• Acta Photonica Sinica
  • Vol. 51, Issue 8, 0851514 (2022)
Dongyu LI1、1、1、1、1、1、1、1、1, Tingting YU1、1、1、1、1、1、1、1、1, Jingtan ZHU1、1、1、1、1、1、1、1、1, and Dan ZHU1、1、1、1、1、1、1、1、1、*
Author Affiliations
  • 11Britton Chance Center for Biomedical Photonics & MoE Key Laboratory for Biomedical Photonics,Wuhan National Laboratory for Optoelectronics,Huazhong University of Science and Technology,Wuhan 430074,China
  • 12Advanced Biomedical Imaging Facility,Huazhong University of Science and Technology,Wuhan 430074,China
  • 13Optics Valley Laboratory,Wuhan 430074,China
  • show less
    DOI: 10.3788/gzxb20225108.0851514 Cite this Article
    Dongyu LI, Tingting YU, Jingtan ZHU, Dan ZHU. In vivo Skull Optical Clearing Technique and its Applications(Invited)[J]. Acta Photonica Sinica, 2022, 51(8): 0851514 Copy Citation Text show less
    References

    [1] S LAVISSE, S GOUTAL, CWIMBERLEY et al. Increased microglial activation in patients with Parkinson disease using [18F]-DPA714 TSPO PET imaging. Parkinsonism & Related Disorders, 82, 29-36(2021).

    [2] T G JOVIN, R G NOGUEIRA, M GLANSBERG et al. Thrombectomy for anterior circulation stroke beyond 6 h from time last known well (AURORA): a systematic review and individual patient data meta-analysis. Lancet, 399, 249-258(2022).

    [3] V L VILLEMAGNE, F BARKHOF, V GARIBOTTO et al. Molecular imaging approaches in dementia. Radiology, 298, 517-530(2021).

    [4] A D JONAS OGIEN, C MAXINE, P JEAN-LUC et al. Line-field confocal optical coherence tomography for three-dimensional skin imaging. Frontiers of Optoelectronics, 13, 381-392(2020).

    [5] Zhongwen CHENG, Haigang MA, Zhiyang WANG et al. In vivo volumetric monitoring of revascularization of traumatized skin using extended depth-of-field photoacoustic microscopy. Frontiers of Optoelectronics, 13, 307-317(2020).

    [6] Xiaohui WANG, Zhipeng LI, Yadan DING et al. Enhanced photothermal-photodynamic therapy for glioma based on near-infrared dye functionalized Fe3O4 superparticles. Chemical Engineering Journal, 381, 122693-122693(2020).

    [7] D V YAKOVLEV, D S FARRAKHOVA, A A SHIRYAEV et al. New approaches to diagnostics and treatment of cholangiocellular cancer based on photonics methods. Frontiers of Optoelectronics, 13, 352-359(2020).

    [8] N KAZACHKINA, J LYMAR, V SHCHESLAVSKIY et al. A pilot study of the dynamics of tissue oxygenation in vivo using time-resolved phosphorescence imaging. Journal of Innovative Optical Health Sciences, 14, 2142001(2021).

    [9] Zhao LEI, Yun ZENG, Xiaofen ZHANG et al. Photoacoustic reporter genes for noninvasive molecular imaging and theranostics. Journal of Innovative Optical Health Sciences, 13, 2030005(2020).

    [10] Wenzhao YANG, S CHEN. Time-gated fluorescence imaging: advances in technology and biological applications. Journal of Innovative Optical Health Sciences, 13, 12-31(2020).

    [11] Guosong HONG, A L ANTARIS, Hongjie DAI. Near-infrared fluorophores for biomedical imaging. Nature Biomedical Engineering, 1, 0010(2017).

    [12] Jianglan FAN, J A RIVERA, Wei SUN et al. High-speed volumetric two-photon fluorescence imaging of neurovascular dynamics. Nature Communication, 11, 6020(2020).

    [13] K KISLER, D LAZIC, M D SWEENEY et al. In vivo imaging and analysis of cerebrovascular hemodynamic responses and tissue oxygenation in the mouse brain. Nature Protocols, 13, 1377-1402(2018).

    [14] Dongyu LI, Qing XIA, Tingting YU et al. Transmissive-detected laser speckle contrast imaging for blood flow monitoring in thick tissue: from Monte Carlo simulation to experimental demonstration. Light: Science & Applications, 10, 241(2021).

    [15] K KISLER, A M NIKOLAKOPOULOU, B V ZLOKOVIC. Microglia have a grip on brain microvasculature. Nature Communications, 12, 5290(2021).

    [16] S LEE, J M NAMGOONG, Y KIM et al. Multimodal imaging of laser speckle contrast imaging combined with mosaic filter-based hyperspectral imaging for precise surgical guidance. IEEE Transactions on Biomedical Engineering, 69, 443-452(2022).

    [17] B MARTINEZ-VEGA, R LEON, H FABELO et al. Oxygen saturation measurement using hyperspectral imaging targeting real-time monitoring, 480-487(2021).

    [18] D RAVI, H FABELO, G M CALLIC et al. Manifold embedding and semantic segmentation for intraoperative guidance with hyperspectral brain imaging. IEEE Transactions on Medical Imaging, 36, 1845-1857(2017).

    [19] A HUSSAIN, W PETERSEN, J STALIEY et al. Quantitative blood oxygen saturation imaging using combined photoacoustics and acousto-optics. Optics Letters, 41, 1720-1723(2016).

    [20] C CHEN, Ruikang WANG. Optical coherence tomography based angiography. Biomedical Optics Express, 8, 1056-1082(2017).

    [21] I MONTEIRO-HENRIQUES, A ROCHA-SOUSA, J BARBOSA-BREDA. Optical coherence tomography angiography changes in cardiovascular systemic diseases and risk factors: a review. Acta Ophthalmologica, 100, e1-e15(2022).

    [22] Bin SUN, Mengran WANG, S A HOERDER et al. Intravital imaging of the murine subventricular zone with three photon microscopy. Cerebral Cortex, 32, 3057-3067(2022).

    [23] Tianyu WANG, C XU. Three-photon neuronal imaging in deep mouse brain. Optica, 7, 947-960(2020).

    [24] Tianyu WANG, D G OUZOUNOV, Chunyan WU et al. Three-photon imaging of mouse brain structure and function through the intact skull. Nature Methods, 15, 789-792(2018).

    [25] S WEISENBURGER, F TEJERA, J DEMAS et al. Volumetric Ca2+ imaging in the mouse brain using hybrid multiplexed sculpted light microscopy. Cell, 177, 1050-1066(2019).

    [26] Wei CHEN, R G NATAN, Yuhan YANG et al. In vivo volumetric imaging of calcium and glutamate activity at synapses with high spatiotemporal resolution. Nature Communications, 12, 6630(2021).

    [27] Fei LU, Jiating CAO, Qinglun SU et al. Recent advances in fluorescence imaging of traumatic brain injury in animal models. Frontiers in Molecular Biosciences, 8, 660993(2021).

    [28] Haochen ZHANG, Dongqin YU, Shuting LIU et al. NIR‐Ⅱ Hydrogen‐Bonded Organic Frameworks (HOFs) used for target‐specific amyloid-β photooxygenation in an alzheimer's disease model. Angewandte Chemie International Edition, 61, e202109068(2022).

    [29] M KNEIPP, J TURNER, H ESTRADA et al. Effects of the murine skull in optoacoustic brain microscopy. Journal of Biophotonics, 9, 117-123(2016).

    [30] Xiaofeng FAN, Weitao ZHENG, D SINGH. Light scattering and surface plasmons on small spherical particles. Light-Science & Applications, 3, e179(2014).

    [31] A HOLTMAAT, T BONHOEFFER, D K CHOW et al. Long-term, high-resolution imaging in the mouse neocortex through a chronic cranial window. Nature Protocols, 4, 1128-1144(2009).

    [32] Guang YANG, Feng PAN, C N PARKHURST et al. Thinned-skull cranial window technique for long-term imaging of the cortex in live mice. Nature Protocols, 5, 201-208(2010).

    [33] Shanshan YANG, Kezhou LIU, Huijie DING et al. Longitudinal in vivo intrinsic optical imaging of cortical blood perfusion and tissue damage in focal photothrombosis stroke model. Journal of Cerebral Blood Flow and Metabolism, 39, 1381-1393(2019).

    [34] R D DORAND, D S BARKAUSKAS, T A EVANS et al. Comparison of intravital thinned skull and cranial window approaches to study CNS immunobiology in the mouse cortex. Intravital, 3, e21978(2014).

    [35] P J DREW, A Y SHIH, J D DRISCOLL et al. Chronic optical access through a polished and reinforced thinned skull. Nature Methods, 7, 981-U960(2010).

    [36] M MANGLANI, D B MCGAVERN. Intravital imaging of neuroimmune interactions through a thinned skull. Current Protocols in Immunology, 120, 24.2.1-24.2.12(2018).

    [37] V COELHO-SANTOS, A A BERTHIAUME, S ORNELAS et al. Imaging the construction of capillary networks in the neonatal mouse brain. Proceedings of the National Academy of Sciences of the United States of America, 118, e2100866118(2021).

    [38] Jingtan ZHU, Xiaomei LIU, Yating DENG et al. Tissue optical clearing for 3D visualization of vascular networks: a review. Vascular Pharmacology, 141, 106905(2021).

    [39] Tingting YU, Jingtan ZHU, Dongyu LI et al. Physical and chemical mechanisms of tissue optical clearing. Iscience, 24, 102178(2021).

    [40] Peng WAN, Jingtan ZHU, Jianyi XU et al. Evaluation of seven optical clearing methods in mouse brain. Neurophotonics, 5, 035007(2018).

    [41] Tingting YU, Jingtan ZHU, Yusha LI et al. RTF: a rapid and versatile tissue optical clearing method. Scientific Reports, 8, 1964(2018).

    [42] Peng WAN, Jingtan ZHU, Tingting YU et al. Comparison of seven optical clearing methods for mouse brain. Neural Imaging and Sensing, 2018, 10481(2018).

    [43] Tingting YU, Jingtan ZHU, Yusha LI et al. ReagentTF: a rapid and versatile optical clearing method for biological imaging. Neural Imaging and Sensing, 10051(2017).

    [44] Tingting YU, Yisong QI, Jianru WANG et al. Rapid and prodium iodide-compatible optical clearing method for brain tissue based on sugar/sugar-alcohol. Journal of Biomedical Optics, 21, 81203(2016).

    [45] V D GENIN, E A GENINA, V V TUCHIN et al. Glycerol effects on optical, weight and geometrical properties of skin tissue. Journal of Innovative Optical Health Sciences, 14, 2142006(2021).

    [46] E A GENINA, Y I SURKOV, I A SEREBRYAKOVA et al. Rapid ultrasound optical clearing of human light and dark skin. IEEE transactions on medical imaging, 39, 3198-3206(2020).

    [47] J ENFIELD, J MCGRATH, S M DALY et al. Enhanced in vivo visualization of the microcirculation by topical application of fructose solution confirmed with correlation mapping optical coherence tomography. Journal of Biomedical Optics, 21, 081212(2016).

    [48] Jing WANG, Yang ZHANG, Tonghui XU et al. An innovative transparent cranial window based on skull optical clearing. Laser Physics Letters, 9, 469-473(2012).

    [49] E A GENINA, A N BASHKATOV, V V TUCHIN. Optical clearing of cranial bone. Advances in Optical Technologies, 2008, 267867(2008).

    [50] Yage CHEN, Shaojun LIU, Hongji LIU et al. Coherent Raman scattering unravelling mechanisms underlying skull optical clearing for through-skull brain imaging. Analytical Chemistry, 91, 9371-9375(2019).

    [51] Dongyu LI, Zheng ZHENG, Tingting YU et al. Visible-near infrared-Ⅱ skull optical clearing window for in vivo cortical vasculature imaging and targeted manipulation. Journal of Biophotonics, 13, e202000142(2020).

    [52] Xiaoquan YANG, Yang ZHANG, Kai ZHAO et al. Skull optical clearing solution for enhancing ultrasonic and photoacoustic imaging. IEEE Transactions on Medical Imaging, 35, 1903-1906(2016).

    [53] Yanjie ZHAO, Tingting YU, Chao ZHANG et al. Skull optical clearing window for in vivo imaging of the mouse cortex at synaptic resolution. Light-Science & Applications, 7, 17153(2018).

    [54] Chao ZHANG, Wwei FENG, Yanjie ZHAO et al. A large, switchable optical clearing skull window for cerebrovascular imaging. Theranostics, 8, 2696-2708(2018).

    [55] Yage CHEN, Shaojun LIU, Hongjie LIU et al. Coherent Raman scattering unravelling mechanisms underlying skull optical clearing for through-skull brain imaging. Analytical Chemistry, 91, 9371-9375(2019).

    [56] K WELSHER, S P SHERLOCK, Hongjie DAI. Deep-tissue anatomical imaging of mice using carbon nanotube fluorophores in the second near-infrared window. Proceedings of the National Academy of Sciences of the United States of America, 108, 8943-8948(2011).

    [57] Hao WAN, Jingying YUE, Shoujun ZHU et al. A bright organic NIR-Ⅱ nanofluorophore for three-dimensional imaging into biological tissues. Nature Communications, 9, 1171(2018).

    [58] Mingxi ZHANG, Jingying YUE, Ran CUI et al. Bright quantum dots emitting at similar to 1,600 nm in the NIR-Ⅱb window for deep tissue fluorescence imaging. Proceedings of the National Academy of Sciences of the United States of America, 115, 6590-6595(2018).

    [59] Wenbin YU, Bin GUO, Hequn ZHANG et al. NIR-Ⅱ fluorescence in vivo confocal microscopy with aggregation-induced emission dots. Science Bulletin, 64, 410-416(2019).

    [60] Ji QI, Chaowei SUN, A ZEBIBULA et al. Real-time and high-resolution bioimaging with bright aggregation-induced emission dots in short-wave infrared region. Advanced Materials, 30, 1706856(2018).

    [61] Wei FENG, Shaojun LIU, Chao ZHANG et al. Comparison of cerebral and cutaneous microvascular dysfunction with the development of type 1 diabetes. Theranostics, 9, 5854-5868(2019).

    [62] Chao ZHANG, Wei FENG, E VODOVOZOVA et al. Photodynamic opening of the blood-brain barrier to high weight molecules and liposomes through an optical clearing skull window. Biomedical Optics Express, 9, 4850-4862(2018).

    [63] Wei FENG, Chao ZHANG, Tingting YU et al. In vivo monitoring blood-brain barrier permeability using spectral imaging through optical clearing skull window. Journal of Biophotonics, 12, e201800330(2019).

    [64] Zhengwu HU, Dongyu LI, Xiang ZHONG et al. In vivo tissue optical clearing assisted through-skull targeted photothrombotic ischemic stroke model in mice. Journal of Biomedical Optics, 27, 065001(2022).

    [65] Dongyu LI, Lu DENG, Zhengwu HU et al. Optical clearing imaging assisted evaluation of urokinase thrombolytic therapy on cerebral vessels with different sizes. Biomedical Optics Express, 13, 3243-3258(2022).

    [66] Lili GUO, Huan XIONG, J I KIM et al. Dynamic rewiring of neural circuits in the motor cortex in mouse models of Parkinson's disease. Nat Neurosci, 18, 1299-1309(2015).

    [67] Y DROMARD, M ARANGO-LIEVANO, P FONTANAUD et al. Dual imaging of dendritic spines and mitochondria in vivo reveals hotspots of plasticity and metabolic adaptation to stress. Neurobiology of Stress, 15, 100402(2021).

    [68] Tonghui XU, Xinzhu YU, A J PERLIK et al. Rapid formation and selective stabilization of synapses for enduring motor memories. Nature, 462, 915-919(2009).

    [69] Xinzhu YU, G WANG, A GILMORE et al. Accelerated experience-dependent pruning of cortical synapses in ephrin-A2 knockout mice. Neuron, 80, 64-71(2013).

    [70] Tonghui XU, Shaofang WANG, R R LALCHANDANI et al. Motor learning in animal models of Parkinson's disease: Aberrant synaptic plasticity in the motor cortex. Mov Disord, 32, 487-497(2017).

    [71] A Y DERBIE. Neural mechanisms of spatial coding : a multimodal imaging study(2021).

    [72] W DENG, K I TSUBOTA. Numerical simulation of the vascular structure dependence of blood flow in the kidney. Medical Engineering & Physics, 104, 103809(2022).

    [73] Y KIM, Y B LEE, S K BAE et al. Development of a photochemical thrombosis investigation system to obtain a rabbit ischemic stroke model. Scientific Reports, 11, 5787(2021).

    [74] M DRAIJER, E HONDEBRINK, T VAN LEEUWEN et al. Review of laser speckle contrast techniques for visualizing tissue perfusion. Lasers in Medical Science, 24, 639-651(2009).

    [75] D A BOAS, A K DUNN. Laser speckle contrast imaging in biomedical optics. Journal of Biomedical Optics, 15, 011109(2010).

    [76] J GUILBERT, M DESJARDINS. Movement correction method for laser speckle contrast imaging of cerebral blood flow in cranial windows in rodents. Journal of Biophotonics, 15, e202100218(2022).

    [77] L KHAODHIAR, T DINH, K T SCHOMACKER et al. The use of medical hyperspectral technology to evaluate microcirculatory changes in diabetic foot ulcers and to predict clinical outcomes. Diabetes Care, 30, 903-910(2007).

    [78] D YUDOVSKY, A NOUVONG, K SCHOMACKER et al. Monitoring temporal development and healing of diabetic foot ulceration using hyperspectral imaging. Journal of Biophotonics, 4, 565-576(2011).

    [79] W RISAU, H WOLBURG. Development of the blood-brain-barrier. Trends in Neurosciences, 13, 174-178(1990).

    [80] N J ABBOTT, I A ROMERO. Transporting therapeutics across the blood-brain barrier. Molecular Medicine Today, 2, 106-113(1996).

    [81] R S EL-BACHA, A MINN. Drug metabolizing enzymes in cerebrovascular endothelial cells afford a metabolic protection to the brain. Molecular and Cellular Biology, 45, 15-23(1999).

    [82] W M PARDRIDGE. Molecular Trojan horses for blood-brain barrier drug delivery. Current Opinion in Pharmacology, 6, 494-500(2006).

    [83] M M PATEL, B M PATEL. Crossing the blood-brain barrier: recent advances in drug delivery to the brain. Cns Drugs, 31, 109-133(2017).

    [84] S MITRAGOTRI. Devices for overcoming biological barriers: The use of physical forces to disrupt the barriers. Advanced Drug Delivery Reviews, 65, 100-103(2013).

    [85] D S HERSH, A S WADAJKAR, N B ROBERTS et al. Evolving drug delivery strategies to overcome the blood brain barrier. Current Pharmaceutical Design, 22, 1177-1193(2016).

    [86] V KIVINIEMI, V KORHONEN, J KORTELAINEN et al. Real-time monitoring of human blood-brain barrier disruption. Plos One, 12, e0174072(2017).

    [87] C POON, D MCMAHON, K HYNYNEN. Noninvasive and targeted delivery of therapeutics to the brain using focused ultrasound. Neuropharmacology, 120, 20-37(2017).

    [88] P S FISHMAN, V FRENKEL. Focused ultrasound: an emerging therapeutic modality for neurologic disease. Neurotherapeutics, 14, 393-404(2017).

    [89] S V DHURIA, L R HANSON, W H FREY. Intranasal delivery to the central nervous system: mechanisms and experimental considerations. Journal of Pharmaceutical Sciences, 99, 1654-1673(2010).

    [90] I KARAISKOS, L GALANI, F BAZIAKA et al. Intraventricular and intrathecal colistin as the last therapeutic resort for the treatment of multidrug-resistant and extensively drug-resistant Acinetobacter baumannii ventriculitis and meningitis: a literature review. International Journal of Antimicrobial Agents, 41, 499-508(2013).

    [91] H HIRSCHBERG, F A UZAL, D CHIGHVINADZE et al. Disruption of the blood-brain barrier following ALA-mediated photodynamic therapy. Lasers Surg Med, 40, 535-542(2008).

    [92] S J MADSEN, H HIRSCHBERG. Site-specific opening of the blood-brain barrier. Journal of Biophotonics, 3, 356-367(2010).

    [93] Chao ZHANG, Wei FENG, Yusha LI et al. Age differences in photodynamic therapy-mediated opening of the blood-brain barrier through the optical clearing skull window in mice. Lasers in Surgery and Medicine, 51, 625-633(2019).

    [94] D BRANT, W D D WATSON, R BUSTO et al. Induction of reproducible brain infarction by photochemically initiated thrombosis. Annals of Neurology, 17, 497-504(1985).

    [95] B D WATSON, R PRADO, A VELOSO et al. Cerebral blood flow restoration and reperfusion injury after ultraviolet laser-facilitated middle cerebral artery recanalization in rat thrombotic stroke. Stroke, 33, 428-434(2002).

    [96] M BACIGALUPPI, A SEMERANO, G S GULLOTTA et al. Insights from thrombi retrieved in stroke due to large vessel occlusion. Journal of Cerebral Blood Flow and Metabolism, 39, 1433-1451(2019).

    [97] V J MARDER, D J CHUTE, S STARKMAN et al. Analysis of thrombi retrieved from cerebral arteries of patients with acute ischemic stroke. Stroke, 37, 2086-2093(2006).

    [98] Y SUN, KUO Yimin, Hongru CHEN et al. A murine photothrombotic stroke model with an increased fibrin content and improved responses to tPA-lytic treatment. Blood Advances, 4, 1222-1231(2020).

    [99] D DAVALOS, J GRUTZENDLER, G YANG et al. ATP mediates rapid microglial response to local brain injury in vivo. Nature Neuroscience, 8, 752-758(2005).

    [100] A NIMMERJAHN, F KIRCHHOFF, F HELMCHEN. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science, 308, 1314-1318(2005).

    [101] Dongyu LI, Zhengwu HU, Hequn ZHANG et al. Solid optical clearing agents based through-Intact-Skull (TIS) window technique for long-term observation of cortical structure and function in mice. bioRxiv(2021).

    [102] Jianrong QIU, Tao HAN, Zhiyu LIU et al. Uniform focusing with an extended depth range and increased working distance for optical coherence tomography by an ultrathin monolith fiber probe. Optics Letters, 45, 976-979(2020).

    [103] Yao YU, Ziyue MENG, Ang LI et al. Monitoring of edema progression in permanent and transient MCAO model using SS-OCT. Journal of Innovative Optical Health Sciences, 14, 2140006(2021).

    [104] Xinjia HAN, Z CHAI, Xingjie PING et al. In vivo two-photon imaging reveals acute cerebral vascular spasm and microthrombosis after mild traumatic brain injury in mice. Frontiers in Neuroscience, 14, 210(2020).

    [105] Huixia REN, Zheng YANG, Chuanming LUO et al. Enriched endogenous Omega-3 fatty acids in mice ameliorate parenchymal cell death after traumatic brain injury. Molecular Neurobiology, 54, 3317-3326(2017).

    [106] T L ROTH, D NAYAK, T ATANASIJEVIC et al. Transcranial amelioration of inflammation and cell death after brain injury. Nature, 505, 223(2014).

    [107] Jinfan ZHANG, Bian LIU, I HONG et al. An ultrasensitive biosensor for high-resolution kinase activity imaging in awake mice. Nature Chemical Biology, 17, 39-46(2021).

    [108] Weijian ZONG, Runlong WU, Mingli LI et al. Fast high-resolution miniature two-photon microscopy for brain imaging in freely behaving mice. Nature Methods, 14, 713-719(2017).

    [109] Jingtao FAN, Jinli SUO, Jiamin WU et al. Video-rate imaging of biological dynamics at centimetre scale and micrometre resolution. Nature Photonics, 13, 809-816(2019).

    [110] L G F SMITH, E MILLIRON, M L HO et al. Advanced neuroimaging in traumatic brain injury: an overview. Neurosurgical Focus, 47, E17(2019).

    [111] A AZIM, B JOSEPH. Surgical critical care therapy: a clinically oriented practical approach(2018).

    [112] Zhongya QIN, Sicong HE, Chao YANG et al. Adaptive optics two-photon microscopy enables near-diffraction-limited and functional retinal imaging in vivo. Light: Science & Applications, 9, 1-11(2020).

    [113] L STREICH, J C BOFFI, Ling WANG et al. High-resolution structural and functional deep brain imaging using adaptive optics three-photon microscopy. Nature Methods, 18, 1253-1258(2021).

    [114] Kai WANG. Deep-learning-enhanced light-field microscopy. Nature Methods, 18, 459-460(2021).

    Dongyu LI, Tingting YU, Jingtan ZHU, Dan ZHU. In vivo Skull Optical Clearing Technique and its Applications(Invited)[J]. Acta Photonica Sinica, 2022, 51(8): 0851514
    Download Citation