Network


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

Hotspot


Dive into the research topics where Louise Gregory is active.

Publication


Featured researches published by Louise Gregory.


The Journal of Clinical Endocrinology and Metabolism | 2012

Genetic Overlap in Kallmann Syndrome, Combined Pituitary Hormone Deficiency, and Septo-Optic Dysplasia

Taneli Raivio; Magdalena Avbelj; Mark J. McCabe; Christopher J. Romero; Andrew A. Dwyer; Johanna Tommiska; Gerasimos P. Sykiotis; Louise Gregory; Daniel Diaczok; Vaitsa Tziaferi; Mariet W. Elting; Raja Padidela; Lacey Plummer; Cecilia Martin; Bihua Feng; Chengkang Zhang; Qun-Yong Zhou; Huaibin Chen; Moosa Mohammadi; Richard Quinton; Yisrael Sidis; Sally Radovick; Mehul T. Dattani; Nelly Pitteloud

CONTEXT Kallmann syndrome (KS), combined pituitary hormone deficiency (CPHD), and septo-optic dysplasia (SOD) all result from development defects of the anterior midline in the human forebrain. OBJECTIVE The objective of the study was to investigate whether KS, CPHD, and SOD have shared genetic origins. DESIGN AND PARTICIPANTS A total of 103 patients with either CPHD (n = 35) or SOD (n = 68) were investigated for mutations in genes implicated in the etiology of KS (FGFR1, FGF8, PROKR2, PROK2, and KAL1). Consequences of identified FGFR1, FGF8, and PROKR2 mutations were investigated in vitro. RESULTS Three patients with SOD had heterozygous mutations in FGFR1; these were either shown to alter receptor signaling (p.S450F, p.P483S) or predicted to affect splicing (c.336C>T, p.T112T). One patient had a synonymous change in FGF8 (c.216G>A, p.T72T) that was shown to affect splicing and ligand signaling activity. Four patients with CPHD/SOD were found to harbor heterozygous rare loss-of-function variants in PROKR2 (p.R85G, p.R85H, p.R268C). CONCLUSIONS Mutations in FGFR1/FGF8/PROKR2 contributed to 7.8% of our patients with CPHD/SOD. These data suggest a significant genetic overlap between conditions affecting the development of anterior midline in the human forebrain.


The Journal of Clinical Endocrinology and Metabolism | 2011

Novel FGF8 mutations associated with recessive holoprosencephaly, craniofacial defects, and hypothalamo-pituitary dysfunction

Mark J. McCabe; Carles Gaston-Massuet; Vaitsa Tziaferi; Louise Gregory; Kyriaki S. Alatzoglou; Massimo Signore; Eduardo Puelles; Dianne Gerrelli; I. Sadaf Farooqi; Jamal Raza; Joanna Walker; Scott I. Kavanaugh; Pei-San Tsai; Nelly Pitteloud; Juan Pedro Martinez-Barbera; Mehul T. Dattani

CONTEXT Fibroblast growth factor (FGF) 8 is important for GnRH neuronal development with human mutations resulting in Kallmann syndrome. Murine data suggest a role for Fgf8 in hypothalamo-pituitary development; however, its role in the etiology of wider hypothalamo-pituitary dysfunction in humans is unknown. OBJECTIVE The objective of this study was to screen for FGF8 mutations in patients with septo-optic dysplasia (n = 374) or holoprosencephaly (HPE)/midline clefts (n = 47). METHODS FGF8 was analyzed by PCR and direct sequencing. Ethnically matched controls were then screened for mutated alleles (n = 480-686). Localization of Fgf8/FGF8 expression was analyzed by in situ hybridization in developing murine and human embryos. Finally, Fgf8 hypomorphic mice (Fgf8(loxPNeo/-)) were analyzed for the presence of forebrain and hypothalamo-pituitary defects. RESULTS A homozygous p.R189H mutation was identified in a female patient of consanguineous parentage with semilobar HPE, diabetes insipidus, and TSH and ACTH insufficiency. Second, a heterozygous p.Q216E mutation was identified in a female patient with an absent corpus callosum, hypoplastic optic nerves, and Moebius syndrome. FGF8 was expressed in the ventral diencephalon and anterior commissural plate but not in Rathkes pouch, strongly suggesting early onset hypothalamic and corpus callosal defects in these patients. This was consolidated by significantly reduced vasopressin and oxytocin staining neurons in the hypothalamus of Fgf8 hypomorphic mice compared with controls along with variable hypothalamo-pituitary defects and HPE. CONCLUSION We implicate FGF8 in the etiology of recessive HPE and potentially septo-optic dysplasia/Moebius syndrome for the first time to our knowledge. Furthermore, FGF8 is important for the development of the ventral diencephalon, hypothalamus, and pituitary.


The Journal of Clinical Endocrinology and Metabolism | 2013

Variations in PROKR2, But Not PROK2, Are Associated With Hypopituitarism and Septo-optic Dysplasia

Mark J. McCabe; Carles Gaston-Massuet; Louise Gregory; Kyriaki S. Alatzoglou; Vaitsa Tziaferi; Oualid Sbai; Philippe Rondard; Koh-hei Masumoto; Mamoru Nagano; Yasufumi Shigeyoshi; Marija Pfeifer; Tony Hulse; Charles Buchanan; Nelly Pitteloud; Juan Pedro Martinez-Barbera; Mehul T. Dattani

Context: Loss-of-function mutations in PROK2 and PROKR2 have been implicated in Kallmann syndrome (KS), characterized by hypogonadotropic hypogonadism and anosmia. Recent data suggest overlapping phenotypes/genotypes between KS and congenital hypopituitarism (CH), including septo-optic dysplasia (SOD). Objective: We screened a cohort of patients with complex forms of CH (n = 422) for mutations in PROK2 and PROKR2. Results: We detected 5 PROKR2 variants in 11 patients with SOD/CH: novel p.G371R and previously reported p.A51T, p.R85L, p.L173R, and p.R268C—the latter 3 being known functionally deleterious variants. Surprisingly, 1 patient with SOD was heterozygous for the p.L173R variant, whereas his phenotypically unaffected mother was homozygous for the variant. We sought to clarify the role of PROKR2 in hypothalamopituitary development through analysis of Prokr2−/− mice. Interestingly, these revealed predominantly normal hypothalamopituitary development and terminal cell differentiation, with the exception of reduced LH; this was inconsistent with patient phenotypes and more analogous to the healthy mother, although she did not have KS, unlike the Prokr2−/− mice. Conclusions: The role of PROKR2 in the etiology of CH, SOD, and KS is uncertain, as demonstrated by no clear phenotype-genotype correlation; loss-of-function variants in heterozygosity or homozygosity can be associated with these disorders. However, we report a phenotypically normal parent, homozygous for p.L173R. Our data suggest that the variants identified herein are unlikely to be implicated in isolation in these disorders; other genetic or environmental modifiers may also impact on the etiology. Given the phenotypic variability, genetic counseling may presently be inappropriate.


Endocrine development | 2012

Pituitary gland development: an update.

Rodrigo E. Bancalari; Louise Gregory; Mark J. McCabe; Mehul T. Dattani

The embryonic development of the pituitary gland involves a complex and highly spatio-temporally regulated network of integrating signalling molecules and transcription factors. Genetic mutations in any of these factors can lead to congenital hypopituitarism in association with a wide spectrum of craniofacial/midline defects ranging from incompatibility with life to holoprosencephaly (HPE) and cleft palate and septo-optic dysplasia (SOD). Increasing evidence supports a genotypic overlap with hypogonadotrophic hypogonadal disorders such as Kallmann syndrome, which is consistent with the known overlap in phenotypes between these disorders. This chapter reviews the cascade of events leading up to the successful development of the pituitary gland and to highlight key areas where genetic variations can occur thus leading to congenital hypopituitarism and associated defects.


Journal of Medical Genetics | 2015

Neuropathy target esterase impairments cause Oliver–McFarlane and Laurence–Moon syndromes

Robert B. Hufnagel; Gavin Arno; Nichole D. Hein; Joshua Hersheson; Megana K. Prasad; Y Anderson; Laura A. Krueger; Louise Gregory; Corinne Stoetzel; Tj Jaworek; Sarah Hull; Abi Li; Plagnol; Cm Willen; Tm Morgan; Cynthia A. Prows; Rs Hegde; Saima Riazuddin; Ga Grabowski; Rudy J. Richardson; K Dieterich; Taosheng Huang; Tamas Revesz; Juan Pedro Martinez-Barbera; Robert A. Sisk; C Jefferies; Henry Houlden; Mehul T. Dattani; John K. Fink; Hélène Dollfus

Background Oliver–McFarlane syndrome is characterised by trichomegaly, congenital hypopituitarism and retinal degeneration with choroidal atrophy. Laurence–Moon syndrome presents similarly, though with progressive spinocerebellar ataxia and spastic paraplegia and without trichomegaly. Both recessively inherited disorders have no known genetic cause. Methods Whole-exome sequencing was performed to identify the genetic causes of these disorders. Mutations were functionally validated in zebrafish pnpla6 morphants. Embryonic expression was evaluated via in situ hybridisation in human embryonic sections. Human neurohistopathology was performed to characterise cerebellar degeneration. Enzymatic activities were measured in patient-derived fibroblast cell lines. Results Eight mutations in six families with Oliver–McFarlane or Laurence–Moon syndrome were identified in the PNPLA6 gene, which encodes neuropathy target esterase (NTE). PNPLA6 expression was found in the developing human eye, pituitary and brain. In zebrafish, the pnpla6 curly-tailed morphant phenotype was fully rescued by wild-type human PNPLA6 mRNA and not by mutation-harbouring mRNAs. NTE enzymatic activity was significantly reduced in fibroblast cells derived from individuals with Oliver–McFarlane syndrome. Intriguingly, adult brain histology from a patient with highly overlapping features of Oliver–McFarlane and Laurence–Moon syndromes revealed extensive cerebellar degeneration and atrophy. Conclusions Previously, PNPLA6 mutations have been associated with spastic paraplegia type 39, Gordon–Holmes syndrome and Boucher–Neuhäuser syndromes. Discovery of these additional PNPLA6-opathies further elucidates a spectrum of neurodevelopmental and neurodegenerative disorders associated with NTE impairment and suggests a unifying mechanism with diagnostic and prognostic importance.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Transcription factor 7-like 1 is involved in hypothalamo–pituitary axis development in mice and humans

Carles Gaston-Massuet; Mark J. McCabe; Valeria Scagliotti; Rodrigo M. Young; Gabriela Carreno; Louise Gregory; Sujatha A. Jayakody; Sara Pozzi; Angelica Gualtieri; Basudha Basu; Markela Koniordou; Chun I. Wu; Rodrigo E. Bancalari; Elisa Rahikkala; Riitta Veijola; Tuija Lopponen; Federica Graziola; J.P.G. Turton; Massimo Signore; Seyedeh Neda Mousavy Gharavy; Nicoletta Charolidi; Sergei Y. Sokol; Cynthia L. Andoniadou; Stephen W. Wilson; Bradley J. Merrill; Mehul T. Dattani; Juan Pedro Martinez-Barbera

Significance The relevance of transcription factor 7-like 1 (TCF7L1) during hypothalamo–pituitary (HP) axis development remains unknown. Using mouse genetics, we show that TCF7L1 acts as a transcriptional repressor to regulate the expression of the hypothalamic signals involved in pituitary formation. In addition, we screened a cohort of human patients with forebrain and/or pituitary defects and report two independent missense variants, p.R92P and p.R400Q, in human TCF7L1. Functional studies in vitro and rescue experiments in zebrafish mutants deficient for tcf7l1a and tcf7l1b show that the p.R92P and p.R400Q variants exhibit reduced repressing activity compared with wild-type TCF7L1. In summary, we identify TCF7L1 as a determinant for the establishment of HP axis development and as a potential candidate gene to be mutated in congenital hypopituitarism. Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/β-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo–pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke’s pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with β-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.


The Journal of Clinical Endocrinology and Metabolism | 2013

Structural pituitary abnormalities associated with CHARGE syndrome.

Louise Gregory; Evelien F. Gevers; Joanne Baker; Tessa Kasia; Kling Chong; Dragana Josifova; María Caimari; Frédéric Bilan; Mark J. McCabe; Mehul T. Dattani

Introduction: CHARGE syndrome is a multisystem disorder that, in addition to Kallmann syndrome/isolated hypogonadotrophic hypogonadism, has been associated with anterior pituitary hypoplasia (APH). However, structural abnormalities such as an ectopic posterior pituitary (EPP) have not yet been described in such patients. Objective: The aims of the study were: 1) to describe the association between CHARGE syndrome and a structurally abnormal pituitary gland; and 2) to investigate whether CHD7 variants, which are identified in 65% of CHARGE patients, are common in septo-optic dysplasia /hypopituitarism. Methods: We describe 2 patients with features of CHARGE and EPP. CHD7 was sequenced in these and other patients with septo-optic dysplasia/hypopituitarism. Results: EPP, APH, and GH, TSH, and probable LH/FSH deficiency were present in 1 patient, and EPP and APH with GH, TSH, LH/FSH, and ACTH deficiency were present in another patient, both of whom had features of CHARGE syndrome. Both had variations in CHD7 that were novel and undetected in control cohorts or in the international database of CHARGE patients, but were also present in their unaffected mothers. No CHD7 variants were detected in the patients with septo-optic dysplasia/hypopituitarism without additional CHARGE features. Conclusion: We report a novel association between CHARGE syndrome and structural abnormalities of the pituitary gland in 2 patients with variations in CHD7 that are of unknown significance. However, CHD7 mutations are an uncommon cause of septo-optic dysplasia or hypopituitarism. Our data suggest the need for evaluation of pituitary function/anatomy in patients with CHARGE syndrome.


Clinical Endocrinology | 2015

The role of the sonic hedgehog signalling pathway in patients with midline defects and congenital hypopituitarism

Louise Gregory; Carles Gaston-Massuet; Cynthia L. Andoniadou; Gabriela Carreno; Emma A Webb; Mark J. McCabe; L Panagiotakopoulos; José W. Saldanha; Helen Spoudeas; J Torpiano; M Rossi; J Raine; Natalie Canham; Juan Pedro Martinez-Barbera; Mehul T. Dattani

The Gli family of zinc finger (GLI) transcription factors mediates the sonic hedgehog signalling pathway (HH) essential for CNS, early pituitary and ventral forebrain development in mice. Human mutations in this pathway have been described in patients with holoprosencephaly (HPE), isolated congenital hypopituitarism (CH) and cranial/midline facial abnormalities. Mutations in Sonic hedgehog (SHH) have been associated with HPE but not CH, despite murine studies indicating involvement in pituitary development.


Clinical Endocrinology | 2016

HESX1 Mutations in Patients with Congenital Hypopituitarism: Variable Phenotypes with the Same Genotype

Qing Fang; Anna Flavia Figueredo Benedetti; Qianyi Ma; Louise Gregory; Jun Li; Mehul T. Dattani; Abdollah Sadeghi-Nejad; Ivo J. P. Arnhold; Berenice B. Mendonca; Sally A. Camper; Luciani R. Carvalho

Mutations in the transcription factor HESX1 can cause isolated growth hormone deficiency (IGHD) or combined pituitary hormone deficiency (CPHD) with or without septo‐optic dysplasia (SOD). So far there is no clear genotype–phenotype correlation.


Molecular and Cellular Endocrinology | 2015

Novel application of luciferase assay for the in vitro functional assessment of KAL1 variants in three females with septo-optic dysplasia (SOD).

Mark J. McCabe; Youli Hu; Louise Gregory; Carles Gaston-Massuet; Kyriaki S. Alatzoglou; José W. Saldanha; Angelica Gualtieri; Ajay Thankamony; Ieuan A. Hughes; Sharron Townshend; Juan Pedro Martinez-Barbera; Pierre-Marc Bouloux; Mehul T. Dattani

KAL1 is implicated in 5% of Kallmann syndrome cases, a disorder which genotypically overlaps with septo-optic dysplasia (SOD). To date, a reporter-based assay to assess the functional consequences of KAL1 mutations is lacking. We aimed to develop a luciferase assay for novel application to functional assessment of rare KAL1 mutations detected in a screen of 422 patients with SOD. Quantitative analysis was performed using L6-myoblasts stably expressing FGFR1, transfected with a luciferase-reporter vector containing elements of the FGF-responsive osteocalcin promoter. The two variants assayed [p.K185N, p.P291T], were detected in three females with SOD (presenting with optic nerve hypoplasia, midline and pituitary defects). Our novel assay revealed significant decreases in transcriptional activity [p.K185N: 21% (p < 0.01); p.P291T: 40% (p < 0.001)]. Our luciferase-reporter assay, developed for assessment of KAL1 mutations, determined that two variants in females with hypopituitarism/SOD are loss-of-function; demonstrating that this assay is suitable for quantitative assessment of mutations in this gene.

Collaboration


Dive into the Louise Gregory's collaboration.

Top Co-Authors

Avatar

Mehul T. Dattani

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

Mark J. McCabe

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kyriaki S. Alatzoglou

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vaitsa Tziaferi

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angelica Gualtieri

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Emma A Webb

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

Helen Spoudeas

Great Ormond Street Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge