1
|
Xu S, Qiu M, Liang L, Chen Y, Wang Y, Wu J, Chen J. Multifunctions of Sustainable Chondroitin Sulfates with Predominant Subtypes and Low Molecular Weights on Neurite Outgrowth. Biomacromolecules 2025; 26:1335-1349. [PMID: 39835408 DOI: 10.1021/acs.biomac.4c01713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Three chondroitin sulfate (CS) analogues with predominant subtypes (A, C, and E) were prepared from engineered Escherichia coli K4 combined with regioselective sulfation. CS with the designed sulfates as the main components was characterized by nuclear magnetic resonance spectroscopy, elementary analysis, and disaccharide analysis. CS prepared from the native or degraded capsular polysaccharide had molecular weights of 1.55 × 104-1.90 × 104 and 5.6 × 103-7.4 × 103, respectively. We found that CS with dual sulfates promoted the outgrowth and survival of hippocampal neurons, whereas CS with monosulfate had an inhibitory effect. CS interacted with the nerve growth factor (NGF) and tyrosine kinase (TrkA), which activated the extracellular signal-regulated kinase (ERK) signaling pathway to modulate the outgrowth of hippocampal neurons. This work clarified the multiple effects of CS on neurite outgrowth based on nonanimal-sourced glycosaminoglycans, which would benefit efforts in discovering their novel functions and therapeutic applications.
Collapse
Affiliation(s)
- Shuqin Xu
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Meiling Qiu
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Liyuan Liang
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yue Chen
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Yajia Wang
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Jing Wu
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Jinghua Chen
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
2
|
Tymanskyj SR, Escorce A, Karthikeyan S, Ma L. Optogenetic control of receptor-mediated growth cone dynamics in neurons. Mol Biol Cell 2025; 36:br5. [PMID: 39705378 PMCID: PMC11809317 DOI: 10.1091/mbc.e23-07-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/22/2024] Open
Abstract
Development of neuronal connections is spatially and temporally controlled by extracellular cues which often activate their cognate cell surface receptors and elicit localized cellular responses. Here, we demonstrate the use of an optogenetic tool to activate receptor signaling locally to induce actin-mediated growth cone remodeling in neurons. Based on the light-induced interaction between Cryptochrome 2 (CRY2) and CIB1, we generated a bicistronic vector to co-expresses CRY2 fused to the intracellular domain of a guidance receptor and a membrane-anchored CIB1. When expressed in primary neurons, activation of the growth inhibitory PlexA4 receptor induced growth cone collapse, while activation of the growth stimulating TrkA receptor increased growth cone size. Moreover, local activation of either receptor not only elicited the predicted response in light-activated growth cones but also an opposite response in neighboring no-light-exposed growth cones of the same neuron. Finally, this tool was used to reorient growth cones toward or away from the site of light activation and to stimulate local actin polymerization for branch initiation along axonal shafts. These studies demonstrate the use of an optogenetic tool for precise spatial and temporal control of receptor signaling in neurons and support its future application in investigating cellular mechanisms of neuronal development and plasticity.
Collapse
Affiliation(s)
- Stephen R. Tymanskyj
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Althea Escorce
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Siddharth Karthikeyan
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Le Ma
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
3
|
Ong Q, Lim LTR, Goh C, Liao Y, Chan SE, Lim CJY, Kam V, Yap J, Tseng T, Desrouleaux R, Wang LC, Ler SG, Lim SL, Kim SY, Sobota RM, Bennett AM, Han W, Yang X. Spatiotemporal control of subcellular O-GlcNAc signaling using Opto-OGT. Nat Chem Biol 2025; 21:300-308. [PMID: 39543398 DOI: 10.1038/s41589-024-01770-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/03/2024] [Indexed: 11/17/2024]
Abstract
The post-translational modification of intracellular proteins through O-linked β-N-acetylglucosamine (O-GlcNAc) is a conserved regulatory mechanism in multicellular organisms. Catalyzed by O-GlcNAc transferase (OGT), this dynamic modification has an essential role in signal transduction, gene expression, organelle function and systemic physiology. Here, we present Opto-OGT, an optogenetic probe that allows for precise spatiotemporal control of OGT activity through light stimulation. By fusing a photosensitive cryptochrome protein to OGT, Opto-OGT can be robustly and reversibly activated with high temporal resolution by blue light and exhibits minimal background activity without illumination. Transient activation of Opto-OGT results in mTORC activation and AMPK suppression, which recapitulate nutrient-sensing signaling. Furthermore, Opto-OGT can be customized to localize to specific subcellular sites. By targeting OGT to the plasma membrane, we demonstrate the downregulation of site-specific AKT phosphorylation and signaling outputs in response to insulin stimulation. Thus, Opto-OGT is a powerful tool for defining the role of O-GlcNAcylation in cell signaling and physiology.
Collapse
Affiliation(s)
- Qunxiang Ong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ler Ting Rachel Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Cameron Goh
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yilie Liao
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Sher En Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Crystal Jing Yi Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Valerie Kam
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jerome Yap
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Tiffany Tseng
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Reina Desrouleaux
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Loo Chien Wang
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siok Ghee Ler
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siew Lan Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sun-Yee Kim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anton M Bennett
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiping Han
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Xiaoyong Yang
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Ong Q, Yi Lim CJ, Liao Y, Tze-Yang Ng J, Rachel Lim LT, Yi Koh SX, En Chan S, Yu Ying PL, Lim H, Ye CR, Wang LC, Ler SG, Sobota RM, Sing Tan Y, Shulman GI, Yang X, Han W. Optogenetic control of Protein Kinase C-epsilon activity reveals its intrinsic signaling properties with spatiotemporal resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631444. [PMID: 39829776 PMCID: PMC11741287 DOI: 10.1101/2025.01.06.631444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The regulation of PKC epsilon (PKCε) and its downstream effects is still not fully understood, making it challenging to develop targeted therapies or interventions. A more precise tool that enables spatiotemporal control of PKCε activity is thus required. Here, we describe a photo-activatable optogenetic PKCε probe (Opto-PKCε) consisting of an engineered PKCε catalytic domain and a blue-light inducible dimerization domain. Molecular dynamics and AlphaFold simulations enable rationalization of the dark-light activity of the optogenetic probe. We first characterize the binding partners of Opto-PKCε, which are similar to those of PKCε. Subsequent validation of the Opto-PKCε tool is performed with phosphoproteome analysis, which reveals that only PKCε substrates are phosphorylated upon light activation. Opto-PKCε could be engineered for recruitment to specific subcellular locations. Activation of Opto-PKCε in isolated hepatocytes reveals its sustained activation at the plasma membrane is required for its phosphorylation of the insulin receptor at Thr1160. In addition, Opto-PKCε recruitment to the mitochondria results in its lowering of the spare respiratory capacity through phosphorylation of complex I NDUFS4. These results demonstrate that Opto-PKCε may have broad applications for the studies of PKCε signaling with high specificity and spatiotemporal resolution.
Collapse
Affiliation(s)
- Qunxiang Ong
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- These authors contributed equally
- Lead contact
| | - Crystal Jing Yi Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- These authors contributed equally
| | - Yilie Liao
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Justin Tze-Yang Ng
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ler Ting Rachel Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shernys Xuan Yi Koh
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sher En Chan
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Pheobe Lee Yu Ying
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Huijun Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chen Rui Ye
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Loo Chien Wang
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Siok Ghee Ler
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, Howard Hughes Medical Institute, Chevy Chase, MD
| | - Xiaoyong Yang
- Departments of Comparative Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520
| | - Weiping Han
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
5
|
Song Y, Huang P, Duan L. Light-Inducible Deformation of Mitochondria in Live Cells. Methods Mol Biol 2025; 2840:185-200. [PMID: 39724353 DOI: 10.1007/978-1-0716-4047-0_14] [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] [Indexed: 12/28/2024]
Abstract
Mitochondria are dynamic organelles with constantly changing morphologies. Despite recent reports indicating that mechanical cues modulate mitochondrial morphologies and functions, there is a lack of methods that can exclusively and precisely exert mechanical forces to and deform mitochondria in live cells. Therefore, how mitochondria sense and respond to mechanical forces remains largely elusive. Optogenetic methods open up new venues for remote and precise manipulation of intracellular activities using light, providing an unprecedented opportunity to establish targeted mechano-stimulation toward mitochondria. This chapter describes the development of a novel optogenetic approach to optically mechanostimulate and induce the deformation of mitochondria. In this approach, light-gated protein-protein heterodimerization recruits force-generating molecular motors to the outer mitochondrial membrane, enabling direct exertion of mechanical force on mitochondria. Details for the design, application, and experimental procedures are laid out in this chapter. This method presents a mitochondria-specific mechano-stimulator for studying the correlation between mitochondrial morphology and functions as well as mitochondrial mechanobiology.
Collapse
Affiliation(s)
- Yutong Song
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Peiyuan Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
6
|
Guo J, Xu Y, Liu J, Hou X. The involvement of lidocaine in amyloid-β1-42-dependent mitochondrial dysfunction and apoptosis in hippocampal neurons via nerve growth factor-protein kinase B pathway. Neuroreport 2024; 35:1123-1132. [PMID: 39445521 DOI: 10.1097/wnr.0000000000002105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
This project is conceived to reveal the role of lidocaine in the process of Alzheimer's disease (AD) and its possible downstream targets. After the employment of AD cell model in mice hippocampal neuronal HT-22 cells in the presence of amyloid-β1-42 (Aβ1-42), Cell Counting Kit-8 method investigated cell viability. Oxidative damage was assayed based on a dichloro-dihydro-fluorescein diacetate fluorescent probe and commercially available kits. The 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbocyanine iodide fluorescent probe estimated mitochondrial function. Terminal-deoxynucleotidyl transferase mediated nick end labeling, western blotting, and immunofluorescence appraised the apoptotic level. Western blot also ascertained the alternations of nerve growth factors (NGF)-protein kinase B (Akt) pathway-related proteins. Aβ1-42 concentration dependently triggered the viability loss, oxidative damage, and apoptosis in HT-22 cells. Lidocaine promoted the viability and reduced the mitochondrial impairment and mitochondria-dependent apoptosis in Aβ1-42-treated HT-22 cells in a concentration-dependent manner. Besides, lidocaine activated the NGF-Akt pathway and NGF absence blocked NGF-Akt pathway, aggravated mitochondrial dysfunction as well as mitochondria-dependent apoptosis in lidocaine-administrated HT-22 cells in response to Aβ1-42. Altogether, these observations concluded that lidocaine might stimulate NGF-Akt pathway to confer protection against mitochondrial impairment and apoptosis in Aβ1-42-mediated cellular model of AD.
Collapse
Affiliation(s)
- Jianlian Guo
- Department of Surgical Anesthesiology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | | | | | | |
Collapse
|
7
|
Liu A, Mohr MA, Hope JM, Wang J, Chen X, Cui B. Light-Inducible Activation of TrkA for Probing Chronic Pain in Mice. ACS Chem Biol 2024; 19:1626-1637. [PMID: 39026469 PMCID: PMC11756861 DOI: 10.1021/acschembio.4c00300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Chronic pain is a prevalent problem that plagues modern society, and better understanding its mechanisms is critical for developing effective therapeutics. Nerve growth factor (NGF) and its primary receptor, Tropomyosin receptor kinase A (TrkA), are known to be potent mediators of chronic pain, but there is a lack of established methods for precisely perturbing the NGF/TrkA signaling pathway in the study of pain and nociception. Optobiological tools that leverage light-induced protein-protein interactions allow for precise spatial and temporal control of receptor signaling. Previously, our lab reported a blue light-activated version of TrkA generated using light-induced dimerization of the intracellular TrkA domain, opto-iTrkA. In this work, we show that opto-iTrkA activation is able to activate endogenous ERK and Akt signaling pathways and causes the retrograde transduction of phospho-ERK signals in dorsal root ganglion (DRG) neurons. Opto-iTrkA activation also sensitizes the transient receptor potential vanilloid 1 (TRPV1) channel in cellular models, further corroborating the physiological relevance of the optobiological stimulus. Finally, we show that opto-iTrkA enables light-inducible potentiation of mechanical sensitization in mice. Light illumination enables nontraumatic and reversible (<2 days) sensitization of mechanical pain in mice transduced with opto-iTrkA, which provides a platform for dissecting TrkA pathways for nociception in vitro and in vivo.
Collapse
Affiliation(s)
- Aofei Liu
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Manuel A Mohr
- Department of Biology, Stanford University, Stanford, California 94305, United States
| | - Jen M Hope
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Jennifer Wang
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, California 94305, United States
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
8
|
Wang Y, Bi Z, Song Y, Duan L, Chen SC. Selective activation of photoactivatable fluorescent protein based on binary holography. BIOMEDICAL OPTICS EXPRESS 2024; 15:3382-3393. [PMID: 38855656 PMCID: PMC11161383 DOI: 10.1364/boe.519531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 06/11/2024]
Abstract
The ability to deliver laser doses to different target locations with high spatial and temporal resolution has been a long-sought goal in photo-stimulation and optogenetics research via, for example, photoactivatable proteins. These light-sensitive proteins undergo conformational changes upon photoactivation, serving functions such as triggering fluorescence, modulating ion channel activities, or initiating biochemical reactions within cells. Conventionally, photo-stimulation on light-sensitive proteins is performed by serially scanning a laser focus or via 2D projection, which is limited by relatively low spatiotemporal resolution. In this work, we present a programmable two-photon stimulation method based on a digital micromirror device (DMD) and binary holography to perform the activation of photoactivatable green fluorescent protein (PAGFP) in live cells. This method achieved grayscale and 3D selective PAGFP activation with subcellular resolution. In the experiments, we demonstrated the 3D activation capability and investigated the diffusion dynamics of activated PAGFP on the cell membrane. A regional difference in cell membrane diffusivity was observed, indicating the great potential of our approach in interrogating the spatiotemporal dynamics of cellular processes inside living cells.
Collapse
Affiliation(s)
- Yintao Wang
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, N.T., Hong Kong SAR, China
- Centre for Perceptual and Interactive Intelligence (CPII), Hong Kong Science Park, N.T., Hong Kong SAR, China
| | - Zhenyu Bi
- Department of Biomedical Engineering, The Chinese University of Hong Kong, N.T., Hong Kong SAR, China
| | - Yutong Song
- Department of Biomedical Engineering, The Chinese University of Hong Kong, N.T., Hong Kong SAR, China
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, N.T., Hong Kong SAR, China
| | - Shih-Chi Chen
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, N.T., Hong Kong SAR, China
- Centre for Perceptual and Interactive Intelligence (CPII), Hong Kong Science Park, N.T., Hong Kong SAR, China
| |
Collapse
|
9
|
Song SY, Jung YW, Shin W, Park M, Lee GW, Jeong S, An S, Kim K, Ko YB, Lee KH, Kang BH, Lee M, Yoo HJ. Endometriosis-Related Chronic Pelvic Pain. Biomedicines 2023; 11:2868. [PMID: 37893241 PMCID: PMC10603876 DOI: 10.3390/biomedicines11102868] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/17/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Endometriosis, which is the presence of endometrial stroma and glands outside the uterus, is one of the most frequently diagnosed gynecologic diseases in reproductive women. Patients with endometriosis suffer from various pain symptoms such as dysmenorrhea, dyspareunia, and chronic pelvic pain. The pathophysiology for chronic pain in patients with endometriosis has not been fully understood. Altered inflammatory responses have been shown to contribute to pain symptoms. Increased secretion of cytokines, angiogenic factors, and nerve growth factors has been suggested to increase pain. Also, altered distribution of nerve fibers may also contribute to chronic pain. Aside from local contributing factors, sensitization of the nervous system is also important in understanding persistent pain in endometriosis. Peripheral sensitization as well as central sensitization have been identified in patients with endometriosis. These sensitizations of the nervous system can also explain increased incidence of comorbidities related to pain such as irritable bowel disease, bladder pain syndrome, and vulvodynia in patients with endometriosis. In conclusion, there are various possible mechanisms behind pain in patients with endometriosis, and understanding these mechanisms can help clinicians understand the nature of the pain symptoms and decide on treatments for endometriosis-related pain symptoms.
