Network


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

Hotspot


Dive into the research topics where Salman Haider is active.

Publication


Featured researches published by Salman Haider.


Journal of Clinical Investigation | 2012

Mutant huntingtin fragmentation in immune cells tracks Huntington’s disease progression

Andreas Weiss; Ulrike Träger; Edward J. Wild; Stephan Grueninger; Ruth Farmer; Christian Landles; Rachael I. Scahill; Nayana Lahiri; Salman Haider; Douglas Macdonald; Chris Frost; Gillian P. Bates; Graeme Bilbe; Rainer Kuhn; Ralph Andre; Sarah J. Tabrizi

Huntingtons disease (HD) is a fatal, inherited neurodegenerative disorder caused by an expanded CAG repeat in the gene encoding huntingtin (HTT). Therapeutic approaches to lower mutant HTT (mHTT) levels are expected to proceed to human trials, but noninvasive quantification of mHTT is not currently possible. The importance of the peripheral immune system in neurodegenerative disease is becoming increasingly recognized. Peripheral immune cells have been implicated in HD pathogenesis, but HTT levels in these cells have not been quantified before. A recently described time-resolved Förster resonance energy transfer (TR-FRET) immunoassay was used to quantify mutant and total HTT protein levels in leukocytes from patients with HD. Mean mHTT levels in monocytes, T cells, and B cells differed significantly between patients with HD and controls and between pre-manifest mutation carriers and those with clinical onset. Monocyte and T cell mHTT levels were significantly associated with disease burden scores and caudate atrophy rates in patients with HD. mHTT N-terminal fragments detected in HD PBMCs may explain the progressive increase in mHTT levels in these cells. These findings indicate that quantification of mHTT in peripheral immune cells by TR-FRET holds significant promise as a noninvasive disease biomarker.


Journal of Clinical Investigation | 2015

Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington’s disease patients

Edward J. Wild; Roberto Boggio; Douglas R. Langbehn; Nicola J. Robertson; Salman Haider; James R. Miller; Henrik Zetterberg; Blair R. Leavitt; Rainer Kuhn; Sarah J. Tabrizi; Douglas Macdonald; Andreas Weiss

BACKGROUND Quantification of disease-associated proteins in the cerebrospinal fluid (CSF) has been critical for the study and treatment of several neurodegenerative disorders; however, mutant huntingtin protein (mHTT), the cause of Huntingtons disease (HD), is at very low levels in CSF and, to our knowledge, has never been measured previously. METHODS We developed an ultrasensitive single-molecule counting (SMC) mHTT immunoassay that was used to quantify mHTT levels in CSF samples from individuals bearing the HD mutation and from control individuals in 2 independent cohorts. RESULTS This SMC mHTT immunoassay demonstrated high specificity for mHTT, high sensitivity with a femtomolar detection threshold, and a broad dynamic range. Analysis of the CSF samples showed that mHTT was undetectable in CSF from all controls but quantifiable in nearly all mutation carriers. The mHTT concentration in CSF was approximately 3-fold higher in patients with manifest HD than in premanifest mutation carriers. Moreover, mHTT levels increased as the disease progressed and were associated with 5-year onset probability. The mHTT concentration independently predicted cognitive and motor dysfunction. Furthermore, the level of mHTT was associated with the concentrations of tau and neurofilament light chain in the CSF, suggesting a neuronal origin for the detected mHTT. CONCLUSIONS We have demonstrated that mHTT can be quantified in CSF from HD patients using the described SMC mHTT immunoassay. Moreover, the level of mHTT detected is associated with proximity to disease onset and diminished cognitive and motor function. The ability to quantify CSF mHTT will facilitate the study of HD, and mHTT quantification could potentially serve as a biomarker for the development and testing of experimental mHTT-lowering therapies for HD. TRIAL REGISTRATION Not applicable. FUNDING CHDI Foundation Inc.; Medical Research Council (MRC) UK; National Institutes for Health Research (NIHR); Rosetrees Trust; Swedish Research Council; and Knut and Alice Wallenberg Foundation.


