2023

Park, K., Kohl, M. M., Kwag, J. (2023) Memory encoding and retrieval by retrosplenial parvalbumin interneurons is impaired in Alzheimer's disease model mice. Current Biology, 34, (doi: 10.1016/j.cub.2023.12.014)

Quillet, R., Gutierrez-Mecinas, M., Polgár, E., Dickie, A. C., Boyle, K. A., Watanabe, M., Todd, A. J. (2023) Synaptic circuits involving gastrin-releasing peptide receptor-expressing neurons in the dorsal horn of the mouse spinal cord. Frontiers in Molecular Neuroscience, 16, (doi: 10.3389/fnmol.2023.1294994)

Andrianova, L., Yanakieva, S., Margetts-Smith, G., Kohli, S., Brady, E. S., Aggleton, J. P., Craig, M. T. (2023) No evidence from complementary data sources of a direct glutamatergic projection from the mouse anterior cingulate area to the hippocampal formation. eLife, 12, (doi: 10.7554/eLife.77364)

Boyle, K. A., Polgár, E., Gutierrez-Mecinas, M., Dickie, A. C., Cooper, A. H., Bell, A. M., Jumolea, M. E., Casas-Benito, A., Watanabe, M., Hughes, D. I., Weir, G. A., Riddell, J. S., Todd, A. J. (2023) Neuropeptide Y-expressing dorsal horn inhibitory interneurons gate spinal pain and itch signalling. eLife, 12, (doi: 10.7554/eLife.86633)

Davis, O. C., Dickie, A. C., Mustapa, M. B., Boyle, K. A., Browne, T. J., Gradwell, M. A., Smith, K. M., Polgár, E., Bell, A. M., Kókai, É., Watanabe, M., Wildner, H., Zeilhofer, H. U., Ginty, D. D., Callister, R. J., Graham, B. A., Todd, A. J., Hughes, D. I. (2023) Calretinin-expressing islet cells are a source of pre- and post-synaptic inhibition of non-peptidergic nociceptor input to the mouse spinal cord. Scientific Reports, 13, (doi: 10.1038/s41598-023-38605-9)

Brady, E. S., Griffiths, J., Andrianova, L., Bielska, M. H., Saito, T., Saido, T. C., Randall, A. D., Tamagnini, F., Witton, J., Craig, M. T. (2023) Alterations to parvalbumin-expressing interneuron function and associated network oscillations in the hippocampal – medial prefrontal cortex circuit during natural sleep in AppNL-G-F/NL-G-F mice. Neurobiology of Disease, 182, pp. 106151. (doi: 10.1016/j.nbd.2023.106151)

Openshaw, R. L., Thomson, D. M., Bristow, G. C., Mitchell, E. J., Pratt, J. A., Morris, B. J., Dawson, N. (2023) 16p11.2 deletion mice exhibit compromised fronto-temporal connectivity, GABAergic dysfunction, and enhanced attentional ability. Communications Biology, 6, (doi: 10.1038/s42003-023-04891-2)

Quillet, R., Dickie, A. C., Polgár, E., Gutierrez-Mecinas, M., Bell, A. M., Goffin, L., Watanabe, M., Todd, A. J. (2023) Characterisation of NPFF-expressing neurons in the superficial dorsal horn of the mouse spinal cord. Scientific Reports, 13, (doi: 10.1038/s41598-023-32720-3)

Panniello, M., Limal, S. A.C., Kohl, M. M. (2023) Imaging somatosensory cortex in rodents. Humana

Gutierrez-Mecinas, M., Kókai, É., Polgár, E., Quillet, R., Titterton, H. F., Weir, G. A., Watanabe, M., Todd, A. J. (2023) Antibodies against the gastrin-releasing peptide precursor pro-gastrin-releasing peptide reveal its expression in the mouse spinal dorsal horn. Neuroscience, 510, pp. 60-71. (doi: 10.1016/j.neuroscience.2022.12.023)

Ronzano, R., Skarlatou, S., Barriga, B. K., Bannatyne, B. A., Bhumbra, G. S., Foster, J. D., Moore, J. D., Lancelin, C., Pocratsky, A. M., Özyurt, M. G., Smith, C. C., Todd, A. J., Maxwell, D. J., Murray, A. J., Pfaff, S. L., Brownstone, R. M., Zampieri, N., Beato, M. (2023) Spinal premotor interneurons controlling antagonistic muscles are spatially intermingled. eLife, 11, (doi: 10.7554/elife.81976)

Visocky, V., Morris, B. J., Dunlop, J., Brandon, N., Sakata, S., Pratt, J. A. (2023) Site-specific inhibition of the thalamic reticular nucleus induces distinct modulations in sleep architecture. European Journal of Neuroscience, (doi: 10.1111/ejn.15908)

Polgár, E., Dickie, A. C., Gutierrez-Mecinas, M., Bell, A. M., Boyle, K. A., Quillet, R., Rashid, E. A., Clark, R. A., German, M. T., Watanabe, M., Riddell, J. S., Todd, A. J. (2023) Grpr expression defines a population of superficial dorsal horn vertical cells that have a role in both itch and pain. Pain, 164, pp. 149-170. (doi: 10.1097/j.pain.0000000000002677)

2022

Kwon, J., Arsenis, C., Suessmilch, M., McColl, A., Cavanagh, J., Morris, B. J. (2022) Differential effects of toll-like receptor activation and differential mediation by MAP kinases of immune responses in microglial cells. Cellular and Molecular Neurobiology, 42, pp. 2655-2671. (doi: 10.1007/s10571-021-01127-x)

Middleton, S. J., Perini, I., Andreas C., T., Weir, G. A., McCann, K., Barry, A. M., Marshall, A., Lee, M., Mayo, L. M., Bohic, M., Baskozos, G., Morrison, I., Löken, L. S., McIntyre, S., Nagi, S. S., Staud, R., Sehlstedt, I., Johnson, R. D., Wessberg, J., Wood, J. N., Woods, C. G., Moqrich, A., Olausson, H., Bennett, D. L. (2022) Nav1.7 is required for normal C-low threshold mechanoreceptor function in humans and mice. Brain, 145, pp. 3637-3653. (doi: 10.1093/brain/awab482)

Sanders, Z. B., Fleming, M. K., Smejka, T., Marzolla, M. C., Zich, C., Rieger, S. W., Lührs, M., Goebel, R., Sampaio-Baptista, C., Johansen-Berg, H. (2022) Self-modulation of motor cortex activity after stroke: a randomized controlled trial. Brain, 145, pp. 3391-3404. (doi: 10.1093/brain/awac239)

Warwick, C., Salsovic, J., Hachisuka, J., Smith, K. M., Sheahan, T. D., Chen, H., Ibinson, J., Koerber, H. R., Ross, S. E. (2022) Cell type-specific calcium imaging of central sensitization in mouse dorsal horn. Nature Communications, 13, (doi: 10.1038/s41467-022-32608-2)

Trendafilova, T. et al. (2022) Sodium-calcium exchanger-3 regulates pain "wind-up": from human psychophysics to spinal mechanisms. Neuron, 110, pp. 2571-2587.e13. (doi: 10.1016/j.neuron.2022.05.017)

Craig, M. T., Witton, J. (2022) A cellular switchboard in memory circuits. Science, 377, pp. 262-263. (doi: 10.1126/science.add2681)

Gradwell, M. A., Smith, K. M., Dayas, C. V., Smith, D. W., Hughes, D. I., Callister, R. J., Graham, B. A. (2022) Altered intrinsic properties and inhibitory connectivity in aged parvalbumin-expressing dorsal horn neurons. Frontiers in Neural Circuits, 16, (doi: 10.3389/fncir.2022.834173)

Baruchin, L. J., Ghezzi, F., Kohl, M. M., Butt, S. J.B. (2022) Contribution of interneuron subtype-specific GABAergic signalling to emergent sensory processing in mouse somatosensory whisker barrel cortex. Cerebral Cortex, 32, pp. 2538-2554. (doi: 10.1093/cercor/bhab363)

Williams, J. A., Huesa, C., Windmill, J. F. C., Purcell, M., Reid, S., Coupaud, S., Riddell, J. S. (2022) Spatiotemporal responses of trabecular and cortical bone to complete spinal cord injury in skeletally mature rats. Bone Reports, 16, (doi: 10.1016/j.bonr.2022.101592)

Hughes, D. I., Todd, A. J. (2022) Visualizing synaptic connectivity using confocal and electron microscopy: neuroanatomical approaches to define spinal circuits. Humana

Willis, A., Pratt, J. A., Morris, B. J. (2022) Enzymatic degradation of cortical perineuronal nets reverses GABAergic interneuron maturation. Molecular Neurobiology, 59, pp. 2874-2893. (doi: 10.1007/s12035-022-02772-z)

El Khoueiry, C., Alba-Delgado, C., Antri, M., Gutierrez-Mecinas, M., Todd, A. J., Artola, A., Dallel, R. (2022) GABAA and glycine receptor-mediated inhibitory synaptic transmission onto adult rat lamina IIi PKCγ-interneurons: pharmacological but not anatomical specialization. Cells, 11, (doi: 10.3390/cells11081356)

Todd, A. J. (2022) An historical perspective: the second order neuron in the pain pathway. Frontiers in Pain Research, 3, (doi: 10.3389/fpain.2022.845211)

Barki, N., Bolognini, D., Börjesson, U., Jenkins, L., Riddell, J., Hughes, D. I., Ulven, T., Hudson, B. D., Rexen Ulven, E., Dekker, N., Tobin, A. B., Milligan, G. (2022) Chemogenetics defines the roles of short chain fatty acid receptors within the gut-brain axis. eLife, 11, (doi: 10.7554/eLife.73777)

Gradwell, M. A., Boyle, K. A., Browne, T. J., Bell, A. M., Leonardo, J., Peralta Reyes, F. S., Dickie, A. C., Smith, K. M., Callister, R. J., Dayas, C. V., Hughes, D. I., Graham, B. A. (2022) Diversity of inhibitory and excitatory parvalbumin interneuron circuits in the dorsal horn. Pain, 163, pp. e432-e452. (doi: 10.1097/j.pain.0000000000002422)

Openshaw, R. L., Pratt, J. A., Morris, B. J. (2022) The schizophrenia risk gene Map2k7 regulates responding in a novel contingency-shifting rodent touchscreen gambling task. Disease Models and Mechanisms, 15, (doi: 10.1242/dmm.049310)

Akam, T., Lustig, A., Rowland, J. M., Kapanaiah, S. K.T., Esteve-Agraz, J., Panniello, M., Márquez, C., Kohl, M. M., Kätzel, D., Costa, R. M., Walton, M. E. (2022) Open-source, Python-based, hardware and software for controlling behavioural neuroscience experiments. eLife, 11, (doi: 10.7554/eLife.67846)

Kókai, É., Alsulaimain, W. A.A., Dickie, A. C., Bell, A. M., Goffin, L., Watanabe, M., Gutierrez-Mecinas, M., Todd, A. J. (2022) Characterisation of deep dorsal horn projection neurons in the spinal cord of the Phox2a::Cre mouse line. Molecular Pain, 18, pp. 17448069221119614. (doi: 10.1177/17448069221119614)

2021

Kwon, J., Suessmilch, M., McColl, A., Cavanagh, J., Morris, B. J. (2021) Distinct trans-placental effects of maternal immune activation by TLR3 and TLR7 agonists: implications for schizophrenia risk. Scientific Reports, 11, (doi: 10.1038/s41598-021-03216-9)

Lazari, A., Salvan, P., Cottaar, M., Papp, D., Jens van der Werf, O., Johnstone, A., Sanders, Z.-B., Sampaio-Baptista, C., Eichert, N., Miyamoto, K., Winkler, A., Callaghan, M. F., Nichols, T. E., Stagg, C. J., Rushworth, M. F.S., Verhagen, L., Johansen-Berg, H. (2021) Reassessing associations between white matter and behaviour with multimodal microstructural imaging. Cortex, 145, pp. 187-200. (doi: 10.1016/j.cortex.2021.08.017)

Xiao, M.-F., Roh, S.-E., Zhou, J., Chien, C.-C., Lucey, B. P., Craig, M. T., Hayes, L. N., Coughlin, J. M., Leweke, F. M., Jia, M., Xu, D., Zhou, W., Talbot Jr, C. C., Arnold, D. B., Staley, M., Jiang, C., Reti, I. M., Sawa, A., Pelkey, K. A., McBain, C. J., Savonenko, A., Worley, P. F. (2021) A biomarker-authenticated model of schizophrenia implicating NPTX2 loss of function. Science Advances, 7, (doi: 10.1126/sciadv.abf6935)

Sampaio-Baptista, C., Neyedli, H. F., Sanders, Z.-B., Biosi, K., Havard, D., Huang, Y., Andersson, J. L.R., Lühr, M., Goebel, R., Johansen-Berg, H. (2021) fMRI neurofeedback in the motor system elicits bidirectional changes in activity and in white matter structure in the adult human brain. Cell Reports, 37, (doi: 10.1016/j.celrep.2021.109890)

Roesl, C., Evans, E. R., Dissanayake, K. N., Boczonadi, V., Jones, R. A., Jordan, G., Ledahawsky, L., Allen, G. C.C., Scott, M., Thomson, A., Wishart, T. M., Hughes, D. I., Mead, R. J., Shone, C. C., Slater, C. R., Gillingwater, T. H., Skehel, P. A., Ribchester, R. R. (2021) Confocal endomicroscopy of neuromuscular junctions stained with physiologically inert protein fragments of tetanus toxin. Biomolecules, 11, (doi: 10.3390/biom11101499)

Thomson, D. M., Mitchell, E. J., Openshaw, R. L., Pratt, J. A., Morris, B. J. (2021) Mice lacking melatonin MT2 receptors exhibit attentional deficits, anxiety and enhanced social interaction. Journal of Psychopharmacology, 35, pp. 1265-1276. (doi: 10.1177/02698811211032439)

Alsulaiman, W. A.A., Quillet, R., Bell, A. M., Dickie, A. C., Polgár, E., Boyle, K. A., Watanabe, M., Roome, R. B., Kania, A., Todd, A., Gutierrez-Mecinas, M. (2021) Characterisation of lamina I anterolateral system neurons that express Cre in a Phox2a-Cre mouse line. Scientific Reports, 11, (doi: 10.1038/s41598-021-97105-w)

Chisholm, K. I., Lo Re, L., Polgár, E., Gutierrez-Mecinas, M., Todd, A. J., McMahon, S. B. (2021) Encoding of cutaneous stimuli by lamina I projection neurons. Pain, 162, pp. 2405-2417. (doi: 10.1097/j.pain.0000000000002226)

Prince, L. Y., Tran, M. M., Grey, D., Saad, L., Chasiotis, H., Kwag, J., Kohl, M. M., Richards, B. A. (2021) Neocortical inhibitory interneuron subtypes are differentially attuned to synchrony- and rate-coded information. Communications Biology, 4, (doi: 10.1038/s42003-021-02437-y)

Chrysostomidou, L., Cooper, A. H., Weir, G. A. (2021) Cellular models of pain: new technologies and their potential to progress preclinical research. Neurobiology of Pain, 10, (doi: 10.1016/j.ynpai.2021.100063)

Warwick, C., Cassidy, C., Hachisuka, J., Wright, M. C., Baumbauer, K. M., Adelman, P. C., Lee, K. H., Smith, K. M., Sheahan, T. D., Ross, S. E., Koerber, H. R. (2021) MrgprdCre lineage neurons mediate optogenetic allodynia through an emergent polysynaptic circuit. Pain, 162, pp. 2120-2131. (doi: 10.1097/j.pain.0000000000002227)

Browne, T. J., Smith, K. M., Gradwell, M. A., Iredale, J. A., Dayas, C. V., Callister, R. J., Hughes, D. I., Graham, B. A. (2021) Spinoparabrachial projection neurons form distinct classes in the mouse dorsal horn. Pain, 162, pp. 1977-1994. (doi: 10.1097/j.pain.0000000000002194)

Nothnagel, N. D., Symon, A., Morgan, A. T., Huber, R., Riddell, J., Goense, J. (2021) VASO-fMRI with Nordic-PCA for laminar sensory testing at 7 Tesla.

Willis, A., Pratt, J. A., Morris, B. J. (2021) BDNF and JNK signalling modulate cortical interneuron and perineuronal net development: implications for schizophrenia-linked 16p11.2 duplication syndrome. Schizophrenia Bulletin, 47, pp. 812-826. (doi: 10.1093/schbul/sbaa139)

Nguyen, E., Lim, G., Ding, H., Hachisuka, J., Ko, M.-C., Ross, S. E. (2021) Morphine acts on spinal dynorphin neurons to cause itch through disinhibition. Science Translational Medicine, 13, (doi: 10.1126/scitranslmed.abc3774)

Barron, H. C., Mars, R. B., Dupret, D., Lerch, J., Sampaio-Baptista, C. (2021) Cross-species neuroscience: closing the explanatory gap. Philosophical Transactions of the Royal Society B: Biological Sciences, 376, (doi: 10.1098/rstb.2019.0633)

2020

Browne, T. J., Hughes, D. I., Dayas, C. V., Callister, R. J., Graham, B. A. (2020) Projection neuron axon collaterals in the dorsal horn: placing a new player in spinal cord pain processing. Frontiers in Physiology, 11, (doi: 10.3389/fphys.2020.560802)

Polgár, E., Bell, A. M., Gutierrez-Mecinas, M., Dickie, A. C., Akar, O., Costreie, M., Watanabe, M., Todd, A. J. (2020) Substance P-expressing neurons in the superficial dorsal horn of the mouse spinal cord: insights into their functions and their roles in synaptic circuits. Neuroscience, 450, pp. 113-125. (doi: 10.1016/j.neuroscience.2020.06.038)

Graham, B. A., Hughes, D. I. (2020) Defining populations of dorsal horn interneurons. Pain, 161, pp. 2434-2436. (doi: 10.1097/j.pain.0000000000002067)

Choi, S., Hachisuka, J., Brett, M. A., Magee, A. R., Omori, Y., Iqbal, N.-u.-A., Zhang, D., DeLisle, M. M., Wolfson, R. L., Bai, L., Santiago, C., Gong, S., Goulding, M., Heintz, N., Koerber, H. R., Ross, S. E., Ginty, D. D. (2020) Parallel ascending spinal pathways for affective touch and pain. Nature, 587, pp. 258-263. (doi: 10.1038/s41586-020-2860-1)

Bell, A. M., Gutierrez-Mecinas, M., Stevenson, A., Casas-Benito, A., Wildner, H., West, S. J., Watanabe, M., Todd, A. J. (2020) Expression of green fluorescent protein defines a specific population of lamina II excitatory interneurons in the GRP::eGFP mouse. Scientific Reports, 10, (doi: 10.1038/s41598-020-69711-7)

Mitchell, E. J., Thomson, D. M., Openshaw, R. L., Bristow, G. C., Dawson, N., Pratt, J. A., Morris, B. (2020) Drug-responsive autism phenotypes in the 16p11.2 deletion mouse model: a central role for gene-environment interactions. Scientific Reports, 10, (doi: 10.1038/s41598-020-69130-8)

Hosseinzadeh, S., Lindsay, S. L., Gallagher, A. G., Wellings, D. A., Riehle, M. O., Riddell, J. S., Barnett, S. C. (2020) A novel poly-ε-lysine based implant, Proliferate®, for promotion of CNS repair following spinal cord injury. Biomaterials Science, 8, pp. 3611-3627. (doi: 10.1039/D0BM00097C)

Tran, M., Braz, J. M., Hamel, K., Kuhn, J., Todd, A. J., Basbaum, A. I. (2020) Ablation of spinal cord estrogen receptor α-expressing interneurons reduces chemically induced modalities of pain and itch. Journal of Comparative Neurology, 528, pp. 1629-1643. (doi: 10.1002/cne.24847)

Hughes, D. I., Todd, A. (2020) Central nervous system targets: inhibitory interneurons in the spinal cord. Neurotherapeutics, 17, pp. 874-885. (doi: 10.1007/s13311-020-00936-0)

Baskozos, G., Sandy-Hindmarch, O., Clark, A. J., Windsor, K., Karlsson, P., Weir, G. A., McDermott, L. A., Burchall, J., Wiberg, A., Furniss, D., Bennett, D. L.H., Schmid, A. B. (2020) Molecular and cellular correlates of human nerve regeneration: ADCYAP1/PACAP enhance nerve outgrowth. Brain, 143, pp. 2009-2026. (doi: 10.1093/brain/awaa163)

Almarzooq, S., Kwon, J., Willis, A., Craig, J., Morris, B. J. (2020) Novel alternatively-spliced exons of the VRK2 gene in mouse brain and microglial cells. Molecular Biology Reports, 47, pp. 5127-5136. (doi: 10.1007/s11033-020-05584-3)

Madden, J.F., Davis, O.C., Boyle, K.A., Iredale, J.A., Browne, T.J., Callister, R.J., Smith, D.W., Jobling, P., Hughes, D.I., Graham, B.A. (2020) Functional and molecular analysis of proprioceptive sensory neuron excitability in mice. Frontiers in Molecular Neuroscience, 13, (doi: 10.3389/fnmol.2020.00036)

Jang, H. J., Chung, H., Rowland, J. M., Richards, B. A., Kohl, M. M., Kwag, J. (2020) Distinct roles of parvalbumin and somatostatin interneurons in gating the synchronization of spike-times in the neocortex. Science Advances, 6, (doi: 10.1126/sciadv.aay5333)

Bristow, G. C., Thomson, D. M., Openshaw, R. L., Mitchell, E. J., Pratt, J. A., Dawson, N., Morris, B. J. (2020) 16p11 duplication disrupts hippocampal-orbitofrontal-amygdala connectivity, revealing a neural circuit endophenotype for schizophrenia. Cell Reports, 31, (doi: 10.1016/j.celrep.2020.107536)

Browne, T. J., Gradwell, M. A., Iredale, J. A., Madden, J. F., Hughes, D. I., Callister, R. J., Dayas, C. V., Graham, B. A. (2020) Transgenic cross-referencing of inhibitory and excitatory interneuron populations to dissect neuronal heterogeneity in the dorsal horn. Frontiers in Molecular Neuroscience, 13, (doi: 10.3389/fnmol.2020.00032)

Chung, H., Park, K., Jang, H. J., Kohl, M. M., Kwag, J. (2020) Dissociation of somatostatin and parvalbumin interneurons circuit dysfunctions underlying hippocampal theta and gamma oscillations impaired by amyloid β oligomers in vivo. Brain Structure and Function, 225, pp. 935-954. (doi: 10.1007/s00429-020-02044-3)

Peirs, C., Dallel, R., Todd, A. J. (2020) Recent advances in our understanding of the organization of dorsal horn neuron populations and their contribution to cutaneous mechanical allodynia. Journal of Neural Transmission, 127, pp. 505-525. (doi: 10.1007/s00702-020-02159-1)

Sampaio-Baptista, C., Vallès, A., Khrapitchev, A. A., Akkermans, G., Winkler, A. M., Foxley, S., Sibson, N. R., Roberts, M., Miller, K., Diamond, M. E., Martens, G. J.M., De Weerd, P., Johansen-Berg, H. (2020) White matter structure and myelin-related gene expression alterations with experience in adult rats. Progress in Neurobiology, 187, (doi: 10.1016/j.pneurobio.2020.101770)

Park, K., Lee, J., Jang, H. J., Richards, B. A., Kohl, M. M., Kwag, J. (2020) Optogenetic activation of parvalbumin and somatostatin interneurons selectively restores theta-nested gamma oscillations and oscillation-induced spike timing-dependent long-term potentiation impaired by amyloid β oligomers. BMC Biology, 18, (doi: 10.1186/s12915-019-0732-7)

Openshaw, R., Thomson, D. M., Thompson, R., Penninger, J. M., Pratt, J. A., Morris, B. J., Dawson, N. (2020) Map2k7 haploinsufficiency induces brain imaging endophenotypes and behavioral phenotypes relevant to schizophrenia. Schizophrenia Bulletin, 46, pp. 211-223. (doi: 10.1093/schbul/sbz044)

Hachisuka, J., Koerber, H. R., Ross, S. E. (2020) Selective-cold output through a distinct subset of lamina I spinoparabrachial neurons. Pain, 161, pp. 185-194. (doi: 10.1097/j.pain.0000000000001710)

Maxwell, D. J., Soteropoulos, D. S. (2020) The mammalian spinal commissural system: properties and functions. Journal of Neurophysiology, 123, pp. 4-21. (doi: 10.1152/jn.00347.2019)

Bell, A. M., Polgár, E., Gutierrez-Mecinas, M., Boyle, K. A., Todd, A. J. (2020) Functional populations among interneurons in the dorsal horn. Academic Press

2019

Pettingill, P., Weir, G. A., Wei, T., Wu, Y., Flower, G., Lalic, T., Handel, A., Duggal, G., Chintawar, S., Cheung, J., Arunasalam, K., Couper, E., Haupt, L. M., Griffiths, L. R., Bassett, A., Cowley, S. A., Zader, M. Z. (2019) A causal role for TRESK loss of function in migraine mechanisms. Brain, 142, pp. 3852-3867. (doi: 10.1093/brain/awz342)

Calvo, M., Davies, A. J., Hébert, H. L., Weir, G. A., Chesler, E. J., Finnerup, N. B., Levitt, R. C., Smith, B. H., Neely, G. G., Costigan, M., Bennett, D. L. (2019) The genetics of neuropathic pain from model organisms to clinical application. Neuron, 104, pp. 637-653. (doi: 10.1016/j.neuron.2019.09.018)

Smith, K. M., Browne, T. J., Davis, O. C.,, Coyle, A., Boyle, K. A., Watanabe, M., Dickinson, S. A., Iredale, J. A., Gradwell, M. A., Jobling, P., Callister, R. J., Dayas, C. V., Hughes, D. I., Graham, B. A. (2019) Calretinin positive neurons form an excitatory amplifier network in the spinal cord dorsal horn. eLife, 8, (doi: 10.7554/eLife.49190)

Harold, A., Amako, Y., Hachisuka, J., Bai, Y., Li, M. Y., Kubat, L., Gravemeyer, J., Franks, J., Gibbs, J. R., Park, H. J., Ezhkova, E., Becker, J. C., Shuda, M. (2019) Conversion of Sox2-dependent Merkel cell carcinoma to a differentiated neuron-like phenotype by T antigen inhibition. Proceedings of the National Academy of Sciences of the United States of America, 116, pp. 20104-20114. (doi: 10.1073/pnas.1907154116)

Graham, B. A., Hughes, D. I. (2019) Rewards, perils and pitfalls of untangling spinal pain circuits. Current Opinion in Physiology, 11, pp. 35-41. (doi: 10.1016/j.cophys.2019.04.015)

Brunton, E. K., Silveira, C., Rosenberg, J., Schiefer, M. A., Riddell, J., Nazarpour, K. (2019) Temporal modulation of the response of sensory fibers to paired-pulse stimulation. IEEE Transactions on Neural Systems and Rehabilitation Engineering, 27, pp. 1676-1683. (doi: 10.1109/TNSRE.2019.2935813)

Gutierrez-Mecinas, M., Bell, A. M., Shepherd, F., Polgár, E., Watanabe, M., Furuta, T., Todd, A. J. (2019) Expression of cholecystokinin by neurons in mouse spinal dorsal horn. Journal of Comparative Neurology, 527, pp. 1857-1871. (doi: 10.1002/cne.24657)

Weir, G. A., Pettingill, P., Wu, Y., Duggal, G., Ilie, A.-S., Akerman, C. J., Cader, M. Z. (2019) The role of TRESK in discrete sensory neuron populations and somatosensory processing. Frontiers in Molecular Neuroscience, 12, (doi: 10.3389/fnmol.2019.00170)

Boyle, K. A., Gradwell, M. A., Yasaka, T., Dickie, A. C., Polgár, E., Ganley, R. P., Orr, D. P.H., Watanabe, M., Abraira, V. E., Kuehn, E. D., Zimmerman, A. L., Ginty, D. D., Callister, R. J., Graham, B. A., Hughes, D. I. (2019) Defining a spinal microcircuit that gates myelinated afferent input: implications for tactile allodynia. Cell Reports, 28, pp. 526-540.e6. (doi: 10.1016/j.celrep.2019.06.040)

McDermott, L. A., Weir, G. A., Themistocleous, A. C., Segerdahl, A. R., Blesneac, I., Basakozos, G., Clark, A. J., Millar, V., Peck, L. J., Ebner, D., Tracey, I., Serra, J., Bennett, D. L. (2019) Defining the functional role of NaV1.7 in human nociception. Neuron, 101, pp. 905-919.e8. (doi: 10.1016/j.neuron.2019.01.047)

Sampaio-Baptista, C., Diosi, K., Johnansen-Berg, H. (2019) Magnetic resonance techniques for imaging white matter. Humana Press

Pratt, J. A., Morris, B., Dawson, N. (2019) Deconstructing Schizophrenia: advances in preclinical models for biomarker identification. Springer

Gutierrez-Mecinas, M., Davis, O., Polgár, E., Shahzad, M., Navarro-Batista, K., Furuta, T., Watanabe, M., Hughes, D. I., Todd, A. J. (2019) Expression of calretinin among different neurochemical classes of interneuron in the superficial dorsal horn of the mouse spinal cord. Neuroscience, 398, pp. 171-181. (doi: 10.1016/j.neuroscience.2018.12.009)

Dickie, A. C., Bell, A. M., Iwagaki, N., Polgár, E., Gutierrez-Mecinas, M., Kelly, R., Lyon, H., Turnbull, K., West, S. J., Etlin, A., Braz, J., Watanabe, M., Bennett, D. L.H., Basbaum, A. I., Riddell, J. S., Todd, A. J. (2019) Morphological and functional properties distinguish the substance P and gastrin-releasing peptide subsets of excitatory interneuron in the spinal cord dorsal horn. Pain, 160, pp. 442-462. (doi: 10.1097/j.pain.0000000000001406)

Openshaw, R. L., Kwon, J., McColl, A., Penninger, J. M., Cavanagh, J., Pratt, J. A., Morris, B. J. (2019) JNK signalling mediates aspects of maternal immune activation: importance of maternal genotype in relation to schizophrenia risk. Journal of Neuroinflammation, 16, (doi: 10.1186/s12974-019-1408-5)

Chong, E. Z., Panniello, M., Barreiros, I., Kohl, M. M., Booth, M. J. (2019) Quasi-simultaneous multiplane calcium imaging of neuronal circuits. Biomedical Optics Express, 10, pp. 267-282. (doi: 10.1364/BOE.10.000267)

Gutierrez-Mecinas, M., Bell, A., Polgár, E., Watanabe, M., Todd, A. J. (2019) Expression of neuropeptide FF defines a population of excitatory interneurons in the superficial dorsal horn of the mouse spinal cord that respond to noxious and pruritic stimuli. Neuroscience, 416, pp. 281-293. (doi: 10.1016/j.neuroscience.2019.08.013)

2018

Bautzova, T., Hockley, J. R.F., Perez-Berezo, T., Pujo, J., Tranter, M. M., Desormeaux, C., Barbaro, M. R., Basso, L., Le Faouder, P., Rolland, C., Malapert, P., Moqrich, A., Eutamene, H., Denadai-Souza, A., Vergnolle, N., Smith, E. S. J., Hughes, D. I., Barbara, G., Dietrich, G., Bulmer, D. C., Cenac, N. (2018) 5-oxoETE triggers nociception in constipation-predominant irritable bowel syndrome through MAS-related G protein–coupled receptor D. Science Signaling, 11, (doi: 10.1126/scisignal.aal2171)

Willis, A., Pratt, J. A., Morris, B. J. (2018) Distortion of protein analysis in primary neuronal cultures by serum albumin from culture medium: a methodological approach to improve target protein quantification. Journal of Neuroscience Methods, 308, pp. 1-5. (doi: 10.1016/j.jneumeth.2018.07.002)

Lazari, A., Koudelka, S., Sampaio-Baptista, C. (2018) Experience-related reductions of myelin and axon diameter in adulthood. Journal of Neurophysiology, 120, pp. 1772-1775. (doi: 10.1152/jn.00070.2018)

Snyder, L. M. et al. (2018) Kappa opioid receptor distribution and function in primary afferents. Neuron, 99, pp. 1274-1288.e6. (doi: 10.1016/j.neuron.2018.08.044)

Sheahan, T. D., Hachisuka, J., Ross, S. E. (2018) Small RNAs, but sizable itch: TRPA1 activation by an extracellular microRNA. Neuron, 99, pp. 421-422. (doi: 10.1016/j.neuron.2018.07.040)

Hachisuka, J., Omori, Y., Chiang, M. C., Gold, M. S., Koerber, H. R., Ross, S. E. (2018) Wind-up in lamina I spinoparabrachial neurons. Pain, 159, pp. 1484-1493. (doi: 10.1097/j.pain.0000000000001229)

Khadria, A., Fleischhauer, J., Boczarow, I., Wilkinson, J. D., Kohl, M. M., Anderson, H. L. (2018) Porphyrin dyes for nonlinear optical imaging of live cells. iScience, 4, pp. 153-163. (doi: 10.1016/j.isci.2018.05.015)

Huang, J., Polgár, E., Solinski, H. J., Mishra, S. K., Tseng, P.-Y., Iwagaki, N., Boyle, K. A., Dickie, A. C., Kriegbaum, M. C., Wildner, H., Zeilhofer, H. U., Watanabe, M., Riddell, J. S., Todd, A. J., Hoon, M. A. (2018) Author correction: circuit dissection of the role of somatostatin in itch and pain. Nature Neuroscience, 21, pp. 894. (doi: 10.1038/s41593-018-0149-6)

Gutierrez-Mecinas, M., Polgár, E., Bell, A. M., Hérau, M., Todd, A. J. (2018) Substance P-expressing excitatory interneurons in the mouse superficial dorsal horn provide a propriospinal input to the lateral spinal nucleus. Brain Structure and Function, 223, pp. 2377-2392. (doi: 10.1007/s00429-018-1629-x)

Neyedli, H. F., Sampaio-Baptista, C., Kirkman, M. A., Havard, D., Lührs, M., Ramsden, K., Flitney, D. D., Clare, S., Goebel, R., Johansen-Berg, H. (2018) Increasing lateralized motor activity in younger and older adults using Real-time fMRI during executed movements. Neuroscience, 378, pp. 165-174. (doi: 10.1016/j.neuroscience.2017.02.010)

Strnadel, J. et al. (2018) Survival of syngeneic and allogeneic iPSC–derived neural precursors after spinal grafting in minipigs. Science Translational Medicine, 10, (doi: 10.1126/scitranslmed.aam6651)

Huang, J., Polgár, E., Solinski, H. J., Mishra, S. K., Tseng, P.-Y., Iwagaki, N., Boyle, K. A., Dickie, A. C., Kriegbaum, M. C., Wildner, H., Zeilhofer, H. U., Watanabe, M., Riddell, J. S., Todd, A. J., Hoon, M. A. (2018) Circuit dissection of the role of somatostatin in itch and pain. Nature Neuroscience, 21, pp. 707-716. (doi: 10.1038/s41593-018-0119-z)

Huang, H., Krogsrud, S. K., Fjell, A. M., Tamnes, C. K., Grydeland, H., Due-Tønnessen, P., Bjørnerud, A., Sampaio-Baptista, C., Andersson, J., Johansen-Berg, H., Walhovd, K. B. (2018) Development of white matter microstructure in relation to verbal and visuospatial working memory—a longitudinal study. PLoS ONE, 13, (doi: 10.1371/journal.pone.0195540)

Hachisuka, J., Chiang, M. C., Ross, S. E. (2018) Itch and neuropathic itch. Pain, 159, pp. 603-609. (doi: 10.1097/j.pain.0000000000001141)

Heath, F., Hurley, S. A., Johansen-Berg, H., Sampaio-Baptista, C. (2018) Advances in noninvasive myelin imaging. Developmental Neurobiology, 78, pp. 136-151. (doi: 10.1002/dneu.22552)

Dawes, J. M. et al. (2018) Immune or genetic-mediated disruption of CASPR2 causes pain hypersensitivity due to enhanced primary afferent excitability. Neuron, 97, pp. 806-822.e10. (doi: 10.1016/j.neuron.2018.01.033)

Sampaio-Baptista, C., Sanders, Z.-B., Johansen-Berg, H. (2018) Structural plasticity in adulthood with motor learning and stroke rehabilitation. Annual Review of Neuroscience, 41, pp. 25-40. (doi: 10.1146/annurev-neuro-080317-062015)

2017

Sampaio-Baptista, C., Johansen-Berg, H. (2017) White matter plasticity in the adult brain. Neuron, 96, pp. 1239-1251. (doi: 10.1016/j.neuron.2017.11.026)

Gradwell, M.A., Boyle, K.A., Callister, R.J., Hughes, D.I., Graham, B.A. (2017) Heteromeric α/β glycine receptors regulate excitability in parvalbumin-expressing dorsal horn neurons through phasic and tonic glycinergic inhibition. Journal of Physiology, 595, pp. 7185-7202. (doi: 10.1113/JP274926)

Kaller, M. S., Lazari, A., Blanco-Duque, C., Sampaio-Baptista, C., Johansen-Berg, H. (2017) Myelin plasticity and behaviour — connecting the dots. Current Opinion in Neurobiology, 47, pp. 86-92. (doi: 10.1016/j.conb.2017.09.014)

Li, J., Han, W., Pelkey, K. A., Duan, J., Mao, X., Wang, Y.-X., Craig, M. T., Dong, L., Petralia, R. S., McBain, C. J., Lu, W. (2017) Molecular dissection of Neuroligin 2 and Slitrk3 reveals an essential framework for GABAergic synapse development. Neuron, 96, pp. 808-826.e8. (doi: 10.1016/j.neuron.2017.10.003)

Boyle, K. A., Gutierrez-Mecinas, M., Polgár, E., Mooney, N., O'Connor, E., Furuta, T., Watanabe, M., Todd, A. J. (2017) A quantitative study of neurochemically-defined populations of inhibitory interneurons in the superficial dorsal horn of the mouse spinal cord. Neuroscience, 363, pp. 120-133. (doi: 10.1016/j.neuroscience.2017.08.044)

Pelkey, K. A., Chittajallu, R., Craig, M. T., Tricoire, L., Wester, J. C., McBain, C. J. (2017) Hippocampal GABAergic inhibitory interneurons. Physiological Reviews, 97, pp. 1619-1747. (doi: 10.1152/physrev.00007.2017)

Weir, G. A., Middleton, S. J., Clark, A. J., Daniel, T., Khovanov, N., McMahon, S. B., Bennett, D. L. (2017) Using an engineered glutamate-gated chloride channel to silence sensory neurons and treat neuropathic pain at the source. Brain, 140, pp. 2570-2585. (doi: 10.1093/brain/awx201)

Tae, H.-S., Smith, K. M., Phillips, A. M., Boyle, K. A., Li, M., Forster, I. C., Hatch, R. J., Richardson, R., Hughes, D. I., Graham, B. A., Petrou, S., Reid, C. A. (2017) Gabapentin modulates HCN4 channel voltage-dependence. Frontiers in Pharmacology, 8, (doi: 10.3389/fphar.2017.00554)

Chittajallu, R., Wester, J.C., Craig, M.T., Barksdale, E., Yuan, X.Q., Akgül, G., Fang, C., Collins, D., Hunt, S., Pelkey, K.A., McBain, C.J. (2017) Afferent specific role of NMDA receptors for the circuit integration of hippocampal neurogliaform cells. Nature Communications, 8, (doi: 10.1038/s41467-017-00218-y)

Kohl, M. M., Kätzel, D. (2017) Optogenetic mapping of neuronal connections and their plasticity. Cambridge University Press,

Xiao, M.-F., Xu, D., Craig, M. T., Pelkey, K. A., Chien, C.-C., Shi, Y., Zhang, J., Resnick, S., Pletnikova, O., Salmon, D., Brewer, J., Edland, S., Wegiel, J., Tycko, B., Savonenko, A., Reeves, R. H., Troncoso, J. C., McBain, C. J., Galasko, D., Worley, P. F. (2017) NPTX2 and cognitive dysfunction in Alzheimer’s disease. eLife, 6, (doi: 10.7554/eLife.23798)

Gutierrez-Mecinas, M., Bell, A. M., Marin, A., Taylor, R., Boyle, K. A., Furuta, T., Watanabe, M., Polgár, E., Todd, A. J. (2017) Preprotachykinin A (PPTA) is expressed by a distinct population of excitatory neurons in the mouse superficial spinal dorsal horn including cells that respond to noxious and pruritic stimuli. Pain, 158, pp. 440-456. (doi: 10.1097/j.pain.0000000000000778)

Brown, T. G., Cogswell, C. J., Wilson, T., Chong, E. Z., Barreiros, I., Li, B., Kohl, M. M., Booth, M. J. (2017) Fast Multiplane Functional Imaging Combining Acousto-optic Switching and Remote Focusing. (doi: 10.1117/12.2251476)

Flynn, J. R., Conn, V., Boyle, K., Hughes, D. I., Watanabe, M., Velasquez, T., Goulding, M. D., Callister, R. J., Graham, B. A. (2017) Anatomical and molecular properties of long descending propriospinal neurons in mice. Frontiers in Neuroanatomy, 11, (doi: 10.3389/fnana.2017.00005)

Hulme, C.H., Brown, S.J., Fuller, H.R., Riddell, J., Osman, A., Chowdhury, J., Kumar, N., Johnson, W.E., Wright, K.T. (2017) The developing landscape of diagnostic and prognostic biomarkers for spinal cord injury in cerebrospinal fluid and blood. Spinal Cord, 55, pp. 114-125. (doi: 10.1038/sc.2016.174)

Lindsay, S. L., Toft, A., Griffin, J., Emraja, A. M.M., Barnett, S. C., Riddell, J. (2017) Human olfactory mesenchymal stromal cell transplants promote remyelination and earlier improvement in gait co-ordination after spinal cord injury. Glia, 65, pp. 639-656. (doi: 10.1002/glia.23117)

Pratt, J., Dawson, N., Morris, B. J., Grent-'T-Jong, T., Roux, F., Uhlhaas, P. J. (2017) Thalamo-cortical communication, glutamatergic neurotransmission and neural oscillations: a unique window into the origins of ScZ? Schizophrenia Research, 180, pp. 4-12. (doi: 10.1016/j.schres.2016.05.013)

Abraira, V. E., Kuehn, E. D., Chirila, A. M., Springel, M. W., Toliver, A. A., Zimmerman, A. L., Orefice, L. L., Boyle, K. A., Bai, L., Song, B. J., Bashista, K. A., O'Neill, T. G., Zhou, J., Tsan, C., Hoynoski, J., Rutlin, M., Kus, L., Niederkofler, V., Watanabe, M., Dymecki, S. M., Nelson, S. B., Heintz, N., Hughes, D. I., Ginty, D. D. (2017) The cellular and synaptic architecture of the mechanosensory dorsal horn. Cell, 168, pp. 295-310.e19. (doi: 10.1016/j.cell.2016.12.010)

Todd, A. J. (2017) Identifying functional populations among the interneurons in laminae I-III of the spinal dorsal horn. Molecular Pain, 13, pp. 1-19. (doi: 10.1177/1744806917693003)

Openshaw, R. L., Thomson, D.M., Penninger, J.M., Pratt, J.A., Morris, B.J. (2017) Mice haploinsufficient for Map2k7, a gene involved in neurodevelopment and risk for schizophrenia, show impaired attention, a vigilance decrement deficit and unstable cognitive processing in an attentional task: impact of minocycline. Psychopharmacology, 234, pp. 293-305. (doi: 10.1007/s00213-016-4463-y)

El-Gaby, M., Kohl, M. M., Paulsen, O. (2017) Optogenetic methods to study lateralized synaptic function. Humana Press

O'Neill, P., Lindsay, S. L., Pantiru, A., Guimond, S. E., Fagoe, N., Verhaagen, J., Turnbull, J. E., Riddell, J. S., Barnett, S. C. (2017) Sulfatase mediated manipulation of the astrocyte-Schwann cell interface. Glia, 65, pp. 19-33. (doi: 10.1002/glia.23047)

2016

Hachisuka, J., Baumbauer, K. M., Omori, Y., Snyder, L. M., Koerber, H. R., Ross, S. E. (2016) Semi-intact ex vivo approach to investigate spinal somatosensory circuits. eLife, 5, (doi: 10.7554/eLife.22866)

Gutierrez-Mecinas, M., Furuta, T., Watanabe, M., Todd, A. J. (2016) A quantitative study of neurochemically-defined excitatory interneuron populations in laminae I-III of the mouse spinal cord. Molecular Pain, 12, pp. 1-18. (doi: 10.1177/1744806916629065)

Bell, A. M., Gutierrez-Mecinas, M., Polgár, E., Todd, A. J. (2016) Spinal neurons that contain gastrin-releasing peptide seldom express Fos or phosphorylate extracellular signal-regulated kinases in response to intradermal chloroquine. Molecular Pain, 12, pp. 1-9. (doi: 10.1177/1744806916649602)

Gutierrez-Mecinas, M., Kuehn, E. D., Abraira, V. E., Polgár, E., Watanabe, M., Todd, A. J. (2016) Immunostaining for Homer reveals the majority of excitatory synapses in laminae I-III of the mouse spinal dorsal horn. Neuroscience, 329, pp. 171-181. (doi: 10.1016/j.neuroscience.2016.05.009)

Dafinca, R., Scaber, J., Ababneh, N.'a., Lalic, T., Weir, G., Christian, H., Vowles, J., Douglas, A. G.L., Fletcher-Jones, A., Browne, C., Nakanishi, M., Turner, M. R., Wade-Martins, R., Cowley, S. A., Talbot, K. (2016) C9orf72 hexanucleotide expansions are associated with altered endoplasmic reticulum calcium homeostasis and stress granule formation in induced pluripotent stem cell-derived neurons from patients with amyotrophic lateral sclerosis and frontotemporal dementia. Stem Cells, 34, pp. 2063-2078. (doi: 10.1002/stem.2388)

Smith, K.M., Boyle, K.A., Mustapa, M., Jobling, P., Callister, R.J., Hughes, D.I., Graham, B.A. (2016) Distinct forms of synaptic inhibition and neuromodulation regulate calretinin positive neuron excitability in the spinal cord dorsal horn. Neuroscience, 326, pp. 10-21. (doi: 10.1016/j.neuroscience.2016.03.058)

Chiang, M.C., Hachisuka, J., Todd, A.J., Ross, S.E. (2016) Erratum. Pain, 157, pp. 992. (doi: 10.1097/j.pain.0000000000000552)

Iwagaki, N., Ganley, R. P., Dickie, A., Polgár, E., Hughes, D. I., Del Rio, P., Revina, Y., Watanabe, M., Todd, A. J., Riddell, J. S. (2016) A combined electrophysiological and morphological study of NPY-expressing inhibitory interneurons in the spinal dorsal horn of the mouse. Pain, 157, pp. 598-612. (doi: 10.1097/j.pain.0000000000000407)

Chiang, M. C., Hachisuka, J., Todd, A. J., Ross, S. E. (2016) Insight into B5-I spinal interneurons and their role in the inhibition of itch and pain. Pain, 157, pp. 544-545. (doi: 10.1097/j.pain.0000000000000474)

Sleigh, J. N., Weir, G. A., Schiavo, G. (2016) A simple, step-by-step dissection protocol for the rapid isolation of mouse dorsal root ganglia. BMC Research Notes, 9, (doi: 10.1186/s13104-016-1915-8)

Krogsrud, S. K., Fjell, A. M., Tamnes, C. K., Grydeland, H., Mork, L., Due-Tønnessen, P., Bjørnerud, A., Sampaio-Baptista, C., Andersson, J., Johansen-Berg, H., Walhovd, K. B. (2016) Changes in white matter microstructure in the developing brain—a longitudinal diffusion tensor imaging study of children from 4 to 11 years of age. NeuroImage, 124, pp. 473-486. (doi: 10.1016/j.neuroimage.2015.09.017)

Todd, A. J. (2016) Anatomy of pain pathways. Wiley Blackwell

Mitchell, E. J., McCallum, S., Dewar, D., Maxwell, D. (2016) Corticospinal and reticulospinal contacts on cervical commissural and long descending propriospinal neurons in the adult rat spinal cord; evidence for powerful reticulospinal connections. PLoS ONE, 11, (doi: 10.1371/journal.pone.0152094)

Mitchell, E. J., Dewar, D., Maxwell, D. J. (2016) Is remodelling of corticospinal tract terminations originating in the intact hemisphere associated with recovery following transient ischaemic stroke in the rat? PLoS ONE, 11, (doi: 10.1371/journal.pone.0152176)

Kido-Nakahara, M., Hachisuka, J., Nakahara, T., Furusyo, N., Shimoda, S., Koto, K., Furue, M. (2016) Pruritus in chronic liver disease: a questionnaire survey in 71 patients. Nishi Nihon Hifuka, 78, pp. 655-659. (doi: 10.2336/nishinihonhifu.78.655)

2015

Tadros, M. A., Lim, R., Hughes, D. I., Brichta, A. M., Callister, R. J. (2015) Electrical maturation of spinal neurons in the human fetus: comparison of ventral and dorsal horn. Journal of Neurophysiology, 114, pp. 2661-2671. (doi: 10.1152/jn.00682.2015)

Smith, K.M., Boyle, K.A., Madden, J.F., Dickinson, S.A., Jobling, P., Callister, R.J., Hughes, D.I., Graham, B.A. (2015) Functional heterogeneity of calretinin-expressing neurons in the mouse superficial dorsal horn: implications for spinal pain processing. Journal of Physiology, 593, pp. 4319-4339. (doi: 10.1113/JP270855)

Nakahara, T., Mitoma, C., Hashimoto-Hachiya, A., Takahara, M., Tsuji, G., Uchi, H., Yan, X., Hachisuka, J., Chiba, T., Esaki, H., Kido-Nakahara, M., Furue, M. (2015) Antioxidant Opuntia ficus-indica extract activates AHR-NRF2 signaling and upregulates filaggrin and loricrin expression in human keratinocytes. Journal of Medicinal Food, 18, pp. 1143-1149. (doi: 10.1089/jmf.2014.3396)

Baumbauer, K. M., DeBerry, J. J., Adelman, P. C., Miller, R. H., Hachisuka, J., Lee, K. H., Ross, S. E., Koerber, H. R., Davis, B. M., Albers, K. M. (2015) Keratinocytes can modulate and directly initiate nociceptive responses. eLife, 4, (doi: 10.7554/eLife.09674)

Shiratori-Hayashi, M., Koga, K., Tozaki-Saitoh, H., Kohro, Y., Toyonaga, H., Yamaguchi, C., Hasegawa, A., Nakahara, T., Hachisuka, J., Akira, S., Okano, H., Furue, M., Inoue, K., Tsuda, M. (2015) STAT3-dependent reactive astrogliosis in the spinal dorsal horn underlies chronic itch. Nature Medicine, 21, pp. 927-931. (doi: 10.1038/nm.3912)

Craig, M. T., McBain, C. J. (2015) Navigating the circuitry of the brain's GPS system: future challenges for neurophysiologists. Hippocampus, 25, pp. 736-743. (doi: 10.1002/hipo.22456)

Ganley, R. P., Iwagaki, N., Del Rio, P., Baseer, N., Dickie, A. C., Boyle, K. A., Polgár, E., Watanabe, M., Abraira, V. E., Zimmerman, A., Riddell, J. S., Todd, A. J. (2015) Inhibitory interneurons that express GFP in the PrP-GFP mouse spinal cord are morphologically heterogeneous, innervated by several classes of primary afferent and include lamina I projection neurons among their postsynaptic targets. Journal of Neuroscience, 35, pp. 7626-7642. (doi: 10.1523/JNEUROSCI.0406-15.2015)

Huma, Z., Ireland, K., Maxwell, D. J. (2015) The spino–bulbar–cerebellar pathway: activation of neurons projecting to the lateral reticular nucleus in the rat in response to noxious mechanical stimuli. Neuroscience Letters, 591, pp. 197-201. (doi: 10.1016/j.neulet.2015.02.047)

Pelkey, K. A., Barksdale, E., Craig, M. T., Yuan, X., Sukumaran, M., Vargish, G. A., Mitchell, R. M., Wyeth, M. S., Petralia, R. S., Chittajallu, R., Karlsson, R.-M., Cameron, H. A., Murata, Y., Colonnese, M. T., Worley, P. F., McBain, C. J. (2015) Pentraxins coordinate excitatory synapse maturation and circuit integration of parvalbumin interneurons. Neuron, 85, pp. 1257-1272. (doi: 10.1016/j.neuron.2015.02.020)

Craig, M. T., McBain, C. J. (2015) Fast gamma oscillations are generated intrinsically in CA1 without the involvement of fast-spiking basket cells. Journal of Neuroscience, 35, pp. 3616-3624. (doi: 10.1523/JNEUROSCI.4166-14.2015)

Sampaio-Baptista, C., Filippini, N., Stagg, C. J., Near, J., Scholz, J., Johansen-Berg, H. (2015) Changes in functional connectivity and GABA levels with long-term motor learning. NeuroImage, 106, pp. 15-20. (doi: 10.1016/j.neuroimage.2014.11.032)

Dawson, N., Morris, B. J., Pratt, J. A. (2015) Functional brain connectivity phenotypes for schizophrenia drug discovery. Journal of Psychopharmacology, 29, pp. 169-177. (doi: 10.1177/0269881114563635)

Dawson, N., Kurihara, M., Thomson, D.M., Winchester, C.L., McVie, A., Hedde, J.R., Randall, A.D., Shen, S., Seymour, P.A., Hughes, Z.A., Dunlop, J., Brown, J.T., Brandon, N.J., Morris, B.J., Pratt, J.A. (2015) Altered functional brain network connectivity and glutamate system function in transgenic mice expressing truncated Disrupted-in-Schizophrenia 1. Translational Psychiatry, 5, (doi: 10.1038/tp.2015.60)

Todd, A. J. (2015) Plasticity of inhibition in the spinal cord. Springer

Cameron, D., Gutierrez-Mecinas, M., Gomez-Lima, M., Watanabe, M., Polgár, E., Todd, A. J. (2015) The organisation of spinoparabrachial neurons in the mouse. Pain, 156, pp. 2061-2071. (doi: 10.1097/j.pain.0000000000000270)

Huma, Z., Maxwell, D. J. (2015) The spino-bulbar-cerebellar pathway: organization and neurochemical properties of spinal cells that project to the lateral reticular nucleus in the rat. Frontiers in Neuroanatomy, 9, (doi: 10.3389/fnana.2015.00001)

Pratt, J. A., Morris, B. J. (2015) The thalamic reticular nucleus: a functional hub for thalamocortical network dysfunction in schizophrenia and a target for drug discovery. Journal of Psychopharmacology, 29, pp. 127-137. (doi: 10.1177/0269881114565805)

2014

Gadalla, K. K.E., Ross, P. D., Riddell, J. S., Bailey, M. E.S., Cobb, S. R. (2014) Gait analysis in a Mecp2 knockout mouse model of Rett syndrome reveals early-onset and progressive motor deficits. PLoS ONE, 9, (doi: 10.1371/journal.pone.0112889)

Morris, B. J., Pratt, J. A. (2014) Novel treatment strategies for schizophrenia from improved understanding of genetic risk. Clinical Genetics, 86, pp. 401-411. (doi: 10.1111/cge.12485)

Baseer, N., Al-Baloushi, A. S., Watanabe, M., Shehab, S. A.S., Todd, A. J. (2014) Selective innervation of NK1 receptor–lacking lamina I spinoparabrachial neurons by presumed nonpeptidergic Aδ nociceptors in the rat. Pain, 155, pp. 2291-2300. (doi: 10.1016/j.pain.2014.08.023)

Shipton, O. A., El-Gaby, M., Apergis-Schoute, J., Deisseroth, K., Bannerman, D. M., Paulsen, O., Kohl, M. M. (2014) Left–right dissociation of hippocampal memory processes in mice. Proceedings of the National Academy of Sciences of the United States of America, 111, pp. 15238-15243. (doi: 10.1073/pnas.1405648111)

Bhumbra, G. S., Bannatyne, B. A., Watanabe, M., Todd, A. J., Maxwell, D. J., Beato, M. (2014) The recurrent case for the Renshaw cell. Journal of Neuroscience, 34, pp. 12919-12932. (doi: 10.1523/JNEUROSCI.0199-14.2014)

Hachisuka, J., Ross, S. E. (2014) Understanding the switch from pain-to-itch in dermatitis. Neuroscience Letters, 579, pp. 188-189. (doi: 10.1016/j.neulet.2014.05.018)

Sampaio-Baptista, C., Scholz, J., Jenkinson, M., Thomas, A. G., Filippini, N., Smit, G., Douaud, G., Johansen-Berg, H. (2014) Gray matter volume is associated with rate of subsequent skill learning after a long term training intervention. NeuroImage, 96, pp. 158-166. (doi: 10.1016/j.neuroimage.2014.03.056)

Winchester, C. L., Pratt, J. A., Morris, B. J. (2014) Risk genes for schizophrenia: translational opportunities for drug discovery. Pharmacology and Therapeutics, 143, pp. 34-50. (doi: 10.1016/j.pharmthera.2014.02.003)

Dawson, N., McDonald, M., Higham, D. J., Morris, B. J., Pratt, J. A. (2014) Subanesthetic ketamine treatment promotes abnormal interactions between neural subsystems and alters the properties of functional brain networks. Neuropsychopharmacology, 39, pp. 1786-1798. (doi: 10.1038/npp.2014.26)

Craig, M. T., McBain, C. J. (2014) The emerging role of GABAB receptors as regulators of network dynamics: fast actions from a 'slow' receptor? Current Opinion in Neurobiology, 26, pp. 15-21. (doi: 10.1016/j.conb.2013.10.002)

Kardon, A. P., Polgár, E., Hachisuka, J., Snyder, L. M., Cameron, D., Savage, S., Cai, X., Karnup, S., Fan, C. R., Hemenway, G. M., Bernard, C. S., Schwartz, E. S., Nagase, H., Schwarzer, C., Watanabe, M., Furuta, T., Kaneko, T., Koerber, H. R., Todd, A. J., Ross, S. E. (2014) Dynorphin acts as a neuromodulator to inhibit itch in the dorsal horn of the spinal cord. Neuron, 82, pp. 573-586. (doi: 10.1016/j.neuron.2014.02.046)

Stagg, C. J., Bachtiar, V., Amadi, U., Gudberg, C. A., Ilie, A. S., Sampaio-Baptista, C., O'Shea, J., Woolrich, M., Smith, S. M. (2014) Local GABA concentration is related to network-level resting functional connectivity. eLife, 3, (doi: 10.7554/eLife.01465)

Banerjee, A., González-Rueda, A., Sampaio-Baptista, C., Paulsen, O., Rodríguez-Moreno, A. (2014) Distinct mechanisms of spike timing-dependent LTD at vertical and horizontal inputs onto L2/3 pyramidal neurons in mouse barrel cortex. Physiological Reports, 2, (doi: 10.1002/phy2.271)

Ross, S. E., Hachisuka, J., Todd, A. J. (2014) Spinal microcircuits and the regulation of itch. CRC Press Taylor & Francis Group

Smith, K. M., Madden, J. F., Callister, R. J., Hughes, D. I., Graham, B. A. (2014) The search for novel analgesics: re-examining spinal cord circuits with new tools. Frontiers in Pharmacology, 5, pp. 1-6. (doi: 10.3389/fphar.2014.00022)

Ijichi, A., Chiba, T., Kudo, K., Hachisuka, J., Nakahara, T., Uchi, H., Takahara, M., Moroi, Y., Furue, M. (2014) Four cases of angiosarcoma in lower leg. Nishi Nihon Hifuka, 76, pp. 27-32. (doi: 10.2336/nishinihonhifu.76.27)

Yasaka, T., Tiong, S. Y.X., Polgár, E., Watanabe, M., Kumamoto, E., Riddell, J. S., Todd, A. J. (2014) A putative relay circuit providing low-threshold mechanoreceptive input to lamina I projection neurons via vertical cells in lamina II of the rat dorsal horn. Molecular Pain, 10, pp. 3. (doi: 10.1186/1744-8069-10-3)

Harris, B. M., Hughes, D. I., Bolton, P. S., Tadross, M. A., Callister, R. J., Graham, B. A. (2014) Contrasting alterations to synaptic and intrinsic properties in upper-cervical superficial dorsal horn neurons following acute neck muscle inflammation. Molecular Pain, 10, (doi: 10.1186/1744-8069-10-25)

Gutierrez-Mecinas, M., Watanabe, M., Todd, A. J. (2014) Expression of gastrin-releasing peptide by excitatory interneurons in the mouse superficial dorsal horn. Molecular Pain, 10, (doi: 10.1186/1744-8069-10-79)

Huma, Z., Du Beau, A., Brown, C., Maxwell, D. J. (2014) Origin and neurochemical properties of bulbospinal neurons projecting to the rat lumbar spinal cord via the medial longitudinal fasciculus and caudal ventrolateral medulla. Frontiers in Neural Circuits, 8, pp. 1-14. (doi: 10.3389/fncir.2014.00040)

Dawson, N., Xiao, X., McDonald, M., Higham, D. J., Morris, B. J., Pratt, J. A. (2014) Sustained NMDA receptor hypofunction induces compromised neural systems integration and schizophrenia-like alterations in functional brain networks. Cerebral Cortex, 24, pp. 452-464. (doi: 10.1093/cercor/bhs322)