Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Carol J. Milligan is active.

Publication


Featured researches published by Carol J. Milligan.


Annals of Neurology | 2014

KCNT1 gain of function in 2 epilepsy phenotypes is reversed by quinidine

Carol J. Milligan; Melody Li; Elena V. Gazina; Sarah E. Heron; Umesh Nair; Chantel Trager; Christopher A. Reid; Anu Venkat; Donald P. Younkin; Dennis J. Dlugos; Slavé Petrovski; David B. Goldstein; Leanne M. Dibbens; Ingrid E. Scheffer; Samuel F. Berkovic; Steven Petrou

Mutations in KCNT1 have been implicated in autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE) and epilepsy of infancy with migrating focal seizures (EIMFS). More recently, a whole exome sequencing study of epileptic encephalopathies identified an additional de novo mutation in 1 proband with EIMFS. We aim to investigate the electrophysiological and pharmacological characteristics of hKCNT1 mutations and examine developmental expression levels.


Annals of Neurology | 2015

Quinidine in the treatment of KCNT1-positive epilepsies

Mohamad A. Mikati; Yong-hui Jiang; Michael P. Carboni; Vandana Shashi; Slavé Petrovski; Rebecca C. Spillmann; Carol J. Milligan; Melody Li; Annette Grefe; Allyn McConkie; Samuel F. Berkovic; Ingrid E. Scheffer; Saul A. Mullen; Melanie J. Bonner; Steven Petrou; David B. Goldstein

We report 2 patients with drug‐resistant epilepsy caused by KCNT1 mutations who were treated with quinidine. Both mutations manifested gain of function in vitro, showing increased current that was reduced by quinidine. One, who had epilepsy of infancy with migrating focal seizures, had 80% reduction in seizure frequency as recorded in seizure diaries, and partially validated by objective seizure evaluation on EEG. The other, who had a novel phenotype, with severe nocturnal focal and secondary generalized seizures starting in early childhood with developmental regression, did not improve. Although quinidine represents an encouraging opportunity for therapeutic benefits, our experience suggests caution in its application and supports the need to identify more targeted drugs for KCNT1 epilepsies. Ann Neurol 2015;78:995–999


Neurology | 2016

A targeted resequencing gene panel for focal epilepsy

Michael S. Hildebrand; Candace T. Myers; Gemma L. Carvill; Brigid M. Regan; John A. Damiano; Saul A. Mullen; Mark R. Newton; Umesh Nair; Elena V. Gazina; Carol J. Milligan; Christopher A. Reid; Steven Petrou; Ingrid E. Scheffer; Samuel F. Berkovic; Mefford Hc

Objectives: We report development of a targeted resequencing gene panel for focal epilepsy, the most prevalent phenotypic group of the epilepsies. Methods: The targeted resequencing gene panel was designed using molecular inversion probe (MIP) capture technology and sequenced using massively parallel Illumina sequencing. Results: We demonstrated proof of principle that mutations can be detected in 4 previously genotyped focal epilepsy cases. We searched for both germline and somatic mutations in 251 patients with unsolved sporadic or familial focal epilepsy and identified 11 novel or very rare missense variants in 5 different genes: CHRNA4, GRIN2B, KCNT1, PCDH19, and SCN1A. Of these, 2 were predicted to be pathogenic or likely pathogenic, explaining ∼0.8% of the cohort, and 8 were of uncertain significance based on available data. Conclusions: We have developed and validated a targeted resequencing panel for focal epilepsies, the most important clinical class of epilepsies, accounting for about 60% of all cases. Our application of MIP technology is an innovative approach that will be advantageous in the clinical setting because it is highly sensitive, efficient, and cost-effective for screening large patient cohorts. Our findings indicate that mutations in known genes likely explain only a small proportion of focal epilepsy cases. This is not surprising given the established clinical and genetic heterogeneity of these disorders and underscores the importance of further gene discovery studies in this complex syndrome.


Methods of Molecular Biology | 2013

Automated planar patch-clamp.

Carol J. Milligan; Clemens Möller

Ion channels are integral membrane proteins that regulate the flow of ions across the plasma membrane and the membranes of intracellular organelles of both excitable and non-excitable cells. Ion channels are vital to a wide variety of biological processes and are prominent components of the nervous system and cardiovascular system, as well as controlling many metabolic functions. Furthermore, ion channels are known to be involved in many disease states and as such have become popular therapeutic targets. For many years now manual patch-clamping has been regarded as one of the best approaches for assaying ion channel function, through direct measurement of ion flow across these membrane proteins. Over the last decade there have been many remarkable breakthroughs in the development of technologies enabling the study of ion channels. One of these breakthroughs is the development of automated planar patch-clamp technology. Automated platforms have demonstrated the ability to generate high-quality data with high throughput capabilities, at great efficiency and reliability. Additional features such as simultaneous intracellular and extracellular perfusion of the cell membrane, current clamp operation, fast compound application, an increasing rate of parallelization, and more recently temperature control have been introduced. Furthermore, in addition to the well-established studies of over-expressed ion channel proteins in cell lines, new generations of planar patch-clamp systems have enabled successful studies of native and primary mammalian cells. This technology is becoming increasingly popular and extensively used both within areas of drug discovery as well as academic research. Many platforms have been developed including NPC-16 Patchliner(®) and SyncroPatch(®) 96 (Nanion Technologies GmbH, Munich), CytoPatch™ (Cytocentrics AG, Rostock), PatchXpress(®) 7000A, IonWorks(®) Quattro and IonWorks Barracuda™, (Molecular Devices, LLC); Dynaflow(®) HT (Cellectricon AB, Mölndal), QPatch HT (Sophion A/S, Copenhagen), IonFlux HT (Fluxion Bioscience Inc, USA), which have demonstrated the capability to generate recordings similar in quality to that of conventional patch clamping. Here we describe features of Nanions NPC-16 Patchliner(®) and processes and protocols suited for this particularly flexible and successful high-throughput automated platform, which is based on planar patch-clamp technology. However, many of the protocols and notes given in this chapter can be applied to other automated patch-clamp platforms, similarly.


Human Mutation | 2017

Purinergic receptors P2RX4 and P2RX7 in familial multiple sclerosis

A. Dessa Sadovnick; Ben J. Gu; Anthony Traboulsee; Cecily Q. Bernales; Irene M. Yee; Maria Criscuoli; Xin Huang; Amber Ou; Carol J. Milligan; Steven Petrou; James S. Wiley; Carles Vilariño-Güell

Genetic variants in the purinergic receptors P2RX4 and P2RX7 have been shown to affect susceptibility to multiple sclerosis (MS). In this study, we set out to evaluate whether rare coding variants of major effect could also be identified in these purinergic receptors. Sequencing analysis of P2RX4 and P2RX7 in 193 MS patients and 100 controls led to the identification of a rare three variant haplotype (P2RX7 rs140915863:C>T [p.T205M], P2RX7 rs201921967:A>G [p.N361S], and P2RX4 rs765866317:G>A [p.G135S]) segregating with disease in a multi‐incident family with six family members diagnosed with MS (logarithm of odds = 3.07). Functional analysis of this haplotype in HEK293 cells revealed impaired P2X7 surface expression (P < 0.01), resulting in over 95% inhibition of adenosine triphosphate (ATP)‐induced pore function (P < 0.001) and a marked reduction in phagocytic ability (P < 0.05). In addition, transfected cells showed 40% increased peak ATP‐induced inward current (P < 0.01), and a greater Ca2+ response to the P2X4 135S variant compared with wild type (P < 0.0001). Our study nominates rare genetic variants in P2RX4 and P2RX7 as major genetic contributors to disease, further supporting a role for these purinergic receptors in MS and the disruption of transmembrane cation channels leading to impairment of phagocytosis as the pathological mechanisms of disease.


Annals of Neurology | 2017

Myoclonus epilepsy and ataxia due to KCNC1 mutation: Analysis of 20 cases and K+ channel properties

Karen L. Oliver; Silvana Franceschetti; Carol J. Milligan; Mikko Muona; Simone Mandelstam; Laura Canafoglia; Anna M. Boguszewska-Chachulska; Amos D. Korczyn; Francesca Bisulli; Carlo Di Bonaventura; Francesca Ragona; Roberto Michelucci; Bruria Ben-Zeev; Rachel Straussberg; Ferruccio Panzica; João Massano; Daniel Friedman; Arielle Crespel; Bernt A. Engelsen; Frederick Andermann; Eva Andermann; Krystyna Spodar; Anetta Lasek-Bal; Patrizia Riguzzi; Elena Pasini; Paolo Tinuper; Laura Licchetta; Elena Gardella; Matthias Lindenau; Annette Wulf

To comprehensively describe the new syndrome of myoclonus epilepsy and ataxia due to potassium channel mutation (MEAK), including cellular electrophysiological characterization of observed clinical improvement with fever.


Journal of Neurochemistry | 2016

Toluene inhalation in adolescent rats reduces flexible behaviour in adulthood and alters glutamatergic and GABAergic signalling.

Teri M. Furlong; Jhodie R. Duncan; Laura H. Corbit; Caroline Rae; Benjamin D. Rowlands; Anthony D. Maher; Fatima A. Nasrallah; Carol J. Milligan; Steven Petrou; Andrew J. Lawrence; Bernard W. Balleine

Toluene is a commonly abused inhalant that is easily accessible to adolescents. Despite the increasing incidence of use, our understanding of its long‐term impact remains limited. Here, we used a range of techniques to examine the acute and chronic effects of toluene exposure on glutameteric and GABAergic function, and on indices of psychological function in adult rats after adolescent exposure. Metabolomics conducted on cortical tissue established that acute exposure to toluene produces alterations in cellular metabolism indicative of a glutamatergic and GABAergic profile. Similarly, in vitro electrophysiology in Xenopus oocytes found that acute toluene exposure reduced NMDA receptor signalling. Finally, in an adolescent rodent model of chronic intermittent exposure to toluene (10 000 ppm), we found that, while toluene exposure did not affect initial learning, it induced a deficit in updating that learning when response‐outcome relationships were reversed or degraded in an instrumental conditioning paradigm. There were also group differences when more effort was required to obtain the reward; toluene‐exposed animals were less sensitive to progressive ratio schedules and to delayed discounting. These behavioural deficits were accompanied by changes in subunit expression of both NMDA and GABA receptors in adulthood, up to 10 weeks after the final exposure to toluene in the hippocampus, prefrontal cortex and ventromedial striatum; regions with recognized roles in behavioural flexibility and decision‐making. Collectively, our data suggest that exposure to toluene is sufficient to induce adaptive changes in glutamatergic and GABAergic systems and in adaptive behaviour that may underlie the deficits observed following adolescent inhalant abuse, including susceptibility to further drug‐use.


Epilepsia | 2016

The antiepileptic medications carbamazepine and phenytoin inhibit native sodium currents in murine osteoblasts.

Sandra J. Petty; Carol J. Milligan; Marian Todaro; Kay L. Richards; Pamuditha K. Kularathna; Charles N. Pagel; Chris French; Elisa L. Hill-Yardin; Terence J. O'Brien; John D. Wark; Eleanor J. Mackie; Steven Petrou

Fracture risk is a serious comorbidity in epilepsy and may relate to the use of antiepileptic drugs (AEDs). Many AEDs inhibit ion channel function, and the expression of these channels in osteoblasts raises the question of whether altered bone signaling increases bone fragility. We aimed to confirm the expression of voltage‐gated sodium (NaV) channels in mouse osteoblasts, and to investigate the action of carbamazepine and phenytoin on NaV channels.


PLOS ONE | 2015

Single Nucleotide Variations in CLCN6 Identified in Patients with Benign Partial Epilepsies in Infancy and/or Febrile Seizures

Toshiyuki Yamamoto; Keiko Shimojima; Noriko Sangu; Yuta Komoike; Atsushi Ishii; Shinpei Abe; Shintaro Yamashita; Katsumi Imai; Tetsuo Kubota; Tatsuya Fukasawa; Tohru Okanishi; Hideo Enoki; Takuya Tanabe; Akira Saito; Toru Furukawa; Toshiaki Shimizu; Carol J. Milligan; Steven Petrou; Sarah E. Heron; Leanne M. Dibbens; Shinichi Hirose; Akihisa Okumura

Nucleotide alterations in the gene encoding proline-rich transmembrane protein 2 (PRRT2) have been identified in most patients with benign partial epilepsies in infancy (BPEI)/benign familial infantile epilepsy (BFIE). However, not all patients harbor these PRRT2 mutations, indicating the involvement of genes other than PRRT2. In this study, we performed whole exome sequencing analysis for a large family affected with PRRT2-unrelated BPEI. We identified a non-synonymous single nucleotide variation (SNV) in the voltage-sensitive chloride channel 6 gene (CLCN6). A cohort study of 48 BPEI patients without PRRT2 mutations revealed a different CLCN6 SNV in a patient, his sibling and his father who had a history of febrile seizures (FS) but not BPEI. Another study of 48 patients with FS identified an additional SNV in CLCN6. Chloride channels (CLCs) are involved in a multitude of physiologic processes and some members of the CLC family have been linked to inherited diseases. However, a phenotypic correlation has not been confirmed for CLCN6. Although we could not detect significant biological effects linked to the identified CLCN6 SNVs, further studies should investigate potential CLCN6 variants that may underlie the genetic susceptibility to convulsive disorders.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Selective NaV1.1 activation rescues Dravet syndrome mice from seizures and premature death

Kay L. Richards; Carol J. Milligan; Robert J. Richardson; Nikola Jancovski; Morten Grunnet; Laura H. Jacobson; Eivind A. B. Undheim; Mehdi Mobli; Chun Yuen Chow; Volker Herzig; Agota Csoti; Gyorgy Panyi; Christopher A. Reid; Glenn F. King; Steven Petrou

Significance Spider venom is a rich source of peptides, many targeting ion channels. We assessed a venom peptide, Hm1a, as a potential targeted therapy for Dravet syndrome, the genetic epilepsy linked to a mutation in the gene encoding the sodium channel alpha subunit NaV1.1. Cell-based assays showed Hm1a was selective for hNaV1.1 over other sodium and potassium channels. Utilizing a mouse model of Dravet syndrome, Hm1a restored inhibitory neuron function and significantly reduced seizures and mortality in heterozygote mice. Evidence from the structure of Hm1a and modeling suggest Hm1a interacts with NaV1.1 inactivation domains, providing a structural correlate of the functional mechanisms. This proof-of-concept study provides a promising strategy for future drug development in genetic epilepsy and other neurogenetic disorders. Dravet syndrome is a catastrophic, pharmacoresistant epileptic encephalopathy. Disease onset occurs in the first year of life, followed by developmental delay with cognitive and behavioral dysfunction and substantially elevated risk of premature death. The majority of affected individuals harbor a loss-of-function mutation in one allele of SCN1A, which encodes the voltage-gated sodium channel NaV1.1. Brain NaV1.1 is primarily localized to fast-spiking inhibitory interneurons; thus the mechanism of epileptogenesis in Dravet syndrome is hypothesized to be reduced inhibitory neurotransmission leading to brain hyperexcitability. We show that selective activation of NaV1.1 by venom peptide Hm1a restores the function of inhibitory interneurons from Dravet syndrome mice without affecting the firing of excitatory neurons. Intracerebroventricular infusion of Hm1a rescues Dravet syndrome mice from seizures and premature death. This precision medicine approach, which specifically targets the molecular deficit in Dravet syndrome, presents an opportunity for treatment of this intractable epilepsy.

Collaboration


Dive into the Carol J. Milligan's collaboration.

Top Co-Authors

Avatar

Steven Petrou

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Christopher A. Reid

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar

Melody Li

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Lawrence

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. Wark

Royal Melbourne Hospital

View shared research outputs
Top Co-Authors

Avatar

Kay L. Richards

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge