211 results on '"Rowitch DH"'
Search Results
2. A Correction to the Research Article Titled: "Neural Stem Cell Engraftment and Myelination in the Human Brain" by N. Gupta, R. G. Henry, J. Strober, S.-M. Kang, D. A. Lim, M. Bucci, E. Caverzasi, L. Gaetano, M. L. Mandelli, T. Ryan, R. Perry, J. Farrell, R. J. Jeremy, M. Ulman, S. L. Huhn, A. J. Barkovich, D. H. Rowitch
- Author
-
Gupta, N, Henry, RG, Strober, J, Kang, SM, Lim, DA, Bucci, M, Caverzasi, E, Gaetano, L, Mandelli, ML, Ryan, T, Perry, R, Farrell, J, Jeremy, RJ, Ulman, M, Huhn, SL, Barkovich, AJ, and Rowitch, DH
- Subjects
Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Neurosciences ,Biological Sciences ,Medical and Health Sciences - Published
- 2012
3. Neural stem cell engraftment and myelination in the human brain (Science Translational Medicine (1988) 17, (351-360))
- Author
-
Gupta, N, Henry, RG, Strober, J, Kang, SM, Lim, DA, Bucci, M, Caverzasi, E, Gaetano, L, Mandelli, ML, Ryan, T, Perry, R, Farrell, J, Jeremy, RJ, Ulman, M, Huhn, SL, Barkovich, AJ, and Rowitch, DH
- Subjects
Biological Sciences ,Medical and Health Sciences - Published
- 2012
4. MC3R links nutritional state to childhood growth and the timing of puberty
- Author
-
Lam, BYH, Williamson, A, Finer, S, Day, FR, Tadross, JA, Gonçalves Soares, A, Wade, K, Sweeney, P, Bedenbaugh, MN, Porter, DT, Melvin, A, Ellacott, KLJ, Lippert, RN, Buller, S, Rosmaninho-Salgado, J, Dowsett, GKC, Ridley, KE, Xu, Z, Cimino, I, Rimmington, D, Rainbow, K, Duckett, K, Holmqvist, S, Khan, A, Dai, X, Bochukova, EG, Genes & Health Research Team, Trembath, RC, Martin, HC, Coll, AP, Rowitch, DH, Wareham, NJ, Van Heel, DA, Timpson, N, Simerly, RB, Ong, KK, Cone, RD, Langenberg, C, Perry, JRB, Yeo, GS, O'Rahilly, S, Lam, BYH [0000-0002-3638-9025], Williamson, A [0000-0002-7599-9301], Finer, S [0000-0002-2684-4653], Day, FR [0000-0003-3789-7651], Tadross, JA [0000-0002-8424-1252], Wade, K [0000-0003-3362-6280], Porter, DT [0000-0002-8042-3251], Cimino, I [0000-0003-1397-5408], Holmqvist, S [0000-0001-6709-6666], Khan, A [0000-0002-5189-6906], Martin, HC [0000-0002-4454-9084], Rowitch, DH [0000-0002-0079-0060], Wareham, NJ [0000-0003-1422-2993], van Heel, DA [0000-0002-0637-2265], Timpson, N [0000-0002-7141-9189], Simerly, RB [0000-0001-5840-0152], Ong, KK [0000-0003-4689-7530], Cone, RD [0000-0003-3333-5651], Langenberg, C [0000-0002-5017-7344], Perry, JRB [0000-0001-6483-3771], Yeo, GS [0000-0001-8823-3615], O'Rahilly, S [0000-0003-2199-4449], and Apollo - University of Cambridge Repository
- Subjects
Aged, 80 and over ,Male ,Menarche ,Time Factors ,Adolescent ,Homozygote ,Puberty ,Hypothalamus ,Nutritional Status ,Estrous Cycle ,Weight Gain ,Melanocortins ,Mice ,Child Development ,Phenotype ,Animals ,Humans ,Female ,Sexual Maturation ,Insulin-Like Growth Factor I ,Child ,Receptor, Melanocortin, Type 3 - Abstract
The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.
- Published
- 2021
- Full Text
- View/download PDF
5. Brain charts for the human lifespan
- Author
-
Bethlehem, RAI, Seidlitz, J, White, SR, Vogel, JW, Anderson, KM, Adamson, C, Adler, S, Alexopoulos, GS, Anagnostou, E, Areces-Gonzalez, A, Astle, DE, Auyeung, B, Ayub, M, Bae, J, Ball, G, Baron-Cohen, S, Beare, R, Bedford, SA, Benegal, V, Beyer, F, Blangero, J, Blesa Cabez, M, Boardman, JP, Borzage, M, Bosch-Bayard, JF, Bourke, N, Calhoun, VD, Chakravarty, MM, Chen, C, Chertavian, C, Chetelat, G, Chong, YS, Cole, JH, Corvin, A, Costantino, M, Courchesne, E, Crivello, F, Cropley, VL, Crosbie, J, Crossley, N, Delarue, M, Delorme, R, Desrivieres, S, Devenyi, GA, Di Biase, MA, Dolan, R, Donald, KA, Donohoe, G, Dunlop, K, Edwards, AD, Elison, JT, Ellis, CT, Elman, JA, Eyler, L, Fair, DA, Feczko, E, Fletcher, PC, Fonagy, P, Franz, CE, Galan-Garcia, L, Gholipour, A, Giedd, J, Gilmore, JH, Glahn, DC, Goodyer, IM, Grant, PE, Groenewold, NA, Gunning, FM, Gur, RE, Gur, RC, Hammill, CF, Hansson, O, Hedden, T, Heinz, A, Henson, RN, Heuer, K, Hoare, J, Holla, B, Holmes, AJ, Holt, R, Huang, H, Im, K, Ipser, J, Jack, CR, Jackowski, AP, Jia, T, Johnson, KA, Jones, PB, Jones, DT, Kahn, RS, Karlsson, H, Karlsson, L, Kawashima, R, Kelley, EA, Kern, S, Kim, KW, Kitzbichler, MG, Kremen, WS, Lalonde, F, Landeau, B, Lee, S, Lerch, J, Lewis, JD, Li, J, Liao, W, Liston, C, Lombardo, MV, Lv, J, Lynch, C, Mallard, TT, Marcelis, M, Markello, RD, Mathias, SR, Mazoyer, B, McGuire, P, Meaney, MJ, Mechelli, A, Medic, N, Misic, B, Morgan, SE, Mothersill, D, Nigg, J, Ong, MQW, Ortinau, C, Ossenkoppele, R, Ouyang, M, Palaniyappan, L, Paly, L, Pan, PM, Pantelis, C, Park, MM, Paus, T, Pausova, Z, Paz-Linares, D, Pichet Binette, A, Pierce, K, Qian, X, Qiu, J, Qiu, A, Raznahan, A, Rittman, T, Rodrigue, A, Rollins, CK, Romero-Garcia, R, Ronan, L, Rosenberg, MD, Rowitch, DH, Salum, GA, Satterthwaite, TD, Schaare, HL, Schachar, RJ, Schultz, AP, Schumann, G, Scholl, M, Sharp, D, Shinohara, RT, Skoog, I, Smyser, CD, Sperling, RA, Stein, DJ, Stolicyn, A, Suckling, J, Sullivan, G, Taki, Y, Thyreau, B, Toro, R, Traut, N, Tsvetanov, KA, Turk-Browne, NB, Tuulari, JJ, Tzourio, C, Vachon-Presseau, E, Valdes-Sosa, MJ, Valdes-Sosa, PA, Valk, SL, van Amelsvoort, T, Vandekar, SN, Vasung, L, Victoria, LW, Villeneuve, S, Villringer, A, Vertes, PE, Wagstyl, K, Wang, YS, Warfield, SK, Warrier, V, Westman, E, Westwater, ML, Whalley, HC, Witte, AV, Yang, N, Yeo, B, Yun, H, Zalesky, A, Zar, HJ, Zettergren, A, Zhou, JH, Ziauddeen, H, Zugman, A, Zuo, XN, Bullmore, ET, Alexander-Bloch, AF, Bethlehem, RAI, Seidlitz, J, White, SR, Vogel, JW, Anderson, KM, Adamson, C, Adler, S, Alexopoulos, GS, Anagnostou, E, Areces-Gonzalez, A, Astle, DE, Auyeung, B, Ayub, M, Bae, J, Ball, G, Baron-Cohen, S, Beare, R, Bedford, SA, Benegal, V, Beyer, F, Blangero, J, Blesa Cabez, M, Boardman, JP, Borzage, M, Bosch-Bayard, JF, Bourke, N, Calhoun, VD, Chakravarty, MM, Chen, C, Chertavian, C, Chetelat, G, Chong, YS, Cole, JH, Corvin, A, Costantino, M, Courchesne, E, Crivello, F, Cropley, VL, Crosbie, J, Crossley, N, Delarue, M, Delorme, R, Desrivieres, S, Devenyi, GA, Di Biase, MA, Dolan, R, Donald, KA, Donohoe, G, Dunlop, K, Edwards, AD, Elison, JT, Ellis, CT, Elman, JA, Eyler, L, Fair, DA, Feczko, E, Fletcher, PC, Fonagy, P, Franz, CE, Galan-Garcia, L, Gholipour, A, Giedd, J, Gilmore, JH, Glahn, DC, Goodyer, IM, Grant, PE, Groenewold, NA, Gunning, FM, Gur, RE, Gur, RC, Hammill, CF, Hansson, O, Hedden, T, Heinz, A, Henson, RN, Heuer, K, Hoare, J, Holla, B, Holmes, AJ, Holt, R, Huang, H, Im, K, Ipser, J, Jack, CR, Jackowski, AP, Jia, T, Johnson, KA, Jones, PB, Jones, DT, Kahn, RS, Karlsson, H, Karlsson, L, Kawashima, R, Kelley, EA, Kern, S, Kim, KW, Kitzbichler, MG, Kremen, WS, Lalonde, F, Landeau, B, Lee, S, Lerch, J, Lewis, JD, Li, J, Liao, W, Liston, C, Lombardo, MV, Lv, J, Lynch, C, Mallard, TT, Marcelis, M, Markello, RD, Mathias, SR, Mazoyer, B, McGuire, P, Meaney, MJ, Mechelli, A, Medic, N, Misic, B, Morgan, SE, Mothersill, D, Nigg, J, Ong, MQW, Ortinau, C, Ossenkoppele, R, Ouyang, M, Palaniyappan, L, Paly, L, Pan, PM, Pantelis, C, Park, MM, Paus, T, Pausova, Z, Paz-Linares, D, Pichet Binette, A, Pierce, K, Qian, X, Qiu, J, Qiu, A, Raznahan, A, Rittman, T, Rodrigue, A, Rollins, CK, Romero-Garcia, R, Ronan, L, Rosenberg, MD, Rowitch, DH, Salum, GA, Satterthwaite, TD, Schaare, HL, Schachar, RJ, Schultz, AP, Schumann, G, Scholl, M, Sharp, D, Shinohara, RT, Skoog, I, Smyser, CD, Sperling, RA, Stein, DJ, Stolicyn, A, Suckling, J, Sullivan, G, Taki, Y, Thyreau, B, Toro, R, Traut, N, Tsvetanov, KA, Turk-Browne, NB, Tuulari, JJ, Tzourio, C, Vachon-Presseau, E, Valdes-Sosa, MJ, Valdes-Sosa, PA, Valk, SL, van Amelsvoort, T, Vandekar, SN, Vasung, L, Victoria, LW, Villeneuve, S, Villringer, A, Vertes, PE, Wagstyl, K, Wang, YS, Warfield, SK, Warrier, V, Westman, E, Westwater, ML, Whalley, HC, Witte, AV, Yang, N, Yeo, B, Yun, H, Zalesky, A, Zar, HJ, Zettergren, A, Zhou, JH, Ziauddeen, H, Zugman, A, Zuo, XN, Bullmore, ET, and Alexander-Bloch, AF
- Abstract
Over the past few decades, neuroimaging has become a ubiquitous tool in basic research and clinical studies of the human brain. However, no reference standards currently exist to quantify individual differences in neuroimaging metrics over time, in contrast to growth charts for anthropometric traits such as height and weight1. Here we assemble an interactive open resource to benchmark brain morphology derived from any current or future sample of MRI data ( http://www.brainchart.io/ ). With the goal of basing these reference charts on the largest and most inclusive dataset available, acknowledging limitations due to known biases of MRI studies relative to the diversity of the global population, we aggregated 123,984 MRI scans, across more than 100 primary studies, from 101,457 human participants between 115 days post-conception to 100 years of age. MRI metrics were quantified by centile scores, relative to non-linear trajectories2 of brain structural changes, and rates of change, over the lifespan. Brain charts identified previously unreported neurodevelopmental milestones3, showed high stability of individuals across longitudinal assessments, and demonstrated robustness to technical and methodological differences between primary studies. Centile scores showed increased heritability compared with non-centiled MRI phenotypes, and provided a standardized measure of atypical brain structure that revealed patterns of neuroanatomical variation across neurological and psychiatric disorders. In summary, brain charts are an essential step towards robust quantification of individual variation benchmarked to normative trajectories in multiple, commonly used neuroimaging phenotypes.
- Published
- 2022
6. Brain charts for the human lifespan (vol 604, pg 525, 2022)
- Author
-
Bethlehem, RAI, Seidlitz, J, White, SR, Vogel, JW, Anderson, KM, Adamson, C, Adler, S, Alexopoulos, GS, Anagnostou, E, Areces-Gonzalez, A, Astle, DE, Auyeung, B, Ayub, M, Bae, J, Ball, G, Baron-Cohen, S, Beare, R, Bedford, SA, Benegal, V, Beyer, F, Blangero, J, Blesa Cabez, M, Boardman, JP, Borzage, M, Bosch-Bayard, JF, Bourke, N, Calhoun, VD, Chakravarty, MM, Chen, C, Chertavian, C, Chetelat, G, Chong, YS, Cole, JH, Corvin, A, Costantino, M, Courchesne, E, Crivello, F, Cropley, VL, Crosbie, J, Crossley, N, Delarue, M, Delorme, R, Desrivieres, S, Devenyi, GA, Di Biase, MA, Dolan, R, Donald, KA, Donohoe, G, Dunlop, K, Edwards, AD, Elison, JT, Ellis, CT, Elman, JA, Eyler, L, Fair, DA, Feczko, E, Fletcher, PC, Fonagy, P, Franz, CE, Galan-Garcia, L, Gholipour, A, Giedd, J, Gilmore, JH, Glahn, DC, Goodyer, IM, Grant, PE, Groenewold, NA, Gunning, FM, Gur, RE, Gur, RC, Hammill, CF, Hansson, O, Hedden, T, Heinz, A, Henson, RN, Heuer, K, Hoare, J, Holla, B, Holmes, AJ, Holt, R, Huang, H, Im, K, Ipser, J, Jack, CR, Jackowski, AP, Jia, T, Johnson, KA, Jones, PB, Jones, DT, Kahn, RS, Karlsson, H, Karlsson, L, Kawashima, R, Kelley, EA, Kern, S, Kim, KW, Kitzbichler, MG, Kremen, WS, Lalonde, F, Landeau, B, Lee, S, Lerch, J, Lewis, JD, Li, J, Liao, W, Liston, C, Lombardo, MV, Lv, J, Lynch, C, Mallard, TT, Marcelis, M, Markello, RD, Mathias, SR, Mazoyer, B, McGuire, P, Meaney, MJ, Mechelli, A, Medic, N, Misic, B, Morgan, SE, Mothersill, D, Nigg, J, Ong, MQW, Ortinau, C, Ossenkoppele, R, Ouyang, M, Palaniyappan, L, Paly, L, Pan, PM, Pantelis, C, Park, MM, Paus, T, Pausova, Z, Paz-Linares, D, Pichet Binette, A, Pierce, K, Qian, X, Qiu, J, Qiu, A, Raznahan, A, Rittman, T, Rodrigue, A, Rollins, CK, Romero-Garcia, R, Ronan, L, Rosenberg, MD, Rowitch, DH, Salum, GA, Satterthwaite, TD, Schaare, HL, Schachar, RJ, Schultz, AP, Schumann, G, Scholl, M, Sharp, D, Shinohara, RT, Skoog, I, Smyser, CD, Sperling, RA, Stein, DJ, Stolicyn, A, Suckling, J, Sullivan, G, Taki, Y, Thyreau, B, Toro, R, Traut, N, Tsvetanov, KA, Turk-Browne, NB, Tuulari, JJ, Tzourio, C, Vachon-Presseau, E, Valdes-Sosa, MJ, Valdes-Sosa, PA, Valk, SL, van Amelsvoort, T, Vandekar, SN, Vasung, L, Victoria, LW, Villeneuve, S, Villringer, A, Vertes, PE, Wagstyl, K, Wang, YS, Warfield, SK, Warrier, V, Westman, E, Westwater, ML, Whalley, HC, Witte, AV, Yang, N, Yeo, B, Yun, H, Zalesky, A, Zar, HJ, Zettergren, A, Zhou, JH, Ziauddeen, H, Zugman, A, Zuo, XN, Bullmore, ET, Alexander-Bloch, AF, Bethlehem, RAI, Seidlitz, J, White, SR, Vogel, JW, Anderson, KM, Adamson, C, Adler, S, Alexopoulos, GS, Anagnostou, E, Areces-Gonzalez, A, Astle, DE, Auyeung, B, Ayub, M, Bae, J, Ball, G, Baron-Cohen, S, Beare, R, Bedford, SA, Benegal, V, Beyer, F, Blangero, J, Blesa Cabez, M, Boardman, JP, Borzage, M, Bosch-Bayard, JF, Bourke, N, Calhoun, VD, Chakravarty, MM, Chen, C, Chertavian, C, Chetelat, G, Chong, YS, Cole, JH, Corvin, A, Costantino, M, Courchesne, E, Crivello, F, Cropley, VL, Crosbie, J, Crossley, N, Delarue, M, Delorme, R, Desrivieres, S, Devenyi, GA, Di Biase, MA, Dolan, R, Donald, KA, Donohoe, G, Dunlop, K, Edwards, AD, Elison, JT, Ellis, CT, Elman, JA, Eyler, L, Fair, DA, Feczko, E, Fletcher, PC, Fonagy, P, Franz, CE, Galan-Garcia, L, Gholipour, A, Giedd, J, Gilmore, JH, Glahn, DC, Goodyer, IM, Grant, PE, Groenewold, NA, Gunning, FM, Gur, RE, Gur, RC, Hammill, CF, Hansson, O, Hedden, T, Heinz, A, Henson, RN, Heuer, K, Hoare, J, Holla, B, Holmes, AJ, Holt, R, Huang, H, Im, K, Ipser, J, Jack, CR, Jackowski, AP, Jia, T, Johnson, KA, Jones, PB, Jones, DT, Kahn, RS, Karlsson, H, Karlsson, L, Kawashima, R, Kelley, EA, Kern, S, Kim, KW, Kitzbichler, MG, Kremen, WS, Lalonde, F, Landeau, B, Lee, S, Lerch, J, Lewis, JD, Li, J, Liao, W, Liston, C, Lombardo, MV, Lv, J, Lynch, C, Mallard, TT, Marcelis, M, Markello, RD, Mathias, SR, Mazoyer, B, McGuire, P, Meaney, MJ, Mechelli, A, Medic, N, Misic, B, Morgan, SE, Mothersill, D, Nigg, J, Ong, MQW, Ortinau, C, Ossenkoppele, R, Ouyang, M, Palaniyappan, L, Paly, L, Pan, PM, Pantelis, C, Park, MM, Paus, T, Pausova, Z, Paz-Linares, D, Pichet Binette, A, Pierce, K, Qian, X, Qiu, J, Qiu, A, Raznahan, A, Rittman, T, Rodrigue, A, Rollins, CK, Romero-Garcia, R, Ronan, L, Rosenberg, MD, Rowitch, DH, Salum, GA, Satterthwaite, TD, Schaare, HL, Schachar, RJ, Schultz, AP, Schumann, G, Scholl, M, Sharp, D, Shinohara, RT, Skoog, I, Smyser, CD, Sperling, RA, Stein, DJ, Stolicyn, A, Suckling, J, Sullivan, G, Taki, Y, Thyreau, B, Toro, R, Traut, N, Tsvetanov, KA, Turk-Browne, NB, Tuulari, JJ, Tzourio, C, Vachon-Presseau, E, Valdes-Sosa, MJ, Valdes-Sosa, PA, Valk, SL, van Amelsvoort, T, Vandekar, SN, Vasung, L, Victoria, LW, Villeneuve, S, Villringer, A, Vertes, PE, Wagstyl, K, Wang, YS, Warfield, SK, Warrier, V, Westman, E, Westwater, ML, Whalley, HC, Witte, AV, Yang, N, Yeo, B, Yun, H, Zalesky, A, Zar, HJ, Zettergren, A, Zhou, JH, Ziauddeen, H, Zugman, A, Zuo, XN, Bullmore, ET, and Alexander-Bloch, AF
- Published
- 2022
7. Single-cell in situ transcriptomic map of astrocyte cortical layer diversity
- Author
-
Bayraktar, OA, Bartels, T, Holmqvist, S, Kleshchevnikov, V, Martirosyan, A, Polioudakis, D, Ben Haim, L, Young, AMH, Batiuk, M, Prakash, K, Brown, A, Roberts, K, Paredes, MF, Kawaguchi, R, Stockley, J, Sabeur, K, Chang, SM, Huang, E, Hutchinson, P, Ullian, EM, Hemberg, M, Geschwind, DH, Coppola, G, Rowitch, DH, and Holt, Matthew
- Subjects
Quantitative Biology::Neurons and Cognition - Abstract
ispartof: NATURE NEUROSCIENCE status: accepted
- Published
- 2019
8. Whole genome sequencing reveals that genetic conditions are frequent in intensively ill children
- Author
-
French, CE, Delon, I, Dolling, H, Sanchis-Juan, A, Shamardina, O, Mégy, K, Abbs, S, Austin, T, Bowdin, S, Branco, RG, Firth, H, Tuna, S, Aitman, TJ, Ashford, S, Astle, WJ, Bennet, DL, Bleda, M, Carss, KJ, Chinnery, PF, Deevi, SVV, Fletcher, D, Gale, DP, Gräf, SF, Hu, F, James, R, Kasanicki, MA, Kingston, N, Koziell, AB, Allen, HL, Maher, ER, Markus, HS, Meacham, S, Morrell, NW, Penkett, CJ, Roberts, I, Smith, KGC, Stark, H, Stirrups, KE, Turro, E, Watkins, H, Williamson, C, Young, T, Bradley, JR, Ouwehand, WH, Raymond, FL, Agrawal, S, Armstrong, R, Beardsall, K, Belteki, G, Bohatschek, M, Broster, S, Campbell, R, Chaudhary, R, Costa, C, D’Amore, A, Fitzsimmons, A, Hague, J, Harley, J, Hoodbhoy, S, Kayani, R, Kelsall, W, Mehta, SG, O’Donnell, R, O’Hare, S, Ogilvy-Stuart, A, Papakostas, S, Park, SM, Parker, A, Pathan, N, Prapa, M, Sammut, A, Sandford, R, Schon, K, Singh, Y, Spike, K, Tavares, ALT, Wari-Pepple, D, Wong, HS, Woods, CG, Rowitch, DH, Raymond, F Lucy [0000-0003-2652-3355], and Apollo - University of Cambridge Repository
- Subjects
Male ,NICU ,medicine.medical_specialty ,Palliative care ,Adolescent ,PICU ,Original ,Critical Illness ,Critically ill children ,Genomics ,Critical Care and Intensive Care Medicine ,Intensive Care Units, Pediatric ,State Medicine ,Cohort Studies ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Anesthesiology ,Intensive care ,Intensive Care Units, Neonatal ,Genetics ,Medicine ,Humans ,Prospective Studies ,Medical diagnosis ,Intensive care medicine ,Prospective cohort study ,Child ,Whole genome sequencing ,Whole Genome Sequencing ,business.industry ,Genetic Diseases, Inborn ,Infant, Newborn ,Infant ,030208 emergency & critical care medicine ,030228 respiratory system ,England ,FOS: Biological sciences ,Child, Preschool ,Female ,business ,Genetic Background ,Cohort study - Abstract
Purpose With growing evidence that rare single gene disorders present in the neonatal period, there is a need for rapid, systematic, and comprehensive genomic diagnoses in ICUs to assist acute and long-term clinical decisions. This study aimed to identify genetic conditions in neonatal (NICU) and paediatric (PICU) intensive care populations. Methods We performed trio whole genome sequence (WGS) analysis on a prospective cohort of families recruited in NICU and PICU at a single site in the UK. We developed a research pipeline in collaboration with the National Health Service to deliver validated pertinent pathogenic findings within 2–3 weeks of recruitment. Results A total of 195 families had whole genome analysis performed (567 samples) and 21% received a molecular diagnosis for the underlying genetic condition in the child. The phenotypic description of the child was a poor predictor of the gene identified in 90% of cases, arguing for gene agnostic testing in NICU/PICU. The diagnosis affected clinical management in more than 65% of cases (83% in neonates) including modification of treatments and care pathways and/or informing palliative care decisions. A 2–3 week turnaround was sufficient to impact most clinical decision-making. Conclusions The use of WGS in intensively ill children is acceptable and trio analysis facilitates diagnoses. A gene agnostic approach was effective in identifying an underlying genetic condition, with phenotypes and symptomatology being primarily used for data interpretation rather than gene selection. WGS analysis has the potential to be a first-line diagnostic tool for a subset of intensively ill children. Electronic supplementary material The online version of this article (10.1007/s00134-019-05552-x) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
9. Kir4.1-Dependent Astrocyte-Fast Motor Neuron Interactions Are Required for Peak Strength
- Author
-
Kelley, Kevin W, Ben Haim, Lucile, Schirmer, Lucas, Tyzack, Giulia, Tolman, Michaela, Miller, John, Tsai, Hui-Hsin, Chang, Sandra M, Molofsky, Anna V, Yang, Yongjie, Patani, Rickie, Lakatos, Andras, Ullian, Erik M, Rowitch, DH, Lakatos, Andras [0000-0002-1301-2292], Rowitch, David [0000-0002-0079-0060], and Apollo - University of Cambridge Repository
- Subjects
Male ,amyotrophic lateral sclerosis ,Potassium Channels ,Kir4.1 ,Cells ,Induced Pluripotent Stem Cells ,Mice, Transgenic ,Neurodegenerative ,Transgenic ,Article ,Mice ,Rare Diseases ,Organ Culture Techniques ,Psychology ,astrocyte diversity ,Animals ,Humans ,Potassium Channels, Inwardly Rectifying ,motor neuron ,Cells, Cultured ,Motor Neurons ,Cultured ,Neurology & Neurosurgery ,Neurosciences ,neurodegeneration ,Newborn ,peak strength ,Brain Disorders ,Inwardly Rectifying ,Animals, Newborn ,Astrocytes ,Neurological ,fast-twitch muscle ,Cognitive Sciences ,Female ,ALS ,spinal cord development - Abstract
Summary Diversified neurons are essential for sensorimotor function, but whether astrocytes become specialized to optimize circuit performance remains unclear. Large fast α-motor neurons (FαMNs) of spinal cord innervate fast-twitch muscles that generate peak strength. We report that ventral horn astrocytes express the inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) around MNs in a VGLUT1-dependent manner. Loss of astrocyte-encoded Kir4.1 selectively altered FαMN size and function and led to reduced peak strength. Overexpression of Kir4.1 in astrocytes was sufficient to increase MN size through activation of the PI3K/mTOR/pS6 pathway. Kir4.1 was downregulated cell autonomously in astrocytes derived from amyotrophic lateral sclerosis (ALS) patients with SOD1 mutation. However, astrocyte Kir4.1 was dispensable for FαMN survival even in the mutant SOD1 background. These findings show that astrocyte Kir4.1 is essential for maintenance of peak strength and suggest that Kir4.1 downregulation might uncouple symptoms of muscle weakness from MN cell death in diseases like ALS., Highlights • Kir4.1 is upregulated in astrocytes around high-activity alpha motor neurons (MNs) • Astrocyte Kir4.1 KO caused decreased peak strength without alpha MN loss • ALS patient-derived astrocytes show cell-autonomous Kir4.1 downregulation • Astrocyte Kir4.1 regulates MN size through PI3K/mTOR/pS6 activation, Kelley et al. show that specialized astrocytes surrounding spinal cord fast α-motor neurons are critical to generate peak strength and that they are compromised by mutations in models of amyotrophic lateral sclerosis.
