gaffke L, Żabińska M, Węgrzyn G, Pierzynowska K. Cellular Organelle-Related Transcriptomic Profile Abnormalities in Neuronopathic Types of Mucopolysaccharidosis: A Comparison with Other Neurodegenerative Diseases. Curr Issues Mol Biol 2024; 46:2678-2700. [PMID: 38534785 PMCID: PMC10968730 DOI: 10.3390/cimb46030169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
Mucopolysaccharidoses (MPS) are a group of diseases caused by mutations in genes encoding lysosomal enzymes that catalyze reactions of glycosaminoglycan (GAG) degradation. As a result, GAGs accumulate in lysosomes, impairing the proper functioning of entire cells and tissues. There are 14 types/subtypes of MPS, which are differentiated by the kind(s) of accumulated GAG(s) and the type of a non-functional lysosomal enzyme. Some of these types (severe forms of MPS types I and II, MPS III, and MPS VII) are characterized by extensive central nervous system disorders. The aim of this work was to identify, using transcriptomic methods, organelle-related genes whose expression levels are changed in neuronopathic types of MPS compared to healthy cells while remaining unchanged in non-neuronopathic types of MPS. The study was conducted with fibroblast lines derived from patients with neuronopathic and non-neuronopathic types of MPS and control (healthy) fibroblasts. Transcriptomic analysis has identified genes related to cellular organelles whose expression is altered. Then, using fluorescence and electron microscopy, we assessed the morphology of selected structures. Our analyses indicated that the genes whose expression is affected in neuronopathic MPS are often associated with the structures or functions of the cell nucleus, endoplasmic reticulum, or Golgi apparatus. Electron microscopic studies confirmed disruptions in the structures of these organelles. Special attention was paid to up-regulated genes, such as PDIA3 and MFGE8, and down-regulated genes, such as ARL6IP6, ABHD5, PDE4DIP, YIPF5, and CLDN11. Of particular interest is also the GM130 (GOLGA2) gene, which encodes golgin A2, which revealed an increased expression in neuronopathic MPS types. We propose to consider the levels of mRNAs of these genes as candidates for biomarkers of neurodegeneration in MPS. These genes may also become potential targets for therapies under development for neurological disorders associated with MPS and candidates for markers of the effectiveness of these therapies. Although fibroblasts rather than nerve cells were used in this study, it is worth noting that potential genetic markers characteristic solely of neurons would be impractical in testing patients, contrary to somatic cells that can be relatively easily obtained from assessed persons.
Collapse
Affiliation(s)
| | | | | | | | - Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (K.W.); (L.G.); (M.Ż.); (G.W.)
| |
Collapse
|
12
|
Boyer CK, Blom SE, Machado AE, Rohli KE, Maxson ME, Stephens SB. Loss of the Golgi-localized v-ATPase subunit does not alter insulin granule formation or pancreatic islet β-cell function. Am J Physiol Endocrinol Metab 2024; 326:E245-E257. [PMID: 38265287 PMCID: PMC11193524 DOI: 10.1152/ajpendo.00342.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/25/2024]
Abstract
Delayed Golgi export of proinsulin has recently been identified as an underlying mechanism leading to insulin granule loss and β-cell secretory defects in type 2 diabetes (T2D). Because acidification of the Golgi lumen is critical for proinsulin sorting and delivery into the budding secretory granule, we reasoned that dysregulation of Golgi pH may contribute to proinsulin trafficking defects. In this report, we examined pH regulation of the Golgi and identified a partial alkalinization of the Golgi lumen in a diabetes model. To further explore this, we generated a β-cell specific knockout (KO) of the v0a2 subunit of the v-ATPase pump, which anchors the v-ATPase to the Golgi membrane. Although loss of v0a2 partially neutralized Golgi pH and was accompanied by distension of the Golgi cisternae, proinsulin export from the Golgi and insulin granule formation were not affected. Furthermore, β-cell function was well preserved. β-cell v0a2 KO mice exhibited normal glucose tolerance in both sexes, no genotypic difference to diet-induced obesity, and normal insulin secretory responses. Collectively, our data demonstrate the v0a2 subunit contributes to β-cell Golgi pH regulation but suggest that additional disturbances to Golgi structure and function contribute to proinsulin trafficking defects in diabetes.NEW & NOTEWORTHY Delayed proinsulin export from the Golgi in diabetic β-cells contributes to decreased insulin granule formation, but the underlying mechanisms are not clear. Here, we explored if dysregulation of Golgi pH can alter Golgi function using β-cell specific knockout (KO) of the Golgi-localized subunit of the v-ATPase, v0a2. We show that partial alkalinization of the Golgi dilates the cisternae, but does not affect proinsulin export, insulin granule formation, insulin secretion, or glucose homeostasis.
Collapse
Affiliation(s)
- Cierra K Boyer
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, United States
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States
| | - Sandra E Blom
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Ashleigh E Machado
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Kristen E Rohli
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States
| | - Michelle E Maxson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States
| |
Collapse
|
13
|
Wang J, Gleeson PA, Fourriere L. Spatial-Temporal Mapping Reveals the Golgi as the Major Processing Site for the Pathogenic Swedish APP Mutation: Familial APP Mutant Shifts the Major APP Processing Site. Traffic 2024; 25:e12932. [PMID: 38528836 DOI: 10.1111/tra.12932] [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: 10/16/2023] [Revised: 02/13/2024] [Accepted: 03/01/2024] [Indexed: 03/27/2024]
Abstract
Alzheimer's disease is associated with increased levels of amyloid beta (Aβ) generated by sequential intracellular cleavage of amyloid precursor protein (APP) by membrane-bound secretases. However, the spatial and temporal APP cleavage events along the trafficking pathways are poorly defined. Here, we use the Retention Using Selective Hooks (RUSH) to compare in real time the anterograde trafficking and temporal cleavage events of wild-type APP (APPwt) with the pathogenic Swedish APP (APPswe) and the disease-protective Icelandic APP (APPice). The analyses revealed differences in the trafficking profiles and processing between APPwt and the APP familial mutations. While APPwt was predominantly processed by the β-secretase, BACE1, following Golgi transport to the early endosomes, the transit of APPswe through the Golgi was prolonged and associated with enhanced amyloidogenic APP processing and Aβ secretion. A 20°C block in cargo exit from the Golgi confirmed β- and γ-secretase processing of APPswe in the Golgi. Inhibition of the β-secretase, BACE1, restored APPswe anterograde trafficking profile to that of APPwt. APPice was transported rapidly through the Golgi to the early endosomes with low levels of Aβ production. This study has revealed different intracellular locations for the preferential cleavage of APPwt and APPswe and Aβ production, and the Golgi as the major processing site for APPswe, findings relevant to understand the molecular basis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jingqi Wang
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Lou Fourriere
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Lacar B, Ferdosi S, Alavi A, Stukalov A, Venkataraman GR, de Geus M, Dodge H, Wu CY, Kivisakk P, Das S, Guturu H, Hyman B, Batzoglou S, Arnold SE, Siddiqui A. Identification of Novel Biomarkers for Alzheimer's Disease and Related Dementias Using Unbiased Plasma Proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574446. [PMID: 38260620 PMCID: PMC10802486 DOI: 10.1101/2024.01.05.574446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Alzheimer's disease (AD) and related dementias (ADRD) is a complex disease with multiple pathophysiological drivers that determine clinical symptomology and disease progression. These diseases develop insidiously over time, through many pathways and disease mechanisms and continue to have a huge societal impact for affected individuals and their families. While emerging blood-based biomarkers, such as plasma p-tau181 and p-tau217, accurately detect Alzheimer neuropthology and are associated with faster cognitive decline, the full extension of plasma proteomic changes in ADRD remains unknown. Earlier detection and better classification of the different subtypes may provide opportunities for earlier, more targeted interventions, and perhaps a higher likelihood of successful therapeutic development. In this study, we aim to leverage unbiased mass spectrometry proteomics to identify novel, blood-based biomarkers associated with cognitive decline. 1,786 plasma samples from 1,005 patients were collected over 12 years from partcipants in the Massachusetts Alzheimer's Disease Research Center Longitudinal Cohort Study. Patient metadata includes demographics, final diagnoses, and clinical dementia rating (CDR) scores taken concurrently. The Proteograph™ Product Suite (Seer, Inc.) and liquid-chromatography mass-spectrometry (LC-MS) analysis were used to process the plasma samples in this cohort and generate unbiased proteomics data. Data-independent acquisition (DIA) mass spectrometry results yielded 36,259 peptides and 4,007 protein groups. Linear mixed effects models revealed 138 differentially abundant proteins between AD and healthy controls. Machine learning classification models for AD diagnosis identified potential candidate biomarkers including MBP, BGLAP, and APoD. Cox regression models were created to determine the association of proteins with disease progression and suggest CLNS1A, CRISPLD2, and GOLPH3 as targets of further investigation as potential biomarkers. The Proteograph workflow provided deep, unbiased coverage of the plasma proteome at a speed that enabled a cohort study of almost 1,800 samples, which is the largest, deep, unbiased proteomics study of ADRD conducted to date.
Collapse
Affiliation(s)
| | | | | | | | | | - Matthijs de Geus
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | - Hiroko Dodge
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | - Chao-Yi Wu
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | - Pia Kivisakk
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | - Sudeshna Das
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | | | - Brad Hyman
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | | | - Steven E. Arnold
- Massachusetts General Hospital (MGH), 55 Fruit Street, Boston, Massachusetts 02114
| | | |
Collapse
|
15
|
Sansaria R, Das KD, Poulose A. Quantification of golgi dispersal and classification using machine learning models. Micron 2024; 176:103547. [PMID: 37839330 DOI: 10.1016/j.micron.2023.103547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023]
Abstract
The Golgi body is a critical organelle in eukaryotic cells responsible for processing and modifying proteins and lipids. Under certain conditions, such as stress, disease, or ageing, the Golgi structure alters. Therefore, understanding the mechanisms that regulate Golgi dispersion has significant research contributions to identifying disease. However, there is a lack of tools to quantify the Golgi dispersion datasets. In this paper, we aim to automate the process of quantification of Golgi dispersion and use extracted features to classify dispersed Golgi images from undispersed Golgi images using machine learning models. First, we collected confocal microscopy images of transiently transfected HeLa cells expressing Galactose-1-phosphate uridylyltransferase (GALT)- green fluorescent protein (GFP) to quantify Golgi dispersal and classification. For the quantification, we introduced automated image processing and segmentation by applying mean and Gaussian filters. Then we used Otsu thresholding on preprocessed images and watershed segmentation to refine the segmentation of dispersed Golgi particles. In the case of classification, we extracted features from the Golgi dispersal images and classified them into empty vector (EV) versus CARP1 ring mutant (CARP1 RM) and empty vector (EV) versus CARP1 wildtype (CARP1 WT) classes. Our approach used machine-learning models, including logistic regression, decision tree, random forest, Naive Bayes, k-Nearest Neighbor (KNN), and gradient boosting for dispersed Golgi image classification. The experiment results show that our quantification technique on Golgi dispersal images reached 65% classification accuracy when the system uses a gradient boosting classifier for EV vs. CARP1 WT classification. Furthermore, our approach achieved 65% classification accuracy using a random forest classifier for EV vs. CARP1 RM classification.
Collapse
Affiliation(s)
- Rutika Sansaria
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram (IISER TVM), Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Alwin Poulose
- School of Data Science, Indian Institute of Science Education and Research Thiruvananthapuram (IISE R TVM), Vithura, Thiruvananthapuram 695551, Kerala, India.
| |
Collapse
|
16
|
Liu J, Hu Z, Huang Y, Zhang Y, Peng D. Study on the protective effect of OM-MSCs on Golgi apparatus after intracerebral hemorrhage in Sprague-Dawley rats. AMERICAN JOURNAL OF STEM CELLS 2023; 12:124-137. [PMID: 38213638 PMCID: PMC10776343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION The present study aimed to assess alterations in apoptosis rate, Golgi morphology and GOLPH3 expression following intracerebral hemorrhage (ICH) both before and after intervention with OM-MSCs. The objective was to investigate the impact of ICH on Golgi apparatus (GA) stress and to explore the potential protective effects of OM-MSCs on GA following ICH. MATERIAL AND METHODS A total of 54 Sprague-Dawley rats were allocated into three experimental groups: sham operation group, ICH group and OM-MSCs group. ICH models were established by collagenase method while OM-MSCs were cultured in vitro. In OM-MSCs intervention group, one million OM-MSCs were stereotactically injected into unilateral striatum of rats 48 hours after ICH modeling while other two groups received an equivalent volume of PBS. Brain tissues were collected at 1 day, 3 day and 7 day post intervention and subsequently assessed for cellular apoptosis, morphological change of GA and expression of GOLPH3. The obtained data were subjected to statistical analysis by SPSS 21.0. RESULTS 1. Apoptosis rate in the 1 d and 3 d ICH groups was significantly higher compared to sham operation group (P < 0.05), but significantly lower compared to OM-MSCs intervention group (P < 0.05). 2. While no noticeable morphological changes were observed in sham operation group, GA in ICH group exhibited a significant increase fragmentation. After OM-MSCs intervention, the fragmentation of GA decreased significantly. 3. On 3 d, expression of GOLPH3 in ICH group was significantly higher than that in sham operation group (P < 0.05) but significantly lower than that of OM-MSCs intervention group (P < 0.05). CONCLUSIONS The rate of apoptosis, fragmentation of GA, and expression of GOLPH3 exhibited significant increases following ICH in SD rats. Conversely, all of these factors demonstrated significant decreases subsequent to early intervention with OM-MSCs, thereby exerting neuroprotective effects.
Collapse
Affiliation(s)
- Junjiang Liu
- Department of Geriatrics, Hunan Provincial People’s Hospital (First Affiliated Hospital of Hunan Normal University)No. 61 Jiefang West Road, Changsha 410011, Hunan, The People’s Republic of China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital of Central South UniversityChangsha 410011, Hunan, The People’s Republic of China
| | - Yan Huang
- Hunan Provincial Maternal and Child Health Care HospitalChangsha 410008, Hunan, The People’s Republic of China
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal UniversityChangsha 410081, Hunan, The People’s Republic of China
- Hunan Provincial University Key Laboratory of The Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical UniversityChangsha 410219, Hunan, The People’s Republic of China
| | - Yidan Zhang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital of Central South UniversityChangsha 410011, Hunan, The People’s Republic of China
| | - Dezhen Peng
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital of Central South UniversityChangsha 410011, Hunan, The People’s Republic of China
| |
Collapse
|
17
|
Guo J, Sun J, Liu D, Liu J, Gui L, Luo M, Kong D, Wusiman S, Yang C, Liu T, Yuan Z, Li R. Developing a Two-Photon "AND" Logic Probe and Its Application in Alzheimer's Disease Differentiation. Anal Chem 2023; 95:16868-16876. [PMID: 37947381 DOI: 10.1021/acs.analchem.3c02634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
In Alzheimer's disease, hypochlorous acid involved in the clearance of invading bacteria or pathogens and butyrylcholinesterase engaged in the hydrolysis of the neurotransmitter acetylcholine are relatively significantly altered. However, there are few dual detection probes for hypochlorous acid and butyrylcholinesterase. In addition, single-response probes suffer from serious off-target effects and near-infrared probes do not easily penetrate the blood-brain barrier due to their excessive molecular weight. In this work, we constructed a two-photon fluorescent probe that recognizes hypochlorous acid and butyrylcholinesterase based on a dual-lock strategy. The thiocarbonyl group is oxidized in the presence of hypochlorous acid, and the hydrolysis occurs at the 7-position ester bond in the existence of butyrylcholinesterase, releasing a strongly fluorescent fluorophore, 4-methylumbelliferone. Excellent imaging was performed in PC12 cells using this probe, and deep two-photon imaging was observed in the brains of AD mice after tail vein injection with this probe. It indicates that the probe can provide a promising tool for the more precise diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jingxuan Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Donghui Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
- School of Pharmacy, Guizhou Medical University, Guiyang 55004, China
| | - Ji Liu
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lijuan Gui
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Man Luo
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Dexin Kong
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Sainaiwaiergul Wusiman
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Chang Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
- School of Pharmacy, Guizhou Medical University, Guiyang 55004, China
| | - Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixi Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| |
Collapse
|
18
|
Patel TA, Kevadiya BD, Bajwa N, Singh PA, Zheng H, Kirabo A, Li YL, Patel KP. Role of Nanoparticle-Conjugates and Nanotheranostics in Abrogating Oxidative Stress and Ameliorating Neuroinflammation. Antioxidants (Basel) 2023; 12:1877. [PMID: 37891956 PMCID: PMC10604131 DOI: 10.3390/antiox12101877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress is a deteriorating condition that arises due to an imbalance between the reactive oxygen species and the antioxidant system or defense of the body. The key reasons for the development of such conditions are malfunctioning of various cell organelles, such as mitochondria, endoplasmic reticulum, and Golgi complex, as well as physical and mental disturbances. The nervous system has a relatively high utilization of oxygen, thus making it particularly vulnerable to oxidative stress, which eventually leads to neuronal atrophy and death. This advances the development of neuroinflammation and neurodegeneration-associated disorders such as Alzheimer's disease, Parkinson's disease, epilepsy, dementia, and other memory disorders. It is imperative to treat such conditions as early as possible before they worsen and progress to irreversible damage. Oxidative damage can be negated by two mechanisms: improving the cellular defense system or providing exogenous antioxidants. Natural antioxidants can normally handle such oxidative stress, but they have limited efficacy. The valuable features of nanoparticles and/or nanomaterials, in combination with antioxidant features, offer innovative nanotheranostic tools as potential therapeutic modalities. Hence, this review aims to represent novel therapeutic approaches like utilizing nanoparticles with antioxidant properties and nanotheranostics as delivery systems for potential therapeutic applications in various neuroinflammation- and neurodegeneration-associated disease conditions.
Collapse
Affiliation(s)
- Tapan A. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Neha Bajwa
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali 140413, Punjab, India; (N.B.); (P.A.S.)
| | - Preet Amol Singh
- University Institute of Pharma Sciences (UIPS), Chandigarh University, Mohali 140413, Punjab, India; (N.B.); (P.A.S.)
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Yu-Long Li
- Department of Emergency Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| |
Collapse
|
19
|
Orobets KS, Karamyshev AL. Amyloid Precursor Protein and Alzheimer's Disease. Int J Mol Sci 2023; 24:14794. [PMID: 37834241 PMCID: PMC10573485 DOI: 10.3390/ijms241914794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders associated with age or inherited mutations. It is characterized by severe dementia in the late stages that affect memory, cognitive functions, and daily life overall. AD progression is linked to the accumulation of cytotoxic amyloid beta (Aβ) and hyperphosphorylated tau protein combined with other pathological features such as synaptic loss, defective energy metabolism, imbalances in protein, and metal homeostasis. Several treatment options for AD are under investigation, including antibody-based therapy and stem cell transplantation. Amyloid precursor protein (APP) is a membrane protein considered to play a main role in AD pathology. It is known that APP in physiological conditions follows a non-amyloidogenic pathway; however, it can proceed to an amyloidogenic scenario, which leads to the generation of extracellular deleterious Aβ plaques. Not all steps of APP biogenesis are clear so far, and these questions should be addressed in future studies. AD is a complex chronic disease with many factors that contribute to disease progression.
Collapse
Affiliation(s)
| | - Andrey L. Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| |
Collapse
|
20
|
Wang P, Anderson DE, Ye Y. PI3K-AKT activation resculpts integrin signaling to drive filamentous tau-induced proinflammatory astrogliosis. Cell Biosci 2023; 13:179. [PMID: 37759245 PMCID: PMC10536728 DOI: 10.1186/s13578-023-01128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Microtubule-binding protein tau is a misfolding-prone protein associated with tauopathies. As tau undergoes cell-to-cell transmission, extracellular tau aggregates convert astrocytes into a pro-inflammatory state via integrin activation, causing them to release unknown neurotoxic factors. RESULTS Here, we combine transcriptomics with isotope labeling-based quantitative mass spectrometry analysis of mouse primary astrocyte secretome to establish PI3K-AKT as a critical differentiator between pathogenic and physiological integrin activation; simultaneous activation of PI3K-AKT and focal adhesion kinase (FAK) in tau fibril-treated astrocytes changes the output of integrin signaling, causing pro-inflammatory gene upregulation, trans-Golgi network restructuring, and altered secretory flow. Furthermore, NCAM1, as a proximal signaling component in tau-stimulated integrin and PI3K-AKT activation, facilitates the secretion of complement C3 as a main neurotoxic factor. Significantly, tau fibrils-associated astrogliosis and C3 secretion can be mitigated by FAK or PI3K inhibitors. CONCLUSIONS These findings reveal an unexpected function for PI3K-AKT in tauopathy-associated reactive astrogliosis, which may be a promising target for anti-inflammation-based Alzheimer's therapy.
Collapse
Affiliation(s)
- Peng Wang
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - D Eric Anderson
- Advanced Mass Spectrometry Core, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
21
|
Toader C, Eva L, Covache-Busuioc RA, Costin HP, Glavan LA, Corlatescu AD, Ciurea AV. Unraveling the Multifaceted Role of the Golgi Apparatus: Insights into Neuronal Plasticity, Development, Neurogenesis, Alzheimer's Disease, and SARS-CoV-2 Interactions. Brain Sci 2023; 13:1363. [PMID: 37891732 PMCID: PMC10605100 DOI: 10.3390/brainsci13101363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
This article critically evaluates the multifunctional role of the Golgi apparatus within neurological paradigms. We succinctly highlight its influence on neuronal plasticity, development, and the vital trafficking and sorting mechanisms for proteins and lipids. The discourse further navigates to its regulatory prominence in neurogenesis and its implications in Alzheimer's Disease pathogenesis. The emerging nexus between the Golgi apparatus and SARS-CoV-2 underscores its potential in viral replication processes. This consolidation accentuates the Golgi apparatus's centrality in neurobiology and its intersections with both neurodegenerative and viral pathologies. In essence, understanding the Golgi's multifaceted functions harbors profound implications for future therapeutic innovations in neurological and viral afflictions.
Collapse
Affiliation(s)
- Corneliu Toader
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
- Department of Vascular Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Lucian Eva
- Faculty of Medicine, “Dunarea de Jos” University of Galati, 800201 Galați, Romania
- Emergency Clinical Hospital “Prof. dr. N. Oblu”, 700309 Iasi, Romania
| | - Razvan-Adrian Covache-Busuioc
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Horia Petre Costin
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Luca-Andrei Glavan
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Antonio Daniel Corlatescu
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
| | - Alexandru Vlad Ciurea
- Department of Neurosurgery, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.T.); (H.P.C.); (L.-A.G.); (A.D.C.); (A.V.C.)
- Neurosurgery Department, Sanador Clinical Hospital, 010991 Bucharest, Romania
| |
Collapse
|
22
|
Fang Z, Chen H. The in vivo drug delivery pattern of the organelle-targeting small molecules. Adv Drug Deliv Rev 2023; 200:115020. [PMID: 37481114 DOI: 10.1016/j.addr.2023.115020] [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: 04/29/2023] [Revised: 07/07/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Eukaryotic cell organelles sustain the life of cells. Their structural changes and dysfunctions can cause abnormal physiological activities and lead to various diseases. Molecular imaging technology enables the visualization of subcellular structures, cells, organs, and the whole living body's structure and metabolism dynamic changes. This could help to reveal the pharmacology mechanisms and drug delivery pathway in vivo. This article discusses the relationship between organelles and human disease, reviews recent probes targeting organelles and their behavior in vivo. We found that mitochondria-targeting probes prefer accumulation in the intestine, heart, and tumor. The lysosome-targeting probe accumulates in the intestine and tumor. Few studies on endoplasmic reticulum- or Golgi apparatus-targeting probes have been reported for in vivo imaging. We hope this review could provide new insights for developing and applying organelle-targeting probes.
Collapse
Affiliation(s)
- Zhao Fang
- Molecular Imaging Center, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hao Chen
- Molecular Imaging Center, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
23
|
Wang P, Anderson DE, Ye Y. PI3K-AKT activation resculpts integrin signaling to drive filamentous tau-induced proinflammatory astrogliosis. RESEARCH SQUARE 2023:rs.3.rs-3253118. [PMID: 37674732 PMCID: PMC10479431 DOI: 10.21203/rs.3.rs-3253118/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Background Microtubule-binding protein tau is a misfolding-prone protein associated with tauopathies. As tau undergoes cell-to-cell transmission, extracellular tau aggregates convert astrocytes into a pro-inflammatory state via integrin activation, causing them to release unknown neurotoxic factors. Results Here, we combine transcriptomics with isotope labeling-based quantitative mass spectrometry analysis of mouse primary astrocyte secretome to establish PI3K-AKT as a critical differentiator between pathogenic and physiological integrin activation; simultaneous activation of PI3K-AKT and focal adhesion kinase (FAK) in tau fibril-treated astrocytes changes the output of integrin signaling, causing pro-inflammatory gene upregulation, trans-Golgi network restructuring, and altered secretory flow. Furthermore, NCAM1, as a proximal signaling component in tau-stimulated integrin and PI3K-AKT activation, facilitates the secretion of complement C3 as a main neurotoxic factor. Significantly, tau fibrils-associated astrogliosis and C3 secretion can be mitigated by FAK or PI3K inhibitors. Conclusions These findings reveal an unexpected function for PI3K-AKT in tauopathy-associated reactive astrogliosis, which may be a promising target for anti-inflammation-based Alzheimer's therapy.
Collapse
Affiliation(s)
- Peng Wang
- National Institute of Diabetes and Digestive and Kidney Diseases
| | - D Eric Anderson
- National Institute of Diabetes and Digestive and Kidney Diseases
| | - Yihong Ye
- National Institute of Diabetes and Digestive and Kidney Diseases
| |
Collapse
|
24
|
Keller T, Trinks N, Brand J, Trippmacher S, Stahlhut P, Albrecht K, Papastavrou G, Koepsell H, Sauer M, Groll J. Design of Nanohydrogels for Targeted Intracellular Drug Transport to the Trans-Golgi Network. Adv Healthc Mater 2023; 12:e2201794. [PMID: 36739269 PMCID: PMC11469190 DOI: 10.1002/adhm.202201794] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/27/2023] [Indexed: 02/06/2023]
Abstract
Nanohydrogels combine advantages of hydrogels and nanoparticles. In particular, they represent promising drug delivery systems. Nanogel synthesis by oxidative condensation of polyglycidol prepolymers, that are modified with thiol groups, results in crosslinking by disulfide bonds. Hereby, biomolecules like the antidiabetic peptide RS1-reg, derived from the regulatory protein RS1 of the Na+ -D-glucose cotransporter SGLT1, can be covalently bound by cysteine residues to the nanogel in a hydrophilic, stabilizing environment. After oral uptake, the acid-stable nanogels protect their loading during gastric passage from proteolytic degradation. Under alkaline conditions in small intestine the nanohydrogels become mucoadhesive, pass the intestinal mucosa and are taken up into small intestinal enterocytes by endocytosis. Using Caco-2 cells as a model for small intestinal enterocytes, by confocal laser scanning microscopy and structured illumination microscopy, the colocalization of fluorescent-labeled RS1-reg with markers of endosomes, lysosomes, and trans-Golgi-network after uptake with polyglycidol-based nanogels formed by precipitation polymerization is demonstrated. This indicates that RS1-reg follows the endosomal pathway. In the following, the design of bespoken nanohydrogels for specific targeting of RS1-reg to its site of action at the trans-Golgi network is described that might also represent a way of targeted transport for other drugs to their targets at the Golgi apparatus.
Collapse
Affiliation(s)
- Thorsten Keller
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Nora Trinks
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jessica Brand
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Steffen Trippmacher
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Krystyna Albrecht
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| | - Georg Papastavrou
- Physical Chemistry IIUniversity of BayreuthUniversitätsstr. 3095440BayreuthGermany
| | - Hermann Koepsell
- Institute of Anatomy and Cell BiologyUniversity of WürzburgKoellikerstraße 697070WürzburgGermany
| | - Markus Sauer
- Department of Biotechnology and BiophysicsUniversity of WürzburgAm Hubland97074WürzburgGermany
| | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and BiofabricationUniversity of WürzburgPleicherwall 297070WürzburgGermany
| |
Collapse
|
25
|
Ghannoum S, Fantini D, Zahoor M, Reiterer V, Phuyal S, Leoncio Netto W, Sørensen Ø, Iyer A, Sengupta D, Prasmickaite L, Mælandsmo GM, Köhn-Luque A, Farhan H. A combined experimental-computational approach uncovers a role for the Golgi matrix protein Giantin in breast cancer progression. PLoS Comput Biol 2023; 19:e1010995. [PMID: 37068117 PMCID: PMC10159355 DOI: 10.1371/journal.pcbi.1010995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 05/04/2023] [Accepted: 03/04/2023] [Indexed: 04/18/2023] Open
Abstract
Our understanding of how speed and persistence of cell migration affects the growth rate and size of tumors remains incomplete. To address this, we developed a mathematical model wherein cells migrate in two-dimensional space, divide, die or intravasate into the vasculature. Exploring a wide range of speed and persistence combinations, we find that tumor growth positively correlates with increasing speed and higher persistence. As a biologically relevant example, we focused on Golgi fragmentation, a phenomenon often linked to alterations of cell migration. Golgi fragmentation was induced by depletion of Giantin, a Golgi matrix protein, the downregulation of which correlates with poor patient survival. Applying the experimentally obtained migration and invasion traits of Giantin depleted breast cancer cells to our mathematical model, we predict that loss of Giantin increases the number of intravasating cells. This prediction was validated, by showing that circulating tumor cells express significantly less Giantin than primary tumor cells. Altogether, our computational model identifies cell migration traits that regulate tumor progression and uncovers a role of Giantin in breast cancer progression.
Collapse
Affiliation(s)
- Salim Ghannoum
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Damiano Fantini
- Department of Urology, Northwestern University, Chicago, Illinois, United States of America
| | - Muhammad Zahoor
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Veronika Reiterer
- Institute of Pathophysiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Santosh Phuyal
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Waldir Leoncio Netto
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Arvind Iyer
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Debarka Sengupta
- Department of Computational Biology, Indraprastha Institute of Information Technology, New Delhi, India
- Centre for Artificial Intelligence, Indraprastha Institute of Information Technology, Delhi, India
| | - Lina Prasmickaite
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Gunhild Mari Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hesso Farhan
- Institute of Basic Medical Sciences, Department of Molecular Medicine, University of Oslo, Oslo, Norway
- Institute of Pathophysiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
26
|
Boyer CK, Bauchle CJ, Zhang J, Wang Y, Stephens SB. Synchronized proinsulin trafficking reveals delayed Golgi export accompanies β-cell secretory dysfunction in rodent models of hyperglycemia. Sci Rep 2023; 13:5218. [PMID: 36997560 PMCID: PMC10063606 DOI: 10.1038/s41598-023-32322-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
The pancreatic islet β-cell's preference for release of newly synthesized insulin requires careful coordination of insulin exocytosis with sufficient insulin granule production to ensure that insulin stores exceed peripheral demands for glucose homeostasis. Thus, the cellular mechanisms regulating insulin granule production are critical to maintaining β-cell function. In this report, we utilized the synchronous protein trafficking system, RUSH, in primary β-cells to evaluate proinsulin transit through the secretory pathway leading to insulin granule formation. We demonstrate that the trafficking, processing, and secretion of the proinsulin RUSH reporter, proCpepRUSH, are consistent with current models of insulin maturation and release. Using both a rodent dietary and genetic model of hyperglycemia and β-cell dysfunction, we show that proinsulin trafficking is impeded at the Golgi and coincides with the decreased appearance of nascent insulin granules at the plasma membrane. Ultrastructural analysis of β-cells from diabetic leptin receptor deficient mice revealed gross morphological changes in Golgi structure, including shortened and swollen cisternae, and partial Golgi vesiculation, which are consistent with defects in secretory protein export. Collectively, this work highlights the utility of the proCpepRUSH reporter in studying proinsulin trafficking dynamics and suggests that altered Golgi export function contributes to β-cell secretory defects in the pathogenesis of Type 2 diabetes.
Collapse
Affiliation(s)
- Cierra K Boyer
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA
| | - Casey J Bauchle
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, IA, 52246, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52246, USA.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Iowa, Iowa City, IA, 52246, USA.
| |
Collapse
|
27
|
Krishna G, Santhoshkumar R, Sivakumar PT, Alladi S, Mahadevan A, Dahale AB, Arshad F, Subramanian S. Pathological (Dis)Similarities in Neuronal Exosome-Derived Synaptic and Organellar Marker Levels Between Alzheimer's Disease and Frontotemporal Dementia. J Alzheimers Dis 2023; 94:S387-S397. [PMID: 36336935 PMCID: PMC10473137 DOI: 10.3233/jad-220829] [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] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) and frontotemporal dementia (FTD) are pathologically distinct neurodegenerative disorders with certain overlap in cognitive and behavioral symptoms. Both AD and FTD are characterized by synaptic loss and accumulation of misfolded proteins, albeit, in different regions of the brain. OBJECTIVE To investigate the synaptic and organellar markers in AD and FTD through assessment of the levels of synaptic protein, neurogranin (Ng) and organellar proteins, mitofusin-2 (MFN-2), lysosomal associated membrane protein-2 (LAMP-2), and golgin A4 from neuronal exosomes. METHODS Exosomes isolated from the plasma of healthy controls (HC), AD and FTD subjects were characterized using transmission electron microscopy. Neurodegenerative status was assessed by measurement of neurofilament light chain (NfL) using Simoa. The pooled exosomal extracts from each group were analyzed for Ng, MFN-2, LAMP-2, and golgin A4 by western blot analysis using enhanced chemiluminescence method of detection. RESULTS The densitometric analysis of immunoreactive bands demonstrated a 65% reduction of Ng in AD and 53% in FTD. Mitochondrial protein MFN-2 showed a significant reduction by 32% in AD and 46% in FTD. Lysosomal LAMP-2 and Golgi complex associated golgin A4 were considerably increased in both AD and FTD. CONCLUSION Changes in Ng may reflect the ongoing synaptic degeneration that are linked to cognitive disturbances in AD and FTD. Importantly, the rate of synaptic degeneration was more pronounced in AD. Changes to a similar extent in both the dementia groups in organellar proteins indicates shared mechanisms of protein accumulation/degradation common to both AD and FTD.
Collapse
Affiliation(s)
- Geethu Krishna
- Department of Neurochemistry, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | - Rashmi Santhoshkumar
- Department of Neuropathology, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | | | - Suvarna Alladi
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | - Ajit B. Dahale
- Department of Psychiatry, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | - Faheem Arshad
- Department of Neurology, National Institute of Mental Health & Neurosciences, Bengaluru, India
| | - Sarada Subramanian
- Department of Neurochemistry, National Institute of Mental Health & Neurosciences, Bengaluru, India
| |
Collapse
|
28
|
Itokazu Y, Fuchigami T, Yu RK. Functional Impairment of the Nervous System with Glycolipid Deficiencies. ADVANCES IN NEUROBIOLOGY 2023; 29:419-448. [PMID: 36255683 PMCID: PMC9793801 DOI: 10.1007/978-3-031-12390-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Patients with nervous system disorders suffer from impaired cognitive, sensory and motor functions that greatly inconvenience their daily life and usually burdens their family and society. It is difficult to achieve functional recovery for the damaged central nervous system (CNS) because of its limited ability to regenerate. Glycosphingolipids (GSLs) are abundant in the CNS and are known to play essential roles in cell-cell recognition, adhesion, signal transduction, and cellular migration, that are crucial in all phases of neurogenesis. Despite intense investigation of CNS regeneration, the roles of GSLs in neural regeneration remain unclear. Here we focus on the respective potentials of glycolipids to promote regeneration and repair of the CNS. Mice lacking glucosylceramide, lactosylceramide or gangliosides show lethal phenotypes. More importantly, patients with ganglioside deficiencies exhibit severe clinical phenotypes. Further, neurodegenerative diseases and mental health disorders are associated with altered GSL expression. Accumulating studies demonstrate that GSLs not only delimit physical regions but also play central roles in the maintenance of the biological functions of neurons and glia. We anticipate that the ability of GSLs to modulate behavior of a variety of molecules will enable them to ameliorate biochemical and neurobiological defects in patients. The use of GSLs to treat such defects in the human CNS will be a paradigm-shift in approach since GSL-replacement therapy has not yet been achieved in this manner clinically.
Collapse
Affiliation(s)
- Yutaka Itokazu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| | - Takahiro Fuchigami
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Robert K Yu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
29
|
Haukedal H, Corsi GI, Gadekar VP, Doncheva NT, Kedia S, de Haan N, Chandrasekaran A, Jensen P, Schiønning P, Vallin S, Marlet FR, Poon A, Pires C, Agha FK, Wandall HH, Cirera S, Simonsen AH, Nielsen TT, Nielsen JE, Hyttel P, Muddashetty R, Aldana BI, Gorodkin J, Nair D, Meyer M, Larsen MR, Freude K. Golgi fragmentation - One of the earliest organelle phenotypes in Alzheimer's disease neurons. Front Neurosci 2023; 17:1120086. [PMID: 36875643 PMCID: PMC9978754 DOI: 10.3389/fnins.2023.1120086] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, with no current cure. Consequently, alternative approaches focusing on early pathological events in specific neuronal populations, besides targeting the well-studied amyloid beta (Aβ) accumulations and Tau tangles, are needed. In this study, we have investigated disease phenotypes specific to glutamatergic forebrain neurons and mapped the timeline of their occurrence, by implementing familial and sporadic human induced pluripotent stem cell models as well as the 5xFAD mouse model. We recapitulated characteristic late AD phenotypes, such as increased Aβ secretion and Tau hyperphosphorylation, as well as previously well documented mitochondrial and synaptic deficits. Intriguingly, we identified Golgi fragmentation as one of the earliest AD phenotypes, indicating potential impairments in protein processing and post-translational modifications. Computational analysis of RNA sequencing data revealed differentially expressed genes involved in glycosylation and glycan patterns, whilst total glycan profiling revealed minor glycosylation differences. This indicates general robustness of glycosylation besides the observed fragmented morphology. Importantly, we identified that genetic variants in Sortilin-related receptor 1 (SORL1) associated with AD could aggravate the Golgi fragmentation and subsequent glycosylation changes. In summary, we identified Golgi fragmentation as one of the earliest disease phenotypes in AD neurons in various in vivo and in vitro complementary disease models, which can be exacerbated via additional risk variants in SORL1.
Collapse
Affiliation(s)
- Henriette Haukedal
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Giulia I Corsi
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg, Denmark
| | - Veerendra P Gadekar
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg, Denmark
| | - Nadezhda T Doncheva
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg, Denmark.,Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Shekhar Kedia
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Noortje de Haan
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abinaya Chandrasekaran
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Pernille Schiønning
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sarah Vallin
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederik Ravnkilde Marlet
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna Poon
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Carlota Pires
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Fawzi Khoder Agha
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Anja Hviid Simonsen
- Danish Dementia Research Centre, Department of Neurology, Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Troels Tolstrup Nielsen
- Danish Dementia Research Centre, Department of Neurology, Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jørgen Erik Nielsen
- Danish Dementia Research Centre, Department of Neurology, Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Poul Hyttel
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ravi Muddashetty
- Institute for Stem Cell Science and Regenerative Medicine, Bengaluru, India
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Gorodkin
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.,Center for Non-coding RNA in Technology and Health, University of Copenhagen, Frederiksberg, Denmark
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, India
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Department of Neurology, Odense University Hospital, Odense, Denmark
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
30
|
Li J, Zhang J, Bui S, Ahat E, Kolli D, Reid W, Xing L, Wang Y. Common Assays in Mammalian Golgi Studies. Methods Mol Biol 2022; 2557:303-332. [PMID: 36512224 DOI: 10.1007/978-1-0716-2639-9_20] [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] [Indexed: 12/15/2022]
Abstract
The Golgi is a complex structure characterized by stacks of tightly aligned flat cisternae. In mammalian cells, Golgi stacks often concentrate in the perinuclear region and link together to form a ribbon. This structure is dynamic to accommodate continuous cargo flow in and out of the Golgi in both directions and undergoes morphological changes under physiological and pathological conditions. The fine, stacked Golgi structure makes it difficult to study by conventional light or even super-resolution microscopy. Furthermore, efforts to understand how Golgi structural dynamics impact cellular processes have been slow because of the knowledge gap in the protein machinery that maintains the complex and dynamic Golgi structure. In this method article, we list the common assays used in our research to help new and established researchers select the most appropriate method to properly address their questions.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Divya Kolli
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Whitney Reid
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lijuan Xing
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
31
|
Chen TT, Lin Y, Zhang S, Liu S, Song L, Zhong W, Luo ZQ, Han A. Atypical Legionella GTPase effector hijacks host vesicular transport factor p115 to regulate host lipid droplet. SCIENCE ADVANCES 2022; 8:eadd7945. [PMID: 36525490 PMCID: PMC9757750 DOI: 10.1126/sciadv.add7945] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/17/2022] [Indexed: 06/17/2023]
Abstract
The intracellular bacterial pathogen Legionella pneumophila uses hundreds of effector proteins to manipulate multiple processes of the host cells to establish a replicative niche known as Legionella-containing vacuole (LCV). Biogenesis of the LCV has been known to depend on host small guanosine triphosphatases (GTPases), but whether bacterial effector GTPases are also involved remains unknown. Here, we show that an ankyrin repeat containing effector LegA15 localizes directly in host lipid droplets (LDs), leading to Golgi apparatus fragmentation of the host cells by hijacking the host vesicular transport factor p115. LegA15 is a GTPase with a unique catalytic mechanism, unlike any eukaryotic small GTPases. Moreover, the effector LegA15 co-opts p115 to modulate homeostasis of the host LDs in its GTPase-dependent manner. Together, our data reveal that an atypical GTPase effector regulates the host LDs through impeding the vesicle secretion system of the host cells for intracellular life cycle of Legionella.
Collapse
Affiliation(s)
- Tao-Tao Chen
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences and Faculty of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- The Key Laboratory of Innate Immune Biology of Fujian Province, Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Yanling Lin
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences and Faculty of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Shijun Zhang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences and Faculty of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Shuxin Liu
- Department of Respiratory Medicine and Center of Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory of Zoonotic Diseases, The First Hospital, Jilin University, Changchun, China
| | - Lei Song
- Department of Respiratory Medicine and Center of Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory of Zoonotic Diseases, The First Hospital, Jilin University, Changchun, China
| | - Wenhong Zhong
- The Key Laboratory of Innate Immune Biology of Fujian Province, Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China
| | - Zhao-Qing Luo
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Aidong Han
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences and Faculty of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
32
|
Common Markers and Small Molecule Inhibitors in Golgi Studies. Methods Mol Biol 2022; 2557:453-493. [PMID: 36512231 PMCID: PMC10178357 DOI: 10.1007/978-1-0716-2639-9_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this chapter, we provide a detailed guide for the application of commonly used small molecules to study Golgi structure and function in vitro. Furthermore, we have curated a concise, validated list of endomembrane markers typically used in downstream assays to examine the consequent effect on the Golgi via microscopy and western blot after drug treatment. This chapter will be useful for researchers beginning their foray into the field of intracellular trafficking and Golgi biology.
Collapse
|
33
|
Parchure A, Tian M, Stalder D, Boyer CK, Bearrows SC, Rohli KE, Zhang J, Rivera-Molina F, Ramazanov BR, Mahata SK, Wang Y, Stephens SB, Gershlick DC, von Blume J. Liquid-liquid phase separation facilitates the biogenesis of secretory storage granules. J Cell Biol 2022; 221:e202206132. [PMID: 36173346 PMCID: PMC9526250 DOI: 10.1083/jcb.202206132] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 02/03/2023] Open
Abstract
Insulin is synthesized by pancreatic β-cells and stored into secretory granules (SGs). SGs fuse with the plasma membrane in response to a stimulus and deliver insulin to the bloodstream. The mechanism of how proinsulin and its processing enzymes are sorted and targeted from the trans-Golgi network (TGN) to SGs remains mysterious. No cargo receptor for proinsulin has been identified. Here, we show that chromogranin (CG) proteins undergo liquid-liquid phase separation (LLPS) at a mildly acidic pH in the lumen of the TGN, and recruit clients like proinsulin to the condensates. Client selectivity is sequence-independent but based on the concentration of the client molecules in the TGN. We propose that the TGN provides the milieu for converting CGs into a "cargo sponge" leading to partitioning of client molecules, thus facilitating receptor-independent client sorting. These findings provide a new receptor-independent sorting model in β-cells and many other cell types and therefore represent an innovation in the field of membrane trafficking.
Collapse
Affiliation(s)
- Anup Parchure
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Meng Tian
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Danièle Stalder
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Cierra K. Boyer
- Departments of Pharmacology and Neuroscience, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA
| | - Shelby C. Bearrows
- Departments of Pharmacology and Neuroscience, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA
| | - Kristen E. Rohli
- Departments of Pharmacology and Neuroscience, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
- Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA
| | - Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI
| | - Felix Rivera-Molina
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Bulat R. Ramazanov
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Sushil K. Mahata
- Department of Medicine, University of California San Diego, La Jolla, CA
- VA San Diego Healthcare System, San Diego, CA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI
| | - Samuel B. Stephens
- Departments of Pharmacology and Neuroscience, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA
| | - David C. Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Julia von Blume
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
34
|
Fasano G, Muto V, Radio FC, Venditti M, Mosaddeghzadeh N, Coppola S, Paradisi G, Zara E, Bazgir F, Ziegler A, Chillemi G, Bertuccini L, Tinari A, Vetro A, Pantaleoni F, Pizzi S, Conti LA, Petrini S, Bruselles A, Prandi IG, Mancini C, Chandramouli B, Barth M, Bris C, Milani D, Selicorni A, Macchiaiolo M, Gonfiantini MV, Bartuli A, Mariani R, Curry CJ, Guerrini R, Slavotinek A, Iascone M, Dallapiccola B, Ahmadian MR, Lauri A, Tartaglia M. Dominant ARF3 variants disrupt Golgi integrity and cause a neurodevelopmental disorder recapitulated in zebrafish. Nat Commun 2022; 13:6841. [PMID: 36369169 PMCID: PMC9652361 DOI: 10.1038/s41467-022-34354-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Vesicle biogenesis, trafficking and signaling via Endoplasmic reticulum-Golgi network support essential developmental processes and their disruption lead to neurodevelopmental disorders and neurodegeneration. We report that de novo missense variants in ARF3, encoding a small GTPase regulating Golgi dynamics, cause a developmental disease in humans impairing nervous system and skeletal formation. Microcephaly-associated ARF3 variants affect residues within the guanine nucleotide binding pocket and variably perturb protein stability and GTP/GDP binding. Functional analysis demonstrates variably disruptive consequences of ARF3 variants on Golgi morphology, vesicles assembly and trafficking. Disease modeling in zebrafish validates further the dominant behavior of the mutants and their differential impact on brain and body plan formation, recapitulating the variable disease expression. In-depth in vivo analyses traces back impaired neural precursors' proliferation and planar cell polarity-dependent cell movements as the earliest detectable effects. Our findings document a key role of ARF3 in Golgi function and demonstrate its pleiotropic impact on development.
Collapse
Affiliation(s)
- Giulia Fasano
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Valentina Muto
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Francesca Clementina Radio
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Martina Venditti
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Niloufar Mosaddeghzadeh
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Simona Coppola
- grid.416651.10000 0000 9120 6856National Center for Rare Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Graziamaria Paradisi
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy ,grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Erika Zara
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy ,grid.7841.aDepartment of Biology and Biotechnology “Charles Darwin”, Università “Sapienza”, Rome, 00185 Italy
| | - Farhad Bazgir
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Alban Ziegler
- grid.7252.20000 0001 2248 3363UFR Santé de l’Université d’Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France ,grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Giovanni Chillemi
- grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy ,grid.5326.20000 0001 1940 4177Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Centro Nazionale delle Ricerche, 70126 Bari, Italy
| | - Lucia Bertuccini
- grid.416651.10000 0000 9120 6856Servizio grandi strumentazioni e core facilities, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Antonella Tinari
- grid.416651.10000 0000 9120 6856Centro di riferimento per la medicina di genere, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Annalisa Vetro
- grid.8404.80000 0004 1757 2304Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children’s Hospital, University of Florence, 50139 Florence, Italy
| | - Francesca Pantaleoni
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Simone Pizzi
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Libenzio Adrian Conti
- grid.414603.4Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Stefania Petrini
- grid.414603.4Confocal Microscopy Core Facility, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Alessandro Bruselles
- grid.416651.10000 0000 9120 6856Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Ingrid Guarnetti Prandi
- grid.12597.380000 0001 2298 9743Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, 01100 Viterbo, Italy
| | - Cecilia Mancini
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Balasubramanian Chandramouli
- grid.431603.30000 0004 1757 1950Super Computing Applications and Innovation, CINECA, 40033 Casalecchio di Reno, Italy
| | - Magalie Barth
- grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Céline Bris
- grid.7252.20000 0001 2248 3363UFR Santé de l’Université d’Angers, INSERM U1083, CNRS UMR6015, MITOVASC, SFR ICAT, F-49000 Angers, France ,grid.411147.60000 0004 0472 0283Département de Génétique, CHU d’Angers, 49000 Angers, France
| | - Donatella Milani
- grid.414818.00000 0004 1757 8749Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Angelo Selicorni
- grid.512106.1Mariani Center for Fragile Children Pediatric Unit, Azienda Socio Sanitaria Territoriale Lariana, 22100 Como, Italy
| | - Marina Macchiaiolo
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Michaela V. Gonfiantini
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Andrea Bartuli
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Riccardo Mariani
- grid.414603.4Department of Laboratories Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Cynthia J. Curry
- grid.266102.10000 0001 2297 6811Genetic Medicine, Dept of Pediatrics, University of California San Francisco, Ca, Fresno, Ca, San Francisco, CA 94143 USA
| | - Renzo Guerrini
- grid.8404.80000 0004 1757 2304Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Meyer Children’s Hospital, University of Florence, 50139 Florence, Italy
| | - Anne Slavotinek
- grid.266102.10000 0001 2297 6811Genetic Medicine, Dept of Pediatrics, University of California San Francisco, Ca, Fresno, Ca, San Francisco, CA 94143 USA
| | - Maria Iascone
- grid.460094.f0000 0004 1757 8431Medical Genetics, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Bruno Dallapiccola
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Mohammad Reza Ahmadian
- grid.411327.20000 0001 2176 9917Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Antonella Lauri
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| | - Marco Tartaglia
- grid.414603.4Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146 Rome, Italy
| |
Collapse
|
35
|
Sato Y, Tsuyusaki M, Takahashi-Iwanaga H, Fujisawa R, Masamune A, Hamada S, Matsumoto R, Tanaka Y, Kakuta Y, Yamaguchi-Kabata Y, Furuse T, Wakana S, Shimura T, Kobayashi R, Shinoda Y, Goitsuka R, Maezawa S, Sadakata T, Sano Y, Furuichi T. Loss of CAPS2/Cadps2 leads to exocrine pancreatic cell injury and intracellular accumulation of secretory granules in mice. Front Mol Biosci 2022; 9:1040237. [DOI: 10.3389/fmolb.2022.1040237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
The type 2 Ca2+-dependent activator protein for secretion (CAPS2/CADPS2) regulates dense-core vesicle trafficking and exocytosis and is involved in the regulated release of catecholamines, peptidergic hormones, and neuromodulators. CAPS2 is expressed in the pancreatic exocrine acinar cells that produce and secrete digestive enzymes. However, the functional role of CAPS2 in vesicular trafficking and/or exocytosis of non-regulatory proteins in the exocrine pancreas remains to be determined. Here, we analyzed the morpho-pathological indicators of the pancreatic exocrine pathway in Cadps2-deficient mouse models using histochemistry, biochemistry, and electron microscopy. We used whole exosome sequencing to identify CADPS2 variants in patients with chronic pancreatitis (CP). Caps2/Cadps2-knockout (KO) mice exhibited morphophysiological abnormalities in the exocrine pancreas, including excessive accumulation of secretory granules (zymogen granules) and their amylase content in the cytoplasm, deterioration of the fine intracellular membrane structures (disorganized rough endoplasmic reticulum, dilated Golgi cisternae, and the appearance of empty vesicles and autophagic-like vacuoles), as well as exocrine pancreatic cell injury, including acinar cell atrophy, increased fibrosis, and inflammatory cell infiltration. Pancreas-specific Cadps2 conditional KO mice exhibited pathological abnormalities in the exocrine pancreas similar to the global Cadps2 KO mice, indicating that these phenotypes were caused either directly or indirectly by CAPS2 deficiency in the pancreas. Furthermore, we identified a rare variant in the exon3 coding region of CADPS2 in a non-alcoholic patient with CP and showed that Cadps2-dex3 mice lacking CAPS2 exon3 exhibited symptoms similar to those exhibited by the Cadps2 KO and cKO mice. These results suggest that CAPS2 is critical for the proper functioning of the pancreatic exocrine pathway, and its deficiency is associated with a risk of pancreatic acinar cell pathology.
Collapse
|
36
|
Wang Y, Gandy S. The Golgi apparatus: Site for convergence of COVID-19 brain fog and Alzheimer's disease? Mol Neurodegener 2022; 17:67. [PMID: 36271398 PMCID: PMC9587685 DOI: 10.1186/s13024-022-00568-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/07/2022] [Accepted: 09/15/2022] [Indexed: 11/10/2022] Open
Affiliation(s)
- Yanzhuang Wang
- Department of Molecular, Cellular, and Developmental Biology and Department of Neurology, University of Michigan, 48109, Ann Arbor, MI, USA.
| | - Sam Gandy
- Departments of Neurology and Psychiatry and the Mount Sinai Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, 10029, New York, NY, USA. .,The James J Peters VA Medical Center, 10468, Bronx, NY, USA.
| |
Collapse
|
37
|
Sengupta R, Mihelc EM, Angel S, Lanman JK, Kuhn RJ, Stahelin RV. Contribution of the Golgi apparatus in morphogenesis of a virus-induced cytopathic vacuolar system. Life Sci Alliance 2022; 5:5/10/e202000887. [PMID: 36137747 PMCID: PMC9500387 DOI: 10.26508/lsa.202000887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/24/2022] Open
Abstract
Electron tomography reveals four classes of cytopathic vesicles-II (CPV-II) stemming from the host Golgi apparatus after Venezuelan equine encephalitis virus infection. The Golgi apparatus (GA) in mammalian cells is pericentrosomally anchored and exhibits a stacked architecture. During infections by members of the alphavirus genus, the host cell GA is thought to give rise to distinct mobile pleomorphic vacuoles known as CPV-II (cytopathic vesicle-II) via unknown morphological steps. To dissect this, we adopted a phased electron tomography approach to image multiple overlapping volumes of a cell infected with Venezuelan equine encephalitis virus (VEEV) and complemented it with localization of a peroxidase-tagged Golgi marker. Analysis of the tomograms revealed a pattern of progressive cisternal bending into double-lamellar vesicles as a central process underpinning the biogenesis and the morphological complexity of this vacuolar system. Here, we propose a model for the conversion of the GA to CPV-II that reveals a unique pathway of intracellular virus envelopment. Our results have implications for alphavirus-induced displacement of Golgi cisternae to the plasma membrane to aid viral egress operating late in the infection cycle.
Collapse
Affiliation(s)
- Ranjan Sengupta
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA .,Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Elaine M Mihelc
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Stephanie Angel
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.,Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Jason K Lanman
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.,The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Robert V Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA .,The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
38
|
Choi W, Kang S, Kim J. New insights into the role of the Golgi apparatus in the pathogenesis and therapeutics of human diseases. Arch Pharm Res 2022; 45:671-692. [PMID: 36178581 DOI: 10.1007/s12272-022-01408-z] [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: 06/22/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022]
Abstract
The Golgi apparatus is an essential cellular organelle that mediates homeostatic functions, including vesicle trafficking and the post-translational modification of macromolecules. Its unique stacked structure and dynamic functions are tightly regulated, and several Golgi proteins play key roles in the functioning of unconventional protein secretory pathways triggered by cellular stress responses. Recently, an increasing number of studies have implicated defects in Golgi functioning in human diseases such as cancer, neurodegenerative, and immunological disorders. Understanding the extraordinary characteristics of Golgi proteins is important for elucidating its associated intracellular signaling mechanisms and has important ramifications for human health. Therefore, analyzing the mechanisms by which the Golgi participates in disease pathogenesis may be useful for developing novel therapeutic strategies. This review articulates the structural features and abnormalities of the Golgi apparatus reported in various diseases and the suspected mechanisms underlying the Golgi-associated pathologies. Furthermore, we review the potential therapeutic strategies based on Golgi function.
Collapse
Affiliation(s)
- Wooseon Choi
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Shinwon Kang
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Jiyoon Kim
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
39
|
Rohli KE, Boyer CK, Bearrows SC, Moyer MR, Elison WS, Bauchle CJ, Blom SE, Zhang J, Wang Y, Stephens SB. ER Redox Homeostasis Regulates Proinsulin Trafficking and Insulin Granule Formation in the Pancreatic Islet β-Cell. FUNCTION 2022; 3:zqac051. [PMID: 36325514 PMCID: PMC9614934 DOI: 10.1093/function/zqac051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/11/2022] [Accepted: 09/21/2022] [Indexed: 01/07/2023] Open
Abstract
Defects in the pancreatic β-cell's secretion system are well-described in type 2 diabetes (T2D) and include impaired proinsulin processing and a deficit in mature insulin-containing secretory granules; however, the cellular mechanisms underlying these defects remain poorly understood. To address this, we used an in situ fluorescent pulse-chase strategy to study proinsulin trafficking. We show that insulin granule formation and the appearance of nascent granules at the plasma membrane are decreased in rodent and cell culture models of prediabetes and hyperglycemia. Moreover, we link the defect in insulin granule formation to an early trafficking delay in endoplasmic reticulum (ER) export of proinsulin, which is independent of overt ER stress. Using a ratiometric redox sensor, we show that the ER becomes hyperoxidized in β-cells from a dietary model of rodent prediabetes and that addition of reducing equivalents restores ER export of proinsulin and insulin granule formation and partially restores β-cell function. Together, these data identify a critical role for the regulation of ER redox homeostasis in proinsulin trafficking and suggest that alterations in ER redox poise directly contribute to the decline in insulin granule production in T2D. This model highlights a critical link between alterations in ER redox and ER function with defects in proinsulin trafficking in T2D. Hyperoxidation of the ER lumen, shown as hydrogen peroxide, impairs proinsulin folding and disulfide bond formation that prevents efficient exit of proinsulin from the ER to the Golgi. This trafficking defect limits available proinsulin for the formation of insulin secretory granules during the development of T2D.
Collapse
Affiliation(s)
- Kristen E Rohli
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Shelby C Bearrows
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Marshall R Moyer
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Weston S Elison
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA
| | - Casey J Bauchle
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Sandra E Blom
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, MI 48103, USA
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
40
|
Sou YS, Yamaguchi J, Kameda H, Masuda K, Maeda Y, Uchiyama Y, Koike M. GPHR-mediated acidification of the Golgi lumen is essential for cholesterol biosynthesis in the brain. FEBS Lett 2022; 596:2873-2888. [PMID: 36056653 DOI: 10.1002/1873-3468.14491] [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: 07/11/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/06/2022]
Abstract
The Golgi pH regulator (GPHR) is essential for maintaining the function and morphology of the Golgi apparatus through the regulation of luminal acidic pH. Abnormal morphology of the Golgi apparatus is associated with neurodegenerative diseases. Here, we found that knockout of GPHR in the mouse brain led to morphological changes in the Golgi apparatus and neurodegeneration, which included brain atrophy, neuronal cell death, and gliosis. Furthermore, in the GPHR knockout mouse brain, transcriptional activity of sterol regulatory element-binding protein 2 (SREBP2) decreased, resulting in a reduction in cholesterol levels. GPHR-deficient cells exhibited suppressed neurite outgrowth, which was recovered by exogenous expression of the active form of SREBP2. Our results show that GPHR-mediated luminal acidification of the Golgi apparatus maintains proper cholesterol levels and, thereby, neuronal morphology.
Collapse
Affiliation(s)
- Yu-Shin Sou
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Junji Yamaguchi
- Laboratory of Morphology and Image Analysis, Research Support Center, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan.,Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Hiroshi Kameda
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Keisuke Masuda
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Yusuke Maeda
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
41
|
Haure-Mirande JV, Audrain M, Ehrlich ME, Gandy S. Microglial TYROBP/DAP12 in Alzheimer's disease: Transduction of physiological and pathological signals across TREM2. Mol Neurodegener 2022; 17:55. [PMID: 36002854 PMCID: PMC9404585 DOI: 10.1186/s13024-022-00552-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 06/27/2022] [Indexed: 02/01/2023] Open
Abstract
TYROBP (also known as DAP12 or KARAP) is a transmembrane adaptor protein initially described as a receptor-activating subunit component of natural killer (NK) cells. TYROBP is expressed in numerous cell types, including peripheral blood monocytes, macrophages, dendritic cells, and osteoclasts, but a key point of recent interest is related to the critical role played by TYROBP in the function of many receptors expressed on the plasma membrane of microglia. TYROBP is the downstream adaptor and putative signaling partner for several receptors implicated in Alzheimer's disease (AD), including SIRP1β, CD33, CR3, and TREM2. TYROBP has received much of its current notoriety because of its importance in brain homeostasis by signal transduction across those receptors. In this review, we provide an overview of evidence indicating that the biology of TYROBP extends beyond its interaction with these four ligand-binding ectodomain-intramembranous domain molecules. In addition to reviewing the structure and localization of TYROBP, we discuss our recent progress using mouse models of either cerebral amyloidosis or tauopathy that were engineered to be TYROBP-deficient or TYROBP-overexpressing. Remarkably, constitutively TYROBP-deficient mice provided a model of genetic resilience to either of the defining proteinopathies of AD. Learning behavior and synaptic electrophysiological function were preserved at normal physiological levels even in the face of robust cerebral amyloidosis (in APP/PSEN1;Tyrobp-/- mice) or tauopathy (in MAPTP301S;Tyrobp-/- mice). A fundamental underpinning of the functional synaptic dysfunction associated with each proteotype was an accumulation of complement C1q. TYROBP deficiency prevented C1q accumulation associated with either proteinopathy. Based on these data, we speculate that TYROBP plays a key role in the microglial sensome and the emergence of the disease-associated microglia (DAM) phenotype. TYROBP may also play a key role in the loss of markers of synaptic integrity (e.g., synaptophysin-like immunoreactivity) that has long been held to be the feature of human AD molecular neuropathology that most closely correlates with concurrent clinical cognitive function.
Collapse
Affiliation(s)
| | - Mickael Audrain
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Department of Psychiatry and the NIA-Designated Mount Sinai Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- James J Peters VA Medical Center, New York, Bronx NY 10468 USA
| |
Collapse
|
42
|
He Z, Liu D, Liu Y, Li X, Shi W, Ma H. Golgi-Targeted Fluorescent Probe for Imaging NO in Alzheimer's Disease. Anal Chem 2022; 94:10256-10262. [PMID: 35815650 DOI: 10.1021/acs.analchem.2c01885] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) is a crucial neurotransmitter participating in many biological processes via nitrosylation reaction. NO produced in diverse subcellular regions also regulates the function of cells in different manners. A Golgi apparatus is rich in nitric oxide synthase and may serve as a potential therapeutic target for Alzheimer's disease (AD). However, due to the lack of an effective tool, it is difficult to reveal the relationship between Golgi-NO and AD. Herein, we report Golgi-NO as the first Golgi-targeted fluorescent probe for sensing and imaging NO in the Golgi apparatus. The probe is designed and synthesized by incorporating 4-sulfamoylphenylamide as a Golgi-targeted moiety to 6-carboxyrhodamine B, generating a fluorophore of Golgi-RhB with modifiable carboxyl, which is then combined with the NO recognition moiety of o-diaminobenzene. The probe shows superior analytical performance including accurate Golgi-targeted ability and high selectivity for NO. Moreover, using the probe, we disclose a significant increase of NO in Golgi apparatus in the AD model. This study provides a competent tool for studying the function and nitrosylation of NO in the Golgi apparatus in related diseases.
Collapse
Affiliation(s)
- Zixu He
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Diankai Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Ya Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohua Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wen Shi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huimin Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
43
|
Suga K, Yamamoto-Hijikata S, Terao Y, Akagawa K, Ushimaru M. Golgi stress induces upregulation of the ER-Golgi SNARE Syntaxin-5, altered βAPP processing, and Caspase-3-dependent apoptosis in NG108-15 cells. Mol Cell Neurosci 2022; 121:103754. [PMID: 35842170 DOI: 10.1016/j.mcn.2022.103754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/29/2022] [Accepted: 07/07/2022] [Indexed: 01/06/2023] Open
Abstract
The involvement of secretory pathways and Golgi dysfunction in neuronal cells during Alzheimer's disease progression is poorly understood. Our previous overexpression and knockdown studies revealed that the intracellular protein level of Syntaxin-5, an endoplasmic reticulum-Golgi soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE), modulates beta-amyloid precursor protein processing in neuronal cells. We recently showed that changes in endogenous Syntaxin-5 protein expression occur under stress induction. Syntaxin-5 was upregulated by endoplasmic reticulum stress but was degraded by Caspase-3 during apoptosis in neuronal cells. In addition, we showed that sustained endoplasmic reticulum stress promotes Caspase-3-dependent apoptosis during the later phase of the endoplasmic reticulum stress response in NG108-15 cells. In this study, to elucidate the consequences of secretory pathway dysfunction in beta-amyloid precursor protein processing that lead to neuronal cell death, we examined the effect of various stresses on endoplasmic reticulum-Golgi SNARE expression and beta-amyloid precursor protein processing. By using compounds to disrupt Golgi function, we show that Golgi stress promotes upregulation of the endoplasmic reticulum-Golgi SNARE Syntaxin-5, and prolonged stress causes Caspase-3-dependent apoptosis. Golgi stress induced intracellular beta-amyloid precursor protein accumulation and a concomitant decrease in total amyloid-beta production. We also examined the protective effect of the chemical chaperone 4-phenylbutylate on changes in amyloid-beta production and the activation of Caspase-3 induced by endoplasmic reticulum and Golgi stress. The compound alleviated the increase in the amyloid-beta 1-42/amyloid-beta 1-40 ratio induced by endoplasmic reticulum and Golgi stress. Furthermore, 4-phenylbutylate could rescue Caspase-3-dependent apoptosis induced by prolonged organelle stress. These results suggest that organelle stress originating from the endoplasmic reticulum and Golgi has a substantial impact on the amyloidogenic processing of beta-amyloid precursor protein and Caspase-3-dependent apoptosis, leading to neuronal cell death.
Collapse
Affiliation(s)
- Kei Suga
- Department of Chemistry, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan; Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan.
| | | | - Yasuo Terao
- Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Kimio Akagawa
- Department of Medical Physiology, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| | - Makoto Ushimaru
- Department of Chemistry, Kyorin University, Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan
| |
Collapse
|
44
|
The Golgi complex: An organelle that determines urothelial cell biology in health and disease. Histochem Cell Biol 2022; 158:229-240. [PMID: 35773494 PMCID: PMC9399047 DOI: 10.1007/s00418-022-02121-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 12/05/2022]
Abstract
The Golgi complex undergoes considerable structural remodeling during differentiation of urothelial cells in vivo and in vitro. It is known that in a healthy bladder the differentiation from the basal to the superficial cell layer leads to the formation of the tightest barrier in our body, i.e., the blood–urine barrier. In this process, urothelial cells start expressing tight junctional proteins, apical membrane lipids, surface glycans, and integral membrane proteins, the uroplakins (UPs). The latter are the most abundant membrane proteins in the apical plasma membrane of differentiated superficial urothelial cells (UCs) and, in addition to well-developed tight junctions, contribute to the permeability barrier by their structural organization and by hindering endocytosis from the apical plasma membrane. By studying the transport of UPs, we were able to demonstrate their differentiation-dependent effect on the Golgi architecture. Although fragmentation of the Golgi complex is known to be associated with mitosis and apoptosis, we found that the process of Golgi fragmentation is required for delivery of certain specific urothelial differentiation cargoes to the plasma membrane as well as for cell–cell communication. In this review, we will discuss the currently known contribution of the Golgi complex to the formation of the blood–urine barrier in normal UCs and how it may be involved in the loss of the blood–urine barrier in cancer. Some open questions related to the Golgi complex in the urothelium will be highlighted.
Collapse
|
45
|
Page KM, McCormack JJ, Lopes-da-Silva M, Patella F, Harrison-Lavoie K, Burden JJ, Quah YYB, Scaglioni D, Ferraro F, Cutler DF. Structure modeling hints at a granular organization of the Golgi ribbon. BMC Biol 2022; 20:111. [PMID: 35549945 PMCID: PMC9102599 DOI: 10.1186/s12915-022-01305-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND In vertebrate cells, the Golgi functional subunits, mini-stacks, are linked into a tri-dimensional network. How this "ribbon" architecture relates to Golgi functions remains unclear. Are all connections between mini-stacks equal? Is the local structure of the ribbon of functional importance? These are difficult questions to address, without a quantifiable readout of the output of ribbon-embedded mini-stacks. Endothelial cells produce secretory granules, the Weibel-Palade bodies (WPB), whose von Willebrand Factor (VWF) cargo is central to hemostasis. The Golgi apparatus controls WPB size at both mini-stack and ribbon levels. Mini-stack dimensions delimit the size of VWF "boluses" whilst the ribbon architecture allows their linear co-packaging, thereby generating WPBs of different lengths. This Golgi/WPB size relationship suits mathematical analysis. RESULTS WPB lengths were quantized as multiples of the bolus size and mathematical modeling simulated the effects of different Golgi ribbon organizations on WPB size, to be compared with the ground truth of experimental data. An initial simple model, with the Golgi as a single long ribbon composed of linearly interlinked mini-stacks, was refined to a collection of mini-ribbons and then to a mixture of mini-stack dimers plus long ribbon segments. Complementing these models with cell culture experiments led to novel findings. Firstly, one-bolus sized WPBs are secreted faster than larger secretory granules. Secondly, microtubule depolymerization unlinks the Golgi into equal proportions of mini-stack monomers and dimers. Kinetics of binding/unbinding of mini-stack monomers underpinning the presence of stable dimers was then simulated. Assuming that stable mini-stack dimers and monomers persist within the ribbon resulted in a final model that predicts a "breathing" arrangement of the Golgi, where monomer and dimer mini-stacks within longer structures undergo continuous linking/unlinking, consistent with experimentally observed WPB size distributions. CONCLUSIONS Hypothetical Golgi organizations were validated against a quantifiable secretory output. The best-fitting Golgi model, accounting for stable mini-stack dimers, is consistent with a highly dynamic ribbon structure, capable of rapid rearrangement. Our modeling exercise therefore predicts that at the fine-grained level the Golgi ribbon is more complex than generally thought. Future experiments will confirm whether such a ribbon organization is endothelial-specific or a general feature of vertebrate cells.
Collapse
Affiliation(s)
- Karen M. Page
- Department of Mathematics, University College London, Gower Street, London, WC1E 6BT UK
| | - Jessica J. McCormack
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Mafalda Lopes-da-Silva
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
- Current address: iNOVA4Health, CEDOC-Chronic Diseases Research Center, NOVA Medical School, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Francesca Patella
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
- Current address: Kinomica, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG UK
| | - Kimberly Harrison-Lavoie
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Jemima J. Burden
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Ying-Yi Bernadette Quah
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Dominic Scaglioni
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| | - Francesco Ferraro
- Department of Biology and Evolution of Marine Organisms, BEOM, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Naples, Italy
| | - Daniel F. Cutler
- MRC Laboratory for Molecular cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
46
|
Tan W, Zhang Q, Quiñones-Frías MC, Hsu AY, Zhang Y, Rodal A, Hong P, Luo HR, Xu B. Enzyme-Responsive Peptide Thioesters for Targeting Golgi Apparatus. J Am Chem Soc 2022; 144:6709-6713. [PMID: 35404599 PMCID: PMC9069992 DOI: 10.1021/jacs.2c02238] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The Golgi apparatus (GA) is the hub of intracellular trafficking, but selectively targeting GA remains a challenge. We show an unconventional types of peptide thioesters, consisting of an aminoethyl thioester and acting as substrates of thioesterases, for instantly targeting the GA of cells. The peptide thioesters, above or below their critical micelle concentrations, enter cells mainly via caveolin-mediated endocytosis or macropinocytosis, respectively. After being hydrolyzed by GA-associated thioesterases, the resulting thiopeptides form dimers and accumulate in the GA. After saturating the GA, the thiopeptides are enriched in the endoplasmic reticulum (ER). Their buildup in ER and GA disrupts protein trafficking, thus leading to cell death via multiple pathways. The peptide thioesters target the GA of a wide variety of cells, including human, murine, and Drosophila cells. Changing d-diphenylalanine to l-diphenylalanine in the peptide maintains the GA-targeting ability. In addition, targeting GA redirects protein (e.g., NRAS) distribution. This work illustrates a thioesterase-responsive and redox-active molecular platform for targeting the GA and controlling cell fates.
Collapse
Affiliation(s)
- Weiyi Tan
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Qiuxin Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | | | - Alan Y. Hsu
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Children’s Hospital Boston and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Yichi Zhang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| | - Avital Rodal
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Pengyu Hong
- Department of Computer Science, Brandeis University, Waltham, MA 02453, USA
| | - Hongbo R. Luo
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Children’s Hospital Boston and Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02453, USA
| |
Collapse
|
47
|
Ahat E, Song Y, Xia K, Reid W, Li J, Bui S, Zhang F, Linhardt RJ, Wang Y. GRASP depletion-mediated Golgi fragmentation impairs glycosaminoglycan synthesis, sulfation, and secretion. Cell Mol Life Sci 2022; 79:199. [PMID: 35312866 PMCID: PMC9164142 DOI: 10.1007/s00018-022-04223-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/02/2022] [Accepted: 02/25/2022] [Indexed: 12/14/2022]
Abstract
Synthesis of glycosaminoglycans, such as heparan sulfate (HS) and chondroitin sulfate (CS), occurs in the lumen of the Golgi, but the relationship between Golgi structural integrity and glycosaminoglycan synthesis is not clear. In this study, we disrupted the Golgi structure by knocking out GRASP55 and GRASP65 and determined its effect on the synthesis, sulfation, and secretion of HS and CS. We found that GRASP depletion increased HS synthesis while decreasing CS synthesis in cells, altered HS and CS sulfation, and reduced both HS and CS secretion. Using proteomics, RNA-seq and biochemical approaches, we identified EXTL3, a key enzyme in the HS synthesis pathway, whose level is upregulated in GRASP knockout cells; while GalNAcT1, an essential CS synthesis enzyme, is robustly reduced. In addition, we found that GRASP depletion decreased HS sulfation via the reduction of PAPSS2, a bifunctional enzyme in HS sulfation. Our study provides the first evidence that Golgi structural defect may significantly alter the synthesis and secretion of glycosaminoglycans.
Collapse
Affiliation(s)
- Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI, 48109-1085, USA
| | - Yuefan Song
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Whitney Reid
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI, 48109-1085, USA
| | - Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI, 48109-1085, USA
| | - Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI, 48109-1085, USA
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, USA.
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, 1105 North University Avenue, Ann Arbor, MI, 48109-1085, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
48
|
Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD, Gao LC. The key roles of organelles and ferroptosis in Alzheimer's disease. J Neurosci Res 2022; 100:1257-1280. [PMID: 35293012 DOI: 10.1002/jnr.25033] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD), an age-related neurodegenerative disease, is a striking global health problem. Ferroptosis is a newly discovered form of cell death characterized by iron-dependent lipid peroxidation products and the accumulation of lethal reactive oxygen species. Strict regulation of iron metabolism is essential to ensure neuronal homeostasis. Excess and deficiency of iron are both associated with neurodegeneration. Studies have shown that oxidative stress caused by cerebral iron metabolism disorders in the body is involved in the process of AD, ferroptosis may play an important role in the pathogenesis of AD, and regulating ferroptosis is expected to be a new direction for the treatment of AD. Various organelles are closely related to ferroptosis: mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosome are involved in the regulation of ferroptosis from the aspects of iron metabolism and redox imbalance. In this review, the relationship between AD and the dysfunction of organelles (including mitochondria, endoplasmic reticulum, lysosome, and Golgi apparatus) and the role of organelles in ferroptosis of AD were reviewed to provide insights for understanding the relationship between organelles and ferroptosis in AD and the treatment of AD.
Collapse
Affiliation(s)
- Hui-Zhi Long
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Yan Cheng
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Zi-Wei Zhou
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Li-Chen Gao
- School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
49
|
Chang HY, Yang WY. Golgi quality control and autophagy. IUBMB Life 2022; 74:361-370. [PMID: 35274438 DOI: 10.1002/iub.2611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 02/12/2022] [Indexed: 11/09/2022]
Abstract
Organelles can easily be disrupted by intracellular and extracellular factors. Studies on ER and mitochondria indicate that a wide range of responses are elicited upon organelle disruption. One response thought to be of particular importance is autophagy. Cells can target entire organelles into autophagosomes for removal. This wholesale nature makes autophagy a robust means for eliminating compromised organelles. Recently, it was demonstrated that the Golgi apparatus is a substrate of autophagy. On the other hand, various reports have shown that components traffic away from the Golgi for elimination in an autophagosome-independent manner when the Golgi apparatus is stressed. Future studies will reveal how these different pieces of machinery coordinate to drive Golgi degradation. Quantitative measurements will be needed to determine how much autophagy contributes to the maintenance of the Golgi apparatus.
Collapse
Affiliation(s)
- Hsiang-Yi Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Wei Yuan Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, College of Life Sciences, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
50
|
Gandy S. A letter concerning "Aducanumab: What About the Patient?". Ann Neurol 2022; 91:732-733. [PMID: 35178727 DOI: 10.1002/ana.26333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/01/2022] [Accepted: 01/05/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Sam Gandy
- Departments of Neurology and Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, 10029.,Research and Development Service, James J Peters VA Medical Center, Bronx, New York, 10468
| |
Collapse
|