Network


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

Hotspot


Dive into the research topics where Natasha Cherman is active.

Publication


Featured researches published by Natasha Cherman.


The New England Journal of Medicine | 2011

A Mosaic Activating Mutation in AKT1 Associated with the Proteus Syndrome

Marjorie J. Lindhurst; Julie C. Sapp; Jamie K. Teer; Jennifer J. Johnston; Erin M. Finn; Kathryn F. Peters; Joyce T. Turner; Jennifer L. Cannons; David P. Bick; Laurel Blakemore; Catherine Blumhorst; Knut Brockmann; Peter Calder; Natasha Cherman; Matthew A. Deardorff; David B. Everman; Gretchen Golas; Robert M. Greenstein; B. Maya Kato; Kim M. Keppler-Noreuil; Sergei A. Kuznetsov; Richard T. Miyamoto; Kurt Newman; David Ng; Kevin O'brien; Steven Rothenberg; Douglas J. Schwartzentruber; Virender Singhal; Roberto Tirabosco; Joseph Upton

BACKGROUND The Proteus syndrome is characterized by the overgrowth of skin, connective tissue, brain, and other tissues. It has been hypothesized that the syndrome is caused by somatic mosaicism for a mutation that is lethal in the nonmosaic state. METHODS We performed exome sequencing of DNA from biopsy samples obtained from patients with the Proteus syndrome and compared the resultant DNA sequences with those of unaffected tissues obtained from the same patients. We confirmed and extended an observed association, using a custom restriction-enzyme assay to analyze the DNA in 158 samples from 29 patients with the Proteus syndrome. We then assayed activation of the AKT protein in affected tissues, using phosphorylation-specific antibodies on Western blots. RESULTS Of 29 patients with the Proteus syndrome, 26 had a somatic activating mutation (c.49G→A, p.Glu17Lys) in the oncogene AKT1, encoding the AKT1 kinase, an enzyme known to mediate processes such as cell proliferation and apoptosis. Tissues and cell lines from patients with the Proteus syndrome harbored admixtures of mutant alleles that ranged from 1% to approximately 50%. Mutant cell lines showed greater AKT phosphorylation than did control cell lines. A pair of single-cell clones that were established from the same starting culture and differed with respect to their mutation status had different levels of AKT phosphorylation. CONCLUSIONS The Proteus syndrome is caused by a somatic activating mutation in AKT1, proving the hypothesis of somatic mosaicism and implicating activation of the PI3K-AKT pathway in the characteristic clinical findings of overgrowth and tumor susceptibility in this disorder. (Funded by the Intramural Research Program of the National Human Genome Research Institute.).


Proceedings of the National Academy of Sciences of the United States of America | 2012

BRD4 is an atypical kinase that phosphorylates Serine2 of the RNA Polymerase II carboxy-terminal domain

Ballachanda N. Devaiah; Brian A. Lewis; Natasha Cherman; Michael C. Hewitt; Brian K. Albrecht; Pamela Gehron Robey; Keiko Ozato; Robert J. Sims; Dinah S. Singer

The bromodomain protein, BRD4, has been identified recently as a therapeutic target in acute myeloid leukemia, multiple myeloma, Burkitt’s lymphoma, NUT midline carcinoma, colon cancer, and inflammatory disease; its loss is a prognostic signature for metastatic breast cancer. BRD4 also contributes to regulation of both cell cycle and transcription of oncogenes, HIV, and human papilloma virus (HPV). Despite its role in a broad range of biological processes, the precise molecular mechanism of BRD4 function remains unknown. We report that BRD4 is an atypical kinase that binds to the carboxyl-terminal domain (CTD) of RNA polymerase II and directly phosphorylates its serine 2 (Ser2) sites both in vitro and in vivo under conditions where other CTD kinases are inactive. Phosphorylation of the CTD Ser2 is inhibited in vivo by a BRD4 inhibitor that blocks its binding to chromatin. Our finding that BRD4 is an RNA polymerase II CTD Ser2 kinase implicates it as a regulator of eukaryotic transcription.