- Published
- 2018
10. Corridors of migrating neurons in the human brain and their decline during infancy
- Author
-
Sanai, N, Nguyen, T, Ihrie, RA, Mirzadeh, Z, Tsai, HH, Wong, M, Gupta, N, Berger, MS, Huang, E, Garcia-Verdugo, JM, Rowitch, DH, and Alvarez-Buylla, A
- Subjects
Adult ,Adolescent ,General Science & Technology ,1.1 Normal biological development and functioning ,Cell Movement ,Underpinning research ,80 and over ,Humans ,Child ,Preschool ,reproductive and urinary physiology ,Cell Proliferation ,Aged ,Neurons ,Pediatric ,Neurosciences ,Brain ,Infant ,Olfactory Pathways ,Middle Aged ,Newborn ,Stem Cell Research ,Brain Disorders ,nervous system ,Neurological ,Stem Cell Research - Nonembryonic - Non-Human - Abstract
The subventricular zone of many adult non-human mammals generates large numbers of new neurons destined for the olfactory bulb. Along the walls of the lateral ventricles, immature neuronal progeny migrate in tangentially oriented chains that coalesce into a rostral migratory stream (RMS) connecting the subventricular zone to the olfactory bulb. The adult human subventricular zone, in contrast, contains a hypocellular gap layer separating the ependymal lining from a periventricular ribbon of astrocytes. Some of these subventricular zone astrocytes can function as neural stem cells in vitro, but their function in vivo remains controversial. An initial report found few subventricular zone proliferating cells and rare migrating immature neurons in the RMS of adult humans. In contrast, a subsequent study indicated robust proliferation and migration in the human subventricular zone and RMS. Here we find that the infant human subventricular zone and RMS contain an extensive corridor of migrating immature neurons before 18 months of age but, contrary to previous reports, this germinal activity subsides in older children and is nearly extinct by adulthood. Surprisingly, during this limited window of neurogenesis, not all new neurons in the human subventricular zone are destined for the olfactory bulb-we describe a major migratory pathway that targets the prefrontal cortex in humans. Together, these findings reveal robust streams of tangentially migrating immature neurons in human early postnatal subventricular zone and cortex. These pathways represent potential targets of neurological injuries affecting neonates. © 2011 Macmillan Publishers Limited. All rights reserved.
- Published
- 2011
- Full Text
- View/download PDF
11. Separated at birth? The functional and molecular divergence of OLIG1 and OLIG2.
- Author
-
Meijer DH, Kane MF, Mehta S, Liu H, Harrington E, Taylor CM, Stiles CD, Rowitch DH, Meijer, Dimphna H, Kane, Michael F, Mehta, Shwetal, Liu, Hongye, Harrington, Emily, Taylor, Christopher M, Stiles, Charles D, and Rowitch, David H
- Abstract
The basic helix-loop-helix transcription factors oligodendrocyte transcription factor 1 (OLIG1) and OLIG2 are structurally similar and, to a first approximation, coordinately expressed in the developing CNS and postnatal brain. Despite these similarities, it was apparent from early on after their discovery that OLIG1 and OLIG2 have non-overlapping developmental functions in patterning, neuron subtype specification and the formation of oligodendrocytes. Here, we summarize more recent insights into the separate roles of these transcription factors in the postnatal brain during repair processes and in neurological disease states, including multiple sclerosis and malignant glioma. We discuss how the unique functions of OLIG1 and OLIG2 may reflect their distinct genetic targets, co-regulator proteins and/or post-translational modifications. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
12. Heparan sulfate sulfatase SULF2 regulates PDGFRα signaling and growth in human and mouse malignant glioma.
- Author
-
Phillips JJ, Huillard E, Robinson AE, Ward A, Lum DH, Polley MY, Rosen SD, Rowitch DH, Werb Z, Phillips, Joanna J, Huillard, Emmanuelle, Robinson, Aaron E, Ward, Anna, Lum, David H, Polley, Mei-Yin, Rosen, Steven D, Rowitch, David H, and Werb, Zena
- Abstract
Glioblastoma (GBM), a uniformly lethal brain cancer, is characterized by diffuse invasion and abnormal activation of multiple receptor tyrosine kinase (RTK) signaling pathways, presenting a major challenge to effective therapy. The activation of many RTK pathways is regulated by extracellular heparan sulfate proteoglycans (HSPG), suggesting these molecules may be effective targets in the tumor microenvironment. In this study, we demonstrated that the extracellular sulfatase, SULF2, an enzyme that regulates multiple HSPG-dependent RTK signaling pathways, was expressed in primary human GBM tumors and cell lines. Knockdown of SULF2 in human GBM cell lines and generation of gliomas from Sulf2(-/-) tumorigenic neurospheres resulted in decreased growth in vivo in mice. We found a striking SULF2 dependence in activity of PDGFRα, a major signaling pathway in GBM. Ablation of SULF2 resulted in decreased PDGFRα phosphorylation and decreased downstream MAPK signaling activity. Interestingly, in a survey of SULF2 levels in different subtypes of GBM, the proneural subtype, characterized by aberrations in PDGFRα, demonstrated the strongest SULF2 expression. Therefore, in addition to its potential as an upstream target for therapy of GBM, SULF2 may help identify a subset of GBMs that are more dependent on exogenous growth factor-mediated signaling. Our results suggest the bioavailability of growth factors from the microenvironment is a significant contributor to tumor growth in a major subset of human GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
13. Epidermal growth factor receptor and Ink4a/Arf: Convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis.
- Author
-
Bachoo RM, Maher EA, Ligon KL, Sharpless NE, Chan SS, You MJ, Tang Y, DeFrances J, Stover E, Weissleder R, Rowitch DH, Louis DN, and DePinho RA
- Published
- 2024
- Full Text
- View/download PDF
14. Oligodendrocyte Slc48a1 (Hrg1) encodes a functional heme transporter required for myelin integrity.
- Author
-
Stockley JH, Vaquie AM, Xu Z, Bartels T, Jordan GD, Holmqvist S, Gunter S, Lam G, Yamamoto D, Pek RH, Chambers IG, Rock AS, Hill M, Zhao C, Dillon S, Franklin RJM, O'Connor R, Bodine DM, Hamza I, and Rowitch DH
- Abstract
Oligodendrocytes (OLs) of the central nervous system require iron for proteolipid biosynthesis during the myelination process. Although most heme is found complexed to hemoglobin in red blood cells, surprisingly, we found that Slc48a1, encoding the heme transporter Hrg1, is expressed at higher levels in OLs than any other cell type in rodent and humans. We confirmed in situ that Hrg1 is expressed in OLs but not their precursors (OPCs) and found that Hrg1 proteins in CNS white matter co-localized within myelin sheaths. In older Hrg1 null mutant mice we observed reduced expression of myelin associated glycoprotein (Mag) and ultrastructural myelin defects reminiscent of Mag-null animals, suggesting myelin adhesion deficiency. Further, we confirmed reduced myelin iron levels in Hrg1 null animals in vivo, and show that OLs in vitro can directly import both the fluorescent heme analogue ZnMP and heme itself, which rescued iron deficiency induced inhibition of OL differentiation in a heme-oxidase-dependent manner. Together these findings indicate OL Hrg1 encodes a functional heme transporter required for myelin integrity., (© 2024 The Author(s). GLIA published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
15. Identifying barriers and opportunities to facilitate the uptake of whole genome sequencing in paediatric haematology and oncology practice.
- Author
-
Bishop M, Vedi A, Bowdin S, Armstrong R, Bartram J, Bentley D, Ross M, Hook CE, Yin Chung BH, Moss P, Rowitch DH, Tarpey P, Behjati S, and Murray MJ
- Subjects
- Humans, Child, Pediatrics, United Kingdom, Hematology, Neoplasms diagnosis, Neoplasms genetics, England, State Medicine, Ireland, Whole Genome Sequencing, Medical Oncology
- Abstract
Background: The clinical utility of whole genome sequencing (WGS) in paediatric cancer has been demonstrated in recent years. WGS has been routinely available in the National Health Service (NHS) England for all children with cancer in England since 2021, but its uptake has been variable geographically. To explore the underlying barriers to routine use of WGS in this population across England and more widely in the United Kingdom (UK) and the Republic of Ireland (ROI), a one-day workshop was held in Cambridge, United Kingdom in October 2022., Methods: Following a series of talks, delegates participated in open, round-table discussions to outline local and broader challenges limiting routine WGS for diagnostic work-up for children with cancer in their Principal Treatment Centres (PTCs) and Genomic Laboratory Hubs (GLHs). Within smaller groups, delegates answered structured questions regarding clinical capability, education and training needs, and workforce competence and requirements. Data was recorded, centrally collated, and analysed following the event using thematic analysis., Results: Sixty participants attended the workshop with broad representation from the 20 PTCs across the UK and ROI and the seven GLHs in England. All healthcare professionals involved in the WGS pathway were represented, including paediatric oncologists, clinical geneticists, clinical scientists, and histopathologists. The main themes highlighted by the group in ensuring equitable access to WGS identified were: lack of knowledge equity between NHS trusts, with a perception of WGS being for research only; and perception of lack of financial support for the clinical process surrounding WGS, including lack of time to take informed consent from patients. The latter also included limited trained staff available for data interpretation, affecting the turnaround time for reporting. Finally, the need for an integrated digital pathway to order, track, and return data to clinicians was highlighted., Conclusion: At the workshop, the general motivation for including WGS in the diagnostic work up for children with cancer was high throughout the UK, however a perceived lack of resources and education opportunities limit the widespread use of this commissioned assay. This workshop has led to some recommendations to increase access to WGS in this population in England and more widely in the devolved national of the UK and the ROI., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Generation of Mammalian Astrocyte Functional Heterogeneity.
- Author
-
Bartels T, Rowitch DH, and Bayraktar OA
- Subjects
- Animals, Brain physiology, Brain embryology, Mammals, Humans, Cell Differentiation, Cell Lineage, Astrocytes physiology
- Abstract
Mammalian astrocytes have regional roles within the brain parenchyma. Indeed, the notion that astrocytes are molecularly heterogeneous could help explain how the central nervous system (CNS) retains embryonic positional information through development into specialized regions into adulthood. A growing body of evidence supports the concept of morphological and molecular differences between astrocytes in different brain regions, which might relate to their derivation from regionally patterned radial glia and/or local neuron inductive cues. Here, we review evidence for regionally encoded functions of astrocytes to provide an integrated concept on lineage origins and heterogeneity to understand regional brain organization, as well as emerging technologies to identify and further investigate novel roles for astrocytes., (Copyright © 2024 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
17. Novel human iPSC models of neuroinflammation in neurodegenerative disease and regenerative medicine.
- Author
-
Summers RA, Fagiani F, Rowitch DH, Absinta M, and Reich DS
- Subjects
- Humans, Cell Differentiation, Neuroinflammatory Diseases immunology, Neuroinflammatory Diseases therapy, Animals, Blood-Brain Barrier immunology, Organoids immunology, Induced Pluripotent Stem Cells immunology, Neurodegenerative Diseases therapy, Neurodegenerative Diseases immunology, Regenerative Medicine
- Abstract
The importance of neuroinflammation in neurodegenerative diseases is becoming increasingly evident, and, in parallel, human induced pluripotent stem cell (hiPSC) models of physiology and pathology are emerging. Here, we review new advancements in the differentiation of hiPSCs into glial, neural, and blood-brain barrier (BBB) cell types, and the integration of these cells into complex organoids and chimeras. These advancements are relevant for modeling neuroinflammation in the context of prevalent neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). With awareness of current limitations, recent progress in the development and application of various hiPSC-derived models shows potential for aiding the identification of candidate therapeutic targets and immunotherapy approaches., Competing Interests: Declaration of interests M.A. has received consultancy and/or speaker honoraria from Abata Therapeutics, Biogen, Sanofi, Immunic Therapeutics, and GSK. D.S.R. has received research funding from Abata Therapeutics and Sanofi. The other authors have nothing to disclose., (Published by Elsevier Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
18. Neuroinflammatory reactive astrocyte formation correlates with adverse outcomes in perinatal white matter injury.
- Author
-
Renz P, Steinfort M, Haesler V, Tscherrig V, Huang EJ, Chavali M, Liddelow S, Rowitch DH, Surbek D, Schoeberlein A, and Brosius Lutz A
- Subjects
- Animals, Mice, Humans, Neuroinflammatory Diseases pathology, Neuroinflammatory Diseases metabolism, Mice, Inbred C57BL, Female, Disease Models, Animal, Mice, Knockout, Infant, Newborn, Astrocytes metabolism, Astrocytes pathology, White Matter pathology, White Matter metabolism, Animals, Newborn
- Abstract
Perinatal white matter injury (WMI) is the leading cause of long-term neurological morbidity in infants born preterm. Neuroinflammation during a critical window of early brain development plays a key role in WMI disease pathogenesis. The mechanisms linking inflammation with the long-term myelination failure that characterizes WMI, however, remain unknown. Here, we investigate the role of astrocyte reactivity in WMI. In an experimental mouse model of WMI, we demonstrate that WMI disease outcomes are improved in mutant mice lacking secretion of inflammatory molecules TNF-α, IL-1α, and C1q known, in addition to other roles, to induce the formation of a neuroinflammatory reactive astrocyte substate. We show that astrocytes express molecular signatures of the neuroinflammatory reactive astrocyte substate in both our WMI mouse model and human tissue affected by WMI, and that this gene expression pattern is dampened in injured mutant mice. Our data provide evidence that a neuroinflammatory reactive astrocyte substate correlates with adverse WMI disease outcomes, thus highlighting the need for further investigation of these cells as potential causal players in WMI pathology., (© 2024 The Author(s). GLIA published by Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
19. Developmental origin of oligodendrocytes determines their function in the adult brain.
- Author
-
Foerster S, Floriddia EM, van Bruggen D, Kukanja P, Hervé B, Cheng S, Kim E, Phillips BU, Heath CJ, Tripathi RB, Call C, Bartels T, Ridley K, Neumann B, López-Cruz L, Crawford AH, Lynch CJ, Serrano M, Saksida L, Rowitch DH, Möbius W, Nave KA, Rasband MN, Bergles DE, Kessaris N, Richardson WD, Bussey TJ, Zhao C, Castelo-Branco G, and Franklin RJM
- Subjects
- Animals, Mice, Cell Differentiation physiology, Mice, Transgenic, Myelin Sheath metabolism, Myelin Sheath physiology, Oligodendroglia physiology, Brain cytology, Brain embryology, Cell Lineage physiology
- Abstract
In the mouse embryonic forebrain, developmentally distinct oligodendrocyte progenitor cell populations and their progeny, oligodendrocytes, emerge from three distinct regions in a spatiotemporal gradient from ventral to dorsal. However, the functional importance of this oligodendrocyte developmental heterogeneity is unknown. Using a genetic strategy to ablate dorsally derived oligodendrocyte lineage cells (OLCs), we show here that the areas in which dorsally derived OLCs normally reside in the adult central nervous system become populated and myelinated by OLCs of ventral origin. These ectopic oligodendrocytes (eOLs) have a distinctive gene expression profile as well as subtle myelination abnormalities. The failure of eOLs to fully assume the role of the original dorsally derived cells results in locomotor and cognitive deficits in the adult animal. This study reveals the importance of developmental heterogeneity within the oligodendrocyte lineage and its importance for homeostatic brain function., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
20. A retroviral link to vertebrate myelination through retrotransposon-RNA-mediated control of myelin gene expression.
- Author
-
Ghosh T, Almeida RG, Zhao C, Mannioui A, Martin E, Fleet A, Chen CZ, Assinck P, Ellams S, Gonzalez GA, Graham SC, Rowitch DH, Stott K, Adams I, Zalc B, Goldman N, Lyons DA, and Franklin RJM
- Subjects
- Animals, Gene Expression, Oligodendroglia metabolism, RNA metabolism, Zebrafish genetics, Anura, Myelin Sheath metabolism, Retroelements genetics
- Abstract
Myelin, the insulating sheath that surrounds neuronal axons, is produced by oligodendrocytes in the central nervous system (CNS). This evolutionary innovation, which first appears in jawed vertebrates, enabled rapid transmission of nerve impulses, more complex brains, and greater morphological diversity. Here, we report that RNA-level expression of RNLTR12-int, a retrotransposon of retroviral origin, is essential for myelination. We show that RNLTR12-int-encoded RNA binds to the transcription factor SOX10 to regulate transcription of myelin basic protein (Mbp, the major constituent of myelin) in rodents. RNLTR12-int-like sequences (which we name RetroMyelin) are found in all jawed vertebrates, and we further demonstrate their function in regulating myelination in two different vertebrate classes (zebrafish and frogs). Our study therefore suggests that retroviral endogenization played a prominent role in the emergence of vertebrate myelin., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Neuron-oligodendrocyte potassium shuttling at nodes of Ranvier protects against inflammatory demyelination.
- Author
-
Kapell H, Fazio L, Dyckow J, Schwarz S, Cruz-Herranz A, Mayer C, Campos J, D'Este E, Möbius W, Cordano C, Pröbstel AK, Gharagozloo M, Zulji A, Narayanan Naik V, Delank A, Cerina M, Müntefering T, Lerma-Martin C, Sonner JK, Sin JH, Disse P, Rychlik N, Sabeur K, Chavali M, Srivastava R, Heidenreich M, Fitzgerald KC, Seebohm G, Stadelmann C, Hemmer B, Platten M, Jentsch TJ, Engelhardt M, Budde T, Nave KA, Calabresi PA, Friese MA, Green AJ, Acuna C, Rowitch DH, Meuth SG, and Schirmer L
- Subjects
- Mice, Animals, Humans, Ranvier's Nodes metabolism, Potassium metabolism, Neurons metabolism, Oligodendroglia metabolism, Encephalomyelitis, Autoimmune, Experimental genetics, Encephalomyelitis, Autoimmune, Experimental metabolism, Multiple Sclerosis genetics, Multiple Sclerosis metabolism
- Abstract
Multiple sclerosis (MS) is a progressive inflammatory demyelinating disease of the CNS. Increasing evidence suggests that vulnerable neurons in MS exhibit fatal metabolic exhaustion over time, a phenomenon hypothesized to be caused by chronic hyperexcitability. Axonal Kv7 (outward-rectifying) and oligodendroglial Kir4.1 (inward-rectifying) potassium channels have important roles in regulating neuronal excitability at and around the nodes of Ranvier. Here, we studied the spatial and functional relationship between neuronal Kv7 and oligodendroglial Kir4.1 channels and assessed the transcriptional and functional signatures of cortical and retinal projection neurons under physiological and inflammatory demyelinating conditions. We found that both channels became dysregulated in MS and experimental autoimmune encephalomyelitis (EAE), with Kir4.1 channels being chronically downregulated and Kv7 channel subunits being transiently upregulated during inflammatory demyelination. Further, we observed that pharmacological Kv7 channel opening with retigabine reduced neuronal hyperexcitability in human and EAE neurons, improved clinical EAE signs, and rescued neuronal pathology in oligodendrocyte-Kir4.1-deficient (OL-Kir4.1-deficient) mice. In summary, our findings indicate that neuron-OL compensatory interactions promoted resilience through Kv7 and Kir4.1 channels and identify pharmacological activation of nodal Kv7 channels as a neuroprotective strategy against inflammatory demyelination.
- Published
- 2023
- Full Text
- View/download PDF
22. Clinical and Neurophysiologic Phenotypes in Neonates With BRAT1 Encephalopathy.
- Author
-
Carapancea E, Cornet MC, Milh M, De Cosmo L, Huang EJ, Granata T, Striano P, Ceulemans B, Stein A, Morris-Rosendahl D, Conti G, Mitra N, Raymond FL, Rowitch DH, Solazzi R, Vercellino F, De Liso P, D'Onofrio G, Boniver C, Danhaive O, Carkeek K, Salpietro V, Weckhuysen S, Fedrigo M, Angelini A, Castellotti B, Lederer D, Benoit V, Raviglione F, Guerrini R, Dilena R, and Cilio MR
- Subjects
- Humans, Apnea, Bradycardia, Seizures genetics, Phenotype, Muscle Hypertonia, Nuclear Proteins genetics, Myoclonus, Hyperekplexia, Brain Diseases diagnosis, Brain Diseases genetics
- Abstract
Background and Objectives: BRAT1 encephalopathy is an ultra-rare autosomal recessive neonatal encephalopathy. We delineate the neonatal electroclinical phenotype at presentation and provide insights for early diagnosis., Methods: Through a multinational collaborative, we studied a cohort of neonates with encephalopathy associated with biallelic pathogenic variants in BRAT1 for whom detailed clinical, neurophysiologic, and neuroimaging information was available from the onset of symptoms. Neuropathologic changes were also analyzed., Results: We included 19 neonates. Most neonates were born at term (16/19) from nonconsanguineous parents. 15/19 (79%) were admitted soon after birth to a neonatal intensive care unit, exhibiting multifocal myoclonus, both spontaneous and exacerbated by stimulation. 7/19 (37%) had arthrogryposis at birth, and all except 1 progressively developed hypertonia in the first week of life. Multifocal myoclonus, which was present in all but 1 infant, was the most prominent manifestation and did not show any EEG correlate in 16/19 (84%). Video-EEG at onset was unremarkable in 14/19 (74%) infants, and 6 (33%) had initially been misdiagnosed with hyperekplexia. Multifocal seizures were observed at a median age of 14 days (range: 1-29). During the first months of life, all infants developed progressive encephalopathy, acquired microcephaly, prolonged bouts of apnea, and bradycardia, leading to cardiac arrest and death at a median age of 3.5 months (range: 20 days to 30 months). Only 7 infants (37%) received a definite diagnosis before death, at a median age of 34 days (range: 25-126), and almost two-thirds (12/19, 63%) were diagnosed 8 days to 12 years postmortem (median: 6.5 years). Neuropathology examination, performed in 3 patients, revealed severely delayed myelination and diffuse astrogliosis, sparing the upper cortical layers., Discussion: BRAT1 encephalopathy is a neonatal-onset, rapidly progressive neurologic disorder. Neonates are often misdiagnosed as having hyperekplexia, and many die undiagnosed. The key phenotypic features are multifocal myoclonus, an organized EEG, progressive, persistent, and diffuse hypertonia, and an evolution into refractory multifocal seizures, prolonged bouts of apnea, bradycardia, and early death. Early recognition of BRAT1 encephalopathy allows for prompt workup, appropriate management, and genetic counseling., (© 2023 American Academy of Neurology.)
- Published
- 2023
- Full Text
- View/download PDF
23. Myeloid cell interferon secretion restricts Zika flavivirus infection of developing and malignant human neural progenitor cells.
- Author
-
Bulstrode H, Girdler GC, Gracia T, Aivazidis A, Moutsopoulos I, Young AMH, Hancock J, He X, Ridley K, Xu Z, Stockley JH, Finlay J, Hallou C, Fajardo T, Fountain DM, van Dongen S, Joannides A, Morris R, Mair R, Watts C, Santarius T, Price SJ, Hutchinson PJA, Hodson EJ, Pollard SM, Mohorianu I, Barker RA, Sweeney TR, Bayraktar O, Gergely F, and Rowitch DH
- Subjects
- Humans, Myeloid Cells, Stem Cells, Interferons, Zika Virus, Zika Virus Infection
- Abstract
Zika virus (ZIKV) can infect human developing brain (HDB) progenitors resulting in epidemic microcephaly, whereas analogous cellular tropism offers treatment potential for the adult brain cancer, glioblastoma (GBM). We compared productive ZIKV infection in HDB and GBM primary tissue explants that both contain SOX2+ neural progenitors. Strikingly, although the HDB proved uniformly vulnerable to ZIKV infection, GBM was more refractory, and this correlated with an innate immune expression signature. Indeed, GBM-derived CD11b+ microglia/macrophages were necessary and sufficient to protect progenitors against ZIKV infection in a non-cell autonomous manner. Using SOX2+ GBM cell lines, we found that CD11b+-conditioned medium containing type 1 interferon beta (IFNβ) promoted progenitor resistance to ZIKV, whereas inhibition of JAK1/2 signaling restored productive infection. Additionally, CD11b+ conditioned medium, and IFNβ treatment rendered HDB progenitor lines and explants refractory to ZIKV. These findings provide insight into neuroprotection for HDB progenitors as well as enhanced GBM oncolytic therapies., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
24. Generation of functional human oligodendrocytes from dermal fibroblasts by direct lineage conversion.
- Author
-
Tanabe K, Nobuta H, Yang N, Ang CE, Huie P, Jordan S, Oldham MC, Rowitch DH, and Wernig M
- Subjects
- Cell Differentiation physiology, Fibroblasts, Humans, Oligodendroglia metabolism, Induced Pluripotent Stem Cells metabolism, Pelizaeus-Merzbacher Disease genetics, Pelizaeus-Merzbacher Disease metabolism, Pelizaeus-Merzbacher Disease therapy
- Abstract
Oligodendrocytes, the myelinating cells of the central nervous system, possess great potential for disease modeling and cell transplantation-based therapies for leukodystrophies. However, caveats to oligodendrocyte differentiation protocols ( Ehrlich et al., 2017; Wang et al., 2013; Douvaras and Fossati, 2015) from human embryonic stem and induced pluripotent stem cells (iPSCs), which include slow and inefficient differentiation, and tumorigenic potential of contaminating undifferentiated pluripotent cells, are major bottlenecks towards their translational utility. Here, we report the rapid generation of human oligodendrocytes by direct lineage conversion of human dermal fibroblasts (HDFs). We show that the combination of the four transcription factors OLIG2, SOX10, ASCL1 and NKX2.2 is sufficient to convert HDFs to induced oligodendrocyte precursor cells (iOPCs). iOPCs resemble human primary and iPSC-derived OPCs based on morphology and transcriptomic analysis. Importantly, iOPCs can differentiate into mature myelinating oligodendrocytes in vitro and in vivo. Finally, iOPCs derived from patients with Pelizaeus Merzbacher disease, a hypomyelinating leukodystrophy caused by mutations in the proteolipid protein 1 (PLP1) gene, showed increased cell death compared with iOPCs from healthy donors. Thus, human iOPCs generated by direct lineage conversion represent an attractive new source for human cell-based disease models and potentially myelinating cell grafts., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
25. Publisher Correction: Brain charts for the human lifespan.
- Author
-
Bethlehem RAI, Seidlitz J, White SR, Vogel JW, Anderson KM, Adamson C, Adler S, Alexopoulos GS, Anagnostou E, Areces-Gonzalez A, Astle DE, Auyeung B, Ayub M, Bae J, Ball G, Baron-Cohen S, Beare R, Bedford SA, Benegal V, Beyer F, Blangero J, Blesa Cábez M, Boardman JP, Borzage M, Bosch-Bayard JF, Bourke N, Calhoun VD, Chakravarty MM, Chen C, Chertavian C, Chetelat G, Chong YS, Cole JH, Corvin A, Costantino M, Courchesne E, Crivello F, Cropley VL, Crosbie J, Crossley N, Delarue M, Delorme R, Desrivieres S, Devenyi GA, Di Biase MA, Dolan R, Donald KA, Donohoe G, Dunlop K, Edwards AD, Elison JT, Ellis CT, Elman JA, Eyler L, Fair DA, Feczko E, Fletcher PC, Fonagy P, Franz CE, Galan-Garcia L, Gholipour A, Giedd J, Gilmore JH, Glahn DC, Goodyer IM, Grant PE, Groenewold NA, Gunning FM, Gur RE, Gur RC, Hammill CF, Hansson O, Hedden T, Heinz A, Henson RN, Heuer K, Hoare J, Holla B, Holmes AJ, Holt R, Huang H, Im K, Ipser J, Jack CR Jr, Jackowski AP, Jia T, Johnson KA, Jones PB, Jones DT, Kahn RS, Karlsson H, Karlsson L, Kawashima R, Kelley EA, Kern S, Kim KW, Kitzbichler MG, Kremen WS, Lalonde F, Landeau B, Lee S, Lerch J, Lewis JD, Li J, Liao W, Liston C, Lombardo MV, Lv J, Lynch C, Mallard TT, Marcelis M, Markello RD, Mathias SR, Mazoyer B, McGuire P, Meaney MJ, Mechelli A, Medic N, Misic B, Morgan SE, Mothersill D, Nigg J, Ong MQW, Ortinau C, Ossenkoppele R, Ouyang M, Palaniyappan L, Paly L, Pan PM, Pantelis C, Park MM, Paus T, Pausova Z, Paz-Linares D, Pichet Binette A, Pierce K, Qian X, Qiu J, Qiu A, Raznahan A, Rittman T, Rodrigue A, Rollins CK, Romero-Garcia R, Ronan L, Rosenberg MD, Rowitch DH, Salum GA, Satterthwaite TD, Schaare HL, Schachar RJ, Schultz AP, Schumann G, Schöll M, Sharp D, Shinohara RT, Skoog I, Smyser CD, Sperling RA, Stein DJ, Stolicyn A, Suckling J, Sullivan G, Taki Y, Thyreau B, Toro R, Traut N, Tsvetanov KA, Turk-Browne NB, Tuulari JJ, Tzourio C, Vachon-Presseau É, Valdes-Sosa MJ, Valdes-Sosa PA, Valk SL, van Amelsvoort T, Vandekar SN, Vasung L, Victoria LW, Villeneuve S, Villringer A, Vértes PE, Wagstyl K, Wang YS, Warfield SK, Warrier V, Westman E, Westwater ML, Whalley HC, Witte AV, Yang N, Yeo B, Yun H, Zalesky A, Zar HJ, Zettergren A, Zhou JH, Ziauddeen H, Zugman A, Zuo XN, Bullmore ET, and Alexander-Bloch AF
- Published
- 2022
- Full Text
- View/download PDF
26. Genetic correlates of phenotypic heterogeneity in autism.
- Author
-
Warrier V, Zhang X, Reed P, Havdahl A, Moore TM, Cliquet F, Leblond CS, Rolland T, Rosengren A, Rowitch DH, Hurles ME, Geschwind DH, Børglum AD, Robinson EB, Grove J, Martin HC, Bourgeron T, and Baron-Cohen S
- Subjects
- Cognition, Female, Humans, Male, Autism Spectrum Disorder genetics, Autistic Disorder genetics, Intellectual Disability genetics
- Abstract
The substantial phenotypic heterogeneity in autism limits our understanding of its genetic etiology. To address this gap, here we investigated genetic differences between autistic individuals (n
max = 12,893) based on core and associated features of autism, co-occurring developmental disabilities and sex. We conducted a comprehensive factor analysis of core autism features in autistic individuals and identified six factors. Common genetic variants were associated with the core factors, but de novo variants were not. We found that higher autism polygenic scores (PGS) were associated with lower likelihood of co-occurring developmental disabilities in autistic individuals. Furthermore, in autistic individuals without co-occurring intellectual disability (ID), autism PGS are overinherited by autistic females compared to males. Finally, we observed higher SNP heritability for autistic males and for autistic individuals without ID. Deeper phenotypic characterization will be critical in determining how the complex underlying genetics shape cognition, behavior and co-occurring conditions in autism., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
27. Refinements and considerations for trio whole-genome sequence analysis when investigating Mendelian diseases presenting in early childhood.
- Author
-
French CE, Dolling H, Mégy K, Sanchis-Juan A, Kumar A, Delon I, Wakeling M, Mallin L, Agrawal S, Austin T, Walston F, Park SM, Parker A, Piyasena C, Bradbury K, Ellard S, Rowitch DH, and Raymond FL
- Abstract
To facilitate early deployment of whole-genome sequencing (WGS) for severely ill children, a standardized pipeline for WGS analysis with timely turnaround and primary care pediatric uptake is needed. We developed a bioinformatics pipeline for comprehensive gene-agnostic trio WGS analysis of children suspected of having an undiagnosed monogenic disease that included detection and interpretation of primary genetic mechanisms of disease, including SNVs/indels, CNVs/SVs, uniparental disomy (UPD), imprinted genes, short tandem repeat expansions, mobile element insertions, SMN1/2 copy number calling, and mitochondrial genome variants. We assessed primary care practitioner experience and competence in a large cohort of 521 families (comprising 90% WGS trios). Children were identified by primary practitioners for recruitment, and we used the UK index of multiple deprivation to confirm lack of patient socio-economic status ascertainment bias. Of the 521 children sequenced, 176 (34%) received molecular diagnoses, with rates as high as 45% for neurology clinics. Twenty-three of the diagnosed cases (13%) required bespoke methods beyond routine SNV/CNV analysis. In our multidisciplinary clinician user experience assessment, both pediatricians and clinical geneticists expressed strong support for rapid WGS early in the care pathway, but requested further training in determining patient selection, consenting, and variant interpretation. Rapid trio WGS provides an efficacious single-pass screening test for children when deployed by primary practitioners in clinical settings that carry high a priori risk for rare pediatric disease presentations., Competing Interests: The authors declare no competing interests., (© 2022 University of Cambridge.)
- Published
- 2022
- Full Text
- View/download PDF
28. Brain charts for the human lifespan.
- Author
-
Bethlehem RAI, Seidlitz J, White SR, Vogel JW, Anderson KM, Adamson C, Adler S, Alexopoulos GS, Anagnostou E, Areces-Gonzalez A, Astle DE, Auyeung B, Ayub M, Bae J, Ball G, Baron-Cohen S, Beare R, Bedford SA, Benegal V, Beyer F, Blangero J, Blesa Cábez M, Boardman JP, Borzage M, Bosch-Bayard JF, Bourke N, Calhoun VD, Chakravarty MM, Chen C, Chertavian C, Chetelat G, Chong YS, Cole JH, Corvin A, Costantino M, Courchesne E, Crivello F, Cropley VL, Crosbie J, Crossley N, Delarue M, Delorme R, Desrivieres S, Devenyi GA, Di Biase MA, Dolan R, Donald KA, Donohoe G, Dunlop K, Edwards AD, Elison JT, Ellis CT, Elman JA, Eyler L, Fair DA, Feczko E, Fletcher PC, Fonagy P, Franz CE, Galan-Garcia L, Gholipour A, Giedd J, Gilmore JH, Glahn DC, Goodyer IM, Grant PE, Groenewold NA, Gunning FM, Gur RE, Gur RC, Hammill CF, Hansson O, Hedden T, Heinz A, Henson RN, Heuer K, Hoare J, Holla B, Holmes AJ, Holt R, Huang H, Im K, Ipser J, Jack CR Jr, Jackowski AP, Jia T, Johnson KA, Jones PB, Jones DT, Kahn RS, Karlsson H, Karlsson L, Kawashima R, Kelley EA, Kern S, Kim KW, Kitzbichler MG, Kremen WS, Lalonde F, Landeau B, Lee S, Lerch J, Lewis JD, Li J, Liao W, Liston C, Lombardo MV, Lv J, Lynch C, Mallard TT, Marcelis M, Markello RD, Mathias SR, Mazoyer B, McGuire P, Meaney MJ, Mechelli A, Medic N, Misic B, Morgan SE, Mothersill D, Nigg J, Ong MQW, Ortinau C, Ossenkoppele R, Ouyang M, Palaniyappan L, Paly L, Pan PM, Pantelis C, Park MM, Paus T, Pausova Z, Paz-Linares D, Pichet Binette A, Pierce K, Qian X, Qiu J, Qiu A, Raznahan A, Rittman T, Rodrigue A, Rollins CK, Romero-Garcia R, Ronan L, Rosenberg MD, Rowitch DH, Salum GA, Satterthwaite TD, Schaare HL, Schachar RJ, Schultz AP, Schumann G, Schöll M, Sharp D, Shinohara RT, Skoog I, Smyser CD, Sperling RA, Stein DJ, Stolicyn A, Suckling J, Sullivan G, Taki Y, Thyreau B, Toro R, Traut N, Tsvetanov KA, Turk-Browne NB, Tuulari JJ, Tzourio C, Vachon-Presseau É, Valdes-Sosa MJ, Valdes-Sosa PA, Valk SL, van Amelsvoort T, Vandekar SN, Vasung L, Victoria LW, Villeneuve S, Villringer A, Vértes PE, Wagstyl K, Wang YS, Warfield SK, Warrier V, Westman E, Westwater ML, Whalley HC, Witte AV, Yang N, Yeo B, Yun H, Zalesky A, Zar HJ, Zettergren A, Zhou JH, Ziauddeen H, Zugman A, Zuo XN, Bullmore ET, and Alexander-Bloch AF
- Subjects
- Body Height, Humans, Magnetic Resonance Imaging methods, Neuroimaging, Brain anatomy & histology, Longevity
- Abstract
Over the past few decades, neuroimaging has become a ubiquitous tool in basic research and clinical studies of the human brain. However, no reference standards currently exist to quantify individual differences in neuroimaging metrics over time, in contrast to growth charts for anthropometric traits such as height and weight
1 . Here we assemble an interactive open resource to benchmark brain morphology derived from any current or future sample of MRI data ( http://www.brainchart.io/ ). With the goal of basing these reference charts on the largest and most inclusive dataset available, acknowledging limitations due to known biases of MRI studies relative to the diversity of the global population, we aggregated 123,984 MRI scans, across more than 100 primary studies, from 101,457 human participants between 115 days post-conception to 100 years of age. MRI metrics were quantified by centile scores, relative to non-linear trajectories2 of brain structural changes, and rates of change, over the lifespan. Brain charts identified previously unreported neurodevelopmental milestones3 , showed high stability of individuals across longitudinal assessments, and demonstrated robustness to technical and methodological differences between primary studies. Centile scores showed increased heritability compared with non-centiled MRI phenotypes, and provided a standardized measure of atypical brain structure that revealed patterns of neuroanatomical variation across neurological and psychiatric disorders. In summary, brain charts are an essential step towards robust quantification of individual variation benchmarked to normative trajectories in multiple, commonly used neuroimaging phenotypes., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
29. Multicenter Consensus Approach to Evaluation of Neonatal Hypotonia in the Genomic Era: A Review.
- Author
-
Morton SU, Christodoulou J, Costain G, Muntoni F, Wakeling E, Wojcik MH, French CE, Szuto A, Dowling JJ, Cohn RD, Raymond FL, Darras BT, Williams DA, Lunke S, Stark Z, Rowitch DH, and Agrawal PB
- Subjects
- Child, Consensus, Genetic Testing methods, Humans, Infant, Infant, Newborn, Multicenter Studies as Topic, Exome Sequencing methods, Intensive Care Units, Neonatal, Muscle Hypotonia diagnosis, Muscle Hypotonia genetics
- Abstract
Importance: Infants with hypotonia can present with a variety of potentially severe clinical signs and symptoms and often require invasive testing and multiple procedures. The wide range of clinical presentations and potential etiologies leaves diagnosis and prognosis uncertain, underscoring the need for rapid elucidation of the underlying genetic cause of disease., Observations: The clinical application of exome sequencing or genome sequencing has dramatically improved the timely yield of diagnostic testing for neonatal hypotonia, with diagnostic rates of greater than 50% in academic neonatal intensive care units (NICUs) across Australia, Canada, the UK, and the US, which compose the International Precision Child Health Partnership (IPCHiP). A total of 74% (17 of 23) of patients had a change in clinical care in response to genetic diagnosis, including 2 patients who received targeted therapy. This narrative review discusses the common causes of neonatal hypotonia, the relative benefits and limitations of available testing modalities used in NICUs, and hypotonia management recommendations., Conclusions and Relevance: This narrative review summarizes the causes of neonatal hypotonia and the benefits of prompt genetic diagnosis, including improved prognostication and identification of targeted treatments which can improve the short-term and long-term outcomes. Institutional resources can vary among different NICUs; as a result, consideration should be given to rule out a small number of relatively unique conditions for which rapid targeted genetic testing is available. Nevertheless, the consensus recommendation is to use rapid genome or exome sequencing as a first-line testing option for NICU patients with unexplained hypotonia. As part of the IPCHiP, this diagnostic experience will be collected in a central database with the goal of advancing knowledge of neonatal hypotonia and improving evidence-based practice.
- Published
- 2022
- Full Text
- View/download PDF
30. Neuroprotective effects of Sonic hedgehog agonist SAG in a rat model of neonatal stroke.
- Author
-
Nguyen V, Chavali M, Larpthaveesarp A, Kodali S, Gonzalez G, Franklin RJM, Rowitch DH, and Gonzalez F
- Subjects
- Animals, Behavior, Animal, Cell Proliferation, Disease Models, Animal, Humans, Infant, Newborn, Infarction, Middle Cerebral Artery complications, Mice, Rats, Rats, Sprague-Dawley, Stroke etiology, Hedgehog Proteins antagonists & inhibitors, Neuroprotective Agents therapeutic use, Small Molecule Libraries therapeutic use, Stroke prevention & control
- Abstract
Background: Neonatal stroke affects 1 in 2800 live births and is a major cause of neurological injury. The Sonic hedgehog (Shh) signaling pathway is critical for central nervous system (CNS) development and has neuroprotective and reparative effects in different CNS injury models. Previous studies have demonstrated beneficial effects of small molecule Shh-Smoothened agonist (SAG) against neonatal cerebellar injury and it improves Down syndrome-related brain structural deficits in mice. Here we investigated SAG neuroprotection in rat models of neonatal ischemia-reperfusion (stroke) and adult focal white matter injury., Methods: We used transient middle cerebral artery occlusion at P10 and ethidium bromide (EB) injection in adult rats to induce damage. Following surgery and SAG or vehicle treatment, we analyzed tissue loss, cell proliferation and fate, and behavioral outcome., Results: We report that a single dose of SAG administered following neonatal stroke preserved brain volume, reduced gliosis, enhanced oligodendrocyte progenitor cell (OPC) and EC proliferation, and resulted in long-term cognitive improvement. Single-dose SAG also promoted proliferation of OPCs following focal demyelination in the adult rat., Conclusions: These findings indicate benefit of one-time SAG treatment post insult in reducing brain injury and improving behavioral outcome after experimental neonatal stroke., Impact: A one-time dose of small molecule Sonic hedgehog agonist protected against neonatal stroke and improved long-term behavioral outcomes in a rat model. This study extends the use of Sonic hedgehog in treating developing brain injury, previously shown in animal models of Down syndrome and cerebellar injury. Sonic hedgehog agonist is one of the most promising therapies in treating neonatal stroke thanks to its safety profile and low dosage., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
31. Evidence for glutamine synthetase function in mouse spinal cord oligodendrocytes.
- Author
-
Ben Haim L, Schirmer L, Zulji A, Sabeur K, Tiret B, Ribon M, Chang S, Lamers WH, Boillée S, Chaumeil MM, and Rowitch DH
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Motor Neurons pathology, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Amyotrophic Lateral Sclerosis pathology, Glutamate-Ammonia Ligase genetics, Glutamate-Ammonia Ligase metabolism, Oligodendroglia metabolism, Spinal Cord metabolism
- Abstract
Glutamine synthetase (GS) is a key enzyme that metabolizes glutamate into glutamine. While GS is highly enriched in astrocytes, expression in other glial lineages has been noted. Using a combination of reporter mice and cell type-specific markers, we show that GS is expressed in myelinating oligodendrocytes (OL) but not oligodendrocyte progenitor cells of the mouse and human ventral spinal cord. To investigate the role of GS in mature OL, we used a conditional knockout (cKO) approach to selectively delete GS-encoding gene (Glul) in OL, which caused a significant decrease in glutamine levels on mouse spinal cord extracts. GS cKO mice (CNP-cre
+ :Glulfl/fl ) showed no differences in motor neuron numbers, size or axon density; OL differentiation and myelination in the ventral spinal cord was normal up to 6 months of age. Interestingly, GS cKO mice showed a transient and specific decrease in peak force while locomotion and motor coordination remained unaffected. Last, GS expression in OL was increased in chronic pathological conditions in both mouse and humans. We found a disease-stage dependent increase of OL expressing GS in the ventral spinal cord of SOD1(G93A) mouse model of amyotrophic lateral sclerosis. Moreover, we showed that GLUL transcripts levels were increased in OL in leukocortical tissue from multiple sclerosis but not control patients. These findings provide evidence towards OL-encoded GS function in spinal cord sensorimotor axis, which is dysregulated in chronic neurological diseases., (© 2021 Wiley Periodicals LLC.)- Published
- 2021
- Full Text
- View/download PDF
32. MC3R links nutritional state to childhood growth and the timing of puberty.
- Author
-
Lam BYH, Williamson A, Finer S, Day FR, Tadross JA, Gonçalves Soares A, Wade K, Sweeney P, Bedenbaugh MN, Porter DT, Melvin A, Ellacott KLJ, Lippert RN, Buller S, Rosmaninho-Salgado J, Dowsett GKC, Ridley KE, Xu Z, Cimino I, Rimmington D, Rainbow K, Duckett K, Holmqvist S, Khan A, Dai X, Bochukova EG, Trembath RC, Martin HC, Coll AP, Rowitch DH, Wareham NJ, van Heel DA, Timpson N, Simerly RB, Ong KK, Cone RD, Langenberg C, Perry JRB, Yeo GS, and O'Rahilly S
- Subjects
- Adolescent, Aged, 80 and over, Animals, Child, Estrous Cycle genetics, Estrous Cycle physiology, Female, Homozygote, Humans, Hypothalamus cytology, Hypothalamus physiology, Insulin-Like Growth Factor I metabolism, Male, Melanocortins metabolism, Menarche genetics, Menarche physiology, Mice, Phenotype, Puberty genetics, Receptor, Melanocortin, Type 3 deficiency, Receptor, Melanocortin, Type 3 genetics, Sexual Maturation genetics, Time Factors, Weight Gain, Child Development physiology, Nutritional Status physiology, Puberty physiology, Receptor, Melanocortin, Type 3 metabolism, Sexual Maturation physiology
- Abstract
The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development
1 . The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2 . Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
33. Nutritional regulation of oligodendrocyte differentiation regulates perineuronal net remodeling in the median eminence.
- Author
-
Kohnke S, Buller S, Nuzzaci D, Ridley K, Lam B, Pivonkova H, Bentsen MA, Alonge KM, Zhao C, Tadross J, Holmqvist S, Shimizu T, Hathaway H, Li H, Macklin W, Schwartz MW, Richardson WD, Yeo GSH, Franklin RJM, Karadottir RT, Rowitch DH, and Blouet C
- Subjects
- Adult, Animals, Cell Lineage, Cell Proliferation, Humans, Male, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Inbred C57BL, Oligodendroglia ultrastructure, Single-Cell Analysis, Transcriptome genetics, Mice, Cell Differentiation, Median Eminence cytology, Nerve Net physiology, Nutritional Physiological Phenomena, Oligodendroglia cytology
- Abstract
The mediobasal hypothalamus (MBH; arcuate nucleus of the hypothalamus [ARH] and median eminence [ME]) is a key nutrient sensing site for the production of the complex homeostatic feedback responses required for the maintenance of energy balance. Here, we show that refeeding after an overnight fast rapidly triggers proliferation and differentiation of oligodendrocyte progenitors, leading to the production of new oligodendrocytes in the ME specifically. During this nutritional paradigm, ME perineuronal nets (PNNs), emerging regulators of ARH metabolic functions, are rapidly remodeled, and this process requires myelin regulatory factor (Myrf) in oligodendrocyte progenitors. In genetically obese ob/ob mice, nutritional regulations of ME oligodendrocyte differentiation and PNN remodeling are blunted, and enzymatic digestion of local PNN increases food intake and weight gain. We conclude that MBH PNNs are required for the maintenance of energy balance in lean mice and are remodeled in the adult ME by the nutritional control of oligodendrocyte differentiation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. A classification of videoconferencing related illness: the Zoomnotic diseases.
- Author
-
Rowitch DH, O'Rahilly S, and Smith GCS
- Subjects
- Humans, Videoconferencing
- Published
- 2021
- Full Text
- View/download PDF
35. Letter to Editor Response to: Is zoomnosis a human-driven human zoonosis? A call for action.
- Author
-
Rowitch DH, O'Rahilly S, and Smith GCS
- Published
- 2021
- Full Text
- View/download PDF
36. Diversity and Function of Glial Cell Types in Multiple Sclerosis.
- Author
-
Schirmer L, Schafer DP, Bartels T, Rowitch DH, and Calabresi PA
- Subjects
- Astrocytes, Humans, Microglia, Neuroglia, Oligodendroglia, Multiple Sclerosis
- Abstract
Glial subtype diversity is an emerging topic in neurobiology and immune-mediated neurological diseases such as multiple sclerosis (MS). We discuss recent conceptual and technological advances that allow a better understanding of the transcriptomic and functional heterogeneity of oligodendrocytes (OLs), astrocytes, and microglial cells under inflammatory-demyelinating conditions. Recent single cell transcriptomic studies suggest the occurrence of novel homeostatic and reactive glial subtypes and provide insight into the molecular events during disease progression. Multiplexed RNA in situ hybridization has enabled 'mapping back' dysregulated gene expression to glial subtypes within the MS lesion microenvironment. These findings suggest novel homeostatic and reactive glial-cell-type functions both in immune-related processes and neuroprotection relevant to understanding the pathology of MS., Competing Interests: Declaration of Interests The authors have no interests to declare., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Reactive astrocyte nomenclature, definitions, and future directions.
- Author
-
Escartin C, Galea E, Lakatos A, O'Callaghan JP, Petzold GC, Serrano-Pozo A, Steinhäuser C, Volterra A, Carmignoto G, Agarwal A, Allen NJ, Araque A, Barbeito L, Barzilai A, Bergles DE, Bonvento G, Butt AM, Chen WT, Cohen-Salmon M, Cunningham C, Deneen B, De Strooper B, Díaz-Castro B, Farina C, Freeman M, Gallo V, Goldman JE, Goldman SA, Götz M, Gutiérrez A, Haydon PG, Heiland DH, Hol EM, Holt MG, Iino M, Kastanenka KV, Kettenmann H, Khakh BS, Koizumi S, Lee CJ, Liddelow SA, MacVicar BA, Magistretti P, Messing A, Mishra A, Molofsky AV, Murai KK, Norris CM, Okada S, Oliet SHR, Oliveira JF, Panatier A, Parpura V, Pekna M, Pekny M, Pellerin L, Perea G, Pérez-Nievas BG, Pfrieger FW, Poskanzer KE, Quintana FJ, Ransohoff RM, Riquelme-Perez M, Robel S, Rose CR, Rothstein JD, Rouach N, Rowitch DH, Semyanov A, Sirko S, Sontheimer H, Swanson RA, Vitorica J, Wanner IB, Wood LB, Wu J, Zheng B, Zimmer ER, Zorec R, Sofroniew MV, and Verkhratsky A
- Subjects
- Animals, Brain Diseases pathology, Brain Injuries pathology, Humans, Spinal Cord Injuries pathology, Aging pathology, Astrocytes pathology, Brain pathology, Spinal Cord pathology
- Abstract
Reactive astrocytes are astrocytes undergoing morphological, molecular, and functional remodeling in response to injury, disease, or infection of the CNS. Although this remodeling was first described over a century ago, uncertainties and controversies remain regarding the contribution of reactive astrocytes to CNS diseases, repair, and aging. It is also unclear whether fixed categories of reactive astrocytes exist and, if so, how to identify them. We point out the shortcomings of binary divisions of reactive astrocytes into good-vs-bad, neurotoxic-vs-neuroprotective or A1-vs-A2. We advocate, instead, that research on reactive astrocytes include assessment of multiple molecular and functional parameters-preferably in vivo-plus multivariate statistics and determination of impact on pathological hallmarks in relevant models. These guidelines may spur the discovery of astrocyte-based biomarkers as well as astrocyte-targeting therapies that abrogate detrimental actions of reactive astrocytes, potentiate their neuro- and glioprotective actions, and restore or augment their homeostatic, modulatory, and defensive functions.
- Published
- 2021
- Full Text
- View/download PDF
38. Wnt-Dependent Oligodendroglial-Endothelial Interactions Regulate White Matter Vascularization and Attenuate Injury.
- Author
-
Chavali M, Ulloa-Navas MJ, Pérez-Borredá P, Garcia-Verdugo JM, McQuillen PS, Huang EJ, and Rowitch DH
- Subjects
- Animals, Axin Protein metabolism, Cell Differentiation physiology, Endothelium, Vascular metabolism, Ferrets, Humans, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Mice, Mice, Transgenic, White Matter metabolism, Endothelial Cells metabolism, Hypoxia metabolism, Oligodendroglia metabolism, White Matter blood supply, Wnt Signaling Pathway physiology
- Abstract
Recent studies have indicated oligodendroglial-vascular crosstalk during brain development, but the underlying mechanisms are incompletely understood. We report that oligodendrocyte precursor cells (OPCs) contact sprouting endothelial tip cells in mouse, ferret, and human neonatal white matter. Using transgenic mice, we show that increased or decreased OPC density results in cognate changes in white matter vascular investment. Hypoxia induced increases in OPC numbers, vessel density and endothelial cell expression of the Wnt pathway targets Apcdd1 and Axin2 in white matter, suggesting paracrine OPC-endothelial signaling. Conditional knockout of OPC Wntless resulted in diminished white matter vascular growth in normoxia, whereas loss of Wnt7a/b function blunted the angiogenic response to hypoxia, resulting in severe white matter damage. These findings indicate that OPC-endothelial cell interactions regulate neonatal white matter vascular development in a Wnt-dependent manner and further suggest this mechanism is important in attenuating hypoxic injury., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Origins and Proliferative States of Human Oligodendrocyte Precursor Cells.
- Author
-
Huang W, Bhaduri A, Velmeshev D, Wang S, Wang L, Rottkamp CA, Alvarez-Buylla A, Rowitch DH, and Kriegstein AR
- Subjects
- Cadherin Related Proteins, Cadherins genetics, Cell Proliferation genetics, Cells, Cultured, Cerebral Cortex embryology, Cerebral Cortex metabolism, Ependymoglial Cells cytology, ErbB Receptors genetics, ErbB Receptors metabolism, HEK293 Cells, Humans, Immunohistochemistry, Oligodendrocyte Precursor Cells cytology, RNA, Small Interfering, RNA-Seq, Single-Cell Analysis, White Matter cytology, White Matter embryology, White Matter metabolism, Cadherins metabolism, Cerebral Cortex cytology, Ependymoglial Cells metabolism, Neurogenesis genetics, Oligodendrocyte Precursor Cells metabolism
- Abstract
Human cerebral cortex size and complexity has increased greatly during evolution. While increased progenitor diversity and enhanced proliferative potential play important roles in human neurogenesis and gray matter expansion, the mechanisms of human oligodendrogenesis and white matter expansion remain largely unknown. Here, we identify EGFR-expressing "Pre-OPCs" that originate from outer radial glial cells (oRGs) and undergo mitotic somal translocation (MST) during division. oRG-derived Pre-OPCs provide an additional source of human cortical oligodendrocyte precursor cells (OPCs) and define a lineage trajectory. We further show that human OPCs undergo consecutive symmetric divisions to exponentially increase the progenitor pool size. Additionally, we find that the OPC-enriched gene, PCDH15, mediates daughter cell repulsion and facilitates proliferation. These findings indicate properties of OPC derivation, proliferation, and dispersion important for human white matter expansion and myelination., Competing Interests: Declaration of Interests A.R.K. is a co-founder and board member of Neurona Therapeutics., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Applying support-vector machine learning algorithms toward predicting host-guest interactions with cucurbit[7]uril.
- Author
-
Tabet A, Gebhart T, Wu G, Readman C, Pierson Smela M, Rana VK, Baker C, Bulstrode H, Anikeeva P, Rowitch DH, and Scherman OA
- Subjects
- Density Functional Theory, Models, Chemical, Support Vector Machine, Benzimidazoles chemistry, Bridged-Ring Compounds chemistry, Heterocyclic Compounds, 3-Ring chemistry, Imidazoles chemistry
- Abstract
Machine learning is a valuable tool in the development of chemical technologies but its applications into supramolecular chemistry have been limited. Here, the utility of kernel-based support vector machine learning using density functional theory calculations as training data is evaluated when used to predict equilibrium binding coefficients of small molecules with cucurbit[7]uril (CB[7]). We find that utilising SVMs may confer some predictive ability. This algorithm was then used to predict the binding of drugs TAK-580 and selumetinib. The algorithm did predict strong binding for TAK-580 and poor binding for selumetinib, and these results were experimentally validated. It was discovered that the larger homologue cucurbit[8]uril (CB[8]) is partial to selumetinib, suggesting an opportunity for tunable release by introducing different concentrations of CB[7] or CB[8] into a hydrogel depot. We qualitatively demonstrated that these drugs may have utility in combination against gliomas. Finally, mass transfer simulations show CB[7] can independently tune the release of TAK-580 without affecting selumetinib. This work gives specific evidence that a machine learning approach to recognition of small molecules by macrocycles has merit and reinforces the view that machine learning may prove valuable in the development of drug delivery systems and supramolecular chemistry more broadly.
- Published
- 2020
- Full Text
- View/download PDF
41. Oxygen Tension and the VHL-Hif1α Pathway Determine Onset of Neuronal Polarization and Cerebellar Germinal Zone Exit.
- Author
-
Kullmann JA, Trivedi N, Howell D, Laumonnerie C, Nguyen V, Banerjee SS, Stabley DR, Shirinifard A, Rowitch DH, and Solecki DJ
- Subjects
- Animals, Cell Differentiation physiology, Female, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Male, Mice, Neurons metabolism, Oxygen, Signal Transduction physiology, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Cell Polarity physiology, Cerebellum growth & development, Cerebellum physiology, Neurogenesis physiology, Neurons cytology
- Abstract
Postnatal brain circuit assembly is driven by temporally regulated intrinsic and cell-extrinsic cues that organize neurogenesis, migration, and axo-dendritic specification in post-mitotic neurons. While cell polarity is an intrinsic organizer of morphogenic events, environmental cues in the germinal zone (GZ) instructing neuron polarization and their coupling during postnatal development are unclear. We report that oxygen tension, which rises at birth, and the von Hippel-Lindau (VHL)-hypoxia-inducible factor 1α (Hif1α) pathway regulate polarization and maturation of post-mitotic cerebellar granule neurons (CGNs). At early postnatal stages with low GZ vascularization, Hif1α restrains CGN-progenitor cell-cycle exit. Unexpectedly, cell-intrinsic VHL-Hif1α pathway activation also delays the timing of CGN differentiation, germinal zone exit, and migration initiation through transcriptional repression of the partitioning-defective (Pard) complex. As vascularization proceeds, these inhibitory mechanisms are downregulated, implicating increasing oxygen tension as a critical switch for neuronal polarization and cerebellar GZ exit., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. Astrocyte layers in the mammalian cerebral cortex revealed by a single-cell in situ transcriptomic map.
- Author
-
Bayraktar OA, Bartels T, Holmqvist S, Kleshchevnikov V, Martirosyan A, Polioudakis D, Ben Haim L, Young AMH, Batiuk MY, Prakash K, Brown A, Roberts K, Paredes MF, Kawaguchi R, Stockley JH, Sabeur K, Chang SM, Huang E, Hutchinson P, Ullian EM, Hemberg M, Coppola G, Holt MG, Geschwind DH, and Rowitch DH
- Subjects
- Animals, Astrocytes metabolism, Brain Mapping, Cerebral Cortex metabolism, Humans, Mice, Neurons metabolism, Astrocytes cytology, Cerebral Cortex cytology, Neurons cytology, Transcriptome
- Abstract
Although the cerebral cortex is organized into six excitatory neuronal layers, it is unclear whether glial cells show distinct layering. In the present study, we developed a high-content pipeline, the large-area spatial transcriptomic (LaST) map, which can quantify single-cell gene expression in situ. Screening 46 candidate genes for astrocyte diversity across the mouse cortex, we identified superficial, mid and deep astrocyte identities in gradient layer patterns that were distinct from those of neurons. Astrocyte layer features, established in the early postnatal cortex, mostly persisted in adult mouse and human cortex. Single-cell RNA sequencing and spatial reconstruction analysis further confirmed the presence of astrocyte layers in the adult cortex. Satb2 and Reeler mutations that shifted neuronal post-mitotic development were sufficient to alter glial layering, indicating an instructive role for neuronal cues. Finally, astrocyte layer patterns diverged between mouse cortical regions. These findings indicate that excitatory neurons and astrocytes are organized into distinct lineage-associated laminae.
- Published
- 2020
- Full Text
- View/download PDF
43. Astrocyte Unfolded Protein Response Induces a Specific Reactivity State that Causes Non-Cell-Autonomous Neuronal Degeneration.
- Author
-
Smith HL, Freeman OJ, Butcher AJ, Holmqvist S, Humoud I, Schätzl T, Hughes DT, Verity NC, Swinden DP, Hayes J, de Weerd L, Rowitch DH, Franklin RJM, and Mallucci GR
- Subjects
- Animals, Endoplasmic Reticulum Stress drug effects, Enzyme Inhibitors pharmacology, In Vitro Techniques, Memory, Mice, Phosphorylation, Protein Biosynthesis, Protein Phosphatase 1 genetics, Protein Phosphatase 1 metabolism, Signal Transduction, Thapsigargin pharmacology, Transcriptome, Tunicamycin pharmacology, Unfolded Protein Response drug effects, Astrocytes metabolism, Eukaryotic Initiation Factor-2B metabolism, Neurodegenerative Diseases metabolism, Prion Diseases metabolism, Synapses metabolism, Unfolded Protein Response physiology, eIF-2 Kinase metabolism
- Abstract
Recent interest in astrocyte activation states has raised the fundamental question of how these cells, normally essential for synapse and neuronal maintenance, become pathogenic. Here, we show that activation of the unfolded protein response (UPR), specifically phosphorylated protein kinase R-like endoplasmic reticulum (ER) kinase (PERK-P) signaling-a pathway that is widely dysregulated in neurodegenerative diseases-generates a distinct reactivity state in astrocytes that alters the astrocytic secretome, leading to loss of synaptogenic function in vitro. Further, we establish that the same PERK-P-dependent astrocyte reactivity state is harmful to neurons in vivo in mice with prion neurodegeneration. Critically, targeting this signaling exclusively in astrocytes during prion disease is alone sufficient to prevent neuronal loss and significantly prolongs survival. Thus, the astrocyte reactivity state resulting from UPR over-activation is a distinct pathogenic mechanism that can by itself be effectively targeted for neuroprotection., Competing Interests: Declaration of Interests The authors declare no competing interests. O.J.F. is now an employee of AstraZeneca., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
44. Cucurbit[8]uril-Derived Graphene Hydrogels.
- Author
-
Rana VK, Tabet A, Vigil JA, Balzer CJ, Narkevicius A, Finlay J, Hallou C, Rowitch DH, Bulstrode H, and Scherman OA
- Abstract
The scalable production of uniformly distributed graphene (GR)-based composite materials remains a sizable challenge. While GR-polymer nanocomposites can be manufactured at a large scale, processing limitations result in poor control over the homogeneity of hydrophobic GR sheets in the matrices. Such processes often result in difficulties controlling stability and avoiding aggregation, therefore eliminating benefits that might have otherwise arisen from the nanoscopic dimensions of GR. Here, we report an exfoliated and stabilized GR dispersion in water. Cucurbit[8]uril (CB[8])-mediated host-guest chemistry was used to obtain supramolecular hydrogels consisting of uniformly distributed GR and guest-functionalized macromolecules. The obtained GR hydrogels show superior bioelectrical properties over identical systems produced without CB[8]. Utilizing such supramolecular interactions with biologically derived macromolecules is a promising approach to stabilize graphene in water and avoid oxidative chemistry.
- Published
- 2019
- Full Text
- View/download PDF
45. Ferret brain possesses young interneuron collections equivalent to human postnatal migratory streams.
- Author
-
Ellis JK, Sorrells SF, Mikhailova S, Chavali M, Chang S, Sabeur K, Mcquillen P, and Rowitch DH
- Subjects
- Animals, Brain metabolism, Caspase 3 metabolism, Cell Movement, Doublecortin Domain Proteins, Doublecortin Protein, Ferrets metabolism, Humans, Interneurons metabolism, Microtubule-Associated Proteins metabolism, Neuropeptides metabolism, Secretagogins metabolism, White Matter cytology, White Matter growth & development, White Matter metabolism, Brain cytology, Brain growth & development, Ferrets anatomy & histology, Interneurons cytology
- Abstract
The human early postnatal brain contains late migratory streams of immature interneurons that are directed to cortex and other focal brain regions. However, such migration is not observed in rodent brain, and whether other small animal models capture this aspect of human brain development is unclear. Here, we investigated whether the gyrencephalic ferret cortex possesses human-equivalent postnatal streams of doublecortin positive (DCX+) young neurons. We mapped DCX+ cells in the brains of ferrets at P20 (analogous to human term gestation), P40, P65, and P90. In addition to the rostral migratory stream, we identified three populations of young neurons with migratory morphology at P20 oriented toward: (a) prefrontal cortex, (b) dorsal posterior sigmoid gyrus, and (c) occipital lobe. These three neuronal collections were all present at P20 and became extinguished by P90 (equivalent to human postnatal age 2 years). DCX+ cells in such collections all expressed GAD67, identifying them as interneurons, and they variously expressed the subtype markers SP8 and secretagogin (SCGN). SCGN+ interneurons appeared in thick sections to be oriented from white matter toward multiple cortical regions, and persistent SCGN-expressing cells were observed in cortex. These findings indicate that ferret is a suitable animal model to study the human-relevant process of late postnatal cortical interneuron integration into multiple regions of cortex., (© 2019 The Authors. The Journal of Comparative Neurology published by Wiley Periodicals, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
46. Oligodendrocyte Death in Pelizaeus-Merzbacher Disease Is Rescued by Iron Chelation.
- Author
-
Nobuta H, Yang N, Ng YH, Marro SG, Sabeur K, Chavali M, Stockley JH, Killilea DW, Walter PB, Zhao C, Huie P Jr, Goldman SA, Kriegstein AR, Franklin RJM, Rowitch DH, and Wernig M
- Subjects
- Animals, Cell Differentiation, Cells, Cultured, Ferroptosis, Humans, Induced Pluripotent Stem Cells drug effects, Induced Pluripotent Stem Cells transplantation, Lipid Peroxidation, Mice, Mice, Mutant Strains, Mutation genetics, Myelin Proteolipid Protein genetics, Oligodendroglia drug effects, Oligodendroglia transplantation, Pelizaeus-Merzbacher Disease genetics, Pelizaeus-Merzbacher Disease pathology, Stem Cell Transplantation, Targeted Gene Repair, Deferiprone therapeutic use, Induced Pluripotent Stem Cells physiology, Iron metabolism, Iron Chelating Agents therapeutic use, Myelin Proteolipid Protein metabolism, Oligodendroglia physiology, Pelizaeus-Merzbacher Disease therapy
- Abstract
Pelizaeus-Merzbacher disease (PMD) is an X-linked leukodystrophy caused by mutations in Proteolipid Protein 1 (PLP1), encoding a major myelin protein, resulting in profound developmental delay and early lethality. Previous work showed involvement of unfolded protein response (UPR) and endoplasmic reticulum (ER) stress pathways, but poor PLP1 genotype-phenotype associations suggest additional pathogenetic mechanisms. Using induced pluripotent stem cell (iPSC) and gene-correction, we show that patient-derived oligodendrocytes can develop to the pre-myelinating stage, but subsequently undergo cell death. Mutant oligodendrocytes demonstrated key hallmarks of ferroptosis including lipid peroxidation, abnormal iron metabolism, and hypersensitivity to free iron. Iron chelation rescued mutant oligodendrocyte apoptosis, survival, and differentiationin vitro, and post-transplantation in vivo. Finally, systemic treatment of Plp1 mutant Jimpy mice with deferiprone, a small molecule iron chelator, reduced oligodendrocyte apoptosis and enabled myelin formation. Thus, oligodendrocyte iron-induced cell death and myelination is rescued by iron chelation in PMD pre-clinical models., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
47. Author Correction: Niche stiffness underlies the ageing of central nervous system progenitor cells.
- Author
-
Segel M, Neumann B, Hill MFE, Weber IP, Viscomi C, Zhao C, Young A, Agley CC, Thompson AJ, Gonzalez GA, Sharma A, Holmqvist S, Rowitch DH, Franze K, Franklin RJM, and Chalut KJ
- Abstract
An Amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
48. Niche stiffness underlies the ageing of central nervous system progenitor cells.
- Author
-
Segel M, Neumann B, Hill MFE, Weber IP, Viscomi C, Zhao C, Young A, Agley CC, Thompson AJ, Gonzalez GA, Sharma A, Holmqvist S, Rowitch DH, Franze K, Franklin RJM, and Chalut KJ
- Subjects
- Animals, Animals, Newborn, Cell Count, Extracellular Matrix pathology, Female, Humans, Membrane Proteins antagonists & inhibitors, Membrane Proteins metabolism, Oligodendroglia pathology, Rats, Adult Stem Cells pathology, Aging pathology, Central Nervous System pathology, Multipotent Stem Cells pathology, Stem Cell Niche physiology
- Abstract
Ageing causes a decline in tissue regeneration owing to a loss of function of adult stem cell and progenitor cell populations
1 . One example is the deterioration of the regenerative capacity of the widespread and abundant population of central nervous system (CNS) multipotent stem cells known as oligodendrocyte progenitor cells (OPCs)2 . A relatively overlooked potential source of this loss of function is the stem cell 'niche'-a set of cell-extrinsic cues that include chemical and mechanical signals3,4 . Here we show that the OPC microenvironment stiffens with age, and that this mechanical change is sufficient to cause age-related loss of function of OPCs. Using biological and synthetic scaffolds to mimic the stiffness of young brains, we find that isolated aged OPCs cultured on these scaffolds are molecularly and functionally rejuvenated. When we disrupt mechanical signalling, the proliferation and differentiation rates of OPCs are increased. We identify the mechanoresponsive ion channel PIEZO1 as a key mediator of OPC mechanical signalling. Inhibiting PIEZO1 overrides mechanical signals in vivo and allows OPCs to maintain activity in the ageing CNS. We also show that PIEZO1 is important in regulating cell number during CNS development. Thus we show that tissue stiffness is a crucial regulator of ageing in OPCs, and provide insights into how the function of adult stem and progenitor cells changes with age. Our findings could be important not only for the development of regenerative therapies, but also for understanding the ageing process itself.- Published
- 2019
- Full Text
- View/download PDF
49. Neuronal vulnerability and multilineage diversity in multiple sclerosis.
- Author
-
Schirmer L, Velmeshev D, Holmqvist S, Kaufmann M, Werneburg S, Jung D, Vistnes S, Stockley JH, Young A, Steindel M, Tung B, Goyal N, Bhaduri A, Mayer S, Engler JB, Bayraktar OA, Franklin RJM, Haeussler M, Reynolds R, Schafer DP, Friese MA, Shiow LR, Kriegstein AR, and Rowitch DH
- Subjects
- Adult, Animals, Astrocytes metabolism, Astrocytes pathology, Autopsy, Cryopreservation, Female, Homeodomain Proteins metabolism, Humans, Macrophages metabolism, Macrophages pathology, Male, Mice, Microglia metabolism, Microglia pathology, Middle Aged, Multiple Sclerosis genetics, Myelin Sheath metabolism, Neurons metabolism, Oligodendroglia metabolism, Oligodendroglia pathology, Phagocytosis, RNA, Small Nuclear analysis, RNA, Small Nuclear genetics, RNA-Seq, Transcriptome genetics, Cell Lineage, Multiple Sclerosis pathology, Neurons pathology
- Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.
- Published
- 2019
- Full Text
- View/download PDF
50. Long-Term Safety, Immunologic Response, and Imaging Outcomes following Neural Stem Cell Transplantation for Pelizaeus-Merzbacher Disease.
- Author
-
Gupta N, Henry RG, Kang SM, Strober J, Lim DA, Ryan T, Perry R, Farrell J, Ulman M, Rajalingam R, Gage A, Huhn SL, Barkovich AJ, and Rowitch DH
- Subjects
- Brain physiology, Child, Preschool, Follow-Up Studies, HLA Antigens immunology, Humans, Infant, Isoantibodies blood, Magnetic Resonance Imaging, Male, Neural Stem Cells cytology, Neural Stem Cells metabolism, Pelizaeus-Merzbacher Disease immunology, Pelizaeus-Merzbacher Disease pathology, Severity of Illness Index, Stem Cell Transplantation adverse effects, Transplantation, Homologous, Treatment Outcome, Brain diagnostic imaging, Neural Stem Cells transplantation, Pelizaeus-Merzbacher Disease therapy
- Abstract
Four boys with Pelizaeus-Merzbacher disease, an X-linked leukodystrophy, underwent transplantation with human allogeneic central nervous system stem cells (HuCNS-SC). Subsequently, all subjects were followed for an additional 4 years in this separate follow-up study to evaluate safety, neurologic function, magnetic resonance imaging (MRI) data, and immunologic response. The neurosurgical procedure, immunosuppression, and HuCNS-SC transplantation were well tolerated and all four subjects were alive at the conclusion of the study period. At year 2, all subjects exhibited diffusion MRI changes at the implantation sites as well as in more distant brain regions. There were persistent, increased signal changes in the three patients who were studied up to year 5. Two of four subjects developed donor-specific HLA alloantibodies, demonstrating that neural stem cells can elicit an immune response when injected into the CNS, and suggesting the importance of monitoring immunologic parameters and identifying markers of engraftment in future studies., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.