Sporadic facial angiofibroma and sporadic angiomyolipoma mimicking tuberous sclerosis complex

2021 ◽  
pp. jmedgenet-2021-108160
Author(s):  
Katarzyna Klonowska ◽  
Elizabeth A. Thiele ◽  
Joannes M. Grevelink ◽  
Aaron R. Thorner ◽  
David J. Kwiatkowski

Tuberous sclerosis complex (TSC) is a genetic syndrome due to mutations in either TSC1 or TSC2, leading to the development of hamartomatous tumours at multiple body sites, including facial skin (facial angiofibroma (FAF)), brain (cortical tubers) and kidney (angiomyolipoma (AML)). In this report, we describe an individual with minimal TSC clinical features, who had ‘no mutation identified’ (NMI) by prior genetic testing in a clinical laboratory. Our massively parallel sequencing (MPS) analysis of multiple samples from different body sites and tumours (including blood, saliva, normal skin, AML and FAF) revealed an extraordinary situation in which FAF and AML had completely independent inactivating biallelic variants in TSC2, not present in other matched samples. This suggests that the two different lesions (AML and FAF) are not due to the same underlying germline or mosaic mutation, rather both are likely sporadic events. This case demonstrates the relevance of thorough clinical examination, high-coverage MPS of multiple tumours and matched normal tissues, and appropriate genetic counselling for individuals with marginal TSC features and possible TSC1 or TSC2 mosaicism.

2021 ◽  
Vol 749 ◽  
pp. 135714
Author(s):  
Kefu Wu ◽  
Jiong Yue ◽  
Kaifeng Shen ◽  
Jiaojiang He ◽  
Gang Zhu ◽  
...  

2018 ◽  
pp. bcr-2018-226244
Author(s):  
Lalit Takia ◽  
Kana Ram Jat ◽  
Anirban Mandal ◽  
Sushil Kumar Kabra

Lymphangioleiomyomatosis (LAM) either sporadic or a part of tuberous sclerosis complex is rare in paediatric age group. Here, we report a case of LAM with tuberous sclerosis in an infant. She was referred to our institute at the age of 4 months as a case of recurrent bilateral pneumothorax requiring intercostal tube drainage. Detailed history revealed that patient was symptomatic since 1 month of age in the form of seizures. She had respiratory symptoms for last 15 days. General physical examination revealed whitish macular patches. Brain imaging was suggestive of cortical tubers and subependymal nodules. The echocardiography showed right atrial rhabdomyoma. Chest CT revealed multiple cysts suggesting LAM. On the basis of above findings, a diagnosis of tuberous sclerosis complex with LAM was made. The infant was started on sirolimus and there was significant clinical and radiological improvement over a period of 2 and half years without any side effects.


2020 ◽  
Author(s):  
Oliver L. Eichmüller ◽  
Nina S. Corsini ◽  
Ábel Vértesy ◽  
Theresa Scholl ◽  
Victoria-Elisabeth Gruber ◽  
...  

SummaryAlthough the intricate and prolonged development of the human brain critically distinguishes it from other mammals1, our current understanding of neurodevelopmental diseases is largely based on work using animal models. Recent studies revealed that neural progenitors in the human brain are profoundly different from those found in rodent animal models2–5. Moreover, post-mortem studies revealed extensive migration of interneurons into the late-gestational and post-natal human prefrontal cortex that does not occur in rodents6. Here, we use cerebral organoids to show that overproduction of mid-gestational human interneurons causes Tuberous Sclerosis Complex (TSC), a severe neuro-developmental disorder associated with mutations in TSC1 and TSC2. We identify a previously uncharacterized population of caudal late interneuron progenitors, the CLIP-cells. In organoids derived from patients carrying heterozygous TSC2 mutations, dysregulation of mTOR signaling leads to CLIP-cell over-proliferation and formation of cortical tubers and subependymal tumors. Surprisingly, second-hit events resulting from copy-neutral loss-of-heterozygosity (cnLOH) are not causative for but occur during the progression of tumor lesions. Instead, EGFR signaling is required for tumor proliferation, opening up a promising approach to treat TSC lesions. Our study demonstrates that the analysis of developmental disorders in organoid models can lead to fundamental insights into human brain development and neuropsychiatric disorders.


2009 ◽  
Vol 4 (1) ◽  
pp. 85-90 ◽  
Author(s):  
Robert J. Bollo ◽  
Jonathan L. Berliner ◽  
Ingeborg Fischer ◽  
Daniel K. Miles ◽  
Elizabeth A. Thiele ◽  
...  

Subependymal giant cell tumors (SGCTs) are observed in 5–20% of patients with tuberous sclerosis complex (TSC) but account for ~ 25% of neurological morbidity. The authors report the case of a 7-year-old girl with TSC and multiple cortical tubers who presented with worsening seizures in the context of the rapid growth of a cystic, calcified, extraventricular SGCT in the right frontal lobe, initially thought to represent a cortical tuber. The tumor and surrounding tubers were excised, and clinical seizures resolved. This is the first report of an extraventricular SGCT in a child with TSC outside the neonatal period.


Epilepsia ◽  
2011 ◽  
Vol 52 (9) ◽  
pp. 1699-1704 ◽  
Author(s):  
Sanghoon Oh ◽  
Tara Stewart ◽  
Ian Miller ◽  
Sanjiv Bhatia ◽  
John Ragheb ◽  
...  

2006 ◽  
Vol 20 (1) ◽  
pp. 1-6 ◽  
Author(s):  
Leandro R. Piedimonte ◽  
Ian K. Wailes ◽  
Howard L. Weiner

Mutations in one of two genes, TSC1 and TSC2, result in a similar disease phenotype by disrupting the normal interaction of their protein products, hamartin and tuberin, which form a functional signaling complex. Disruption of these genes in the brain results in abnormal cellular differentiation, migration, and proliferation, giving rise to the characteristic brain lesions of tuberous sclerosis complex (TSC) called cortical tubers. The most devastating complications of TSC affect the central nervous system and include epilepsy, mental retardation, autism, and glial tumors. Relevant animal models, including conventional and conditional knockout mice, are valuable tools for studying the normal functions of tuberin and hamartin and the way in which disruption of their expression gives rise to the variety of clinical features that characterize TSC. In the future, these animals will be invaluable preclinical models for the development of highly specific and efficacious treatments for children affected with TSC.


2005 ◽  
Vol 5 (4) ◽  
pp. 136-138 ◽  
Author(s):  
Carl E. Stafstrom

Abnormal Cortical Cells and Astrocytomas in the Eker Rat Model of Tuberous Sclerosis Complex Takahashi DK, Dinday MT, Barbaro NM, Baraban SC Epilepsia 2004;45:1525–1530 Purpose In patients with tuberous sclerosis complex (TSC), a wide range of neurologic abnormalities develop, including mental retardation and seizures. Brains from TSC patients are characterized by the presence of cortical tubers, large dysmorphic neurons, and abnormal cytomegalic cells. Although analysis of human TSC brain samples led to the identification of these abnormal cell types, very little is known about how these cells function. In an effort to model TSC-associated CNS abnormalities (and ultimately to analyze the electrophysiologic properties of abnormal cells), we examined Eker rats carrying a Tsc2 mutation. Anatomic studies, including standard histologic stains and immunocytochemistry, were performed on young Eker rats exposed to a carcinogen in utero or on aged untreated Eker rats (18–24 months old). Methods Pregnant TSC2+/– females were injected once a day with hydroquinone, and offspring were killed at postnatal day P14 or P28. Coronal tissue sections throughout the CNS were prepared and stained for cresyl violet. In separate studies, brains of old untreated Eker rats were sectioned for anatomic analysis by using standard immunohistochemical techniques. Results Tissue sections stained with cresyl violet did not reveal any gross differences between hydroquinonetreated Eker ( Tsc2 Ek/+) rats and siblings ( Tsc2+/+). However, two classes of abnormal giant cells were observed in brain sections from untreated aged Eker rats: 1) large dysmorphic pyramid-like cells immunoreactive for NeuN, tuberin, and EAAC-1 in layers IV to VI; and 2) abnormal cytomegalic cells immunoreactive for glial fibrillary acidic protein, vimentin, and nestin in deep cortical layers or along the white matter. In addition, large subependymal astrocytomas were observed in four animals. Conclusions Our data suggest that cortical tuber formation in Eker rats is a rare event and that prenatal exposure to a nongenotoxic carcinogen such as hydroquinone is not sufficient to induce tuber formation. However, with advanced age, an increased likelihood of astrocytoma formation and the emergence of dysmorphic neurons and cytomegalic cells in the Eker rat brain might exist; each of these abnormalities mimics those seen clinically and could contribute to neurologic problems associated with TSC. Further analysis of this rodent model may be warranted. Morphology of Cerebral Lesions in the Eker Rat Model of Tuberous Sclerosis Wenzel HJ, Patel LS, Robbins CA, Emmi A, Yeung RS, Schwartzkroin PA Acta Neuropathol (Berl) 2004;108:97–108 Tuberous sclerosis (TSC) is an autosomal dominant disorder, caused by mutations of either the TSC1 or TSC2 gene. Characteristic brain pathologies (including cortical tubers and subependymal hamartomas/giant astrocytomas) are thought to cause epilepsy, as well as other neurologic dysfunction. The Eker rat, which carries a spontaneous germline mutation of the TSC2 gene ( Tsc2+/+), provides a unique animal model in which to study the relation between TSC cortical pathologies and epilepsy. In the present study, we analyzed the seizure propensity and histopathologic features of a modified Eker rat preparation, in which early postnatal irradiation was used as a “second hit” stimulus in an attempt to exacerbate cortical malformations and increase seizure propensity. Irradiated Eker rats had a tendency toward lower seizure thresholds (latencies to flurothyl-induced seizures) than seen in nonirradiated Eker rats (significant difference) or irradiated wild-type rats (nonsignificant difference). The majority of irradiated Eker rats exhibited dysplastic cytomegalic neurons and giant astrocyte-like cells, similar to cytopathologies observed in TSC lesions of patients. The most prominent features in these brains were hamartoma-like lesions involving large eosinophilic cells, similar to giant tuber cells in human TSC. In some cells from these hamartomas, immunocytochemistry revealed features of both neuronal and glial phenotypes, suggesting an undifferentiated or immature cell population. Both normal-appearing and dysmorphic neurons, as well as cells in the hamartomas, exhibited immunopositivity for tuberin, the protein product of the TSC2 gene.


Epilepsia ◽  
2009 ◽  
Vol 50 (12) ◽  
pp. 2648-2651 ◽  
Author(s):  
Catherine J. Chu-Shore ◽  
Matthew P. Frosch ◽  
Patricia E. Grant ◽  
Elizabeth A. Thiele

2001 ◽  
Vol 159 (4) ◽  
pp. 1541-1554 ◽  
Author(s):  
Robin Kyin ◽  
Yue Hua ◽  
Marianna Baybis ◽  
Bernd Scheithauer ◽  
Dennis Kolson ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document