Collapse
Affiliation(s)
- Soo Youn Song
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7 ro, Sejong 30099, Republic of Korea; (S.Y.S.); (Y.W.J.); (W.S.)
| | - Ye Won Jung
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7 ro, Sejong 30099, Republic of Korea; (S.Y.S.); (Y.W.J.); (W.S.)
| | - WonKyo Shin
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7 ro, Sejong 30099, Republic of Korea; (S.Y.S.); (Y.W.J.); (W.S.)
| | - Mia Park
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Geon Woo Lee
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Soohwa Jeong
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Sukjeong An
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Kyoungmin Kim
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Young Bok Ko
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Ki Hwan Lee
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Byung Hun Kang
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Mina Lee
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Hospital, 33, Munhwa-ro, Jung-gu, Daejeon 2868, Republic of Korea; (M.P.); (G.W.L.); (S.J.); (S.A.); (K.K.); (Y.B.K.); (K.H.L.); (B.H.K.); (M.L.)
| | - Heon Jong Yoo
- Department of Obstetrics & Gynecology, Chungnam National University School of Medicine, Chungnam National University Sejong Hospital, 20, Bodeum 7 ro, Sejong 30099, Republic of Korea; (S.Y.S.); (Y.W.J.); (W.S.)
| |
Collapse
|
10
|
Li J, Wu C, Zeng M, Zhang Y, Wei D, Sun J, Fan H. Functional material-mediated wireless physical stimulation for neuro-modulation and regeneration. J Mater Chem B 2023; 11:9056-9083. [PMID: 37649427 DOI: 10.1039/d3tb01354e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Nerve injuries and neurological diseases remain intractable clinical challenges. Despite the advantages of stem cell therapy in treating neurological disorders, uncontrollable cell fates and loss of cell function in vivo are still challenging. Recently, increasing attention has been given to the roles of external physical signals, such as electricity and ultrasound, in regulating stem cell fate as well as activating or inhibiting neuronal activity, which provides new insights for the treatment of neurological disorders. However, direct physical stimulations in vivo are short in accuracy and safety. Functional materials that can absorb energy from a specific physical field exerted in a wireless way and then release another localized physical signal hold great advantages in mediating noninvasive or minimally invasive accurate indirect physical stimulations to promote the therapeutic effect on neurological disorders. In this review, the mechanism by which various physical signals regulate stem cell fate and neuronal activity is summarized. Based on these concepts, the approaches of using functional materials to mediate indirect wireless physical stimulation for neuro-modulation and regeneration are systematically reviewed. We expect that this review will contribute to developing wireless platforms for neural stimulation as an assistance for the treatment of neurological diseases and injuries.
Collapse
Affiliation(s)
- Jialu Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
- Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, Sichuan, China
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
11
|
Mwaura AN, Marshall N, Anglesio MS, Yong PJ. Neuroproliferative dyspareunia in endometriosis and vestibulodynia. Sex Med Rev 2023; 11:323-332. [PMID: 37544766 DOI: 10.1093/sxmrev/qead033] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023]
Abstract
INTRODUCTION Endometriosis is a common cause of deep dyspareunia, while provoked vestibulodynia is a common cause of superficial dyspareunia. The etiology of dyspareunia in both conditions is multifactorial and may include the role of local nerve growth (neurogenesis or neuroproliferation) that sensitizes pelvic structures and leads to pain with contact. OBJECTIVES To review the evidence for neuroproliferative dyspareunia in endometriosis and provoked vestibulodynia. METHODS Narrative review. RESULTS The pelvic peritoneum and vulvar vestibule receive somatic and autonomic innervation. Various markers have been utilized for nerve subtypes, including pan-neuronal markers and those specific for sensory and autonomic nerve fibers. The nerve growth factor family includes neurotrophic factors, such as nerve growth factor and brain-derived neurotrophic factor, and their receptors. Studies of endometriosis and provoked vestibulodynia have demonstrated the presence of nerve fibers around endometriosis epithelium/stroma in the pelvic peritoneum and within the vulvar vestibule. The number of nerve fibers is higher in these pain conditions as compared with control tissue. Nerve growth factor expression by endometriosis stroma and by immune cells in the vulvar vestibule may be involved in local neuroproliferation. Local inflammation is implicated in this neuroproliferation, with potential roles of interleukin 1β and mast cells in both conditions. Several studies have shown a correlation between nerve fibers around endometriosis and dyspareunia severity, but studies are lacking in provoked vestibulodynia. There are several possible clinical ramifications of neuroproliferative dyspareunia in endometriosis and provoked vestibulodynia, in terms of history, examination, biopsy, and surgical and medical treatment. CONCLUSIONS A neuroproliferative subtype of dyspareunia may be implicated in endometriosis and provoked vestibulodynia. Additional research is needed to validate this concept and to integrate it into clinical studies. Neuroproliferative pathways could serve as novel therapeutic targets for the treatment of dyspareunia in endometriosis and provoked vestibulodynia.
Collapse
Affiliation(s)
- Agnes N Mwaura
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, V6H 3N1, Canada
| | - Nisha Marshall
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, V6H 3N1, Canada
| | - Michael S Anglesio
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, V6H 3N1, Canada
| | - Paul J Yong
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, V6H 3N1, Canada
| |
Collapse
|
12
|
Pal AA, Benman W, Mumford TR, Huang Z, Chow BY, Bugaj LJ. Optogenetic clustering and membrane translocation of the BcLOV4 photoreceptor. Proc Natl Acad Sci U S A 2023; 120:e2221615120. [PMID: 37527339 PMCID: PMC10410727 DOI: 10.1073/pnas.2221615120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/25/2023] [Indexed: 08/03/2023] Open
Abstract
Optogenetic tools respond to light through one of a small number of behaviors including allosteric changes, dimerization, clustering, or membrane translocation. Here, we describe a new class of optogenetic actuator that simultaneously clusters and translocates to the plasma membrane in response to blue light. We demonstrate that dual translocation and clustering of the BcLOV4 photoreceptor can be harnessed for novel single-component optogenetic tools, including for control of the entire family of epidermal growth factor receptor (ErbB1-4) tyrosine kinases. We further find that clustering and membrane translocation are mechanistically linked. Stronger clustering increased the magnitude of translocation and downstream signaling, increased sensitivity to light by ~threefold-to-fourfold, and decreased the expression levels needed for strong signal activation. Thus light-induced clustering of BcLOV4 provides a strategy to generate a new class of optogenetic tools and to enhance existing ones.
Collapse
Affiliation(s)
- Ayush Aditya Pal
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - William Benman
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Thomas R. Mumford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Zikang Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Brian Y. Chow
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Lukasz J. Bugaj
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA19104
- Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
13
|
Koga T, Ito H, Iwaoka Y, Noshita T, Tai A. Neurite Outgrowth-Promoting Compounds from the Petals of Paeonia lactiflora in PC12 Cells. Molecules 2022; 27:molecules27227670. [PMID: 36431771 PMCID: PMC9692541 DOI: 10.3390/molecules27227670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
Isorhamnetin-3-O-glucoside and astragalin, flavonol glucosides, were isolated from the petals of Paeonia lactiflora as neurite outgrowth-promoting compounds. Isoquercitrin, formed by demethylating the B ring of isorhamnetin-3-O-glucoside or by adding a hydroxyl group to the B ring of astragalin, was evaluated for neurite outgrowth-promoting activity and was compared with the activities of isorhamnetin-3-O-glucoside and astragalin. The activities of isorhamnetin, kaempferol, and quercetin, aglycones corresponding to isorhamnetin-3-O-glucoside, astragalin, and isoquercitrin, respectively, were also evaluated. Isorhamnetin-3-O-glucoside and astragalin showed much stronger neurite outgrowth-promoting activities than the activities of the other tested flavonoids. They exhibited relatively weak anti-oxidant activities and moderate AChE inhibitory activities compared to the activities of the other tested flavonoids. Isorhamnetin-3-O-glucoside and astragalin promoted morphological neurite outgrowth and the expression of neurofilaments induced by NGF in PC12 cells. Isorhamnetin-3-O-glucoside and astragalin might be candidate compounds as neural differentiation agents in peripheral nerves and functional food ingredients preventing cognitive decline.
Collapse
Affiliation(s)
- Takeru Koga
- Graduate School of Advanced Technology and Science, Tokushima University, 2-1 Minamijosanjima-cho, Tokushima 770-8506, Japan
| | - Hideyuki Ito
- Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Yuji Iwaoka
- Faculty of Health and Welfare Science, Okayama Prefectural University, 111 Kuboki, Soja, Okayama 719-1197, Japan
| | - Toshiro Noshita
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka-cho, Shobara, Hiroshima 727-0023, Japan
- Department of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu 509-0293, Japan
| | - Akihiro Tai
- Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka-cho, Shobara, Hiroshima 727-0023, Japan
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1 Minamijosanjima-cho, Tokushima 770-8513, Japan
- Correspondence:
| |
Collapse
|
14
|
Tymanskyj SR, Curran BM, Ma L. Selective axonal transport through branch junctions is directed by growth cone signaling and mediated by KIF1/kinesin-3 motors. Cell Rep 2022; 39:110748. [PMID: 35476993 PMCID: PMC9097860 DOI: 10.1016/j.celrep.2022.110748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/27/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Development and function of nerve cells rely on the orchestration of microtubule-based transport from the cell body into distal axonal terminals. Neurons often have highly elaborate branches innervating multiple targets, but how protein or membrane cargos navigate through branch junctions to specific branch targets is unknown. Here, we demonstrate that anterograde transport of membrane vesicles through axonal branch junctions is highly selective, which is influenced by branch length and more strongly by growth cone motility. Using an optogenetic tool, we demonstrate that signaling from the growth cone can rapidly direct transport through branch junctions. We further demonstrate that such transport selectivity is differentially regulated for different vesicles and mediated by the KIF1/kinesin-3 family motors. We propose that this transport regulation through branch junctions could broadly impact neuronal development, function, and regeneration.