Journal of Bone and Mineral Research | 2012

Denosumab treatment for fibrous dysplasia

Alison M. Boyce; William H. Chong; Jack Yao; Rachel I. Gafni; Marilyn H. Kelly; Christine E. Chamberlain; Carol W. Bassim; Natasha Cherman; Michelle Ellsworth; Josephine Z. Kasa-Vubu; Frances A. Farley; Alfredo A. Molinolo; Nisan Bhattacharyya; Michael T. Collins

Fibrous dysplasia (FD) is a skeletal disease caused by somatic activating mutations of the cyclic adenosine monophosphate (cAMP)‐regulating protein, α‐subunit of the Gs stimulatory protein (Gsα). These mutations lead to replacement of normal bone by proliferative osteogenic precursors, resulting in deformity, fracture, and pain. Medical treatment has been ineffective in altering the disease course. Receptor activator of NF‐κB ligand (RANKL) is a cell‐surface protein involved in many cellular processes, including osteoclastogenesis, and is reported to be overexpressed in FD‐like bone cells. Denosumab is a humanized monoclonal antibody to RANKL approved for treatment of osteoporosis and prevention of skeletal‐related events from bone metastases. We present the case of a 9‐year‐old boy with severe FD who was treated with denosumab for a rapidly expanding femoral lesion. Immunohistochemical staining on a pretreatment bone biopsy specimen revealed marked RANKL expression. He was started on monthly denosumab, with an initial starting dose of 1 mg/kg and planned 0.25 mg/kg dose escalations every 3 months. Over 7 months of treatment he showed marked reduction in pain, bone turnover markers (BTMs), and tumor growth rate. Denosumab did not appear to impair healing of a femoral fracture that occurred while on treatment. With initiation of treatment he developed hypophosphatemia and secondary hyperparathyroidism, necessitating supplementation with phosphorus, calcium, and calcitriol. BTMs showed rapid and sustained suppression. With discontinuation there was rapid and dramatic rebound of BTMs with cross‐linked C‐telopeptide (reflecting osteoclast activity) exceeding pretreatment levels, accompanied by severe hypercalcemia. In this child, denosumab lead to dramatic reduction of FD expansion and FD‐related bone pain. Denosumab was associated with clinically significant disturbances of mineral metabolism both while on treatment and after discontinuation. Denosumab treatment of FD warrants further study to confirm efficacy and determine potential morbidity, as well as to determine the mechanism of RANKL in the pathogenesis of FD and related bone marrow stromal cell diseases.


Journal of Bone and Mineral Research | 2012

Mechanism of FGF23 processing in fibrous dysplasia

Nisan Bhattacharyya; Malgorzata Wiench; Claudia E Dumitrescu; Brian M Connolly; Thomas H. Bugge; Himatkumar V Patel; Rachel I. Gafni; Natasha Cherman; Monique E. Cho; Gordon L. Hager; Michael T. Collins

Fibroblast growth factor‐23 (FGF23) is a phosphate‐ and vitamin D‐regulating hormone derived from osteoblasts/osteocytes that circulates in both active (intact, iFGF23) and inactive (C‐terminal, cFGF23) forms. O‐glycosylation by O‐glycosyl transferase N‐acetylgalactosaminyltransferase 3 (ppGalNAcT3) and differential cleavage by furin have been shown to be involved in regulating the ratio of active to inactive FGF23. Elevated iFGF23 levels are observed in a number of hypophosphatemic disorders, such as X‐linked, autosomal recessive, and autosomal dominant hypophosphatemic rickets, whereas low iFGF23 levels are found in the hyperphosphatemic disorder familial tumoral calcinosis/hyperphosphatemic hyperostosis syndrome. Fibrous dysplasia of bone (FD) is associated with increased total FGF23 levels (cFGF23 + iFGF23); however, classic hypophosphatemic rickets is uncommon. Our results suggest that it can be explained by increased FGF23 cleavage leading to an increase in inactive cFGF23 relative to active iFGF23. Given the fact that FD is caused by activating mutations in the small G‐protein Gsα that results in increased cyclic adenosine monophosphate (cAMP) levels, we postulated that there may be altered FGF23 cleavage in FD and that the mechanism may involve alterations in cAMP levels and ppGalNacT3 and furin activities. Analysis of blood specimens from patients with FD confirmed that the elevated total FGF23 levels are the result of proportionally increased cFGF23 levels, consistent with less glycosylation and enhanced cleavage by furin. Analysis of primary cell lines of normal and mutation‐harboring bone marrow stromal cells (BMSCs) from patients with FD demonstrated that BMSCs harboring the causative Gsα mutation had higher cAMP levels, lower ppGalNAcT3, and higher furin activity. These data support the model wherein glycosylation by ppGalNAcT3 inhibits FGF23 cleavage by furin and suggest that FGF23 processing is a regulated process that controls overall FGF23 activity in FD patients.


Bone | 2003

Osteoclastogenesis in fibrous dysplasia of bone: in situ and in vitro analysis of IL-6 expression

Mara Riminucci; Sergei A. Kuznetsov; Natasha Cherman; Alessandro Corsi; Paolo Bianco; P. Gehron Robey

Fibrous dysplasia of bone (FD) is caused by somatic mutations of the GNAS1 gene, which lead to constitutive activation of adenylyl cyclase and overproduction of cAMP in osteogenic cells. Previous in vitro studies using nonclonal, heterogeneous strains of FD-derived cells suggested that IL-6 might play a critical role in promoting excess osteoclastogenesis in FD. In this study, we investigated IL-6 expression in FD in situ and its relationship to the actual patterns of osteoclastogenesis within the abnormal tissue. We found that osteoclastogenesis is not spatially restricted to bone surfaces in FD but occurs to a large extent ectopicly in the fibrous tissue, where stromal cells diffusely express IL-6 mRNA and exhibit a characteristic cell morphology. We also observed specific expression of IL-6 mRNA in a proportion of osteoclasts, suggesting that an autocrine/paracrine loop may contribute to osteoclastogenesis in vivo in FD, as in some other bone diseases, including Pagets disease. We also generated homogeneous, clonally derived strains of wild-type and GNAS1-mutated stromal cells from the same individual, parent FD lesions. In this way, we could show that mutated stromal cells produce IL-6 at a basal magnitude and rate that are significantly higher than in the cognate wild-type cells. Conversely, wild-type cells respond to db-cAMP with a severalfold increase in magnitude and rate of IL-6 production, whereas mutant strains remain essentially unresponsive. Our data establish a direct link between GNAS1 mutations in stromal cells and IL-6 production but also define the complexity of the role of IL-6 in regulating osteoclastogenesis in FD in vivo. Here, patterns of osteoclastogenesis and bone resorption reflect not only the cell-autonomous effects of GNAS1 mutations in osteogenic cells (including IL-6 production) but also the local and systemic context to which non-osteogenic cells, local proportions of wild-type vs mutated cells, and systemic hormones contribute.


Journal of Bone and Mineral Research | 2008

Age-dependent demise of GNAS-mutated skeletal stem cells and "normalization" of fibrous dysplasia of bone.

Sergei A. Kuznetsov; Natasha Cherman; Mara Riminucci; Michael T. Collins; Pamela Gehron Robey; Paolo Bianco

We studied the role of somatic mosaicism in fibrous dysplasia of bone (FD) within the context of skeletal (“mesenchymal”) stem cells by assessing the frequency of mutated colony forming unit‐fibroblasts (CFU‐Fs) from FD lesions, and in some cases, from unaffected sites, in a series of patients. There was a tight inverse correlation between the percentage mutant CFU‐F versus age, suggesting demise of mutant stem cells caused by exuberant apoptosis noted in samples from young patients. In older patients, either partially or completely normal bone/marrow histology was observed. On in vivo transplantation, FD ossicles were generated only by cell strains in which mutant CFU‐Fs were identified. Strains that lacked mutant CFU‐F (but were mutation positive) failed to regenerate an FD ossicle. These data indicate that GNAS mutations are only pathogenic when in clonogenic skeletal stem cells. From these data, we have evolved the novel concept of “normalization” of FD. As a lesion ages, mutant stem cells fail to self‐renew, and their progeny are consumed by apoptosis, whereas residual normal stem cells survive, self‐renew, and enable formation of a normal structure. This suggests that activating GNAS mutations disrupt a pathway that is required for skeletal stem cell self‐renewal.


Stem Cells and Development | 2011

In Vivo Bone Formation by Progeny of Human Embryonic Stem Cells

Sergei A. Kuznetsov; Natasha Cherman; Pamela Gehron Robey

The derivation of osteogenic cells from human embryonic stem cells (hESCs) or from induced pluripotent stem cells for bone regeneration would be a welcome alternative to the use of adult stem cells. In an attempt to promote hESC osteogenic differentiation, cells of the HSF-6 line were cultured in differentiating conditions in vitro for prolonged periods of time ranging from 7 to 14.5 weeks, followed by in vivo transplantation into immunocompromised mice in conjunction with hydroxyapatite/tricalcium phosphate ceramic powder. Twelve different medium compositions were tested, along with a number of other variables in culture parameters. In differentiating conditions, HSF-6-derived cells demonstrated an array of diverse phenotypes reminiscent of multiple tissues, but after a few passages, acquired a more uniform, fibroblast-like morphology. Eight to 16 weeks post-transplantation, a group of transplants revealed the formation of histologically proven bone of human origin, including broad areas of multiple intertwining trabeculae, which represents by far the most extensive in vivo bone formation by the hESC-derived cells described to date. Knockout-Dulbeccos modified Eagles medium-based media with fetal bovine serum, dexamethasone, and ascorbate promoted more frequent bone formation, while media based on α-modified minimum essential medium promoted teratoma formation in 12- to 20-week-old transplants. Transcription levels of pluripotency-related (octamer binding protein 4, Nanog), osteogenesis-related (collagen type I, Runx2, alkaline phosphatase, and bone sialoprotein), and chondrogenesis-related (collagen types II and X, and aggrecan) genes were not predictive of either bone or teratoma formation. The most extensive bone was formed by the strains that, following 4 passages in monolayer conditions, were cultured for 23 to 25 extra days on the surface of hydroxyapatite/tricalcium phosphate particles, suggesting that coculturing of hESC-derived cells with osteoconductive material may increase their osteogenic potential. While none of the conditions tested in this study, and elsewhere, ensured consistent bone formation by hESC-derived cells, our results may elucidate further directions toward the construction of bone on the basis of hESCs or an individuals own induced pluripotent stem cells.


Journal of Bone and Mineral Research | 2001

Gnathodiaphyseal Dysplasia: A Syndrome of Fibro‐Osseous Lesions of Jawbones, Bone Fragility, and Long Bone Bowing

Mara Riminucci; Michael T. Collins; Alessandro Corsi; A. Boyde; Mark D. Murphey; Shlomo Wientroub; Sergei A. Kuznetsov; Natasha Cherman; Pamela Gehron Robey; Paolo Bianco

We report an unusual generalized skeletal syndrome characterized by fibro‐osseous lesions of the jawbones with a prominent psammomatoid body component, bone fragility, and bowing/sclerosis of tubular bones. The case fits with the emerging profile of a distinct syndrome with similarities to previously reported cases, some with an autosomal dominant inheritance and others sporadic. We suggest that the syndrome be named gnathodiaphyseal dysplasia. The patient had been diagnosed previously with polyostotic fibrous dysplasia (PFD) elsewhere, but further clinical evaluation, histopathological study, and mutation analysis excluded this diagnosis. In addition to providing a novel observation of an as yet poorly characterized syndrome, the case illustrates the need for stringent diagnostic criteria for FD. The jaw lesions showed fibro‐osseous features with the histopathological characteristics of cemento‐ossifying fibroma, psammomatoid variant. This case emphasizes that the boundaries between genuine GNAS1 mutation‐positive FD and other fibro‐osseous lesions occurring in the jawbones should be kept sharply defined, contrary to a prevailing tendency in the literature. A detailed pathological study revealed previously unreported features of cemento‐ossifying fibroma, including the participation of myofibroblasts and the occurrence of psammomatoid bodies and aberrant mineralization, within the walls of blood vessels. Transplantation of stromal cells grown from the lesion into immunocompromised mice resulted in a close mimicry of the native lesion, including the sporadic formation of psammomatoid bodies, suggesting an intrinsic abnormality of bone‐forming cells.


Blood | 2015

Bone marrow skeletal stem/progenitor cell defects in dyskeratosis congenita and telomere biology disorders

Arun Balakumaran; Prasun J. Mishra; Edyta Pawelczyk; Sayuri Yoshizawa; Brian J. Sworder; Natasha Cherman; Sergei A. Kuznetsov; Paolo Bianco; Neelam Giri; Sharon A. Savage; Glenn Merlino; Bogdan Dumitriu; Cynthia E. Dunbar; Neal S. Young; Blanche P. Alter; Pamela Gehron Robey

Dyskeratosis congenita (DC) is an inherited multisystem disorder, characterized by oral leukoplakia, nail dystrophy, and abnormal skin pigmentation, as well as high rates of bone marrow (BM) failure, solid tumors, and other medical problems such as osteopenia. DC and telomere biology disorders (collectively referred to as TBD here) are caused by germline mutations in telomere biology genes leading to very short telomeres and limited proliferative potential of hematopoietic stem cells. We found that skeletal stem cells (SSCs) within the BM stromal cell population (BMSCs, also known as BM-derived mesenchymal stem cells), may contribute to the hematologic phenotype. TBD-BMSCs exhibited reduced clonogenicity, spontaneous differentiation into adipocytes and fibrotic cells, and increased senescence in vitro. Upon in vivo transplantation into mice, TBD-BMSCs failed to form bone or support hematopoiesis, unlike normal BMSCs. TERC reduction (a TBD-associated gene) in normal BMSCs by small interfering TERC-RNA (siTERC-RNA) recapitulated the TBD-BMSC phenotype by reducing proliferation and secondary colony-forming efficiency, and by accelerating senescence in vitro. Microarray profiles of control and siTERC-BMSCs showed decreased hematopoietic factors at the messenger RNA level and decreased secretion of factors at the protein level. These findings are consistent with defects in SSCs/BMSCs contributing to BM failure in TBD.


Journal of Bone and Mineral Research | 2006

Clinical Vignette: Monostotic Fibrous Dysplasia of the Proximal Femur and Liposclerosing Myxofibrous Tumor: Which One Is Which?†

Alessandro Corsi; Fernando De Maio; Ernesto Ippolito; Natasha Cherman; Pamela Gehron Robey; Mara Riminucci; Paolo Bianco

Clinical, histological, and genetic studies of two cases of isolated fibro‐osseous lesions of the femur in adults show the overlap between monostotic fibrous dysplasia (MFD) of the proximal femur and the so‐called liposclerosing myxofibrous tumor. The two cases highlight how the incomplete understanding of the natural history of MFD may result in diagnostic pitfalls or incorrect classification of individual lesions.

Collaboration


Dive into the Natasha Cherman's collaboration.

Top Co-Authors

Avatar

Pamela Gehron Robey

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael T. Collins

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Sergei A. Kuznetsov

Novosibirsk State University

View shared research outputs
Top Co-Authors

Avatar

Mara Riminucci

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Paolo Bianco

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paolo Bianco

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alessandro Corsi

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Glenn Merlino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marilyn H. Kelly

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge