108 results on '"Jeffrey W. Innis"'
Search Results
2. AHDC1 missense mutations in Xia-Gibbs syndrome
- Author
-
Michael M. Khayat, Jianhong Hu, Yunyun Jiang, He Li, Varuna Chander, Moez Dawood, Adam W. Hansen, Shoudong Li, Jennifer Friedman, Laura Cross, Emilia K. Bijlsma, Claudia A.L. Ruivenkamp, Francis H. Sansbury, Jeffrey W. Innis, Jessica Omark O’Shea, Qingchang Meng, Jill A. Rosenfeld, Kirsty McWalter, Michael F. Wangler, James R. Lupski, Jennifer E. Posey, David Murdock, and Richard A. Gibbs
- Subjects
AHDC1 ,Xia-Gibbs syndrome ,missense mutation ,de novo mutation ,Genetics ,QH426-470 - Abstract
Summary: Xia-Gibbs syndrome (XGS; MIM: 615829) is a phenotypically heterogeneous neurodevelopmental disorder (NDD) caused by newly arising mutations in the AT-Hook DNA-Binding Motif-Containing 1 (AHDC1) gene that are predicted to lead to truncated AHDC1 protein synthesis. More than 270 individuals have been diagnosed with XGS worldwide. Despite the absence of an independent assay for AHDC1 protein function to corroborate potential functional consequences of rare variant genetic findings, there are also reports of individuals with XGS-like trait manifestations who have de novo missense AHDC1 mutations and who have been provided a molecular diagnosis of the disorder. To investigate a potential contribution of missense mutations to XGS, we mapped the missense mutations from 10 such individuals to the AHDC1 conserved protein domain structure and detailed the observed phenotypes. Five newly identified individuals were ascertained from a local XGS Registry, and an additional five were taken from external reports or databases, including one publication. Where clinical data were available, individuals with missense mutations all displayed phenotypes consistent with those observed in individuals with AHDC1 truncating mutations, including delayed motor milestones, intellectual disability (ID), hypotonia, and speech delay. A subset of the 10 reported missense mutations cluster in two regions of the AHDC1 protein with known conserved domains, likely representing functional motifs. Variants outside the clustered regions score lower for computational prediction of their likely damaging effects. Overall, de novo missense variants in AHDC1 are likely diagnostic of XGS when in silico analysis of their position relative to conserved regions is considered together with disease trait manifestations.
- Published
- 2021
- Full Text
- View/download PDF
3. Natural history of ROHHAD syndrome: development of severe insulin resistance and fatty liver disease over time
- Author
-
Abdel Wahab Jalal Eldin, Dilara Tombayoglu, Laura Butz, Alison Affinati, Rasimcan Meral, Mehmet Selman Ontan, Kelly Walkovich, Maria Westerhoff, Jeffrey W. Innis, Neehar D. Parikh, and Elif A. Oral
- Subjects
ROHHAD ,Insulin resistance ,GLP-1 agonists ,Obesity ,Atypical diabetes ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Absract Background Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation (ROHHAD) is a rare syndrome with unknown etiology. Metabolic abnormalities are not known to be part of the syndrome. We present one of the oldest cases reported in the literature, who developed severe metabolic abnormalities and hepatic disease suggesting that these features may be part of the syndrome. Case presentation A 27-year-old woman, diagnosed with ROHHAD syndrome at age 15, who previously developed diabetes insipidus, growth hormone deficiency, hyperprolactinemia, and hypothyroidism in her first decade of life. This was followed by insulin resistance, NAFLD, liver fibrosis, and splenomegaly before age 14 years. Her regimen included a short course of growth hormone, and cyclic estrogen and progesterone. Her metabolic deterioration continued despite treatment with metformin. Interestingly, she had a favorable response to liraglutide therapy despite having a centrally mediated cause for her obesity. At age 26, a 1.6 cm lesion was found incidentally in her liver. Liver biopsy showed hepatocellular carcinoma which was successfully treated with radiofrequency ablation. Conclusion Metabolic abnormalities, Insulin resistance and fatty liver disease are potentially part of the ROHHAD syndrome that may develop over time. GLP1 agonists were reasonably effective to treat insulin resistance and hyperphagia. Patients with ROHHAD may benefit from close follow up in regards to liver disease.
- Published
- 2019
- Full Text
- View/download PDF
4. Potential association of LMNA-associated generalized lipodystrophy with juvenile dermatomyositis
- Author
-
Melis Sahinoz, Shafaq Khairi, Ashley Cuttitta, Graham F. Brady, Amit Rupani, Rasimcan Meral, Marwan K. Tayeh, Peedikayil Thomas, Meredith Riebschleger, Sandra Camelo-Piragua, Jeffrey W. Innis, M. Bishr Omary, Daniel E. Michele, and Elif A. Oral
- Subjects
Laminopathies ,Myositis ,LMNA ,Whole exome sequencing ,P.T10I ,Muscle biopsy ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Abstract Background Juvenile dermatomyositis (JDM) is an auto-immune muscle disease which presents with skin manifestations and muscle weakness. At least 10% of the patients with JDM present with acquired lipodystrophy. Laminopathies are caused by mutations in the lamin genes and cover a wide spectrum of diseases including muscular dystrophies and lipodystrophy. The p.T10I LMNA variant is associated with a phenotype of generalized lipodystrophy that has also been called atypical progeroid syndrome. Case presentation A previously healthy female presented with bilateral proximal lower extremity muscle weakness at age 4. She was diagnosed with JDM based on her clinical presentation, laboratory tests and magnetic resonance imaging (MRI). She had subcutaneous fat loss which started in her extremities and progressed to her whole body. At age 7, she had diabetes, hypertriglyceridemia, low leptin levels and low body fat on dual energy X-ray absorptiometry (DEXA) scan, and was diagnosed with acquired generalized lipodystrophy (AGL). Whole exome sequencing (WES) revealed a heterozygous c.29C > T; p.T10I missense pathogenic variant in LMNA, which encodes lamins A and C. Muscle biopsy confirmed JDM rather than muscular dystrophy, showing perifascicular atrophy and perivascular mononuclear cell infiltration. Immunofluroscence of skin fibroblasts confirmed nuclear atypia and fragmentation. Conclusions This is a unique case with p.T10I LMNA variant displaying concurrent JDM and AGL. This co-occurrence raises the intriguing possibility that LMNA, and possibly p.T10I, may have a pathogenic role in not only the occurrence of generalized lipodystrophy, but also juvenile dermatomyositis. Careful phenotypic characterization of additional patients with laminopathies as well as individuals with JDM is warranted.
- Published
- 2018
- Full Text
- View/download PDF
5. Design and Outcomes of a Novel Multidisciplinary Ophthalmic Genetics Clinic
- Author
-
Bela Parekh, Adelyn Beil, Bridget Blevins, Adam Jacobson, Pamela Williams, Jeffrey W. Innis, Amanda Barone Pritchard, and Lev Prasov
- Subjects
ophthalmic genetics ,medical genetics ,inherited ocular disorders ,microphthalmia ,congenital cataracts ,optic neuropathy ,anterior segment dysgenesis ,Bosch–Boonstra–Schaaf syndrome ,nystagmus ,Genetics ,Genetics (clinical) - Abstract
The Multidisciplinary Ophthalmic Genetics Clinic (MOGC) at the University of Michigan Kellogg Eye Center aims to provide medical and ophthalmic genetics care to patients with inherited ocular conditions. We have developed a clinical and referral workflow where each patient undergoes coordinated evaluation by our multidisciplinary team followed by discussions on diagnosis, prognosis, and genetic testing. Testing approaches are specific to each patient and can be targeted (single-gene, gene panel), broad (chromosomal microarray, whole-exome sequencing), or a combination. We hypothesize that this clinic model improves patient outcomes and quality of care. A retrospective chart review of patients in the MOGC from July 2020 to October 2022 revealed that the most common referral diagnoses were congenital cataracts, optic neuropathy, and microphthalmia, with 52% syndromic cases. Within this patient cohort, we saw a 76% uptake for genetic testing, among which 33% received a diagnostic test result. Our results support a tailored approach to genetic testing for specific conditions. Through case examples, we highlight the power and impact of our clinic. By integrating ophthalmic care with medical genetics and counseling, the MOGC has not only helped solve individual patient diagnostic challenges but has aided the greater population in novel genetic discoveries and research towards targeted therapeutics.
- Published
- 2023
- Full Text
- View/download PDF
6. A Novel 13q12 Microdeletion Associated with Familial Syndromic Corneal Opacification
- Author
-
Jasmine Y. Serpen, William Presley, Adelyn Beil, Stephen T. Armenti, Kayla Johnson, Shahzad I. Mian, Jeffrey W. Innis, and Lev Prasov
- Subjects
Genetics ,13q12.11 microdeletion ,corneal opacification ,tracheomalacia ,laryngomalacia ,hearing loss ,XPO4 ,LATS2 ,ZDHHC20 ,IFT88 ,SMAD signaling ,Genetics (clinical) - Abstract
Progressive corneal opacification can result from multiple etiologies, including corneal dystrophies or systemic and genetic diseases. We describe a novel syndrome featuring progressive epithelial and anterior stromal opacification in a brother and sister and their mildly affected father, with all three family members having sensorineural hearing loss and two also with tracheomalacia/laryngomalacia. All carried a 1.2 Mb deletion at chromosome 13q12.11, with no other noteworthy co-segregating variants identified on clinical exome or chromosomal microarray. RNAseq analysis from an affected corneal epithelial sample from the proband’s brother revealed downregulation of XPO4, IFT88, ZDHHC20, LATS2, SAP18, and EEF1AKMT1 within the microdeletion interval, with no notable effect on the expression of nearby genes. Pathway analysis showed upregulation of collagen metabolism and extracellular matrix (ECM) formation/maintenance, with no significantly down-regulated pathways. Analysis of overlapping deletions/variants demonstrated that deleterious variants in XPO4 were found in patients with laryngomalacia and sensorineural hearing loss, with the latter phenotype also being a feature of variants in the partially overlapping DFNB1 locus, yet none of these had reported corneal phenotypes. Together, these data define a novel microdeletion-associated syndromic progressive corneal opacification and suggest that a combination of genes within the microdeletion may contribute to ECM dysregulation leading to pathogenesis.
- Published
- 2023
7. Phenotypic spectrum and transcriptomic profile associated with germline variants in TRAF7
- Author
-
Robert A. Hegele, Maria Iascone, Kevin A. Shapiro, Nicolas Chatron, Marwan Shinawi, Joel Charrow, Jeffrey W. Innis, Luitgard Graul-Neumann, Joanna Goes Castro Meira, Anna Lehman, Dawn L. Earl, Victoria R. Sanders, Shannon Rego, David A. Sweetser, Clémantine Dimartino, Wilhelmina S. Kerstjens-Frederikse, Antonio Vitobello, Davor Lessel, Daniel Grinberg, Laurence Faivre, Ryan Peretz, Katherine M. Christensen, Emma Reesor, Erin Beaver, Elizabeth Wohler, Margot R.F. Reijnders, Deborah Barbouth, Anna Cereda, Kaja Kristine Selmer, Melissa A. Walker, Barbro Stadheim, Alessandro Serretti, Helen Kingston, Jill Clayton-Smith, Raymond Lewandowski, Bernarda Lozić, Robert Stratton, Amelia Kirby, Anne H. O’Donnell-Luria, Sara Gabbiadini, Susanna Balcells, Myriam Oufadem, Christel Thauvin, Maha Aly, Wendy K. Chung, Susan M. White, Lauren C. Briere, Thomas Smol, Stanislas Lyonnet, Roberto Colombo, Catherine E. Keegan, Marie T. McDonald, Melanie Parisot, Tiong Yang Tan, Brian Wong, Christopher T. Gordon, Magnus Dehli Vigeland, Frances A. High, Emily Bryant, Audrey Labalme, Nara Sobreira, Arnold Munnich, Jeanne Amiel, Dayna Morel Swols, Raquel Rabionet, Laura Castilla-Vallmanya, Jennifer Heeley, Gunnar Houge, Michael J. Gambello, Bernardo Blanco-Sánchez, Lynn Pais, Olena M. Vaske, Roser Urreizti, Alison Wray, Veronique Pingault, Damien Sanlaville, John Christodoulou, John Millichap, Valérie Cormier-Daire, Parul Jayakar, Helen Cox, Frédéric Tran Mau-Them, Belinda Chong, Victoria Mok Siu, Anne Slavotinek, Antonie J. van Essen, Ingvild Aukrust, Lorne A. Clarke, Rachel Gannaway, Anne Dieux-Coeslier, Patrick Nitschké, Tony Yao, Simon Sadedin, Danielle Karlowicz, Christelle Rougeot, Christine Bole-Feysot, Sandra Yang, Megan T. Cho, Gaetan Lesca, Christiane Zweier, Castilla-Vallmanya L., Selmer K.K., Dimartino C., Rabionet R., Blanco-Sanchez B., Yang S., Reijnders M.R.F., van Essen A.J., Oufadem M., Vigeland M.D., Stadheim B., Houge G., Cox H., Kingston H., Clayton-Smith J., Innis J.W., Iascone M., Cereda A., Gabbiadini S., Chung W.K., Sanders V., Charrow J., Bryant E., Millichap J., Vitobello A., Thauvin C., Mau-Them F.T., Faivre L., Lesca G., Labalme A., Rougeot C., Chatron N., Sanlaville D., Christensen K.M., Kirby A., Lewandowski R., Gannaway R., Aly M., Lehman A., Clarke L., Graul-Neumann L., Zweier C., Lessel D., Lozic B., Aukrust I., Peretz R., Stratton R., Smol T., Dieux-Coeslier A., Meira J., Wohler E., Sobreira N., Beaver E.M., Heeley J., Briere L.C., High F.A., Sweetser D.A., Walker M.A., Keegan C.E., Jayakar P., Shinawi M., Kerstjens-Frederikse W.S., Earl D.L., Siu V.M., Reesor E., Yao T., Hegele R.A., Vaske O.M., Rego S., Shapiro K.A., Wong B., Gambello M.J., McDonald M., Karlowicz D., Colombo R., Serretti A., Pais L., O'Donnell-Luria A., Wray A., Sadedin S., Chong B., Tan T.Y., Christodoulou J., White S.M., Slavotinek A., Barbouth D., Morel Swols D., Parisot M., Bole-Feysot C., Nitschke P., Pingault V., Munnich A., Cho M.T., Cormier-Daire V., Balcells S., Lyonnet S., Grinberg D., Amiel J., Urreizti R., Gordon C.T., MUMC+: DA KG Polikliniek (9), and RS: FHML non-thematic output
- Subjects
0301 basic medicine ,NF-KAPPA-B ,PROTEIN ,030105 genetics & heredity ,medicine.disease_cause ,Germline ,Transcriptome ,ACTIVATION ,POLYUBIQUITINATION ,Missense mutation ,Exome ,Genetics (clinical) ,Genetics ,Sanger sequencing ,Mutation ,leads ,Necrosi ,craniofacial development ,Phenotype ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,intellectual disability ,patent ductus arteriosu ,symbols ,Mutation, Missense ,Biology ,traf7 ,Article ,akt1 ,target ,03 medical and health sciences ,symbols.namesake ,Necrosis ,patent ductus arteriosus ,medicine ,Humans ,blepharophimosi ,Tumors ,MUTATIONS ,Fibroblasts ,medicine.disease ,Blepharophimosis ,TRAF7 ,blepharophimosis ,GENOMIC ANALYSIS ,Germ Cells ,030104 developmental biology ,MENINGIOMAS - Abstract
PURPOSE: Somatic variants in tumor necrosis factor receptor-associated factor 7 (TRAF7) cause meningioma, while germline variants have recently been identified in seven patients with developmental delay and cardiac, facial, and digital anomalies. We aimed to define the clinical and mutational spectrum associated with TRAF7 germline variants in a large series of patients, and to determine the molecular effects of the variants through transcriptomic analysis of patient fibroblasts.METHODS: We performed exome, targeted capture, and Sanger sequencing of patients with undiagnosed developmental disorders, in multiple independent diagnostic or research centers. Phenotypic and mutational comparisons were facilitated through data exchange platforms. Whole-transcriptome sequencing was performed on RNA from patient- and control-derived fibroblasts.RESULTS: We identified heterozygous missense variants in TRAF7 as the cause of a developmental delay-malformation syndrome in 45 patients. Major features include a recognizable facial gestalt (characterized in particular by blepharophimosis), short neck, pectus carinatum, digital deviations, and patent ductus arteriosus. Almost all variants occur in the WD40 repeats and most are recurrent. Several differentially expressed genes were identified in patient fibroblasts.CONCLUSION: We provide the first large-scale analysis of the clinical and mutational spectrum associated with the TRAF7 developmental syndrome, and we shed light on its molecular etiology through transcriptome studies.
- Published
- 2020
8. AHDC1 missense mutations in Xia-Gibbs syndrome
- Author
-
Moez Dawood, James R. Lupski, Emilia K. Bijlsma, Michael M. Khayat, Shoudong Li, Jeffrey W. Innis, Jessica Omark O’Shea, Laura A Cross, Richard A. Gibbs, Jennifer Friedman, Francis H. Sansbury, Kirsty McWalter, Adam W. Hansen, Michael F. Wangler, Yunyun Jiang, Jill A. Rosenfeld, Jianhong Hu, David R. Murdock, Claudia A. L. Ruivenkamp, Jennifer E. Posey, He Li, Qingchang Meng, and Varuna Chander
- Subjects
Genetics ,AHDC1 ,Xia-Gibbs syndrome ,In silico ,missense mutation ,Protein domain ,Biology ,QH426-470 ,medicine.disease ,Phenotype ,Hypotonia ,Article ,de novo mutation ,Neurodevelopmental disorder ,Speech delay ,medicine ,Molecular Medicine ,Missense mutation ,medicine.symptom ,Gene ,Genetics (clinical) - Abstract
Summary Xia-Gibbs syndrome (XGS; MIM: 615829) is a phenotypically heterogeneous neurodevelopmental disorder (NDD) caused by newly arising mutations in the AT-Hook DNA-Binding Motif-Containing 1 (AHDC1) gene that are predicted to lead to truncated AHDC1 protein synthesis. More than 270 individuals have been diagnosed with XGS worldwide. Despite the absence of an independent assay for AHDC1 protein function to corroborate potential functional consequences of rare variant genetic findings, there are also reports of individuals with XGS-like trait manifestations who have de novo missense AHDC1 mutations and who have been provided a molecular diagnosis of the disorder. To investigate a potential contribution of missense mutations to XGS, we mapped the missense mutations from 10 such individuals to the AHDC1 conserved protein domain structure and detailed the observed phenotypes. Five newly identified individuals were ascertained from a local XGS Registry, and an additional five were taken from external reports or databases, including one publication. Where clinical data were available, individuals with missense mutations all displayed phenotypes consistent with those observed in individuals with AHDC1 truncating mutations, including delayed motor milestones, intellectual disability (ID), hypotonia, and speech delay. A subset of the 10 reported missense mutations cluster in two regions of the AHDC1 protein with known conserved domains, likely representing functional motifs. Variants outside the clustered regions score lower for computational prediction of their likely damaging effects. Overall, de novo missense variants in AHDC1 are likely diagnostic of XGS when in silico analysis of their position relative to conserved regions is considered together with disease trait manifestations.
- Published
- 2021
9. Genotype-phenotype analysis of 523 patients by genetics evaluation and clinical exome sequencing
- Author
-
Megan Glassford, Jeffrey W. Innis, Donna M. Martin, Shane C. Quinonez, John A. Bernat, Stephanie L. Bielas, Rachel Fisher, Ayesha Ahmad, Catherine E. Keegan, Mark N. Ziats, Lauren Turner, Nicholas L Harris, Lauren Seemann, Tessa B Marzulla, Jacob D Ogle, Mark C. Hannibal, Natasha E. Weiser, Kristen N. Lee, Joseph E. Jacher, and Bridget C. O’Connor
- Subjects
medicine.medical_specialty ,Referral ,DNA Mutational Analysis ,MEDLINE ,Article ,Genotype phenotype ,Congenital Abnormalities ,03 medical and health sciences ,0302 clinical medicine ,Pediatric genetics ,Predictive Value of Tests ,030225 pediatrics ,Exome Sequencing ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Exome sequencing ,Genetic Association Studies ,Retrospective Studies ,Genetics ,business.industry ,Pathogenic mutation ,Phenotype ,Pediatrics, Perinatology and Child Health ,Mutation ,Medical genetics ,business ,030217 neurology & neurosurgery - Abstract
BACKGROUND As clinical exome sequencing (CES) becomes more common, understanding which patients are most likely to benefit and in what manner is critical for the general pediatrics community to appreciate. METHODS Five hundred and twenty-three patients referred to the Pediatric Genetics clinic at Michigan Medicine were systematically phenotyped by the presence or absence of abnormalities for 13 body/organ systems by a Clinical Genetics team. All patients then underwent CES. RESULTS Overall, 30% of patients who underwent CES had an identified pathogenic mutation. The most common phenotypes were developmental delay (83%), neuromuscular system abnormalities (81%), and multiple congenital anomalies (42%). In all, 67% of patients had a variant of uncertain significance (VUS) or gene of uncertain significance (GUS); 23% had no variants reported. There was a significant difference in the average number of body systems affected among these groups (pathogenic 5.89, VUS 6.0, GUS 6.12, and no variant 4.6; P
- Published
- 2019
10. Expanding the phenotypic spectrum of MBOAT7‐related intellectual disability
- Author
-
Jeffrey W. Innis, Joseph E. Jacher, Nikita Roy, and Mohammad Ghaziuddin
- Subjects
Pediatrics ,medicine.medical_specialty ,Adolescent ,Developmental Disabilities ,Cellular and Molecular Neuroscience ,Seizures ,Intellectual Disability ,Angelman syndrome ,Exome Sequencing ,Intellectual disability ,medicine ,Humans ,Attention deficit hyperactivity disorder ,Exome ,Global developmental delay ,Genetics (clinical) ,business.industry ,Homozygote ,Macrocephaly ,Chromosome Mapping ,Membrane Proteins ,medicine.disease ,Hyperintensity ,Psychiatry and Mental health ,Phenotype ,Attention Deficit Disorder with Hyperactivity ,Autism spectrum disorder ,Mutation ,Female ,medicine.symptom ,business ,Acyltransferases ,Anxiety disorder - Abstract
MBOAT7 gene pathogenic variants are a newly discovered and rare cause for intellectual disability, autism spectrum disorder (ASD), seizures, truncal hypotonia, appendicular hypertonia, and below average head sizes (ranging from -1 to -3 standard deviations). There have been only 16 individuals previously reported who have MBOAT7-related intellectual disability, all of whom were younger than 10 years old and from consanguineous relationships. Thus, there is a lack of phenotypic information for adolescent and adult individuals with this disorder. Medical genetics and psychiatric evaluations in a 14-year-old female patient with a history of global developmental delay, intellectual disability, overgrowth with macrocephaly, metrorrhagia, seizures, basal ganglia hyperintensities, nystagmus, strabismus with amblyopia, ASD, anxiety, attention deficit hyperactivity disorder (ADHD), aggressive outbursts, and hyperphagia included a karyotype, methylation polymerase chain reaction for Prader-Willi/Angelman syndrome, chromosome microarray, and whole exome sequencing (WES), ADOS2, and ADI-R. WES identified a homozygous, likely pathogenic variant in the MBOAT7 gene (c.855-2A>G). This is the oldest known patient with MBOAT7-related intellectual disability, whose unique features compared with previously described individuals include overgrowth with macrocephaly, metrorrhagia, ophthalmological abnormalities, basal ganglia hyperintensities, unspecified anxiety disorder, and ADHD; combined type; and hyperphagia with the absence of appendicular hypertonia and cortical atrophy. More individuals need to be identified in order to delineate the full clinical spectrum of this disorder.
- Published
- 2019
11. Transcriptome analysis of MBD5-associated neurodevelopmental disorder (MAND) neural progenitor cells reveals dysregulation of autism-associated genes
- Author
-
Steven D Klein, Jeffrey W. Innis, Ying Yang, Sureni V. Mullegama, Toshihiko Ezashi, Sarah H. Elsea, Stephen R. Williams, Yuchen Tian, Chad R. Haldeman-Englert, Frank J. Probst, and Julian A. Martinez-Agosto
- Subjects
Autism Spectrum Disorder ,Science ,Induced Pluripotent Stem Cells ,Primary Cell Culture ,Gene Expression ,Stem cells ,Biology ,Article ,Transcriptome ,Craniofacial Abnormalities ,Neurodevelopmental disorder ,Neural Stem Cells ,Intellectual Disability ,medicine ,Genetics ,Humans ,Abnormalities, Multiple ,Epigenetics ,RNA-Seq ,Autistic Disorder ,Induced pluripotent stem cell ,Multidisciplinary ,Gene Expression Profiling ,Cell Differentiation ,Mand ,medicine.disease ,DNA-Binding Proteins ,Phenotype ,Neurology ,Hippo signaling ,Autism spectrum disorder ,Neurodevelopmental Disorders ,Chromosomes, Human, Pair 2 ,Medicine ,Autism ,Chromosome Deletion ,Neuroscience ,Signal Transduction - Abstract
MBD5-associated neurodevelopmental disorder (MAND) is an autism spectrum disorder (ASD) characterized by intellectual disability, motor delay, speech impairment and behavioral problems; however, the biological role of methyl-CpG-binding domain 5, MBD5, in neurodevelopment and ASD remains largely undefined. Hence, we created neural progenitor cells (NPC) derived from individuals with chromosome 2q23.1 deletion and conducted RNA-seq to identify differentially expressed genes (DEGs) and the biological processes and pathways altered in MAND. Primary skin fibroblasts from three unrelated individuals with MAND and four unrelated controls were converted into induced pluripotent stem cell (iPSC) lines, followed by directed differentiation of iPSC to NPC. Transcriptome analysis of MAND NPC revealed 468 DEGs (q
- Published
- 2021
12. Cardiac phenotype in familial partial lipodystrophy
- Author
-
Banu Sarer Yurekli, Nicole M. Bhave, Marwan K. Tayeh, Hakan Oral, Ebru Özpelit, Ilgin Yildirim Simsir, Elif A. Oral, Zeynep Şıklar, José Jalife, Rita Hench, Ramazan Gen, Abdelwahab Jalal Eldin, Andre Monteiro da Rocha, Baris Akinci, Rasimcan Meral, Adam H. Neidert, Nilufer Ozdemir Kutbay, Suleyman Cem Adiyaman, Jeffrey W. Innis, and Ege Üniversitesi
- Subjects
Adult ,medicine.medical_specialty ,lipodystrophy ,Adolescent ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Induced Pluripotent Stem Cells ,LMNA ,030209 endocrinology & metabolism ,Laminopathy ,arrhythmia ,Article ,Afterdepolarization ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Humans ,atrial fibrillation ,Aged ,Retrospective Studies ,conduction disease ,business.industry ,Retrospective cohort study ,Atrial fibrillation ,Middle Aged ,medicine.disease ,Familial partial lipodystrophy ,Lamin Type A ,Lipodystrophy, Familial Partial ,Phenotype ,030220 oncology & carcinogenesis ,Mutation ,Cardiology ,cardiovascular system ,Female ,Lipodystrophy ,Cardiac monitoring ,business - Abstract
Objectives LMNA variants have been previously associated with cardiac abnormalities independent of lipodystrophy. We aimed to assess cardiac impact of familial partial lipodystrophy (FPLD) to understand the role of laminopathy in cardiac manifestations. Study design Retrospective cohort study. Methods Clinical data from 122 patients (age range: 13-77, 101 females) with FPLD were analysed. Mature human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from a patient with an LMNA variant were studied as proof-of-concept for future studies. Results Subjects with LMNA variants had a higher prevalence of overall cardiac events than others. The likelihood of having an arrhythmia was significantly higher in patients with LMNA variants (OR: 3.77, 95% CI: 1.45-9.83). These patients were at higher risk for atrial fibrillation or flutter (OR: 5.78, 95% CI: 1.04-32.16). The time to the first arrhythmia was significantly shorter in the LMNA group, with a higher HR of 3.52 (95% CI: 1.34-9.27). Non-codon 482 LMNA variants were more likely to be associated with cardiac events (vs. 482 LMNA: OR: 4.74, 95% CI: 1.41-15.98 for arrhythmia; OR: 17.67, 95% CI: 2.45-127.68 for atrial fibrillation or flutter; OR: 5.71, 95% CI: 1.37-23.76 for conduction disease). LMNA mutant hiPSC-CMs showed a higher frequency of spontaneous activity and shorter action potential duration. Functional syncytia of hiPSC-CMs displayed several rhythm alterations such as early afterdepolarizations, spontaneous quiescence and spontaneous tachyarrhythmia, and significantly slower recovery in chronotropic changes induced by isoproterenol exposure. Conclusions Our results highlight the need for vigilant cardiac monitoring in FPLD, especially in patients with LMNA variants who have an increased risk of developing cardiac arrhythmias. in addition, hiPSC-CMs can be studied to understand the basic mechanisms for the arrhythmias in patients with lipodystrophy to understand the impact of specific mutations., NIH Clinical and Translational Science AwardsUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USA [UL1TR000433]; Nutrition Obesity Research Centers [P30 DK089503]; NIH institutional grantUnited States Department of Health & Human ServicesNational Institutes of Health (NIH) - USA [DK034933]; White Point Foundation of Turkey; Morton S. and Henrietta K. Sellner Professorship in Human Genetics, Infrastructure and data management support has been provided by the NIH Clinical and Translational Science Awards grant UL1TR000433, the Nutrition Obesity Research Centers grant P30 DK089503, and NIH institutional grant DK034933. Finally, the work was supported by generous gifts to the Lipodystrophy Fund at the University of Michigan made by the Sopha family, and the White Point Foundation of Turkey. JWI acknowledges support from the Morton S. and Henrietta K. Sellner Professorship in Human Genetics.
- Published
- 2021
13. The SRG rat, a Sprague-Dawley Rag2/Il2rg double-knockout validated for human tumor oncology studies
- Author
-
Jaya Sangodkar, Rita Tohme, Sam Moody, Matthew D. Galsky, Analisa DiFeo, Marwan K. Tayeh, B. Mark Evers, Fallon K. Noto, Caitlin M. O’Connor, Bisoye Towobola Adedeji, Ming Tong, Tseten Yeshi Jamling, Kajari Bhattacharya, Lauren Hurst, Goutham Narla, Sudeh Izadmehr, Jyothsna Narla, Eric M. Ostertag, Amit R. Rupani, Christopher B. McClain, Xiaohua Gao, Jack Crawford, Jeffrey W. Innis, Erika Hanson, and Kristin LeSueur
- Subjects
0301 basic medicine ,Oncology ,Lung Neoplasms ,Rodent ,Colorectal cancer ,Physiology ,NK cells ,Lung and Intrathoracic Tumors ,Rats, Sprague-Dawley ,Prostate cancer ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Immune Physiology ,Cellular types ,Medicine and Health Sciences ,Multidisciplinary ,biology ,Prostate Cancer ,Immune cells ,Prostate Diseases ,Animal Models ,medicine.anatomical_structure ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Experimental pathology ,Medicine ,White blood cells ,Interleukin Receptor Common gamma Subunit ,Research Article ,medicine.medical_specialty ,Cell biology ,Blood cells ,Science ,Urology ,Immunology ,Spleen ,Mouse Models ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Malignant Tumors ,RAG2 ,biology.animal ,Internal medicine ,Cell Line, Tumor ,medicine ,Carcinoma ,Animals ,Humans ,Colorectal Cancer ,Biology and life sciences ,Cancers and Neoplasms ,Neoplasms, Experimental ,medicine.disease ,Xenograft Model Antitumor Assays ,Rats ,Non-Small Cell Lung Cancer ,Genitourinary Tract Tumors ,030104 developmental biology ,Animal cells ,Cell culture ,Animal Studies ,Gene Deletion - Abstract
We have created the immunodeficient SRG rat, a Sprague-Dawley Rag2/Il2rg double knockout that lacks mature B cells, T cells, and circulating NK cells. This model has been tested and validated for use in oncology (SRG OncoRat®). The SRG rat demonstrates efficient tumor take rates and growth kinetics with different human cancer cell lines and PDXs. Although multiple immunodeficient rodent strains are available, some important human cancer cell lines exhibit poor tumor growth and high variability in those models. The VCaP prostate cancer model is one such cell line that engrafts unreliably and grows irregularly in existing models but displays over 90% engraftment rate in the SRG rat with uniform growth kinetics. Since rats can support much larger tumors than mice, the SRG rat is an attractive host for PDX establishment. Surgically resected NSCLC tissue from nine patients were implanted in SRG rats, seven of which engrafted and grew for an overall success rate of 78%. These developed into a large tumor volume, over 20,000 mm3 in the first passage, which would provide an ample source of tissue for characterization and/or subsequent passage into NSG mice for drug efficacy studies. Molecular characterization and histological analyses were performed for three PDX lines and showed high concordance between passages 1, 2 and 3 (P1, P2, P3), and the original patient sample. Our data suggest the SRG OncoRat is a valuable tool for establishing PDX banks and thus serves as an alternative to current PDX mouse models hindered by low engraftment rates, slow tumor growth kinetics, and multiple passages to develop adequate tissue banks.
- Published
- 2020
14. Histone H3.3 beyond cancer: Germline mutations in
- Author
-
Laura, Bryant, Dong, Li, Samuel G, Cox, Dylan, Marchione, Evan F, Joiner, Khadija, Wilson, Kevin, Janssen, Pearl, Lee, Michael E, March, Divya, Nair, Elliott, Sherr, Brieana, Fregeau, Klaas J, Wierenga, Alexandrea, Wadley, Grazia M S, Mancini, Nina, Powell-Hamilton, Jiddeke, van de Kamp, Theresa, Grebe, John, Dean, Alison, Ross, Heather P, Crawford, Zoe, Powis, Megan T, Cho, Marcia C, Willing, Linda, Manwaring, Rachel, Schot, Caroline, Nava, Alexandra, Afenjar, Davor, Lessel, Matias, Wagner, Thomas, Klopstock, Juliane, Winkelmann, Claudia B, Catarino, Kyle, Retterer, Jane L, Schuette, Jeffrey W, Innis, Amy, Pizzino, Sabine, Lüttgen, Jonas, Denecke, Tim M, Strom, Kristin G, Monaghan, Zuo-Fei, Yuan, Holly, Dubbs, Renee, Bend, Jennifer A, Lee, Michael J, Lyons, Julia, Hoefele, Roman, Günthner, Heiko, Reutter, Boris, Keren, Kelly, Radtke, Omar, Sherbini, Cameron, Mrokse, Katherine L, Helbig, Sylvie, Odent, Benjamin, Cogne, Sandra, Mercier, Stephane, Bezieau, Thomas, Besnard, Sebastien, Kury, Richard, Redon, Karit, Reinson, Monica H, Wojcik, Katrin, Õunap, Pilvi, Ilves, A Micheil, Innes, Kristin D, Kernohan, Gregory, Costain, M Stephen, Meyn, David, Chitayat, Elaine, Zackai, Anna, Lehman, Hilary, Kitson, Martin G, Martin, Julian A, Martinez-Agosto, Stan F, Nelson, Christina G S, Palmer, Jeanette C, Papp, Neil H, Parker, Janet S, Sinsheimer, Eric, Vilain, Jijun, Wan, Amanda J, Yoon, Allison, Zheng, Elise, Brimble, Giovanni Battista, Ferrero, Francesca Clementina, Radio, Diana, Carli, Sabina, Barresi, Alfredo, Brusco, Marco, Tartaglia, Jennifer Muncy, Thomas, Luis, Umana, Marjan M, Weiss, Garrett, Gotway, K E, Stuurman, Michelle L, Thompson, Kirsty, McWalter, Constance T R M, Stumpel, Servi J C, Stevens, Alexander P A, Stegmann, Kristian, Tveten, Arve, Vøllo, Trine, Prescott, Christina, Fagerberg, Lone Walentin, Laulund, Martin J, Larsen, Melissa, Byler, Robert Roger, Lebel, Anna C, Hurst, Joy, Dean, Samantha A, Schrier Vergano, Jennifer, Norman, Saadet, Mercimek-Andrews, Juanita, Neira, Margot I, Van Allen, Nicola, Longo, Elizabeth, Sellars, Raymond J, Louie, Sara S, Cathey, Elly, Brokamp, Delphine, Heron, Molly, Snyder, Adeline, Vanderver, Celeste, Simon, Xavier, de la Cruz, Natália, Padilla, J Gage, Crump, Wendy, Chung, Benjamin, Garcia, Hakon H, Hakonarson, and Elizabeth J, Bhoj
- Subjects
endocrine system ,SciAdv r-articles ,Forkhead Transcription Factors ,Neurodegenerative Diseases ,Zebrafish Proteins ,Histones ,fluids and secretions ,mental disorders ,Genetics ,Animals ,Humans ,Molecular Biology ,reproductive and urinary physiology ,Germ-Line Mutation ,Zebrafish ,Research Articles ,Research Article - Abstract
Germ line mutations in H3F3A and H3F3B cause a previously unidentified neurodevelopmental syndrome., Although somatic mutations in Histone 3.3 (H3.3) are well-studied drivers of oncogenesis, the role of germline mutations remains unreported. We analyze 46 patients bearing de novo germline mutations in histone 3 family 3A (H3F3A) or H3F3B with progressive neurologic dysfunction and congenital anomalies without malignancies. Molecular modeling of all 37 variants demonstrated clear disruptions in interactions with DNA, other histones, and histone chaperone proteins. Patient histone posttranslational modifications (PTMs) analysis revealed notably aberrant local PTM patterns distinct from the somatic lysine mutations that cause global PTM dysregulation. RNA sequencing on patient cells demonstrated up-regulated gene expression related to mitosis and cell division, and cellular assays confirmed an increased proliferative capacity. A zebrafish model showed craniofacial anomalies and a defect in Foxd3-derived glia. These data suggest that the mechanism of germline mutations are distinct from cancer-associated somatic histone mutations but may converge on control of cell proliferation.
- Published
- 2020
15. Histone H3.3 beyond cancer: Germline mutations in Histone 3 Family 3A and 3B cause a previously unidentified neurodegenerative disorder in 46 patients
- Author
-
Thomas Besnard, Kristian Tveten, Hilary F Kitson, Jennifer A. Lee, Brieana Fregeau, Rachel Schot, Khadija Wilson, Katrin Õunap, Juliane Winkelmann, Anna Lehman, Nicola Longo, Servi J. C. Stevens, Megan T. Cho, Christina G.S. Palmer, Causes Study, Giovanni Battista Ferrero, Joy Dean, Lone W. Laulund, Grazia M.S. Mancini, Matias Wagner, Martin G. Martin, Sabine Lüttgen, Elizabeth J. Bhoj, Amanda J. Yoon, Thomas Klopstock, Janet S. Sinsheimer, Eric Vilain, Sébastien Küry, Francesca Clementina Radio, Jiddeke M. van de Kamp, Cameron Mrokse, Hakon Hakonarson, Samuel G. Cox, Jeanette C. Papp, Margot I. Van Allen, Raymond J. Louie, Constance T. R. M. Stumpel, Evan F. Joiner, Juanita Neira, Arve Vøllo, Amy Pizzino, Kelly Radtke, Celeste Simon, Michelle L. Thompson, Allison Zheng, Omar Sherbini, Marcia C. Willing, Tim M. Strom, Benjamin Garcia, Sara S. Cathey, Theresa A. Grebe, Dong Li, Marjan M. Weiss, Marco Tartaglia, Laura M Bryant, Sandra Mercier, Katherine L. Helbig, Martin Jakob Larsen, Ddd Study, Alexandrea Wadley, Alexander P.A. Stegmann, Sabina Barresi, A. Micheil Innes, Elaine H. Zackai, Gregory Costain, Davor Lessel, Molly Snyder, Heather P. Crawford, Richard Redon, Pearl Lee, Melissa Byler, Holly Dubbs, J. Gage Crump, K. E. Stuurman, Boris Keren, Stéphane Bézieau, Stan F. Nelson, Kristin G. Monaghan, Michael J. Lyons, Jeffrey W. Innis, Anna C.E. Hurst, Elizabeth A. Sellars, Samantha A. Schrier Vergano, Saadet Mercimek-Andrews, Monica H. Wojcik, Alison Ross, Heiko Reutter, Zuo-Fei Yuan, Dylan M. Marchione, Renee Bend, Diana Carli, Zöe Powis, Neil H. Parker, Jennifer Muncy Thomas, Luis A. Umaña, Adeline Vanderver, Julia Hoefele, Linda Manwaring, Christina Fagerberg, Elly Brokamp, M. Stephen Meyn, Pilvi Ilves, Xavier de la Cruz, Nina Powell-Hamilton, Caroline Nava, Garrett Gotway, Karit Reinson, Kristin D. Kernohan, Jennifer Norman, Alexandra Afenjar, Benjamin Cogné, Delphine Héron, Roman Günthner, Alfredo Brusco, John Dean, Kevin A. Janssen, Robert Roger Lebel, Divya Nair, Jijun Wan, Julian A. Martinez-Agosto, Elliott H. Sherr, Kyle Retterer, Claudia B. Catarino, Michael E. March, Natalia Padilla, Elise Brimble, Sylvie Odent, Jane L. Schuette, David Chitayat, Klaas J. Wierenga, Kirsty McWalter, Trine Prescott, Jonas Denecke, Wendy K. Chung, Human genetics, Amsterdam Neuroscience - Complex Trait Genetics, Amsterdam Gastroenterology Endocrinology Metabolism, Klinische Genetica, MUMC+: DA KG Polikliniek (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Lab Centraal Lab (9), and Clinical Genetics
- Subjects
metabolism [Zebrafish Proteins] ,RESIDUE ,metabolism [Histones] ,GENES ,Somatic cell ,CODE ,cancer mutation ,histone ,Biology ,VARIANTS ,medicine.disease_cause ,progressive neurologic dysfunction ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Germline mutation ,SDG 3 - Good Health and Well-being ,histone, neurodevelopmental disorder, progressive neurologic dysfunction, congenital anomalies, cancer mutation ,medicine ,Animals ,Humans ,H3-3A protein, human ,metabolism [Zebrafish] ,TRANSCRIPTION ,PHOSPHORYLATION ,Gene ,Zebrafish ,Germ-Line Mutation ,030304 developmental biology ,Genetics ,genetics [Zebrafish] ,0303 health sciences ,Multidisciplinary ,foxd3 protein, zebrafish ,congenital anomalies ,Forkhead Transcription Factors ,Zebrafish Proteins ,biology.organism_classification ,genetics [Histones] ,neurodevelopmental disorder ,H3F3B ,Histone ,genetics [Forkhead Transcription Factors] ,genetics [Neurodegenerative Diseases] ,biology.protein ,ddc:500 ,Carcinogenesis ,030217 neurology & neurosurgery - Abstract
Germ line mutations in H3F3A and H3F3B cause a previously unidentified neurodevelopmental syndrome. Although somatic mutations in Histone 3.3 (H3.3) are well-studied drivers of oncogenesis, the role of germline mutations remains unreported. We analyze 46 patients bearing de novo germline mutations in histone 3 family 3A (H3F3A) or H3F3B with progressive neurologic dysfunction and congenital anomalies without malignancies. Molecular modeling of all 37 variants demonstrated clear disruptions in interactions with DNA, other histones, and histone chaperone proteins. Patient histone posttranslational modifications (PTMs) analysis revealed notably aberrant local PTM patterns distinct from the somatic lysine mutations that cause global PTM dysregulation. RNA sequencing on patient cells demonstrated up-regulated gene expression related to mitosis and cell division, and cellular assays confirmed an increased proliferative capacity. A zebrafish model showed craniofacial anomalies and a defect in Foxd3-derived glia. These data suggest that the mechanism of germline mutations are distinct from cancer-associated somatic histone mutations but may converge on control of cell proliferation
- Published
- 2020
16. Interstitial microdeletion of the 1p34.3p34.2 region
- Author
-
Jeffrey W. Innis and Joseph E. Jacher
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Microarray ,030105 genetics & heredity ,chromosome microarray ,Clinical Reports ,Aberrant subclavian artery ,03 medical and health sciences ,Genetics ,Medicine ,Global developmental delay ,Molecular Biology ,Kyphoscoliosis ,Genetics (clinical) ,Clinical Report ,SNIP1 ,medicine.diagnostic_test ,business.industry ,Chromosome ,microdeletions ,medicine.disease ,Real-time polymerase chain reaction ,1p34.3 ,1p34.2 ,business ,Haploinsufficiency ,Fluorescence in situ hybridization - Abstract
Background Interstitial microdeletions of chromosome 1p34.3p34.2 are rare, but are continuing to be identified by the use of chromosome microarray. There have been fewer than 10 individuals identified who have deletions of the 1p34.3p34.2 region; all of these previously described individuals have deletions of the AGO1, AGO3, GRIK3, SLC2A1, or RIMS3 genes. Haploinsufficiency of these genes has been associated with neurodevelopmental delays. Methods Chromosome microarray, quantitative PCR, and fluorescence in situ hybridization were performed with DNA extracted from peripheral blood. Results Chromosome microarray identified a 2.3 Mb 1p34.3p34.2 one copy deletion in our patient with global developmental delay, mild intellectual disability, delayed bone age, bilateral vesicoureteral reflux, vocal cord paralysis, right aberrant subclavian artery, kyphoscoliosis, bilateral metatarsus adductus, and valgus knee deformity. This deletion was confirmed by quantitative PCR and does not include the AGO1, AGO3, GRIK3, SLC2A1, or RIMS3 genes. Subsequent FISH testing of the parents was negative. Conclusion Haploinsufficiency of the 1p34.3p34.2 region, including the SNIP1 gene and excluding the five genes listed above, is responsible for the neurocognitive delays and other symptoms as identified in our patient.
- Published
- 2018
17. Compound heterozygosity for loss-of-function FARSB variants in a patient with classic features of recessive aminoacyl-tRNA synthetase-related disease
- Author
-
Stephanie N. Oprescu, Anthony Antonellis, Jeffrey W. Innis, Andrea Amalfitano, Laurie B. Griffin, and Amer Heider
- Subjects
Male ,0301 basic medicine ,Biology ,Compound heterozygosity ,Article ,Amino Acyl-tRNA Synthetases ,03 medical and health sciences ,chemistry.chemical_compound ,Loss of Function Mutation ,Locus heterogeneity ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Gene ,Genetics (clinical) ,Loss function ,chemistry.chemical_classification ,Aminoacyl tRNA synthetase ,medicine.disease ,Phenotype ,Amino acid ,030104 developmental biology ,Gene Expression Regulation ,chemistry ,Transfer RNA ,Phenylalanine-tRNA Ligase - Abstract
Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed enzymes that ligate amino acids onto tRNA molecules. Genes encoding ARSs have been implicated in phenotypically diverse dominant and recessive human diseases. The charging of tRNA(PHE) with phenylalanine is performed by a tetrameric enzyme that contains two alpha (FARSA) and two beta (FARSB) subunits. To date, mutations in the genes encoding these subunits (FARSA and FARSB) have not been implicated in any human disease. Here, we describe a patient with a severe, lethal, multisystem, developmental phenotype who was compound heterozygous for FARSB variants: p.Thr256Met and p.His496Lysfs*14. Expression studies using fibroblasts isolated from the proband revealed a severe depletion of both FARSB and FARSA protein levels. These data indicate that the FARSB variants destabilize total phenylalanyl-tRNA synthetase levels, thus causing a loss-of-function effect. Importantly, our patient shows strong phenotypic overlap with patients that have recessive diseases associated with other ARS loci; these observations strongly support the pathogenicity of the identified FARSB variants and are consistent with the essential function of phenylalanyl-tRNA synthetase in human cells. In sum, our clinical, genetic, and functional analyses revealed the first FARSB variants associated with a human disease phenotype and expand the locus heterogeneity of ARS-related human disease.
- Published
- 2018
18. Untangling the Heterogeneity of Acquired Generalized Lipodystrophy
- Author
-
Umit Cavdar, Jeffrey W. Innis, Marwan K. Tayeh, Elif A. Oral, David Frame, David A. Fox, Hari S. Conjeevaram, Maria Cristina Foss de Freitas, Baris Akinci, Robert J. Fontana, and Kelly Walkovich
- Subjects
Novel Insights From the Clinic into the Development of Metabolic Disease: Case Reports ,Adipose Tissue, Appetite, and Obesity ,Computer science ,Endocrinology, Diabetes and Metabolism ,medicine ,Computational biology ,Acquired generalized lipodystrophy ,medicine.disease ,AcademicSubjects/MED00250 - Abstract
Acquired generalized lipodystrophy (AGL) is characterized by extensive adipose tissue loss, subsequent development of metabolic disease with severe insulin resistance and hypertriglyceridemia and a varying spectrum of autoimmune or immune-dysregulatory features. We recently evaluated twelve patients (8 females, 4 males; age range: 14 - 54 years). The reported onset of fat loss varies from age 2 to 31 years of age but most patients presenting between age 3 and 19 years. The age of diagnosis varies from 4 to 53 years. Clinical panniculitis was reported in 4 patients. Although adipose tissue loss occurred in a generalized fashion, several patients had measured body fat as high as 35 percent on DEXA scans with leptinemia also covering a spectrum from undetectable levels to as high as 6.2 ng/ml. All patients had insulin resistance. Eight patients had diabetes (4 had type 1 diabetes). Dyslipidemia was diagnosed in 11 patients. Hepatic steatosis and/or elevated liver function tests were detected in all subjects. The liver disease was varied and had distinctive characteristics of episodic increases in liver tests in 4 with development of portal hypertension and splenomegaly. These patients had periportal lymphocytic and eosinophilic infiltration on liver biopsies when undertaken during transaminitis and developed portal hypertension, splenomegaly and biliary obstruction while preserving synthetic liver function over time, suggesting that their clinical course may be consistent with nodular regenerative hyperplasia (NRH). Patients also developed a wide range of immune-dysregulatory features including IgA deficiency, common variable immunodeficiency, autoimmune hives, angioedema, eczematous dermatitis, autoimmune hypothyroidism, Graves’ disease, atrophic gastritis, juvenile dermatomyositis, rheumatoid arthritis, polyarthritis, immune thrombocytopenia, cutaneous morphea, lentigo, peripheral T cell lymphoma, graft versus host disease, hemophagocytic syndrome, interstitial lung disease, autoimmune hemolytic anemia, autoimmune colitis, Crohn disease, and periodic fevers. Low complement 4 levels were detected in 5 patients, and 3 had low complement 3 levels. Genetic workup revealed CTLA-4 haploinsufficiency in one case and variants of uncertain significance in P4HA3, TTN, TNFRSF13B, and NLRP3 genes in several others. One distinctive patient was heterozygous for a pathogenic LRBA variant. The exact etiology of AGL is unknown and heterogeneity creates a diagnostic challenge. While panniculitis is a distinct initial presentation in some cases, immune dysregulation affecting multiple organs with accompanying NRH may constitute a new subgroup of AGL. Immune check-point perturbation via gremlin mutation may also lead to AGL. Collective review of cases with predetermined clinical and laboratory evaluation criteria may be helpful to describe subgroups of AGL.
- Published
- 2021
19. Metreleptin therapy for nonalcoholic steatohepatitis: Open-label therapy interventions in two different clinical settings
- Author
-
Nazanene H. Esfandiari, Charles Burant, Angela Subauste, Christos S. Mantzoros, Nevin Ajluni, Adam H. Neidert, Baris Akinci, Rasimcan Meral, Jeffrey W. Innis, Rita Hench, Thomas L. Chenevert, Akin Eraslan, Amit R. Rupani, Hero K. Hussain, Diana Rus, Hari S. Conjeevaram, Elif A. Oral, Marwan K. Tayeh, and Barbara J. McKenna
- Subjects
Leptin ,Male ,medicine.medical_specialty ,Leptin Deficiency ,Lipodystrophy ,medicine.diagnostic_test ,business.industry ,Partial Lipodystrophy ,General Medicine ,medicine.disease ,Gastroenterology ,Article ,Metreleptin ,chemistry.chemical_compound ,Insulin resistance ,chemistry ,Non-alcoholic Fatty Liver Disease ,Liver biopsy ,Internal medicine ,medicine ,Humans ,Steatosis ,business - Abstract
BACKGROUND. Recombinant leptin therapy reverses nonalcoholic steatohepatitis (NASH) in leptin-deficient lipodystrophy. We inquired if leptin therapy would improve nonalcoholic steatohepatitis in more common forms of this heterogeneous condition. METHODS. Nine male patients with relative leptin deficiency (level < 25th percentile of body mass index- and gender-matched United States population) and biopsy-proven NASH and 23 patients with partial lipodystrophy and NASH were recruited for two distinctive open-label trials. Participants received leptin therapy in the form of metreleptin for 12 months. The primary endpoints were the global nonalcoholic steatohepatitis scores from paired liver biopsies scored blindly. FINDINGS. Of 9 participants recruited in the relative leptin deficiency treatment study, 7 completed 12-months of therapy. Mean global NASH scores were reduced from 8 ± 3 to 5 ± 2 (range: from 1 to 6, P = 0.004). In the partial lipodystrophy study, 19 of 22 subjects completed 12 months of treatment, and 18 completed a second liver biopsy. Global NASH scores also reduced significantly from 6 ± 2 to 5 ± 2 (range: from −2 to 4, P = 0.008). In both studies, the predominant changes were in steatosis and hepatic injury scores. CONCLUSION. Our findings show that patients with NASH associated with both relative leptin deficiency and partial lipodystrophy have reductions in hepatic steatosis and injury in response to exogenous leptin therapy. Moreover, leptin deficiency may have regulatory effects in mediating fat deposition and ensuing injury in the liver. TRIAL REGISTRATION. ClinicalTrials.gov NCT00596934 and NCT01679197.
- Published
- 2021
20. Integrative Clinical Genomics of Metastatic Cancer
- Author
-
Jessica Everett, Xuhong Cao, Jeffrey W. Innis, Rajen Mody, Francis P. Worden, Yi-Mi Wu, David Smith, Javed Siddiqui, Joseph Vijai, Erica Rabban, Moshe Talpaz, Kenneth Offit, Elena M. Stoffel, Daniel F. Hayes, J. Scott Roberts, Ajjai Alva, Chandan Kumar-Sinha, Erin F. Cobain, Robert J. Lonigro, Nithya Ramnath, Marcin Cieślik, Laurence H. Baker, David R. Lucas, Ann F. Schott, Dan R. Robinson, Mark M. Zalupski, Scott A. Tomlins, Victoria M. Raymond, Pankaj Vats, Arul M. Chinnaiyan, Scott M. Schuetze, Lakshmi P. Kunju, and Rashmi Chugh
- Subjects
Adult ,Male ,0301 basic medicine ,DNA Repair ,Class I Phosphatidylinositol 3-Kinases ,Genetics, Medical ,Ubiquitin-Protein Ligases ,precision medicine ,Genomics ,clinical sequencing ,transcriptome sequencing ,Bioinformatics ,DNA sequencing ,Article ,Metastasis ,whole exome sequencing ,Transcriptome ,03 medical and health sciences ,Germline mutation ,CDKN2A ,Neoplasms ,Exome Sequencing ,Tumor Microenvironment ,medicine ,Cyclin-Dependent Kinase Inhibitor p18 ,PTEN ,Humans ,Neoplasm Metastasis ,Cyclin-Dependent Kinase Inhibitor p16 ,Germ-Line Mutation ,Exome sequencing ,metastatic cancers ,Multidisciplinary ,biology ,PTEN Phosphohydrolase ,High-Throughput Nucleotide Sequencing ,medicine.disease ,3. Good health ,Retinoblastoma Binding Proteins ,030104 developmental biology ,biology.protein ,Female ,Tumor Suppressor Protein p53 - Abstract
SUMMARY Metastasis is the primary cause of cancer-related deaths. While The Cancer Genome Atlas (TCGA) has sequenced primary tumor types obtained from surgical resections, much less comprehensive molecular analysis is available from clinically acquired metastatic cancers. Here, we perform whole exome and transcriptome sequencing of 500 adult patients with metastatic solid tumors of diverse lineage and biopsy site. The most prevalent genes somatically altered in metastatic cancer included TP53, CDKN2A, PTEN, PIK3CA, and RB1. Putative pathogenic germline variants were present in 12.2% of cases of which 75% were related to defects in DNA repair. RNA sequencing complemented DNA sequencing for the identification of gene fusions, pathway activation, and immune profiling. Integrative sequence analysis provides a clinically relevant, multi-dimensional view of the complex molecular landscape and microenvironment of metastatic cancers.
- Published
- 2017
21. Natural history of ROHHAD syndrome: development of severe insulin resistance and fatty liver disease over time
- Author
-
Rasimcan Meral, Kelly Walkovich, Alison H. Affinati, Neehar D. Parikh, Mehmet Selman Ontan, Laura Butz, Jeffrey W. Innis, Dilara Tombayoglu, Abdel Wahab Jalal Eldin, Elif A. Oral, and Maria Westerhoff
- Subjects
medicine.medical_specialty ,Case Report ,Gastroenterology ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,Growth hormone deficiency ,Liver disease ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,ROHHAD ,medicine ,Obesity ,lcsh:RC648-665 ,medicine.diagnostic_test ,GLP-1 agonists ,business.industry ,Fatty liver ,General Medicine ,medicine.disease ,Atypical diabetes ,Hypoventilation ,Liver biopsy ,medicine.symptom ,business - Abstract
Background:Rapid-onset obesity with hypothalamic dysfunction, hypoventilation, and autonomic dysregulation (ROHHAD) is a rare syndrome with unknown etiology. Metabolic abnormalities are not known to be part of the syndrome. We present one of the oldest cases reported in the literature, who developed severe metabolic abnormalities and hepatic disease suggesting that these features may be part of the syndrome.Case presentation:A 27-year-old woman, diagnosed with ROHHAD syndrome at age 15, who previously developed diabetes insipidus, growth hormone deficiency, hyperprolactinemia, and hypothyroidism in her first decade of life. This was followed by insulin resistance, NAFLD, liver fibrosis, and splenomegaly before age 14 years. Her regimen included a short course of growth hormone, and cyclic estrogen and progesterone. Her metabolic deterioration continued despite treatment with metformin. Interestingly, she had a favorable response to liraglutide therapy despite having a centrally mediated cause for her obesity. At age 26, a 1.6 cm lesion was found incidentally in her liver. Liver biopsy showed hepatocellular carcinoma which was successfully treated with radiofrequency ablation.Conclusion:Metabolic abnormalities, Insulin resistance and fatty liver disease are potentially part of the ROHHAD syndrome that may develop over time. GLP1 agonists were reasonably effective to treat insulin resistance and hyperphagia. Patients with ROHHAD may benefit from close follow up in regards to liver disease.Keywords:Atypical diabetes; GLP-1 agonists; Insulin resistance; Obesity; ROHHAD.
- Published
- 2019
22. Nine de novo duplications affecting both maternal and paternal chromosomes and an inherited 15q11.2 deletion, in a patient with developmental delay
- Author
-
Marwan K. Tayeh, Tracy Rocco, Jeffrey W. Innis, Leslie Ernst, Thomas W. Glover, and Todd Ackley
- Subjects
Genetics ,developmental delay ,Zygote ,Chromosomal microduplication ,multiple congenital anomalies ,Period (gene) ,General Medicine ,Case Reports ,Biology ,Bioinformatics ,de novo copy number variations - Abstract
Key Clinical Message A patient with developmental delay and nine, de novo, tandem duplications affecting eight different chromosomes that arose on both maternal and paternal chromosomes indicating a vulnerable zygotic or early postzygotic period of development for these errors, potentially affected by genetic and nongenetic factors.
- Published
- 2015
23. A Novel Generalized Lipodystrophy-Associated Progeroid Syndrome Due to Recurrent Heterozygous LMNA p.T10I Mutation
- Author
-
Yulia Tikhonovich, Masako Ueda, Nivedita Patni, Elif A. Oral, Elaine Cochran, Joseph Peeden, Priti Lal, Iram Hussain, Abhimanyu Garg, Cynthia Melissa Valerio, Daniel J. Rader, Ekaterina Sorkina, Anders R.L. Meyer, Jeffrey W. Innis, Beverley Adams-Huet, Marwan K. Tayeh, Milena Gurgel Teles, Sarah Stender, Amelio F. Godoy-Matos, Rebecca J. Brown, Elisabeth Klouda, Anatoly Tiulpakov, and Robert A. Hegele
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,030209 endocrinology & metabolism ,Gene mutation ,Biochemistry ,Gastroenterology ,LMNA ,03 medical and health sciences ,Metreleptin ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Absorptiometry, Photon ,Progeria ,Lipodystrophy, Congenital Generalized ,Internal medicine ,Medicine ,Humans ,Child ,Acanthosis nigricans ,Clinical Research Articles ,integumentary system ,Anthropometry ,business.industry ,Generalized lipodystrophy ,Myocardium ,Biochemistry (medical) ,nutritional and metabolic diseases ,Middle Aged ,medicine.disease ,Lamin Type A ,Magnetic Resonance Imaging ,Mandibuloacral dysplasia ,030104 developmental biology ,Phenotype ,chemistry ,Mutation ,Female ,Lipodystrophy ,business - Abstract
Background Lamin A/C (LMNA) gene mutations cause a heterogeneous group of progeroid disorders, including Hutchinson-Gilford progeria syndrome, mandibuloacral dysplasia, and atypical progeroid syndrome (APS). Five of the 31 previously reported patients with APS harbored a recurrent de novo heterozygous LMNA p.T10I mutation. All five had generalized lipodystrophy, as well as similar metabolic and clinical features, suggesting a distinct progeroid syndrome. Methods We report nine new patients and follow-up of two previously reported patients with the heterozygous LMNA p.T10I mutation and compare their clinical and metabolic features with other patients with APS. Results Compared with other patients with APS, those with the heterozygous LMNA p.T10I mutation were younger in age but had increased prevalence of generalized lipodystrophy, diabetes mellitus, acanthosis nigricans, hypertriglyceridemia, and hepatomegaly, together with higher fasting serum insulin and triglyceride levels and lower serum leptin and high-density lipoprotein cholesterol levels. Prominent clinical features included mottled skin pigmentation, joint contractures, and cardiomyopathy resulting in cardiac transplants in three patients at ages 13, 33, and 47 years. Seven patients received metreleptin therapy for 0.5 to 16 years with all, except one noncompliant patient, showing marked improvement in metabolic complications. Conclusions Patients with the heterozygous LMNA p.T10I mutation have distinct clinical features and significantly worse metabolic complications compared with other patients with APS as well as patients with Hutchinson-Gilford progeria syndrome. We propose that they be recognized as having generalized lipodystrophy-associated progeroid syndrome. Patients with generalized lipodystrophy-associated progeroid syndrome should undergo careful multisystem assessment at onset and yearly metabolic and cardiac evaluation, as hyperglycemia, hypertriglyceridemia, hepatic steatosis, and cardiomyopathy are the major contributors to morbidity and mortality.
- Published
- 2017
24. Chromosomal Imbalances in Patients with Congenital Cardiac Defects: A Meta-analysis Reveals Novel Potential Critical Regions Involved in Heart Development
- Author
-
Judith A. Goodship, Aoy Tomita-Mitchell, Marwan K. Tayeh, Jeffrey W. Innis, Thor Thorsson, Mark W. Russell, Ana Töpf, Rachel Soemedi, Todd Ackley, Bernard Keavney, Jill A. Rosenfeld, William W. Russell, Nour El-Kashlan, Sarah B. Geisler, and Jonathan Levine
- Subjects
Genetics ,Heart development ,business.industry ,General Medicine ,Bioinformatics ,Critical regions ,Meta-analysis ,Pediatrics, Perinatology and Child Health ,Cardiac defects ,Medicine ,Radiology, Nuclear Medicine and imaging ,Surgery ,In patient ,Cardiology and Cardiovascular Medicine ,business ,Gene - Abstract
Congenital cardiac defects (CCD) represent the most common group of birth defects, estimated at 8 per 1000 births. Genetic characterization of patients and families with cardiac defects has identified a number of genes required for heart development. Yet, mutations affecting known genes still
- Published
- 2014
25. Human HOX gene disorders
- Author
-
Shane C. Quinonez and Jeffrey W. Innis
- Subjects
animal structures ,Genotype ,Endocrinology, Diabetes and Metabolism ,Penetrance ,Biology ,Biochemistry ,Congenital Abnormalities ,Mice ,Endocrinology ,Germline mutation ,Genetics ,Animals ,Humans ,Inheritance Patterns ,Exome ,Allele ,Hox gene ,Molecular Biology ,Gene ,Germ-Line Mutation ,Genome, Human ,Genes, Homeobox ,Genetic Diseases, Inborn ,Genetic Variation ,Syndrome ,Phenotype ,Disease Models, Animal ,Multigene Family ,Vertebrates - Abstract
The Hox genes are an evolutionarily conserved family of genes, which encode a class of important transcription factors that function in numerous developmental processes. Following their initial discovery, a substantial amount of information has been gained regarding the roles Hox genes play in various physiologic and pathologic processes. These processes range from a central role in anterior-posterior patterning of the developing embryo to roles in oncogenesis that are yet to be fully elucidated. In vertebrates there are a total of 39 Hox genes divided into 4 separate clusters. Of these, mutations in 10 Hox genes have been found to cause human disorders with significant variation in their inheritance patterns, penetrance, expressivity and mechanism of pathogenesis. This review aims to describe the various phenotypes caused by germline mutation in these 10 Hox genes that cause a human phenotype, with specific emphasis paid to the genotypic and phenotypic differences between allelic disorders. As clinical whole exome and genome sequencing is increasingly utilized in the future, we predict that additional Hox gene mutations will likely be identified to cause distinct human phenotypes. As the known human phenotypes closely resemble gene-specific murine models, we also review the homozygous loss-of-function mouse phenotypes for the 29 Hox genes without a known human disease. This review will aid clinicians in identifying and caring for patients affected with a known Hox gene disorder and help recognize the potential for novel mutations in patients with phenotypes informed by mouse knockout studies.
- Published
- 2014
26. ARHGDIA mutations cause nephrotic syndrome via defective RHO GTPase signaling
- Author
-
Virginia Vega-Warner, Toby W. Hurd, Friedhelm Hildebrandt, Jeffrey W. Innis, Neveen A. Soliman, Weibin Zhou, Christian Faul, Yaacov Frishberg, Bodo B. Beck, Olivier Gribouval, Katrina A. Diaz, Gil Chernin, Zhe Han, Shawn Levy, Edgar A. Otto, Dominik S. Schoeb, Heike Goebel, Heon Yung Gee, Joseph Washburn, Pawaree Saisawat, Sivakumar Natarajan, Lutz T. Weber, Hanan M. Fathy, Shazia Ashraf, Bugsu Ovunc, Humphrey Fang, Roger C. Wiggins, Svjetlana Lovric, Corinne Antignac, and Julia Hoefele
- Subjects
rac1 GTP-Binding Protein ,Nephrotic Syndrome ,RHOA ,Mutation, Missense ,RAC1 ,CDC42 ,GTPase ,medicine.disease_cause ,Consanguinity ,Cell Movement ,Protein Interaction Mapping ,medicine ,Animals ,Humans ,Missense mutation ,cdc42 GTP-Binding Protein ,Cells, Cultured ,Genetic Association Studies ,Zebrafish ,rho Guanine Nucleotide Dissociation Inhibitor alpha ,Mutation ,Base Sequence ,biology ,Podocytes ,Homozygote ,Chromosome Mapping ,Sequence Analysis, DNA ,General Medicine ,Phenotype ,Protein Transport ,Cdc42 GTP-Binding Protein ,Case-Control Studies ,Gene Knockdown Techniques ,biology.protein ,Cancer research ,rhoA GTP-Binding Protein ,Protein Binding ,Signal Transduction ,Research Article - Abstract
Nephrotic syndrome (NS) is divided into steroid-sensitive (SSNS) and -resistant (SRNS) variants. SRNS causes end-stage kidney disease, which cannot be cured. While the disease mechanisms of NS are not well understood, genetic mapping studies suggest a multitude of unknown single-gene causes. We combined homozygosity mapping with whole-exome resequencing and identified an ARHGDIA mutation that causes SRNS. We demonstrated that ARHGDIA is in a complex with RHO GTPases and is prominently expressed in podocytes of rat glomeruli. ARHGDIA mutations (R120X and G173V) from individuals with SRNS abrogated interaction with RHO GTPases and increased active GTP-bound RAC1 and CDC42, but not RHOA, indicating that RAC1 and CDC42 are more relevant to the pathogenesis of this SRNS variant than RHOA. Moreover, the mutations enhanced migration of cultured human podocytes; however, enhanced migration was reversed by treatment with RAC1 inhibitors. The nephrotic phenotype was recapitulated in arhgdia-deficient zebrafish. RAC1 inhibitors were partially effective in ameliorating arhgdia-associated defects. These findings identify a single-gene cause of NS and reveal that RHO GTPase signaling is a pathogenic mediator of SRNS.
- Published
- 2013
27. Spectrum of disease associated with partial lipodystrophy: lessons from a trial cohort
- Author
-
Peedikayil E. Thomas, Thomas L. Chenevert, Amit R. Rupani, Frank DiPaola, Colleen Buggs-Saxton, M. Bishr Omary, Adam H. Neidert, Eric D. Buras, Rasimcan Meral, Graham F. Brady, Jeffrey W. Innis, Marwan K. Tayeh, Nevin Ajluni, Elif A. Oral, Barbara J. McKenna, Hari S. Conjeevaram, and Jeffrey F. Horowitz
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Lipodystrophy ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Gastroenterology ,Article ,LMNA ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Atypia ,Humans ,Nuclear atypia ,Child ,medicine.diagnostic_test ,business.industry ,Fatty liver ,Partial Lipodystrophy ,Magnetic resonance imaging ,Middle Aged ,medicine.disease ,Lipodystrophy, Familial Partial ,030104 developmental biology ,Cross-Sectional Studies ,Mood disorders ,Physical therapy ,Body Composition ,Biomarker (medicine) ,Female ,business - Abstract
SummaryContext Partial lipodystrophy (PL) is associated with metabolic co-morbidities but may go undiagnosed as the disease spectrum is not fully described. Objective The objective of the study was to define disease spectrum in PL using genetic, clinical (historical, morphometric) and laboratory characteristics. Design Cross-sectional evaluation. Participants Twenty-three patients (22 with familial, one acquired, 78·3% female, aged 12–64 years) with PL and non-alcoholic fatty liver disease (NAFLD). Measurements Genetic, clinical and laboratory characteristics, body composition indices, liver fat content by magnetic resonance imaging (MRI), histopathological and immunofluorescence examinations of liver biopsies. Results Seven patients displayed heterozygous pathogenic variants in LMNA. Two related patients had a heterozygous, likely pathogenic novel variant of POLD1 (NM002691·3: c.3199 G>A; p.E1067K). Most patients had high ratios (>1·5) of percentage fat trunk to percentage fat legs (FMR) when compared to reference normals. Liver fat quantified using MR Dixon method was high (11·3 ± 6·3%) and correlated positively with haemoglobin A1c and triglycerides while leg fat by dual-energy X-ray absorptiometry (DEXA) correlated negatively with triglycerides. In addition to known metabolic comorbidities; chronic pain (78·3%), hypertension (56·5%) and mood disorders (52·2%) were highly prevalent. Mean NAFLD Activity Score (NAS) was 5 ± 1 and 78·3% had fibrosis. LMNA-immunofluorescence staining from select patients (including one with the novel POLD1 variant) showed a high degree of nuclear atypia and disorganization. Conclusions Partial lipodystrophy is a complex multi-system disorder. Metabolic parameters correlate negatively with extremity fat and positively with liver fat. DEXA-based FMR may prove useful as a diagnostic tool. Nuclear disorganization and atypia may be a common biomarker even in the absence of pathogenic variants in LMNA.
- Published
- 2016
28. HOXA13 Hand-Foot-Genital Syndrome and Guttmacher Syndrome
- Author
-
Jeffrey W. Innis
- Subjects
medicine.medical_specialty ,Guttmacher syndrome ,business.industry ,medicine ,Hand-Foot-Genital Syndrome ,medicine.disease ,business ,Dermatology - Published
- 2016
29. De novo missense variants in PPP2R5D are associated with intellectual disability, macrocephaly, hypotonia, and autism
- Author
-
Jason Carmichael, Jeffrey W. Innis, Donald S. Petrey, Linshan Shang, Katrina Haude, Jane L. Schuette, Megan T. Cho, Lindsay B. Henderson, Wendy K. Chung, Kyle Retterer, Margaret Pearson, Chin-To Fong, Leandra Folk, Julie Lundberg, Shailesh Asaikar, Kristin G. Monaghan, Natasha Shur, Yvonne W. Wu, and Natalie Hauser
- Subjects
0301 basic medicine ,Male ,Autism Spectrum Disorder ,Autism ,DNA Mutational Analysis ,Intellectual disabilities ,Missense mutation ,2.1 Biological and endogenous factors ,Protein Phosphatase 2 ,Aetiology ,Child ,Genetics (clinical) ,Genetics ,Pediatric ,Single Nucleotide ,Hypotonia ,PPP2R5D ,Mental Health ,Protein phosphatase ,Child, Preschool ,Whole-exome sequencing ,Muscle Hypotonia ,Female ,Cognitive Sciences ,medicine.symptom ,Adolescent ,Protein subunit ,Intellectual and Developmental Disabilities (IDD) ,1.1 Normal biological development and functioning ,Phosphatase ,Mutation, Missense ,Biology ,Polymorphism, Single Nucleotide ,Article ,Chromatin remodeling ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Underpinning research ,Intellectual Disability ,medicine ,Humans ,Genetic Predisposition to Disease ,Autistic Disorder ,Polymorphism ,Preschool ,Protein kinase B ,GSK3B ,Genetic Association Studies ,De novo mutations ,Neurology & Neurosurgery ,Neurosciences ,Infant ,Protein phosphatase 2 ,Megalencephaly ,Brain Disorders ,030104 developmental biology ,Mutation ,Missense - Abstract
Protein phosphatase 2A (PP2A) is a heterotrimeric protein serine/threonine phosphatase and is involved in a broad range of cellular processes. PPP2R5D is a regulatory B subunit of PP2A and plays an important role in regulating key neuronal and developmental regulation processes such as PI3K/AKT and glycogen synthase kinase 3 beta (GSK3β)-mediated cell growth, chromatin remodeling, and gene transcriptional regulation. Using whole-exome sequencing (WES), we identified four de novo variants in PPP2R5D in a total of seven unrelated individuals with intellectual disability (ID) and other shared clinical characteristics, including autism spectrum disorder, macrocephaly, hypotonia, seizures, and dysmorphic features. Among the four variants, two have been previously reported and two are novel. All four amino acids are highly conserved among the PP2A subunit family, and all change a negatively charged acidic glutamic acid (E) to a positively charged basic lysine (K) and are predicted to disrupt the PP2A subunit binding and impair the dephosphorylation capacity. Our data provides further support for PPP2R5D as a genetic cause of ID.
- Published
- 2016
30. Microcephaly, intellectual impairment, bilateral vesicoureteral reflux, distichiasis, and glomuvenous malformations associated with a 16q24.3 contiguous gene deletion and a Glomulin mutation
- Author
-
Susan L. Dagenais, Jose L. Garcia-Perez, Jeffrey W. Innis, Thomas W. Glover, Miikka Vikkula, Pascal Brouillard, Peter J. Strouse, and Matthew G. Butler
- Subjects
Proband ,Pathology ,medicine.medical_specialty ,Microcephaly ,DNA Copy Number Variations ,Genotype ,Distichiasis ,Locus (genetics) ,Gene mutation ,Vesicoureteral reflux ,Article ,Intellectual Disability ,Internal medicine ,Genetics ,medicine ,Humans ,Lymphedema ,Child ,Genetics (clinical) ,Adaptor Proteins, Signal Transducing ,Paraganglioma, Extra-Adrenal ,Vesico-Ureteral Reflux ,Eyelashes ,business.industry ,Chromosome Mapping ,Forkhead Transcription Factors ,Glomus Tumor ,medicine.disease ,Magnetic Resonance Imaging ,Receptor, TIE-2 ,Glomuvenous malformation ,medicine.anatomical_structure ,Endocrinology ,Female ,Eyelid ,business ,Chromosomes, Human, Pair 16 ,Gene Deletion - Abstract
Two hereditary syndromes, lymphedema-distichiasis syndrome (LD) and blepharo-chelio-dontic (BCD) syndrome include the aberrant growth of eyelashes from the meibomian glands, known as distichiasis. LD is an autosomal dominant syndrome primarily characterized by distichiasis and the onset of lymphedema usually during puberty. Mutations in the forkhead transcription factor FOXC2 are the only known cause of LD. BCD syndrome consists of autosomal dominant abnormalities of the eyelid, lip, and teeth, and the etiology remains unknown. In this report, we describe a proband that presented with distichiasis, microcephaly, bilateral grade IV vesicoureteral reflux requiring ureteral re-implantation, mild intellectual impairment and apparent glomuvenous malformations. Distichiasis was present in three generations of the proband’s maternal side of the family. The glomuvenous malformations were severe in the proband, and maternal family members exhibited lower extremity varicosities of variable degree. A GLMN (glomulin) gene mutation was identified in the proband that accounts for the observed glomuvenous malformations; no other family member could be tested. TIE2 sequencing revealed no mutations. In the proband, an additional submicroscopic 265 kb contiguous gene deletion was identified in 16q24.3, located 609 kb distal to the FOXC2 locus, which was inherited from the proband’s mother. The deletion includes the C16ORF95, FBXO31, MAP1LC3B, and ZCCHC14 loci and 115 kb of a gene desert distal to FOXC2 and FOXL1. Thus, it is likely that the microcephaly, distichiasis, vesicoureteral and intellectual impairment in this family may be caused by the deletion of one or more of these genes and/or deletion of distant cis-regulatory elements of FOXC2 expression.
- Published
- 2012
31. Multiple congenital anomalies and developmental delay in a boy associated with a de novo 16p13.3 deletion
- Author
-
Ram Iyer, Jeffrey W. Innis, Shane C. Quinonez, Todd Ackley, and Marc E. Nelson
- Subjects
Male ,Candidate gene ,DNA Copy Number Variations ,Developmental Disabilities ,Biology ,Craniosynostosis ,Open Reading Frames ,Pregnancy ,Genetics ,medicine ,Humans ,Abnormalities, Multiple ,Copy-number variation ,Global developmental delay ,Child ,Genetics (clinical) ,Comparative Genomic Hybridization ,Infant, Newborn ,Infant ,Chromosome ,medicine.disease ,Female ,Chromosome Deletion ,medicine.symptom ,Chordee ,Haploinsufficiency ,Chromosomes, Human, Pair 16 ,Comparative genomic hybridization - Abstract
We describe a patient with multiple congenital anomalies including tracheobronchomalacia, CT-proven metopic craniosynostosis, glandular hypospadias and severe ventral chordee, torticollis, esotropia, strabismus, fifth finger clinodactyly, hallux valgus, and global developmental delay. Using high resolution chromosomal microarray analysis, we identified a de novo deletion of 555 kb on chromosome 16p13.3, 444 kb telomeric to the CREBBP gene and 623 kb centromeric of PKD1. Review of the literature revealed numerous reports of individuals with deletions involving adjacent regions including CREBBP, but only one overlapping with this isolated region of 16p13.3. Haploinsufficiency for one or more of the 25 candidate genes in the deleted genomic region may be responsible for these clinical features. No copy number variants (CNVs) span the entire region, but several small CNVs within the 555 kb genomic region reduce the likelihood for effects due to haploinsufficiency to 18 genes.
- Published
- 2011
32. A complex 6p25 rearrangement in a child with multiple epiphyseal dysplasia
- Author
-
Jeffrey W. Innis, Jirair K. Bedoyan, Marci M. Lesperance, Todd Ackley, Vinod K. Misra, and Ramaswamy K. Iyer
- Subjects
Molecular Sequence Data ,Copy number analysis ,Single-nucleotide polymorphism ,Biology ,Osteochondrodysplasias ,Polymorphism, Single Nucleotide ,Article ,Multiple epiphyseal dysplasia ,Duane Retraction Syndrome ,Genetics ,medicine ,Humans ,SNP ,Child ,Hearing Loss ,Gene ,In Situ Hybridization, Fluorescence ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,Repetitive Sequences, Nucleic Acid ,Chromosome Aberrations ,Gene Rearrangement ,Comparative Genomic Hybridization ,Base Sequence ,Forkhead Transcription Factors ,Gene rearrangement ,medicine.disease ,Dual-Specificity Phosphatases ,Mitogen-Activated Protein Kinase Phosphatases ,Chromosomes, Human, Pair 6 ,SNP array ,Comparative genomic hybridization - Abstract
Genomic rearrangements are increasingly recognized as important contributors to human disease. Here we report on an 11½-year-old child with myopia, Duane retraction syndrome, bilateral mixed hearing loss, skeletal anomalies including multiple epiphyseal dysplasia, and global developmental delay, and a complex 6p25 genomic rearrangement. We have employed oligonucleotide-based comparative genomic hybridization arrays (aCGH) of different resolutions (44 and 244K) as well as a 1 M single nucleotide polymorphism (SNP) array to analyze this complex rearrangement. Our analyses reveal a complex rearrangement involving a ∼2.21 Mb interstitial deletion, a ∼240 kb terminal deletion, and a 70–80 kb region in between these two deletions that shows maintenance of genomic copy number. The interstitial deletion contains eight known genes, including three Forkhead box containing (FOX) transcription factors (FOXQ1, FOXF2, and FOXC1). The region maintaining genomic copy number partly overlaps the dual specificity protein phosphatase 22 (DUSP22) gene. Array analyses suggest a homozygous loss of genomic material at the 5′ end of DUSP22, which was corroborated using TaqMan® copy number analysis. It is possible that this homozygous genomic loss may render both copies of DUSP22 or its products non-functional. Our analysis suggests a rearrangement mechanism distinct from a previously reported replication-based error-prone mechanism without template switching for a specific 6p25 rearrangement with a 1.22 Mb interstitial deletion. Our study demonstrates the utility and limitations of using oligonucleotide-based aCGH and SNP array technologies of increasing resolutions in order to identify complex DNA rearrangements and gene disruptions. © 2010 Wiley-Liss, Inc.
- Published
- 2010
33. Expanded HOXA13 polyalanine tracts in a monotreme
- Author
-
Jessica A. Lehoczky and Jeffrey W. Innis
- Subjects
Homeodomain Proteins ,Repetitive Sequences, Amino Acid ,Lineage (genetic) ,biology ,Reproduction ,Tachyglossidae ,Molecular Sequence Data ,Vertebrate ,Zoology ,Monotreme ,biology.organism_classification ,Article ,Evolution, Molecular ,biology.animal ,Echidna ,Animals ,Amino Acid Sequence ,Peptides ,Sequence Alignment ,Phylogeny ,Ecology, Evolution, Behavior and Systematics ,HOXA13 ,Developmental Biology - Abstract
SUMMARY The N-terminal region of human HOXA13 has seven discrete polyalanine tracts. Our previous analysis of these tracts in multiple major vertebrate clades suggested that three are mammal-specific. We now report the N-terminal HOXA13 repetitive tract structures in the monotreme Tachyglossus aculeatus (echidna). Contrary to our expectations, echidna HOXA13 possesses a unique set of polyalanine tracts and an unprecedented polyglycine tract. The data support the conclusion that the emergence of expanded polyalanine tracts in proteins occurred very early in the stem lineage that gave rise to mammals, between 162 and 315Ma.
- Published
- 2008
34. BAC transgenic analysis reveals enhancers sufficient forHoxa13and neighborhood gene expression in mouse embryonic distal limbs and genital bud
- Author
-
Jeffrey W. Innis and Jessica A. Lehoczky
- Subjects
Genetically modified mouse ,Chromosomes, Artificial, Bacterial ,Limb Buds ,Transgene ,Embryonic Development ,Mice, Transgenic ,Biology ,Article ,Mice ,Genes, Reporter ,Gene expression ,Gene duplication ,Animals ,Genitalia ,Transgenes ,Enhancer ,Gene ,Ecology, Evolution, Behavior and Systematics ,Homeodomain Proteins ,Gene Expression Regulation, Developmental ,Molecular biology ,Embryonic stem cell ,Enhancer Elements, Genetic ,Lac Operon ,DNA, Intergenic ,HOXA13 ,Developmental Biology - Abstract
We previously demonstrated that a ~1 Mb domain of genes upstream of and including Hoxa13 is co-expressed in the developing mouse limbs and genitalia. A very highly conserved non-coding sequence, mmA13CNS, located ~350 kb upstream of the Hoxa13 gene, was shown to be insufficient in transgenic mice to direct precise Hoxa13-like expression in the limb buds or genital bud, although some LacZ expression from the transgene was found in those tissues. In this report, we used overlapping β-globin minimal promoter LacZ recombinant BAC mouse transgenes encompassing mmA13CNS to localize genital bud and distal limb enhancers. Hoxa13-like embryonic genital bud expression was observed with both BACs, suggesting that a genital bud enhancer lies within the region of BAC overlap. In contrast, at least two separate regions of sequence remote to the HoxA cluster are required to drive Hoxa13-like expression in developing distal limbs. Given that the paralogous posterior HoxD and neighboring genes have been shown to be under the influence of long-range distal limb and genital bud enhancers, we hypothesize that both HoxA and HoxD long-range enhancers have one ancestral origin, which diverged in both sequence and function after the HoxA/D cluster duplication.
- Published
- 2008
35. Developmental Biology: Frontiers for Clinical Genetics: Limb development: molecular dysmorphology is at hand!
- Author
-
Douglas P. Mortlock and Jeffrey W. Innis
- Subjects
Bone development ,Genetics ,Limb development ,Anatomy ,Congenital disease ,Biology ,Neuroscience ,Genetics (clinical) - Abstract
We present a review of limb development integrating current molecular information and selected genetic disorders to illustrate the advances made in this field over the last few years. With this knowledge, clinical geneticists can now begin to consider molecular mechanisms and pathways when investigating patients with limb malformation syndromes.
- Published
- 2008
36. Molecular characterization of HOXA13 polyalanine expansion proteins in hand-foot-genital syndrome
- Author
-
Mark Born, Michael Ludwig, Jörg Dötsch, Boris Utsch, Jeffrey W. Innis, Heiko Reutter, Ricardo Gonzalez, Kenneth Galbraith, and Colleen D. McCabe
- Subjects
Male ,Cytoplasm ,Protein Folding ,Foot Deformities, Congenital ,Lactams, Macrocyclic ,Biology ,Transfection ,chemistry.chemical_compound ,Mutant protein ,Chlorocebus aethiops ,Benzoquinones ,Genetics ,medicine ,Animals ,Humans ,Hand-Foot-Genital Syndrome ,Genetics (clinical) ,Cell Nucleus ,Homeodomain Proteins ,COS cells ,Syndrome ,Geldanamycin ,medicine.disease ,Cell biology ,HOXD13 ,chemistry ,Urogenital Abnormalities ,COS Cells ,Female ,Protein folding ,Peptides ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,Hand Deformities, Congenital - Abstract
We report on a father and daughter with hand-foot-genital syndrome (HFGS) with typical skeletal and genitourinary anomalies due to a 14-residue polyalanine expansion in HOXA13. This is the largest (32 residues) polyalanine tract so far described for any polyalanine mutant protein. Polyalanine expansion results in protein misfolding, cytoplasmic aggregation and degradation; however, HOXA13 polyalanine expansions appear to act as loss of function mutations in contrast to gain of function for HOXD13 polyalanine expansions. To address this paradox we examined the cellular consequences of polyalanine expansions on HOXA13 protein using COS cell transfection and immunocytochemistry. HOXA13 polyalanine expansion proteins form cytoplasmic aggregates, and distribution between cytoplasmic aggregates or the nucleus is polyalanine tract size-dependent. Geldanamycin, an Hsp90 inhibitor, reduces the steady-state abundance of all polyalanine-expanded proteins in transfected cells. We also found that wild-type HOXA13 or HOXD13 proteins are sequestered in HOXA13 polyalanine expansion cytoplasmic aggregates. Thus, the difference between HOXA13 polyalanine expansion loss-of-function and HOXD13 polyalanine expansion dominant-negative effect is not the ability to aggregate wild-type group 13 paralogs but perhaps to variation in activities associated with refolding, aggregation or degradation of the proteins.
- Published
- 2007
37. Integrative clinical sequencing in the management of children and young adults with refractory or relapsed cancer
- Author
-
Fengyun Su, Jeffrey W. Innis, Rui Wang, Andrew C. Harris, Raymond J. Hutchinson, Aghiad Chamdin, Carrie L. Kitko, Moshe Talpaz, Kevin Frank, Xuhong Cao, Gregory A. Yanik, Victoria M. Raymond, Arul M. Chinnaiyan, Raja Rabah, Robert J. Lonigro, Valerie P. Castle, John R. Prensner, Patricia L. Robertson, Elena M. Stoffel, J. Scott Roberts, Sung Choi, Rama Jasty Rao, Jonathan R. Dillman, Dan R. Robinson, Sameek Roychowdhury, Yu Ning, James A. Connelly, Yi-Mi Wu, Irfan A. Asangani, Nallasivam Palanisamy, John E. Levine, Pankaj Vats, Jessica Everett, Rajen Mody, and L.P. Kunju
- Subjects
medicine.medical_specialty ,Pathology ,Adolescent ,Genetic counseling ,Psychological intervention ,Genetic Counseling ,Article ,Young Adult ,Internal medicine ,Neoplasms ,Outcome Assessment, Health Care ,medicine ,Humans ,Family ,Molecular Targeted Therapy ,Young adult ,Child ,Exome ,Chromosome Aberrations ,Incidental Findings ,business.industry ,Remission Induction ,Infant, Newborn ,Cancer ,Infant ,General Medicine ,Consecutive case series ,Sequence Analysis, DNA ,Precision medicine ,medicine.disease ,Child, Preschool ,Hematologic Neoplasms ,Feasibility Studies ,Observational study ,Gene Fusion ,Neoplasm Recurrence, Local ,business - Abstract
Importance Cancer is caused by a diverse array of somatic and germline genomic aberrations. Advances in genomic sequencing technologies have improved the ability to detect these molecular aberrations with greater sensitivity. However, integrating them into clinical management in an individualized manner has proven challenging. Objective To evaluate the use of integrative clinical sequencing and genetic counseling in the assessment and treatment of children and young adults with cancer. Design, Setting, and Participants Single-site, observational, consecutive case series (May 2012-October 2014) involving 102 children and young adults (mean age, 10.6 years; median age, 11.5 years, range, 0-22 years) with relapsed, refractory, or rare cancer. Exposures Participants underwent integrative clinical exome (tumor and germline DNA) and transcriptome (tumor RNA) sequencing and genetic counseling. Results were discussed by a precision medicine tumor board, which made recommendations to families and their physicians. Main Outcomes and Measures Proportion of patients with potentially actionable findings, results of clinical actions based on integrative clinical sequencing, and estimated proportion of patients or their families at risk of future cancer. Results Of the 104 screened patients, 102 enrolled with 91 (89%) having adequate tumor tissue to complete sequencing. Only the 91 patients were included in all calculations, including 28 (31%) with hematological malignancies and 63 (69%) with solid tumors. Forty-two patients (46%) had actionable findings that changed their cancer management: 15 of 28 (54%) with hematological malignancies and 27 of 63 (43%) with solid tumors. Individualized actions were taken in 23 of the 91 (25%) based on actionable integrative clinical sequencing findings, including change in treatment for 14 patients (15%) and genetic counseling for future risk for 9 patients (10%). Nine of 91 (10%) of the personalized clinical interventions resulted in ongoing partial clinical remission of 8 to 16 months or helped sustain complete clinical remission of 6 to 21 months. All 9 patients and families with actionable incidental genetic findings agreed to genetic counseling and screening. Conclusions and Relevance In this single-center case series involving young patients with relapsed or refractory cancer, incorporation of integrative clinical sequencing data into clinical management was feasible, revealed potentially actionable findings in 46% of patients, and was associated with change in treatment and family genetic counseling for a small proportion of patients. The lack of a control group limited assessing whether better clinical outcomes resulted from this approach than outcomes that would have occurred with standard care.
- Published
- 2015
38. A genomic approach to the identification and characterization of HOXA13 functional binding elements
- Author
-
Colleen D. McCabe and Jeffrey W. Innis
- Subjects
Transcriptional Activation ,Chromatin Immunoprecipitation ,Cesium ,Biology ,Response Elements ,Article ,Histones ,Mice ,03 medical and health sciences ,Dimethyl Adipimidate ,0302 clinical medicine ,Chlorides ,Genes, Reporter ,Multienzyme Complexes ,Transcription (biology) ,Genetics ,Animals ,Pyrophosphatases ,Hox gene ,Gene ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Binding Sites ,Phosphoric Diester Hydrolases ,Intron ,Acetylation ,Genomics ,Molecular biology ,Chromatin ,Up-Regulation ,3. Good health ,Cross-Linking Reagents ,Histone ,Phosphodiesterase I ,NIH 3T3 Cells ,biology.protein ,Chromatin immunoprecipitation ,030217 neurology & neurosurgery - Abstract
HOX proteins are important transcriptional regulators in mammalian embryonic development and are dysregulated in human cancers. However, there are few known direct HOX target genes and their mechanisms of regulation are incompletely understood. To isolate and characterize gene segments through which HOX proteins regulate transcription we used cesium chloride centrifugation-based chromatin purification and immunoprecipitation (ChIP). From NIH 3T3-derived HOXA13-FLAG expressing cells, 33% of randomly selected, ChIP clones were reproducibly enriched. Hox-enriched fragments (HEFs) were more AT-rich compared with cloned fragments that failed reproducible ChIP. All HEFs augmented transcription of a heterologous promoter upon coexpression with HOXA13. One HEF was from intron 2 of Enpp2, a gene highly upregulated in these cells and has been implicated in cell motility. Using Enpp2 as a candidate direct target, we identified three additional HEFs upstream of the transcription start site. HOXA13 upregulated transcription from an Enpp2 promoter construct containing these sites, and each site was necessary for full HOXA13-induced expression. Lastly, given that HOX proteins have been demonstrated to interact with histone deacetylases and/or CBP, we explored whether histone acetylation changed at Enpp2 upon HOXA13-induced activation. No change in the general histone acetylation state was observed. Our results support models in which occupation of multiple HOX binding sites is associated with highly activated genes.
- Published
- 2005
39. TBX5 Genetic Testing Validates Strict Clinical Criteria for Holt-Oram Syndrome
- Author
-
Craig T. Basson, Martin G. St. John-Sutton, Joseph Lee, Mary Ella M Pierpont, Jie He, Robert H. Spencer, Gail E. Graham, Lionel Van Maldergem, Fred Gilbert, Mark C. Hannibal, Wendy E. Smith, Deborah A. McDermott, Salim Aftimos, Matthew Weber, Alan L. Shanske, Martina Brueckner, Annick Raas-Rothschild, Darrel Waggoner, Michael Bressan, and Jeffrey W. Innis
- Subjects
Heart Defects, Congenital ,Pediatrics ,medicine.medical_specialty ,Genotype ,Heart disease ,DNA Mutational Analysis ,Population ,medicine ,Humans ,Abnormalities, Multiple ,Genetic Testing ,education ,Genotyping ,DNA Primers ,Genes, Dominant ,Genetic testing ,education.field_of_study ,Holt–Oram syndrome ,Base Sequence ,medicine.diagnostic_test ,business.industry ,Dysostosis ,Syndrome ,medicine.disease ,Surgery ,Phenotype ,Mutation ,Pediatrics, Perinatology and Child Health ,Cohort ,T-Box Domain Proteins ,business ,Hand Deformities, Congenital - Abstract
Holt-Oram syndrome (HOS) is an autosomal dominant heart-hand syndrome characterized by congenital heart disease (CHD) and upper limb deformity, and caused by mutations in the TBX5 gene. To date, the sensitivity of TBX5 genetic testing for HOS has been unclear. We now report mutational analyses of a nongenetically selected population of 54 unrelated individuals who were consecutively referred to our center with a clinical diagnosis of HOS. TBX5 mutational analyses were performed in all individuals, and clinical histories and findings were reviewed for each patient without reference to the genotypes. Twenty-six percent of the complete cohort was shown to have mutations of the TBX5 gene. However, among those subjects for whom clinical review demonstrated that their presentations met strict diagnostic criteria for HOS, TBX5 mutations were identified in 74%. No mutations were identified in those subjects who did not meet these criteria. Thus, these studies validate our clinical diagnostic criteria for HOS including an absolute requirement for preaxial radial ray upper limb malformation. Accordingly, TBX5 genotyping has high sensitivity and specificity for HOS if stringent diagnostic criteria are used in assigning the clinical diagnosis.
- Published
- 2005
40. Phenotypic heterogeneity in the presentation of d-2-hydroxyglutaric aciduria in monozygotic twins
- Author
-
Jeffrey W. Innis, Eduard A. Struys, Gajja S. Salomons, Cornelis Jakobs, Vinod K. Misra, Thomas W. Glover, William E. O'Brien, and VU University medical center
- Subjects
Endocrinology, Diabetes and Metabolism ,Biology ,Compound heterozygosity ,Biochemistry ,Glutarates ,Endocrinology ,Locus heterogeneity ,Genetics ,medicine ,Humans ,Epigenetics ,Allele ,Molecular Biology ,DNA Primers ,Base Sequence ,Genetic heterogeneity ,Metabolic disorder ,Twins, Monozygotic ,medicine.disease ,Phenotype ,Inborn error of metabolism ,Child, Preschool ,Female ,Metabolism, Inborn Errors - Abstract
d-2-Hydroxyglutaric aciduria (d-2-HGA) is a very rare autosomal recessive metabolic disorder that has recently been associated with mutations in the d -2-hydroxyglutarate dehydrogenase gene. The biochemical phenotype of d-2-HGA is defined by the accumulation of abnormal amounts of d-2-hydroxyglutarate in cerebrospinal fluid, blood, and urine while the clinical phenotype can vary from a severe epileptic encephalopathy to normal. The basis for this phenotypic variation is not well-defined. We report a set of 4 1 2 -year-old monozygotic (MZ) female twins with d-2-hydroxyglutaric aciduria who are shown to be compound heterozygotes for c.326–327dupTC, p.Glu110ArgfsX19, and c.1123G→T, p.Asp375Tyr mutations in the d -2-hydroxyglutarate dehydrogenase gene, but who have remarkably different clinical phenotypes. One twin presented with multiple congenital anomalies, severe developmental delay, and abnormal neuroradiological findings, while the other had normal neurocognitive and neuroradiological phenotypes, without concomitant congenital abnormalities. Monozygosity of these twins implies that the differences in the clinical phenotype arise from postzygotic genetic changes, epigenetic differences, or environmental factors that influence the phenotypic response to biochemical perturbation rather than allelic or locus heterogeneity. Though the mechanistic role of these factors in d-2-HGA is far from apparent, the discordance in the phenotypes of these siblings establishes that these factors are at least as important as the nature of the mutant alleles in influencing the progression of the disorder.
- Published
- 2005
41. Group 13 HOX proteins interact with the MH2 domain of R-Smads and modulate Smad transcriptional activation functions independent of HOX DNA-binding capability
- Author
-
Joanne H. Heaton, Melissa E. Williams, Jeffrey W. Innis, Thomas D. Gelehrter, and Thomas M. Williams
- Subjects
Smad5 Protein ,Transcriptional Activation ,animal structures ,Smad Proteins ,SMAD ,Biology ,Trithorax-group proteins ,DNA-binding protein ,Article ,Cell Line ,Smad1 Protein ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transforming Growth Factor beta ,Two-Hybrid System Techniques ,Genetics ,Animals ,Humans ,Smad3 Protein ,Hox gene ,Transcription factor ,030304 developmental biology ,Gene Library ,Homeodomain Proteins ,0303 health sciences ,Effector ,Extremities ,Phosphoproteins ,Fusion protein ,Cell biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Mice, Inbred C57BL ,embryonic structures ,Bone Morphogenetic Proteins ,Trans-Activators ,Homeobox ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Interactions with co-factors provide a means by which HOX proteins exert specificity. To identify candidate protein interactors of HOXA13, we created and screened an E11.5-E12.5, distal limb bud yeast two-hybrid prey library. Among the interactors, we isolated the BMP-signaling effector Smad5, which interacted with the paralogous HOXD13 but not with HOXA11 or HOXA9, revealing unique interaction capabilities of the AbdB-like HOX proteins. Using deletion mutants, we determined that the MH2 domain of Smad5 is necessary for HOXA13 interaction. This is the first report demonstrating an interaction between HOX proteins and the MH2 domain of Smad proteins. HOXA13 and HOXD13 also bind to other BMP and TGF-beta/Activin-regulated Smad proteins including Smad1 and Smad2, but not Smad4. Furthermore, HOXD13 could be co-immunoprecipitated with Smad1 from cells. Expression of HOXA13, HOXD13 or a HOXD13 homeodomain mutant (HOXD13(IQNAAA)) antagonized TGF-beta-stimulated transcriptional activation of the pAdtrack-3TP-Lux reporter vector in Mv1Lu cells as well as the Smad3/Smad4-activated pTRS6-E1b promoter in Hep3B cells. Finally, using mammalian one-hybrid assay, we show that transcriptional activation by a GAL4/Smad3-C-terminus fusion protein is specifically inhibited by HOXA13. Our results identify a new co-factor for HOX group 13 proteins and suggest that HOX proteins may modulate Smad-mediated transcriptional activity through protein-protein interactions without the requirement for HOX monomeric DNA-binding capability.
- Published
- 2005
42. Candidate downstream regulated genes of HOX group 13 transcription factors with and without monomeric DNA binding capability
- Author
-
Thomas M. Williams, Melissa E. Williams, Jeffrey W. Innis, Samir M. Hanash, Kevin T. McDonagh, David E. Misek, and Rork Kuick
- Subjects
animal structures ,Recombinant Fusion Proteins ,Genetic Vectors ,Homeodomain ,Biology ,Mice ,03 medical and health sciences ,Limb bud ,Transcriptional regulation ,Animals ,Hox gene ,Gene ,Transcription factor ,Molecular Biology ,In Situ Hybridization ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,Homeodomain Proteins ,Mice, Knockout ,Genetics ,0303 health sciences ,Muscle cell differentiation ,Microarray analysis techniques ,Gene Expression Profiling ,030302 biochemistry & molecular biology ,Gene Expression Regulation, Developmental ,Reproducibility of Results ,Extremities ,DNA ,Cell Biology ,Hox ,Realizators ,HOXA13 ,embryonic structures ,NIH 3T3 Cells ,Homeobox ,Female ,Target genes ,Protein Binding ,Transcription Factors ,Developmental Biology - Abstract
Hox genes encode transcription factors that regulate the morphogenesis of developing embryos. In mammals, knowledge of the genetic pathways, including the possible direct or indirect targets, regulated by HOX proteins is extremely limited. To identify the downstream genes regulated by posterior HOX proteins, we expressed HOXA13 in mouse embryonic fibroblasts lacking paralog group 13 expression using a bicistronic HOXA13/EGFP retroviral vector. Microarray analysis identified 68 genes with significant, reproducible RNA expression changes (50 activated; 18 repressed) in stable HOXA13-expressing cells. Genes with the GO annotation terms “extracellular matrix” and “basement membrane” were greatly overrepresented, and several were shown to be regulated by HOX proteins in other studies. Among the genes strongly activated by HOXA13 were Enpp2, a bifunctional enzyme known to modulate tumor and normal cell motility and which is expressed in precartilaginous condensations; Fhl1, a transcription factor implicated in muscle cell differentiation and development; and M32486, a putative integral membrane molecule expressed in the female reproductive tract. Expression differences in the HOXA13-expressing cells were confirmed for selected downstream genes using semi-quantitative RT-PCR, and in vivo coexpression with Hoxa13 in the limb interdigital mesenchyme was demonstrated for many. For two candidates, Igfbp4 and Fstl, interdigital limb bud expression was reduced in Hoxa13 mutants. To explore whether paralogous and nonparalogous HOX proteins could regulate the same genes, we created new HOX cell lines and examined the expression of selected genes identified by the HOXA13 screen. HOXD13 similarly activated/repressed 6 tested candidates, demonstrating that multiple downstream genetic pathways may be regulated by paralog HOX proteins. In contrast, HOXA9 was only able to repress expression of some gene targets. A HOXD13 mutant, HOXD13IQN > AAA, incapable of monomeric DNA-binding, activated the expression of 5 HOXA13-upregulated genes; but was incapable of repressing the expression of Ngef and Casp8ap2. Our results suggest that HOX protein–protein interactions without direct HOX DNA-binding may play a larger role in HOX transcriptional regulation than generally assumed, and DNA-binding appears critical for repression.
- Published
- 2005
- Full Text
- View/download PDF
43. Range of HOX/TALE superclass associations and protein domain requirements for HOXA13:MEIS interaction
- Author
-
Thomas M. Williams, Jeffrey W. Innis, and Melissa E. Williams
- Subjects
Male ,Plasma protein binding ,Mice ,0302 clinical medicine ,Cluster Analysis ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Hox gene ,Peptide sequence ,In Situ Hybridization ,Genetics ,0303 health sciences ,Limb bud ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Regulation, Developmental ,Genitalia, Female ,Hox ,Immunohistochemistry ,HOXA13 ,Neoplasm Proteins ,Cell biology ,Müllerian duct ,030220 oncology & carcinogenesis ,embryonic structures ,Female ,MEIS ,Protein Binding ,animal structures ,Molecular Sequence Data ,TALE superclass ,Protein domain ,Homeodomain ,Sequence alignment ,Biology ,03 medical and health sciences ,Two-Hybrid System Techniques ,Animals ,Immunoprecipitation ,Amino Acid Sequence ,Transcription factor ,Molecular Biology ,AbdB-like ,DNA Primers ,030304 developmental biology ,Homeodomain Proteins ,Cofactor ,Extremities ,Cell Biology ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Repressor Proteins ,NIH 3T3 Cells ,Homeobox ,Sequence Alignment ,Transcription Factors ,Superclass ,Developmental Biology - Abstract
AbdB-like HOX proteins form DNA-binding complexes with the TALE superclass proteins MEIS1A and MEIS1B, and trimeric complexes have been identified in nuclear extracts that include a second TALE protein, PBX. Thus, soluble DNA-independent protein–protein complexes exist in mammals. The extent of HOX/TALE superclass interactions, protein structural requirements, and sites of in vivo cooperative interaction have not been fully explored. We show that Hoxa13 and Hoxd13 expression does not overlap with that of Meis1–3 in the developing limb; however, coexpression occurs in the developing male and female reproductive tracts (FRTs). We demonstrate that both HOXA13 and HOXD13 associate with MEIS1B in mammalian and yeast cells, and that HOXA13 can interact with all MEIS proteins but not more diverged TALE superclass members. In addition, the C-terminal domains (CTDs) of MEIS1A (18 amino acids) and MEIS1B (93 amino acids) are necessary for HOXA13 interaction; for MEIS1B, this domain was also sufficient. We also show by yeast two-hybrid assay that MEIS proteins can interact with anterior HOX proteins, but for some, additional N-terminal MEIS sequences are required for interaction. Using deletion mutants of HOXA13 and HOXD13, we provide evidence for multiple HOX peptide domains interacting with MEIS proteins. These data suggest that HOX:MEIS interactions may extend to non-AbdB-like HOX proteins in solution and that differences may exist in the MEIS peptide domains utilized by different HOX groups. Finally, the capability of multiple HOX domains to interact with MEIS C-terminal sequences implies greater complexity of the HOX:MEIS protein–protein interactions and a larger role for variation of HOX amino-terminal sequences in specificity of function.
- Published
- 2005
- Full Text
- View/download PDF
44. Conserved expression domains for genes upstream and within the HoxA and HoxD clusters suggests a long-range enhancer existed before cluster duplication
- Author
-
Jessica A. Lehoczky, Jeffrey W. Innis, and Melissa E. Williams
- Subjects
Homeodomain Proteins ,Regulation of gene expression ,Genetics ,Sequence analysis ,Intron ,Gene Expression Regulation, Developmental ,Mice, Transgenic ,Biology ,Introns ,Conserved sequence ,Evolution, Molecular ,Mice ,Enhancer Elements, Genetic ,Multigene Family ,Gene duplication ,Trans-Activators ,Animals ,Humans ,Enhancer ,Hox gene ,Gene ,Ecology, Evolution, Behavior and Systematics ,Body Patterning ,Developmental Biology - Abstract
The posterior HoxA and HoxD genes are essential in appendicular development. Studies have demonstrated that a "distal limb enhancer," remotely located upstream of the HoxD complex, is required to drive embryonic autopod expression of the posterior Hox genes as well as the two additional non-Hox genes in the region: Evx2 and Lnp. Our work demonstrates a similar mode of regulation for Hoxa13 and four upstream genes: Evx1, Hibadh, Tax1bp, and Jaz1. These genes all show embryonic (E11.5-E13.5) distal limb and genital bud expression, suggesting the existence of a nearby enhancer influencing the expression of a domain of genes. Comparative sequence analysis between homologous human and mouse genomic sequence upstream of Hoxa13 revealed a remote 2.25-kb conserved noncoding sequence (mmA13CNS) within the fourth intron of the Hibadh gene. mmA13CNS shares a common 131-bp core identity within a conserved noncoding sequence upstream of Hoxd13, which is located within the previously identified distal limb enhancer critical region. To test the function of this conserved sequence, we created mmA13CNS-Hsp86-lacZ transgenic mice. mmA13CNS directed a wide range of tissue expression, including the central nervous system, developing olfactory tissue, limb, and genital bud. Limb and genital bud expression directed by mmA13CNS is not identical to the patterns exhibited by Hoxa13/Evx1/Hibadh/Tax1bp1/Jaz1, suggesting that mmA13CNS is not sufficient to fully recapitulate their expression in those tissues. The Evx1- and Evx2-like central nervous system expression observed in these mice suggests that the long-range regulatory element(s) for the Hox cluster existed before the cluster duplication.
- Published
- 2004
45. Polyalanine expansion in HOXA13: three new affected families and the molecular consequences in a mouse model
- Author
-
Zhi Chen, Jeffrey W. Innis, Thomas M. Williams, Stefan Mundlos, Douglas P. Mortlock, Davor Mikulic, Michael Ludwig, Katarina Lehmann, Zhihong Shao, Michael W. Glynn, Melissa E. Williams, Colleen D. Doyle, and Boris Utsch
- Subjects
Male ,Protein Denaturation ,Foot Deformities, Congenital ,RNA Splicing ,Molecular Sequence Data ,Mutant ,Biology ,Mice ,Exon ,Trinucleotide Repeats ,Genetics ,Protein biosynthesis ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,Genetics (clinical) ,Loss function ,Homeodomain Proteins ,Recombination, Genetic ,Base Sequence ,Stem Cells ,Translation (biology) ,Syndrome ,General Medicine ,Embryo, Mammalian ,Molecular biology ,Mice, Mutant Strains ,Pedigree ,Protein Biosynthesis ,RNA splicing ,Female ,Peptides ,Homologous recombination ,Hand Deformities, Congenital ,HOXA13 - Abstract
Polyalanine expansions in two of three large imperfect trinucleotide repeats encoded by the first exon of HOXA13 have been reported in hand-foot-genital syndrome (HFGS). Here we report additional families with expansions in the third repeat of 11 and 12 alanine residues, the latter being the largest expansion reported. We also report a patient with a novel, de novo 8-alanine expansion in the first large repeat. Thus, expansions in all three large HOXA13 polyalanine repeats can cause HFGS. To determine the molecular basis for impaired HOXA13 function, we performed homologous recombination in ES cells in mice to expand the size of the third largest polyalanine tract by 10 residues (HOXA13(ALA28)). Mutant mice were indistinguishable from Hoxa13 null mice. Mutant limb buds had normal steady-state Hoxa13 RNA expression, normal mRNA splicing and reduced levels of steady-state protein. In vitro translation efficiency of the HOXA13(ALA28) protein was normal. Thus, loss of function is secondary to a reduction in the in vivo abundance of the expanded protein likely due to degradation.
- Published
- 2004
46. Tibial aplasia, lower extremity mirror image polydactyly, brachyphalangy, craniofacial dysmorphism and genital hypoplasia: further delineation and mutational analysis
- Author
-
Stephanie Burns Wechsler, Jessica A. Lehoczky, Jeffrey W. Innis, and Judith G. Hall
- Subjects
Tibial aplasia ,business.industry ,Preaxial polydactyly ,General Medicine ,Anatomy ,Pathology and Forensic Medicine ,Mirror image polydactyly ,Mutational analysis ,stomatognathic diseases ,Facial dysmorphism ,Genital hypoplasia ,Pediatrics, Perinatology and Child Health ,Medicine ,business ,Genetics (clinical) ,Craniofacial dysmorphism - Abstract
Congenital anomalies involving tibial aplasia are rare. Recently, four children with an unusual combination of limb anomalies, facial dysmorphism and genital hypoplasia have been reported. All affected children reported were male. One case noted father to son transmission, implying autosomal dominan
- Published
- 2004
47. Microcephaly, jejunal atresia, aberrant right bronchus, ocular anomalies, and XY sex reversal
- Author
-
Jessica A. Lehoczky, Jeffrey W. Innis, Mansoor Mohammed, Catherine E. Keegan, Steven M. Archer, William B. Dobyns, and Eric Vilain
- Subjects
Male ,Microcephaly ,medicine.medical_specialty ,Disorders of Sex Development ,Gene Dosage ,Intestinal Atresia ,Gonadal dysgenesis ,Bronchi ,Biology ,Polymerase Chain Reaction ,Internal medicine ,Testis ,medicine ,Humans ,Abnormalities, Multiple ,Testosterone ,Eye Abnormalities ,Genetics (clinical) ,Gonadal Dysgenesis, 46,XY ,Sexual differentiation ,Corectopia ,Intestinal atresia ,Infant ,Nucleic Acid Hybridization ,Dihydrotestosterone ,Sex reversal ,medicine.disease ,Jejunum ,Endocrinology ,Receptors, Androgen ,Atresia ,medicine.symptom ,Digestive System Abnormalities ,medicine.drug - Abstract
We present a patient with microcephaly, jejunal atresia, aberrant right tracheobronchial tree, mild left blepharoptosis, and corectopia (irregular pupil), left sectoral iris stromal hypoplasia and peripheral anterior synechia, and 46,XY sex reversal. Testosterone and dihydrotestosterone (DHT) levels were within normal limits for a male infant at 3 weeks of age. Gonadectomy at age 18 months revealed immature testis tissue and no evidence of Müllerian structures. PCR amplification of the androgen receptor (AR) gene and flanking genomic regions revealed no evidence for deletion. Array-comparative genomic hybridization (array-CGH) for assessment of gene dosage in other regions of the genome was normal. This patient represents a multiple anomaly disorder similar to intestinal atresia-ocular anomalies-microcephaly syndrome (MIM#243605) but incorporating 46,XY sex reversal with testicular tissue, demonstrating a defect in the sexual differentiation pathway.
- Published
- 2004
48. Juberg-Hayward syndrome: Report of a new patient with severe phenotype and novel clinical features
- Author
-
Peter Hedera and Jeffrey W. Innis
- Subjects
medicine.medical_specialty ,Microcephaly ,Adolescent ,Genotype ,Developmental Disabilities ,Elbow ,Absent thumb ,Tongue ,Internal medicine ,Frenulum ,medicine ,Humans ,Child ,Genetics (clinical) ,medicine.diagnostic_test ,business.industry ,Karyotype ,Anatomy ,Orofaciodigital Syndromes ,medicine.disease ,Cleft Palate ,Phenotype ,medicine.anatomical_structure ,Endocrinology ,Synostosis ,Thumb ,Child, Preschool ,Juberg Hayward syndrome ,business ,Follow-Up Studies ,Fluorescence in situ hybridization - Abstract
We report a patient with severe mental retardation (MR), microcephaly, Dandy-Walker malformation, bilateral lip/palate clefts, hypertrophied sublingual frenulum, lobular tongue, absent thumbs, and other skeletal abnormalities, including Y-shaped metacarpals and urogenital abnormalities. High-resolution karyotype and subtelomeric fluorescence in situ hybridization were normal. We propose that his clinical picture is most consistent with Juberg-Hayward or orocraniodigital syndrome. Several clinical features present in our patient (unilateral distal displacement of elbow position, second-site radioulnar synostosis, bilateral Y-shaped metacarpal, lobular tongue, hypertrophic frenuli, Dandy-Walker malformation) have not previously been reported in this condition, thus expanding the phenotypic spectrum of this rare condition. The presence of these novel findings suggests possible overlap with other syndromes, such as orofaciodigital and Malpuech syndromes.
- Published
- 2003
49. Integrative biology and the developing limb bud1
- Author
-
Jeffrey W. Innis, Sharon L.R. Kardia, and Elliott H. Margulies
- Subjects
Genetics ,Limb bud ,Expression (architecture) ,Gene expression ,Identification (biology) ,Computational biology ,Biology ,Hox gene ,Gene ,Ecology, Evolution, Behavior and Systematics ,Function (biology) ,HOXA13 ,Developmental Biology - Abstract
SUMMARY The identification or selective construction of mutations within genes has allowed researchers to explore the downstream effects of gene disruption. Although these approaches have been successful, a limitation in our assessment of the consequences of conditional changes, and thereby our understanding of roles or function of genes, limits the degree to which we examine the effects of our manipulations. It is also clear that linear associations are incorrect models for describing development, and newer methods now give us an opportunity to practice an integrative biology. In our attempts to explore the consequences of Hoxa13 disruption in mice and humans, it has become clear that a better understanding of the consequences of gene alteration may be achievable by taking a broader approach with a long-term view. Fundamental questions regarding Hox gene function in vertebrates, including those related to the number of target genes; the degree of overlap of target gene regulation among paralogs; the magnitude of modulation exerted; and the identity of genes that are activated versus repressed need to be explored if a more thorough mechanistic understanding is to be achieved. To begin to address these questions, we undertook a comprehensive analysis of the expression of genes within developing limb buds of mice, and here we present some of our preliminary results. Our efforts will further (1) the exploration of the broader genetic relationships of expressed genes, (2) the determination of parallels or variations in target usage for a given gene in different tissues and between different organisms, (3) the evaluation of limb patterning mechanisms in other animal model systems, and (4) the exploration of gene expression hierarchies regulated by HOX proteins in developmental systems.
- Published
- 2002
50. Possible third case of Lin-Gettig syndrome
- Author
-
Peter Hedera and Jeffrey W. Innis
- Subjects
Heart Septal Defects, Ventricular ,Male ,Pathology ,medicine.medical_specialty ,Corpus callosum ,Craniosynostosis ,Craniosynostoses ,Camptodactyly ,Intellectual Disability ,medicine ,Humans ,Abnormalities, Multiple ,Eye Abnormalities ,Genetics (clinical) ,medicine.diagnostic_test ,business.industry ,Hypogonadism ,Dysostosis ,Eye movement ,Karyotype ,Syndrome ,medicine.disease ,Subtelomere ,Child, Preschool ,Karyotyping ,Agenesis of Corpus Callosum ,medicine.symptom ,business ,Hand Deformities, Congenital ,Fluorescence in situ hybridization - Abstract
We report a patient with craniosynostosis, severe mental retardation, absence of the corpus callosum, camptodactyly, hypogonadism, and ventricular septal defect. We propose that he has Lin-Gettig syndrome and that he is the third reported patient with this entity. Our patient also had additional phenotypic features, including palatal cleft and absent rapid eye movement (REM) sleep that were not present in the two previously described patients with this syndrome. High-resolution karyotype and subtelomeric fluorescence in situ hybridization (FISH) for cryptic telomeric rearrangement were normal. The existence of an unrelated patient with Lin-Gettig syndrome supports that this is a separate and distinct clinical entity.
- Published
- 2002
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.