British Journal of Clinical Pharmacology | 2015

An exploratory double‐blind, randomized clinical trial with selisistat, a SirT1 inhibitor, in patients with Huntington's disease

Sigurd D. Süssmuth; Salman Haider; G. Bernhard Landwehrmeyer; Ruth Farmer; Chris Frost; Giovanna Tripepi; Claus A. Andersen; Marco Di Bacco; Claudia Lamanna; Enrica Diodato; Luisa Massai; Daniela Diamanti; Elisa Mori; Letizia Magnoni; Jens Dreyhaupt; Karin Schiefele; David Craufurd; Carsten Saft; Monika Rudzińska; Danuta Ryglewicz; Michael Orth; Sebastian Brzozy; Anna Baran; Giuseppe Pollio; Ralph Andre; Sarah J. Tabrizi; Borje Darpo; G Westerberg

AIMS Selisistat, a selective SirT1 inhibitor is being developed as a potentially disease-modifying therapeutic for Huntingtons disease (HD). This was the first study of selisistat in HD patients and was primarily aimed at development of pharmacodynamic biomarkers. METHODS This was a randomized, double-blind, placebo-controlled, multicentre exploratory study. Fifty-five male and female patients in early stage HD were randomized to receive 10 mg or 100 mg of selisistat or placebo once daily for 14 days. Blood sampling, clinical and safety assessments were conducted throughout the study. Candidate pharmacodynamic markers included circulating soluble huntingtin and innate immune markers. RESULTS Selisistat was found to be safe and well tolerated, and systemic exposure parameters showed that the average steady-state plasma concentration achieved at the 10 mg dose level (125 nm) was comparable with the IC50 for SirT1 inhibition. No adverse effects on motor, cognitive or functional readouts were recorded. While circulating levels of soluble huntingtin were not affected by selisistat in this study, the biological samples collected have allowed development of assay technology for use in future studies. No effects on innate immune markers were seen. CONCLUSIONS Selisistat was found to be safe and well tolerated in early stage HD patients at plasma concentrations within the anticipated therapeutic concentration range.


Brain | 2015

Altered PDE10A expression detectable early before symptomatic onset in Huntington’s disease

Flavia Niccolini; Salman Haider; Tiago Reis Marques; Nils Muhlert; Andri C. Tziortzi; Graham Searle; Sridhar Natesan; Paola Piccini; Shitij Kapur; Eugenii A. Rabiner; Roger N. Gunn; Sarah J. Tabrizi; Marios Politis

There is an urgent need for early biomarkers and novel disease-modifying therapies in Huntingtons disease. Huntingtons disease pathology involves the toxic effect of mutant huntingtin primarily in striatal medium spiny neurons, which highly express phosphodiesterase 10A (PDE10A). PDE10A hydrolyses cAMP/cGMP signalling cascades, thus having a key role in the regulation of striatal output, and in promoting neuronal survival. PDE10A could be a key therapeutic target in Huntingtons disease. Here, we used combined positron emission tomography (PET) and multimodal magnetic resonance imaging to assess PDE10A expression in vivo in a unique cohort of 12 early premanifest Huntingtons disease gene carriers with a mean estimated 90% probability of 25 years before the predicted onset of clinical symptoms. We show bidirectional changes in PDE10A expression in premanifest Huntingtons disease gene carriers, which are associated with the probability of symptomatic onset. PDE10A expression in early premanifest Huntingtons disease was decreased in striatum and pallidum and increased in motor thalamic nuclei, compared to a group of matched healthy controls. Connectivity-based analysis revealed prominent PDE10A decreases confined in the sensorimotor-striatum and in striatonigral and striatopallidal projecting segments. The ratio between higher PDE10A expression in motor thalamic nuclei and lower PDE10A expression in striatopallidal projecting striatum was the strongest correlate with higher probability of symptomatic conversion in early premanifest Huntingtons disease gene carriers. Our findings demonstrate in vivo, a novel and earliest pathophysiological mechanism underlying Huntingtons disease with direct implications for the development of new pharmacological treatments, which can promote neuronal survival and improve outcome in Huntingtons disease gene carriers.


Movement Disorders | 2014

Cerebellar abnormalities in Huntington's disease: A role in motor and psychiatric impairment?

Em Rees; Ruth Farmer; James H. Cole; Salman Haider; Alexandra Durr; Bernhard Landwehrmeyer; Rachael I. Scahill; Sarah J. Tabrizi; Nicola Z. Hobbs

The cerebellum has received limited attention in Huntingtons disease (HD), despite signs of possible cerebellar dysfunction, including motor incoordination and impaired gait, which are currently attributed to basal ganglia atrophy and disrupted fronto‐striatal circuits. This study is the first to investigate a potential contribution of macro‐ and microstructural cerebellar damage to clinical manifestations of HD. T1‐ and diffusion‐weighted 3T magnetic resonance imaging (MRI) scans were obtained from 12 controls and 22 early‐stage HD participants. Manual delineation and voxel‐based morphometry were used to assess between‐group differences in cerebellar volume, and diffusion metrics were compared between groups within the cerebellar gray and white matter. Associations between these imaging measures and clinical scores were examined within the HD group. Reduced paravermal volume was detected in HD compared with controls using voxel‐based morphometry (P < 0.05), but no significant volumetric differences were found using manual delineation. Diffusion abnormalities were detected in both cerebellar gray matter and white matter. Smaller cerebellar volumes, although not significantly reduced, were significantly associated with impaired gait and psychiatric morbidity and of borderline significance with pronate/supinate‐hand task performance. Abnormal cerebellar diffusion was associated with increased total motor score, impaired saccade initiation, tandem walking, and timed finger tapping. In conclusion, atrophy of the paravermis, possibly encompassing the cerebellar nuclei, and microstructural abnormalities within the cerebellum may contribute to HD neuropathology. Aberrant cerebellar diffusion and reduced cerebellar volume together associate with impaired motor function and increased psychiatric symptoms in stage I HD, potentially implicating the cerebellum more centrally in HD presentation than previously recognized.


Drug Discovery Today | 2014

Biomarker development for Huntington's disease

Ralph Andre; Rachael I. Scahill; Salman Haider; Sarah J. Tabrizi

Huntingtons disease (HD) is a fatal inherited neurodegenerative disorder, treatment to slow the progression of which has not yet been found. Human clinical trials to test a number of therapeutic strategies are underway or imminent, facilitated in part by the recent development of biomarkers that might be used as surrogate endpoints in such trials. However, although much progress in developing HD biomarkers has been made, ongoing work seeks to improve the sensitivity and reliability of current measures, and to demonstrate that they correspond to clear meaningful benefit to patients. Of particular importance is the identification of state biomarkers that can be used in pre-manifest HD gene carriers to test therapies hoped to delay symptom onset in these individuals. Functional, neuroimaging and biochemical biomarkers continue to be investigated for use in the development of disease-modifying treatments of HD.


BMC Biochemistry | 2013

Development of an ELISA assay for the quantification of soluble huntingtin in human blood cells

Luisa Massai; Lara Petricca; Letizia Magnoni; Luca Rovetini; Salman Haider; Ralph Andre; Sarah J. Tabrizi; Sigurd D. Süssmuth; Bernhard Landwehrmeyer; Andrea Caricasole; Giuseppe Pollio; Simonetta Bernocco

BackgroundHuntington’s disease (HD) is a monogenic disorder caused by an aberrant expansion of CAG repeats in the huntingtin gene (HTT). Pathogenesis is associated with expression of the mutant (mHTT) protein in the CNS, with its levels most likely related to disease progression and symptom severity. Since non-invasive methods to quantify HTT in the CNS do not exist, measuring amount of soluble HTT in peripheral cells represents an important step in development of disease-modifying interventions in HD.ResultsAn ELISA assay using commercially available antibodies was developed to quantify HTT levels in complex matrices like mammalian cell cultures lysates and human samples. The immunoassay was optimized using a recombinant full-length HTT protein, and validated both on wild-type and mutant HTT species. The ability of the assay to detect significant variations of soluble HTT levels was evaluated using an HSP90 inhibitor that is known to enhance HTT degradation. Once optimized, the bioassay was applied to peripheral blood mononuclear cells (PBMCs) from HD patients, demonstrating good potential in tracking the disease course.ConclusionsThe method described here represents a validated, simple and rapid bio-molecular assay to evaluate soluble HTT levels in blood cells as useful tool in disease and pharmacodynamic marker identification for observational and clinical trials.


Journal of the Neurological Sciences | 2016

Loss of extra-striatal phosphodiesterase 10A expression in early premanifest Huntington's disease gene carriers

Heather Wilson; Flavia Niccolini; Salman Haider; Tiago Reis Marques; Gennaro Pagano; Christopher Coello; Sridhar Natesan; Shitij Kapur; Eugenii A. Rabiner; Roger N. Gunn; Sarah J. Tabrizi; Marios Politis

Huntingtons disease (HD) is a monogenic neurodegenerative disorder with an underlying pathology involving the toxic effect of mutant huntingtin protein primarily in striatal and cortical neurons. Phosphodiesterase 10A (PDE10A) regulates intracellular signalling cascades, thus having a key role in promoting neuronal survival. Using positron emission tomography (PET) with [(11)C]IMA107, we investigated the in vivo extra-striatal expression of PDE10A in 12 early premanifest HD gene carriers. Image processing and kinetic modelling was performed using MIAKAT™. Parametric images of [(11)C]IMA107 non-displaceable binding potential (BPND) were generated from the dynamic [(11)C]IMA107 scans using the simplified reference tissue model with the cerebellum as the reference tissue for nonspecific binding. We set a threshold criterion for meaningful quantification of [(11)C]IMA107 BPND at 0.30 in healthy control data; regions meeting this criterion were designated as regions of interest (ROIs). MRI-based volumetric analysis showed no atrophy in ROIs. We found significant differences in mean ROIs [(11)C]IMA107 BPND between HD gene carriers and healthy controls. HD gene carriers had significant loss of PDE10A within the insular cortex and occipital fusiform gyrus compared to healthy controls. Insula and occipital fusiform gyrus are important brain areas for the regulation of cognitive and limbic function that is impaired in HD. Our findings suggest that dysregulation of PDE10A-mediated intracellular signalling could be an early phenomenon in the course of HD with relevance also for extra-striatal brain areas.


