1
|
Chen J, Sun H, Han L, Gu X, Hao X, Fu Y, Weng Z, Xiong Y, Liu B, Zhang H, He Y, Li H. Analysis of Genotypes and Phenotypes in Chinese Patients With Tuberous Sclerosis Complex Harboring Novel Variants of TSC1 and TSC2 Genes. Int J Genomics 2025; 2025:6963280. [PMID: 40376498 PMCID: PMC12081158 DOI: 10.1155/ijog/6963280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/28/2025] [Indexed: 05/18/2025] Open
Abstract
Background: This study aimed to assess the pathogenicity of newly identified tuberous sclerosis Complex 1 (TSC1) and TSC2 variants, contributing definitive evidence for the diagnosis of TSC. Methods: A total of 103 TSC patients underwent TSC genetic testing using standardized protocols, and genetic testing was extended to their respective families. Analysis of genetic testing results considered clinical phenotype and gene pathogenicity based on the 2012 revision of the International Society of TSC. Results: Among participants, 12 exhibited previously unreported variants of TSC1 or TSC2 gene absent in relevant databases. All 12 clinically diagnosed TSC patients presented typical phenotypes, such as brain lesions and skin changes. Notably, there were 2 variants of TSC1 gene and 10 variants of TSC2 gene, encompassing 8 frameshift variants, 2 nonsense variants, and 2 missense variants. Conclusions: This study broadens the spectrum of variants of TSC1 and TSC2 genes, reaffirming the clinical diagnosis of patients through genetic testing.
Collapse
Affiliation(s)
- Jian Chen
- Department of Echocardiography, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Hairui Sun
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Ling Han
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Gu
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Hao
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Yuwei Fu
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Zongjie Weng
- Department of Ultrasonography, Fujian Maternal and Child Hospital, Fuzhou, Fujian, China
| | - Yi Xiong
- Department of Ultrasonography, Shenzhen People's Hospital, Shenzhen, China
| | - Baomin Liu
- Department of Ultrasonography, The Second Affiliated Hospital, Medical College of Xi'an Jiao Tong University, Xi'an, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing An Zhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yihua He
- Maternal-Fetal Consultation Center of Congenital Heart Disease, Department of Echocardiography, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Hong Li
- Department of Ultrasound, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chinese Medicine Hospital), Shenzhen, China
| |
Collapse
|
2
|
Kim SY. Insights into Tuberous Sclerosis Complex : From Genes to Clinics. J Korean Neurosurg Soc 2025; 68:321-337. [PMID: 40090343 PMCID: PMC12062541 DOI: 10.3340/jkns.2025.0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder caused by pathogenic variants of TSC1 or TSC2 genes, leading to dysregulation of the mammalian target of rapamycin (mTOR) pathway. This dysregulation results in the formation of organ-specific tumors and neurological manifestations such as seizures, intellectual disability, and developmental delays. These characteristic clinical features are crucial for diagnosis, and genetic testing is playing an increasingly significant role. Long-term disease monitoring and appropriate interventions by multidisciplinary experts, including the use of mTOR inhibitors and promising therapeutic agents based on disease pathomechanisms, are essential for effective TSC management and improved clinical outcomes.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul National University Children’s Hospital, Seoul, Korea
| |
Collapse
|
3
|
Hartung J, Müller C, Calkhoven CF. The dual role of the TSC complex in cancer. Trends Mol Med 2025; 31:452-465. [PMID: 39488444 DOI: 10.1016/j.molmed.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The tuberous sclerosis complex (TSC1/TSC2/TBC1D7) primarily functions to inhibit the mechanistic target of rapamycin complex 1 (mTORC1), a crucial regulator of cell growth. Mutations in TSC1 or TSC2 cause tuberous sclerosis complex (TSC), a rare autosomal dominant genetic disorder marked by benign tumors in multiple organs that rarely progress to malignancy. Traditionally, TSC proteins are considered tumor suppressive due to their inhibition of mTORC1 and other mechanisms. However, more recent studies have shown that TSC proteins can also promote tumorigenesis in certain cancer types. In this review, we explore the composition and function of the TSC protein complex, the roles of its individual components in cancer biology, and potential future therapeutic targeting strategies.
Collapse
Affiliation(s)
- Josephine Hartung
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
4
|
Feliciano DM, Bordey A. TSC-mTORC1 Pathway in Postnatal V-SVZ Neurodevelopment. Biomolecules 2025; 15:573. [PMID: 40305300 PMCID: PMC12024678 DOI: 10.3390/biom15040573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
In restricted regions of the rodent brain, neurogenesis persists throughout life, hinting that perhaps similar phenomena may exist in humans. Neural stem cells (NSCs) that reside within the ventricular-subventricular zone (V-SVZ) continually produce functional cells, including neurons that integrate into the olfactory bulb circuitry. The ability to achieve this feat is based on genetically encoded transcriptional programs that are controlled by environmentally regulated post-transcriptional signaling pathways. One such pathway that molds V-SVZ neurogenesis is the mTOR pathway. This pathway integrates nutrient sufficiency with growth factor signaling to control distinct steps of neurogenesis. Alterations in mTOR pathway signaling occur in numerous neurodevelopmental disorders. Here, we provide a narrative review for the role of the mTOR pathway in this process and discuss the use of this region to study the mTOR pathway in both health and disease.
Collapse
Affiliation(s)
- David M. Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC 29634-0314, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| | - Angelique Bordey
- Departments of Neurosurgery, and Cellular & Molecular Physiology, Wu Tsai Institute, Yale University School of Medicine, New Haven, CT 06520-8082, USA;
| |
Collapse
|
5
|
Chalkley MBL, Guerin LN, Iyer T, Mallahan S, Nelson S, Sahin M, Hodges E, Ess KC, Ihrie RA. Human TSC2 mutant cells exhibit aberrations in early neurodevelopment accompanied by changes in the DNA Methylome. Hum Mol Genet 2025; 34:684-698. [PMID: 39877967 PMCID: PMC11973902 DOI: 10.1093/hmg/ddae199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a debilitating developmental disorder characterized by a variety of clinical manifestations. While benign tumors in the heart, lungs, kidney, and brain are all hallmarks of the disease, the most severe symptoms of TSC are often neurological, including seizures, autism, psychiatric disorders, and intellectual disabilities. TSC is caused by loss of function mutations in the TSC1 or TSC2 genes and consequent dysregulation of signaling via mechanistic Target of Rapamycin Complex 1 (mTORC1). While TSC neurological phenotypes are well-documented, it is not yet known how early in neural development TSC1/2-mutant cells diverge from the typical developmental trajectory. Another outstanding question is the contribution of homozygous-mutant cells to disease phenotypes and whether phenotypes are also present in the heterozygous-mutant populations that comprise the vast majority of cells in patients. Using TSC patient-derived isogenic induced pluripotent stem cells (iPSCs) with defined genetic changes, we observed aberrant early neurodevelopment in vitro, including misexpression of key proteins associated with lineage commitment and premature electrical activity. These alterations in differentiation were coincident with hundreds of differentially methylated DNA regions, including loci associated with key genes in neurodevelopment. Collectively, these data suggest that mutation or loss of TSC2 affects gene regulation and expression at earlier timepoints than previously appreciated, with implications for whether and how prenatal treatment should be pursued.
Collapse
Affiliation(s)
- Mary-Bronwen L Chalkley
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
| | - Lindsey N Guerin
- Department of Biochemistry, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
| | - Tenhir Iyer
- Department of Pediatrics, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, Tennessee, 37232, United States of America
| | - Samantha Mallahan
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
| | - Sydney Nelson
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Harvard Medical School, 25 Shattuck Street, Boston, Massachusetts, 02115, United States of America
| | - Emily Hodges
- Department of Biochemistry, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, TN, 37232, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Kevin C Ess
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
- Department of Pediatrics, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, Tennessee, 37232, United States of America
- Department of Pediatrics - Neurology, University of Colorado Anschutz Medical Campus, 13123 E. 16th Ave., Aurora, Colorado, 80045, United States of America
| | - Rebecca A Ihrie
- Department of Cell & Developmental Biology, Vanderbilt University School of Medicine, 1161 21st Ave S, Nashville, Tennessee, 37232, United States of America
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2220 Pierce Ave, Nashville, TN, 37232, USA
- Department of Pediatrics - Neurology, University of Colorado Anschutz Medical Campus, 13123 E. 16th Ave., Aurora, Colorado, 80045, United States of America
- Department of Neurological Surgery, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, Tennessee, 37232, United States of America
| |
Collapse
|
6
|
Ritter DM, Twardowski S, Franz DN. Treating subependymal giant cell astrocytoma in patients with tuberous sclerosis complex: an update of the literature. Expert Rev Neurother 2025; 25:389-396. [PMID: 40052854 DOI: 10.1080/14737175.2025.2472922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Subependymal giant cell astrocytomas (SEGAs) are one of the predominant features of the tuberous sclerosis complex (TSC). Before the use of mTOR inhibitors (mTORi; everolimus and sirolimus) in TSC, many patients had to undergo surgical operations (both preemptively and emergently). However, with mTORis gaining increased use, the role of medical and surgical therapy in SEGA is unclear. AREAS COVERED The authors have based this review on publications listed in PubMed that delve into the role of surgery and mTORi in the treatment of SEGAs. EXPERT OPINION There is no sizable head-to-head comparison of surgery and medical therapy in treating SEGA. Factors that reduce the ability to do these types of studies are the lack of uniform diagnosis of SEGA, provider preference for treatment, and variability in each treatment group (dosing of mTORis and various surgical providers). However, with the safety of mTORi, the authors recommend starting mTORi therapy for any growth in a nodule on serial scans and relying on surgery only for failed mTORi therapy.
Collapse
Affiliation(s)
- David M Ritter
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sara Twardowski
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - David N Franz
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
7
|
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant inherited disease characterized by systemic hamartomas, neuropsychiatric symptoms known as TAND (TSC-associated neuropsychiatric disorders), and vitiligo. These symptoms are attributed to the constant activation of mechanistic target of rapamycin complex 1 (mTORC1) caused by genetic mutations in the causative genes TSC1 or TSC2. The elucidation of the pathogenesis of this disease and advances in diagnostic technologies have led to dramatic changes in the diagnosis and treatment of TSC. Diagnostic criteria have been created at a global level, and mTORC1 inhibitors have emerged as therapeutic agents for this disease. Previously, the treatment strategy was limited to symptomatic treatments such as surgery. Inhibitors of mTORC1 are effective against all symptoms of TSC, but they also have systemic side effects. Therefore, the need for a cross-disciplinary, collaborative medical care system has increased, resulting in the establishment of a practice structure known as the "TSC Board." Furthermore, to reduce the side effects of systemic administration of mTORC1 inhibitors, a topical formulation of mTORC1 inhibitor was developed in Japan for the treatment of skin lesions caused by TSC. This report summarizes the pathogenesis and current status of TSC and the contribution of the Neurocutaneous Syndrome Policy Research Group to the policies of the Ministry of Health, Labor, and Welfare with respect to this rare, intractable disease.
Collapse
Affiliation(s)
- Mari Wataya-Kaneda
- Department of Neurocutaneous Medicine, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Boccatonda A, Marcellini MM, Ruggeri E, Felicani C, Brighenti A, Loiacono R, Ercolani G, Serra C. Ceus features of liver pecoma: a case report and literature review. J Ultrasound 2025; 28:261-268. [PMID: 39557792 PMCID: PMC11947360 DOI: 10.1007/s40477-024-00973-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Perivascular epithelioid cell neoplasms (PEComas) and epithelioid angiomyolipomas (EAMLs) are two different denominations for the same "mesenchymal tumor composed of histologically and immunohistochemically distinctive perivascular epithelioid cells". Hepatic PEComa/EAML is a very rare neoplasm, and only 29 case reports of hepatic PEComa and 25 of hepatic EAML have been reported in the current literature. A clear female predominance with a mean age at diagnosis of 42.5 years old can be observed by literature review. Ultrasound (US) examination was the first-line diagnostic technique in most of the cases of hepatic PEComa, but it was documented in very few cases of hepatic EAML. A great variability in the ultrasonographic B-mode, color Doppler and contrast-enhanced ultrasonography (CEUS) features of hepatic PEComa/EAML emerges. Computed tomography and magnetic resonance were the most common used techniques to confirm the nature of the hepatic lesion, even if the anatomo-pathological examination was the only technique to display a certainty diagnosis and to differentiate hepatic PEComa/EAML from benign and malignant hepatic lesions. The great majority of hepatic PEComas/EAMLs are surgically treated without any adjuvant therapy.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Diagnostic and Therapeutic Interventional Ultrasound Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti n 9, 40138, Bologna, Italy.
| | | | - Eugenio Ruggeri
- Diagnostic and Therapeutic Interventional Ultrasound Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti n 9, 40138, Bologna, Italy
| | - Cristina Felicani
- Medicina ad Indirizzo Metabolico Nutrizionale. Policlinico di Modena, AOU Modena, Modena, Italy
| | - Alice Brighenti
- Diagnostic and Therapeutic Interventional Ultrasound Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti n 9, 40138, Bologna, Italy
| | - Rossella Loiacono
- Diagnostic and Therapeutic Interventional Ultrasound Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti n 9, 40138, Bologna, Italy
| | - Giorgio Ercolani
- General and Oncologic Surgery, Morgagni-Pierantoni Hospital, AUSL Romagna, Via Forlanini 34, 47121, Forlì, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Via Zamboni 33, 40126, Bologna, Italy
| | - Carla Serra
- Diagnostic and Therapeutic Interventional Ultrasound Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, via Massarenti n 9, 40138, Bologna, Italy
| |
Collapse
|
9
|
Do J, Hashmi S, Northrup H, Farach LS, Pearson D, Richardson K. Parenting Stress in Tuberous Sclerosis Complex. Pediatr Neurol 2025; 164:25-34. [PMID: 39837130 DOI: 10.1016/j.pediatrneurol.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a multisystemic genetic disorder with clinical variability. As the needs of children with TSC may differ, parenting demands may similarly differ. Characterizing parenting stress, or emotional maladaptation from parenting duties, can enable health care providers to assist parents of children with TSC. METHODS Multiple methods were used to survey 269 parents of children (aged zero to 12 years) with TSC. The Parenting Stress Index (PSI) measured parenting stress, and children's TSC clinical, seizure, and neuropsychiatric features were obtained. A qualitative free response item asked parents to describe significant parenting stressors for a child with TSC. RESULTS Half of the participants achieved a clinically relevant PSI composite score with a high representation of the PSI subdomain, Parent-Child Dysfunction. Parents reported higher stress for children with certain skin and ocular TSC features, intractable epilepsy with or without status epilepticus, developmental delay or intellectual disability, TSC-associated neuropsychiatric disorders, and parent race and income. These variables accounted for 46% of PSI variability. Thematic analyses identified stressors consistent with survey findings and other novel constructs like uncertainty and a lack of personal or health care support. CONCLUSIONS These results could be used to improve parenting stress by counseling on the dermatologic and ophthalmologic TSC findings, anticipatory guidance on seizure symptoms, maximizing early childhood intervention or related therapies, and providing psychosocial assessment to all parents with a low threshold for referral to a mental health specialist. These considerations may ameliorate parenting stress and ultimately improve quality of life for families and patients with TSC.
Collapse
Affiliation(s)
- Jenny Do
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas.
| | - Syed Hashmi
- Department of Pediatrics, Pediatric Research Center, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children's Memorial Hermann Hospital, Houston, Texas
| | - Laura S Farach
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children's Memorial Hermann Hospital, Houston, Texas
| | - Deborah Pearson
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, Texas
| | - Kate Richardson
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston (UTHealth) and Children's Memorial Hermann Hospital, Houston, Texas
| |
Collapse
|
10
|
Buchert R, Burkhalter MD, Huridou C, Sofan L, Roser T, Cremer K, Alvi JR, Efthymiou S, Froukh T, Gulieva S, Guliyeva U, Hamdallah M, Holder-Espinasse M, Kaiyrzhanov R, Klingler D, Koko M, Matthies L, Park J, Sturm M, Velic A, Spranger S, Sultan T, Engels H, Lerche H, Houlden H, Pagnamenta AT, Borggraefe I, Weber Y, Bonnen PE, Maroofian R, Riess O, Weber JJ, Philipp M, Haack TB. Bi-allelic KICS2 mutations impair KICSTOR complex-mediated mTORC1 regulation, causing intellectual disability and epilepsy. Am J Hum Genet 2025; 112:374-393. [PMID: 39824192 PMCID: PMC11866974 DOI: 10.1016/j.ajhg.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/20/2025] Open
Abstract
Nutrient-dependent mTORC1 regulation upon amino acid deprivation is mediated by the KICSTOR complex, comprising SZT2, KPTN, ITFG2, and KICS2, recruiting GATOR1 to lysosomes. Previously, pathogenic SZT2 and KPTN variants have been associated with autosomal recessive intellectual disability and epileptic encephalopathy. We identified bi-allelic KICS2 variants in eleven affected individuals presenting with intellectual disability and epilepsy. These variants partly affected KICS2 stability, compromised KICSTOR complex formation, and demonstrated a deleterious impact on nutrient-dependent mTORC1 regulation of 4EBP1 and S6K. Phosphoproteome analyses extended these findings to show that KICS2 variants changed the mTORC1 proteome, affecting proteins that function in translation, splicing, and ciliogenesis. Depletion of Kics2 in zebrafish resulted in ciliary dysfunction consistent with a role of mTORC1 in cilia biology. These in vitro and in vivo functional studies confirmed the pathogenicity of identified KICS2 variants. Our genetic and experimental data provide evidence that variants in KICS2 are a factor involved in intellectual disability due to its dysfunction impacting mTORC1 regulation and cilia biology.
Collapse
Affiliation(s)
- Rebecca Buchert
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University of Tübingen, Tübingen, Germany
| | - Chrisovalantou Huridou
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Linda Sofan
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Timo Roser
- Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Kirsten Cremer
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Javeria Raza Alvi
- Department of Pediatric Neurology, Institute of Child Health, Children's Hospital Lahore, Lahore, Pakistan
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tawfiq Froukh
- Department of Biotechnology and Genetic Engineering, Philadelphia University, Amman, Jordan
| | | | | | - Moath Hamdallah
- Pediatrics Department, An-Najah National University Hospital, Nablus, Palestine
| | - Muriel Holder-Espinasse
- Clinical Genetics Department, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK
| | - Rauan Kaiyrzhanov
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Doreen Klingler
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Mahmoud Koko
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lars Matthies
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Joohyun Park
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Marc Sturm
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ana Velic
- Proteome Center Tübingen, University of Tübingen, Tübingen, Germany
| | | | - Tipu Sultan
- Department of Pediatric Neurology, Institute of Child Health, Children's Hospital Lahore, Lahore, Pakistan
| | - Hartmut Engels
- Institute of Human Genetics, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Alistair T Pagnamenta
- NIHR Oxford Biomedical Research Centre, Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ingo Borggraefe
- Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Yvonne Weber
- Department of Neurology and Epileptology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Penelope E Bonnen
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Center for Rare Disease, University of Tübingen, Tübingen, Germany; Genomics for Health in Africa (GHA), Africa-Europe Cluster of Research Excellence (CoRE)
| | - Jonasz J Weber
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Division of Pharmacogenomics, University of Tübingen, Tübingen, Germany
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany; Center for Rare Disease, University of Tübingen, Tübingen, Germany; Genomics for Health in Africa (GHA), Africa-Europe Cluster of Research Excellence (CoRE).
| |
Collapse
|
11
|
Wang S, Ma R, Gao C, Tian YN, Hu RG, Zhang H, Li L, Li Y. Unraveling the function of TSC1-TSC2 complex: implications for stem cell fate. Stem Cell Res Ther 2025; 16:38. [PMID: 39901197 PMCID: PMC11792405 DOI: 10.1186/s13287-025-04170-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Tuberous sclerosis complex is a genetic disorder caused by mutations in the TSC1 or TSC2 genes, affecting multiple systems. These genes produce proteins that regulate mTORC1 activity, essential for cell function and metabolism. While mTOR inhibitors have advanced treatment, maintaining long-term therapeutic success is still challenging. For over 20 years, significant progress has linked TSC1 or TSC2 gene mutations in stem cells to tuberous sclerosis complex symptoms. METHODS A comprehensive review was conducted using databases like Web of Science, Google Scholar, PubMed, and Science Direct, with search terms such as "tuberous sclerosis complex," "TSC1," "TSC2," "stem cell," "proliferation," and "differentiation." Relevant literature was thoroughly analyzed and summarized to present an updated analysis of the TSC1-TSC2 complex's role in stem cell fate determination and its implications for tuberous sclerosis complex. RESULTS The TSC1-TSC2 complex plays a crucial role in various stem cells, such as neural, germline, nephron progenitor, intestinal, hematopoietic, and mesenchymal stem/stromal cells, primarily through the mTOR signaling pathway. CONCLUSIONS This review aims shed light on the role of the TSC1-TSC2 complex in stem cell fate, its impact on health and disease, and potential new treatments for tuberous sclerosis complex.
Collapse
Affiliation(s)
- Shuang Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruishuang Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chong Gao
- School of Medicine, Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yu-Nong Tian
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong-Gui Hu
- State Key Laboratory of Brain-Machine Intelligence, Liangzhu Laboratory, School of Medicine, Zhejiang University, Zhejiang, China.
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Lan Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Yue Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
12
|
Kratz CP. Re-envisioning genetic predisposition to childhood and adolescent cancers. Nat Rev Cancer 2025; 25:109-128. [PMID: 39627375 DOI: 10.1038/s41568-024-00775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 01/31/2025]
Abstract
Although cancer is rare in children and adolescents, it remains a leading cause of death within this age range, and genetic predisposition is the main known risk factor. Since the discovery of retinoblastoma-predisposing RB1 pathogenic germline variants in 1985, several additional high-penetrance cancer predisposition genes (CPGs) have been identified. Although few clinically recognizable genetic conditions display moderate cancer phenotypes, burden testing has revealed low-to-moderate penetrance CPGs. In addition to germline pathogenic variants in CPGs, postzygotic somatic mosaic CPG pathogenic variants acquired during embryonic development are increasingly recognized as factors that predispose children and adolescents to malignancies. Genome-wide association studies of various childhood and adolescent cancer types have identified some common low-risk cancer susceptibility alleles. Although the clinical utility of polygenic risk scores is currently limited in children and adolescents, polygenic risk scores developed for adults can predict subsequent cancer risks in childhood and adolescent cancer survivors. In this Review, I describe our current knowledge of genetic predisposition to childhood and adolescent cancers. Survival rates in children and adolescents with cancer and CPGs are often poor, necessitating better integration of genomic testing into clinical care to improve cancer prevention, surveillance and therapies.
Collapse
Affiliation(s)
- Christian P Kratz
- Department of Paediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
13
|
Zhao N, Xiong Q, Li P, Chen G, Xiao H, Wu C. TSC complex decrease the expression of mTOR by regulated miR-199b-3p. Sci Rep 2025; 15:1892. [PMID: 39806027 PMCID: PMC11730325 DOI: 10.1038/s41598-025-85706-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
The TSC complex formed by TSC1 and TSC2 is the most important upstream negative regulator of mTORC1. Genetic variations in either TSC1 or TSC2 cause tuberous sclerosis complex (TSC) disease which is a rare autosomal dominant disorder resulting in impairment of multiple organ systems. In this study, besides a reported variation, c.2509_2512del (p.Asn837Valfs*11, p.N837fs) in TSC1, we found a de novo TSC2 variation c.1113delG (p.Gln371Hisfs*18, p.Q371fs), which these two mutation influence the formation of TSC complex. We found that the decrease of TSC complex with the appearance of the decreased miR-199b-3p expression. At the same time, the reduction of miR-199b-3p increased the expression of mTOR and the activation of mTORC1 and mTORC2, the additional miR-199b-3p caused the decrease the expression of mTOR and the activation of mTORC1 and mTORC2. In brief, our results may illustrate a novel mechanism of TSC caused by variations in either TSC1 or TSC2, and a new mTOR expression regulator, miR-199b-3p.
Collapse
Affiliation(s)
- Na Zhao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
- Department of Pathology, The Second Hospital of ShanXi Medical University, No.382 WuYi Road, Tai Yuan, ShanXi, Taiyuan, 030000, China
| | - Qiuhong Xiong
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Ping Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Guangxin Chen
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China
| | - Han Xiao
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| | - Changxin Wu
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
14
|
Ihnen SKZ, Alperin S, Capal JK, Cohen AL, Peters JM, Bebin EM, Northrup HA, Sahin M, Krueger DA. Accumulated seizure burden predicts neurodevelopmental outcome at 36 months of age in patients with tuberous sclerosis complex. Epilepsia 2025; 66:117-133. [PMID: 39470995 PMCID: PMC11742629 DOI: 10.1111/epi.18172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE Epilepsy and intellectual disability are common in tuberous sclerosis complex (TSC). Although early life seizures and intellectual disability are known to be correlated in TSC, the differential effects of age at seizure onset and accumulated seizure burden on development remain unclear. METHODS Daily seizure diaries, serial neurodevelopmental testing, and brain magnetic resonance imaging were analyzed for 129 TSC patients followed from 0 to 36 months. We used machine learning to identify subgroups of patients based on neurodevelopmental test scores at 36 months of age and assessed the stability of those subgroups at 12 months. We tested the ability of candidate biomarkers to predict 36-month neurodevelopmental subgroup using univariable and multivariable logistic regression. Candidate biomarkers included age at seizure onset, accumulated seizure burden, tuber volume, sex, and earlier neurodevelopmental test scores. RESULTS Patients clustered into two neurodevelopmental subgroups at 36 months of age, higher and lower scoring. Subgroup was mostly (75%) the same at 12 months. Significant univariable effects on subgroup were seen only for accumulated seizure burden (largest effect), earlier test scores, and tuber volume. Neither age at seizure onset nor sex significantly distinguished 36-month subgroups, although for girls but not boys there was a significant effect of age at seizure onset. In the multivariable model, accumulated seizure burden and earlier test scores together predicted 36-month neurodevelopmental group with 82% accuracy and an area under the curve of .86. SIGNIFICANCE These results untangle the contributions of age at seizure onset and accumulated seizure burden to neurodevelopmental outcomes in young children with TSC. Accumulated seizure burden, rather than the age at seizure onset, most accurately predicts neurodevelopmental outcome at 36 months of age. These results emphasize the need to manage seizures aggressively during the first 3 years of life for patients with TSC, not only to promote seizure control but to optimize cognitive function.
Collapse
Affiliation(s)
- S. Katie Z. Ihnen
- Division of NeurologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Samuel Alperin
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Perelman School of Medicine at University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jamie K. Capal
- Department of NeurologyUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Alexander L. Cohen
- Department of NeurologyBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Computational Radiology Laboratory, Department of RadiologyBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Laboratory for Brain Network Imaging and Modulation, Center for Brain Circuit Therapeutics, Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jurriaan M. Peters
- Department of NeurologyBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Localization Laboratory, Division of Epilepsy and Clinical NeurophysiologyBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - E. Martina Bebin
- University of Alabama at BirminghamDepartment of Neurology, Epilepsy DivisionBirminghamAlabamaUSA
| | - Hope A. Northrup
- Department of PediatricsMcGovern Medical School at University of Texas Health Science Center at Houston and Children's Memorial Hermann HospitalHoustonTexasUSA
| | - Mustafa Sahin
- Department of NeurologyBoston Children's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Darcy A. Krueger
- Division of NeurologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | | |
Collapse
|
15
|
Akbari A, Villanueva CR, Hes O, Williamson SR, Kandukuri S, Sharma S, Aditi A, Pivovarcikova K, Argani P, Mohanty SK, Asrani K, Lotan TL. Genetic validation of a TSC2 immunohistochemistry assay in TSC/mTOR-pathway altered renal tumors. Hum Pathol 2024; 154:105693. [PMID: 39571694 DOI: 10.1016/j.humpath.2024.105693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Pathogenic mutations in the genes associated with tuberous sclerosis complex (TSC)/mTOR pathway are linked to histologically diverse renal cell neoplasms, including eosinophilic solid and cystic renal cell carcinoma (ESC RCC), low grade oncocytic tumor (LOT), eosinophilic vacuolated tumor (EVT), and xanthomatous giant cell renal cell carcinoma (XGC RCC). Here, we validate a TSC2 immunohistochemistry (IHC) assay by comparison to genomic data in these neoplasms. Automated TSC2 IHC was performed on formalin-fixed paraffin embedded (FFPE) tissues from 38 genetically-confirmed TSC/mTOR-associated renal tumors (6 ESCs, 16 EVTs, 13 LOTs, 2 XGC and 1 clear cell RCC) and visually scored in a semi-dichotomous fashion compared to internal control tissue. The positive predictive value (PPV) of TSC2 protein loss for underlying pathogenic mutation in TSC2 was 92% (11/12), while the negative predictive value (NPV) of intact TSC2 by IHC for lack of underlying pathogenic mutation in TSC2 was 81% (21/26). Intact TSC2 by IHC was 95% (21/22) specific for absence of underlying pathogenic TSC2 mutation. All the cases lacking TSC2 mutation with intact TSC2 protein had an underlying mutation in TSC1, MTOR or PIK3CA. Loss of TSC2 was 77% (10/13) sensitive for underlying TSC2 truncation mutations and 33% (1/3) sensitive for underlying TSC2 missense mutations. Overall, 73% (8/11) tumors with TSC2 IHC loss and underlying pathogenic alterations in TSC2 showed heterogeneous protein loss, with rare interspersed positively staining tumor cells. These data support TSC2 IHC as a potentially useful assay for the diagnostic workup of renal tumors suspected to belong to the TSC/mTOR-associated subgroups.
Collapse
Affiliation(s)
- Amir Akbari
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Pathology, University of Southern California Keck School of Medicine, USA.
| | | | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | | | - Shivani Kandukuri
- Department of Pathology, University of Southern California Keck School of Medicine, USA
| | | | | | - Kristyna Pivovarcikova
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pedram Argani
- Department of Pathology, Johns Hopkins School of Medicine, USA
| | - Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, OR, India
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins School of Medicine, USA.
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Urology, Johns Hopkins School of Medicine, USA; Department of Oncology, Johns Hopkins School of Medicine, USA.
| |
Collapse
|
16
|
Hernández-Ramírez LC, Perez-Rivas LG, Theodoropoulou M, Korbonits M. An Update on the Genetic Drivers of Corticotroph Tumorigenesis. Exp Clin Endocrinol Diabetes 2024; 132:678-696. [PMID: 38830604 DOI: 10.1055/a-2337-2265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
The genetic landscape of corticotroph tumours of the pituitary gland has dramatically changed over the last 10 years. Somatic changes in the USP8 gene account for the most common genetic defect in corticotrophinomas, especially in females, while variants in TP53 or ATRX are associated with a subset of aggressive tumours. Germline defects have also been identified in patients with Cushing's disease: some are well-established (MEN1, CDKN1B, DICER1), while others are rare and could represent coincidences. In this review, we summarise the current knowledge on the genetic drivers of corticotroph tumorigenesis, their molecular consequences, and their impact on the clinical presentation and prognosis.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, LMU Klinikum, LMU München, Munich 80336, Germany
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
17
|
Dufner-Almeida LG, Cardozo LFM, Schwind MR, Carvalho D, Almeida JPG, Cappellano AM, Alegria TGP, Nanhoe S, Nellist M, Passos-Bueno MR, Chiavegatto S, Silva NS, Rosemberg S, Pereira APA, Antoniuk SA, Haddad LA. Molecular and Functional Assessment of TSC1 and TSC2 in Individuals with Tuberous Sclerosis Complex. Genes (Basel) 2024; 15:1432. [PMID: 39596632 PMCID: PMC11593644 DOI: 10.3390/genes15111432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 11/29/2024] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant neurodevelopmental disorder and multisystem disease caused by pathogenic DNA alterations in the TSC1 and TSC2 tumor suppressor genes. A molecular genetic diagnosis of TSC confirms the clinical diagnosis, facilitating the implementation of appropriate care and surveillance. TSC1 and TSC2 encode the core components of the TSC1/2 complex (TSC1/2), a negative regulator of the mechanistic target of rapamycin (MTOR) complex 1 (TORC1). Functional analysis of the effects of TSC1 and TSC2 variants on TORC1 activity can help establish variant pathogenicity. We searched for pathogenic alterations to TSC1 and TSC2 in DNA isolated from 116 individuals with a definite clinical diagnosis of TSC. Missense variants and in-frame deletions were functionally assessed. Pathogenic DNA alterations were identified in 106 cases (91%); 18 (17%) in TSC1 and 88 (83%) in TSC2. Of these, 35 were novel. Disruption of TSC1/2 activity was demonstrated for seven TSC2 variants. Molecular diagnostics confirms the clinical diagnosis of TSC in a large proportion of cases. Functional assessment can help establish variant pathogenicity and is a useful adjunct to DNA analysis.
Collapse
Affiliation(s)
- Luiz Gustavo Dufner-Almeida
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Department of Clinical Genetics, Erasmus Medical Center, 3015 Rotterdam, The Netherlands
| | - Laís F. M. Cardozo
- Pediatric Neurology Center, Department of Pediatrics, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba 80060-900, Brazil (S.A.A.)
| | - Mariana R. Schwind
- Pediatric Neurology Center, Department of Pediatrics, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba 80060-900, Brazil (S.A.A.)
| | - Danielly Carvalho
- Pediatric Neurology Center, Department of Pediatrics, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba 80060-900, Brazil (S.A.A.)
| | - Juliana Paula G. Almeida
- Division of Neurology, Department of Pediatrics, Santa Casa de Misericórdia, São Paulo 01221-010, Brazil
| | - Andrea Maria Cappellano
- Grupo de Apoio ao Adolescente e à Criança com Câncer, Instituto de Oncologia Pediátrica, Universidade Federal de São Paulo, São Paulo 04039-001, Brazil
| | - Thiago G. P. Alegria
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Santoesha Nanhoe
- Department of Clinical Genetics, Erasmus Medical Center, 3015 Rotterdam, The Netherlands
| | - Mark Nellist
- Department of Clinical Genetics, Erasmus Medical Center, 3015 Rotterdam, The Netherlands
| | - Maria Rita Passos-Bueno
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| | - Silvana Chiavegatto
- Department of Pharmacology, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, Brazil
- Department of Psychiatry, Instituto de Psiquiatria, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-903, Brazil
| | - Nasjla S. Silva
- Grupo de Apoio ao Adolescente e à Criança com Câncer, Instituto de Oncologia Pediátrica, Universidade Federal de São Paulo, São Paulo 04039-001, Brazil
| | - Sérgio Rosemberg
- Division of Neurology, Department of Pediatrics, Santa Casa de Misericórdia, São Paulo 01221-010, Brazil
| | - Ana Paula A. Pereira
- Department of Psychology, Universidade Federal do Paraná, Curitiba 80060-000, Brazil
| | - Sérgio Antônio Antoniuk
- Pediatric Neurology Center, Department of Pediatrics, Hospital de Clínicas, Universidade Federal do Paraná, Curitiba 80060-900, Brazil (S.A.A.)
| | - Luciana A. Haddad
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
18
|
Alesi N, Asrani K, Lotan TL, Henske EP. The Spectrum of Renal "TFEopathies": Flipping the mTOR Switch in Renal Tumorigenesis. Physiology (Bethesda) 2024; 39:0. [PMID: 39012319 DOI: 10.1152/physiol.00026.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
The mammalian target of Rapamycin complex 1 (mTORC1) is a serine/threonine kinase that couples nutrient and growth factor signaling to the cellular control of metabolism and plays a fundamental role in aberrant proliferation in cancer. mTORC1 has previously been considered an "on/off" switch, capable of phosphorylating the entire pool of its substrates when activated. However, recent studies have indicated that mTORC1 may be active toward its canonical substrates, eukaryotic translation initiation factor 4E-binding protein 1 (4EBP1) and S6 kinase (S6K), involved in mRNA translation and protein synthesis, and inactive toward TFEB and TFE3, transcription factors involved in the regulation of lysosome biogenesis, in several pathological contexts. Among these conditions are Birt-Hogg-Dubé syndrome (BHD) and, recently, tuberous sclerosis complex (TSC). Furthermore, increased TFEB and TFE3 nuclear localization in these syndromes, and in translocation renal cell carcinomas (tRCC), drives mTORC1 activity toward the canonical substrates, through the transcriptional activation of the Rag GTPases, thereby positioning TFEB and TFE3 upstream of mTORC1 activity toward 4EBP1 and S6K. The expanding importance of TFEB and TFE3 in the pathogenesis of these renal diseases warrants a novel clinical grouping that we term "TFEopathies." Currently, there are no therapeutic options directly targeting TFEB and TFE3, which represents a challenging and critically required avenue for cancer research.
Collapse
Affiliation(s)
- Nicola Alesi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Elizabeth P Henske
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
19
|
Jones VM, Thompson LDR, Pettus JR, Green DC, Lefferts JA, Shah PS, Tsongalis GJ, Sajed DP, Guilmette JM, Lewis JS, Fisch AS, Tafe LJ, Kerr DA. Angiomyolipomatous Lesions of the Nasal Cavity (Sinonasal Angioleiomyoma with Adipocytic Differentiation): A Multi-Institutional Immunohistochemical and Molecular Study. Head Neck Pathol 2024; 18:93. [PMID: 39400771 PMCID: PMC11473525 DOI: 10.1007/s12105-024-01700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Mesenchymal neoplasms composed of vascular, smooth muscle, and adipocytic components are uncommon in the nasal cavity. While angioleiomyoma (AL) is a smooth muscle tumor in the Head & Neck WHO classification, it is considered of pericytic origin in the Skin as well as Soft Tissue and Bone classifications. For nasal AL with an adipocytic component, the terms AL with adipocytic differentiation and angiomyolipoma (AML) have been applied, among others. AML is a type of perivascular epithelioid cell tumor (PEComa), most often arising in the kidney, sometimes associated with the tuberous sclerosis complex (TSC). It is uncertain whether nasal cavity AML and AL are best considered hamartomas or neoplasms, as their genetics are largely unexplored. METHODS We performed a multi-institutional retrospective study of nasal cavity mesenchymal lesions. Patient demographics, clinical histories, and histologic and immunohistochemical findings were collected. DNA and RNA were extracted from formalin-fixed, paraffin-embedded tissue and analyzed by SNP-based chromosomal microarray, targeted RNA fusion sequencing, and whole-exome sequencing. RESULTS Fifteen lesions (3-42 mm) were identified, predominantly in male (87%) patients with a median age of 60. Patients typically presented with obstructive symptoms, and none had a history of TSC. One AL was a recurrence from six years prior; 11 cases showed no recurrence (median 4.7 years, range: 0.88-12.4). Morphologically, 11 AML contained 30-80% smooth muscle, 10-25% vasculature, and 2-60% adipose tissue, while four AL contained 70-80% smooth muscle and 20-30% vasculature. Other histologic observations included ulceration, thrombosis, inflammation, myxoid change, senescent nuclei, and extramedullary hematopoiesis; no well-developed epithelioid cell morphology was identified. Immunohistochemically, all cases were positive for smooth muscle markers (actin, desmin, and/or caldesmon) and negative for melanocytic markers. Molecular analysis revealed loss of 3p and 11q in a single AML. No other known pathogenic copy number or molecular alterations were seen, including in TSC1/2, TFE3, or NOTCH2. CONCLUSION Nasal cavity AML lacks morphologic, immunophenotypic, and genetic features of PEComa family AML. The significant histologic overlap between nasal AML and AL without distinguishing molecular features in either entity suggests "sinonasal angioleiomyoma with adipocytic differentiation" may be the most appropriate terminology for hybrid vascular and smooth muscle lesions containing adipocytic components.
Collapse
Affiliation(s)
- Victoria M Jones
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | | | - Jason R Pettus
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Donald C Green
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Joel A Lefferts
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Parth S Shah
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Gregory J Tsongalis
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Dipti P Sajed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Julie M Guilmette
- Department of Pathology, Hôpital Charles-Lemoyne, Faculty of Medicine and Health Sciences, University of Sherbrooke, Greenfield Park, QC, Canada
| | - James S Lewis
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Adam S Fisch
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura J Tafe
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Darcy A Kerr
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA.
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
20
|
Chung CWT, Bournazos AM, Chan LCD, Sarkozy V, Lawson J, Kennedy SE, Cooper ST, Kirk EP, Mowat D. Deep Sequencing and Phenotyping in an Australian Tuberous Sclerosis Complex "No Mutations Identified" Cohort. Mol Genet Genomic Med 2024; 12:e70017. [PMID: 39352229 PMCID: PMC11443604 DOI: 10.1002/mgg3.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/08/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
Tuberous sclerosis complex (TSC) is a variable multisystem disorder. The "no mutations identified" (NMI) group are reportedly phenotypically milder than those with an identified molecular cause, and often have mosaic or intronic variants not detected by standard sequencing methods. METHODS We describe the phenotypes in an Australian TSC NMI group (n = 18) and a molecular testing strategy implementable in a diagnostic laboratory. Massively parallel sequencing (MPS) of the whole genomic regions of TSC1 and TSC2 was performed using DNA extracted from multiple tissue samples per participant. RESULTS Our study showed that the phenotype in TSC NMI individuals can be similar to those with heterozygous, particularly TSC1, variants. Although neurodevelopmental outcomes can be less severe, the number of organ systems involved was similar to the non-mosaic groups. A diagnostic yield of 72% (13/18) was achieved, with the majority (10/13) being mosaic variants and the remainder heterozygous variants missed on previous testing. CONCLUSION Testing DNA from multiple tissue samples allowed for validation of otherwise discarded low-level mosaic variants and detection of mosaic variants by MPS without excessive cost or the need for specialised techniques. Implementing this approach in a diagnostic setting is viable and allows optimal clinical care of patients with NMI TSC.
Collapse
Affiliation(s)
- Clara W T Chung
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - Adam M Bournazos
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- The Children's Medical Research Institute, Westmead, New South Wales, Australia
- Specialty of Child and Adolescent Health, Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Lok Chi Denise Chan
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - Vanessa Sarkozy
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - John Lawson
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Sean E Kennedy
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| | - Sandra T Cooper
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- The Children's Medical Research Institute, Westmead, New South Wales, Australia
- Specialty of Child and Adolescent Health, Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia
| | - Edwin P Kirk
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
- NSW Health Pathology Randwick Genomics Laboratory, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - David Mowat
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Tuberous Sclerosis Management Clinic, Sydney Children's Hospital, Randwick, New South Wales, Australia
- Discipline of Paediatrics and Child Health, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, New South Wales, Australia
| |
Collapse
|
21
|
Jones VM, Thompson LDR, Pettus JR, Green DC, Lefferts JA, Shah PS, Tsongalis GJ, Sajed DP, Guilmette JM, Lewis JS, Fisch AS, Tafe LJ, Kerr DA. Angiomyolipomatous Lesions of the Nasal Cavity (Sinonasal Angioleiomyoma with Adipocytic Differentiation): A Multi-Institutional Immunohistochemical and Molecular Study. RESEARCH SQUARE 2024:rs.3.rs-4843357. [PMID: 39281855 PMCID: PMC11398573 DOI: 10.21203/rs.3.rs-4843357/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Purpose Mesenchymal neoplasms composed of vascular, smooth muscle, and adipocytic components are uncommon in the nasal cavity. While angioleiomyoma (AL) is a smooth muscle tumor in the Head & Neck WHO classification, it is considered of pericytic origin in the Skin as well as Soft Tissue and Bone classifications. For nasal AL with an adipocytic component, the terms AL with adipocytic differentiation and angiomyolipoma (AML) have been applied, among others. AML is a type of perivascular epithelioid cell tumor (PEComa), most often arising in the kidney, sometimes associated with the tuberous sclerosis complex (TSC). It is uncertain whether nasal cavity AML and AL are best considered hamartomas or neoplasms, as their genetics are largely unexplored. Methods We performed a multi-institutional retrospective study of nasal cavity mesenchymal lesions. Patient demographics, clinical histories, and histologic and immunohistochemical findings were collected. DNA and RNA were extracted from formalin-fixed, paraffin-embedded tissue and analyzed by SNP-based chromosomal microarray, targeted RNA fusion sequencing, and whole-exome sequencing. Results Fifteen lesions (3 to 42 mm) were identified predominantly in male (87%) patients with a median age of 60. Patients typically presented with obstructive symptoms, and none had a history of TSC. One AL was a recurrence from six years prior; 11 cases showed no recurrence (median 4.7 years, range: 0.88-12.4). Morphologically, 11 AMLs contained 30-80% smooth muscle, 10-25% vasculature, and 2-60% adipose tissue, while four ALs contained 70-80% smooth muscle and 20-30% vasculature. Other histologic observations included surface ulceration, vascular thrombosis, chronic inflammation, and myxoid change; no well-developed epithelioid cell morphology was identified. Immunohistochemically, all cases were positive for smooth muscle markers (actin and/or desmin) and negative for melanocytic markers. Molecular analysis revealed loss of 3p and 11q in a single AML. No other known pathogenic copy number or molecular alterations were seen, including in TSC1/2, TFE3, or NOTCH2. Conclusion Nasal cavity AML lacks morphologic, immunophenotypic, and genetic features of PEComa family AMLs. The significant histologic overlap between nasal AML and AL without distinguishing molecular features in either entity suggests "sinonasal angioleiomyoma with adipocytic differentiation" may be the most appropriate terminology for hybrid vascular and smooth muscle lesions containing adipocytic components.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Adam S Fisch
- Massachusetts General Hospital, Harvard Medical School
| | | | | |
Collapse
|
22
|
Devkota S, Bhatta OP, Kalikote A, Gyawali P, Lamichhane S. Atypical case of tuberous sclerosis with isolated neurologic findings: A case report. Clin Case Rep 2024; 12:e9379. [PMID: 39229295 PMCID: PMC11369680 DOI: 10.1002/ccr3.9379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/28/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
Tuberous sclerosis (TSC) is an autosomal dominant neurocutaneous disorder. This case highlights rare isolated neurologic finding in a TSC patient emphasizing the need for heightened suspicion even in the absence of any cutaneous findings and family history.
Collapse
Affiliation(s)
- Shritik Devkota
- Department of Radiodiagnosis and ImagingAnil Baghi HospitalPunjabIndia
- Department of Radiodiagnosis and ImagingPostgraduate Institute of Medical Education and ResearchChandigarhIndia
| | | | | | - Prakash Gyawali
- Department of Emergency MedicineSukraraj Tropical and Infectious HospitalKathmandu
| | - Samiksha Lamichhane
- Department of Radiodiagnosis and ImagingBP Koirala Institute of Health SciencesDharanNepal
| |
Collapse
|
23
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
24
|
Chalkley MBL, Guerin LN, Iyer T, Mallahan S, Nelson S, Sahin M, Hodges E, Ess KC, Ihrie RA. Human TSC2 Mutant Cells Exhibit Aberrations in Early Neurodevelopment Accompanied by Changes in the DNA Methylome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597443. [PMID: 38895266 PMCID: PMC11185654 DOI: 10.1101/2024.06.04.597443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tuberous Sclerosis Complex (TSC) is a debilitating developmental disorder characterized by a variety of clinical manifestations. While benign tumors in the heart, lungs, kidney, and brain are all hallmarks of the disease, the most severe symptoms of TSC are often neurological, including seizures, autism, psychiatric disorders, and intellectual disabilities. TSC is caused by loss of function mutations in the TSC1 or TSC2 genes and consequent dysregulation of signaling via mechanistic Target of Rapamycin Complex 1 (mTORC1). While TSC neurological phenotypes are well-documented, it is not yet known how early in neural development TSC1/2-mutant cells diverge from the typical developmental trajectory. Another outstanding question is the contribution of homozygous-mutant cells to disease phenotypes and whether such phenotypes are also seen in the heterozygous-mutant populations that comprise the vast majority of cells in patients. Using TSC patient-derived isogenic induced pluripotent stem cells (iPSCs) with defined genetic changes, we observed aberrant early neurodevelopment in vitro, including misexpression of key proteins associated with lineage commitment and premature electrical activity. These alterations in differentiation were coincident with hundreds of differentially methylated DNA regions, including loci associated with key genes in neurodevelopment. Collectively, these data suggest that mutation or loss of TSC2 affects gene regulation and expression at earlier timepoints than previously appreciated, with implications for whether and how prenatal treatment should be pursued.
Collapse
Affiliation(s)
- Mary-Bronwen L. Chalkley
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Lindsey N. Guerin
- Department of Biochemistry, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Tenhir Iyer
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Samantha Mallahan
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Sydney Nelson
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emily Hodges
- Department of Biochemistry, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Kevin C. Ess
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Denver, Colorado, United States of America
| | - Rebecca A. Ihrie
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
25
|
Lazea C, Țaranu I, Bolboacă SD. Exploring Cardiovascular Involvement in Tuberous Sclerosis: Insights for Pediatric Clinicians. CHILDREN (BASEL, SWITZERLAND) 2024; 11:674. [PMID: 38929253 PMCID: PMC11201926 DOI: 10.3390/children11060674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
Tuberous sclerosis is a rare genetic disorder involving mainly the nervous and cardiovascular systems. The early recognition of the cardiovascular manifestations by the pediatrician allows an appropriate management and therefore enhances the quality of life of the affected children. Cardiac rhabdomyomas and the associated arrhythmias are the first cardiac features and they might represent a diagnosis challenge given their wide spectrum of clinical manifestations. We aimed to provide the paediatric practitioners with current knowledge regarding the cardiovascular complications in children with tuberous sclerosis. We overviewed the antenatal and postnatal evolution of cardiovascular manifestations, the systematic screening and long-term follow-up strategy of cardiac rhabdomyomas and arrhythmias in children with tuberous sclerosis.
Collapse
Affiliation(s)
- Cecilia Lazea
- Pediatric Clinic 1, Emergency Pediatric Hospital, Calea Moților, No. 68, 400370 Cluj-Napoca, Romania;
- Department Mother and Child, Iuliu Hațieganu University of Medicine and Pharmacy, Calea Moților, No. 68, 400370 Cluj-Napoca, Romania
| | - Ioana Țaranu
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Louis Pasteur Str., No. 6, 400349 Cluj-Napoca, Romania;
| | - Sorana D. Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, Louis Pasteur Str., No. 6, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
26
|
Xu W, Ma R, Li Y, Hu Z, Zhang G, Hu J, Hei Y, Yang X. Perivascular epithelioid cell tumor of the lacrimal gland. Orbit 2024; 43:362-365. [PMID: 36734431 DOI: 10.1080/01676830.2022.2141804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/25/2022] [Indexed: 02/04/2023]
Abstract
A 33-year-old lady was referred to our clinic with a history of left upper eyelid swelling and difficulty to open her left eye for more than 4 years. Her globe was in infero-nasal displacement, and ocular movement was not full, with restriction to every direction. No clinical sign including eye redness, pain or visual loss was found on examination. Exophthalmometry confirmed 2 mm of left-sided proptosis. Computed tomography (CT) imaging of the orbit obtained in axial and coronal planes depicted an irregular and heterogeneous soft-tissue density mass in the left lacrimal gland, measuring 25 × 17 mm. Magnetic resonance imaging (MRI) revealed the mass of slightly longer T1 and slightly longer/shorter T2 signal, and Gd-enhanced MRI showed a significant enhancement. The tumor was first presumably diagnosed of pleomorphic adenomas. A surgery was applied to remove the tumor completely. Histopathological evaluation led to the diagnosis of PEComa.
Collapse
Affiliation(s)
- Wenqin Xu
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Rui Ma
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Yueyue Li
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Zhicha Hu
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | | | - Jian Hu
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Yan Hei
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Xinji Yang
- Department of Orbital Disease, Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
27
|
Nóbrega IDS, Teles e Silva AL, Yokota-Moreno BY, Sertié AL. The Importance of Large-Scale Genomic Studies to Unravel Genetic Risk Factors for Autism. Int J Mol Sci 2024; 25:5816. [PMID: 38892002 PMCID: PMC11172008 DOI: 10.3390/ijms25115816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Autism spectrum disorder (ASD) is a common and highly heritable neurodevelopmental disorder. During the last 15 years, advances in genomic technologies and the availability of increasingly large patient cohorts have greatly expanded our knowledge of the genetic architecture of ASD and its neurobiological mechanisms. Over two hundred risk regions and genes carrying rare de novo and transmitted high-impact variants have been identified. Additionally, common variants with small individual effect size are also important, and a number of loci are now being uncovered. At the same time, these new insights have highlighted ongoing challenges. In this perspective article, we summarize developments in ASD genetic research and address the enormous impact of large-scale genomic initiatives on ASD gene discovery.
Collapse
Affiliation(s)
| | | | | | - Andréa Laurato Sertié
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, Rua Comendador Elias Jafet, 755. Morumbi, São Paulo 05653-000, Brazil; (I.d.S.N.); (A.L.T.e.S.); (B.Y.Y.-M.)
| |
Collapse
|
28
|
杨 晓. [Sperm Mosaic Variants and Their Influence on the Offspring]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:535-541. [PMID: 38948294 PMCID: PMC11211766 DOI: 10.12182/20240560507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Indexed: 07/02/2024]
Abstract
Genomic mosaicism arising from mosaic variants is a phenomenon that describes the presence of a cell or cell populations with different genome compositions from the germline cells of an individual. It comprises all types of genetic variants. A large proportion of childhood genetic disorders are defined as being de novo, meaning that the disease-causing mutations are only detected in the proband, not in any of the parents. Population studies show that 80% of the de novo mutations arise from the paternal haplotype, that is, from paternal sperm mosaicism. This review provides a summary of the types and detection strategies of sperm mosaicism. In addition, it provides discussions on how recent studies demonstrated that genomic mosaic mutations in parents, especially those in the paternal sperms, could be inherited by the offspring and cause childhood disorders. According to the previous findings of the author's research team, sperm mosaicism derived from early embryogenesis and primordial germ cell stages can explain 5% to 20% of the de novo mutations related to clinical phenotypes and can serve as an important predictor of both rare and complex disorders. Sperm mosaicism shows great potential for clinical genetic diagnosis and consultations. Based on the published literature, the author suggests that, large-scale screening for de novo sperm mosaic mutations and population-based genetic screening should be conducted in future studies, which will greatly enhance the risk assessment in the offspring and effectively improve the genetic health at the population level. Implementation of direct sperm detection for de novo mutations will significantly increase the efficiency of the stratification of patient cohorts and improve recurrence risk assessment for future births. Future research in the field should be focused on the impact of environmental and lifestyle factors on the health of the offspring through sperms and their modeling of mutation signatures. In addition, targeted in vitro modeling of sperm mutations will also be a promising direction.
Collapse
Affiliation(s)
- 晓旭 杨
- 犹他大学 (盐湖城 UT 84112)University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
29
|
Ramírez-Rentería C, Hernández-Ramírez LC. Genetic diagnosis in acromegaly and gigantism: From research to clinical practice. Best Pract Res Clin Endocrinol Metab 2024; 38:101892. [PMID: 38521632 DOI: 10.1016/j.beem.2024.101892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
It is usually considered that only 5% of all pituitary neuroendocrine tumours are due to inheritable causes. Since this estimate was reported, however, multiple genetic defects driving syndromic and nonsyndromic somatotrophinomas have been unveiled. This heterogeneous genetic background results in overlapping phenotypes of GH excess. Genetic tests should be part of the approach to patients with acromegaly and gigantism because they can refine the clinical diagnoses, opening the possibility to tailor the clinical conduct to each patient. Even more, genetic testing and clinical screening of at-risk individuals have a positive impact on disease outcomes, by allowing for the timely detection and treatment of somatotrophinomas at early stages. Future research should focus on determining the actual frequency of novel genetic drivers of somatotrophinomas in the general population, developing up-to-date disease-specific multi-gene panels for clinical use, and finding strategies to improve access to modern genetic testing worldwide.
Collapse
Affiliation(s)
- Claudia Ramírez-Rentería
- Unidad de Investigación Médica en Enfermedades Endocrinas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Laura C Hernández-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
30
|
Karalis V, Wood D, Teaney NA, Sahin M. The role of TSC1 and TSC2 proteins in neuronal axons. Mol Psychiatry 2024; 29:1165-1178. [PMID: 38212374 DOI: 10.1038/s41380-023-02402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
Tuberous Sclerosis Complex 1 and 2 proteins, TSC1 and TSC2 respectively, participate in a multiprotein complex with a crucial role for the proper development and function of the nervous system. This complex primarily acts as an inhibitor of the mechanistic target of rapamycin (mTOR) kinase, and mutations in either TSC1 or TSC2 cause a neurodevelopmental disorder called Tuberous Sclerosis Complex (TSC). Neurological manifestations of TSC include brain lesions, epilepsy, autism, and intellectual disability. On the cellular level, the TSC/mTOR signaling axis regulates multiple anabolic and catabolic processes, but it is not clear how these processes contribute to specific neurologic phenotypes. Hence, several studies have aimed to elucidate the role of this signaling pathway in neurons. Of particular interest are axons, as axonal defects are associated with severe neurocognitive impairments. Here, we review findings regarding the role of the TSC1/2 protein complex in axons. Specifically, we will discuss how TSC1/2 canonical and non-canonical functions contribute to the formation and integrity of axonal structure and function.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Delaney Wood
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Nicole A Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Kioutchoukova I, Foster D, Thakkar R, Ciesla C, Cabassa JS, Strouse J, Kurz H, Lucke-Wold B. Neurocutaneous Diseases: Diagnosis, Management, and Treatment. J Clin Med 2024; 13:1648. [PMID: 38541874 PMCID: PMC10971194 DOI: 10.3390/jcm13061648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 01/05/2025] Open
Abstract
Neurocutaneous disorders, also known as phakomatoses, are congenital and acquired syndromes resulting in simultaneous neurologic and cutaneous involvement. In several of these conditions, the genetic phenomenon is understood, providing a pivotal role in the development of therapeutic options. This review encompasses the discussion of the genetic and clinical involvement of neurocutaneous disorders, and examines clinical management and treatment options. With the current advances in genetics, the role of precision medicine and targeted therapy play a substantial role in addressing the management of these conditions. The interconnectedness between therapeutic options highlights the importance of precision medicine in treating each disorder's unique molecular pathway. This review provides an extensive synthesis of ongoing and current therapeutics in the management of such clinically unique and challenging conditions.
Collapse
Affiliation(s)
- Ivelina Kioutchoukova
- College of Medicine, University of Florida, Gainsville, FL 32610, USA; (I.K.); (R.T.); (H.K.)
| | - Devon Foster
- Herbert Wertheim College of Medicine, Miami, FL 33199, USA; (D.F.); (C.C.); (J.S.C.); (J.S.)
| | - Rajvi Thakkar
- College of Medicine, University of Florida, Gainsville, FL 32610, USA; (I.K.); (R.T.); (H.K.)
| | - Christopher Ciesla
- Herbert Wertheim College of Medicine, Miami, FL 33199, USA; (D.F.); (C.C.); (J.S.C.); (J.S.)
| | - Jake Salvatore Cabassa
- Herbert Wertheim College of Medicine, Miami, FL 33199, USA; (D.F.); (C.C.); (J.S.C.); (J.S.)
| | - Jacob Strouse
- Herbert Wertheim College of Medicine, Miami, FL 33199, USA; (D.F.); (C.C.); (J.S.C.); (J.S.)
| | - Hayley Kurz
- College of Medicine, University of Florida, Gainsville, FL 32610, USA; (I.K.); (R.T.); (H.K.)
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
32
|
Man A, Di Scipio M, Grewal S, Suk Y, Trinari E, Ejaz R, Whitney R. The Genetics of Tuberous Sclerosis Complex and Related mTORopathies: Current Understanding and Future Directions. Genes (Basel) 2024; 15:332. [PMID: 38540392 PMCID: PMC10970281 DOI: 10.3390/genes15030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/02/2024] [Accepted: 03/02/2024] [Indexed: 06/14/2024] Open
Abstract
The mechanistic target of rapamycin (mTOR) pathway serves as a master regulator of cell growth, proliferation, and survival. Upregulation of the mTOR pathway has been shown to cause malformations of cortical development, medically refractory epilepsies, and neurodevelopmental disorders, collectively described as mTORopathies. Tuberous sclerosis complex (TSC) serves as the prototypical mTORopathy. Characterized by the development of benign tumors in multiple organs, pathogenic variants in TSC1 or TSC2 disrupt the TSC protein complex, a negative regulator of the mTOR pathway. Variants in critical domains of the TSC complex, especially in the catalytic TSC2 subunit, correlate with increased disease severity. Variants in less crucial exons and non-coding regions, as well as those undetectable with conventional testing, may lead to milder phenotypes. Despite the assumption of complete penetrance, expressivity varies within families, and certain variants delay disease onset with milder neurological effects. Understanding these genotype-phenotype correlations is crucial for effective clinical management. Notably, 15% of patients have no mutation identified by conventional genetic testing, with the majority of cases postulated to be caused by somatic TSC1/TSC2 variants which present complex diagnostic challenges. Advancements in genetic testing, prenatal screening, and precision medicine hold promise for changing the diagnostic and treatment paradigm for TSC and related mTORopathies. Herein, we explore the genetic and molecular mechanisms of TSC and other mTORopathies, emphasizing contemporary genetic methods in understanding and diagnosing the condition.
Collapse
Affiliation(s)
- Alice Man
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Matteo Di Scipio
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Shan Grewal
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Yujin Suk
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Elisabetta Trinari
- Division of Developmental Pediatrics, Department of Pediatrics, McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Resham Ejaz
- Division of Genetics, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Pediatrics, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
33
|
Auen T, Linde E. An Autopsy Case of Abdominal Aortic Aneurysm in a Pediatric Decedent With Tuberous Sclerosis Complex. Am J Forensic Med Pathol 2024; 45:72-76. [PMID: 37486961 DOI: 10.1097/paf.0000000000000860] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
ABSTRACT Vascular involvement in tuberous sclerosis complex (TSC) is uncommon and even more so in pediatric patients. When asymptomatic, these vascular abnormalities carry increased risk of rupture with increased morbidity and mortality. Here, we describe a case of a ruptured unrecognized abdominal aortic aneurysm in an 11-month-old patient with a history of TSC. The abdominal aortic aneurysm was discovered at autopsy and highlights the rarity of abdominal aortic aneurysm in pediatric patients diagnosed with TSC and the importance of screening for associated aneurysmal disease in the pediatric population with TSC. Furthermore, the extensive retroperitoneal hemorrhage seen in this case also highlights a rare but potential mimic of abuse in the pediatric population.
Collapse
Affiliation(s)
- Thomas Auen
- From the Department of Pathology and Microbiology, Nebraska Medical Center, University of Nebraska Medical Center
| | - Erin Linde
- Physicians Laboratory Services, Omaha, NE
| |
Collapse
|
34
|
Yeşil Ş, Kurucu B, Hamamcı MB, Yılmaz Ş, Şahin G. Treatment of tuberous sclerosis complex manifestations in children with mTOR inhibitors. Childs Nerv Syst 2024; 40:831-837. [PMID: 37947859 DOI: 10.1007/s00381-023-06218-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
PURPOSE Tuberous sclerosis complex (TSC) is a rare autosomal dominant genetic disorder that affects multiple organ systems. Mutations in the TSC1 and TSC2 genes result in the constitutive hyperactivation of the mammalian target of rapamycin (mTOR) pathway, contributing to the growth of benign tumors or hamartomas in various organs. Due to the implication of mTOR pathway dysregulation in the disease pathology, increasing evidence supports the use of mTOR inhibitors for treating multiple manifestations of TSC. METHODS In this study, we conducted a retrospective analysis of clinical findings and treatment data from 38 patients diagnosed with tuberous sclerosis who were followed up in the Pediatric Oncology Clinic between 2010 and 2020. We collected information on patients' ages, genders, affected sites, familial history, imaging findings, presence of tumors, and treatments. RESULTS Among the patients, nine individuals with TSC manifestations were treated with mTOR inhibitors. Specifically, everolimus was successfully administered to five patients with inborn cardiac rhabdomyoma causing hemodynamic impairment. In addition, two patients with refractory seizures received everolimus in combination with anti-epileptic drugs. A patient with renal angiomyolipomas larger than 3 cm was treated with everolimus, while a patient with extensive facial angiofibroma received topical sirolimus. All patients tolerated the mTOR inhibitors well, and the side effects were deemed acceptable. CONCLUSION The utilization of mTOR inhibition in TSC is expected to become more prevalent in clinical practice, as current research is anticipated to provide a better understanding of the therapeutic roles of these treatments in TSC.
Collapse
Affiliation(s)
- Şule Yeşil
- Department of Pediatric Hematology and Oncology, Ankara Etlik Integrated Health Campus, Ankara, Turkey.
| | - Burçak Kurucu
- Department of Pediatric Hematology and Oncology, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Melda Berber Hamamcı
- Department of Pediatric Hematology and Oncology, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Şükriye Yılmaz
- Department of Pediatric Radiology, Ankara Etlik Integrated Health Campus, Ankara, Turkey
| | - Gürses Şahin
- Dr. Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Department of Pediatric Hematology and Oncology, Ankara, Turkey
| |
Collapse
|
35
|
Long HY, Qian ZP, Lan Q, Xu YJ, Da JJ, Yu FX, Zha Y. Human pluripotent stem cell-derived kidney organoids: Current progress and challenges. World J Stem Cells 2024; 16:114-125. [PMID: 38455108 PMCID: PMC10915962 DOI: 10.4252/wjsc.v16.i2.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
Human pluripotent stem cell (hPSC)-derived kidney organoids share similarities with the fetal kidney. However, the current hPSC-derived kidney organoids have some limitations, including the inability to perform nephrogenesis and lack of a corticomedullary definition, uniform vascular system, and coordinated exit pathway for urinary filtrate. Therefore, further studies are required to produce hPSC-derived kidney organoids that accurately mimic human kidneys to facilitate research on kidney development, regeneration, disease modeling, and drug screening. In this review, we discussed recent advances in the generation of hPSC-derived kidney organoids, how these organoids contribute to the understanding of human kidney development and research in disease modeling. Additionally, the limitations, future research focus, and applications of hPSC-derived kidney organoids were highlighted.
Collapse
Affiliation(s)
- Hong-Yan Long
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Zu-Ping Qian
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Qin Lan
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Yong-Jie Xu
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Jing-Jing Da
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Fu-Xun Yu
- Key Laboratory of Diagnosis and Treatment of Pulmonary Immune Diseases, National Health Commission, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Yan Zha
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China.
| |
Collapse
|
36
|
Garcia ABDM, Viola GD, Corrêa BDS, Fischer TDS, Pinho MCDF, Rodrigues GM, Ashton-Prolla P, Rosset C. An overview of actionable and potentially actionable TSC1 and TSC2 germline variants in an online Database. Genet Mol Biol 2024; 46:e20230132. [PMID: 38373162 PMCID: PMC10876083 DOI: 10.1590/1678-4685-gmb-2023-0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/26/2023] [Indexed: 02/21/2024] Open
Abstract
Tuberous Sclerosis Complex (TSC) is caused by loss of function germline variants in the TSC1 or TSC2 tumor suppressor genes. Genetic testing for the detection of pathogenic variants in either TSC1 or TSC2 was implemented as a diagnostic criterion for TSC. However, TSC molecular diagnosis can be challenging due to the absence of variant hotspots and the high number of variants described. This review aimed to perform an overview of TSC1/2 variants submitted in the ClinVar database. Variants of uncertain significance (VUS), missense and single nucleotide variants were the most frequent in clinical significance (37-40%), molecular consequence (37%-39%) and variation type (82%-83%) categories in ClinVar in TSC1 and TSC2 variants, respectively. Frameshift and nonsense VUS have potential for pathogenic reclassification if further functional and segregation studies were performed. Indeed, there were few functional assays deposited in the database and literature. In addition, we did not observe hotspots for variation and many variants presented conflicting submissions regarding clinical significance. This study underscored the importance of disseminating molecular diagnostic results in a public database to render the information largely accessible and promote accurate diagnosis. We encourage the performance of functional studies evaluating the pathogenicity of TSC1/2 variants.
Collapse
Affiliation(s)
- Arthur Bandeira de Mello Garcia
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Guilherme Danielski Viola
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Bruno da Silveira Corrêa
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Taís da Silveira Fischer
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
| | - Maria Clara de Freitas Pinho
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Centro Universitário CESUCA, Cachoeirinha, RS, Brazil
| | - Grazielle Motta Rodrigues
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| | - Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Departamento de Genética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Clévia Rosset
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Ciências Médicas, Porto Alegre, RS, Brazil
| |
Collapse
|
37
|
Na B, Shah S, Nghiemphu PL. Cancer Predisposition Syndromes in Neuro-oncology. Semin Neurol 2024; 44:16-25. [PMID: 38096910 DOI: 10.1055/s-0043-1777702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Although most primary central and peripheral nervous system (NS) tumors occur sporadically, there are a subset that may arise in the context of a cancer predisposition syndrome. These syndromes occur due to a pathogenic mutation in a gene that normally functions as a tumor suppressor. With increased understanding of the molecular pathogenesis of these tumors, more people have been identified with a cancer predisposition syndrome. Identification is crucial, as this informs surveillance, diagnosis, and treatment options. Moreover, relatives can also be identified through genetic testing. Although there are many cancer predisposition syndromes that increase the risk of NS tumors, in this review, we focus on three of the most common cancer predisposition syndromes, neurofibromatosis type 1, neurofibromatosis type 2, and tuberous sclerosis complex type 1 and type 2, emphasizing the clinical manifestations, surveillance guidelines, and treatment options.
Collapse
Affiliation(s)
- Brian Na
- Department of Neurology, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Shilp Shah
- Department of Bioengineering, UCLA Samueli School of Engineering, Los Angeles, California
| | | |
Collapse
|
38
|
López-Aranda MF, Bach K, Bui R, Phan M, Lu O, Thadani C, Luchetti A, Mandanas R, Herrera I, López-Ávalos MD, Silva AJ. Early Post-Natal Immune Activation Leads to Object Memory Deficits in Female Tsc2+/- Mice: The Importance of Including Both Sexes in Neuroscience Research. Biomedicines 2024; 12:203. [PMID: 38255309 PMCID: PMC10813674 DOI: 10.3390/biomedicines12010203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
There is evidence that viral infections during pre-natal development constitute a risk factor for neuropsychiatric disorders and lead to learning and memory deficits. However, little is known about why viral infections during early post-natal development have a different impact on learning and memory depending on the sex of the subject. We previously showed that early post-natal immune activation induces hippocampal-dependent social memory deficits in a male, but not in a female, mouse model of tuberous sclerosis complex (TSC; Tsc2+/- mice). Here, we explored the impact of a viral-like immune challenge in object memory. We demonstrate that early post-natal immune activation (during the first 2 weeks of life) leads to object memory deficits in female, but not male, mice that are heterozygous for a gene responsible for tuberous sclerosis complex (Tsc2+/- mice), while no effect was observed in wild type (WT) mice. Moreover, we found that the same immune activation in Tsc2+/- adult mice was not able to cause object memory deficits in females, which suggests that the early post-natal development stage constitutes a critical window for the effects of immune challenge on adult memory. Also, our results suggest that mTOR plays a critical role in the observed deficit in object memory in female Tsc2+/- mice. These results, together with previous results published by our laboratory, showing sex-specific memory deficits due to early post-natal immune activation, reinforce the necessity of using both males and females for research studies. This is especially true for studies related to immune activation, since the higher levels of estrogens in females are known to affect inflammation and to provide neuroprotection.
Collapse
Affiliation(s)
- Manuel F. López-Aranda
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, 29590 Málaga, Spain
| | - Karen Bach
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Raymond Bui
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Miranda Phan
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Odilia Lu
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Chirag Thadani
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Rochelle Mandanas
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - Isaiah Herrera
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| | - María Dolores López-Ávalos
- Departamento de Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, 29590 Málaga, Spain
| | - Alcino J. Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095, USA (A.J.S.)
| |
Collapse
|
39
|
Bosetti C, Ferrini L, Ferrari AR, Bartolini E, Calderoni S. Children with Autism Spectrum Disorder and Abnormalities of Clinical EEG: A Qualitative Review. J Clin Med 2024; 13:279. [PMID: 38202286 PMCID: PMC10779511 DOI: 10.3390/jcm13010279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/22/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
Over the last decade, the comorbidity between Autism Spectrum Disorder (ASD) and epilepsy has been widely demonstrated, and many hypotheses regarding the common neurobiological bases of these disorders have been put forward. A variable, but significant, prevalence of abnormalities on electroencephalogram (EEG) has been documented in non-epileptic children with ASD; therefore, several scientific studies have recently tried to demonstrate the role of these abnormalities as a possible biomarker of altered neural connectivity in ASD individuals. This narrative review intends to summarize the main findings of the recent scientific literature regarding abnormalities detected with standard EEG in children/adolescents with idiopathic ASD. Research using three different databases (PubMed, Scopus and Google Scholar) was conducted, resulting in the selection of 10 original articles. Despite an important lack of studies on preschoolers and a deep heterogeneity in results, some authors speculated on a possible association between EEG abnormalities and ASD characteristics, in particular, the severity of symptoms. Although this correlation needs to be more strongly elucidated, these findings may encourage future studies aimed at demonstrating the role of electrical brain abnormalities as an early biomarker of neural circuit alterations in ASD, highlighting the potential diagnostic, prognostic and therapeutic value of EEG in this field.
Collapse
Affiliation(s)
- Chiara Bosetti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Luca Ferrini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
| | - Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Tuscany PhD Programme in Neurosciences, 50139 Florence, Italy
| | - Sara Calderoni
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (C.B.); (L.F.); (A.R.F.); (S.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
40
|
Roberds SL, Fuchs Z, Cassidy EM, Metzger S, Abi A, Pounders AJ, Aguiar DJ. The role of the TSC Alliance in advancing therapy development: a patient organization perspective. THERAPEUTIC ADVANCES IN RARE DISEASE 2024; 5:26330040241265411. [PMID: 39070094 PMCID: PMC11273576 DOI: 10.1177/26330040241265411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024]
Abstract
Tuberous sclerosis complex (TSC) is a genetic disease leading to malformations, or tubers, in the cerebral cortex and growth of tumors, most frequently in the brain, heart, kidneys, skin, and lungs. Changes in the brain caused by TSC usually have the biggest negative impact on quality of life. Approximately 85% of individuals with TSC have epilepsy, and TSC-associated neuropsychiatric disorders (TAND) affect nearly all individuals with TSC in some way. TSC Alliance's research strategy is built upon both funding and catalyzing research. Through grants, the organization provides funding directly to researchers through a competitive application process. The organization has also built a set of resources available to researchers worldwide, including a Natural History Database, Biosample Repository, and Preclinical Consortium. These resources catalyze research because they are available to qualified academic or industry researchers around the world, enabling an almost unlimited number of scientists to access data and resources to enable and accelerate research on TSC. This research strategy continues to be shaped by the needs and priorities of the TSC community, working toward a future where everyone affected by TSC can live their fullest lives.
Collapse
Affiliation(s)
| | - Zoë Fuchs
- TSC Alliance, Silver Spring, MD, USA
| | | | | | - Ayat Abi
- TSC Alliance, Silver Spring, MD, USA
| | | | | |
Collapse
|
41
|
Hu JS, Malik R, Sohal VS, Rubenstein JL, Vogt D. Tsc1 Loss in VIP-Lineage Cortical Interneurons Results in More VIP+ Interneurons and Enhanced Excitability. Cells 2023; 13:52. [PMID: 38201256 PMCID: PMC10777938 DOI: 10.3390/cells13010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway is a powerful regulator of cell proliferation, growth, synapse maintenance and cell fate. While intensely studied for its role in cancer, the role of mTOR signaling is just beginning to be uncovered in specific cell types that are implicated in neurodevelopmental disorders. Previously, loss of the Tsc1 gene, which results in hyperactive mTOR, was shown to affect the function and molecular properties of GABAergic cortical interneurons (CINs) derived from the medial ganglionic eminence. To assess if other important classes of CINs could be impacted by mTOR dysfunction, we deleted Tsc1 in a caudal ganglionic eminence-derived interneuron group, the vasoactive intestinal peptide (VIP)+ subtype, whose activity disinhibits local circuits. Tsc1 mutant VIP+ CINs reduced their pattern of apoptosis from postnatal days 15-20, resulting in increased VIP+ CINs. The mutant CINs exhibited synaptic and electrophysiological properties that could contribute to the high rate of seizure activity in humans that harbor Tsc1 mutations.
Collapse
Affiliation(s)
- Jia Sheng Hu
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ruchi Malik
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
| | - Vikaas S. Sohal
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
- Center for Integrative Neuroscience, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
- Sloan-Swartz Center for Theoretical Neurobiology, University of California San Francisco, 1550 4th St., San Francisco, CA 94158, USA
| | - John L. Rubenstein
- Department of Psychiatry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Daniel Vogt
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI 49503, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
42
|
Riley VA, Shankar V, Holmberg JC, Sokolov AM, Neckles VN, Williams K, Lyman R, Mackay TF, Feliciano DM. Tsc2 coordinates neuroprogenitor differentiation. iScience 2023; 26:108442. [PMID: 38107199 PMCID: PMC10724693 DOI: 10.1016/j.isci.2023.108442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/22/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem cells (NSCs) of the ventricular-subventricular zone (V-SVZ) generate numerous cell types. The uncoupling of mRNA transcript availability and translation occurs during the progression from stem to differentiated states. The mTORC1 kinase pathway acutely controls proteins that regulate mRNA translation. Inhibiting mTORC1 during differentiation is hypothesized to be critical for brain development since somatic mutations of mTORC1 regulators perturb brain architecture. Inactivating mutations of TSC1 or TSC2 genes cause tuberous sclerosis complex (TSC). TSC patients have growths near the striatum and ventricles. Here, it is demonstrated that V-SVZ NSC Tsc2 inactivation causes striatal hamartomas. Tsc2 removal altered translation factors, translatomes, and translational efficiency. Single nuclei RNA sequencing following in vivo loss of Tsc2 revealed changes in NSC activation states. The inability to decouple mRNA transcript availability and translation delayed differentiation leading to the retention of immature phenotypes in hamartomas. Taken together, Tsc2 is required for translational repression and differentiation.
Collapse
Affiliation(s)
- Victoria A. Riley
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | - Vijay Shankar
- Department of Biochemistry and Genetics, Clemson University, Clemson, SC, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, USA
| | | | - Aidan M. Sokolov
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | | | - Kaitlyn Williams
- Clemson University Genomics and Bioinformatics Facility (CUGBF), Clemson University, Clemson, SC, USA
| | - Rachel Lyman
- Department of Biochemistry and Genetics, Clemson University, Clemson, SC, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, USA
| | - Trudy F.C. Mackay
- Department of Biochemistry and Genetics, Clemson University, Clemson, SC, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, USA
| | - David M. Feliciano
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, USA
| |
Collapse
|
43
|
Vitale G, Terrone G, Vitale S, Vitulli F, Aiello S, Bravaccio C, Pisano S, Bove I, Rizzo F, Seetahal-Maraj P, Wiese T. The Evolving Landscape of Therapeutics for Epilepsy in Tuberous Sclerosis Complex. Biomedicines 2023; 11:3241. [PMID: 38137462 PMCID: PMC10741146 DOI: 10.3390/biomedicines11123241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a rare multisystem genetic disorder characterized by benign tumor growth in multiple organs, including the brain, kidneys, heart, eyes, lungs, and skin. Pathogenesis stems from mutations in either the TSC1 or TSC2 gene, which encode the proteins hamartin and tuberin, respectively. These proteins form a complex that inhibits the mTOR pathway, a critical regulator of cell growth and proliferation. Disruption of the tuberin-hamartin complex leads to overactivation of mTOR signaling and uncontrolled cell growth, resulting in hamartoma formation. Neurological manifestations are common in TSC, with epilepsy developing in up to 90% of patients. Seizures tend to be refractory to medical treatment with anti-seizure medications. Infantile spasms and focal seizures are the predominant seizure types, often arising in early childhood. Drug-resistant epilepsy contributes significantly to morbidity and mortality. This review provides a comprehensive overview of the current state of knowledge regarding the pathogenesis, clinical manifestations, and treatment approaches for epilepsy and other neurological features of TSC. While narrative reviews on TSC exist, this review uniquely synthesizes key advancements across the areas of TSC neuropathology, conventional and emerging pharmacological therapies, and targeted treatments. The review is narrative in nature, without any date restrictions, and summarizes the most relevant literature on the neurological aspects and management of TSC. By consolidating the current understanding of TSC neurobiology and evidence-based treatment strategies, this review provides an invaluable reference that highlights progress made while also emphasizing areas requiring further research to optimize care and outcomes for TSC patients.
Collapse
Affiliation(s)
- Giovanni Vitale
- Neuroscience and Rare Diseases, Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), F. Hoffmann–La Roche, 4070 Basel, Switzerland
| | - Gaetano Terrone
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Samuel Vitale
- School of Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy;
| | - Francesca Vitulli
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, University of Naples Federico II, 80138 Naples, Italy (I.B.)
| | - Salvatore Aiello
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Carmela Bravaccio
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Simone Pisano
- Department of Translational Medical Sciences, Child and Adolescent Neuropsychiatry, University of Naples Federico II, 80138 Naples, Italy; (G.T.); (C.B.)
| | - Ilaria Bove
- Department of Neurosciences and Reproductive and Dental Sciences, Division of Neurosurgery, University of Naples Federico II, 80138 Naples, Italy (I.B.)
| | - Francesca Rizzo
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy;
| | | | - Thomas Wiese
- Neuroscience and Rare Diseases, Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), F. Hoffmann–La Roche, 4070 Basel, Switzerland
| |
Collapse
|
44
|
Cockerell I, Christensen J, Hoei-Hansen CE, Holst L, Grenaa Frederiksen M, Issa-Epe AI, Nedregaard B, Solhoff R, Heimdal K, Johannessen Landmark C, Lund C, Nærland T. Effectiveness and safety of everolimus treatment in patients with tuberous sclerosis complex in real-world clinical practice. Orphanet J Rare Dis 2023; 18:377. [PMID: 38042867 PMCID: PMC10693167 DOI: 10.1186/s13023-023-02982-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/18/2023] [Indexed: 12/04/2023] Open
Abstract
BACKGROUND The randomised double-blinded placebo-controlled EXIST-1-3 studies have showed everolimus effective with adverse effects reported as acceptable in treatment of symptoms in patients with tuberous sclerosis complex (TSC), although evidence of outcomes in clinical practice remains limited. This study aimed to investigate, in clinical practice, the effectiveness and safety of everolimus for epilepsy, renal angiomyolipoma (rAML), and subependymal giant cell astrocytoma (SEGA) in patients with TSC. RESULTS The study included 64 patients with TSC (median age: 19, range 0.9-54 years) receiving everolimus treatment (Norway: n = 35; Denmark: n = 29). Among 45 patients with epilepsy, 14 (31%) were responders experiencing ≥ 50% reduction in seizure frequency in the last 3 months of treatment compared with the last 3 months before treatment. Nineteen (42%) patients changed their anti-seizure medications (ASMs). Responders were more common among patients < 18 years (46%) than among patients ≥ 18 years (14%, p = 0.03). In 29 patients with rAML, everolimus reduced (≥ 30% decrease) and stabilized (< 20% increase, ≤ 30% decrease) longest diameter of rAML in 38% and 59%, respectively, after a mean treatment duration of 37 months. SEGA volume was reduced in three patients by 71%, 43%, and 48% after 39, 34, and 82 months. Adverse effects were reported in 61 of 64 patients (95%) after a median treatment duration of 31 months (range 0-106), with oral ulceration/stomatitis (63%) and upper respiratory tract infections (38%) being the most common. The most common laboratory abnormalities were increased cholesterol (41%), anaemia (30%), and leucopoenia (25%). Grade 3-4 adverse effects were reported in 36% of cases, and life-threatening conditions were reported in two patients. Nine patients discontinued everolimus treatment. CONCLUSIONS Seizure reduction in this study sample was consistent with results from EXIST, but might be lower than expected, given that changes in concomitant ASMs are part of clinical practice. Seizure reduction was associated with younger age. As with EXIST, everolimus reduced or stabilised rAML size in most patients. SEGA volume was reduced in all three patients. Close follow-up is needed for this group, especially for children and patients who may not be able to report adverse effects.
Collapse
Affiliation(s)
- Ine Cockerell
- Department of Rare Disorders and Disabilities, National Centre for Rare Epilepsy-Related Disorders, Oslo University Hospital, Pb 4950, 0424, Nydalen, Oslo, Norway.
| | - Jakob Christensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Neurology, Aarhus University Hospital, Affiliated Member of the European Reference Network EpiCARE, Aarhus, Denmark
| | - Christina E Hoei-Hansen
- Department of Clinical Medicine, Copenhagen University, Copenhagen, Denmark
- Department of Paediatrics, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Lotte Holst
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Aart Imran Issa-Epe
- Section of Abdominal Radiology, Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Bård Nedregaard
- Section of Neuroradiology, Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Ragnar Solhoff
- Department of Neurology, Sørlandet Hospital, Arendal, Norway
| | - Ketil Heimdal
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Cecilie Johannessen Landmark
- Department of Pharmacy, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
- The National Center for Epilepsy (SSE), Member of the ERN EpiCare, Oslo University Hospital, Oslo, Norway
- Section for Clinical Pharmacology, SSE, Department of Pharmacology, Oslo University Hospital, Oslo, Norway
| | - Caroline Lund
- Department of Rare Disorders and Disabilities, National Centre for Rare Epilepsy-Related Disorders, Oslo University Hospital, Pb 4950, 0424, Nydalen, Oslo, Norway
| | - Terje Nærland
- K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- NevSom, Department of Rare Disorders and Disabilities, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
45
|
Li HP, Huang CY, Lui KW, Chao YK, Yeh CN, Lee LY, Huang Y, Lin TL, Kuo YC, Huang MY, Fan HC, Lin AC, Hsieh CH, Chang KP, Lin CY, Wang HM, Chao M, Liu JS, Chang YS, Hsu CL. Nasopharyngeal carcinoma patient-derived xenograft mouse models reveal potential drugs targeting cell cycle, mTOR, and autophagy pathways. Transl Oncol 2023; 38:101785. [PMID: 37713975 PMCID: PMC10509698 DOI: 10.1016/j.tranon.2023.101785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is associated with Epstein-Barr virus (EBV) infection. To test preclinical NPC drugs, we established two patient-derived xenograft (PDX) mouse models, EBV-positive PDX-B13 and EBV-negative PDX-Li41, for drug screening. METHODS Based on next generation sequencing (NGS) studies, PDX-B13 had CCND1 copy number (CN) gain but CDKN2A CN loss, whereas PDX-Li41 had CDKN2A and RB1 CN loss, TSC1 (negative regulator of mTOR) frameshift deletion mutation, and increased activation of mTOR, a serine/threonine kinase that governs metabolism, autophagy, and apoptosis. Increased mTOR was also associated with poor NPC prognosis. RESULTS Everolimus, an mTOR inhibitor, suppressed tumor growth in the two PDX NPC models and had an additive antitumor effect with palbociclib, a CDK4/6 inhibitor. PDX tumors treated with various drugs or untreated were subjected to RNA sequencing, transcriptome profile analysis, and selective Western blotting to understand the interactions between these drugs and gene expression profiles. Palbociclib also suppressed EB viral nuclear antigen (EBNA1) expression in PDX-B13. Everolimus together with autophagy inhibitor, hydroxychloroquine, had additive anti-tumor effect on PDX-B13 tumor. Immunohistochemistry revealed that high mTOR levels were correlated with poor overall survival in patients with metastatic NPC (N = 90). CONCLUSIONS High mTOR levels are a poor prognostic factor in NPC, and cell cycle, mTOR and autophagy pathways may serve as therapeutic targets in NPC. In addition, PDX models can be used for efficiently testing potential NPC drugs.
Collapse
Affiliation(s)
- Hsin-Pai Li
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Kar-Wai Lui
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yin-Kai Chao
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chun-Nan Yeh
- Department of General Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Tung-Liang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yung-Chia Kuo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Mei-Yuan Huang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hsien-Chi Fan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - An-Chi Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chien-Yu Lin
- Department of Radiation, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hung-Ming Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Mei Chao
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Jai-Shin Liu
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Yu-Sun Chang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan; School of Medicine, Chang Gung University, Taoyuan 33305, Taiwan.
| |
Collapse
|
46
|
Frappaolo A, Giansanti MG. Using Drosophila melanogaster to Dissect the Roles of the mTOR Signaling Pathway in Cell Growth. Cells 2023; 12:2622. [PMID: 37998357 PMCID: PMC10670727 DOI: 10.3390/cells12222622] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023] Open
Abstract
The evolutionarily conserved target of rapamycin (TOR) serine/threonine kinase controls eukaryotic cell growth, metabolism and survival by integrating signals from the nutritional status and growth factors. TOR is the catalytic subunit of two distinct functional multiprotein complexes termed mTORC1 (mechanistic target of rapamycin complex 1) and mTORC2, which phosphorylate a different set of substrates and display different physiological functions. Dysregulation of TOR signaling has been involved in the development and progression of several disease states including cancer and diabetes. Here, we highlight how genetic and biochemical studies in the model system Drosophila melanogaster have been crucial to identify the mTORC1 and mTORC2 signaling components and to dissect their function in cellular growth, in strict coordination with insulin signaling. In addition, we review new findings that involve Drosophila Golgi phosphoprotein 3 in regulating organ growth via Rheb-mediated activation of mTORC1 in line with an emerging role for the Golgi as a major hub for mTORC1 signaling.
Collapse
Affiliation(s)
- Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| |
Collapse
|
47
|
Karita H, Tsuda K, Kono M, Yamamoto T, Ihara S. Neoadjuvant Therapy with Everolimus for Subependymal Giant Cell Astrocytoma: A Case Report. NMC Case Rep J 2023; 10:291-297. [PMID: 37953906 PMCID: PMC10635902 DOI: 10.2176/jns-nmc.2022-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/31/2023] [Indexed: 11/14/2023] Open
Abstract
Direct surgical resection remains to be the standard treatment for tuberous sclerosis complex (TSC) with subependymal giant cell astrocytoma (SEGA). Medical therapy with everolimus (mammalian target of rapamycin inhibitor or mTOR) serves as a second-line treatment for patients with SEGA who are determined to be ineligible for surgical resection. Some recent studies have reported that neoadjuvant therapy for SEGA may be a useful, novel treatment. In this study, we herein present a case of SEGA and demonstrate the efficacy of preoperative everolimus therapy. We have also examined the utility and safety of neoadjuvant therapy for SEGA and investigated four previously reported cases of preoperative administration of mTOR inhibitors. In these cases, everolimus was administered preoperatively to shrink the tumor although the duration of treatment varied. Afterward, gross total tumor removal was conducted in all the cases. No postoperative complications were reported during the follow-up period. These findings indicate that neoadjuvant therapy with an mTOR inhibitor can be a potential treatment for SEGA. The findings of this present study also suggested that a short administration period of about 2 months may be sufficient to achieve preoperative tumor reduction.
Collapse
Affiliation(s)
- Hiroki Karita
- Department of Neurosurgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Kyoji Tsuda
- Department of Neurosurgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Maya Kono
- Department of Neurosurgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Tetsuya Yamamoto
- Department of Neurosurgery, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Satoshi Ihara
- Department of Neurosurgery, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| |
Collapse
|
48
|
Chalkley MBL, Mersfelder RB, Sundberg M, Armstrong LC, Sahin M, Ihrie RA, Ess KC. Non-canonical functions of a mutant TSC2 protein in mitotic division. PLoS One 2023; 18:e0292086. [PMID: 37792789 PMCID: PMC10550124 DOI: 10.1371/journal.pone.0292086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/12/2023] [Indexed: 10/06/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a debilitating developmental disorder characterized by a variety of clinical manifestations. TSC is caused by mutations in the TSC1 or TSC2 genes, which encode the hamartin/tuberin proteins respectively. These proteins function as a heterodimer that negatively regulates the mechanistic Target of Rapamycin Complex 1 (mTORC1). TSC research has focused on the effects of mTORC1, a critical signaling hub, on regulation of diverse cell processes including metabolism, cell growth, translation, and neurogenesis. However, non-canonical functions of TSC2 are not well studied, and the potential disease-relevant biological mechanisms of mutations affecting these functions are not well understood. We observed aberrant multipolar mitotic division, a novel phenotype, in TSC2 mutant iPSCs. The multipolar phenotype is not meaningfully affected by treatment with the inhibitor rapamycin. We further observed dominant negative activity of the mutant form of TSC2 in producing the multipolar division phenotype. These data expand the knowledge of TSC2 function and pathophysiology which will be highly relevant to future treatments for patients with TSC.
Collapse
Affiliation(s)
- Mary-Bronwen L. Chalkley
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Rachel B. Mersfelder
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Maria Sundberg
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Laura C. Armstrong
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rebecca A. Ihrie
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Kevin C. Ess
- Department of Cell & Developmental Biology, School of Medicine Basic Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
49
|
Elbeltagy M, Abbassy M. Neurofibromatosis type1, type 2, tuberous sclerosis and Von Hippel-Lindau disease. Childs Nerv Syst 2023; 39:2791-2806. [PMID: 37819506 DOI: 10.1007/s00381-023-06160-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/16/2023] [Indexed: 10/13/2023]
Abstract
Neurocutaneous syndromes (also known as phakomatoses) are heterogenous group of disorders that involve derivatives of the neuroectoderm. Each disease has diagnostic and pathognomonic criteria, once identified, thorough clinical examination to the patient and the family members should be done. Magnetic resonance imaging (MRI) is used to study the pathognomonic findings withing the CNS (Evans et al. in Am J Med Genet A 152A:327-332, 2010). This chapter includes the 4 most common syndromes faced by neurosurgeons and neurologists; neurofibromatosis types 1 and 2, tuberous sclerosis and Von Hippel-Lindau disease. Each syndrome has specific genetic anomaly that involves a tumor suppressor gene and the loss of inhibition of specific pathways. The result is a spectrum of cutaneous manifestations and neoplasms.
Collapse
Affiliation(s)
- M Elbeltagy
- Department of Neurosurgery, Cairo University, 1 University Street, Giza Governorate, 12613, Egypt.
- Department of Neurosurgery, Children's Cancer Hospital Egypt, Sekat Hadid Al Mahger, Zeinhom, El Sayeda Zeinab, Cairo Governorate, 4260102, Egypt.
| | - M Abbassy
- Department of Neurosurgery, Children's Cancer Hospital Egypt, Sekat Hadid Al Mahger, Zeinhom, El Sayeda Zeinab, Cairo Governorate, 4260102, Egypt
- Department of Neurosurgery, Alexandria University, 22 El-Gaish Rd, Al Azaritah WA Ash Shatebi, Bab Sharqi, Alexandria Governorate, 5424041, Egypt
| |
Collapse
|
50
|
Ura H, Togi S, Niida Y. Target-capture full-length double-stranded cDNA long-read sequencing through Nanopore revealed novel intron retention in patient with tuberous sclerosis complex. Front Genet 2023; 14:1256064. [PMID: 37829285 PMCID: PMC10565506 DOI: 10.3389/fgene.2023.1256064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a relatively common autosomal dominant disorder characterized by multiple dysplastic organ lesions and neuropsychiatric symptoms caused by loss-of-function mutation of either TSC1 or TSC2. The genetic diagnosis of inherited diseases, including TSC, in the clinical field is widespread using next-generation sequencing. The mutations in protein-coding exon tend to be verified because mutations directly cause abnormal protein. However, it is relatively difficult to verify mutations in the intron region because it is required to investigate whether the intron mutations affect the abnormal splicing of transcripts. In this study, we developed a target-capture full-length double-stranded cDNA sequencing method using Nanopore long-read sequencer (Nanopore long-read target sequencing). This method revealed the occurrence of intron mutation in the TSC2 gene and found that the intron mutation produces novel intron retention splicing transcripts that generate truncated proteins. The protein-coding transcripts were decreased due to the expression of the novel intron retention transcripts, which caused TSC in patients with the intron mutation. Our results indicate that Nanopore long-read target sequencing is useful for the detection of mutations and confers information on the full-length alternative splicing of transcripts for genetic diagnosis.
Collapse
Affiliation(s)
- Hiroki Ura
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada, Ishikawa, Japan
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Sumihito Togi
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada, Ishikawa, Japan
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada, Ishikawa, Japan
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|