Collapse
Affiliation(s)
- Stephen R Tymanskyj
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bridget M Curran
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Le Ma
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
15
|
Neurite Outgrowth-Promoting Compounds from Cockscomb Hydrolysate. Nutrients 2022; 14:nu14071422. [PMID: 35406035 PMCID: PMC9002945 DOI: 10.3390/nu14071422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/01/2023] Open
Abstract
Cockscomb hydrolysate was found to have neurite outgrowth-promoting activity in PC12 cells. To investigate the neurite outgrowth-promoting compounds derived from cockscomb hydrolysate, bioassay-guided purification was carried out. Purified active fractions were obtained by liquid–liquid partition, followed by column chromatography. High-performance liquid chromatography and proton nuclear magnetic resonance analyses of the purified active fractions clarified that the main compounds are threonine, alanine, valine, and methionine. By screening for 20 kinds of amino acids, it was shown that valine and methionine, but not threonine and alanine, have neurite outgrowth-promoting activity. The results of activity evaluation of the mixture of amino acids indicated that alanine enhanced the activity of valine and that the mixture of valine and methionine showed a higher ratio of neurite formation than did each of them alone. On the other hand, dipeptides formed by valine and methionine showed weak neurite outgrowth-promoting activity. A mixture of threonine, alanine, valine, and methionine at the same concentrations as those in cockscomb hydrolysate showed neurite outgrowth-promoting activity comparable to that of cockscomb hydrolysate although threonine, alanine, valine, and methionine alone did not show activity at their concentrations in cockscomb hydrolysate. Therefore, the strong neurite outgrowth-promoting activity of cockscomb hydrolysate was considered to be due to the synergistic effect of threonine, alanine, valine, and methionine.
Collapse
|
16
|
Crossman SH, Janovjak H. Light-activated receptor tyrosine kinases: Designs and applications. Curr Opin Pharmacol 2022; 63:102197. [PMID: 35245796 DOI: 10.1016/j.coph.2022.102197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 11/03/2022]
Abstract
Receptor tyrosine kinases (RTKs) are a large and essential membrane receptor family. The molecular mechanisms and physiological consequences of RTK activation depend on, for example, ligand identity, subcellular localization, and developmental or disease stage. In the past few years, genetically-encoded light-activated RTKs (Opto-RTKs) have been developed to dissect these complexities by providing reversible and spatio-temporal control over cell signaling. These methods have very recently matured to include highly-sensitive multi-color actuators. The new ability to regulate RTK activity with high precision has been recently harnessed to gain mechanistic insights in subcellular, tissue, and animal models. Because of their sophisticated engineering, Opto-RTKs may only mirror some aspects of natural activation mechanisms but nevertheless offer unique opportunities to study RTK signaling and physiology.
Collapse
Affiliation(s)
- Samuel H Crossman
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia; European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia; European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 15 Innovation Walk, Clayton, Victoria 3800, Australia; Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Sturt Road, Bedford Park, South Australia 5042, Australia.
| |
Collapse
|
17
|
Mirzayi P, Shobeiri P, Kalantari A, Perry G, Rezaei N. Optogenetics: implications for Alzheimer's disease research and therapy. Mol Brain 2022; 15:20. [PMID: 35197102 PMCID: PMC8867657 DOI: 10.1186/s13041-022-00905-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD), a critical neurodegenerative condition, has a wide range of effects on brain activity. Synaptic plasticity and neuronal circuits are the most vulnerable in Alzheimer's disease, but the exact mechanism is unknown. Incorporating optogenetics into the study of AD has resulted in a significant leap in this field during the last decades, kicking off a revolution in our knowledge of the networks that underpin cognitive functions. In Alzheimer's disease, optogenetics can help to reduce and reverse neural circuit and memory impairments. Here we review how optogenetically driven methods have helped expand our knowledge of Alzheimer's disease, and how optogenetic interventions hint at a future translation into therapeutic possibilities for further utilization in clinical settings. In conclusion, neuroscience has witnessed one of its largest revolutions following the introduction of optogenetics into the field.
Collapse
Affiliation(s)
- Parsa Mirzayi
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
| |
Collapse
|
18
|
Zhu D, Johnson HJ, Chen J, Schaffer DV. Optogenetic Application to Investigating Cell Behavior and Neurological Disease. Front Cell Neurosci 2022; 16:811493. [PMID: 35273478 PMCID: PMC8902366 DOI: 10.3389/fncel.2022.811493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
Cells reside in a dynamic microenvironment that presents them with regulatory signals that vary in time, space, and amplitude. The cell, in turn, interprets these signals and accordingly initiates downstream processes including cell proliferation, differentiation, migration, and self-organization. Conventional approaches to perturb and investigate signaling pathways (e.g., agonist/antagonist addition, overexpression, silencing, knockouts) are often binary perturbations that do not offer precise control over signaling levels, and/or provide limited spatial or temporal control. In contrast, optogenetics leverages light-sensitive proteins to control cellular signaling dynamics and target gene expression and, by virtue of precise hardware control over illumination, offers the capacity to interrogate how spatiotemporally varying signals modulate gene regulatory networks and cellular behaviors. Recent studies have employed various optogenetic systems in stem cell, embryonic, and somatic cell patterning studies, which have addressed fundamental questions of how cell-cell communication, subcellular protein localization, and signal integration affect cell fate. Other efforts have explored how alteration of signaling dynamics may contribute to neurological diseases and have in the process created physiologically relevant models that could inform new therapeutic strategies. In this review, we focus on emerging applications within the expanding field of optogenetics to study gene regulation, cell signaling, neurodevelopment, and neurological disorders, and we comment on current limitations and future directions for the growth of the field.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
| | - Hunter J. Johnson
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
- Graduate Program in Bioengineering, University of California, San Francisco, San Francisco, CA, United States
- Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA, United States
| | - Jun Chen
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
| | - David V. Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, United States
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- *Correspondence: David V. Schaffer
| |
Collapse
|
19
|
Zhang W, Zhao S, Lu L, Fan Z, Ye S. Activation of neurotrophin signalling with light‑inducible receptor tyrosine kinases. Mol Med Rep 2022; 25:70. [PMID: 35014690 PMCID: PMC8767455 DOI: 10.3892/mmr.2022.12586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/17/2020] [Indexed: 11/05/2022] Open
Abstract
Optogenetics combined with protein engineering based on natural light-sensitive dimerizing proteins has evolved as a powerful strategy to study cellular functions. The present study focused on tropomyosin kinase receptors (Trks) that have been engineered to be light-sensitive. Trk belongs to the superfamily of receptor tyrosine kinases (RTKs), which are single-pass transmembrane receptors that are activated by natural ligands and serve crucial roles in cellular growth, differentiation, metabolism and motility. However, functional variations exist among receptors fused with light-sensitive proteins. The present study proposed a signal transduction model for light-induced receptor activation. This model is based on analysis of previous light-induced Trk receptors reported to date and comparisons to the activation mechanism of natural receptors. In this model, quantitative differences on the dimerization induced from either top-to-bottom or bottom-to-up may lead to the varying amplitude of intracellular signals. We hypothesize that the top-to-bottom propagation is more favourable for activation and yields better results compared with the bottom-to-top direction. The careful delineation of the dimerization mechanisms fine-tuning activation will guide future design for an optimum cellular output with the precision of light.
Collapse
Affiliation(s)
- Wei Zhang
- Anesthesiology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China
| | - Shu Zhao
- School of Life Science, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Linjie Lu
- Institute of Genetics, Molecular and Cellular Biology, University of Strasbourg, Illkirch 67400, France
| | - Zhimin Fan
- Anesthesiology Department, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China
| | - Shixin Ye
- Institut National de la Sante et de la Recherche Medicale (INSERM) U1195, Bicetre Hospital, Paris‑Saclay University, Le Kremlin-Bicêtre 94276, France
| |
Collapse
|
20
|
Huang P, Zhao Z, Duan L. Optogenetic activation of intracellular signaling based on light-inducible protein-protein homo-interactions. Neural Regen Res 2022; 17:25-30. [PMID: 34100422 PMCID: PMC8451544 DOI: 10.4103/1673-5374.314293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dynamic protein-protein interactions are essential for proper cell functioning. Homo-interaction events—physical interactions between the same type of proteins—represent a pivotal subset of protein-protein interactions that are widely exploited in activating intracellular signaling pathways. Capacities of modulating protein-protein interactions with spatial and temporal resolution are greatly desired to decipher the dynamic nature of signal transduction mechanisms. The emerging optogenetic technology, based on genetically encoded light-sensitive proteins, provides promising opportunities to dissect the highly complex signaling networks with unmatched specificity and spatiotemporal precision. Here we review recent achievements in the development of optogenetic tools enabling light-inducible protein-protein homo-interactions and their applications in optical activation of signaling pathways.
Collapse
Affiliation(s)
- Peiyuan Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong Special Administrative Region, China
| | - Zhihao Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong Special Administrative Region, China
| | - Liting Duan
- Department of Biomedical Engineering; Shun Hing Institute of Advanced Engineering (SHIAE), The Chinese University of Hong Kong, Sha Tin, Hong Kong Special Administrative Region, China
| |
Collapse
|
21
|
Zhou JJ, Li H, Li L, Li Y, Wang PH, Meng XM, He JG. CYLD mediates human pulmonary artery smooth muscle cell dysfunction in congenital heart disease-associated pulmonary arterial hypertension. J Cell Physiol 2021; 236:6297-6311. [PMID: 33507567 DOI: 10.1002/jcp.30298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 11/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a common complication of congenital heart disease (CHD). Deubiquitinase cylindromatosis (CYLD) has been reported to significantly aggravate vascular smooth muscle cell (VSMC) phenotypic transformation, proliferation, and migration. Here, we aimed to further investigate its roles and underlying mechanisms in the CHD-PAH development. The expression of CYLD in the lung tissues from CHD-PAH patients and monocrotaline (MCT) plus aortocaval (AV)-induced PAH rats, pulmonary artery smooth muscle cells (PASMCs) from MCT-AV-induced PAH rats, and human PASMCs (HPASMCs) was evaluated. After infection with CYLD siRNA or pcNDA3.1-CYLD, the proliferation, migration, and apoptosis of HPASMCs were measured using an EdU assay, transwell and scratch wound healing assays, and flow cytometric assay, respectively. An adeno-associated virus (AAV) vector encoding CYLD was used to suppress CYLD expression by being intratracheally instilled in rats 7 days before MCT-AV treatment. The results showed that CYLD was increased in the lung tissues from CHD-PAH patients and MCT-AV-induced PAH rats, and in PASMCs from MCT-AV-induced PAH rats. The contractile-type HPASMCs expressed low levels of CYLD, while the proliferative synthetic-type HPASMCs expressed high levels of CYLD. In addition, CYLD could mediate HPASMC dysfunction, which regulated HPASMC phenotypic transformation and proliferation via the modulation of p38 and ERK activation, while CYLD regulated HPASMC migration via the modulation of p38 activation. In vivo results demonstrated that the local suppression of CYLD expression could attenuate the increased levels of PAH and its associated pulmonary vascular remodeling in MCT-AV-induced PAH rats. Collectively, these results indicated that CYLD might be a potential novel therapeutic target for the prevention of PAH and pulmonary vascular remodeling in CHD-PAH through the modulation of HPASMC dysfunction.
Collapse
Affiliation(s)
- Jing-Jing Zhou
- Center of Pulmonary Vascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huang Li
- Department of Cardiology, Guangdong Cardiovascular Institute Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Li Li
- Department of Pathology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Li
- The Animal Experimental Centre, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei-He Wang
- The Animal Experimental Centre, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xian-Min Meng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Guo He
- Center of Pulmonary Vascular Disease, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Ueki R, Hayashi S, Tsunoda M, Akiyama M, Liu H, Ueno T, Urano Y, Sando S. Nongenetic control of receptor signaling dynamics using a DNA-based optochemical tool. Chem Commun (Camb) 2021; 57:5969-5972. [PMID: 34027523 DOI: 10.1039/d1cc01968f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Optochemical tools that can modulate the activity of the target protein provide an opportunity for studying and regulating the related biological processes. Here we present a DNA-based nongenetic optochemical tool that can control the dynamics of growth factor signaling. This photo-caged mimicry of growth factor can be a promising tool for elucidating a linkage between the dynamics of signaling and the resulting biological outcomes, as well as for manipulating cellular functions and the fate of living cells.
Collapse
Affiliation(s)
- Ryosuke Ueki
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Shota Hayashi
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Masaya Tsunoda
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Momoko Akiyama
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Hanrui Liu
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan and Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Shinsuke Sando
- Department of Chemistry and Biotechnology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan and Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
23
|
Park SW, Park S, Choi HK, Park HJ, Yu W, Kim HS, Jeon M, Chung SC, Ban K, Moon S, Bae YM. Blue laser-induced selective vasorelaxation by the activation of NOSs. Microvasc Res 2021; 136:104165. [PMID: 33845105 DOI: 10.1016/j.mvr.2021.104165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 10/21/2022]
Abstract
Phototherapy has been tried for treating cardiovascular diseases. In particular, ultraviolet and blue visible lights were suggested to be useful due to their nitric oxide (NO)-production ability in the skin. However, the effects of blue light on the arterial contractility are controversial. Here, we hypothesized that appropriate protocol of blue laser can induce selective vasorelaxation by activating vasodilating signaling molecules in arteries. Using organ chamber arterial mechanics, NO assay, Matrigel assay, and microarray, we showed that a 200-Hz, 300-μs, 445-nm pulsed-laser (total energy of 600 mJ; spot size 4 mm) induced selective vasorelaxation, without vasocontraction in rat mesenteric arteries. The laser stimulation increased NO production in the cord blood-endothelial progenitor cells (CB-EPCs). Both the laser-induced vasorelaxation and NO production were inhibited by a non-selective, pan-NO synthase inhibitor, L-NG-Nitro arginine methyl ester. Microarray study in CB-EPCs suggested up-regulation of cryptochrome (CRY)2 as well as NO synthase (NOS)1 and NOSTRIN (NOS trafficking) by the laser. In conclusion, this study suggests that the 445-nm blue puled-laser can induce vasorelaxation possibly via the CRY photoreceptors and NOSs activation. The blue laser-therapy would be useful for treating systemic hypertension as well as improving local blood flow depending on the area of irradiation.
Collapse
Affiliation(s)
- Sang Woong Park
- Medical Services, Eulji University, Seongnam, Gyeonggi-do 13135, South Korea
| | - Soonjung Park
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, South Korea
| | - Hea Kyung Choi
- Medical Services, Eulji University, Seongnam, Gyeonggi-do 13135, South Korea
| | - Hyun Ji Park
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
| | - Wonjong Yu
- Department of Physical Therapy, Eulji University, 13135, South Korea
| | - Hyung-Sik Kim
- Department of Biomedical Engineering, BK21+ Research Institute of Biomedical Engineering, School of ICT Convergence Engineering, College of Science & Technology, Konkuk University Chungju, Chungbuk 380-701, South Korea
| | - Mina Jeon
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea
| | - Soon-Cheol Chung
- Department of Biomedical Engineering, BK21+ Research Institute of Biomedical Engineering, School of ICT Convergence Engineering, College of Science & Technology, Konkuk University Chungju, Chungbuk 380-701, South Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong Special Administrative Region
| | - Sunghwan Moon
- Department of Stem Cell Biology, School of Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 143-701, South Korea; Division of Stem Cell Research, T&R Biofab Co. Ltd, Seongnam-si 13494, Republic of Korea.
| | - Young Min Bae
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju, South Korea.
| |
Collapse
|
24
|
Song Y, Huang P, Liu X, Zhao Z, Wang Y, Cui B, Duan L. Light-inducible deformation of mitochondria in live cells. Cell Chem Biol 2021; 29:109-119.e3. [PMID: 34157274 DOI: 10.1016/j.chembiol.2021.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/30/2021] [Accepted: 05/26/2021] [Indexed: 12/22/2022]
Abstract
Mitochondria, the powerhouse of the cell, are dynamic organelles that undergo constant morphological changes. Increasing evidence indicates that mitochondria morphologies and functions can be modulated by mechanical cues. However, the mechano-sensing and -responding properties of mitochondria and the relation between mitochondrial morphologies and functions are unclear due to the lack of methods to precisely exert mechano-stimulation on and deform mitochondria inside live cells. Here, we present an optogenetic approach that uses light to induce deformation of mitochondria by recruiting molecular motors to the outer mitochondrial membrane via light-activated protein-protein hetero-dimerization. Mechanical forces generated by motor proteins distort the outer membrane, during which the inner mitochondrial membrane can also be deformed. Moreover, this optical method can achieve subcellular spatial precision and be combined with different optical dimerizers and molecular motors. This method presents a mitochondria-specific mechano-stimulator for studying mitochondria mechanobiology and the interplay between mitochondria shapes and functions.
Collapse
Affiliation(s)
- Yutong Song
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Peiyuan Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Xiaoying Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Zhihao Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Yijin Wang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA.
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China; Shun Hing Institute of Advanced Engineering (SHIAE), The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China.
| |
Collapse
|
25
|
Optogenetic Control of the Canonical Wnt Signaling Pathway During Xenopus laevis Embryonic Development. J Mol Biol 2021; 433:167050. [PMID: 34019868 DOI: 10.1016/j.jmb.2021.167050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/21/2022]
Abstract
Optogenetics uses light-inducible protein-protein interactions to precisely control the timing, localization, and intensity of signaling activity. The precise spatial and temporal resolution of this emerging technology has proven extremely attractive to the study of embryonic development, a program faithfully replicated to form the same organism from a single cell. We have previously performed a comparative study for optogenetic activation of receptor tyrosine kinases, where we found that the cytoplasm-to-membrane translocation-based optogenetic systems outperform the membrane-anchored dimerization systems in activating the receptor tyrosine kinase signaling in live Xenopus embryos. Here, we determine if this engineering strategy can be generalized to other signaling pathways involving membrane-bound receptors. As a proof of concept, we demonstrate that the cytoplasm-to-membrane translocation of the low-density lipoprotein receptor-related protein-6 (LRP6), a membrane-bound coreceptor for the canonical Wnt pathway, triggers Wnt activity. Optogenetic activation of LRP6 leads to axis duplication in developing Xenopus embryos, indicating that the cytoplasm-to-membrane translocation of the membrane-bound receptor could be a generalizable strategy for the construction of optogenetic systems.
Collapse
|
26
|
Kramer MM, Lataster L, Weber W, Radziwill G. Optogenetic Approaches for the Spatiotemporal Control of Signal Transduction Pathways. Int J Mol Sci 2021; 22:5300. [PMID: 34069904 PMCID: PMC8157557 DOI: 10.3390/ijms22105300] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 02/06/2023] Open
Abstract
Biological signals are sensed by their respective receptors and are transduced and processed by a sophisticated intracellular signaling network leading to a signal-specific cellular response. Thereby, the response to the signal depends on the strength, the frequency, and the duration of the stimulus as well as on the subcellular signal progression. Optogenetic tools are based on genetically encoded light-sensing proteins facilitating the precise spatiotemporal control of signal transduction pathways and cell fate decisions in the absence of natural ligands. In this review, we provide an overview of optogenetic approaches connecting light-regulated protein-protein interaction or caging/uncaging events with steering the function of signaling proteins. We briefly discuss the most common optogenetic switches and their mode of action. The main part deals with the engineering and application of optogenetic tools for the control of transmembrane receptors including receptor tyrosine kinases, the T cell receptor and integrins, and their effector proteins. We also address the hallmarks of optogenetics, the spatial and temporal control of signaling events.
Collapse
Affiliation(s)
- Markus M. Kramer
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
- SGBM—Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Levin Lataster
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
| | - Wilfried Weber
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
- SGBM—Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Gerald Radziwill
- Faculty of Biology and Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany; (M.M.K.); (L.L.); (W.W.)
| |
Collapse
|
27
|
Astragalus membranaceus Injection Protects Retinal Ganglion Cells by Regulating the Nerve Growth Factor Signaling Pathway in Experimental Rat Traumatic Optic Neuropathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2020:2429843. [PMID: 33381196 PMCID: PMC7762646 DOI: 10.1155/2020/2429843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Activation of the nerve growth factor (NGF) signaling pathway is a potential method of treatment for retinal ganglion cell (RGC) loss due to traumatic optic neuropathy (TON). The present study aimed to explore the biological effects of injecting Astragalus membranaceus (A. mem) on RGCs in an experimental TON model. Adult male Wistar rats were randomly divided into three groups: sham-operated (SL), model (ML), and A. mem injection (AL). The left eyes of the rats were considered the experimental eyes, and the right eyes served as the controls. AL rats received daily intraperitoneal injections of A. mem (3 mL/kg), whereas ML and SL rats were administered the same volume of normal saline. The TON rat model was induced by optic nerve (ON) transverse quantitative traction. After two-week administration, the number of RGCs was determined using retrograde labeling with Fluoro-Gold. The protein levels of NGF, tyrosine kinase receptor A (TrkA), c-Jun N-terminal protein kinase (JNK), JNK phosphorylation (p-JNK), and nuclear factor kappa-B (NF-κB) were assessed using western blotting. The levels of p75 neurotrophin receptor (p75NTR) and NF-κB DNA binding were examined using real-time PCR and an electrophoretic mobility shift assay. In addition, the concentrations of JNK and p-JNK were assessed using an enzyme-linked immunosorbent assay. Results. The number of RGCs in ML was found to be significantly decreased (P < 0.01) relative to both AL and SL, together with the downregulation of NGF (P < 0.01), TrkA (P < 0.05), and NF-κB (P < 0.01); upregulation of p75NTR mRNA (P < 0.01); and increased protein levels of JNK (P < 0.05) and p-JNK (P < 0.05). Treatment using A. mem injection significantly preserved the density of RGCs in rats with experimental TON and markedly upregulated the proteins of NGF (P < 0.01), TrkA (P < 0.05), and NF-κB (P < 0.01) and downregulated the mRNA level of p75NTR(P < 0.01), as well as the proteins of JNK (P < 0.05) and p-JNK (P < 0.01). Thus, A. mem injection could reduce RGC death in TON induced by ON transverse quantitative traction by stimulating the NGF signaling pathway.
Collapse
|
28
|
Photosensitive tyrosine analogues unravel site-dependent phosphorylation in TrkA initiated MAPK/ERK signaling. Commun Biol 2020; 3:706. [PMID: 33239753 PMCID: PMC7689462 DOI: 10.1038/s42003-020-01396-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/14/2020] [Indexed: 01/01/2023] Open
Abstract
Tyrosine kinase A (TrkA) is a membrane receptor which, upon ligand binding, activates several pathways including MAPK/ERK signaling, implicated in a spectrum of human pathologies; thus, TrkA is an emerging therapeutic target in treatment of neuronal diseases and cancer. However, mechanistic insights into TrKA signaling are lacking due to lack of site-dependent phosphorylation control. Here we engineer two light-sensitive tyrosine analogues, namely p-azido-L-phenylalanine (AzF) and the caged-tyrosine (ONB), through amber codon suppression to optically manipulate the phosphorylation state of individual intracellular tyrosines in TrkA. We identify TrkA-AzF and ONB mutants, which can activate the ERK pathway in the absence of NGF ligand binding through light control. Our results not only reveal how TrkA site-dependent phosphorylation controls the defined signaling process, but also extend the genetic code expansion technology to enable regulation of receptor-type kinase activation by optical control at the precision of a single phosphorylation site. It paves the way for comprehensive analysis of kinase-associated pathways as well as screening of compounds intervening in a site-directed phosphorylation pathway for targeted therapy. Using genetic code expansion, Zhao, Shi et al. generate light-sensitive tyrosine analogues to obtain insights into the activation of the NGF receptor, TrkA. They identify light-sensitive and NGF-insensitive phosphorylation sites, validating the approach and providing insights into TrkA signaling
Collapse
|
29
|
Neurite Outgrowth-Promoting Activity of Compounds in PC12 Cells from Sunflower Seeds. Molecules 2020; 25:molecules25204748. [PMID: 33081156 PMCID: PMC7587564 DOI: 10.3390/molecules25204748] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 11/21/2022] Open
Abstract
In the current super-aging society, the establishment of methods for prevention and treatment of Alzheimer’s disease (AD) is an urgent task. One of the causes of AD is thought to be a decrease in the revel of nerve growth factor (NGF) in the brain. Compounds showing NGF-mimicking activity and NGF-enhancing activity have been examined as possible agents for improving symptoms. In the present study, sunflower seed extract was found to have neurite outgrowth-promoting activity, which is an NGF-enhancing activity, in PC12 cells. To investigate neurite outgrowth-promoting compounds from sunflower seed extract, bioassay-guided purification was carried out. The purified active fraction was obtained by liquid-liquid partition followed by some column chromatographies. Proton nuclear magnetic resonance and gas chromatography-mass spectrometry analyses of the purified active fraction indicated that the fraction was a mixture of β-sitosterol, stigmasterol and campesterol, with β-sitosterol being the main component. Neurite outgrowth-promoting activities of β-sitosterol, stigmasterol, campesterol and cholesterol were evaluated in PC12 cells. β-Sitosterol and stigmasterol showed the strongest activity of the four sterol compounds (β-sitosterol ≈ stigmasterol > campesterol > cholesterol), and cholesterol did not show any activity. The results indicated that β-sitosterol was the major component responsible for the neurite outgrowth-promoting activity of sunflower seeds. Results of immunostaining also showed that promotion by β-sitosterol of neurite formation induced by NGF was accompanied by neurofilament expression. β-Sitosterol, which showed NGF-enhancing activity, might be a candidate ingredient in food for prevention of AD.
Collapse
|
30
|
Leopold AV, Verkhusha VV. Light control of RTK activity: from technology development to translational research. Chem Sci 2020; 11:10019-10034. [PMID: 33209247 PMCID: PMC7654314 DOI: 10.1039/d0sc03570j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Inhibition of receptor tyrosine kinases (RTKs) by small molecule inhibitors and monoclonal antibodies is used to treat cancer. Conversely, activation of RTKs with their ligands, including growth factors and insulin, is used to treat diabetes and neurodegeneration. However, conventional therapies that rely on injection of RTK inhibitors or activators do not provide spatiotemporal control over RTK signaling, which results in diminished efficiency and side effects. Recently, a number of optogenetic and optochemical approaches have been developed that allow RTK inhibition or activation in cells and in vivo with light. Light irradiation can control RTK signaling non-invasively, in a dosed manner, with high spatio-temporal precision, and without the side effects of conventional treatments. Here we provide an update on the current state of the art of optogenetic and optochemical RTK technologies and the prospects of their use in translational studies and therapy.
Collapse
Affiliation(s)
- Anna V Leopold
- Medicum , Faculty of Medicine , University of Helsinki , Helsinki 00290 , Finland
| | - Vladislav V Verkhusha
- Medicum , Faculty of Medicine , University of Helsinki , Helsinki 00290 , Finland
- Department of Anatomy and Structural Biology and Gruss-Lipper Biophotonics Center , Albert Einstein College of Medicine , Bronx , NY 10461 , USA .
| |
Collapse
|
31
|
Kainrath S, Janovjak H. Design and Application of Light-Regulated Receptor Tyrosine Kinases. Methods Mol Biol 2020; 2173:233-246. [PMID: 32651922 DOI: 10.1007/978-1-0716-0755-8_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Understanding how the activity of membrane receptors and cellular signaling pathways shapes cell behavior is of fundamental interest in basic and applied research. Reengineering receptors to react to light instead of their cognate ligands allows for generating defined signaling inputs with high spatial and temporal precision and facilitates the dissection of complex signaling networks. Here, we describe fundamental considerations in the design of light-regulated receptor tyrosine kinases (Opto-RTKs) and appropriate control experiments. We also introduce methods for transient receptor expression in HEK293 cells, quantitative assessment of signaling activity in reporter gene assays, semiquantitative assessment of (in)activation time courses through Western blot (WB) analysis, and easy to implement light stimulation hardware.
Collapse
Affiliation(s)
- Stephanie Kainrath
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria.,Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.,European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton, VIC, Australia
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia. .,European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, Clayton, VIC, Australia.
| |
Collapse
|
32
|
Construction of Light-Activated Neurotrophin Receptors Using the Improved Light-Induced Dimerizer (iLID). J Mol Biol 2020; 432:3739-3748. [PMID: 32335036 PMCID: PMC9879133 DOI: 10.1016/j.jmb.2020.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
Receptor tyrosine kinases (RTKs) play crucial roles in human health, and their misregulation is implicated in disorders ranging from neurodegenerative diseases to cancers. The highly conserved mechanism of activation of RTKs makes them especially appealing candidates for control via optogenetic dimerization methods. This work offers a strategy for using the improved light-induced dimer (iLID) system with a constructed tandem dimer of its binding partner nano (tdnano) to build light-activatable versions of RTKs. In the absence of light, the iLID-RTK is cytosolic, monomeric, and inactive. Under blue light, the iLID + tdnano system recruits two copies of iLID-RTK to tdnano, dimerizing, and activating the RTK. We demonstrate that iLID opto-iTrkA and opto-iTrkB are capable of reproducing downstream ERK and Akt signaling only in the presence of tdnano. We further show with our opto-iTrkA that the system is compatible with multi-day and population-level activation of TrkA in PC12 cells. By leveraging genetic targeting of tdnano, we achieve RTK activation at a specific subcellular location even with whole-cell illumination, allowing us to confidently probe the impact of context on signaling outcome.
Collapse
|
33
|
Huang P, Liu A, Song Y, Hope JM, Cui B, Duan L. Optical Activation of TrkB Signaling. J Mol Biol 2020; 432:3761-3770. [PMID: 32422149 DOI: 10.1016/j.jmb.2020.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 01/04/2023]
Abstract
Brain-derived neurotrophic factor, via activation of tropomyosin receptor kinase B (TrkB), plays a critical role in neuronal proliferation, differentiation, survival, and death. Dysregulation of TrkB signaling is implicated in neurodegenerative disorders and cancers. Precise activation of TrkB signaling with spatial and temporal resolution is greatly desired to study the dynamic nature of TrkB signaling and its role in related diseases. Here we develop different optogenetic approaches that use light to activate TrkB signaling. Utilizing the photosensitive protein Arabidopsis thaliana cryptochrome 2, the light-inducible homo-interaction of the intracellular domain of TrkB in the cytosol or on the plasma membrane is able to induce the activation of downstream MAPK/ERK and PI3K/Akt signaling as well as the neurite outgrowth of PC12 cells. Moreover, we prove that such strategies are generalizable to other optical homo-dimerizers by demonstrating the optical TrkB activation based on the light-oxygen-voltage domain of aureochrome 1 from Vaucheria frigida. The results open up new possibilities of many other optical platforms to activate TrkB signaling to fulfill customized needs. By comparing all the different strategies, we find that the cryptochrome 2-integrated approach to achieve light-induced cell membrane recruitment and homo-interaction of intracellular domain of TrkB is most efficient in activating TrkB signaling. The optogenetic strategies presented are promising tools to investigate brain-derived neurotrophic factor/TrkB signaling with tight spatial and temporal control.
Collapse
Affiliation(s)
- Peiyuan Huang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Aofei Liu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Yutong Song
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China
| | - Jen M Hope
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Liting Duan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, China.
| |
Collapse
|
34
|
Krishnamurthy VV, Fu J, Oh TJ, Khamo J, Yang J, Zhang K. A Generalizable Optogenetic Strategy to Regulate Receptor Tyrosine Kinases during Vertebrate Embryonic Development. J Mol Biol 2020; 432:3149-3158. [PMID: 32277988 DOI: 10.1016/j.jmb.2020.03.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 12/22/2022]
Abstract
Ligand-independent activation of receptor tyrosine kinases (RTKs) allows for dissecting out the receptor-specific signaling outcomes from the pleiotropic effects of the ligands. In this regard, RTK intracellular domains (ICD) are of interest due to their ability to recapitulate signaling activity in a ligand-independent manner when fused to chemical or optical dimerizing domains. A common strategy for synthetic activation of RTKs involves membrane tethering of dimerizer-RTK ICD fusions. Depending on the intrinsic signaling capacity, however, this approach could entail undesirable baseline signaling activity in the absence of stimulus, thereby diminishing the system's sensitivity. Here, we observed toxicity in early Xenopus laevis embryos when using such a conventional optogenetic design for the fibroblast growth factor receptor (FGFR). To surpass this challenge, we developed a cytoplasm-to-membrane translocation approach, where FGFR ICD is recruited from the cytoplasm to the plasma membrane by light, followed by its subsequent activation via homo-association. This strategy results in the optical activation of FGFR with low background activity and high sensitivity, which allows for the light-mediated formation of ectopic tail-like structures in developing X. laevis embryos. We further generalized this strategy by developing optogenetic platforms to control three neurotrophic tropomyosin receptor kinases, TrkA, TrkB, and TrkC. We envision that these ligand-independent optogenetic RTKs will provide useful toolsets for the delineation of signaling sub-circuits in developing vertebrate embryos.
Collapse
Affiliation(s)
- Vishnu V Krishnamurthy
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jia Fu
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Teak-Jung Oh
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - John Khamo
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA.
| | - Kai Zhang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
35
|
Wei Y, Liang Y, Lin H, Dai Y, Yao S. Autonomic nervous system and inflammation interaction in endometriosis-associated pain. J Neuroinflammation 2020; 17:80. [PMID: 32145751 PMCID: PMC7060607 DOI: 10.1186/s12974-020-01752-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Endometriosis is a chronic inflammatory disease. Pain is the most common symptom in endometriosis. Endometriosis-associated pain is caused by inflammation, and is related to aberrant innervation. Although the specific mechanism between endometriosis-associated pain and the interaction of aberrant innervation and inflammation remains unclear, many studies have confirmed certain correlations between them. In addition, we found that some chronic inflammatory autoimmune diseases (AIDs) such as inflammatory bowel disease (IBD) and rheumatoid arthritis (RA) share similar characteristics: the changes in dysregulation of inflammatory factors as well as the function and innervation of the autonomic nervous system (ANS). The mechanisms underlying the interaction between the ANS and inflammation have provided new advances among these disorders. Therefore, the purpose of this review is to compare the changes in inflammation and ANS in endometriosis, IBD, and RA; and to explore the role and possible mechanism of sympathetic and parasympathetic nerves in endometriosis-associated inflammation by referring to IBD and RA studies to provide some reference for further endometriosis research and treatment.
Collapse
Affiliation(s)
- Yajing Wei
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China
| | - Haishan Lin
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510089, China
| | - Yujing Dai
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510089, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Sun Yat-Sen University, No. 58, the 2nd Zhongshan Road, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
36
|
Abstract
Optogenetic approaches are transforming quantitative studies of cell-signaling systems. A recently developed photoswitchable mitogen-activated protein kinase kinase 1 (MEK1) enzyme (psMEK) short-circuits the highly conserved Extracellular Signal-Regulated Kinase (ERK)-signaling cascade at the most proximal step of effector kinase activation. However, since this optogenetic tool relies on phosphorylation-mimicking substitutions in the activation loop of MEK, its catalytic activity is predicted to be substantially lower than that of wild-type MEK that has been phosphorylated at these residues. Here, we present evidence that psMEK indeed has suboptimal functionality in vivo and propose a strategy to circumvent this limitation by harnessing gain-of-function, destabilizing mutations in MEK. Specifically, we demonstrate that combining phosphomimetic mutations with additional mutations in MEK, chosen for their activating potential, restores maximal kinase activity in vitro. We establish that this modification can be tuned by the choice of the destabilizing mutation and does not interfere with reversible activation of psMEK in vivo in both Drosophila and zebrafish. To illustrate the types of perturbations enabled by optimized psMEK, we use it to deliver pulses of ERK activation during zebrafish embryogenesis, revealing rheostat-like responses of an ERK-dependent morphogenetic event.
Collapse
|
37
|
Hu W, Li Q, Li B, Ma K, Zhang C, Fu X. Optogenetics sheds new light on tissue engineering and regenerative medicine. Biomaterials 2019; 227:119546. [PMID: 31655444 DOI: 10.1016/j.biomaterials.2019.119546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 01/23/2023]
Abstract
Optogenetics has demonstrated great potential in the fields of tissue engineering and regenerative medicine, from basic research to clinical applications. Spatiotemporal encoding during individual development has been widely identified and is considered a novel strategy for regeneration. A as a noninvasive method with high spatiotemporal resolution, optogenetics are suitable for this strategy. In this review, we discuss roles of dynamic signal coding in cell physiology and embryonic development. Several optogenetic systems are introduced as ideal optogenetic tools, and their features are compared. In addition, potential applications of optogenetics for tissue engineering are discussed, including light-controlled genetic engineering and regulation of signaling pathways. Furthermore, we present how emerging biomaterials and photoelectric technologies have greatly promoted the clinical application of optogenetics and inspired new concepts for optically controlled therapies. Our summation of currently available data conclusively demonstrates that optogenetic tools are a promising method for elucidating and simulating developmental processes, thus providing vast prospects for tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Wenzhi Hu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medicine Science, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, PR China; Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China
| | - Qiankun Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medicine Science, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, PR China; Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China
| | - Bingmin Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medicine Science, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, PR China; Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China
| | - Kui Ma
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China
| | - Cuiping Zhang
- Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China.
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medicine Science, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, PR China; Key Laboratory of Tissue Repair and Regeneration of PLA and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Fourth Medical Center, Chinese PLA General Hospital, 100048, Beijing, PR China.
| |
Collapse
|
38
|
Di Donato M, Cernera G, Migliaccio A, Castoria G. Nerve Growth Factor Induces Proliferation and Aggressiveness In Prostate Cancer Cells. Cancers (Basel) 2019; 11:E784. [PMID: 31174415 PMCID: PMC6627659 DOI: 10.3390/cancers11060784] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
Resistance to hormone therapy and disease progression is the major challenge in clinical management of prostate cancer (PC). Drugs currently used in PC therapy initially show a potent antitumor effects, but PC gradually develops resistance, relapses and spreads. Most patients who fail primary therapy and have recurrences eventually develop castration-resistant prostate cancer (CRPC), which is almost incurable. The nerve growth factor (NGF) acts on a variety of non-neuronal cells by activating the NGF tyrosine-kinase receptor, tropomyosin receptor kinase A (TrkA). NGF signaling is deregulated in PC. In androgen-dependent PC cells, TrkA mediates the proliferative action of NGF through its crosstalk with the androgen receptor (AR). Epithelial PC cells, however, acquire the ability to express NGF and TrkA, as the disease progresses, indicating a role for NGF/TrkA axis in PC progression and androgen-resistance. We here report that once activated by NGF, TrkA mediates proliferation, invasiveness and epithelial-mesenchymal transition (EMT) in various CRPC cells. NGF promotes organoid growth in 3D models of CRPC cells, and specific inhibition of TrkA impairs all these responses. Thus TrkA represents a new biomarker to target in CRPC.
Collapse
Affiliation(s)
- Marzia Di Donato
- Department of Precision Medicine-University of Campania 'L. Vanvitelli'-via L. De Crecchio, 7-80138 Naples, Italy.
| | - Gustavo Cernera
- Department of Precision Medicine-University of Campania 'L. Vanvitelli'-via L. De Crecchio, 7-80138 Naples, Italy.
| | - Antimo Migliaccio
- Department of Precision Medicine-University of Campania 'L. Vanvitelli'-via L. De Crecchio, 7-80138 Naples, Italy.
| | - Gabriella Castoria
- Department of Precision Medicine-University of Campania 'L. Vanvitelli'-via L. De Crecchio, 7-80138 Naples, Italy.
| |
Collapse
|
39
|
Jeknić S, Kudo T, Covert MW. Techniques for Studying Decoding of Single Cell Dynamics. Front Immunol 2019; 10:755. [PMID: 31031756 PMCID: PMC6470274 DOI: 10.3389/fimmu.2019.00755] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
Cells must be able to interpret signals they encounter and reliably generate an appropriate response. It has long been known that the dynamics of transcription factor and kinase activation can play a crucial role in selecting an individual cell's response. The study of cellular dynamics has expanded dramatically in the last few years, with dynamics being discovered in novel pathways, new insights being revealed about the importance of dynamics, and technological improvements increasing the throughput and capabilities of single cell measurements. In this review, we highlight the important developments in this field, with a focus on the methods used to make new discoveries. We also include a discussion on improvements in methods for engineering and measuring single cell dynamics and responses. Finally, we will briefly highlight some of the many challenges and avenues of research that are still open.
Collapse
Affiliation(s)
- Stevan Jeknić
- Department of Bioengineering, Stanford University, Stanford, CA, United States.,Allen Discovery Center for Systems Modeling of Infection, Stanford, CA, United States
| | - Takamasa Kudo
- Allen Discovery Center for Systems Modeling of Infection, Stanford, CA, United States.,Department of Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA, United States.,Allen Discovery Center for Systems Modeling of Infection, Stanford, CA, United States.,Department of Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| |
Collapse
|
40
|
Membrane-Associated, Not Cytoplasmic or Nuclear, FGFR1 Induces Neuronal Differentiation. Cells 2019; 8:cells8030243. [PMID: 30875802 PMCID: PMC6468866 DOI: 10.3390/cells8030243] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/04/2019] [Accepted: 03/08/2019] [Indexed: 01/22/2023] Open
Abstract
The intracellular transport of receptor tyrosine kinases results in the differential activation of various signaling pathways. In this study, optogenetic stimulation of fibroblast growth factor receptor type 1 (FGFR1) was performed to study the effects of subcellular targeting of receptor kinases on signaling and neurite outgrowth. The catalytic domain of FGFR1 fused to the algal light-oxygen-voltage-sensing (LOV) domain was directed to different cellular compartments (plasma membrane, cytoplasm and nucleus) in human embryonic kidney (HEK293) and pheochromocytoma (PC12) cells. Blue light stimulation elevated the pERK and pPLCγ1 levels in membrane-opto-FGFR1-transfected cells similarly to ligand-induced receptor activation; however, no changes in pAKT levels were observed. PC12 cells transfected with membrane-opto-FGFR1 exhibited significantly longer neurites after light stimulation than after growth factor treatment, and significantly more neurites extended from their cell bodies. The activation of cytoplasmic FGFR1 kinase enhanced ERK signaling in HEK293 cells but not in PC12 cells and did not induce neuronal differentiation. The stimulation of FGFR1 kinase in the nucleus also did not result in signaling changes or neurite outgrowth. We conclude that FGFR1 kinase needs to be associated with membranes to induce the differentiation of PC12 cells mainly via ERK activation.
Collapse
|