Network


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

Hotspot


Dive into the research topics where Andreas C. Themistocleous is active.

Publication


Featured researches published by Andreas C. Themistocleous.


Practical Neurology | 2014

The clinical approach to small fibre neuropathy and painful channelopathy

Andreas C. Themistocleous; Juan D. Ramirez; Jordi Serra; David L. H. Bennett

Small fibre neuropathy (SFN) is characterised by structural injury selectively affecting small diameter sensory and/or autonomic axons. The clinical presentation is dominated by pain. SFN complicates a number of common diseases such as diabetes mellitus and is likely to be increasingly encountered. The diagnosis of SFN is demanding as clinical features can be vague and nerve conduction studies normal. New diagnostic techniques, in particular measurement of intraepidermal nerve fibre density, have significantly improved the diagnostic efficiency of SFN. Management is focused on the treatment of the underlying cause and analgesia, as there is no neuroprotective therapy. A recent and significant advance is the finding that a proportion of cases labelled as idiopathic SFN are in fact associated with gain of function mutations of the voltage-gated sodium channels Nav1.7 and Nav1.8 (encoded by the genes SCN9A and SCN10A, respectively). There is a further group of heritable painful conditions in which gain of function mutations in ion channels alter excitability of sensory neurones but do not cause frank axon degeneration; these include mutations in Nav1.7 (causing erythromelalgia and paroxysmal extreme pain disorder) and TRPA1 (resulting in familial episodic pain disorder). These conditions are exceptionally rare but have provided great insight into the nociceptive system as well as yielding potential analgesic drug targets. In patients with no pre-existing risk factor, the investigation of an underlying cause of SFN should be systematic and appropriate for the patient population. In this review, we focus on how to incorporate recent developments in the diagnosis and pathophysiology of SFN into clinical practice.


Pain | 2016

The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy

Andreas C. Themistocleous; Juan D. Ramirez; Pallai Shillo; Jonathan G. Lees; Dinesh Selvarajah; Christine A. Orengo; Solomon Tesfaye; Andrew S.C. Rice; David L. H. Bennett

Abstract Disabling neuropathic pain (NeuP) is a common sequel of diabetic peripheral neuropathy (DPN). We aimed to characterise the sensory phenotype of patients with and without NeuP, assess screening tools for NeuP, and relate DPN severity to NeuP. The Pain in Neuropathy Study (PiNS) is an observational cross-sectional multicentre study. A total of 191 patients with DPN underwent neurological examination, quantitative sensory testing, nerve conduction studies, and skin biopsy for intraepidermal nerve fibre density assessment. A set of questionnaires assessed the presence of pain, pain intensity, pain distribution, and the psychological and functional impact of pain. Patients were divided according to the presence of DPN, and thereafter according to the presence and severity of NeuP. The DN4 questionnaire demonstrated excellent sensitivity (88%) and specificity (93%) in screening for NeuP. There was a positive correlation between greater neuropathy severity (r = 0.39, P < 0.01), higher HbA1c (r = 0.21, P < 0.01), and the presence (and severity) of NeuP. Diabetic peripheral neuropathy sensory phenotype is characterised by hyposensitivity to applied stimuli that was more marked in the moderate/severe NeuP group than in the mild NeuP or no NeuP groups. Brush-evoked allodynia was present in only those with NeuP (15%); the paradoxical heat sensation did not discriminate between those with (40%) and without (41.3%) NeuP. The “irritable nociceptor” subgroup could only be applied to a minority of patients (6.3%) with NeuP. This study provides a firm basis to rationalise further phenotyping of painful DPN, for instance, stratification of patients with DPN for analgesic drug trials.


Nature Genetics | 2015

Transcriptional regulator PRDM12 is essential for human pain perception

Ya Chun Chen; Michaela Auer-Grumbach; Shinya Matsukawa; Manuela Zitzelsberger; Andreas C. Themistocleous; Tim M. Strom; Chrysanthi Samara; Adrian W Moore; Lily Ting-Yin Cho; Gareth T. Young; Caecilia Weiss; Maria Schabhüttl; Rolf Stucka; Annina B. Schmid; Yesim Parman; Luitgard Graul-Neumann; Wolfram Heinritz; Eberhard Passarge; Rosemarie Watson; Jens Michael Hertz; Ute Moog; Manuela Baumgartner; Enza Maria Valente; Diego Pereira; Carlos Martín Restrepo; Istvan Katona; Marina Dusl; Claudia Stendel; Thomas Wieland; Fay Stafford

Pain perception has evolved as a warning mechanism to alert organisms to tissue damage and dangerous environments. In humans, however, undesirable, excessive or chronic pain is a common and major societal burden for which available medical treatments are currently suboptimal. New therapeutic options have recently been derived from studies of individuals with congenital insensitivity to pain (CIP). Here we identified 10 different homozygous mutations in PRDM12 (encoding PRDI-BF1 and RIZ homology domain-containing protein 12) in subjects with CIP from 11 families. Prdm proteins are a family of epigenetic regulators that control neural specification and neurogenesis. We determined that Prdm12 is expressed in nociceptors and their progenitors and participates in the development of sensory neurons in Xenopus embryos. Moreover, CIP-associated mutants abrogate the histone-modifying potential associated with wild-type Prdm12. Prdm12 emerges as a key factor in the orchestration of sensory neurogenesis and may hold promise as a target for new pain therapeutics.


American Journal of Human Genetics | 2016

Rare Variants in MME, Encoding Metalloprotease Neprilysin, Are Linked to Late-Onset Autosomal-Dominant Axonal Polyneuropathies.

Michaela Auer-Grumbach; Stefan Toegel; Maria Schabhüttl; Daniela Weinmann; Catharina Chiari; David L. H. Bennett; Christian Beetz; Dennis Klein; Peter Andersen; Ilka Böhme; Regina Fink-Puches; Michael Gonzalez; Matthew Harms; William W. Motley; Mary M. Reilly; Wilfried Renner; Sabine Rudnik-Schöneborn; Beate Schlotter-Weigel; Andreas C. Themistocleous; Jochen H. Weishaupt; Albert C. Ludolph; Thomas Wieland; Feifei Tao; Lisa Abreu; Reinhard Windhager; Manuela Zitzelsberger; Tim M. Strom; Thomas Walther; Steven S. Scherer; Stephan Züchner

Axonal polyneuropathies are a frequent cause of progressive disability in the elderly. Common etiologies comprise diabetes mellitus, paraproteinaemia, and inflammatory disorders, but often the underlying causes remain elusive. Late-onset axonal Charcot-Marie-Tooth neuropathy (CMT2) is an autosomal-dominantly inherited condition that manifests in the second half of life and is genetically largely unexplained. We assumed age-dependent penetrance of mutations in a so far unknown gene causing late-onset CMT2. We screened 51 index case subjects with late-onset CMT2 for mutations by whole-exome (WES) and Sanger sequencing and subsequently queried WES repositories for further case subjects carrying mutations in the identified candidate gene. We studied nerve pathology and tissue levels and function of the abnormal protein in order to explore consequences of the mutations. Altogether, we observed heterozygous rare loss-of-function and missense mutations in MME encoding the metalloprotease neprilysin in 19 index case subjects diagnosed with axonal polyneuropathies or neurodegenerative conditions involving the peripheral nervous system. MME mutations segregated in an autosomal-dominant fashion with age-related incomplete penetrance and some affected individuals were isolated case subjects. We also found that MME mutations resulted in strongly decreased tissue availability of neprilysin and impaired enzymatic activity. Although neprilysin is known to degrade β-amyloid, we observed no increased amyloid deposition or increased incidence of dementia in individuals with MME mutations. Detection of MME mutations is expected to increase the diagnostic yield in late-onset polyneuropathies, and it will be tempting to explore whether substances that can elevate neprilysin activity could be a rational option for treatment.


Pain | 2017

Stratifying patients with peripheral neuropathic pain based on sensory profiles: algorithm and sample size recommendations

Jan Vollert; Christoph Maier; Nadine Attal; David L. Bennett; Didier Bouhassira; Elena K. Enax-Krumova; Nanna Brix Finnerup; Rainer Freynhagen; Janne Gierthmühlen; Maija Haanpää; Per Hansson; Philipp Hüllemann; Troels Staehelin Jensen; Walter Magerl; Juan D. Ramirez; Andrew S.C. Rice; Sigrid Schuh-Hofer; Märta Segerdahl; Jordi Serra; Pallai Shillo; Soeren Sindrup; Solomon Tesfaye; Andreas C. Themistocleous; Thomas R. Tölle; Rolf-Detlef Treede; Ralf Baron

Abstract In a recent cluster analysis, it has been shown that patients with peripheral neuropathic pain can be grouped into 3 sensory phenotypes based on quantitative sensory testing profiles, which are mainly characterized by either sensory loss, intact sensory function and mild thermal hyperalgesia and/or allodynia, or loss of thermal detection and mild mechanical hyperalgesia and/or allodynia. Here, we present an algorithm for allocation of individual patients to these subgroups. The algorithm is nondeterministic—ie, a patient can be sorted to more than one phenotype—and can separate patients with neuropathic pain from healthy subjects (sensitivity: 78%, specificity: 94%). We evaluated the frequency of each phenotype in a population of patients with painful diabetic polyneuropathy (n = 151), painful peripheral nerve injury (n = 335), and postherpetic neuralgia (n = 97) and propose sample sizes of study populations that need to be screened to reach a subpopulation large enough to conduct a phenotype-stratified study. The most common phenotype in diabetic polyneuropathy was sensory loss (83%), followed by mechanical hyperalgesia (75%) and thermal hyperalgesia (34%, note that percentages are overlapping and not additive). In peripheral nerve injury, frequencies were 37%, 59%, and 50%, and in postherpetic neuralgia, frequencies were 31%, 63%, and 46%. For parallel study design, either the estimated effect size of the treatment needs to be high (>0.7) or only phenotypes that are frequent in the clinical entity under study can realistically be performed. For crossover design, populations under 200 patients screened are sufficient for all phenotypes and clinical entities with a minimum estimated treatment effect size of 0.5.


Neuron | 2018

Immune or Genetic-Mediated Disruption of CASPR2 Causes Pain Hypersensitivity Due to Enhanced Primary Afferent Excitability

John M. Dawes; Greg A. Weir; Steven J. Middleton; Ryan Patel; Kim I. Chisholm; Liam J. Peck; Joseph Sheridan; Akila Shakir; Leslie Jacobson; Maria Gutierrez-Mecinas; J Galino; Jan Walcher; Johannes Kühnemund; Hannah Kuehn; Maria D. Sanna; Bethan Lang; Alex J. Clark; Andreas C. Themistocleous; Noboru Iwagaki; Steven West; Karolina Werynska; Liam Carroll; Teodora Trendafilova; David A. Menassa; Maria Pia Giannoccaro; Ester Coutinho; Ilaria Cervellini; Damini Tewari; Camilla Buckley; M. Isabel Leite

Summary Human autoantibodies to contactin-associated protein-like 2 (CASPR2) are often associated with neuropathic pain, and CASPR2 mutations have been linked to autism spectrum disorders, in which sensory dysfunction is increasingly recognized. Human CASPR2 autoantibodies, when injected into mice, were peripherally restricted and resulted in mechanical pain-related hypersensitivity in the absence of neural injury. We therefore investigated the mechanism by which CASPR2 modulates nociceptive function. Mice lacking CASPR2 (Cntnap2−/−) demonstrated enhanced pain-related hypersensitivity to noxious mechanical stimuli, heat, and algogens. Both primary afferent excitability and subsequent nociceptive transmission within the dorsal horn were increased in Cntnap2−/− mice. Either immune or genetic-mediated ablation of CASPR2 enhanced the excitability of DRG neurons in a cell-autonomous fashion through regulation of Kv1 channel expression at the soma membrane. This is the first example of passive transfer of an autoimmune peripheral neuropathic pain disorder and demonstrates that CASPR2 has a key role in regulating cell-intrinsic dorsal root ganglion (DRG) neuron excitability.


Brain | 2018

A brain-based pain facilitation mechanism contributes to painful diabetic polyneuropathy.

Andrew R. Segerdahl; Andreas C. Themistocleous; Dean Fido; David L. Bennett; Irene Tracey

Diabetic polyneuropathy is a leading cause of chronic neuropathic pain, but the mechanism underlying this link is unknown. Using a multimodal neuroimaging approach, Segerdahl et al. show dysfunction of the descending pain modulatory system in those patients with neuropathic pain, which is associated with amplified brain activity in response to painful stimuli.


Brain | 2017

Chronic non-freezing cold injury results in neuropathic pain due to a sensory neuropathy

Tom A Vale; Mkael Symmonds; Michael Polydefkis; Kelly Byrnes; Andrew S.C. Rice; Andreas C. Themistocleous; David L. Bennett

Non-freezing cold injury, first described during World War I and known colloquially as ‘trench foot’, remains a common cause of disabling, career-ending pain in soldiers. Vale et al. show that this pain is due to an acquired sensory neuropathy, providing an evidence-based rationale for its diagnosis and treatment.


Pain | 2017

Rare NaV1.7 variants associated with painful diabetic peripheral neuropathy.

Iulia Blesneac; Andreas C. Themistocleous; Carl Fratter; Linus J. Conrad; Juan D. Ramirez; James J. Cox; Solomon Tesfaye; Pallai Shillo; Andrew S.C. Rice; Stephen J. Tucker; David L. Bennett

Abstract Diabetic peripheral neuropathy (DPN) is a common disabling complication of diabetes. Almost half of the patients with DPN develop neuropathic pain (NeuP) for which current analgesic treatments are inadequate. Understanding the role of genetic variability in the development of painful DPN is needed for improved understanding of pain pathogenesis for better patient stratification in clinical trials and to target therapy more appropriately. Here, we examined the relationship between variants in the voltage-gated sodium channel NaV1.7 and NeuP in a deeply phenotyped cohort of patients with DPN. Although no rare variants were found in 78 participants with painless DPN, we identified 12 rare NaV1.7 variants in 10 (out of 111) study participants with painful DPN. Five of these variants had previously been described in the context of other NeuP disorders and 7 have not previously been linked to NeuP. Those patients with rare variants reported more severe pain and greater sensitivity to pressure stimuli on quantitative sensory testing. Electrophysiological characterization of 2 of the novel variants (M1852T and T1596I) demonstrated that gain of function changes as a consequence of markedly impaired channel fast inactivation. Using a structural model of NaV1.7, we were also able to provide further insight into the structural mechanisms underlying fast inactivation and the role of the C-terminal domain in this process. Our observations suggest that rare NaV1.7 variants contribute to the development NeuP in patients with DPN. Their identification should aid understanding of sensory phenotype, patient stratification, and help target treatments effectively.


Neurology | 2014

Null mutation in SCN9A in which noxious stimuli can be detected in the absence of pain.

Juan D. Ramirez; Abdella M. Habib; James J. Cox; Andreas C. Themistocleous; Stephen B. McMahon; John N. Wood; David L. H. Bennett

The term nociception was originally defined by Sherrington1 as the neural process by which high-threshold stimuli (which cause tissue injury) are detected. This is distinct from pain, which is defined by the quality of the sensory percept (i.e., unpleasant), is not always evoked by a noxious stimulus, and can also be experienced in the absence of a noxious stimulus. In man, recessive loss of function mutations in the SCN9A gene, encoding the α subunit of Nav1.7, results in anosmia and the congenital inability to experience pain,2–4 but there have been few reports of detailed sensory testing in these patients.

Collaboration


Dive into the Andreas C. Themistocleous's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Solomon Tesfaye

Royal Hallamshire Hospital

View shared research outputs
Top Co-Authors

Avatar

Maria Schabhüttl

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adrian W Moore

Western General Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge