1
|
Zavala-Vargas DI, Visoso-Carbajal G, Cedillo-Barrón L, Filisola-Villaseñor JG, Rosales-Ramirez R, Ludert JE, Morales-Ríos E. Interaction of the Zika virus with the cytoplasmic dynein-1. Virol J 2023; 20:43. [PMID: 36879270 PMCID: PMC9987375 DOI: 10.1186/s12985-023-01992-6] [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: 12/19/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Zika virus (ZIKV) infection is a major public health threat, making the study of its biology a matter of great importance. By analyzing the viral-host protein interactions, new drug targets may be proposed. In this work, we showed that human cytoplasmic dynein-1 (Dyn) interacts with the envelope protein (E) of ZIKV. Biochemical evidence indicates that the E protein and the dimerization domain of the heavy chain of Dyn binds directly without dynactin or any cargo adaptor. Analysis of this interactions in infected Vero cells by proximity ligation assay suggest that the E-Dyn interaction is dynamic and finely tuned along the replication cycle. Altogether, our results suggest new steps in the replication cycle of the ZIKV for virion transport and indicate a suitable molecular target to modulate infection by ZIKV.
Collapse
Affiliation(s)
- Dan Israel Zavala-Vargas
- Department of Biochemistry, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | - Giovani Visoso-Carbajal
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | - Leticia Cedillo-Barrón
- Department of Molecular Biomedicine, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | | | - Romel Rosales-Ramirez
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | - Juan E Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico
| | - Edgar Morales-Ríos
- Department of Biochemistry, Center for Research and Advanced Studies (Cinvestav), 07360, Mexico City, Mexico.
| |
Collapse
|
2
|
Yuan J, Huang X, Zhao Y, Gu J, Yuan Y, Liu Z, Zou H, Bian J. Rat Hepatocytes Mitigate Cadmium Toxicity by Forming Annular Gap Junctions and Degrading Them via Endosome-Lysosome Pathway. Int J Mol Sci 2022; 23:ijms232415607. [PMID: 36555247 PMCID: PMC9778680 DOI: 10.3390/ijms232415607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Gap junction protein connexin 43 (Cx43) plays a critical role in gap junction communication in rat hepatocytes. However, those located between hepatocytes are easily internalized following exposure to poisons. Herein, we investigated the potential of buffalo rat liver 3A (BRL 3A) cells to generate annular gap junctions (AGJs) proficient at alleviating cadmium (Cd) cytotoxic injury through degradation via an endosome-lysosome pathway. Our results showed that Cd-induced damage of liver microtubules promoted Cx43 internalization and increased Cx43 phosphorylation at Ser373 site. Furthermore, we established that Cd induced AGJs generation in BRL 3A cells, and AGJs were subsequently degraded through the endosome-lysosome pathway. Overall, our results suggested that Cx43 internalization and the generation of AGJs were cellular protective mechanisms to alleviate Cd toxicity in rat hepatocytes.
Collapse
Affiliation(s)
- Junzhao Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xiaoqian Huang
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yumeng Zhao
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.Z.); (J.B.)
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: (H.Z.); (J.B.)
| |
Collapse
|
3
|
van der Horst J, Rognant S, Hellsten Y, Aalkjær C, Jepps TA. Dynein Coordinates β2-Adrenoceptor-Mediated Relaxation in Normotensive and Hypertensive Rat Mesenteric Arteries. Hypertension 2022; 79:2214-2227. [PMID: 35929419 DOI: 10.1161/hypertensionaha.122.19351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The voltage-gated potassium channel (Kv)7.4 and Kv7.5 channels contribute to the β-adrenoceptor-mediated vasodilatation. In arteries from hypertensive rodents, the Kv7.4 channel is downregulated and function attenuated, which contributes to the reduced β-adrenoceptor-mediated vasodilatation observed in these arteries. Recently, we showed that disruption of the microtubule network, with colchicine, or inhibition of the microtubule motor protein, dynein, with ciliobrevin D, enhanced the membrane abundance and function of Kv7.4 channels in rat mesenteric arteries. This study aimed to determine whether these pharmacological compounds can improve Kv7.4 function in third-order mesenteric arteries from the spontaneously hypertensive rat, thereby restoring the β-adrenoceptor-mediated vasodilatation. METHODS Wire and intravital myography was performed on normotensive and hypertensive male rat mesenteric arteries and immunostaining was performed on isolated smooth muscle cells from the same arteries. RESULTS Using wire and intravital microscopy, we show that ciliobrevin D enhanced the β-adrenoceptor-mediated vasodilatation by isoprenaline. This effect was inhibited partially by the Kv7 channel blocker linopirdine and was dependent on an increased functional contribution of the β2-adrenoceptor to the isoprenaline-mediated relaxation. In mesenteric arteries from the spontaneously hypertensive rat, ciliobrevin D and colchicine both improved the isoprenaline-mediated vasorelaxation and relaxation to the Kv7.2 -7.5 activator, ML213. Immunostaining confirmed ciliobrevin D enhanced the membrane abundance of Kv7.4. As well as an increase in the function of Kv7.4, the functional changes were associated with an increase in the contribution of β2-adrenoceptor following isoprenaline treatment. Immunostaining experiments showed ciliobrevin D prevented isoprenaline-mediated internalizationof the β2-adrenoceptor. CONCLUSIONS Overall, these data show that colchicine and ciliobrevin D can induce a β2-adrenoceptor-mediated vasodilatation in arteries from the spontaneously hypertensive rat as well as reinstating Kv7.4 channel function.
Collapse
Affiliation(s)
- Jennifer van der Horst
- Department of Biomedical Sciences (J.v.d.H., S.R., C.A., T.A.J.), University of Copenhagen, Denmark.,The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (J.v.d.H., Y.H.), University of Copenhagen, Denmark
| | - Salomé Rognant
- Department of Biomedical Sciences (J.v.d.H., S.R., C.A., T.A.J.), University of Copenhagen, Denmark
| | - Ylva Hellsten
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (J.v.d.H., Y.H.), University of Copenhagen, Denmark
| | - Christian Aalkjær
- Department of Biomedical Sciences (J.v.d.H., S.R., C.A., T.A.J.), University of Copenhagen, Denmark.,Department of Biomedicine, Aarhus University, Denmark (C.A.)
| | - Thomas A Jepps
- Department of Biomedical Sciences (J.v.d.H., S.R., C.A., T.A.J.), University of Copenhagen, Denmark
| |
Collapse
|
4
|
Cui L, Li H, Xi Y, Hu Q, Liu H, Fan J, Xiang Y, Zhang X, Shui W, Lai Y. Vesicle trafficking and vesicle fusion: mechanisms, biological functions, and their implications for potential disease therapy. MOLECULAR BIOMEDICINE 2022; 3:29. [PMID: 36129576 PMCID: PMC9492833 DOI: 10.1186/s43556-022-00090-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
Intracellular vesicle trafficking is the fundamental process to maintain the homeostasis of membrane-enclosed organelles in eukaryotic cells. These organelles transport cargo from the donor membrane to the target membrane through the cargo containing vesicles. Vesicle trafficking pathway includes vesicle formation from the donor membrane, vesicle transport, and vesicle fusion with the target membrane. Coat protein mediated vesicle formation is a delicate membrane budding process for cargo molecules selection and package into vesicle carriers. Vesicle transport is a dynamic and specific process for the cargo containing vesicles translocation from the donor membrane to the target membrane. This process requires a group of conserved proteins such as Rab GTPases, motor adaptors, and motor proteins to ensure vesicle transport along cytoskeletal track. Soluble N-ethyl-maleimide-sensitive factor (NSF) attachment protein receptors (SNARE)-mediated vesicle fusion is the final process for vesicle unloading the cargo molecules at the target membrane. To ensure vesicle fusion occurring at a defined position and time pattern in eukaryotic cell, multiple fusogenic proteins, such as synaptotagmin (Syt), complexin (Cpx), Munc13, Munc18 and other tethering factors, cooperate together to precisely regulate the process of vesicle fusion. Dysfunctions of the fusogenic proteins in SNARE-mediated vesicle fusion are closely related to many diseases. Recent studies have suggested that stimulated membrane fusion can be manipulated pharmacologically via disruption the interface between the SNARE complex and Ca2+ sensor protein. Here, we summarize recent insights into the molecular mechanisms of vesicle trafficking, and implications for the development of new therapeutics based on the manipulation of vesicle fusion.
Collapse
|
5
|
Mayya C, Naveena AH, Sinha P, Wunder C, Johannes L, Bhatia D. The roles of dynein and myosin VI motor proteins in endocytosis. J Cell Sci 2022; 135:274777. [DOI: 10.1242/jcs.259387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ABSTRACT
Endocytosis is indispensable for multiple cellular processes, including signalling, cell adhesion, migration, as well as the turnover of plasma membrane lipids and proteins. The dynamic interplay and regulation of different endocytic entry routes requires multiple cytoskeletal elements, especially motor proteins that bind to membranes and transport vesicles along the actin and microtubule cytoskeletons. Dynein and kinesin motor proteins transport vesicles along microtubules, whereas myosins drive vesicles along actin filaments. Here, we present a brief overview of multiple endocytic pathways and our current understanding of the involvement of these motor proteins in the regulation of the different cellular entry routes. We particularly focus on structural and mechanistic details of the retrograde motor proteins dynein and myosin VI (also known as MYO6), along with their adaptors, which have important roles in the early events of endocytosis. We conclude by highlighting the key challenges in elucidating the involvement of motor proteins in endocytosis and intracellular membrane trafficking.
Collapse
Affiliation(s)
- Chaithra Mayya
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - A. Hema Naveena
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Pankhuri Sinha
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| | - Christian Wunder
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, 26 rue d'Ulm, 75248 Paris CEDEX 05, France
| | - Dhiraj Bhatia
- Biological Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
- Center for Biomedical Engineering, Indian Institute of Technology Gandhinagar, Palaj, 382355 Gandhinagar, India
| |
Collapse
|
6
|
Pan H, Chai W, Liu X, Yu T, Sun L, Yan M. DYNC1H1 regulates NSCLC cell growth and metastasis by IFN-γ-JAK-STAT signaling and is associated with an aberrant immune response. Exp Cell Res 2021; 409:112897. [PMID: 34717919 DOI: 10.1016/j.yexcr.2021.112897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 02/07/2023]
Abstract
It is urgent to identify new biomarkers and therapeutic targets to ameliorate the clinical prognosis of patients with lung cancer. The functional significance and molecular mechanism of dynein cytoplasmic 1 heavy chain 1 (DYNC1H1) in nonsmall cell lung cancer (NSCLC) progression is still elusive. In our current study, publicly available data and Western blotting experiments confirmed that DYNC1H1 expression was upregulated in lung cancer samples compared with noncancerous samples. Quantitative real-time PCR (qPCR) results indicated that high DYNC1H1 expression in lung cancer tissues was significantly associated with clinical tumor stage and distal metastasis; moreover, its high expression was negatively correlated with prognosis. Functional experiments demonstrated that DYNC1H1 loss of function caused a significant decrease in cell viability and cell proliferative ability, inhibition of the cell cycle, and promotion of both migration potential and invasion potential in vitro. Animal experiments by tail vein injection of lung cancer cells showed that DYNC1H1 knockdown significantly decreased lung cancer metastasis. Mechanistically, the results from a human protein array showed changes in the IFN-γ-JAK-STAT signaling pathway, and analysis of The Cancer Genome Atlas (TCGA) immune data demonstrated that disturbance of the immune microenvironment might be involved in the impaired growth and metastatic ability mediated by DYNC1H1 loss in NSCLC. DYNC1H1 might serve as a promising biological marker of prognosis and a potential clinical therapeutic target for patients with NSCLC.
Collapse
Affiliation(s)
- Hongyu Pan
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenjun Chai
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoli Liu
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tao Yu
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei Sun
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Mingxia Yan
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Yuan J, Zhao Y, Bai Y, Gu J, Yuan Y, Liu X, Liu Z, Zou H, Bian J. Cadmium induces endosomal/lysosomal enlargement and blocks autophagy flux in rat hepatocytes by damaging microtubules. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112993. [PMID: 34808507 DOI: 10.1016/j.ecoenv.2021.112993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/31/2021] [Accepted: 11/11/2021] [Indexed: 06/13/2023]
Abstract
Acute exposure to cadmium (Cd) causes vacuolar degeneration in buffalo rat liver 3 A (BRL 3 A) cells. The present study aimed to determine the relationship between Cd-induced microtubule damage and intracellular vacuolar degeneration. Western blotting results showed that Cd damaged the microtubule network and downregulated the expression of microtubule-associated proteins-kinesin-1 heavy chain (KIF5B), γ-tubulin, and acetylated α-tubulin in BRL 3 A cells. Immunofluorescence staining revealed that Cd inhibited interactions between α-tubulin and microtubule-associated protein 4 (MAP4) as well as KIF5B. Increasing Cd concentrations decreased the levels of the lipid kinase, PIKfyve, which regulates the activity of endosome-lysosome fission. Immunofluorescence and transmission electron microscopy revealed vacuole-like organelles that were late endosomes and lysosomes. The PIKfyve inhibitor, YM201636, and the microtubule depolymerizer, nocodazole, aggravated Cd-induced endosome-lysosome enlargement. Knocking down the kif5b gene that encodes KIF5B intensified the enlargement of endosome-lysosomes and expression of early endosome antigen 1 (EEA1), Ras-related protein Rab-7a (RAB7), and lysosome-associated membrane glycoprotein 2 (LAMP2). Nocodazole, YM201636, and the knockdown of kif5b blocked autophagic flux. We concluded that Cd-induced damage to the microtubule network is the main reason for endosome-lysosome enlargement and autophagic flux blockage in BRL 3 A cells, and kinesin-1 plays a critical role in this process.
Collapse
Affiliation(s)
- Junzhao Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Yumeng Zhao
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yuni Bai
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, 12 Wenhui East Road, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
8
|
Microfilaments and microtubules alternately coordinate the multi-step endosomal trafficking of Classical Swine Fever Virus. J Virol 2021; 95:JVI.02436-20. [PMID: 33627389 PMCID: PMC8139654 DOI: 10.1128/jvi.02436-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cytoskeleton, as a ubiquitous structure in the cells, plays an important role in the process of virus entry, replication, and survival. However, the action mechanism of cytoskeleton in the invasion of Pestivirus into host cells remains unclear. In this study, we systematically dissected the key roles of the main cytoskeleton components, microfilaments and microtubules in the endocytosis of porcine Pestivirus, Classical swine fever virus (CSFV). We observed the dynamic changes of actin filaments in CSFV entry. Confocal microscopy showed that CSFV invasion induced the dissolution and aggregation of stress fibers, resulting in the formation of lamellipodia and filopodia. Chemical inhibitors and RNA interference were used to find that the dynamic changes of actin were caused by EGFR-PI3K/MAPK-RhoA/Rac1/Cdc42-cofilin signaling pathway, which regulates the microfilaments to help CSFV entry. Furthermore, co-localization of the microfilaments with clathrin and Rab5 (early endosome), as well as microtubules with Rab7 (late endosome) and Lamp1 (lysosome) revealed that microfilaments were activated and rearranged to help CSFV trafficking to early endosome after endocytosis. Subsequently, recruitment of microtubules by CSFV also assisted membrane fusion of the virions from late endosome to lysosome with the help of a molecular motor, dynein. Unexpectedly, vimentin, which is an intermediate filament, had no effect on CSFV entry. Taken together, our findings comprehensively revealed the molecular mechanisms of cytoskeletal components that regulated CSFV endocytosis and facilitated further understanding of Pestivirus entry, which would be conducive to explore antiviral molecules to control classical swine fever.IMPORTANCEEndocytosis, an essential biological process mediating cellular internalization events, is often exploited by pathogens for their entry into target cells. Previously, we have reported different mechanisms of CSFV endocytosis into the porcine epithelial cells (PK-15) and macrophages (3D4/21); however, the details of microfilaments/microtubules mediated virus migration within the host cells remained to be elucidated. In this study, we found that CSFV infection induced rearrangement of actin filaments regulated by cofilin through EGFR-PI3K/MAPK-RhoA/Rac1/Cdc42 pathway. Furthermore, we found that CSFV particles were trafficked along actin filaments in early and late endosomes, and through microtubules in lysosomes after entry. Here, we provide for the first time a comprehensive description of the cytoskeleton that facilitates entry and intracellular transport of highly pathogenic swine virus. Results from this study will greatly contribute to the understanding of virus-induced early and complex changes in host cells that are important in CSFV pathogenesis.
Collapse
|
9
|
Wang L, Yan M, Wong CKC, Ge R, Wu X, Sun F, Cheng CY. Microtubule-associated proteins (MAPs) in microtubule cytoskeletal dynamics and spermatogenesis. Histol Histopathol 2020; 36:249-265. [PMID: 33174615 DOI: 10.14670/hh-18-279] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The microtubule (MT) cytoskeleton in Sertoli cells, a crucial cellular structure in the seminiferous epithelium of adult mammalian testes that supports spermatogenesis, was studied morphologically decades ago. However, its biology, in particular the involving regulatory biomolecules and the underlying mechanism(s) in modulating MT dynamics, are only beginning to be revealed in recent years. This lack of studies in delineating the biology of MT cytoskeletal dynamics undermines other studies in the field, in particular the plausible therapeutic treatment and management of male infertility and fertility since studies have shown that the MT cytoskeleton is one of the prime targets of toxicants. Interestingly, much of the information regarding the function of actin-, MT- and intermediate filament-based cytoskeletons come from studies using toxicant models including some genetic models. During the past several years, there have been some advances in studying the biology of MT cytoskeleton in the testis, and many of these studies were based on the use of pharmaceutical/toxicant models. In this review, we summarize the results of these findings, illustrating the importance of toxicant/pharmaceutical models in unravelling the biology of MT dynamics, in particular the role of microtubule-associated proteins (MAPs), a family of regulatory proteins that modulate MT dynamics but also actin- and intermediate filament-based cytoskeletons. We also provide a timely hypothetical model which can serve as a guide to design functional experiments to study how the MT cytoskeleton is regulated during spermatogenesis through the use of toxicants and/or pharmaceutical agents.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ming Yan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chris K C Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Renshan Ge
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA.,The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
10
|
Spinner MA, Pinter K, Drerup CM, Herman TG. A Conserved Role for Vezatin Proteins in Cargo-Specific Regulation of Retrograde Axonal Transport. Genetics 2020; 216:431-445. [PMID: 32788307 PMCID: PMC7536845 DOI: 10.1534/genetics.120.303499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Active transport of organelles within axons is critical for neuronal health. Retrograde axonal transport, in particular, relays neurotrophic signals received by axon terminals to the nucleus and circulates new material among enpassant synapses. A single motor protein complex, cytoplasmic dynein, is responsible for nearly all retrograde transport within axons: its linkage to and transport of diverse cargos is achieved by cargo-specific regulators. Here, we identify Vezatin as a conserved regulator of retrograde axonal transport. Vertebrate Vezatin (Vezt) is required for the maturation and maintenance of cell-cell junctions and has not previously been implicated in axonal transport. However, a related fungal protein, VezA, has been shown to regulate retrograde transport of endosomes in hyphae. In a forward genetic screen, we identified a loss-of-function mutation in the Drosophila vezatin-like (vezl) gene. We here show that vezl loss prevents a subset of endosomes, including signaling endosomes containing activated BMP receptors, from initiating transport out of motor neuron terminal boutons. vezl loss also decreases the transport of endosomes and dense core vesicles, but not mitochondria, within axon shafts. We disrupted vezt in zebrafish and found that vezt loss specifically impairs the retrograde axonal transport of late endosomes, causing their accumulation in axon terminals. Our work establishes a conserved, cargo-specific role for Vezatin proteins in retrograde axonal transport.
Collapse
Affiliation(s)
- Michael A Spinner
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403
| | - Katherine Pinter
- Unit on Neuronal Cell Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Catherine M Drerup
- Unit on Neuronal Cell Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Tory G Herman
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403
| |
Collapse
|
11
|
Lu Y, West RJH, Pons M, Sweeney ST, Gao FB. Ik2/TBK1 and Hook/Dynein, an adaptor complex for early endosome transport, are genetic modifiers of FTD-associated mutant CHMP2B toxicity in Drosophila. Sci Rep 2020; 10:14221. [PMID: 32848189 PMCID: PMC7450086 DOI: 10.1038/s41598-020-71097-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in CHMP2B, encoding a protein in the endosomal sorting complexes required for transport (ESCRT) machinery, causes frontotemporal dementia linked to chromosome 3 (FTD3). FTD, the second most common form of pre-senile dementia, can also be caused by genetic mutations in other genes, including TANK-binding kinase 1 (TBK1). How FTD-causing disease genes interact is largely unknown. We found that partial loss function of Ik2, the fly homologue of TBK1 also known as I-kappaB kinase ε (IKKε), enhanced the toxicity of mutant CHMP2B in the fly eye and that Ik2 overexpression suppressed the effect of mutant CHMP2B in neurons. Partial loss of function of Spn-F, a downstream phosphorylation target of Ik2, greatly enhanced the mutant CHMP2B phenotype. An interactome analysis to understand cellular processes regulated by Spn-F identified a network of interacting proteins including Spn-F, Ik2, dynein light chain, and Hook, an adaptor protein in early endosome transport. Partial loss of function of dynein light chain or Hook also enhanced mutant CHMP2B toxicity. These findings identify several evolutionarily conserved genes, including ik2/TBK1, cut up (encoding dynein light chain) and hook, as genetic modifiers of FTD3-associated mutant CHMP2B toxicity and implicate early endosome transport as a potential contributing pathway in FTD.
Collapse
Affiliation(s)
- Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ryan J H West
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Marine Pons
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Sean T Sweeney
- Department of Biology, University of York, York, YO10 5DD, UK
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
12
|
Mattera R, Williamson CD, Ren X, Bonifacino JS. The FTS-Hook-FHIP (FHF) complex interacts with AP-4 to mediate perinuclear distribution of AP-4 and its cargo ATG9A. Mol Biol Cell 2020; 31:963-979. [PMID: 32073997 PMCID: PMC7185972 DOI: 10.1091/mbc.e19-11-0658] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/05/2020] [Accepted: 02/12/2020] [Indexed: 01/08/2023] Open
Abstract
The heterotetrameric adaptor protein complex 4 (AP-4) is a component of a protein coat associated with the trans-Golgi network (TGN). Mutations in AP-4 subunits cause a complicated form of autosomal-recessive hereditary spastic paraplegia termed AP-4-deficiency syndrome. Recent studies showed that AP-4 mediates export of the transmembrane autophagy protein ATG9A from the TGN to preautophagosomal structures. To identify additional proteins that cooperate with AP-4 in ATG9A trafficking, we performed affinity purification-mass spectrometry followed by validation of the hits by biochemical and functional analyses. This approach resulted in the identification of the fused toes homolog-Hook-FHIP (FHF) complex as a novel AP-4 accessory factor. We found that the AP-4-FHF interaction is mediated by direct binding of the AP-4 μ4 subunit to coiled-coil domains in the Hook1 and Hook2 subunits of FHF. Knockdown of FHF subunits resulted in dispersal of AP-4 and ATG9A from the perinuclear region of the cell, consistent with the previously demonstrated role of the FHF complex in coupling organelles to the microtubule (MT) retrograde motor dynein-dynactin. These findings thus uncover an additional mechanism for the distribution of ATG9A within cells and provide further evidence for a role of protein coats in coupling transport vesicles to MT motors.
Collapse
Affiliation(s)
- Rafael Mattera
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Chad D. Williamson
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Xuefeng Ren
- Department of Molecular and Cell Biology and California Institute of Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720
| | - Juan S. Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
13
|
Qiu R, Zhang J, Xiang X. The splicing-factor Prp40 affects dynein-dynactin function in Aspergillus nidulans. Mol Biol Cell 2020; 31:1289-1301. [PMID: 32267207 PMCID: PMC7353152 DOI: 10.1091/mbc.e20-03-0166] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The multi-component cytoplasmic dynein transports cellular cargoes with the help of another multi-component complex dynactin, but we do not know enough about factors that may affect the assembly and functions of these proteins. From a genetic screen for mutations affecting early-endosome distribution in Aspergillus nidulans, we identified the prp40AL438* mutation in Prp40A, a homologue of Prp40, an essential RNA-splicing factor in the budding yeast. Prp40A is not essential for splicing, although it associates with the nuclear splicing machinery. The prp40AL438* mutant is much healthier than the ∆prp40A mutant, but both mutants exhibit similar defects in dynein-mediated early-endosome transport and nuclear distribution. In the prp40AL438* mutant, the frequency but not the speed of dynein-mediated early-endosome transport is decreased, which correlates with a decrease in the microtubule plus-end accumulations of dynein and dynactin. Within the dynactin complex, the actin-related protein Arp1 forms a mini-filament. In a pull-down assay, the amount of Arp1 pulled down with its pointed-end protein Arp11 is lowered in the prp40AL438* mutant. In addition, we found from published interactome data that a mammalian Prp40 homologue PRPF40A interacts with Arp1. Thus, Prp40 homologues may regulate the assembly or function of dynein–dynactin and their mechanisms deserve to be further studied.
Collapse
Affiliation(s)
- Rongde Qiu
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Jun Zhang
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Xin Xiang
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| |
Collapse
|
14
|
Otamendi A, Perez-de-Nanclares-Arregi E, Oiartzabal-Arano E, Cortese MS, Espeso EA, Etxebeste O. Developmental regulators FlbE/D orchestrate the polarity site-to-nucleus dynamics of the fungal bZIP transcription factor FlbB. Cell Mol Life Sci 2019; 76:4369-4390. [PMID: 31065746 PMCID: PMC11105705 DOI: 10.1007/s00018-019-03121-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
Abstract
Permanently polarized cells have developed transduction mechanisms linking polarity sites with gene regulation in the nucleus. In neurons, one mechanism is based on long-distance retrograde migration of transcription factors (TFs). Aspergillus nidulans FlbB is the only known fungal TF shown to migrate retrogradely to nuclei from the polarized region of fungal cells known as hyphae. There, FlbB controls developmental transitions by triggering the production of asexual multicellular structures. FlbB dynamics in hyphae is orchestrated by regulators FlbE and FlbD. At least three FlbE domains are involved in the acropetal transport of FlbB, with a final MyoE/actin filament-dependent step from the subapex to the apex. Experiments employing a T2A viral peptide-containing chimera (FlbE::mRFP::T2A::FlbB::GFP) suggest that apical FlbB/FlbE interaction is inhibited to initiate a dynein-dependent FlbB transport to nuclei. FlbD controls the nuclear accumulation of FlbB through a cMyb domain and a C-terminal LxxLL motif. Overall, results elucidate a highly dynamic pattern of FlbB interactions, which enable timely developmental induction. Furthermore, this system establishes a reference for TF-based long-distance signaling in permanently polarized cells.
Collapse
Affiliation(s)
- Ainara Otamendi
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Elixabet Perez-de-Nanclares-Arregi
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Elixabet Oiartzabal-Arano
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Marc S Cortese
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain
| | - Eduardo A Espeso
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Oier Etxebeste
- Biochemistry II Laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of The Basque Country, Manuel de Lardizabal, 3, 20018, San Sebastian, Spain.
| |
Collapse
|
15
|
Zeng Y, Qian H, Wu Z, Marangoni D, Sieving PA, Bush RA. AAVrh-10 transduces outer retinal cells in rodents and rabbits following intravitreal administration. Gene Ther 2019; 26:386-398. [PMID: 31308478 PMCID: PMC11388630 DOI: 10.1038/s41434-019-0094-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/09/2019] [Accepted: 04/28/2019] [Indexed: 01/11/2023]
Abstract
Recombinant adeno-associated virus (rAAV) has been widely used for gene delivery in animal models and successfully applied in clinical trials for treating inherited retinal disease. Although subretinal delivery of AAVs can effectively transduce photoreceptors and/or retinal pigmental epithelium (RPE), cells most affected by inherited retinal diseases, the procedure is invasive and complicated, and only delivers the gene to a limited retinal area. AAVs can also be delivered intravitreally to the retina, a much less invasive nonsurgical procedure. However, intravitreal administration of non-modified AAV serotypes tends to transduce only ganglion cells and inner nuclear layer cells. To date, most non-modified AAV serotypes that have been identified are incapable of efficiently transducing photoreceptors and/or RPE when delivered intravitreally. In this study, we investigate the retinal tropism of AAVrh10 vector administered by intravitreal injection to mouse, rat, and rabbit eyes. Our results demonstrate that AAVrh10 is capable of transducing not only inner retinal cells, but also outer retinal cells in all three species, though the transduction efficiency in rabbit was low. In addition, AAVrh10 preferentially transduced outer retinal cells in mouse models of retinal disease. Therefore, AAVrh10 vector could be a useful candidate to intravitreally deliver genes to photoreceptor and RPE cells.
Collapse
Affiliation(s)
- Yong Zeng
- Section on Translational Research for Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Haohua Qian
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijian Wu
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dario Marangoni
- Section on Translational Research for Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Paul A Sieving
- Section on Translational Research for Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ronald A Bush
- Section on Translational Research for Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
16
|
Logan CM, Menko AS. Microtubules: Evolving roles and critical cellular interactions. Exp Biol Med (Maywood) 2019; 244:1240-1254. [PMID: 31387376 DOI: 10.1177/1535370219867296] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microtubules are cytoskeletal elements known as drivers of directed cell migration, vesicle and organelle trafficking, and mitosis. In this review, we discuss new research in the lens that has shed light into further roles for stable microtubules in the process of development and morphogenesis. In the lens, as well as other systems, distinct roles for characteristically dynamic microtubules and stabilized populations are coming to light. Understanding the mechanisms of microtubule stabilization and the associated microtubule post-translational modifications is an evolving field of study. Appropriate cellular homeostasis relies on not only one cytoskeletal element, but also rather an interaction between cytoskeletal proteins as well as other cellular regulators. Microtubules are key integrators with actin and intermediate filaments, as well as cell–cell junctional proteins and other cellular regulators including myosin and RhoGTPases to maintain this balance.Impact statementThe role of microtubules in cellular functioning is constantly expanding. In this review, we examine new and exciting fields of discovery for microtubule’s involvement in morphogenesis, highlight our evolving understanding of differential roles for stabilized versus dynamic subpopulations, and further understanding of microtubules as a cellular integrator.
Collapse
Affiliation(s)
- Caitlin M Logan
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - A Sue Menko
- Pathology Anatomy and Cell Biology Department, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Aspergillus nidulans in the post-genomic era: a top-model filamentous fungus for the study of signaling and homeostasis mechanisms. Int Microbiol 2019; 23:5-22. [DOI: 10.1007/s10123-019-00064-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/30/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023]
|
18
|
Lapitz A, Arbelaiz A, Olaizola P, Aranburu A, Bujanda L, Perugorria MJ, Banales JM. Extracellular Vesicles in Hepatobiliary Malignancies. Front Immunol 2018; 9:2270. [PMID: 30369925 PMCID: PMC6194158 DOI: 10.3389/fimmu.2018.02270] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022] Open
Abstract
Primary hepatobiliary malignancies include a heterogeneous group of cancers with dismal prognosis, among which hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and hepatoblastoma (HB) stand out. These tumors mainly arise from the malignant transformation of hepatocytes, cholangiocytes (bile duct epithelial cells) or hepatoblasts (embryonic liver progenitor cells), respectively. Early diagnosis, prognosis prediction and effective therapies are still a utopia for these diseases. Extracellular vesicles (EVs) are small membrane-enclosed spheres secreted by cells and present in biological fluids. They contain multiple types of biomolecules, such as proteins, RNA, DNA, metabolites and lipids, which make them a potential source of biomarkers as well as regulators of human pathobiology. In this review, the role of EVs in the pathogenesis of hepatobiliary cancers and their potential usefulness as disease biomarkers are highlighted. Moreover, the therapeutic value of EV regulation is discussed and future directions on basic and clinical research are indicated.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Extracellular Vesicles/genetics
- Extracellular Vesicles/immunology
- Extracellular Vesicles/metabolism
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- MicroRNAs/genetics
- MicroRNAs/immunology
- Models, Genetic
- Models, Immunological
Collapse
Affiliation(s)
- Ainhoa Lapitz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Ander Arbelaiz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Paula Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Aitziber Aranburu
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Maria J. Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
19
|
Qiu R, Zhang J, Xiang X. p25 of the dynactin complex plays a dual role in cargo binding and dynactin regulation. J Biol Chem 2018; 293:15606-15619. [PMID: 30143531 DOI: 10.1074/jbc.ra118.004000] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/23/2018] [Indexed: 01/13/2023] Open
Abstract
Cytoplasmic dynein binds its cargoes via the dynactin complex and cargo adapters, and the dynactin pointed-end protein p25 is required for dynein-dynactin binding to the early endosomal dynein adapter HookA (Hook in the fungus Aspergillus nidulans). However, it is unclear whether the HookA-dynein-dynactin interaction requires p27, another pointed-end protein forming heterodimers with p25 within vertebrate dynactin. Here, live-cell imaging and biochemical pulldown experiments revealed that although p27 is a component of the dynactin complex in A. nidulans, it is dispensable for dynein-dynactin to interact with ΔC-HookA (cytosolic HookA lacking its early endosome-binding C terminus) and is not critical for dynein-mediated early endosome transport. Using mutagenesis, imaging, and biochemical approaches, we found that several p25 regions are required for the ΔC-HookA-dynein-dynactin interaction, with the N terminus and loop1 being the most critical regions. Interestingly, p25 was also important for the microtubule (MT) plus-end accumulation of dynactin. This p25 function in dynactin localization also involved p25's N terminus and the loop1 critical for the ΔC-HookA-dynein-dynactin interaction. Given that dynactin's MT plus-end localization does not require HookA and that the kinesin-1-dependent plus-end accumulation of dynactin is unnecessary for the ΔC-HookA-dynein-dynactin interaction, our results indicate that p25 plays a dual role in cargo binding and dynactin regulation. As cargo adapters are implicated in dynein activation via binding to dynactin's pointed end to switch the conformation of p150, a major dynactin component, our results suggest p25 as a critical pointed-end protein involved in this process.
Collapse
Affiliation(s)
- Rongde Qiu
- From the Department of Biochemistry and Molecular Biology, the Uniformed Services University-F. Edward Hébert School of Medicine, Bethesda, Maryland 20814
| | - Jun Zhang
- From the Department of Biochemistry and Molecular Biology, the Uniformed Services University-F. Edward Hébert School of Medicine, Bethesda, Maryland 20814
| | - Xin Xiang
- From the Department of Biochemistry and Molecular Biology, the Uniformed Services University-F. Edward Hébert School of Medicine, Bethesda, Maryland 20814
| |
Collapse
|
20
|
The actin capping protein in Aspergillus nidulans enhances dynein function without significantly affecting Arp1 filament assembly. Sci Rep 2018; 8:11419. [PMID: 30061726 PMCID: PMC6065395 DOI: 10.1038/s41598-018-29818-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/18/2018] [Indexed: 11/08/2022] Open
Abstract
The minus-end-directed microtubule motor cytoplasmic dynein requires the dynactin complex for in vivo functions. The backbone of the vertebrate dynactin complex is the Arp1 (actin-related protein 1) mini-filament whose barbed end binds to the heterodimeric actin capping protein. However, it is unclear whether the capping protein is a dynactin component in lower eukaryotic organisms, especially because it does not appear to be a component of the budding yeast dynactin complex. Here our biochemical data show that the capping protein is a component of the dynactin complex in the filamentous fungus Aspergillus nidulans. Moreover, deletion of the gene encoding capping protein alpha (capA) results in a defect in both nuclear distribution and early-endosome transport, two dynein-mediated processes. However, the defect in either process is less severe than that exhibited by a dynein heavy chain mutant or the ∆p25 mutant of dynactin. In addition, loss of capping protein does not significantly affect the assembly of the dynactin Arp1 filament or the formation of the dynein-dynactin-∆C-HookA (Hook in A. nidulans) complex. These results suggest that fungal capping protein is not important for Arp1 filament assembly but its presence is required for enhancing dynein function in vivo.
Collapse
|
21
|
Morlon-Guyot J, Berry L, Sauquet I, Singh Pall G, El Hajj H, Meissner M, Daher W. Conditional knock-down of a novel coccidian protein leads to the formation of aberrant apical organelles and abrogates mature rhoptry positioning in Toxoplasma gondii. Mol Biochem Parasitol 2018; 223:19-30. [DOI: 10.1016/j.molbiopara.2018.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/23/2018] [Accepted: 06/23/2018] [Indexed: 01/21/2023]
|
22
|
Riquelme M, Aguirre J, Bartnicki-García S, Braus GH, Feldbrügge M, Fleig U, Hansberg W, Herrera-Estrella A, Kämper J, Kück U, Mouriño-Pérez RR, Takeshita N, Fischer R. Fungal Morphogenesis, from the Polarized Growth of Hyphae to Complex Reproduction and Infection Structures. Microbiol Mol Biol Rev 2018; 82:e00068-17. [PMID: 29643171 PMCID: PMC5968459 DOI: 10.1128/mmbr.00068-17] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Filamentous fungi constitute a large group of eukaryotic microorganisms that grow by forming simple tube-like hyphae that are capable of differentiating into more-complex morphological structures and distinct cell types. Hyphae form filamentous networks by extending at their tips while branching in subapical regions. Rapid tip elongation requires massive membrane insertion and extension of the rigid chitin-containing cell wall. This process is sustained by a continuous flow of secretory vesicles that depends on the coordinated action of the microtubule and actin cytoskeletons and the corresponding motors and associated proteins. Vesicles transport cell wall-synthesizing enzymes and accumulate in a special structure, the Spitzenkörper, before traveling further and fusing with the tip membrane. The place of vesicle fusion and growth direction are enabled and defined by the position of the Spitzenkörper, the so-called cell end markers, and other proteins involved in the exocytic process. Also important for tip extension is membrane recycling by endocytosis via early endosomes, which function as multipurpose transport vehicles for mRNA, septins, ribosomes, and peroxisomes. Cell integrity, hyphal branching, and morphogenesis are all processes that are largely dependent on vesicle and cytoskeleton dynamics. When hyphae differentiate structures for asexual or sexual reproduction or to mediate interspecies interactions, the hyphal basic cellular machinery may be reprogrammed through the synthesis of new proteins and/or the modification of protein activity. Although some transcriptional networks involved in such reprogramming of hyphae are well studied in several model filamentous fungi, clear connections between these networks and known determinants of hyphal morphogenesis are yet to be established.
Collapse
Affiliation(s)
- Meritxell Riquelme
- Department of Microbiology, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada, Baja California, Mexico
| | - Jesús Aguirre
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Salomon Bartnicki-García
- Department of Microbiology, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada, Baja California, Mexico
| | - Gerhard H Braus
- Department of Molecular Microbiology and Genetics and Goettingen Center for Molecular Biosciences (GZMB), University of Goettingen, Goettingen, Germany
| | - Michael Feldbrügge
- Institute for Microbiology, Heinrich Heine University Düsseldorf, Cluster of Excellence on Plant Sciences, Düsseldorf, Germany
| | - Ursula Fleig
- Institute for Functional Genomics of Microorganisms, Heinrich Heine University Düsseldorf, Cluster of Excellence on Plant Sciences, Düsseldorf, Germany
| | - Wilhelm Hansberg
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Alfredo Herrera-Estrella
- Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Mexico
| | - Jörg Kämper
- Karlsruhe Institute of Technology-South Campus, Institute for Applied Biosciences, Karlsruhe, Germany
| | - Ulrich Kück
- Ruhr University Bochum, Lehrstuhl für Allgemeine und Molekulare Botanik, Bochum, Germany
| | - Rosa R Mouriño-Pérez
- Department of Microbiology, Centro de Investigación Científica y de Educación Superior de Ensenada, Ensenada, Baja California, Mexico
| | - Norio Takeshita
- University of Tsukuba, Faculty of Life and Environmental Sciences, Tsukuba, Japan
| | - Reinhard Fischer
- Karlsruhe Institute of Technology-South Campus, Institute for Applied Biosciences, Karlsruhe, Germany
| |
Collapse
|
23
|
Hernández-González M, Bravo-Plaza I, Pinar M, de los Ríos V, Arst HN, Peñalva MA. Endocytic recycling via the TGN underlies the polarized hyphal mode of life. PLoS Genet 2018; 14:e1007291. [PMID: 29608571 PMCID: PMC5880334 DOI: 10.1371/journal.pgen.1007291] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/06/2018] [Indexed: 12/13/2022] Open
Abstract
Intracellular traffic in Aspergillus nidulans hyphae must cope with the challenges that the high rates of apical extension (1μm/min) and the long intracellular distances (>100 μm) impose. Understanding the ways in which the hyphal tip cell coordinates traffic to meet these challenges is of basic importance, but is also of considerable applied interest, as fungal invasiveness of animals and plants depends critically upon maintaining these high rates of growth. Rapid apical extension requires localization of cell-wall-modifying enzymes to hyphal tips. By combining genetic blocks in different trafficking steps with multidimensional epifluorescence microscopy and quantitative image analyses we demonstrate that polarization of the essential chitin-synthase ChsB occurs by indirect endocytic recycling, involving delivery/exocytosis to apices followed by internalization by the sub-apical endocytic collar of actin patches and subsequent trafficking to TGN cisternae, where it accumulates for ~1 min before being re-delivered to the apex by a RAB11/TRAPPII-dependent pathway. Accordingly, ChsB is stranded at the TGN by Sec7 inactivation but re-polarizes to the apical dome if the block is bypassed by a mutation in geaAgea1 that restores growth in the absence of Sec7. That polarization is independent of RAB5, that ChsB predominates at apex-proximal cisternae, and that upon dynein impairment ChsB is stalled at the tips in an aggregated endosome indicate that endocytosed ChsB traffics to the TGN via sorting endosomes functionally located upstream of the RAB5 domain and that this step requires dynein-mediated basipetal transport. It also requires RAB6 and its effector GARP (Vps51/Vps52/Vps53/Vps54), whose composition we determined by MS/MS following affinity chromatography purification. Ablation of any GARP component diverts ChsB to vacuoles and impairs growth and morphology markedly, emphasizing the important physiological role played by this pathway that, we propose, is central to the hyphal mode of growth. Filamentous fungi form long tubular cells, called hyphae, which grow rapidly by apical extension, enabling these sessile organisms to explore substrates and facilitating tissue invasion in the case of pathogenic species. Because the shape of the hyphae is determined by an external cell wall, hyphal growth requires that cell-wall sculpting enzymes polarize to the tips. Endocytosis is essential for hyphal growth, and it was suspected that this results from its participation in a recycling pathway that takes up cell-wall enzymes from the plasma membrane and re-delivers them to the apex. Here we track the trafficking of a chitin synthase (a cell-wall modifying enzyme) to demonstrate that it is polarized by endocytic recycling. This chitin synthase is delivered by exocytosis to the apex, but diffuses away until being captured by a subapical collar of actin patches (sites of endocytosis) from where it reaches a sorting endosome before undergoing transport to the nearest trans-Golgi cisternae and incorporating into secretory vesicles that re-deliver the enzyme to the apex. Because impairing transit across this pathway compromises apical extension markedly and results in severe morphological defects, the pathway could be manipulated to prevent fungal pathogenicity of plants and humans, an enormous burden on human welfare.
Collapse
Affiliation(s)
- Miguel Hernández-González
- Department of Cellular and Molecular Biology and Intradepartmental WhiteBiotech Unit, Centro de Investigaciones Biológicas del Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, Madrid, Spain
| | - Ignacio Bravo-Plaza
- Department of Cellular and Molecular Biology and Intradepartmental WhiteBiotech Unit, Centro de Investigaciones Biológicas del Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, Madrid, Spain
| | - Mario Pinar
- Department of Cellular and Molecular Biology and Intradepartmental WhiteBiotech Unit, Centro de Investigaciones Biológicas del Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, Madrid, Spain
| | - Vivian de los Ríos
- Proteomics Facility, Centro de Investigaciones Biológicas del Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, Madrid, Spain
| | - Herbert N. Arst
- Section of Microbiology, Imperial College London, Flowers Building, Armstrong Road, London, United Kingdom
| | - Miguel A. Peñalva
- Department of Cellular and Molecular Biology and Intradepartmental WhiteBiotech Unit, Centro de Investigaciones Biológicas del Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu, Madrid, Spain
- * E-mail:
| |
Collapse
|
24
|
Xiang X. Nuclear movement in fungi. Semin Cell Dev Biol 2017; 82:3-16. [PMID: 29241689 DOI: 10.1016/j.semcdb.2017.10.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 12/22/2022]
Abstract
Nuclear movement within a cell occurs in a variety of eukaryotic organisms including yeasts and filamentous fungi. Fungal molecular genetic studies identified the minus-end-directed microtubule motor cytoplasmic dynein as a critical protein for nuclear movement or orientation of the mitotic spindle contained in the nucleus. Studies in the budding yeast first indicated that dynein anchored at the cortex via its anchoring protein Num1 exerts pulling force on an astral microtubule to orient the anaphase spindle across the mother-daughter axis before nuclear division. Prior to anaphase, myosin V interacts with the plus end of an astral microtubule via Kar9-Bim1/EB1 and pulls the plus end along the actin cables to move the nucleus/spindle close to the bud neck. In addition, pushing or pulling forces generated from cortex-linked polymerization or depolymerization of microtubules drive nuclear movements in yeasts and possibly also in filamentous fungi. In filamentous fungi, multiple nuclei within a hyphal segment undergo dynein-dependent back-and-forth movements and their positioning is also influenced by cytoplasmic streaming toward the hyphal tip. In addition, nuclear movement occurs at various stages of fungal development and fungal infection of plant tissues. This review discusses our current understanding on the mechanisms of nuclear movement in fungal organisms, the importance of nuclear positioning and the regulatory strategies that ensure the proper positioning of nucleus/spindle.
Collapse
Affiliation(s)
- Xin Xiang
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, USA.
| |
Collapse
|
25
|
Etxebeste O, Espeso EA. Neurons show the path: tip-to-nucleus communication in filamentous fungal development and pathogenesis. FEMS Microbiol Rev 2017; 40:610-24. [PMID: 27587717 DOI: 10.1093/femsre/fuw021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2016] [Indexed: 01/11/2023] Open
Abstract
Multiple fungal species penetrate substrates and accomplish host invasion through the fast, permanent and unidirectional extension of filamentous cells known as hyphae. Polar growth of hyphae results, however, in a significant increase in the distance between the polarity site, which also receives the earliest information about ambient conditions, and nuclei, where adaptive responses are executed. Recent studies demonstrate that these long distances are overcome by signal transduction pathways which convey sensory information from the polarity site to nuclei, controlling development and pathogenesis. The present review compares the striking connections of the mechanisms for long-distance communication in hyphae with those from neurons, and discusses the importance of their study in order to understand invasion and dissemination processes of filamentous fungi, and design strategies for developmental control in the future.
Collapse
Affiliation(s)
- Oier Etxebeste
- Biochemistry II laboratory, Department of Applied Chemistry, Faculty of Chemistry, University of the Basque Country (UPV/EHU), 20018 San Sebastian, Spain
| | - Eduardo A Espeso
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
26
|
Peñalva MA, Zhang J, Xiang X, Pantazopoulou A. Transport of fungal RAB11 secretory vesicles involves myosin-5, dynein/dynactin/p25, and kinesin-1 and is independent of kinesin-3. Mol Biol Cell 2017; 28:947-961. [PMID: 28209731 PMCID: PMC5385943 DOI: 10.1091/mbc.e16-08-0566] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 01/31/2017] [Accepted: 02/07/2017] [Indexed: 12/02/2022] Open
Abstract
In Aspergillus nidulans, the distribution of exocytic carriers involves interplay between kinesin-1, myosin-5, and dynein. Engagement of the dynein complex to these carriers requires dynactin p25, but, unlike that of early endosomes, it does not require the Hook complex. Hyphal tip cells of the fungus Aspergillus nidulans are useful for studying long-range intracellular traffic. Post-Golgi secretory vesicles (SVs) containing the RAB11 orthologue RabE engage myosin-5 as well as plus end– and minus end–directed microtubule motors, providing an experimental system with which to investigate the interplay between microtubule and actin motors acting on the same cargo. By exploiting the fact that depolymerization of F-actin unleashes SVs focused at the apex by myosin-5 to microtubule-dependent motors, we establish that the minus end–directed transport of SVs requires the dynein/dynactin supercomplex. This minus end–directed transport is largely unaffected by genetic ablation of the Hook complex adapting early endosomes (EEs) to dynein but absolutely requires p25 in dynactin. Thus dynein recruitment to two different membranous cargoes, namely EEs and SVs, requires p25, highlighting the importance of the dynactin pointed-end complex to scaffold cargoes. Finally, by studying the behavior of SVs and EEs in null and rigor mutants of kinesin-3 and kinesin-1 (UncA and KinA, respectively), we demonstrate that KinA is the major kinesin mediating the anterograde transport of SVs. Therefore SVs arrive at the apex of A. nidulans by anterograde transport involving cooperation of kinesin-1 with myosin-5 and can move away from the apex powered by dynein.
Collapse
Affiliation(s)
- Miguel A Peñalva
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
| | - Jun Zhang
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799
| | - Xin Xiang
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799
| | - Areti Pantazopoulou
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid 28040, Spain
| |
Collapse
|
27
|
Wen Q, Tang EI, Xiao X, Gao Y, Chu DS, Mruk DD, Silvestrini B, Cheng CY. Transport of germ cells across the seminiferous epithelium during spermatogenesis-the involvement of both actin- and microtubule-based cytoskeletons. Tissue Barriers 2016; 4:e1265042. [PMID: 28123928 DOI: 10.1080/21688370.2016.1265042] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/16/2022] Open
Abstract
The transport of germ cells from the base of the seminiferous epithelium toward the luminal edge of the tubule lumen in the adluminal compartment during the epithelial cycle is an essential cellular event to support spermatogenesis. Thus, fully developed elongated spermatids (i.e., spermatozoa) can be released at spermiation in late stage VIII in rodents versus late stage II in humans. Earlier studies to examine the molecular mechanism(s) that support germ cell transport, most notably the transport of preleptotene spermatocytes across the blood-testis barrier (BTB), and the transport of elongating spermatids across the adluminal compartment during spermiogenesis, is focused on the adhesion protein complexes at the cell-cell interface. It is generally accepted that cell junctions at the Sertoli cell-cell interface at the BTB, including the actin-based tight junction (TJ), basal ectoplasmic specialization (basal ES, a testis-specific adherens junction) and gap junction (GJ), as well as the intermediate filament-based desmosome undergo constant remodeling to accommodate the transport of preleptotene spermatocytes across the barrier. On the other hand, similar junction dynamics (i.e., disassembly, reassembly and stabilization/maintenance) take place at the Sertoli-spermatid interface. Emerging evidence has shown that junction dynamics at the Sertoli cell-cell vs. Sertoli-germ cell interface are supported by the 2 intriguingly coordinated cytoskeletons, namely the F-actin- and microtubule (MT)-based cytoskeletons. Herein, we provide a brief summary and critically evaluate the recent findings. We also provide an updated hypothetical concept regarding germ cell transport in the testis utilizing the MT-conferred tracks and the MT-specific motor proteins. Furthermore, this cellular event is also supported by the F-actin-based cytoskeleton.
Collapse
Affiliation(s)
- Qing Wen
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| | - Elizabeth I Tang
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| | - Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, NY, USA; Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Ying Gao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| | - Darren S Chu
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| | - Dolores D Mruk
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| | | | - C Yan Cheng
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council , New York, NY, USA
| |
Collapse
|
28
|
Salogiannis J, Reck-Peterson SL. Hitchhiking: A Non-Canonical Mode of Microtubule-Based Transport. Trends Cell Biol 2016; 27:141-150. [PMID: 27665063 DOI: 10.1016/j.tcb.2016.09.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/29/2016] [Accepted: 09/02/2016] [Indexed: 01/01/2023]
Abstract
The long-range movement of organelles, vesicles, and macromolecular complexes by microtubule-based transport is crucial for cell growth and survival. The canonical view of intracellular transport is that each cargo directly recruits molecular motors via cargo-specific adaptor molecules. Recently, a new paradigm called 'hitchhiking' has emerged: some cargos can achieve motility by interacting with other cargos that have already recruited molecular motors. In this way, cargos are co-transported together and their movements are directly coupled. Cargo hitchhiking was discovered in fungi. However, the observation that organelle dynamics are coupled in mammalian cells suggests that this paradigm may be evolutionarily conserved. We review here the data for hitchhiking and discuss the biological significance of this non-canonical mode of microtubule-based transport.
Collapse
Affiliation(s)
- John Salogiannis
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Samara L Reck-Peterson
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Pallesi-Pocachard E, Bazellieres E, Viallat-Lieutaud A, Delgrossi MH, Barthelemy-Requin M, Le Bivic A, Massey-Harroche D. Hook2, a microtubule-binding protein, interacts with Par6α and controls centrosome orientation during polarized cell migration. Sci Rep 2016; 6:33259. [PMID: 27624926 PMCID: PMC5021942 DOI: 10.1038/srep33259] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Polarity protein complexes function during polarized cell migration and a subset of these proteins localizes to the reoriented centrosome during this process. Despite these observations, the mechanisms behind the recruitment of these polarity complexes such as the aPKC/PAR6α complex to the centrosome are not well understood. Here we identify Hook2 as an interactor for the aPKC/PAR6α complex that functions to localize this complex at the centrosome. We first demonstrate that Hook2 is essential for the polarized Golgi re-orientation towards the migration front. Depletion of Hook2 results in a decrease of PAR6α at the centrosome during cell migration, while overexpression of Hook2 in cells induced the formation of aggresomes with the recruitment of PAR6α, aPKC and PAR3. In addition, we demonstrate that the interaction between the C-terminal domain of Hook2 and the aPKC-binding domain of PAR6α localizes PAR6α to the centrosome during cell migration. Our data suggests that Hook2, a microtubule binding protein, plays an important role in the regulation of PAR6α recruitment to the centrosome to bridge microtubules and the aPKC/PAR complex. This data reveals how some of the polarity protein complexes are recruited to the centrosome and might regulate pericentriolar and microtubule organization and potentially impact on polarized migration.
Collapse
Affiliation(s)
- Emilie Pallesi-Pocachard
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Elsa Bazellieres
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Annelise Viallat-Lieutaud
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Marie-Hélène Delgrossi
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Magali Barthelemy-Requin
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - André Le Bivic
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| | - Dominique Massey-Harroche
- Aix-Marseille Univ, CNRS, UMR 7288, Developmental Biology Institute of Marseille (IBDM), case 907, 13288 Marseille, cedex 09, France
| |
Collapse
|
30
|
Rab5 and its effector FHF contribute to neuronal polarity through dynein-dependent retrieval of somatodendritic proteins from the axon. Proc Natl Acad Sci U S A 2016; 113:E5318-27. [PMID: 27559088 PMCID: PMC5018783 DOI: 10.1073/pnas.1601844113] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An open question in cell biology is how the general intracellular transport machinery is adapted to perform specialized functions in polarized cells such as neurons. Here we illustrate this adaptation by elucidating a role for the ubiquitous small GTPase Ras-related protein in brain 5 (Rab5) in neuronal polarity. We show that inactivation or depletion of Rab5 in rat hippocampal neurons abrogates the somatodendritic polarity of the transferrin receptor and several glutamate receptor types, resulting in their appearance in the axon. This loss of polarity is not caused primarily by increased transport from the soma to the axon but rather by decreased retrieval from the axon to the soma. Retrieval is also dependent on the Rab5 effector Fused Toes (FTS)-Hook-FTS and Hook-interacting protein (FHIP) (FHF) complex, which interacts with the minus-end-directed microtubule motor dynein and its activator dynactin to drive a population of axonal retrograde carriers containing somatodendritic proteins toward the soma. These findings emphasize the importance of both biosynthetic sorting and axonal retrieval for the polarized distribution of somatodendritic receptors at steady state.
Collapse
|
31
|
Olenick MA, Tokito M, Boczkowska M, Dominguez R, Holzbaur ELF. Hook Adaptors Induce Unidirectional Processive Motility by Enhancing the Dynein-Dynactin Interaction. J Biol Chem 2016; 291:18239-51. [PMID: 27365401 DOI: 10.1074/jbc.m116.738211] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Indexed: 11/06/2022] Open
Abstract
Cytoplasmic dynein drives the majority of minus end-directed vesicular and organelle motility in the cell. However, it remains unclear how dynein is spatially and temporally regulated given the variety of cargo that must be properly localized to maintain cellular function. Recent work has suggested that adaptor proteins provide a mechanism for cargo-specific regulation of motors. Of particular interest, studies in fungal systems have implicated Hook proteins in the regulation of microtubule motors. Here we investigate the role of mammalian Hook proteins, Hook1 and Hook3, as potential motor adaptors. We used optogenetic approaches to specifically recruit Hook proteins to organelles and observed rapid transport of peroxisomes to the perinuclear region of the cell. This rapid and efficient translocation of peroxisomes to microtubule minus ends indicates that mammalian Hook proteins activate dynein rather than kinesin motors. Biochemical studies indicate that Hook proteins interact with both dynein and dynactin, stabilizing the formation of a supramolecular complex. Complex formation requires the N-terminal domain of Hook proteins, which resembles the calponin-homology domain of end-binding (EB) proteins but cannot bind directly to microtubules. Single-molecule motility assays using total internal reflection fluorescence microscopy indicate that both Hook1 and Hook3 effectively activate cytoplasmic dynein, inducing longer run lengths and higher velocities than the previously characterized dynein activator bicaudal D2 (BICD2). Together, these results suggest that dynein adaptors can differentially regulate dynein to allow for organelle-specific tuning of the motor for precise intracellular trafficking.
Collapse
Affiliation(s)
- Mara A Olenick
- From the Department of Physiology and Pennsylvania Muscle Institute and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mariko Tokito
- From the Department of Physiology and Pennsylvania Muscle Institute and
| | | | - Roberto Dominguez
- From the Department of Physiology and Pennsylvania Muscle Institute and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Erika L F Holzbaur
- From the Department of Physiology and Pennsylvania Muscle Institute and Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
32
|
Yao X, Arst HN, Wang X, Xiang X. Discovery of a vezatin-like protein for dynein-mediated early endosome transport. Mol Biol Cell 2015; 26:3816-27. [PMID: 26378255 PMCID: PMC4626066 DOI: 10.1091/mbc.e15-08-0602] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 09/06/2015] [Indexed: 11/11/2022] Open
Abstract
In filamentous fungi, dynein moves early endosomes away from the hyphal tip. Aspergillus genetics is used to identify a vezatin-like protein, VezA, which is critical for dynein-mediated transport of early endosomes. VezA localizes to the hyphal tip in an actin-dependent manner and regulates the interaction between dynein and early endosomes. Early endosomes are transported bidirectionally by cytoplasmic dynein and kinesin-3, but how the movements are regulated in vivo remains unclear. Here our forward genetic study led to the discovery of VezA, a vezatin-like protein in Aspergillus nidulans, as a factor critical for early endosome distribution. Loss of vezA causes an abnormal accumulation of early endosomes at the hyphal tip, where microtubule plus ends are located. This abnormal accumulation depends on kinesin-3 and is due to a decrease in the frequency but not the speed of dynein-mediated early endosome movement. VezA-GFP signals are enriched at the hypha tip in an actin-dependent manner but are not obviously associated with early endosomes, thus differing from the early endosome association of the cargo adapter HookA (Hook in A. nidulans). On loss of VezA, HookA associates normally with early endosomes, but the interaction between dynein-dynactin and the early-endosome-bound HookA is significantly decreased. However, VezA is not required for linking dynein-dynactin to the cytosolic ∆C-HookA, lacking the cargo-binding C-terminus. These results identify VezA as a novel regulator required for the interaction between dynein and the Hook-bound early endosomes in vivo.
Collapse
Affiliation(s)
- Xuanli Yao
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences-F. Edward Hébert School of Medicine, Bethesda, MD 20814
| | - Herbert N Arst
- Microbiology Section, Department of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Xiangfeng Wang
- School of Plant Sciences, University of Arizona, Tucson, AZ 85721
| | - Xin Xiang
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences-F. Edward Hébert School of Medicine, Bethesda, MD 20814
| |
Collapse
|
33
|
Wang B, Li K, Jin M, Qiu R, Liu B, Oakley BR, Xiang X. The Aspergillus nidulans bimC4 mutation provides an excellent tool for identification of kinesin-14 inhibitors. Fungal Genet Biol 2015; 82:51-5. [PMID: 26117688 DOI: 10.1016/j.fgb.2015.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 11/30/2022]
Abstract
Centrosome amplification is a hallmark of many types of cancer cells, and clustering of multiple centrosomes is critical for cancer cell survival and proliferation. Human kinesin-14 HSET/KFIC1 is essential for centrosome clustering, and its inhibition leads to the specific killing of cancer cells with extra centrosomes. Since kinesin-14 motor domains are conserved evolutionarily, we conceived a strategy of obtaining kinesin-14 inhibitors using Aspergillus nidulans, based on the previous result that loss of the kinesin-14 KlpA rescues the non-viability of the bimC4 kinesin-5 mutant at 42 °C. However, it was unclear whether alteration of BimC or any other non-KlpA protein would be a major factor reversing the lethality of the bimC4 mutant. Here we performed a genome-wide screen for bimC4 suppressors and obtained fifteen suppressor strains. None of the suppressor mutations maps to bimC. The vast majority of them contain mutations in the klpA gene, most of which are missense mutations affecting the C-terminal motor domain. Our study confirms that the bimC4 mutant is suitable for a cell-based screen for chemical inhibitors of kinesin-14. Since the selection is based on enhanced growth rather than diminished growth, cytotoxic compounds can be excluded.
Collapse
Affiliation(s)
- Betsy Wang
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States; Richard Montgomery High School, Rockville, MD, United States
| | - Kristin Li
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States; River Hill High School, Clarksville, MD, United States; USU Summer Research Training Program (USRTP), United States
| | - Max Jin
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States; Wootton High School, Rockville, MD, United States
| | - Rongde Qiu
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States
| | - Bo Liu
- Department of Plant Biology, UC Davis, Davis, CA, United States
| | - Berl R Oakley
- Department of Molecular Biosciences, College of Liberal Arts and Sciences, The University of Kansas, Lawrence, KS, United States
| | - Xin Xiang
- Department of Biochemistry and Molecular Biology, The Uniformed Services University of the Health Sciences - F. Edward Hébert School of Medicine, Bethesda, MD, United States.
| |
Collapse
|