Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters











Publication year range
1.
Handb Clin Neurol ; 204: 95-109, 2024.
Article in English | MEDLINE | ID: mdl-39322397

ABSTRACT

Intracranial calcification (ICC) occurs in many neurologic disorders both acquired and genetic. In some inherited white matter disorders, it is a common or even invariable feature where the presence and pattern of calcification provides an important pointer to the specific diagnosis. This is particularly the case for the Aicardi-Goutières syndrome (AGS) and for Coats plus (CP) and leukoencephalopathy with calcifications and cysts (LCC), which are discussed in detail in this chapter. AGS is a genetic disorder of type 1 interferon regulation, caused by mutations in any of the nine genes identified to date. In its classic form, AGS has very characteristic clinical and neuroimaging features which will be discussed here. LCC is a purely neurologic disorder caused by mutations in the SNORD118 gene, whereas CP is a multisystem disorder of telomere function that may result from mutations in the CTC1, POT1, or STN genes. In spite of the different pathogenetic basis for LCC and CP, they share remarkably similar neuroimaging and neuropathologic features. Cockayne syndrome, in which ICC is usually present, is discussed elsewhere in this volume. ICC may occur as an occasional feature of many other white matter diseases, including Alexander disease, Krabbe disease, X-ALD, and occulodentodigital dysplasia.


Subject(s)
Calcinosis , Leukoencephalopathies , Nervous System Malformations , Humans , Calcinosis/genetics , Calcinosis/diagnostic imaging , Calcinosis/pathology , Leukoencephalopathies/genetics , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/pathology , Nervous System Malformations/genetics , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/pathology , Child , Central Nervous System Cysts/genetics , Central Nervous System Cysts/diagnostic imaging , Central Nervous System Cysts/pathology , Autoimmune Diseases of the Nervous System
2.
Cureus ; 16(9): e69158, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39268024

ABSTRACT

Coats plus syndrome (CPS) is an exceedingly rare genetic disorder associated with premature telomere shortening. The syndrome, also known as cerebroretinal microangiopathy with calcifications and cysts, has a multisystemic manifestation. It may present as brain abnormalities, seizures, osteopenia, prenatal and postnatal growth deficiency, and portal hypertension, among others. Up to 40% of affected individuals manifest recurrent gastrointestinal (GI) bleeding which can be life-threatening in some cases. Treatment for GI bleeding is not standardized and is therefore individualized based on the patient's clinical status, comorbidities, and resource availability. We herein present a case of a 20-year-old female with CPS and a two-year history of severe recurrent GI bleeding unable to be identified by conventional endoscopy. This report highlights successful laparoscopic assisted enteroscopy with enterectomy as a novel diagnostic and therapeutic modality in this population.

3.
J Vitreoretin Dis ; 7(6): 562-564, 2023.
Article in English | MEDLINE | ID: mdl-37974921

ABSTRACT

Purpose: To present a case of retinal vascular disease characterized primarily by capillary nonperfusion in an adult with Coats plus syndrome (CPS). Methods: A case and its findings were analyzed. Results: A 38-year-old woman with a history of poliosis, thrombocytopenia, seizures, and white-matter brain lesions was referred for evaluation of bilateral blurred central vision. Fluorescein angiography showed extensive bilateral retinal capillary nonperfusion with retinal arteriolitis in the right eye. Genetic testing found 2 pathological mutations in the conserved telomere maintenance component 1 (CTC1) gene, diagnostic of CPS. Conclusions: Genetic testing may be diagnostic in patients who present with retinal vascular disease and systemic disease suggestive of CPS.

4.
Genes (Basel) ; 14(9)2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37761957

ABSTRACT

Leukoencephalopathy with calcifications and cysts (LCC) is a rare autosomal recessive disorder showing a pediatric or adult onset. First described in 1996 by Labrune and colleagues, it was only in 2016 that bi-allelic variants in a non-protein coding gene, SNORD118, were found as the cause for LCC, differentiating this syndrome from coats plus (CP). SNORD118 transcribes for a small nucleolar RNA, which is necessary for correct ribosome biogenesis, hence the classification of LCC among ribosomopathies. The syndrome is characterized by a combination of white matter hyperintensities, calcifications, and cysts on brain MRI with varying neurological signs. Corticosteroids, surgery, and recently bevacizumab, have been tried with unclear results since the natural history of the disease remains elusive. To date, 67 patients with a pediatric onset of disease have been described in the literature, with a clinical-radiological follow-up carried out in only eleven of them. We described the clinical-radiological follow-up from birth to almost five years of age of a late-preterm patient diagnosed with LCC and carried out a thorough overview of pediatric patients described in the literature. It is important to gather serial clinical-radiological data from other patients to depict the natural history of this disease, aiming to deeply depict genotype-phenotype correlations and make the role of new therapeutics clearer.

5.
J Vitreoretin Dis ; 7(1): 74-78, 2023.
Article in English | MEDLINE | ID: mdl-37008390

ABSTRACT

Purpose: A premature infant was diagnosed with Coats plus syndrome based on a genetic evaluation showing biallelic heterozygous pathogenic CTC1 variants. Methods: A case study was performed, including findings and interventions. Results: A premature infant born 30 weeks gestational age weighing 817 g was evaluated for retinopathy of prematurity at 35 weeks corrected gestational age. An initial dilated fundus examination showed an exudative retinal detachment (RD) in the right eye and avascularity post-equatorially in the left eye with telangiectasias and aneurysmal dilations. Genetic evaluation showed biallelic heterozygous pathogenic CTC1 variants, diagnostic of Coats plus syndrome. Sequential examination under anesthesia with fluorescein showed progressive ischemia despite confluent photocoagulation. Conclusions: CTC1 gene variants manifest as Coats plus syndrome, which has a clinical appearance consistent with retinovascular ischemia, capillary remodeling, aneurysmal dilation, and exudative RD. Systemic and local corticosteroids in conjunction with peripheral laser ablation decreased vascular exudation and avoided intraocular intervention.

6.
Am J Ophthalmol Case Rep ; 28: 101713, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36177296

ABSTRACT

Purpose: To describe a case of Coats Plus Syndrome (CPS), a vision and life threatening disease belonging to a family of diseases known as the Telomere Biology Disorders. Observations: A 15-year-old girl with a history of small for gestational age, short stature, microcephaly, thinning/greying of scalp hair, skin hyperpigmentation, nail ridging, and multiple pathological fractures presented with bilateral Coats-like retinopathy. We discovered a new observation of multiple peripheral pinpoint retinal pigment epithelial detachments (PEDs). Further genetic testing revealed CTC1 gene mutation and she was diagnosed with Coats plus syndrome with features of dyskeratosis congenita, a telomere biology disorder. Conclusion and importance: Patients with bilateral Coats-like retinopathy and associated systemic features suggestive of CPS should be evaluated through genetic testing to diagnose this disease and treat vision and life threatening manifestations as early as possible. In this report, we also document, for the first time, multiple pinpoint PEDs that could be related to an accelerated aging process with telomere dysfunction.

7.
Genes (Basel) ; 13(8)2022 08 05.
Article in English | MEDLINE | ID: mdl-36011306

ABSTRACT

Coats plus (CP) syndrome is an inherited autosomal recessive condition that results from mutations in the conserved telomere maintenance component 1 gene (CTC1). The CTC1 protein functions as a part of the CST protein complex, a protein heterotrimer consisting of CTC1-STN1-TEN1 which promotes telomere DNA synthesis and inhibits telomerase-mediated telomere elongation. However, it is unclear how CTC1 mutations may have an effect on telomere structure and function. For that purpose, we established the very first induced pluripotent stem cell lines (iPSCs) from a compound heterozygous patient with CP carrying deleterious mutations in both alleles of CTC1. Telomere dysfunction and chromosomal instability were assessed in both circulating lymphocytes and iPSCs from the patient and from healthy controls of similar age. The circulating lymphocytes and iPSCs from the CP patient were characterized by their higher telomere length heterogeneity and telomere aberrations compared to those in control cells from healthy donors. Moreover, in contrast to iPSCs from healthy controls, the high levels of telomerase were associated with activation of the alternative lengthening of telomere (ALT) pathway in CP-iPSCs. This was accompanied by inappropriate activation of the DNA repair proteins γH2AX, 53BP1, and ATM, as well as with accumulation of DNA damage, micronuclei, and anaphase bridges. CP-iPSCs presented features of cellular senescence and increased radiation sensitivity. Clonal dicentric chromosomes were identified only in CP-iPSCs after exposure to radiation, thus mirroring the role of telomere dysfunction in their formation. These data demonstrate that iPSCs derived from CP patients can be used as a model system for molecular studies of the CP syndrome and underscores the complexity of telomere dysfunction associated with the defect of DNA repair machinery in the CP syndrome.


Subject(s)
DNA Repair-Deficiency Disorders , Induced Pluripotent Stem Cells , Telomerase , Ataxia , Brain Neoplasms , Calcinosis , Central Nervous System Cysts , Humans , Induced Pluripotent Stem Cells/metabolism , Leukoencephalopathies , Muscle Spasticity , Retinal Diseases , Seizures , Telomerase/genetics , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis/genetics
8.
Ophthalmic Genet ; 43(4): 543-549, 2022 08.
Article in English | MEDLINE | ID: mdl-35416114

ABSTRACT

BACKGROUND: Coats plus syndrome or cerebroretinal microangiopathy with calcifications and cysts (CMCC) is an exceedingly rare autosomal recessive disorder that predominantly affects the microvasculature in the retina, brain, bones, and gastrointestinal system. Unlike Coats disease, CMCC is bilateral and affects multiple organ systems. MATERIALS AND METHODS: Case report. RESULTS: We report the case of two brothers with Coats Plus syndrome who presented with variable phenotypic expression. One sibling (Patient 1) was thought to have atypical retinopathy of prematurity and was only diagnosed with Coats plus after his older brother (Patient 2) presented with a seizure and a left upper extremity tremor at 4 years of age. The CTC1 mutation was confirmed in both patients. Aggressive treatment with laser photocoagulation and intravitreal bevacizumab dramatically improved the retinal vascular and exudative changes. CONCLUSION: Coats Plus syndrome can have a variable phenotypic presentation, including retinal vascular findings. This rare genetic disease should be in the differential diagnosis in patients who present with atypical retinal pathology, including Retinopathy of Prematurity, Familial Exudative Vitreoretinopathy, or Coats disease associated with non-specific multiorgan abnormalities.


Subject(s)
Central Nervous System Cysts , Leukoencephalopathies , Retinal Telangiectasis , Retinopathy of Prematurity , Ataxia , Brain Neoplasms , Calcinosis , Central Nervous System Cysts/genetics , Humans , Infant, Newborn , Laser Coagulation , Leukoencephalopathies/genetics , Male , Muscle Spasticity , Retinal Diseases , Retinal Telangiectasis/diagnosis , Retinal Telangiectasis/genetics , Retinal Telangiectasis/therapy , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/genetics , Seizures
9.
BMC Pediatr ; 22(1): 119, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260125

ABSTRACT

BACKGROUND: Coats plus syndrome, cerebroretinal microangiopathy with calcifications and cysts, is a rare disease with autosomal recessive pattern occurring due to a mutation in CTC1, encoding conserved telomere maintenance component 1, gene. Besides retinal involvement, abnormalities in brain and osteopenia, serious life-threatening bleeding in gastrointestinal tract and portal hypertension can be observed. CASE PRESENTATION: A 6-year-old girl with Coats plus syndrome presented to the pediatric emergency department with vomiting blood and blood in stool. An upper and lower gastrointestinal endoscopy revealed esophageal varices and vascular telangiectasia in the pyloric antrum, duodenum, and colon. She received palliative care and the bleeding was stopped after receiving intravenous octreotide. She then was followed in the pediatric gastroenterology, neurology, and ophthalmology clinics. She was later hospitalized and admitted to the intensive care unit as she continued to have intermittent gastrointestinal system bleeding. She eventually died due to severe gastrointestinal system bleeding. CONCLUSIONS: Coats plus syndrome can lead to life-threatening gastrointestinal bleeding and portal hypertension. As Coats plus syndrome is quite rare, there is little published data on this syndrome. This report presents a case of Coats plus syndrome as a rare cause of gastrointestinal bleeding and portal hypertension.


Subject(s)
Central Nervous System Cysts , Hypertension, Portal , Ataxia , Brain Neoplasms , Calcinosis , Central Nervous System Cysts/genetics , Child , Female , Gastrointestinal Hemorrhage/etiology , Humans , Leukoencephalopathies , Muscle Spasticity , Retinal Diseases , Seizures
10.
Turk J Pediatr ; 64(1): 166-170, 2022.
Article in English | MEDLINE | ID: mdl-35286046

ABSTRACT

BACKGROUND: Cerebroretinal microangiopathy with calcifications and cysts formerly known as Coats plus syndrome is a rare multisystemic autosomal recessive disease that affects the eyes, brain, bone, and gastrointestinal system. Intestinal telangiectasia are components of vascular malformations characterized by gastrointestinal system bleedings. Recurrent gastrointestinal system bleedings have been reported as being due to hepatic failure or vascular malformations of the gastrointestinal system tract. CASE: Here we report a patient who presented with recurrent gastrointestinal system bleeding episodes, bilateral exudative retinopathy, intracranial calcification and was diagnosed with Coats plus syndrome. Recurrent gastrointestinal system bleeding was controlled by monthly octreotide treatment. CONCLUSIONS: Coats plus syndrome presenting with vascular malformations should always be kept in mind in a patient with recurrent gastrointestinal bleeding and accompanying systemic physical findings. Octreotide treatment is an important option for patients with life threatening gastrointestinal system bleeding. Long term use of octreotide treatment can be used successfully in selected pediatric cases.


Subject(s)
Central Nervous System Cysts , Vascular Malformations , Ataxia , Brain Neoplasms , Calcinosis , Child , Gastrointestinal Hemorrhage/diagnosis , Gastrointestinal Hemorrhage/etiology , Humans , Leukoencephalopathies , Muscle Spasticity , Octreotide/therapeutic use , Retinal Diseases , Seizures , Syndrome
11.
Horm Res Paediatr ; 94(11-12): 448-455, 2021.
Article in English | MEDLINE | ID: mdl-34706368

ABSTRACT

Coats plus syndrome is an autosomal recessive multisystemic and pleiotropic disorder affecting the eyes, brain, bone, and gastrointestinal tract, usually caused by compound heterozygous variants of the conserved telomere maintenance component 1 gene (CTC1), involved in telomere homeostasis and replication. So far, most reported patients are compound heterozygous for a truncating mutation and a missense variant. The phenotype is believed to result from telomere dysfunction, with accumulation of DNA damage, cellular senescence, and stem cell depletion. Here, we report a 23-year-old female with prenatal and postnatal growth retardation, microcephaly, osteopenia, recurrent fractures, intracranial calcification, leukodystrophy, parenchymal brain cysts, bicuspid aortic valve, and primary ovarian failure. She carries a previously reported maternally inherited pathogenic variant in exon 5 (c.724_727del, p.(Lys242Leufs*41)) and a novel, paternally inherited splice site variant (c.1617+5G>T; p.(Lys480Asnfs*17)) in intron 9. CTC1 transcript analysis showed that the latter resulted in skipping of exon 9. A trace of transcripts was normally spliced resulting in the presence of a low level of wild-type CTC1 transcripts. We speculate that ovarian failure is caused by telomere shortening or chromosome cohesion failure in oocytes and granulosa cells, with early decrease in follicular reserve. This is the first patient carrying 2 truncating CTC1 variants and the first presenting primary ovarian failure.


Subject(s)
Calcinosis , Central Nervous System Cysts , Leukoencephalopathies , Ataxia/genetics , Ataxia/pathology , Brain Neoplasms , Calcinosis/genetics , Central Nervous System Cysts/genetics , Central Nervous System Cysts/pathology , Female , Humans , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Muscle Spasticity , Mutation , Retinal Diseases , Seizures , Telomere-Binding Proteins/genetics
12.
Best Pract Res Clin Haematol ; 34(2): 101282, 2021 06.
Article in English | MEDLINE | ID: mdl-34404536

ABSTRACT

Telomeropathies or telomere biology disorders (TBDs) are a group of rare diseases characterised by altered telomere maintenance. Most patients with TBDs show pathogenic variants of genes that encode factors involved in the prevention of telomere shortening. Particularly in adults, TBDs mostly present themselves with heterogeneous clinical features that often include bone marrow failure, hepatopathies, interstitial lung disease and other organ sites. Different degrees of severity are also observed among patients with TBDs, ranging from very severe syndromes manifesting themselves in early childhood, such as Revesz syndrome, Hoyeraal-Hreidarsson syndrome, and Coats plus disease, to dyskeratosis congenita (DKC) and adult-onset "cryptic" forms of TBD, which often affect fewer organ systems. Overall, the most relevant clinical complications of TBD are bone marrow failure, lung fibrosis, and liver cirrhosis. In this review, we summarise recent advances in the management and treatment of TBD and provide a brief overview of the various treatment approaches.


Subject(s)
Dyskeratosis Congenita , Intellectual Disability , Microcephaly , Adult , Child, Preschool , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/therapy , Fetal Growth Retardation , Humans , Telomere
13.
Mol Genet Genomic Med ; 9(12): e1708, 2021 12.
Article in English | MEDLINE | ID: mdl-34110109

ABSTRACT

AIM: Coats plus syndrome (CP) is a rare autosomal recessive disorder, characterised by retinal telangiectasia exudates (Coats disease), leukodystrophy, distinctive intracranial calcification and cysts, as well as extra-neurological features including abnormal vasculature of the gastrointestinal tract, portal hypertension and osteopenia with a tendency to fractures. CP most frequently occurs due to loss-of-function mutations in CTC1. The encoded protein CTC1 constitutes part of the CST (CTC1-STN1-TEN1) complex, and three patients have been described with CP due to biallelic mutations in STN1. Together with the identification of homozygosity for a specific loss-of-function mutation in POT1 in a sibling pair, these observations highlight a defect in the maintenance of telomere integrity as the cause of CP, although the precise mechanism leading to the micro-vasculopathy seen at a pathological level remains unclear. Here, we present the investigation of a fourth child who presented to us with retinal exudates, intracranial calcifications and developmental delay, in keeping with a diagnosis of CP, and later went on to develop pancytopenia and gastrointestinal bleeding. Genome sequencing revealed compound heterozygous variants in STN1 as the likely genetic cause of CP in this present case. METHODS: We assessed the phenotype to be CP and undertook targeted sequencing. RESULTS: Whilst sequencing of CTC1 and POT1 was normal, we identified novel compound heterozygous variants in STN1 (previous gene symbol OBFC1): one loss-of-function--c.894dup (p.(Asp299Argfs*58)); and one missense--c.707T>C (p.(Leu236Pro)). CONCLUSION: Given the clinical phenotype and identified variants we suggest that this is only the fourth patient reported to date with CP due to mutations in STN1.


Subject(s)
Ataxia/diagnosis , Ataxia/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Calcinosis/diagnosis , Calcinosis/genetics , Central Nervous System Cysts/diagnosis , Central Nervous System Cysts/genetics , Genetic Predisposition to Disease , Heterozygote , Leukoencephalopathies/diagnosis , Leukoencephalopathies/genetics , Muscle Spasticity/diagnosis , Muscle Spasticity/genetics , Mutation , Retinal Diseases/diagnosis , Retinal Diseases/genetics , Seizures/diagnosis , Seizures/genetics , Telomere-Binding Proteins/genetics , Alleles , Child , DNA Mutational Analysis , Genetic Association Studies , Humans , Magnetic Resonance Angiography , Male , Models, Molecular , Neuroimaging , Phenotype , Protein Conformation , Structure-Activity Relationship , Telomere-Binding Proteins/chemistry , Tomography, X-Ray Computed
14.
J Pediatr ; 230: 55-61.e4, 2021 03.
Article in English | MEDLINE | ID: mdl-32971146

ABSTRACT

OBJECTIVE: To describe the clinical features, therapeutic interventions, and patient outcomes of gastrointestinal (GI) hemorrhage in individuals with a telomere biology disorder, including dyskeratosis congenita, Hoyeraal-Hreidarsson syndrome, Revesz syndrome, and Coats plus. STUDY DESIGN: Clinical Care Consortium for Telomere Associated Ailments members were invited to contribute data on individuals with telomere biology disorders at their institutions who experienced GI bleeding. Patient demographic, laboratory, imaging, procedural, and treatment information and outcomes were extracted from the medical record. RESULTS: Sixteen patients who experienced GI hemorrhage were identified at 11 centers. Among 14 patients who underwent genetic testing, 8 had mutations in TINF2, 4 had mutations in CTC1 or STN1, and 1 patient each had a mutation in TERC and RTEL1. Ten patients had a history of hematopoietic cell transplantation. The patients with Coats plus and those without Coats plus had similar clinical features and courses. Angiodysplasia of the stomach and/or small bowel was described in 8 of the 12 patients who underwent endoscopy; only 4 had esophageal varices. Various medical interventions were trialed. No single intervention was uniformly associated with cessation of bleeding, although 1 patient had a sustained response to treatment with bevacizumab. Recurrence was common, and the overall long-term outcome for affected patients was poor. CONCLUSIONS: GI bleeding in patients with telomere biology disorders is associated with significant morbidity and with vascular ectasias rather than varices.


Subject(s)
Gastrointestinal Hemorrhage/etiology , Telomere/genetics , Adolescent , Adult , Ataxia/complications , Ataxia/genetics , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/genetics , Bone Marrow/abnormalities , Brain Neoplasms/complications , Brain Neoplasms/genetics , Calcinosis/complications , Calcinosis/genetics , Central Nervous System Cysts/complications , Central Nervous System Cysts/genetics , Child , Child, Preschool , Dyskeratosis Congenita/complications , Dyskeratosis Congenita/genetics , Female , Fetal Growth Retardation/genetics , Gastrointestinal Hemorrhage/genetics , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Male , Microcephaly/complications , Microcephaly/genetics , Muscle Spasticity/complications , Muscle Spasticity/genetics , Mutation , Retina , Retinal Diseases/complications , Retinal Diseases/genetics , Seizures/complications , Seizures/genetics , Telomere/metabolism , Telomere/pathology , Young Adult
15.
Ophthalmic Genet ; 42(1): 79-83, 2021 02.
Article in English | MEDLINE | ID: mdl-33034244

ABSTRACT

BACKGROUND: Coats plus syndrome is a rare multisystem disorder, and is also a telomere-related disorder caused by CTC1 gene mutation. We reported ophthalmic findings in a Chinese child with genetically confirmed Coats plus syndrome. MATERIALS AND METHODS: The comprehensive ophthalmic findings were presented, as well as treatment history and systemic manifestations. In addition, genetic testing was performed to confirm the diagnosis. RESULTS: Examination under anesthesia showed notable retinal vasculopathy, including vascular tortuosity and dilation, abnormal vascular anastomosis, retinal telangiectasias and mild exudation, extensive peripheral avascularity, as well as the presence of retinal neovascularization. The patient developed vitreous hemorrhage and tractional retinal detachment, and then underwent vitrectomy. Meanwhile, the patient was noted to have growth retardation and leukoencephalopathy. Gene testing identified a compound heterozygous mutation in CTC1 gene: a novel splicing site mutation (c.33 + 1 G > T) and a deletion mutation (c.2954_2956del, p.C985del), which were inherited from his mother and father, respectively. CONCLUSIONS: The present report expanded the genotype and phenotype spectrum of CTC1 gene associated with Coats plus syndrome.


Subject(s)
Ataxia/pathology , Brain Neoplasms/pathology , Calcinosis/pathology , Central Nervous System Cysts/pathology , Leukoencephalopathies/pathology , Muscle Spasticity/pathology , Mutation , Phenotype , Retinal Diseases/pathology , Seizures/pathology , Telomere-Binding Proteins/genetics , Ataxia/genetics , Brain Neoplasms/genetics , Calcinosis/genetics , Central Nervous System Cysts/genetics , Child, Preschool , Humans , Infant , Leukoencephalopathies/genetics , Male , Muscle Spasticity/genetics , Retinal Diseases/genetics , Seizures/genetics
16.
Am J Med Genet A ; 185(1): 15-25, 2021 01.
Article in English | MEDLINE | ID: mdl-33029936

ABSTRACT

Biallelic mutations in SNORD118, encoding the small nucleolar RNA U8, cause leukoencephalopathy with calcifications and cysts (LCC). Given the difficulty in interpreting the functional consequences of variants in nonprotein encoding genes, and the high allelic polymorphism across SNORD118 in controls, we set out to provide a description of the molecular pathology and clinical spectrum observed in a cohort of patients with LCC. We identified 64 affected individuals from 56 families. Age at presentation varied from 3 weeks to 67 years, with disease onset after age 40 years in eight patients. Ten patients had died. We recorded 44 distinct, likely pathogenic, variants in SNORD118. Fifty two of 56 probands were compound heterozygotes, with parental consanguinity reported in only three families. Forty nine of 56 probands were either heterozygous (46) or homozygous (three) for a mutation involving one of seven nucleotides that facilitate a novel intramolecular interaction between the 5' end and 3' extension of precursor-U8. There was no obvious genotype-phenotype correlation to explain the marked variability in age at onset. Complementing recently published functional analyses in a zebrafish model, these data suggest that LCC most often occurs due to combinatorial severe and milder mutations, with the latter mostly affecting 3' end processing of precursor-U8.


Subject(s)
Calcinosis/genetics , Genetic Association Studies , Leukoencephalopathies/genetics , RNA, Small Nucleolar/genetics , Adolescent , Adult , Aged , Animals , Calcinosis/complications , Calcinosis/pathology , Child , Child, Preschool , Consanguinity , Disease Models, Animal , Female , Heterozygote , Humans , Infant , Infant, Newborn , Leukoencephalopathies/complications , Leukoencephalopathies/pathology , Male , Middle Aged , Pathology, Molecular , Young Adult , Zebrafish/genetics
17.
Rom J Ophthalmol ; 65(3): 247-254, 2021.
Article in English | MEDLINE | ID: mdl-35036645

ABSTRACT

We present the case of a 20-year-old girl with severe combined seronegative immunodeficiency who developed a bilateral decrease in visual acuity due to retinal necrosis. After further investigations, increased serological viral levels of Cytomegalovirus (CMV) were detected and confirmed the diagnosis of CMV retinitis in both eyes. After three weeks of systemic therapy with oral valganciclovir, her condition improved, with the best corrected visual acuity of the most affected eye changing from finger counting at presentation to 6/ 12. Although financial matters determined her to discontinue the antiviral treatment after three months, her ophthalmological status remained stable, and she resumed therapy after four weeks of pause. At the four months follow-up, despite an unchanged visual function, her general condition deteriorated. In the absence of appropriate treatment for her immunodeficiency both the patient's ophthalmological and systemic prognosis were poor.


Subject(s)
Cytomegalovirus Retinitis , HIV Infections , Adult , Antiviral Agents/therapeutic use , Cytomegalovirus Retinitis/diagnosis , Cytomegalovirus Retinitis/drug therapy , Female , Ganciclovir/therapeutic use , HIV Infections/drug therapy , Humans , Retrospective Studies , Young Adult
18.
Am J Med Genet A ; 182(9): 2139-2144, 2020 09.
Article in English | MEDLINE | ID: mdl-32627942

ABSTRACT

The role of the CTC1-STN1-TEN1 (CST) complex in Coats plus syndrome (CP), as well as other telomeropathy-phenotypes and disorders of genome instability is well documented. We report an Indian child with a clinical diagnosis of CP who presented to us with retinal exudates, extensive cerebral calcification, developmental delay and severe anemia consequent upon chronic gastrointestinal (GI) bleeding. Whole exome sequencing revealed compound heterozygous variants in STN1 as the probable genetic cause leading to CP in the present case. Of the two variants, the nonsense variant c.397C>T (p.Arg133*) was a truncating variant leading to loss of full protein length whereas the second variant c.985G>C (p.Ala329Pro) was novel and neither reported in ExAC, 1KGP or gnomAD. The deleteriousness of the novel variant was explored through molecular dynamics simulation analysis where p.Ala329Pro mutation affected C-terminal domain interaction between STN1 and TEN1 complex. Hormonal therapy using ethinyl estradiol and norethisterone was apparently associated with a clinically useful, although poorly sustained, decrease in blood transfusion requirement in the proband.


Subject(s)
Ataxia/genetics , Brain Neoplasms/genetics , Calcinosis/genetics , Central Nervous System Cysts/genetics , Leukoencephalopathies/genetics , Muscle Spasticity/genetics , Retinal Diseases/genetics , Seizures/genetics , Telomere-Binding Proteins/genetics , Asian People/genetics , Ataxia/pathology , Brain Neoplasms/pathology , Calcinosis/pathology , Central Nervous System Cysts/pathology , DNA Replication/genetics , Female , Humans , Infant , Leukoencephalopathies/pathology , Muscle Spasticity/pathology , Mutation/genetics , Phenotype , Retinal Diseases/pathology , Seizures/pathology , Telomere/genetics , Telomere Homeostasis/genetics
19.
Ophthalmic Genet ; 41(4): 363-367, 2020 08.
Article in English | MEDLINE | ID: mdl-32543263

ABSTRACT

Coats plus syndrome (CP) is a rare condition characterized by bilateral exudative retinal telangiectasias with associated systemic disorders primarily affecting the brain, bone and gastrointestinal tract due to a mutation in the CTC1 gene. CTC1 mutations are also known to cause dyskeratosis congenita (DC), which is an inherited bone marrow failure syndrome characterized by skin pigmentation abnormalities, nail dystrophy, and oral leukoplakia. This is the first reported case of a patient diagnosed with both CP and DC caused by compound heterozygous CTC1 gene mutations. Moreover, one of the variant mutations found in this patient has never been published before.


Subject(s)
Ataxia/pathology , Brain Neoplasms/pathology , Calcinosis/pathology , Central Nervous System Cysts/pathology , Dyskeratosis Congenita/pathology , Leukoencephalopathies/pathology , Muscle Spasticity/pathology , Mutation , Retinal Diseases/pathology , Seizures/pathology , Telomere-Binding Proteins/genetics , Ataxia/complications , Ataxia/genetics , Brain Neoplasms/complications , Brain Neoplasms/genetics , Calcinosis/complications , Calcinosis/genetics , Central Nervous System Cysts/complications , Central Nervous System Cysts/genetics , Dyskeratosis Congenita/complications , Dyskeratosis Congenita/genetics , Female , Humans , Infant , Leukoencephalopathies/complications , Leukoencephalopathies/genetics , Muscle Spasticity/complications , Muscle Spasticity/genetics , Prognosis , Retinal Diseases/complications , Retinal Diseases/genetics , Seizures/complications , Seizures/genetics
20.
Expert Rev Hematol ; 12(12): 1037-1052, 2019 12.
Article in English | MEDLINE | ID: mdl-31478401

ABSTRACT

Introduction: Telomere biology disorders (TBDs) encompass a group of illnesses caused by germline mutations in genes regulating telomere maintenance, resulting in very short telomeres. Possible TBD manifestations range from complex multisystem disorders with onset in childhood such as dyskeratosis congenita (DC), Hoyeraal-Hreidarsson syndrome, Revesz syndrome and Coats plus to adults presenting with one or two DC-related features.Areas covered: The discovery of multiple genetic causes and inheritance patterns has led to the recognition of a spectrum of clinical features affecting multiple organ systems. Patients with DC and associated TBDs are at high risk of bone marrow failure, cancer, liver and pulmonary disease. Recently, vascular diseases, including pulmonary arteriovenous malformations and gastrointestinal telangiectasias, have been recognized as additional manifestations. Diagnostics include detection of very short leukocyte telomeres and germline genetic testing. Hematopoietic cell transplantation and lung transplantation are the only current therapeutic modalities but are complicated by numerous comorbidities. This review summarizes the pathophysiology underlying TBDs, associated clinical features, management recommendations and therapeutic options.Expert opinion: Understanding TBDs as complex, multisystem disorders with a heterogenous genetic background and diverse phenotypes, highlights the importance of clinical surveillance and the urgent need to develop new therapeutic strategies to improve health outcomes.


Subject(s)
Dyskeratosis Congenita , Germ-Line Mutation , Telomere , Dyskeratosis Congenita/diagnosis , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/metabolism , Dyskeratosis Congenita/pathology , Humans , Telomere/genetics , Telomere/metabolism , Telomere/pathology
SELECTION OF CITATIONS
SEARCH DETAIL