1
|
Yan G, Zhou J, Yin J, Gao D, Zhong X, Deng X, Kang H, Sun A. Membrane Ruffles: Composition, Function, Formation and Visualization. Int J Mol Sci 2024; 25:10971. [PMID: 39456754 PMCID: PMC11507850 DOI: 10.3390/ijms252010971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Membrane ruffles are cell actin-based membrane protrusions that have distinct structural characteristics. Linear ruffles with columnar spike-like and veil-like structures assemble at the leading edge of cell membranes. Circular dorsal ruffles (CDRs) have no supporting columnar structures but their veil-like structures, connecting from end to end, present an enclosed ring-shaped circular outline. Membrane ruffles are involved in multiple cell functions such as cell motility, macropinocytosis, receptor internalization, fluid viscosity sensing in a two-dimensional culture environment, and protecting cells from death in response to physiologically compressive loads. Herein, we review the state-of-the-art knowledge on membrane ruffle structure and function, the growth factor-induced membrane ruffling process, and the growth factor-independent ruffling mode triggered by calcium and other stimulating factors, together with the respective underlying mechanisms. We also summarize the inhibitors used in ruffle formation studies and their specificity. In the last part, an overview is given of the various techniques in which the membrane ruffles have been visualized up to now.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongyan Kang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| | - Anqiang Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China; (G.Y.); (J.Z.); (J.Y.); (D.G.); (X.Z.); (X.D.)
| |
Collapse
|
2
|
Bayati A, McPherson PS. Alpha-synuclein, autophagy-lysosomal pathway, and Lewy bodies: Mutations, propagation, aggregation, and the formation of inclusions. J Biol Chem 2024; 300:107742. [PMID: 39233232 PMCID: PMC11460475 DOI: 10.1016/j.jbc.2024.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
Research into the pathophysiology of Parkinson's disease (PD) is a fast-paced pursuit, with new findings about PD and other synucleinopathies being made each year. The involvement of various lysosomal proteins, such as TFEB, TMEM175, GBA, and LAMP1/2, marks the rising awareness about the importance of lysosomes in PD and other neurodegenerative disorders. This, along with recent developments regarding the involvement of microglia and the immune system in neurodegenerative diseases, has brought about a new era in neurodegeneration: the role of proinflammatory cytokines on the nervous system, and their downstream effects on mitochondria, lysosomal degradation, and autophagy. More effort is needed to understand the interplay between neuroimmunology and disease mechanisms, as many of the mechanisms remain enigmatic. α-synuclein, a key protein in PD and the main component of Lewy bodies, sits at the nexus between lysosomal degradation, autophagy, cellular stress, neuroimmunology, PD pathophysiology, and disease progression. This review revisits some fundamental knowledge about PD while capturing some of the latest trends in PD research, specifically as it relates to α-synuclein.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill, University, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. Nat Commun 2024; 15:4926. [PMID: 38858371 PMCID: PMC11164928 DOI: 10.1038/s41467-024-49350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 05/30/2024] [Indexed: 06/12/2024] Open
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D Romero
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rey A Carabeo
- Department of Pathology, Microbiology, and Immunology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Romero MD, Carabeo RA. Dynamin-dependent entry of Chlamydia trachomatis is sequentially regulated by the effectors TarP and TmeA. RESEARCH SQUARE 2023:rs.3.rs-3376558. [PMID: 37841835 PMCID: PMC10571596 DOI: 10.21203/rs.3.rs-3376558/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Chlamydia invasion of epithelial cells is a pathogen-driven process involving two functionally distinct effectors - TarP and TmeA. They collaborate to promote robust actin dynamics at sites of entry. Here, we extend studies on the molecular mechanism of invasion by implicating the host GTPase dynamin 2 (Dyn2) in the completion of pathogen uptake. Importantly, Dyn2 function is modulated by TarP and TmeA at the levels of recruitment and activation through oligomerization, respectively. TarP-dependent recruitment requires phosphatidylinositol 3-kinase and the small GTPase Rac1, while TmeA has a post-recruitment role related to Dyn2 oligomerization. This is based on the rescue of invasion duration and efficiency in the absence of TmeA by the Dyn2 oligomer-stabilizing small molecule activator Ryngo 1-23. Notably, Dyn2 also regulated turnover of TarP- and TmeA-associated actin networks, with disrupted Dyn2 function resulting in aberrant turnover dynamics, thus establishing the interdependent functional relationship between Dyn2 and the effectors TarP and TmeA.
Collapse
Affiliation(s)
- Matthew D. Romero
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Rey A. Carabeo
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
5
|
Liu LZ, Wang B, Zhang R, Wu Z, Huang Y, Zhang X, Zhou J, Yi J, Shen J, Li MY, Dong M. The activated CD36-Src axis promotes lung adenocarcinoma cell proliferation and actin remodeling-involved metastasis in high-fat environment. Cell Death Dis 2023; 14:548. [PMID: 37612265 PMCID: PMC10447533 DOI: 10.1038/s41419-023-06078-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
Obesity/overweight and lipid metabolism disorders have become increased risk factors for lung cancer. Fatty acid translocase CD36 promotes cellular uptake of fatty acids. Whether and how CD36 facilitates lung adenocarcinoma (LUAD) growth in high-fat environment is unknown. Here, we demonstrated that palmitic acid (PA) or high-fat diet (HFD) promoted LUAD cell proliferation and metastasis in a CD36-dependent manner. Mechanistically, CD36 translocated from cytoplasm to cell membrane and interacted with Src kinase upon PA stimulation in human LUAD cells. Akt and ERK, downstream of Src, were then activated to mediate LUAD cell proliferation and metastasis. Furthermore, PA treatment promoted CD36 sarcolemmal translocation, where it activated Rac1 and upregulated MMP-9 through Src-Akt/ERK pathway, resulting in redistribution of cortactin, N-WASP and Arp2/3, and finally led to occurrence of finger-like protrusions of actin on cell surface to enhance cell metastasis. Compared with normal-chew diet (NCD) mice, the HFD group exhibited higher level of blood free fatty acid (FFA) and cholesterol (TC), developed larger xenograft LUAD tumors and enhanced tumor cell metastatic potential, which were accompanied by obvious sarcolemmal actin remodeling and were blocked by simultaneous CD36 knockdown in LUAD cells. Consistently, xenografted and tail vein-injected scramble-RNA-A549 cells but not CD36-shRNA-A549 in HFD mice formed metastatic LUAD tumors on the lung. CD36 inhibitor SSO significantly inhibited LUAD cell metastasis to the lung. Collectively, CD36 initiates Src signaling to promote LUAD cell proliferation and actin remodeling-involved metastasis under high-fat environment. Our study provides the new insights that CD36 is a valid target for LUAD therapy.
Collapse
Affiliation(s)
- Li-Zhong Liu
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Bowen Wang
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Guangdong Medical Academic Exchange Center, Yuexiu District, Guangzhou, Guangdong, China
| | - Rui Zhang
- GuangZhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
| | - Zangshu Wu
- GuangZhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
| | - Yuxi Huang
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Xiaoyang Zhang
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Jiaying Zhou
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Junbo Yi
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518055, Guangdong, China
| | - Jian Shen
- GuangZhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
| | - Ming-Yue Li
- GuangZhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming Dong
- GuangZhou National Laboratory, Guangzhou International Bio Island, No. 9 XingDaoHuanBei Road, Guangzhou, 510005, Guangdong, China.
| |
Collapse
|
6
|
Wang Y, Ma L, Jia S, Liu D, Gu H, Wei X, Ma W, Luo W, Bai Y, Wang W, Yuan Z. Serum exosomal coronin 1A and dynamin 2 as neural tube defect biomarkers. J Mol Med (Berl) 2022; 100:1307-1319. [PMID: 35915349 PMCID: PMC9402777 DOI: 10.1007/s00109-022-02236-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/18/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022]
Abstract
Abstract No highly specific and sensitive biomarkers have been identified for early diagnosis of neural tube defects (NTDs). In this study, we used proteomics to identify novel proteins specific for NTDs. Our findings revealed three proteins showing differential expression during fetal development. In a rat model of NTDs, we used western blotting to quantify proteins in maternal serum exosomes on gestational days E18, E16, E14, and E12, in serum on E18 and E12, in neural tubes on E18 and E12, and in fetal neural exosomes on E18. The expression of coronin 1A and dynamin 2 was exosome-specific and associated with spina bifida aperta embryogenesis. Furthermore, coronin 1A and dynamin 2 were significantly downregulated in maternal serum exosomes (E12–E18), neural tubes, and fetal neural exosomes. Although downregulation was also observed in serum, the difference was not significant. Differentially expressed proteins were further analyzed in the serum exosomes of pregnant women during gestational weeks 12–40 using enzyme-linked immunosorbent assays. The findings revealed that coronin 1A and dynamin 2 showed potential diagnostic efficacy during gestational weeks 12–40, particularly during early gestation (12–18 weeks). Therefore, these two targets are used as candidate NTD screening and diagnostic biomarkers during early gestation. Key messages We used proteomics to identify novel proteins specific for NTDs. CORO1A and DNM2 showed exosome-specific expression and were associated with SBA. CORO1A and DNM2 were downregulated in maternal serum exosomes and FNEs. CORO1A and DNM2 showed good diagnostic efficacy for NTDs during early gestation. These two targets may have applications as NTD screening and diagnostic biomarkers.
Supplementary information The online version contains supplementary material available at 10.1007/s00109-022-02236-w.
Collapse
Affiliation(s)
- Yanfu Wang
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China.,Department of Pediatric Surgery, Neonatal Surgery, Shengjing Hospital, China Medical University, Shenyang, People's Republic of China
| | - Ling Ma
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China.,Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, People's Republic of China
| | - Shanshan Jia
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Dan Liu
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Hui Gu
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Xiaowei Wei
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Wei Ma
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Wenting Luo
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Yuzuo Bai
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Weilin Wang
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China
| | - Zhengwei Yuan
- Department of Pediatric Surgery, Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, People's Republic of China.
| |
Collapse
|
7
|
Bayati A, Banks E, Han C, Luo W, Reintsch WE, Zorca CE, Shlaifer I, Del Cid Pellitero E, Vanderperre B, McBride HM, Fon EA, Durcan TM, McPherson PS. Rapid macropinocytic transfer of α-synuclein to lysosomes. Cell Rep 2022; 40:111102. [PMID: 35858558 DOI: 10.1016/j.celrep.2022.111102] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/10/2022] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
The nervous system spread of alpha-synuclein fibrils is thought to cause Parkinson's disease (PD) and other synucleinopathies; however, the mechanisms underlying internalization and cellular spread are enigmatic. Here, we use confocal and superresolution microscopy, subcellular fractionation, and electron microscopy (EM) of immunogold-labeled α-synuclein preformed fibrils (PFFs) to demonstrate that this form of the protein undergoes rapid internalization and is targeted directly to lysosomes in as little as 2 min. Uptake of PFFs is disrupted by macropinocytic inhibitors and circumvents classical endosomal pathways. Immunogold-labeled PFFs are seen at the highly curved inward edge of membrane ruffles, in newly formed macropinosomes, in multivesicular bodies and in lysosomes. While most fibrils remain in lysosomes, a portion is transferred to neighboring naive cells along with markers of exosomes. These data indicate that PFFs use a unique internalization mechanism as a component of cell-to-cell propagation.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Emily Banks
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Chanshuai Han
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Wen Luo
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Wolfgang E Reintsch
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Cornelia E Zorca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Irina Shlaifer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Esther Del Cid Pellitero
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Benoit Vanderperre
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, QC, Canada
| | - Heidi M McBride
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Thomas M Durcan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada.
| |
Collapse
|
8
|
Tian X, Bunda P, Ishibe S. Podocyte Endocytosis in Regulating the Glomerular Filtration Barrier. Front Med (Lausanne) 2022; 9:801837. [PMID: 35223901 PMCID: PMC8866310 DOI: 10.3389/fmed.2022.801837] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/06/2022] [Indexed: 12/26/2022] Open
Abstract
Endocytosis is a mechanism that internalizes and recycles plasma membrane components and transmembrane receptors via vesicle formation, which is mediated by clathrin-dependent and clathrin-independent signaling pathways. Podocytes are specialized, terminally differentiated epithelial cells in the kidney, located on the outermost layer of the glomerulus. These cells play an important role in maintaining the integrity of the glomerular filtration barrier in conjunction with the adjacent basement membrane and endothelial cell layers within the glomerulus. An intact podocyte endocytic machinery appears to be necessary for maintaining podocyte function. De novo pathologic human genetic mutations and loss-of-function studies of critical podocyte endocytosis genes in genetically engineered mouse models suggest that this pathway contributes to the pathophysiology of development and progression of proteinuria in chronic kidney disease. Here, we review the mechanism of cellular endocytosis and its regulation in podocyte injury in the context of glomerular diseases. A thorough understanding of podocyte endocytosis may shed novel insights into its biological function in maintaining a functioning filter and offer potential targeted therapeutic strategies for proteinuric glomerular diseases.
Collapse
Affiliation(s)
| | | | - Shuta Ishibe
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
9
|
Avila H, Truong A, Tyrpak D, Lee SJ, Lei S, Li Y, Okamoto C, Hamm-Alvarez S, MacKay JA. Intracellular Dynamin Elastin-like Polypeptides Assemble into Rodlike, Spherical, and Reticular Dynasomes. Biomacromolecules 2022; 23:265-275. [PMID: 34914359 PMCID: PMC9159747 DOI: 10.1021/acs.biomac.1c01251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dynamin (DNM) is a family of large GTPases possessing a unique mechanical ability to "pinch" off vesicles entering cells. DNM2 is the most ubiquitously expressed member of the DNM family. We developed a novel tool based on elastin-like polypeptide (ELP) technology to quickly, precisely, and reversibly modulate the structure of DNM2. ELPs are temperature-sensitive biopolymers that self-assemble into microdomains above sharp transition temperatures. When linked together, DNM2 and a temperature-sensitive ELP fusion organize into a range of distinct temperature-dependent structures above a sharp transition temperature, which were not observed with wild-type DNM2 or a temperature-insensitive ELP fusion control. The structures comprised three different morphologies, which were prevalent at different temperature ranges. The size of these structures was influenced by an inhibitor of the DNM2 GTPase activity, dynasore; furthermore, they appear to entrap co-expressed cytosolic ELPs. Having demonstrated an unexpected diversity of morphologically distinct structures, DNM2-ELP fusions may have applications in the exploration of dynamin-dependent biology.
Collapse
Affiliation(s)
- Hugo Avila
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Anh Truong
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - David Tyrpak
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Shin-Jae Lee
- USC Viterbi School of Engineering, Department of Biomedical Engineering, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Siqi Lei
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Yaocun Li
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Curtis Okamoto
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - Sarah Hamm-Alvarez
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089,USC Keck School of Medicine, Department of Ophthalmology, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| | - J. Andrew MacKay
- USC School of Pharmacy, Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089,USC Viterbi School of Engineering, Department of Biomedical Engineering, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089,USC Keck School of Medicine, Department of Ophthalmology, University of Southern California School of Pharmacy, 1985 Zonal Ave., PSC 306A, Los Angeles, CA, 90089
| |
Collapse
|
10
|
Abstract
The distinct movements of macropinosome formation and maturation have corresponding biochemical activities which occur in a defined sequence of stages and transitions between those stages. Each stage in the process is regulated by variously phosphorylated derivatives of phosphatidylinositol (PtdIns) which reside in the cytoplasmic face of the membrane lipid bilayer. PtdIns derivatives phosphorylated at the 3' position of the inositol moiety, called 3' phosphoinositides (3'PIs), regulate different stages of the sequence. 3'PIs are synthesized by numerous phosphoinositide 3'-kinases (PI3K) and other lipid kinases and phosphatases, which are themselves regulated by small GTPases of the Ras superfamily. The combined actions of these enzymes localize four principal species of 3'PI to distinct domains of the plasma membrane or to discrete organelles, with distinct biochemical activities confined to those domains. Phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) and phosphatidylinositol (3,4)-bisphosphate (PtdIns(3,4)P2) regulate the early stages of macropinosome formation, which include cell surface ruffling and constrictions of circular ruffles which close into macropinosomes. Phosphatidylinositol 3-phosphate (PtdIns3P) regulates macropinosome fusion with other macropinosomes and early endocytic organelles. Phosphatidylinositol (3,5)-bisphosphate (PtdIns(3,5)P2) mediates macropinosome maturation and shrinkage, through loss of ions and water, and subsequent traffic to lysosomes. The different characteristic rates of macropinocytosis in different cell types indicate levels of regulation which may be governed by the cell's capacity to generate 3'PIs.
Collapse
Affiliation(s)
- Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Nobukazu Araki
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Miki, Kagawa, Japan
| |
Collapse
|
11
|
Concomitant and decoupled effects of cigarette smoke and SCAL1 upregulation on oncogenic phenotypes and ROS detoxification in lung adenocarcinoma cells. Sci Rep 2021; 11:18345. [PMID: 34526564 PMCID: PMC8443756 DOI: 10.1038/s41598-021-97869-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, with smoking as its primary predisposing factor. Although carcinogens in cigarettes are known to cause oncogenic DNA alterations, analyses of patient cohorts revealed heterogeneous genetic aberrations with no clear driver mutations. The contribution of noncoding RNAs (ncRNAs) in the pathogenesis of lung cancer has since been demonstrated. Their dysregulation has been linked to cancer initiation and progression. A novel long noncoding RNA (lncRNA) called smoke and cancer-associated lncRNA 1 (SCAL1) was recently found upregulated in smoke-exposed adenocarcinomic alveolar epithelial cells. The present study characterized the phenotypic consequences of SCAL1 overexpression and knockdown using A549 cells as model system, with or without prior exposure to cigarette smoke extract (CSE). Increase in SCAL1 levels either by CSE treatment or SCAL1 overexpression led to increased cell migration, extensive cytoskeletal remodeling, and resistance to apoptosis. Further, SCAL1 levels were negatively correlated with intracellular levels of reactive oxygen species (ROS). In contrast, SCAL1 knockdown showed converse results for these assays. These results confirm the oncogenic function of SCAL1 and its role as a CSE-activated lncRNA that mediates ROS detoxification in A549 cells, thereby allowing them to develop resistance to and survive smoke-induced toxicity.
Collapse
|
12
|
Trochet D, Bitoun M. A review of Dynamin 2 involvement in cancers highlights a promising therapeutic target. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:238. [PMID: 34294140 PMCID: PMC8296698 DOI: 10.1186/s13046-021-02045-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 12/23/2022]
Abstract
Dynamin 2 (DNM2) is an ubiquitously expressed large GTPase well known for its role in vesicle formation in endocytosis and intracellular membrane trafficking also acting as a regulator of cytoskeletons. During the last two decades, DNM2 involvement, through mutations or overexpression, emerged in an increasing number of cancers and often associated with poor prognosis. A wide panel of DNM2-dependent processes was described in cancer cells which explains DNM2 contribution to cancer pathomechanisms. First, DNM2 dysfunction may promote cell migration, invasion and metastasis. Second, DNM2 acts on intracellular signaling pathways fostering tumor cell proliferation and survival. Relative to these roles, DNM2 was demonstrated as a therapeutic target able to reduce cell proliferation, induce apoptosis, and reduce the invasive phenotype in a wide range of cancer cells in vitro. Moreover, proofs of concept of therapy by modulation of DNM2 expression was also achieved in vivo in several animal models. Consequently, DNM2 appears as a promising molecular target for the development of anti-invasive agents and the already provided proofs of concept in animal models represent an important step of preclinical development.
Collapse
Affiliation(s)
- Delphine Trochet
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France
| | - Marc Bitoun
- Centre de Recherche en Myologie, Sorbonne Université, Inserm, UMRS 974, Institut de Myologie, F-75013, Paris, France.
| |
Collapse
|
13
|
Zdżalik-Bielecka D, Poświata A, Kozik K, Jastrzębski K, Schink KO, Brewińska-Olchowik M, Piwocka K, Stenmark H, Miączyńska M. The GAS6-AXL signaling pathway triggers actin remodeling that drives membrane ruffling, macropinocytosis, and cancer-cell invasion. Proc Natl Acad Sci U S A 2021; 118:e2024596118. [PMID: 34244439 PMCID: PMC8285903 DOI: 10.1073/pnas.2024596118] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AXL, a member of the TAM (TYRO3, AXL, MER) receptor tyrosine kinase family, and its ligand, GAS6, are implicated in oncogenesis and metastasis of many cancer types. However, the exact cellular processes activated by GAS6-AXL remain largely unexplored. Here, we identified an interactome of AXL and revealed its associations with proteins regulating actin dynamics. Consistently, GAS6-mediated AXL activation triggered actin remodeling manifested by peripheral membrane ruffling and circular dorsal ruffles (CDRs). This further promoted macropinocytosis that mediated the internalization of GAS6-AXL complexes and sustained survival of glioblastoma cells grown under glutamine-deprived conditions. GAS6-induced CDRs contributed to focal adhesion turnover, cell spreading, and elongation. Consequently, AXL activation by GAS6 drove invasion of cancer cells in a spheroid model. All these processes required the kinase activity of AXL, but not TYRO3, and downstream activation of PI3K and RAC1. We propose that GAS6-AXL signaling induces multiple actin-driven cytoskeletal rearrangements that contribute to cancer-cell invasion.
Collapse
Affiliation(s)
- Daria Zdżalik-Bielecka
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland;
| | - Agata Poświata
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kamila Kozik
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kamil Jastrzębski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Kay Oliver Schink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | | | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379 Oslo, Norway
| | - Marta Miączyńska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland;
| |
Collapse
|
14
|
Kadrmas JL, Beckerle MC, Yoshigi M. Genetic analyses in mouse fibroblast and melanoma cells demonstrate novel roles for PDGF-AB ligand and PDGF receptor alpha. Sci Rep 2020; 10:19303. [PMID: 33168840 PMCID: PMC7653911 DOI: 10.1038/s41598-020-75774-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 10/14/2020] [Indexed: 01/21/2023] Open
Abstract
Platelet Derived Growth Factor Receptor (PDGFR) signaling is a central mitogenic pathway in development, as well as tissue repair and homeostasis. The rules governing the binding of PDGF ligand to the receptor to produce activation and downstream signaling have been well defined over the last several decades. In cultured cells after a period of serum deprivation, treatment with PDGF leads to the rapid formation of dramatic, actin-rich Circular Dorsal Ruffles (CDRs). Using CDRs as a robust visual readout of early PDGFR signaling, we have identified several contradictory elements in the widely accepted model of PDGF activity. Employing CRISPR/Cas9 gene editing to disrupt the Pdgfra gene in two different murine cell lines, we show that in addition to the widely accepted function for PDGFR-beta in CDR formation, PDGFR-alpha is also clearly capable of eliciting CDRs. Moreover, we demonstrate activity for heterodimeric PDGF-AB ligand in the vigorous activation of PDGFR-beta homodimers to produce CDRs. These findings are key to a more complete understanding of PDGF ligand-receptor interactions and their downstream signaling consequences. This knowledge will allow for more rigorous experimental design in future studies of PDGFR signaling and its contributions to development and disease.
Collapse
Affiliation(s)
- Julie L Kadrmas
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, 84112, USA. .,Department of Oncological Sciences, The University of Utah, Salt Lake City, UT, 84112, USA.
| | - Mary C Beckerle
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT, 84112, USA. .,School of Biological Sciences, The University of Utah, Salt Lake City, UT, 84112, USA.
| | - Masaaki Yoshigi
- Department of Pediatrics, The University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
15
|
Sajed R, Saeednejad Zanjani L, Rahimi M, Mansoori M, Zarnani AH, Madjd Z, Ghods R. Overexpression and translocation of dynamin 2 promotes tumor aggressiveness in breast carcinomas. EXCLI JOURNAL 2020; 19:1423-1435. [PMID: 33250680 PMCID: PMC7689243 DOI: 10.17179/excli2020-2762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022]
Abstract
Dynamin 2 is a GTPase protein that has been implicated in cancer progression through its various roles such as endocytosis, morphogenesis, epithelial-mesenchymal transition (EMT), cellular contractions, and focal adhesion maturation. The increased expression levels of this molecule have been demonstrated with the development of several cancers such as prostate, pancreas, and bladder. However, its clinical significance in breast cancer is unclear yet. In the present study, the membranous, cytoplasmic, and nuclear expression levels of dynamin 2 molecule were evaluated for the first time, using immunohistochemistry (IHC) on tissue microarray (TMA) slides in 113 invasive breast cancer tissues. Moreover, afterward, the association between the dynamin 2 expression and clinicopathological features was determined. Our finding showed that, a higher nuclear expression of dynamin 2 is significantly associated with an increase in tumor stage (P = 0.05), histological grade (P = 0.001), and age of the patients (P = 0.03). In addition, analysis of the cytoplasmic expression levels of this molecule revealed that, there was a statistically significant difference between the expression levels of dynamin 2 among the different breast cancer subtypes (P = 0.003). Moreover, a significant association was found between the increased expression of dynamin 2 membranous and vascular invasion (VI) (P = 0.02). We showed that dynamin 2 protein expression has an association with more aggressive tumor behavior and more advanced disease in the patients with breast cancer; therefore, dynamin 2 molecule could be considered as an indicator of disease progression and aggressiveness.
Collapse
Affiliation(s)
- Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Mandana Rahimi
- Hasheminejad Kidney Center, Pathology Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Mansoori
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran.,Reproductive Immunology Research Center, Avicenna Research Institute (ACECR), Tehran, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medicine Sciences (IUMS), Tehran, Iran.,Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
16
|
Lian Y, Wen D, Meng X, Wang X, Li H, Hao L, Xue H, Zhao J. Inhibition of invadopodia formation by diosgenin in tumor cells. Oncol Lett 2020; 20:283. [PMID: 33014161 PMCID: PMC7520800 DOI: 10.3892/ol.2020.12148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Diosgenin is a type of steroid extracted from the rhizome of Dioscorea plants. In traditional Chinese medicine, Dioscorea has the effect of ‘eliminating phlegm, promoting digestion, relaxing tendons, promoting blood circulation and inhibiting malaria’. Recent studies have confirmed that diosgenin exhibits a number of pharmacological effects, including antitumor activities. Through its antitumor effect, diosgenin is able to block tumor progression and increase the survival rate of patients with cancer; ultimately improving their quality of life. However, the mechanism underlying its pharmacological action remains unclear. Once tumor cells reach a metastatic phase, it can be fatal. Increased migration and invasiveness are the hallmarks of metastatic tumor cells. Invadopodia formation is key to maintaining the high migration and invasive ability of tumor cells. Invadopodia are a type of membrane structure process rich in filamentous-actin and are common in highly invasive tumor cells. In addition to actin, numerous actin regulators, including cortical actin-binding protein (Cortactin), accumulate in invadopodia. Cortactin is a microfilament actin-binding protein with special repetitive domains that are directly involved in the formation of the cortical microfilament actin cell skeleton. Cortactin is also one of the main substrates of intracellular Src-type tyrosine protein kinases and represents a highly conserved family of intracellular cortical signaling proteins. In recent years, great progress has been made in understanding the role of Cortactin and its molecular mechanism in cell motility. However, the diosgenin-Cortactin-invadopodia mechanism is still under investigation. Therefore, the present review focused on the current research on the regulation of invadopodia by diosgenin via Cortactin.
Collapse
Affiliation(s)
- Yaxin Lian
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dezhong Wen
- Department of Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoting Meng
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaozhen Wang
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongcheng Li
- GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin 130021, P.R. China
| | - Liming Hao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hui Xue
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Zhao
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
17
|
Lin SS, Hsieh TL, Liou GG, Li TN, Lin HC, Chang CW, Wu HY, Yao CK, Liu YW. Dynamin-2 Regulates Postsynaptic Cytoskeleton Organization and Neuromuscular Junction Development. Cell Rep 2020; 33:108310. [DOI: 10.1016/j.celrep.2020.108310] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 11/30/2022] Open
|
18
|
Burton KM, Cao H, Chen J, Qiang L, Krueger EW, Johnson KM, Bamlet WR, Zhang L, McNiven MA, Razidlo GL. Dynamin 2 interacts with α-actinin 4 to drive tumor cell invasion. Mol Biol Cell 2020; 31:439-451. [PMID: 31967944 PMCID: PMC7185896 DOI: 10.1091/mbc.e19-07-0395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/23/2019] [Accepted: 01/15/2020] [Indexed: 12/21/2022] Open
Abstract
The large GTPase Dynamin 2 (Dyn2) is known to increase the invasiveness of pancreatic cancer tumor cells, but the mechanisms by which Dyn2 regulates changes in the actin cytoskeleton to drive cell migration are still unclear. Here we report that a direct interaction between Dyn2 and the actin-bundling protein alpha-actinin (α-actinin) 4 is critical for tumor cell migration and remodeling of the extracellular matrix in pancreatic ductal adenocarcinoma (PDAC) cells. The direct interaction is mediated through the C-terminal tails of both Dyn2 and α-actinin 4, and these proteins interact at invasive structures at the plasma membrane. While Dyn2 binds directly to both α-actinin 1 and α-actinin 4, only the interaction with α-actinin 4 is required to promote tumor cell invasion. Specific disruption of the Dyn2-α-actinin 4 interaction blocks the ability of PDAC cells to migrate in either two dimensions or invade through extracellular matrix as a result of impaired invadopodia stability. Analysis of human PDAC tumor tissue additionally reveals that elevated α-actinin 4 or Dyn2 expression are predictive of poor survival. Overall, these data demonstrate that Dyn2 regulates cytoskeletal dynamics, in part, by interacting with the actin-binding protein α-actinin 4 during tumor cell invasion.
Collapse
Affiliation(s)
- Kevin M. Burton
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
| | - Hong Cao
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Jing Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Li Qiang
- Mayo Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Eugene W. Krueger
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | | | - William R. Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905
| | - Lizhi Zhang
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN 55905
| | - Mark A. McNiven
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Gina L. Razidlo
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
19
|
Dynamins 2 and 3 control the migration of human megakaryocytes by regulating CXCR4 surface expression and ITGB1 activity. Blood Adv 2019; 2:3540-3552. [PMID: 30538113 DOI: 10.1182/bloodadvances.2018021923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/04/2018] [Indexed: 12/14/2022] Open
Abstract
Megakaryocyte (MK) migration from the bone marrow periosteal niche toward the vascular niche is a prerequisite for proplatelet extension and release into the circulation. The mechanism for this highly coordinated process is poorly understood. Here we show that dynasore (DNSR), a small-molecule inhibitor of dynamins (DNMs), or short hairpin RNA knockdown of DNM2 and DNM3 impairs directional migration in a human MK cell line or MKs derived from cultured CD34+ cells. Because cell migration requires actin cytoskeletal rearrangements, we measured actin polymerization and the activity of cytoskeleton regulator RhoA and found them to be decreased after inhibition of DNM2 and DNM3. Because SDF-1α is important for hematopoiesis, we studied the expression of its receptor CXCR4 in DNSR-treated cells. CXCR4 expression on the cell surface was increased, at least partially because of slower endocytosis and internalization after SDF-1α treatment. Combined inhibition of DNM2 and DNM3 or forced expression of dominant-negative Dnm2-K44A or GTPase-defective DNM3 diminished β1 integrin (ITGB1) activity. DNSR-treated MKs showed an abnormally clustered staining pattern of Rab11, a marker of recycling endosomes. This suggests decreased recruitment of the recycling pathway in DNSR-treated cells. Altogether, we show that the GTPase activity of DNMs, which governs endocytosis and regulates cell receptor trafficking, exerts control on MK migration toward SDF-1α gradients, such as those originating from the vascular niche. DNMs play a critical role in MKs by triggering membrane-cytoskeleton rearrangements downstream of CXCR4 and integrins.
Collapse
|
20
|
Eschenburg S, Reubold TF. Modulation of dynamin function by small molecules. Biol Chem 2018; 399:1421-1432. [PMID: 30067507 DOI: 10.1515/hsz-2018-0257] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 07/17/2018] [Indexed: 02/05/2023]
Abstract
Dynamins are essential as membrane remodelers in various cellular processes, like receptor-mediated endocytosis, synaptic vesicle recycling and spermatogenesis. Moreover, dynamin is involved in the internalization of numerous viruses and in the motility of several cancer cell lines. As tools for dissecting the underlying mechanisms of these important biological processes and as potential future therapeutics, small molecules have been developed in the last two decades that modulate the functions of dynamin. In this review we give an overview of the compound classes that are currently in use and describe how they affect dynamin function.
Collapse
Affiliation(s)
- Susanne Eschenburg
- Medizinische Hochschule Hannover, Institut für Biophysikalische Chemie, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - Thomas F Reubold
- Medizinische Hochschule Hannover, Institut für Biophysikalische Chemie, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| |
Collapse
|
21
|
Syrovatkina V, Huang XY. Signaling mechanisms and physiological functions of G-protein Gα 13 in blood vessel formation, bone homeostasis, and cancer. Protein Sci 2018; 28:305-312. [PMID: 30345641 DOI: 10.1002/pro.3531] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/08/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
Heterotrimeric G-proteins are cellular signal transducers. They mainly relay signals from G-protein-coupled receptors (GPCRs). GPCRs function as guanine nucleotide-exchange factors to active these G-proteins. Based on the sequence and functional similarities, these G-proteins are grouped into four subfamilies: Gs , Gi , Gq , and G12/13 . The G12/13 subfamily consists of two members: G12 and G13 . G12/13 -mediated signaling pathways play pivotal roles in a variety of physiological processes, while aberrant regulation of this pathway has been identified in various human diseases. Here we summarize the signaling mechanisms and physiological functions of Gα13 in blood vessel formation and bone homeostasis. We further discuss the expanding roles of Gα13 in cancers, serving as oncogenes as well as tumor suppressors.
Collapse
Affiliation(s)
- Viktoriya Syrovatkina
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, New York, 10065
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, New York, 10065
| |
Collapse
|
22
|
Zhang K, Lyu W, Yu J, Koleske AJ. Abl2 is recruited to ventral actin waves through cytoskeletal interactions to promote lamellipodium extension. Mol Biol Cell 2018; 29:2863-2873. [PMID: 30256707 PMCID: PMC6249870 DOI: 10.1091/mbc.e18-01-0044] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 08/28/2018] [Accepted: 09/19/2018] [Indexed: 01/05/2023] Open
Abstract
Abl family nonreceptor tyrosine kinases regulate changes in cell shape and migration. Abl2 localizes to dynamic actin-rich protrusions, such as lamellipodia in fibroblasts and dendritic spines in neurons. Abl2 interactions with cortactin, an actin filament stabilizer, are crucial for the formation and stability of actin-rich structures, but Abl2:cortactin-positive structures have not been characterized with high spatiotemporal resolution in cells. Using total internal reflection fluorescence microscopy, we demonstrate that Abl2 colocalizes with cortactin at wave-like structures within lamellum and lamellipodium tips. Abl2 and cortactin within waves are focal and transient, extend to the outer edge of lamella, and serve as the base for lamellipodia protrusions. Abl2-positive foci colocalize with integrin β3 and paxillin, adhesive markers of the lamellum-lamellipodium interface. Cortactin-positive waves still form in Abl2 knockout cells, but the lamellipodium size is significantly reduced. This deficiency is restored following Abl2 reexpression. Complementation analyses revealed that the Abl2 C-terminal half, which contains domains that bind actin and microtubules, is necessary and sufficient for recruitment to the wave-like structures and to support normal lamellipodium size, while the kinase domain-containing N-terminal half does not impact lamellipodium size. Together, this work demonstrates that Abl2 is recruited with cortactin to actin waves through cytoskeletal interactions to promote lamellipodium extension.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cell Biology, Yale University, New Haven, CT 06520
| | - Wanqing Lyu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
| | - Ji Yu
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Anthony J. Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520
- Department of Neuroscience, Yale University, New Haven, CT 06520
| |
Collapse
|
23
|
Condon ND, Heddleston JM, Chew TL, Luo L, McPherson PS, Ioannou MS, Hodgson L, Stow JL, Wall AA. Macropinosome formation by tent pole ruffling in macrophages. J Cell Biol 2018; 217:3873-3885. [PMID: 30150290 PMCID: PMC6219714 DOI: 10.1083/jcb.201804137] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 12/26/2022] Open
Abstract
Condon et al. use lattice light-sheet microscopy to analyze live macrophages and
define a new model of macropinosome formation and closure through tent pole
ruffles. The ruffles, which are enhanced by LPS and regulated by Rab13, are
erected and supported by F-actin tent poles that cross over and twist to
constrict the forming macropinosomes. Pathogen-mediated activation of macrophages arms innate immune responses that
include enhanced surface ruffling and macropinocytosis for environmental
sampling and receptor internalization and signaling. Activation of macrophages
with bacterial lipopolysaccharide (LPS) generates prominent dorsal ruffles,
which are precursors for macropinosomes. Very rapid, high-resolution imaging of
live macrophages with lattice light sheet microscopy (LLSM) reveals new features
and actions of dorsal ruffles, which redefine the process of macropinosome
formation and closure. We offer a new model in which ruffles are erected and
supported by F-actin tent poles that cross over and twist to constrict the
forming macropinosomes. This process allows for formation of large
macropinosomes induced by LPS. We further describe the enrichment of active
Rab13 on tent pole ruffles and show that CRISPR deletion of Rab13 results in
aberrant tent pole ruffles and blocks the formation of large LPS-induced
macropinosomes. Based on the exquisite temporal and spatial resolution of LLSM,
we can redefine the ruffling and macropinosome processes that underpin innate
immune responses.
Collapse
Affiliation(s)
- Nicholas D Condon
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Australia
| | - John M Heddleston
- Advanced Imaging Center, Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA
| | - Teng-Leong Chew
- Advanced Imaging Center, Howard Hughes Medical Institute Janelia Research Campus, Ashburn, VA
| | - Lin Luo
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Australia
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Maria S Ioannou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY
| | - Jennifer L Stow
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Australia
| | - Adam A Wall
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation and Disease Research, The University of Queensland, Brisbane, Australia
| |
Collapse
|
24
|
Bertier L, Hebbrecht T, Mettepenningen E, De Wit N, Zwaenepoel O, Verhelle A, Gettemans J. Nanobodies targeting cortactin proline rich, helical and actin binding regions downregulate invadopodium formation and matrix degradation in SCC-61 cancer cells. Biomed Pharmacother 2018; 102:230-241. [DOI: 10.1016/j.biopha.2018.03.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/10/2018] [Accepted: 03/12/2018] [Indexed: 01/19/2023] Open
|
25
|
DePasquale JA. Apical surface ring formation in
Cyprinus carpio
scale epidermis. ACTA ZOOL-STOCKHOLM 2018. [DOI: 10.1111/azo.12256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Makaraci P, Kim K. trans-Golgi network-bound cargo traffic. Eur J Cell Biol 2018; 97:137-149. [PMID: 29398202 DOI: 10.1016/j.ejcb.2018.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Cargo following the retrograde trafficking are sorted at endosomes to be targeted the trans-Golgi network (TGN), a central receiving organelle. Though molecular requirements and their interaction networks have been somewhat established, the complete understanding of the intricate nature of their action mechanisms in every step of the retrograde traffic pathway remains unachieved. This review focuses on elucidating known functions of key regulators, including scission factors at the endosome and tethering/fusion mediators at the receiving dock, TGN, as well as a diverse range of cargo.
Collapse
Affiliation(s)
- Pelin Makaraci
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National Ave., Springfield, MO 65807, USA.
| |
Collapse
|
27
|
Abstract
Actin remodeling plays an essential role in diverse cellular processes such as cell motility, vesicle trafficking or cytokinesis. The scaffold protein and actin nucleation promoting factor Cortactin is present in virtually all actin-based structures, participating in the formation of branched actin networks. It has been involved in the control of endocytosis, and vesicle trafficking, axon guidance and organization, as well as adhesion, migration and invasion. To migrate and invade through three-dimensional environments, cells have developed specialized actin-based structures called invadosomes, a generic term to designate invadopodia and podosomes. Cortactin has emerged as a critical regulator of invadosome formation, function and disassembly. Underscoring this role, Cortactin is frequently overexpressed in several types of invasive cancers. Herein we will review the roles played by Cortactin in these specific invasive structures.
Collapse
Affiliation(s)
- Pauline Jeannot
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester , Manchester M20 4BX, UK
| | - Arnaud Besson
- CRCT INSERM UMR1037, Université Toulouse III Paul Sabatier , CNRS ERL5294, Toulouse, France.,LBCMCP , Centre de Biologie Intégrative, Université de Toulouse , CNRS, UPS, Toulouse Cedex, France
| |
Collapse
|
28
|
Schneider DJ, Speth JM, Penke LR, Wettlaufer SH, Swanson JA, Peters-Golden M. Mechanisms and modulation of microvesicle uptake in a model of alveolar cell communication. J Biol Chem 2017; 292:20897-20910. [PMID: 29101235 PMCID: PMC5743066 DOI: 10.1074/jbc.m117.792416] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/27/2017] [Indexed: 12/31/2022] Open
Abstract
Extracellular vesicles, including exosomes and shed microvesicles (MVs), can be internalized by recipient cells to modulate function. Although the mechanism by which extracellular vesicles are internalized is incompletely characterized, it is generally considered to involve endocytosis and an initial surface-binding event. Furthermore, modulation of uptake by microenvironmental factors is largely unstudied. Here, we used flow cytometry, confocal microscopy, and pharmacologic and molecular targeting to address these gaps in knowledge in a model of pulmonary alveolar cell-cell communication. Alveolar macrophage-derived MVs were fully internalized by alveolar epithelial cells in a time-, dose-, and temperature-dependent manner. Uptake was dependent on dynamin and actin polymerization. However, it was neither saturable nor dependent on clathrin or receptor binding. Internalization was enhanced by extracellular proteins but was inhibited by cigarette smoke extract via oxidative disruption of actin polymerization. We conclude that MV internalization occurs via a pathway more consistent with fluid-phase than receptor-dependent endocytosis and is subject to bidirectional modulation by relevant pathologic perturbations.
Collapse
Affiliation(s)
| | | | - Loka R Penke
- From the Division of Pulmonary and Critical Care Medicine
| | | | - Joel A Swanson
- Department of Microbiology and Immunology, and
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Marc Peters-Golden
- From the Division of Pulmonary and Critical Care Medicine,
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
29
|
G-Protein Gα 13 Functions with Abl Kinase to Regulate Actin Cytoskeletal Reorganization. J Mol Biol 2017; 429:3836-3849. [PMID: 29079481 DOI: 10.1016/j.jmb.2017.10.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 11/23/2022]
Abstract
Heterotrimeric G-proteins are essential cellular signal transducers. One of the G-proteins, Gα13, is critical for actin cytoskeletal reorganization, cell migration, cell proliferation, and apoptosis. Previously, we have shown that Gα13 is essential for both G-protein-coupled receptor and receptor tyrosine kinase-induced actin cytoskeletal reorganization such as dynamic dorsal ruffle turnover and cell migration. However, the mechanism by which Gα13 signals to actin cytoskeletal reorganization is not completely understood. Here we show that Gα13 directly interacts with Abl tyrosine kinase, which is a critical regulator of actin cytoskeleton. This interaction is critical for Gα13-induced dorsal ruffle turnover, endothelial cell remodeling, and cell migration. Our data uncover a new molecular signaling pathway by which Gα13 controls actin cytoskeletal reorganization.
Collapse
|
30
|
Abstract
During macropinocytosis, cells remodel their morphologies for the uptake of extracellular matter. This endocytotic mechanism relies on the collapse and closure of precursory structures, which are propagating actin-based, ring-shaped vertical undulations at the dorsal (top) cell membrane, a.k.a. circular dorsal ruffles (CDRs). As such, CDRs are essential to a range of vital and pathogenic processes alike. Here we show, based on both experimental data and theoretical analysis, that CDRs are propagating fronts of actin polymerization in a bistable system. The theory relies on a novel mass-conserving reaction–diffusion model, which associates the expansion and contraction of waves to distinct counter-propagating front solutions. Moreover, the model predicts that under a change in parameters (for example, biochemical conditions) CDRs may be pinned and fluctuate near the cell boundary or exhibit complex spiral wave dynamics due to a wave instability. We observe both phenomena also in our experiments indicating the conditions for which macropinocytosis is suppressed. Circular dorsal ruffles (CDRs) are important for the vesicular uptake of extracellular matter, but the basis of their wave dynamics is not understood. Here, the authors propose and experimentally test a bistable reaction-diffusion system, which they show accounts for the typical CDR expansion and shrinkage and for aberrant formation of pinned waves and spirals.
Collapse
|
31
|
Atherton P, Lausecker F, Harrison A, Ballestrem C. Low-intensity pulsed ultrasound promotes cell motility through vinculin-controlled Rac1 GTPase activity. J Cell Sci 2017; 130:2277-2291. [PMID: 28576970 DOI: 10.1242/jcs.192781] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/29/2017] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a therapy used clinically to promote healing. Using live-cell imaging we show that LIPUS stimulation, acting through integrin-mediated cell-matrix adhesions, rapidly induces Rac1 activation associated with dramatic actin cytoskeleton rearrangements. Our study demonstrates that the mechanosensitive focal adhesion (FA) protein vinculin, and both focal adhesion kinase (FAK, also known as PTK2) and Rab5 (both the Rab5a and Rab5b isoforms) have key roles in regulating these effects. Inhibiting the link of vinculin to the actin-cytoskeleton abolished LIPUS sensing. We show that this vinculin-mediated link was not only critical for Rac1 induction and actin rearrangements, but was also important for the induction of a Rab5-dependent increase in the number of early endosomes. Expression of dominant-negative Rab5, or inhibition of endocytosis with dynasore, also blocked LIPUS-induced Rac1 signalling events. Taken together, our data show that LIPUS is sensed by cell matrix adhesions through vinculin, which in turn modulates a Rab5-Rac1 pathway to control ultrasound-mediated endocytosis and cell motility. Finally, we demonstrate that a similar FAK-Rab5-Rac1 pathway acts to control cell spreading upon fibronectin.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Andrew Harrison
- Bioventus Cooperatief, Taurusavenue 31, 2132 LS Hoofddorp, The Netherlands
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| |
Collapse
|
32
|
Valdivia A, Goicoechea SM, Awadia S, Zinn A, Garcia-Mata R. Regulation of circular dorsal ruffles, macropinocytosis, and cell migration by RhoG and its exchange factor, Trio. Mol Biol Cell 2017; 28:1768-1781. [PMID: 28468978 PMCID: PMC5491185 DOI: 10.1091/mbc.e16-06-0412] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 11/11/2022] Open
Abstract
The small GTPase RhoG and its exchange factor, Trio, regulate the formation and size of circular dorsal ruffles and associated functions, including macropinocytosis and cell migration. Circular dorsal ruffles (CDRs) are actin-rich structures that form on the dorsal surface of many mammalian cells in response to growth factor stimulation. CDRs represent a unique type of structure that forms transiently and only once upon stimulation. The formation of CDRs involves a drastic rearrangement of the cytoskeleton, which is regulated by the Rho family of GTPases. So far, only Rac1 has been consistently associated with CDR formation, whereas the role of other GTPases in this process is either lacking or inconclusive. Here we show that RhoG and its exchange factor, Trio, play a role in the regulation of CDR dynamics, particularly by modulating their size. RhoG is activated by Trio downstream of PDGF in a PI3K- and Src-dependent manner. Silencing RhoG expression decreases the number of cells that form CDRs, as well as the area of the CDRs. The regulation of CDR area by RhoG is independent of Rac1 function. In addition, our results show the RhoG plays a role in the cellular functions associated with CDR formation, including macropinocytosis, receptor internalization, and cell migration. Taken together, our results reveal a novel role for RhoG in the regulation of CDRs and the cellular processes associated with their formation.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606.,Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322
| | | | - Sahezeel Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Ashtyn Zinn
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| |
Collapse
|
33
|
Busija AR, Patel HH, Insel PA. Caveolins and cavins in the trafficking, maturation, and degradation of caveolae: implications for cell physiology. Am J Physiol Cell Physiol 2017; 312:C459-C477. [PMID: 28122734 DOI: 10.1152/ajpcell.00355.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/09/2023]
Abstract
Caveolins (Cavs) are ~20 kDa scaffolding proteins that assemble as oligomeric complexes in lipid raft domains to form caveolae, flask-shaped plasma membrane (PM) invaginations. Caveolae ("little caves") require lipid-lipid, protein-lipid, and protein-protein interactions that can modulate the localization, conformational stability, ligand affinity, effector specificity, and other functions of proteins that are partners of Cavs. Cavs are assembled into small oligomers in the endoplasmic reticulum (ER), transported to the Golgi for assembly with cholesterol and other oligomers, and then exported to the PM as an intact coat complex. At the PM, cavins, ~50 kDa adapter proteins, oligomerize into an outer coat complex that remodels the membrane into caveolae. The structure of caveolae protects their contents (i.e., lipids and proteins) from degradation. Cellular changes, including signal transduction effects, can destabilize caveolae and produce cavin dissociation, restructuring of Cav oligomers, ubiquitination, internalization, and degradation. In this review, we provide a perspective of the life cycle (biogenesis, degradation), composition, and physiologic roles of Cavs and caveolae and identify unanswered questions regarding the roles of Cavs and cavins in caveolae and in regulating cell physiology.1.
Collapse
Affiliation(s)
- Anna R Busija
- Department of Anesthesiology, University of California, San Diego, La Jolla, California.,Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Hemal H Patel
- Department of Anesthesiology, University of California, San Diego, La Jolla, California
| | - Paul A Insel
- Department of Medicine, University of California, San Diego, La Jolla, California; and .,Department of Pharmacology, University of California, San Diego, La Jolla, California
| |
Collapse
|
34
|
Edwards BS, Clay CM, Ellsworth BS, Navratil AM. Functional Role of Gonadotrope Plasticity and Network Organization. Front Endocrinol (Lausanne) 2017; 8:223. [PMID: 28936197 PMCID: PMC5595155 DOI: 10.3389/fendo.2017.00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/16/2017] [Indexed: 11/18/2022] Open
Abstract
Gonadotrope cells of the anterior pituitary are characterized by their ability to mount a cyclical pattern of gonadotropin secretion to regulate gonadal function and fertility. Recent in vitro and in vivo evidence suggests that gonadotropes exhibit dramatic remodeling of the actin cytoskeleton following gonadotropin-releasing hormone (GnRH) exposure. GnRH engagement of actin is critical for gonadotrope function on multiple levels. First, GnRH-induced cell movements lead to spatial repositioning of the in vivo gonadotrope network toward vascular endothelium, presumably to access the bloodstream for effective hormone release. Interestingly, these plasticity changes can be modified depending on the physiological status of the organism. Additionally, GnRH-induced actin assembly appears to be fundamental to gonadotrope signaling at the level of extracellular signal-regulated kinase (ERK) activation, which is a well-known regulator of luteinizing hormone (LH) β-subunit synthesis. Last, GnRH-induced cell membrane projections are capable of concentrating LHβ-containing vesicles and disruption of the actin cytoskeleton reduces LH secretion. Taken together, gonadotrope network positioning and LH synthesis and secretion are linked to GnRH engagement of the actin cytoskeleton. In this review, we will cover the dynamics and organization of the in vivo gonadotrope cell network and the mechanisms of GnRH-induced actin-remodeling events important in ERK activation and subsequently hormone secretion.
Collapse
Affiliation(s)
- Brian S. Edwards
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| | - Colin M. Clay
- Department of Biomedical Science, Colorado State University, Fort Collins, CO, United States
| | - Buffy S. Ellsworth
- Department of Physiology, Southern Illinois University Carbondale, Carbondale, IL, United States
| | - Amy M. Navratil
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
- *Correspondence: Amy M. Navratil,
| |
Collapse
|
35
|
Abstract
Genetic studies of hereditary forms of nephrotic syndrome have identified several proteins that are involved in regulating the permselective properties of the glomerular filtration system. Further extensive research has elucidated the complex molecular basis of the glomerular filtration barrier and clearly established the pivotal role of podocytes in the pathophysiology of glomerular diseases. Podocyte architecture is centred on focal adhesions and slit diaphragms - multiprotein signalling hubs that regulate cell morphology and function. A highly interconnected actin cytoskeleton enables podocytes to adapt in order to accommodate environmental changes and maintain an intact glomerular filtration barrier. Actin-based endocytosis has now emerged as a regulator of podocyte integrity, providing an impetus for understanding the precise mechanisms that underlie the steady-state control of focal adhesion and slit diaphragm components. This Review outlines the role of actin dynamics and endocytosis in podocyte biology, and discusses how molecular heterogeneity in glomerular disorders could be exploited to deliver more rational therapeutic interventions, paving the way for targeted medicine in nephrology.
Collapse
|
36
|
Syrovatkina V, Alegre KO, Dey R, Huang XY. Regulation, Signaling, and Physiological Functions of G-Proteins. J Mol Biol 2016; 428:3850-68. [PMID: 27515397 DOI: 10.1016/j.jmb.2016.08.002] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/31/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022]
Abstract
Heterotrimeric guanine-nucleotide-binding regulatory proteins (G-proteins) mainly relay the information from G-protein-coupled receptors (GPCRs) on the plasma membrane to the inside of cells to regulate various biochemical functions. Depending on the targeted cell types, tissues, and organs, these signals modulate diverse physiological functions. The basic schemes of heterotrimeric G-proteins have been outlined. In this review, we briefly summarize what is known about the regulation, signaling, and physiological functions of G-proteins. We then focus on a few less explored areas such as the regulation of G-proteins by non-GPCRs and the physiological functions of G-proteins that cannot be easily explained by the known G-protein signaling pathways. There are new signaling pathways and physiological functions for G-proteins to be discovered and further interrogated. With the advancements in structural and computational biological techniques, we are closer to having a better understanding of how G-proteins are regulated and of the specificity of G-protein interactions with their regulators.
Collapse
Affiliation(s)
- Viktoriya Syrovatkina
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Kamela O Alegre
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Raja Dey
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
37
|
Burke RT, Orth JD. Through the Looking Glass: Time-lapse Microscopy and Longitudinal Tracking of Single Cells to Study Anti-cancer Therapeutics. J Vis Exp 2016. [PMID: 27213923 DOI: 10.3791/53994] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The response of single cells to anti-cancer drugs contributes significantly in determining the population response, and therefore is a major contributing factor in the overall outcome. Immunoblotting, flow cytometry and fixed cell experiments are often used to study how cells respond to anti-cancer drugs. These methods are important, but they have several shortcomings. Variability in drug responses between cancer and normal cells, and between cells of different cancer origin, and transient and rare responses are difficult to understand using population averaging assays and without being able to directly track and analyze them longitudinally. The microscope is particularly well suited to image live cells. Advancements in technology enable us to routinely image cells at a resolution that enables not only cell tracking, but also the observation of a variety of cellular responses. We describe an approach in detail that allows for the continuous time-lapse imaging of cells during the drug response for essentially as long as desired, typically up to 96 hr. Using variations of the approach, cells can be monitored for weeks. With the employment of genetically encoded fluorescent biosensors numerous processes, pathways and responses can be followed. We show examples that include tracking and quantification of cell growth and cell cycle progression, chromosome dynamics, DNA damage, and cell death. We also discuss variations of the technique and its flexibility, and highlight some common pitfalls.
Collapse
Affiliation(s)
- Russell T Burke
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder
| | - James D Orth
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder;
| |
Collapse
|
38
|
Zimnicka AM, Husain YS, Shajahan AN, Sverdlov M, Chaga O, Chen Z, Toth PT, Klomp J, Karginov AV, Tiruppathi C, Malik AB, Minshall RD. Src-dependent phosphorylation of caveolin-1 Tyr-14 promotes swelling and release of caveolae. Mol Biol Cell 2016; 27:2090-106. [PMID: 27170175 PMCID: PMC4927282 DOI: 10.1091/mbc.e15-11-0756] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/05/2016] [Indexed: 11/18/2022] Open
Abstract
Src-induced phosphorylation of Cav-1 is analyzed using live TIRF and FRET microscopy, as well as by biochemical analysis. Cav1 phosphorylation destabilizes plasma membrane–associated Cav-1 oligomers and thereby is crucial for regulating the fission of caveolae from the plasma membrane in vascular endothelial cells. Caveolin 1 (Cav1) is a required structural component of caveolae, and its phosphorylation by Src is associated with an increase in caveolae-mediated endocytosis. Here we demonstrate, using quantitative live-cell 4D, TIRF, and FRET imaging, that endocytosis and trafficking of caveolae are associated with a Cav1 Tyr-14 phosphorylation-dependent conformational change, which spatially separates, or loosens, Cav1 molecules within the oligomeric caveolar coat. When tracked by TIRF and spinning-disk microscopy, cells expressing phosphomimicking Cav1 (Y14D) mutant formed vesicles that were greater in number and volume than with Y14F-Cav1-GFP. Furthermore, we observed in HEK cells cotransfected with wild-type, Y14D, or Y14F Cav1-CFP and -YFP constructs that FRET efficiency was greater with Y14F pairs than with Y14D, indicating that pY14-Cav1 regulates the spatial organization of Cav1 molecules within the oligomer. In addition, albumin-induced Src activation or direct activation of Src using a rapamycin-inducible Src construct (RapR-Src) led to an increase in monomeric Cav1 in Western blots, as well as a simultaneous increase in vesicle number and decrease in FRET intensity, indicative of a Src-mediated conformational change in CFP/YFP-tagged WT-Cav1 pairs. We conclude that phosphorylation of Cav1 leads to separation or “spreading” of neighboring negatively charged N-terminal phosphotyrosine residues, promoting swelling of caveolae, followed by their release from the plasma membrane.
Collapse
Affiliation(s)
- Adriana M Zimnicka
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Yawer S Husain
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Ayesha N Shajahan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Maria Sverdlov
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Oleg Chaga
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Zhenlong Chen
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
| | - Peter T Toth
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Jennifer Klomp
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Andrei V Karginov
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Asrar B Malik
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612 Center for Lung and Vascular Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612
| | - Richard D Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612 Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612 Center for Lung and Vascular Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
39
|
Marie-Anaïs F, Mazzolini J, Herit F, Niedergang F. Dynamin-Actin Cross Talk Contributes to Phagosome Formation and Closure. Traffic 2016; 17:487-99. [DOI: 10.1111/tra.12386] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/02/2016] [Accepted: 02/02/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Florence Marie-Anaïs
- Inserm U1016, Institut Cochin; Paris France
- CNRS, UMR 8104; Paris France
- Université Paris Descartes; Sorbonne Paris Cité; Paris France
| | - Julie Mazzolini
- Inserm U1016, Institut Cochin; Paris France
- CNRS, UMR 8104; Paris France
- Université Paris Descartes; Sorbonne Paris Cité; Paris France
- Current address: Centre for Neuroregeneration; The University of Edinburgh; Edinburgh UK
| | - Floriane Herit
- Inserm U1016, Institut Cochin; Paris France
- CNRS, UMR 8104; Paris France
- Université Paris Descartes; Sorbonne Paris Cité; Paris France
| | - Florence Niedergang
- Inserm U1016, Institut Cochin; Paris France
- CNRS, UMR 8104; Paris France
- Université Paris Descartes; Sorbonne Paris Cité; Paris France
| |
Collapse
|
40
|
Hetzenecker S, Helenius A, Krzyzaniak MA. HCMV Induces Macropinocytosis for Host Cell Entry in Fibroblasts. Traffic 2016; 17:351-68. [PMID: 26650385 DOI: 10.1111/tra.12355] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/30/2015] [Accepted: 11/30/2015] [Indexed: 12/14/2022]
Abstract
Human cytomegalovirus (HCMV) is an important and widespread pathogen in the human population. While infection by this β-herpesvirus in endothelial, epithelial and dendritic cells depends on endocytosis, its entry into fibroblasts is thought to occur by direct fusion of the viral envelope with the plasma membrane. To characterize individual steps during entry in primary human fibroblasts, we employed quantitative assays as well as electron, fluorescence and live cell microscopy in combination with a variety of inhibitory compounds. Our results showed that while infectious entry was pH- and clathrin-independent, it required multiple, endocytosis-related factors and processes. The virions were found to undergo rapid internalization into large vacuoles containing internalized fluid and endosome markers. The characteristics of the internalization process fulfilled major criteria for macropinocytosis. Moreover, we found that soon after addition to fibroblasts the virus rapidly triggered the formation of circular dorsal ruffles in the host cell followed by the generation of large macropinocytic vacuoles. This distinctive form of macropinocytosis has been observed especially in primary cells but has not previously been reported in response to virus stimulation.
Collapse
Affiliation(s)
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland
| | - Magdalena Anna Krzyzaniak
- Institute of Biochemistry, ETH Zurich, Zurich 8093, Switzerland.,Department of Biomedicine, University of Basel, Basel 40001, Switzerland
| |
Collapse
|
41
|
Edwards BS, Dang AK, Murtazina DA, Dozier MG, Whitesell JD, Khan SA, Cherrington BD, Amberg GC, Clay CM, Navratil AM. Dynamin Is Required for GnRH Signaling to L-Type Calcium Channels and Activation of ERK. Endocrinology 2016; 157:831-43. [PMID: 26696122 PMCID: PMC4733113 DOI: 10.1210/en.2015-1575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have shown that GnRH-mediated engagement of the cytoskeleton induces cell movement and is necessary for ERK activation. It also has previously been established that a dominant negative form of the mechano-GTPase dynamin (K44A) attenuates GnRH activation of ERK. At present, it is not clear at what level these cellular events might be linked. To explore this, we used live cell imaging in the gonadotrope-derived αT3-1 cell line to determine that dynamin-green fluorescent protein accumulated in GnRH-induced lamellipodia and plasma membrane protrusions. Coincident with translocation of dynamin-green fluorescent protein to the plasma membrane, we demonstrated that dynamin colocalizes with the actin cytoskeleton and the actin binding protein, cortactin at the leading edge of the plasma membrane. We next wanted to assess the physiological significance of these findings by inhibiting dynamin GTPase activity using dynasore. We find that dynasore suppresses activation of ERK, but not c-Jun N-terminal kinase, after exposure to GnRH agonist. Furthermore, exposure of αT3-1 cells to dynasore inhibited GnRH-induced cyto-architectural rearrangements. Recently it has been discovered that GnRH induced Ca(2+) influx via the L-type Ca(2+) channels requires an intact cytoskeleton to mediate ERK phosphorylation. Interestingly, not only does dynasore attenuate GnRH-mediated actin reorganization, it also suppresses Ca(2+) influx through L-type Ca(2+) channels visualized in living cells using total internal reflection fluorescence microscopy. Collectively, our data suggest that GnRH-induced membrane remodeling events are mediated in part by the association of dynamin and cortactin engaging the actin cytoskeleton, which then regulates Ca(2+) influx via L-type channels to facilitate ERK phosphorylation.
Collapse
Affiliation(s)
- Brian S Edwards
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - An K Dang
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Dilyara A Murtazina
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Melissa G Dozier
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Jennifer D Whitesell
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Shaihla A Khan
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Brian D Cherrington
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Gregory C Amberg
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Colin M Clay
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| | - Amy M Navratil
- Department of Zoology and Physiology (B.S.E., M.G.D., S.A.K., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; Department of Biomedical Sciences (A.K.D., D.A.M., G.C.A., C.M.C.), Colorado State University, Fort Collins, Colorado 80523; and Department of Research and Development (J.D.W.), Allen Institute for Brain Science, Seattle, Washington 98103
| |
Collapse
|
42
|
Mascia A, Gentile F, Izzo A, Mollo N, De Luca M, Bucci C, Nitsch L, Calì G. Rab7 Regulates CDH1 Endocytosis, Circular Dorsal Ruffles Genesis, and Thyroglobulin Internalization in a Thyroid Cell Line. J Cell Physiol 2015; 231:1695-708. [PMID: 26599499 DOI: 10.1002/jcp.25267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/23/2015] [Indexed: 01/02/2023]
Abstract
Rab7 regulates the biogenesis of late endosomes, lysosomes, and autophagosomes. It has been proposed that a functional and physical interaction exists between Rab7 and Rac1 GTPases in CDH1 endocytosis and ruffled border formation. In FRT cells over-expressing Rab7, increased expression and activity of Rac1 was observed, whereas a reduction of Rab7 expression by RNAi resulted in reduced Rac1 activity, as measured by PAK1 phosphorylation. We found that CDH1 endocytosis was extremely reduced only in Rab7 over-expressing cells but was unchanged in Rab7 silenced cells. In Rab7 under or over-expressing cells, Rab7 and LC3B-II co-localized and co-localization in large circular structures occurred only in Rab7 over-expressing cells. These large circular structures occurred in about 10% of the cell population; some of them (61%) showed co-localization of Rab7 with cortactin and f-actin and were identified as circular dorsal ruffles (CDRs), the others as mature autophagosomes. We propose that the over-expression of Rab7 is sufficient to induce CDRs. Furthermore, in FRT cells, we found that the expression of the insoluble/active form of Rab7, rather than Rab5, or Rab8, was inducible by cAMP and that cAMP-stimulated FRT cells showed increased PAK1 phosphorylation and were no longer able to endocytose CDH1. Finally, we demonstrated that Rab7 over-expressing cells are able to endocytose exogenous thyroglobulin via pinocytosis/CDRs more efficiently than control cells. We propose that the major thyroglobulin endocytosis described in thyroid autonomous adenomas due to Rab7 increased expression, occurs via CDRs. J. Cell. Physiol. 231: 1695-1708, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anna Mascia
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| | - Flaviana Gentile
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| | - Antonella Izzo
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Nunzia Mollo
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Maria De Luca
- Department of Biological and Environmental Sciences and Technologies, University of Salento (DiSTeBA), Lecce, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento (DiSTeBA), Lecce, Italy
| | - Lucio Nitsch
- Department of Molecular Medicine and Medical Biotechnologies, University "Federico II", Napoli, Italy
| | - Gaetano Calì
- IEOS Istituto di Endocrinologia e Oncologia Sperimentale "G. Salvatore", National Council of Research, Napoli, Italy
| |
Collapse
|
43
|
Ito A, Shimazu T, Maeda S, Shah AA, Tsunoda T, Iemura SI, Natsume T, Suzuki T, Motohashi H, Yamamoto M, Yoshida M. The subcellular localization and activity of cortactin is regulated by acetylation and interaction with Keap1. Sci Signal 2015; 8:ra120. [DOI: 10.1126/scisignal.aad0667] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Loss of the obscurin-RhoGEF downregulates RhoA signaling and increases microtentacle formation and attachment of breast epithelial cells. Oncotarget 2015; 5:8558-68. [PMID: 25261370 PMCID: PMC4226704 DOI: 10.18632/oncotarget.2338] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Obscurins are RhoGEF-containing proteins whose downregulation has been implicated in the development and progression of breast cancer. Herein, we aim to elucidate the mechanism for increased motility of obscurin-deficient cells. We show that shRNA-mediated obscurin downregulation in MCF10A cells leads to >50% reduction in RhoA activity relative to scramble control (shCtrl), as well as decreased phosphorylation of RhoA effectors, including myosin light chain phosphatase, myosin light chain, lim kinase, and cofilin, in both attached and suspended cells. These alterations result in decreased actomyosin contractility, allowing suspended cells to escape detachment-induced apoptosis. Moreover, ~40% of shObsc-expressing cells, but only ~10% of shCtrl-expressing cells, extend microtentacles, tubulin-based projections that mediate the attachment of circulating tumor cells to endothelium. Indeed, we show that MCF10A cells expressing shObsc attach in vitro more readily than shCtrl cells, an advantage that persists following taxane exposure. Overall, our data suggest that loss of obscurins may represent a substantial selective advantage for breast epithelial cells during metastasis, and that treatment with paclitaxel may exacerbate this advantage by preferentially allowing obscurin-deficient, stem-like cells to attach to the endothelium of distant sites, a first step towards colonizing metastatic tumors.
Collapse
|
45
|
Inoue K, Ishibe S. Podocyte endocytosis in the regulation of the glomerular filtration barrier. Am J Physiol Renal Physiol 2015; 309:F398-405. [PMID: 26084928 DOI: 10.1152/ajprenal.00136.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/15/2015] [Indexed: 02/06/2023] Open
Abstract
Severe defects in the glomerular filtration barrier result in nephrotic syndrome, which is characterized by massive proteinuria. The podocyte, a specialized epithelial cell with interdigitating foot processes separated by a slit diaphragm, plays a vital role in regulating the passage of proteins from the capillary lumen to Bowman's space. Recent findings suggest a critical role for endocytosis in podocyte biology as highlighted by genetic mouse models of disease and human genetic mutations that result in the loss of the integrity of the glomerular filtration barrier. In vitro podocyte studies have also unraveled a plethora of constituents that are differentially internalized to maintain homeostasis. These observations provide a framework and impetus for understanding the precise regulation of podocyte endocytic machinery in both health and disease.
Collapse
Affiliation(s)
- Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
46
|
Dutta D, Donaldson JG. Search for inhibitors of endocytosis: Intended specificity and unintended consequences. CELLULAR LOGISTICS 2014; 2:203-208. [PMID: 23538558 PMCID: PMC3607622 DOI: 10.4161/cl.23967] [Citation(s) in RCA: 345] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We discuss here the variety of approaches that have been taken to inhibit different forms of endocytosis. Typically, both non-specific and specific chemical inhibitors of endocytosis are tried in order to “classify” entry of a new plasma membrane protein into one of the various types of endocytosis. This classification can be confirmed through genetic approaches of protein depletion or overexpression of mutants of known endocytosis machinery components. Although some new compounds have been designed to be selective in biochemical assays, we caution investigators to be alert to the unintended consequences that sometimes arise when these compounds are applied to intact cells.
Collapse
Affiliation(s)
- Dipannita Dutta
- Cell Biology & Physiology Center; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD USA
| | | |
Collapse
|
47
|
Shin NY, Choi H, Neff L, Wu Y, Saito H, Ferguson SM, De Camilli P, Baron R. Dynamin and endocytosis are required for the fusion of osteoclasts and myoblasts. ACTA ACUST UNITED AC 2014; 207:73-89. [PMID: 25287300 PMCID: PMC4195819 DOI: 10.1083/jcb.201401137] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dynamin function is essential for cell–cell fusion in both osteoclast precursors and myoblasts in part through its effects on endocytosis. Cell–cell fusion is an evolutionarily conserved process that leads to the formation of multinucleated myofibers, syncytiotrophoblasts and osteoclasts, allowing their respective functions. Although cell–cell fusion requires the presence of fusogenic membrane proteins and actin-dependent cytoskeletal reorganization, the precise machinery allowing cells to fuse is still poorly understood. Using an inducible knockout mouse model to generate dynamin 1– and 2–deficient primary osteoclast precursors and myoblasts, we found that fusion of both cell types requires dynamin. Osteoclast and myoblast cell–cell fusion involves the formation of actin-rich protrusions closely associated with clathrin-mediated endocytosis in the apposed cell. Furthermore, impairing endocytosis independently of dynamin also prevented cell–cell fusion. Since dynamin is involved in both the formation of actin-rich structures and in endocytosis, our results indicate that dynamin function is central to the osteoclast precursors and myoblasts fusion process, and point to an important role of endocytosis in cell–cell fusion.
Collapse
Affiliation(s)
- Nah-Young Shin
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hyewon Choi
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Lynn Neff
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yumei Wu
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Hiroaki Saito
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shawn M Ferguson
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510
| | - Pietro De Camilli
- Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Department of Cell Biology and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510 Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Roland Baron
- Department of Medicine, Harvard Medical School, Boston, MA 02115 Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| |
Collapse
|
48
|
Devarajan PV, Jain S, Dutta R. Infectious Diseases: Need for Targeted Drug Delivery. TARGETED DRUG DELIVERY : CONCEPTS AND DESIGN 2014. [PMCID: PMC7122176 DOI: 10.1007/978-3-319-11355-5_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infectious diseases are a leading cause of death worldwide, with the constant fear of global epidemics. It is indeed an irony that the reticuloendothelial system (RES), the body’s major defence system, is the primary site for intracellular infections which are more difficult to treat. Pro-inflammatory M1 macrophages play an important role in defence. However, ingenious pathogen survival mechanisms including phagolysosome destruction enable their persistence. Microbial biofilms present additional challenges. Low intracellular drug concentrations, drug efflux by efflux pumps and/or enzymatic degradation, emergence of multi-drug resistance (MDR), are serious limitations of conventional therapy. Targeted delivery using nanocarriers, and passive and active targeting strategies could provide quantum increase in intracellular drug concentration. Receptor mediated endocytosis using appropriate ligands is a viable approach. Liposomes and polymeric/lipidic nanoparticles, dendrimers micelles and micro/nanoemulsions could all be relied upon. Specialised targeting approaches are demonstrated for important diseases like tuberculosis, HIV and Malaria. Application of targeted delivery in the treatment of veterinary infections is exemplified and future possibilities indicated. The chapter thus provides an overview on important aspects of infectious diseases and the challenges therein, while stressing on the promise of targeted drug delivery in augmenting therapy of infectious diseases.
Collapse
Affiliation(s)
- Padma V. Devarajan
- grid.44871.3e0000000106680201Institute of Chemical Technology, Department of Pharmaceutical Sciences and Technology, Mumbai, India
| | - Sanyog Jain
- grid.419631.8000000008877852XNational Institute of Pharmaceutical Education and Research (NIPER), Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, Mohali, Punjab India
| | | |
Collapse
|
49
|
Chou AM, Sem KP, Wright GD, Sudhaharan T, Ahmed S. Dynamin1 is a novel target for IRSp53 protein and works with mammalian enabled (Mena) protein and Eps8 to regulate filopodial dynamics. J Biol Chem 2014; 289:24383-96. [PMID: 25031323 DOI: 10.1074/jbc.m114.553883] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Filopodia are dynamic actin-based structures that play roles in processes such as cell migration, wound healing, and axonal guidance. Cdc42 induces filopodial formation through IRSp53, an Inverse-Bin-Amphiphysins-Rvs (I-BAR) domain protein. Previous work from a number of laboratories has shown that IRSp53 generates filopodia by coupling membrane protrusion with actin dynamics through its Src homology 3 domain binding partners. Here, we show that dynamin1 (Dyn1), the large guanosine triphosphatase, is an interacting partner of IRSp53 through pulldown and Förster resonance energy transfer analysis, and we explore its role in filopodial formation. In neuroblastoma cells, Dyn1 localizes to filopodia, associated tip complexes, and the leading edge just behind the anti-capping protein mammalian enabled (Mena). Dyn1 knockdown reduces filopodial formation, which can be rescued by overexpressing wild-type Dyn1 but not the GTPase mutant Dyn1-K44A and the loss-of-function actin binding domain mutant Dyn1-K/E. Interestingly, dynasore, an inhibitor of Dyn GTPase, also reduced filopodial number and increased their lifetime. Using rapid time-lapse total internal reflection fluorescence microscopy, we show that Dyn1 and Mena localize to filopodia only during initiation and assembly. Dyn1 actin binding domain mutant inhibits filopodial formation, suggesting a role in actin elongation. In contrast, Eps8, an actin capping protein, is seen most strongly at filopodial tips during disassembly. Taken together, the results suggest IRSp53 partners with Dyn1, Mena, and Eps8 to regulate filopodial dynamics.
Collapse
Affiliation(s)
- Ai Mei Chou
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Kai Ping Sem
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Graham Daniel Wright
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Thankiah Sudhaharan
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Sohail Ahmed
- From the Institute of Medical Biology, Immunos, 8A Biomedical Grove, Singapore 138648, Singapore
| |
Collapse
|
50
|
Wang QC, Liu J, Wang ZB, Zhang Y, Duan X, Cui XS, Kim NH, Sun SC. Dynamin 2 regulates actin-mediated spindle migration in mouse oocytes. Biol Cell 2014; 106:193-202. [DOI: 10.1111/boc.201400007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/09/2014] [Indexed: 02/01/2023]
Affiliation(s)
- Qiao-Chu Wang
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing 210095 China
| | - Jun Liu
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing 210095 China
| | - Zhen-Bo Wang
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing 100101 China
| | - Yu Zhang
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing 210095 China
| | - Xing Duan
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing 210095 China
| | - Xiang-Shun Cui
- Department of Animal Sciences; Chungbuk National University; Cheongju Chungbuk 361-763 Korea
| | - Nam-Hyung Kim
- Department of Animal Sciences; Chungbuk National University; Cheongju Chungbuk 361-763 Korea
| | - Shao-Chen Sun
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing 210095 China
| |
Collapse
|