Journal of Neurology, Neurosurgery, and Psychiatry | 2015

Short-interval observational data to inform clinical trial design in Huntington's disease

Nicola Z. Hobbs; Ruth Farmer; Em Rees; James H. Cole; Salman Haider; Ian B. Malone; Reiner Sprengelmeyer; Hans J. Johnson; Hans-Peter Mueller; Sigurd D. Süssmuth; Raymund A.C. Roos; Alexandra Durr; Chris Frost; Rachael I. Scahill; Bernhard Landwehrmeyer; Sarah J. Tabrizi

Objectives To evaluate candidate outcomes for disease-modifying trials in Huntingtons disease (HD) over 6-month, 9-month and 15-month intervals, across multiple domains. To present guidelines on rapid efficacy readouts for disease-modifying trials. Methods 40 controls and 61 patients with HD, recruited from four EU sites, underwent 3 T MRI and standard clinical and cognitive assessments at baseline, 6 and 15 months. Neuroimaging analysis included global and regional change in macrostructure (atrophy and cortical thinning), and microstructure (diffusion metrics). The main outcome was longitudinal effect size (ES) for each outcome. Such ESs can be used to calculate sample-size requirements for clinical trials for hypothesised treatment efficacies. Results Longitudinal changes in macrostructural neuroimaging measures such as caudate atrophy and ventricular expansion were significantly larger in HD than controls, giving rise to consistently large ES over the 6-month, 9-month and 15-month intervals. Analogous ESs for cortical metrics were smaller with wide CIs. Microstructural (diffusion) neuroimaging metrics ESs were also typically smaller over the shorter intervals, although caudate diffusivity metrics performed strongly over 9 and 15 months. Clinical and cognitive outcomes exhibited small longitudinal ESs, particularly over 6-month and 9-month intervals, with wide CIs, indicating a lack of precision. Conclusions To exploit the potential power of specific neuroimaging measures such as caudate atrophy in disease-modifying trials, we propose their use as (1) initial short-term readouts in early phase/proof-of-concept studies over 6 or 9 months, and (2) secondary end points in efficacy studies over longer periods such as 15 months.


Journal of Huntington's disease | 2015

Analysis of White Adipose Tissue Gene Expression Reveals CREB1 Pathway Altered in Huntington’s Disease

Andrew C. McCourt; Jennifer Parker; Edina Silajdžić; Salman Haider; Huma Sethi; Sarah J. Tabrizi; Thomas T. Warner; Maria Björkqvist

BACKGROUND In addition to classical neurological symptoms, Huntingtons disease (HD) is complicated by peripheral pathology and both the mutant gene and the protein are found in cells and tissues throughout the body. Despite the adipose tissue gene expression alterations described in HD mouse models, adipose tissue and its gene expression signature have not been previously explored in human HD. OBJECTIVE We investigated gene expression signatures in subcutaneous adipose tissue obtained from control subjects, premanifest HD gene carriers and manifest HD subjects with the aim to identify gene expression changes and signalling pathway alterations in adipose tissue relevant to HD. METHODS Gene expression was assessed using Affymetrix GeneChip® Human Gene 1.0 ST Array. Target genes were technically validated using real-time quantitative PCR and the expression signature was validated in an independent subject cohort. RESULTS In subcutaneous adipose tissue, more than 500 genes were significantly different in premanifest HD subjects as compared to healthy controls. Pathway analysis suggests that the differentially expressed genes found here in HD adipose tissue are involved in fatty acid metabolism pathways, angiotensin signalling pathways and immune pathways. Transcription factor analysis highlights CREB1. Using RT-qPCR, we found that MAL2, AGTR2, COBL and the transcription factor CREB1 were significantly upregulated, with CREB1 and AGT also being significantly upregulated in a separate cohort. CONCLUSIONS Distinct gene expression profiles can be seen in HD subcutaneous adipose tissue, with CREB1 highlighted as a key transcription factor.

Collaboration


Dive into the Salman Haider's collaboration.

Top Co-Authors

Avatar

Sarah J. Tabrizi

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralph Andre

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Edward J. Wild

University College London

View shared research outputs
Top Co-Authors

Avatar

Em Rees

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Nicola Z. Hobbs

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge