Network


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

Hotspot


Dive into the research topics where Melissa A. Parisi is active.

Publication


Featured researches published by Melissa A. Parisi.


Nature Genetics | 2006

The centrosomal protein nephrocystin-6 is mutated in Joubert syndrome and activates transcription factor ATF4

John A. Sayer; Edgar A. Otto; John F. O'Toole; Gudrun Nürnberg; Michael A. Kennedy; Christian F. W. Becker; Hans Christian Hennies; Juliana Helou; Massimo Attanasio; Blake V. Fausett; Boris Utsch; Hemant Khanna; Yan Liu; Iain A. Drummond; Isao Kawakami; Takehiro Kusakabe; Motoyuki Tsuda; Li Ma; Hwankyu Lee; Ronald G. Larson; Susan J. Allen; Christopher J. Wilkinson; Erich A. Nigg; Chengchao Shou; Concepción Lillo; David S. Williams; Bernd Hoppe; Markus J. Kemper; Thomas J. Neuhaus; Melissa A. Parisi

The molecular basis of nephronophthisis, the most frequent genetic cause of renal failure in children and young adults, and its association with retinal degeneration and cerebellar vermis aplasia in Joubert syndrome are poorly understood. Using positional cloning, we here identify mutations in the gene CEP290 as causing nephronophthisis. It encodes a protein with several domains also present in CENPF, a protein involved in chromosome segregation. CEP290 (also known as NPHP6) interacts with and modulates the activity of ATF4, a transcription factor implicated in cAMP-dependent renal cyst formation. NPHP6 is found at centrosomes and in the nucleus of renal epithelial cells in a cell cycle–dependent manner and in connecting cilia of photoreceptors. Abrogation of its function in zebrafish recapitulates the renal, retinal and cerebellar phenotypes of Joubert syndrome. Our findings help establish the link between centrosome function, tissue architecture and transcriptional control in the pathogenesis of cystic kidney disease, retinal degeneration, and central nervous system development.


Nature Genetics | 2007

Mutations in the gene encoding the basal body protein RPGRIP1L, a nephrocystin-4 interactor, cause Joubert syndrome.

Heleen H. Arts; Dan Doherty; Sylvia E. C. van Beersum; Melissa A. Parisi; Stef J.F. Letteboer; Nicholas T. Gorden; Theo A. Peters; Tina Märker; Krysta Voesenek; Aileen Kartono; Hamit Özyürek; Federico M. Farin; Hester Y. Kroes; Uwe Wolfrum; Han G. Brunner; Frans P.M. Cremers; Ian A. Glass; N.V.A.M. Knoers; Ronald Roepman

Protein-protein interaction analyses have uncovered a ciliary and basal body protein network that, when disrupted, can result in nephronophthisis (NPHP), Leber congenital amaurosis, Senior-Løken syndrome (SLSN) or Joubert syndrome (JBTS). However, details of the molecular mechanisms underlying these disorders remain poorly understood. RPGRIP1-like protein (RPGRIP1L) is a homolog of RPGRIP1 (RPGR-interacting protein 1), a ciliary protein defective in Leber congenital amaurosis. We show that RPGRIP1L interacts with nephrocystin-4 and that mutations in the gene encoding nephrocystin-4 (NPHP4) that are known to cause SLSN disrupt this interaction. RPGRIP1L is ubiquitously expressed, and its protein product localizes to basal bodies. Therefore, we analyzed RPGRIP1L as a candidate gene for JBTS and identified loss-of-function mutations in three families with typical JBTS, including the characteristic mid-hindbrain malformation. This work identifies RPGRIP1L as a gene responsible for JBTS and establishes a central role for cilia and basal bodies in the pathophysiology of this disorder.


American Journal of Medical Genetics Part A | 2004

Molar Tooth Sign of the Midbrain-Hindbrain Junction: Occurrence in Multiple Distinct Syndromes

Joseph G. Gleeson; Lesley C. Keeler; Melissa A. Parisi; Sarah E. Marsh; Phillip F. Chance; Ian A. Glass; John M. Graham; Bernard L. Maria; A. James Barkovich; William B. Dobyns

The Molar Tooth Sign (MTS) is defined by an abnormally deep interpeduncular fossa; elongated, thick, and mal‐oriented superior cerebellar peduncles; and absent or hypoplastic cerebellar vermis that together give the appearance of a “molar tooth” on axial brain MRI through the junction of the midbrain and hindbrain (isthmus region). It was first described in Joubert syndrome (JS) where it is present in the vast majority of patients with this diagnosis. We previously showed that the MTS is a component of several other syndromes, including Dekaban–Arima (DAS), Senior–Löken, and COACH (cerebellar vermis hypoplasia (CVH), oligophrenia, ataxia, coloboma, and hepatic fibrosis). Here we present evidence that the MTS is seen together with polymicrogyria, Váradi–Papp syndrome (Orofaciodigital VI (OFD VI)), and a new syndrome with encephalocele and cortical renal cysts. We also present a new patient with COACH syndrome plus the MTS. We propose that the MTS is found in multiple distinct clinical syndromes that may share common developmental mechanisms. Proper classification of patients with these variants of the MTS will be essential for localization and identification of mutant genes.


Current Opinion in Pediatrics | 2000

Genetics of Hirschsprung disease.

Melissa A. Parisi; Raj P. Kapur

Hirschsprung disease (HSCR), or congenital intestinal aganglionosis, is a relatively common disorder of neural crest migration. It has a strong genetic basis, although simple Mendelian inheritance is rarely observed. Hirschsprung disease is associated with several other anomalies and syndromes, and animal models for these conditions exist. Mutations in the RET gene are responsible for approximately half of familial cases and a smaller fraction of sporadic cases. Mutations in genes that encode RET ligands (GDNF and NTN); components of another signaling pathway (EDNRB, EDN3, ECE-1); and the transcription factor, SOX10, have been identified in HSCR patients. A subset of these mutations is associated with anomalies of pigmentation and/or hearing loss. For almost every HSCR gene, incomplete penetrance of the HSCR phenotype has been observed, probably due to genetic modifier loci. Thus, HSCR has become a model of a complex polygenic disorder in which the interplay of different genes is currently being elucidated.


The Lancet | 2001

Association of germline mutation in the PTEN tumour suppressor gene and Proteus and Proteus-like syndromes.

Xiao Ping Zhou; Heather Hampel; Hannelore Thiele; Robert J. Gorlin; Raoul C. M. Hennekam; Melissa A. Parisi; Robin M. Winter; Charis Eng

The molecular aetiology of Proteus syndrome (PS) remains elusive. Germline mutations in PTEN cause Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome, which are hereditary hamartoma syndromes. Some features-eg, macrocephaly, lipomatosis, and vascular malformations-can be seen in all three syndromes. We examined PTEN in patients with PS and undefined Proteus-like syndromes (PS-like) and identified de-novo germline mutations in two of nine patients with PS and three of five patients with PS-like. Germline PTEN mutation analysis should be done in individuals with PS and PS-like because of its association with increased risk of cancer development and potential of germline-mutation transmission.


American Journal of Human Genetics | 2008

CC2D2A Is Mutated in joubert Syndrome and Interacts with the Ciliopathy-Associated Basal Body Protein CEP290

Nicholas T. Gorden; Heleen H. Arts; Melissa A. Parisi; Karlien L.M. Coene; Stef J.F. Letteboer; Sylvia E. C. van Beersum; Dorus A. Mans; Abigail Hikida; Melissa L. Eckert; Dana M. Knutzen; Abdulrahman Alswaid; Hamit Özyürek; Sel Dibooglu; Edgar A. Otto; Yangfan Liu; Erica E. Davis; Carolyn M. Hutter; Theo K. Bammler; Frederico M. Farin; Michael O. Dorschner; Meral Topçu; Elaine H. Zackai; Phillip Rosenthal; Kelly N. Owens; Nicholas Katsanis; John B. Vincent; Friedhelm Hildebrandt; Edwin W. Rubel; David W. Raible; Nine V.A.M. Knoers

Joubert syndrome and related disorders (JSRD) are primarily autosomal-recessive conditions characterized by hypotonia, ataxia, abnormal eye movements, and intellectual disability with a distinctive mid-hindbrain malformation. Variable features include retinal dystrophy, cystic kidney disease, and liver fibrosis. JSRD are included in the rapidly expanding group of disorders called ciliopathies, because all six gene products implicated in JSRD (NPHP1, AHI1, CEP290, RPGRIP1L, TMEM67, and ARL13B) function in the primary cilium/basal body organelle. By using homozygosity mapping in consanguineous families, we identify loss-of-function mutations in CC2D2A in JSRD patients with and without retinal, kidney, and liver disease. CC2D2A is expressed in all fetal and adult tissues tested. In ciliated cells, we observe localization of recombinant CC2D2A at the basal body and colocalization with CEP290, whose cognate gene is mutated in multiple hereditary ciliopathies. In addition, the proteins can physically interact in vitro, as shown by yeast two-hybrid and GST pull-down experiments. A nonsense mutation in the zebrafish CC2D2A ortholog (sentinel) results in pronephric cysts, a hallmark of ciliary dysfunction analogous to human cystic kidney disease. Knockdown of cep290 function in sentinel fish results in a synergistic pronephric cyst phenotype, revealing a genetic interaction between CC2D2A and CEP290 and implicating CC2D2A in cilium/basal body function. These observations extend the genetic spectrum of JSRD and provide a model system for studying extragenic modifiers in JSRD and other ciliopathies.


American Journal of Medical Genetics Part C-seminars in Medical Genetics | 2009

Clinical and molecular features of Joubert syndrome and related disorders

Melissa A. Parisi

Joubert syndrome (JBTS; OMIM 213300) is a rare, autosomal recessive disorder characterized by a specific congenital malformation of the hindbrain and a broad spectrum of other phenotypic findings that is now known to be caused by defects in the structure and/or function of the primary cilium. The complex hindbrain malformation that is characteristic of JBTS can be identified on axial magnetic resonance imaging and is known as the molar tooth sign (MTS); other diagnostic criteria include intellectual disability, hypotonia, and often, abnormal respiratory pattern and/or abnormal eye movements. In addition, a broad spectrum of other anomalies characterize Joubert syndrome and related disorders (JSRD), and may include retinal dystrophy, ocular coloboma, oral frenulae and tongue tumors, polydactyly, cystic renal disease (including cystic dysplasia or juvenile nephronophthisis), and congenital hepatic fibrosis. The clinical course can be variable, but most children with this condition survive infancy to reach adulthood. At least eight genes cause JSRD, with some genotype–phenotype correlations emerging, including the association between mutations in the MKS3 gene and hepatic fibrosis characteristic of the JSRD subtype known as COACH syndrome. Several of the causative genes for JSRD are implicated in other ciliary disorders, such as juvenile nephronophthisis and Meckel syndrome, illustrating the close association between these conditions and their overlapping clinical features that reflect a shared etiology involving the primary cilium.


European Journal of Human Genetics | 2007

Joubert syndrome (and related disorders) (OMIM 213300)

Melissa A. Parisi; Dan Doherty; Phillip F. Chance; Ian A. Glass

Joubert syndrome (JS) and related disorders are characterized by the ‘molar tooth sign’ (cerebellar vermis hypoplasia and brainstem anomalies) on MRI, hypotonia, developmental delay, ataxia, irregular breathing pattern and abnormal eye movements. Combinations of additional features such as polydactyly, ocular coloboma, retinal dystrophy, renal disease, hepatic fibrosis, encephalocele, and other brain malformations define clinical sub-types. Recent identification of the NPHP1, AHI1, and CEP290 genes has started to reveal the molecular basis of JS, which may implicate the primary cilium in these disorders. Additional genes remain to be identified.


American Journal of Human Genetics | 2005

Constitutional Rearrangement of the Architectural Factor HMGA2: A Novel Human Phenotype Including Overgrowth and Lipomas

Azra H. Ligon; Steven D.P. Moore; Melissa A. Parisi; Matthew E. Mealiffe; David J. Harris; Heather L. Ferguson; Bradley J. Quade; Cynthia C. Morton

Although somatic mutations in a number of genes have been associated with development of human tumors, such as lipomas, relatively few examples exist of germline mutations in these genes. Here we describe an 8-year-old boy who has a de novo pericentric inversion of chromosome 12, with breakpoints at p11.22 and q14.3, and a phenotype including extreme somatic overgrowth, advanced endochondral bone and dental ages, a cerebellar tumor, and multiple lipomas. His chromosomal inversion was found to truncate HMGA2, a gene that encodes an architectural factor involved in the etiology of many benign mesenchymal tumors and that maps to the 12q14.3 breakpoint. Similar truncations of murine Hmga2 in transgenic mice result in somatic overgrowth and, in particular, increased abundance of fat and lipomas, features strikingly similar to those observed in the child. This represents the first report of a constitutional rearrangement affecting HMGA2 and demonstrates the role of this gene in human growth and development. Systematic genetic analysis and clinical studies of this child may offer unique insights into the role of HMGA2 in adipogenesis, osteogenesis, and general growth control.


Journal of Medical Genetics | 2005

AHI1 mutations cause both retinal dystrophy and renal cystic disease in Joubert syndrome

Melissa A. Parisi; Dan Doherty; Melissa L. Eckert; Dennis W. W. Shaw; H. Ozyurek; S. Aysun; O. Giray; A. Al Swaid; S. Al Shahwan; N. Dohayan; E. Bakhsh; O. S. Indridason; William B. Dobyns; Craig L. Bennett; Phillip F. Chance; Ian A. Glass

Background: Joubert syndrome (JS) is an autosomal recessive disorder characterised by hypotonia, ataxia, mental retardation, altered respiratory pattern, abnormal eye movements, and a brain malformation known as the molar tooth sign (MTS) on cranial MRI. Four genetic loci have been mapped, with two genes identified (AHI1 and NPHP1). Methods: We screened a cohort of 117 JS subjects for AHI1 mutations by a combination of haplotype analysis and sequencing of the gene, and for the homozygous NPHP1 deletion by sequencing and marker analysis. Results: We identified a total of 15 novel AHI1 mutations in 13 families, including nonsense, missense, splice site, and insertion mutations, with some clustering in the WD40 domains. Eight families were consanguineous, but no single founder mutation was apparent. In addition to the MTS, retinal dystrophy was present in 11 of 12 informative families; however, no subjects exhibited variable features of JS such as polydactyly, encephalocele, colobomas, or liver fibrosis. In contrast to previous reports, we identified two families with affected siblings who developed renal disease consistent with nephronophthisis (NPH) in their 20s. In addition, two individuals with classic NPH were found to have homozygous NPHP1 deletions. Conclusions: Overall, 11% of subjects had AHI1 mutations, while ∼2% had the NPHP1 deletion, representing a total of less than 15% in a large JS cohort. Some preliminary genotype-phenotype correlations are possible, notably the association of renal impairment, specifically NPH, in those with NPHP1 deletions. Subjects with AHI1 mutations may be at risk of developing both retinal dystrophy and progressive kidney disease.

Collaboration


Dive into the Melissa A. Parisi's collaboration.

Top Co-Authors

Avatar

Ian A. Glass

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Dan Doherty

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meral Gunay-Aygun

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William A. Gahl

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Raj P. Kapur

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Meghana Vemulapalli

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Thierry Vilboux

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

William B. Dobyns

Seattle Children's Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian G. Phelps

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge