294 results on '"William H. McAlister"'
Search Results
2. Lamin B receptor-related disorder is associated with a spectrum of skeletal dysplasia phenotypes
- Author
-
Cori DeSanto, Beth A. Kozel, Ebtesam M. Abdalla, Asmaa K. Amin, Marwan Shinawi, Lauréane Mittaz-Crettol, Belinda Campos-Xavier, Andrea Superti-Furga, Sheila Unger, Eliza R Thompson, Ganka Douglas, David B. Wilson, William H. McAlister, Beryl Royer-Bertrand, and Lisa E. Kratz
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Receptors, Cytoplasmic and Nuclear ,030209 endocrinology & metabolism ,Biology ,Lamin B receptor ,Osteochondrodysplasias ,Short stature ,Evolution, Molecular ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Lymphocytes ,Heterochromatin organization ,Child ,Exome sequencing ,Base Sequence ,Infant, Newborn ,Genetic Variation ,Infant ,medicine.disease ,Hypoplasia ,Pedigree ,Phenotype ,030104 developmental biology ,Endocrinology ,Dysplasia ,Child, Preschool ,Pelger–Huet anomaly ,Female ,medicine.symptom - Abstract
LBR (Lamin B Receptor) encodes a bifunctional protein important for cholesterol biosynthesis and heterochromatin organization on the inner nuclear membrane. Pathogenic variants in LBR are associated with marked phenotypic variability, ranging from the benign Pelger-Huet anomaly to lethal Greenberg Dysplasia. We performed trio exome sequencing (ES) on two patients with atypical variants of skeletal dysplasia and their unaffected parents. Patient 1 exhibited frontal bossing, mid-face hypoplasia, short stature with rhizomelic limb shortening, and relative macrocephaly at birth. Although remained short, Patient 1 later showed spontaneous improvement in her skeletal findings. Exome sequencing revealed two novel variants in LBR, c.1504C > G (p.Arg502Gly) in exon 12 and c.1748G > T (p.Arg583Leu) in exon 14, which were inherited from her unaffected father and mother, respectively. Sterol analysis revealed an increased level of cholesta‑8,14‑dien‑3β‑ol to 2.9% of total sterols, consistent with a functional deficiency of 3β‑hydroxysterol Δ14‑reductase. Patient 2 presented at birth with short stature and marked rhizomelic limb shortening but later exhibited decreasing severity of shortening of the long bones and improvement in the radiographic skeletal abnormalities although he continued to be significantly short at age 10 years. Exome sequencing revealed that Patient 2 is homozygous for a pathogenic variant c.1534C > T (p.Arg512Trp) in exon 12 of LBR, which was inherited from his unaffected consanguineous parents. This report provides further evidence for a phenotypic spectrum of LBR-associated disorders and expands the genotypic spectrum by describing 3 novel disease-causing variants that have not been previously associated with a disease. Moreover, our data on Patient 1 demonstrate that variants throughout the gene appear to influence both the sterol reductase and nuclear functions of LBR.
- Published
- 2019
- Full Text
- View/download PDF
3. Asfotase alfa for infants and young children with hypophosphatasia: 7 year outcomes of a single-arm, open-label, phase 2 extension trial
- Author
-
Kenji P Fujita, John W. Taylor, William H. McAlister, Dawn Phillips, Jill H. Simmons, Michael P. Whyte, Nada J. Salman, Nick Bishop, Scott Moseley, and Mairead McGinn
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Fever ,Recombinant Fusion Proteins ,Endocrinology, Diabetes and Metabolism ,Population ,Hypophosphatasia ,030209 endocrinology & metabolism ,Bayley Scales of Infant Development ,Bone and Bones ,Craniosynostoses ,03 medical and health sciences ,Calcification, Physiologic ,Child Development ,0302 clinical medicine ,Endocrinology ,Internal Medicine ,Humans ,Medicine ,Enzyme Replacement Therapy ,Respiratory function ,030212 general & internal medicine ,Toddler ,Child ,education ,Adverse effect ,Respiratory Tract Infections ,education.field_of_study ,business.industry ,Infant, Newborn ,Infant ,Alkaline Phosphatase ,medicine.disease ,Treatment Outcome ,Tolerability ,Child, Preschool ,Immunoglobulin G ,Asfotase alfa ,Female ,business - Abstract
Summary Background Our previous phase 2, open-label study of 11 infants and young children with life-threatening perinatal or infantile hypophosphatasia showed 1 year safety and efficacy of asfotase alfa, an enzyme replacement therapy. We aimed to report the long-term outcomes over approximately 7 years of treatment. Methods We did a prespecified, end of study, 7 year follow-up of our single-arm, open-label, phase 2 trial in which children aged 3 years or younger with life-threatening perinatal or infantile hypophosphatasia were recruited from ten hospitals (six in the USA, two in the UK, one in Canada, and one in the United Arab Emirates). Patients received asfotase alfa (1 mg/kg three times per week subcutaneously, adjusted to 3 mg/kg three times per week if required) for up to 7 years (primary treatment period plus extension phase) or until the product became commercially available; dosage adjustments were made at each visit according to changes in the patient's weight. The primary objectives of this extension study were to assess the long-term tolerability of asfotase alfa, defined as the number of patients with one or more treatment-emergent adverse events, and skeletal manifestations associated with hypophosphatasia, evaluated using the Radiographic Global Impression of Change (RGI-C) scale (−3 indicating severe worsening, and +3 complete or near-complete healing). Respiratory support, growth, and cognitive and motor functions were also evaluated. All efficacy and safety analyses were done in all patients who received any asfotase alfa (full-analysis population). This study and extension phase are registered with ClinicalTrials.gov , number NCT01205152 , and EudraCT, number 2009-009369-32. Findings 11 participants were recruited between Oct 6, 2008, and Dec 4, 2009. Ten patients completed a 6 month treatment period and entered the extension phase; nine received asfotase alfa for at least 6 years and completed the study, with four being treated for more than 7 years. Skeletal healing was sustained over 7 years of treatment; all evaluable patients had RGI-C scores of at least +2 at year 6 (n=9; median score +2·0 [range 2·0–3·0]) and year 7 (n=7; median score +2·3 [2·0–3·0]). No patient who completed the study required respiratory support after year 4. Weight Z scores improved to within normal range from year 3 to study end; length or height Z scores improved but remained below normal. Age-equivalent scores on gross motor, fine motor, and cognitive subscales of the Bayley Scales of Infant and Toddler Development also improved. All 11 patients had at least one treatment-emergent adverse event. The most common adverse events were pyrexia (eight [73%] of 11 patients), upper respiratory tract infection (eight [73%]), craniosynostosis (seven [64%]), and pneumonia (seven [64%]). Serious adverse events related to asfotase alfa occurred in three (27%) patients (severe chronic hepatitis; moderate immediate post-injection reaction; and severe craniosynostosis with severe conductive deafness). Interpretation Patients with perinatal or infantile hypophosphatasia treated with asfotase alfa for up to 7 years showed early, sustained improvements in skeletal mineralisation. Respiratory function, growth, and cognitive and motor function also improved, and asfotase alfa was generally well tolerated. Funding Alexion Pharmaceuticals, Inc.
- Published
- 2019
- Full Text
- View/download PDF
4. Periarticular calcifications containing giant pseudo-crystals of francolite in skeletal fluorosis from 1,1-difluoroethane 'huffing'
- Author
-
Nilton, Salles Rosa Neto, Daniel, Englert, William H, McAlister, Steven, Mumm, David, Mills, Deborah J, Veis, Alan, Burshell, Alan, Boyde, and Michael P, Whyte
- Subjects
Bone Diseases, Metabolic ,Histology ,Hydrocarbons, Fluorinated ,Physiology ,Endocrinology, Diabetes and Metabolism ,Osteoarthritis ,Calcinosis ,Humans ,Hypophosphatasia ,Female ,Alkaline Phosphatase ,Osteosclerosis - Abstract
Inhalant use disorder is a psychiatric condition characterized by repeated deliberate inhalation from among a broad range of household and industrial chemical products with the intention of producing psychoactive effects. In addition to acute intoxication, prolonged inhalation of fluorinated compounds can cause skeletal fluorosis (SF). We report a young woman referred for hypophosphatasemia and carrying a heterozygous ALPL gene variant (c.457TC, p.Trp153Arg) associated with hypophosphatasia, the heritable metabolic bone disease featuring impaired skeletal mineralization, who instead suffered from SF. Manifestations of her SF included recurrent articular pain, axial osteosclerosis, elevated bone mineral density, maxillary exostoses, and multifocal periarticular calcifications. SF was suspected when a long history was discovered of 'huffing' a computer cleaner containing 1,1-difluoroethane. Investigation revealed markedly elevated serum and urine levels of F
- Published
- 2022
- Full Text
- View/download PDF
5. Vitamin B
- Author
-
Michael P, Whyte, Jennifer D, May, William H, McAlister, Katherine, Burgener, Samuel R, Cortez, Raymond, Kreienkamp, Olivia, Castro, Rachel, Verzola, Ana Solis, Zavala, Christopher C, McPherson, Gary S, Gottesman, Karen L, Ericson, Stephen P, Coburn, and Ana Maria, Arbelaez
- Subjects
Male ,Pyridoxal ,Pregnancy ,Infant, Newborn ,Humans ,Hypophosphatasia ,Female ,Vitamins ,Alkaline Phosphatase ,Vitamin B 6 ,Phosphates - Abstract
Pyridoxal 5'-phosphate (PLP), the principal circulating form of vitamin B
- Published
- 2021
6. Coalescing expansile skeletal disease: Delineation of an extraordinary osteopathy involving the IFITM5 mutation of osteogenesis imperfecta type V
- Author
-
Gary S. Gottesman, Margaret Huskey, Shenghui Duan, James Aronson, William H. McAlister, Karen L. Clements, Steven Mumm, Michael P. Whyte, Vinieth N. Bijanki, Horacio Plotkin, Marina Stolina, Robert S. Weinstein, Deborah Wenkert, and Katherine L Madson
- Subjects
0301 basic medicine ,Adult ,Male ,Hyperostosis ,Pathology ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Bone and Bones ,Bone remodeling ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,medicine ,Humans ,Child ,Periosteum ,Hyaline cartilage ,business.industry ,Ossification ,Cartilage ,Infant ,Membrane Proteins ,Middle Aged ,Osteogenesis Imperfecta ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Osteogenesis imperfecta ,Mutation ,Heterotopic ossification ,Female ,medicine.symptom ,business ,5' Untranslated Regions - Abstract
In 2003, we briefly reported the remarkable osteopathy of a 12-year-old boy who at age two months began fracturing his limbs with subsequent hyperplastic callus formation and expansion and fusion of appendicular bones. By age ten years he had coalesced his lumbosacral spine, pelvis, femurs, and leg and foot bones as a single structure. Computed tomography of expanded bone revealed a thin cortical shell, diminished irregular trabeculae, and cystic areas. Histopathology featured foci of woven bone, densely packed osteocytes, cartilage, fibrovascular tissue, and massive fat deposition in the marrow space lacking hematogenous precursor cells. Bone turnover markers indicated accelerated remodeling and the few radiographically assessable appendicular bones improved during brief adherence to alendronate therapy. Following puberty, serum multiplex biomarker profiling confirmed accelerated bone turnover. At age 23 years, macrospecimens from leg amputation revealed ossification along capsular tissue together with hyaline cartilage degeneration. Concurrently, the life-long course of this same disorder was delineated in an unrelated woman until her death at age 51 years. Both patients demonstrated the radiographic hallmarks and harbored the heterozygous point mutation (c.-14C>T) in the 5′-UTR of IFITM5 associated with osteogenesis imperfecta type V (OI-V). Herein, we detail the clinical, radiological, histopathological, biochemical, and molecular findings and discuss the etiology and pathogenesis of this extraordinary osteopathy that we call coalescing expansile skeletal disease.
- Published
- 2020
7. Juvenile Paget’s Disease From Heterozygous Mutation of SP7 Encoding Osterix (Specificity Protein 7, Transcription Factor Sp7)
- Author
-
Nori Kurihara, Homer Sedighi, Steven Mumm, Michael P. Whyte, William H. McAlister, Deborah J. Veis, G. David Roodman, Vinieth N. Bijanki, Philippe M. Campeau, Angela Nenninger, Gary S. Gottesman, and Shenghui Duan
- Subjects
0301 basic medicine ,musculoskeletal diseases ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Article ,Bone remodeling ,03 medical and health sciences ,Osteosclerosis ,Young Adult ,0302 clinical medicine ,Osteoprotegerin ,Osteoclast ,Internal medicine ,Gene duplication ,Medicine ,Missense mutation ,Humans ,Sequence Deletion ,biology ,business.industry ,Homozygote ,RANK Ligand ,Osteoblast ,medicine.disease ,Osteitis Deformans ,3. Good health ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,RANKL ,Sp7 Transcription Factor ,Child, Preschool ,Mutation ,biology.protein ,Female ,business ,Transcription Factors - Abstract
Juvenile Paget's disease (JPD) became in 1974 the commonly used name for ultra-rare heritable occurrences of rapid bone remodeling throughout of the skeleton that present in infancy or early childhood as fractures and deformity hallmarked biochemically by marked elevation of serum alkaline phosphatase (ALP) activity (hyperphosphatasemia). Untreated, JPD can kill during childhood or young adult life. In 2002, we reported that homozygous deletion of the gene called tumor necrosis factor receptor superfamily, member 11B (TNFRSF11B) encoding osteoprotegerin (OPG) explained JPD in Navajos. Soon after, other bi-allelic loss-of-function TNFRSF11B defects were identified in JPD worldwide. OPG inhibits osteoclastogenesis and osteoclast activity by decoying receptor activator of nuclear factor κ-B (RANK) ligand (RANKL) away from its receptor RANK. Then, in 2014, we reported JPD in a Bolivian girl caused by a heterozygous activating duplication within TNFRSF11A encoding RANK. Herein, we identify mutation of a third gene underlying JPD. An infant girl began atraumatic fracturing of her lower extremity long-bones. Skull deformity and mild hearing loss followed. Our single investigation of the patient, when she was 15 years-of-age, showed generalized osteosclerosis and hyperostosis. DXA revealed a Z-score of +5.1 at her lumbar spine and T-score of +3.3 at her non-dominant wrist. Biochemical studies were consistent with positive mineral balance and several markers of bone turnover were elevated and included striking hyperphosphatasemia. Iliac crest histopathology was consistent with rapid skeletal remodeling. Measles virus transcripts, common in classic Paget's disease of bone, were not detected in circulating mononuclear cells. Then, reportedly, she responded to several months of alendronate therapy with less skeletal pain and correction of hyperphosphatasemia but had been lost to our follow-up. After we detected no defect in TNFRSF11A or B, trio exome sequencing revealed a de novo heterozygous missense mutation (c.926C>G; p.S309W) within SP7 encoding the osteoblast transcription factor osterix (specificity protein 7, transcription factor SP7). Thus, mutation of SP7 represents a third genetic cause of JPD.
- Published
- 2020
8. Persistent idiopathic hyperphosphatasemia from bone alkaline phosphatase in a healthy boy
- Author
-
Per Magnusson, Nina S. Ma, Gary S. Gottesman, William H. McAlister, Karen L. Ericson, Angela Nenninger, Steven Mumm, Michael P. Whyte, and Vinieth N. Bijanki
- Subjects
0301 basic medicine ,Gene isoform ,Male ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Hypophosphatasia ,030209 endocrinology & metabolism ,Isozyme ,Bone and Bones ,03 medical and health sciences ,0302 clinical medicine ,N-terminal telopeptide ,Internal medicine ,medicine ,Humans ,Renal Insufficiency, Chronic ,Child ,Minerals ,Chemistry ,ALPL ,Osteoblast ,medicine.disease ,Alkaline Phosphatase ,Isoenzymes ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Alkaline phosphatase ,Type I collagen - Abstract
Alkaline phosphatase (ALP) in humans comprises a family of four cell-surface phosphomonoester phosphohydrolase isozymes. Three genes separately encode the "tissue-specific" ALPs whereas the fourth gene encodes ubiquitous homodimeric "tissue-nonspecific" ALP (TNSALP) richly expressed in bone, liver, kidney, and developing teeth. TNSALP monomers have five putative N-linked glycosylation sites where different post-translational modifications account for this isozyme's distinctive physicochemical properties in different organs. Three bone-derived TNSALP (BALP) isoforms (B/I, B1, and B2) are present in healthy serum, whereas a fourth BALP isoform (B1x) can circulate in chronic kidney disease. Herein, we report a healthy boy with persistent hyperphosphatasemia due to BALP levels two- to threefold higher than age-appropriate reference values. High-performance liquid chromatography, electrophoresis, heat inactivation, catalysis inhibition, and polyethylene glycol precipitation revealed increased serum B/I, B1, and B2 differing from patterns found in skeletal diseases. B/I was ~23-fold elevated. Absence of mental retardation and physical stigmata excluded Mabry syndrome, the ALP-anchoring disorder causing hyperphosphatasemia. Routine biochemical studies indicated intact mineral homeostasis. Serum N-terminal propeptide of type I procollagen (P1NP) level was normal, but C-terminal cross-linking telopeptide of type I collagen (CTX) level was elevated. However, radiological studies showed no evidence for a generalized skeletal disturbance. Circulating pyridoxal 5'-phosphate, a TNSALP natural substrate, was not low despite the laboratory hyperphosphatasemia, thereby suggesting BALP phosphohydrolase activity was not elevated endogenously. Mutation analysis of the ALPL gene encoding TNSALP revealed no defect. His non-consanguineous healthy parents had serum total ALP activity and BALP protein levels that were normal. Our patient's sporadic idiopathic hyperphosphatasemia could reflect altered post-translational modification together with increased expression and/or impaired degradation of BALP.
- Published
- 2020
9. Healing of vitamin D deficiency rickets complicating hypophosphatasia suggests a role beyond circulating mineral sufficiency for vitamin D in musculoskeletal health
- Author
-
Karen E. Mack, Elizabeth L. Lin, Donna Griffin, William H. McAlister, Gary S. Gottesman, Steven Mumm, Michael P. Whyte, and Vinieth N. Bijanki
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Parathyroid hormone ,Hypophosphatasia ,030209 endocrinology & metabolism ,Rickets ,vitamin D deficiency ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Vitamin D and neurology ,Humans ,Vitamin D ,Osteomalacia ,Minerals ,business.industry ,ALPL ,Infant ,medicine.disease ,Alkaline Phosphatase ,030104 developmental biology ,Endocrinology ,chemistry ,Hypercalcemia ,Female ,business ,Cholecalciferol - Abstract
Hypophosphatasia (HPP) is the metabolic bone disease caused by loss-of-function mutation(s) of the ALPL gene that encodes the cell-surface tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP). In HPP, extracellular accumulation of inorganic pyrophosphate (PPi), a TNSALP natural substrate and inhibitor of biomineralization, often leads to rickets or osteomalacia despite normal or sometimes elevated circulating levels of calcium (Ca) and inorganic phosphate (Pi). We report an infant girl with vitamin D deficiency rickets subsequently healed by cholecalciferol administration alone before receiving TNSALP-replacement therapy for accompanying HPP. Throughout her clinical course, circulating Ca and Pi levels were normal or elevated. At presentation with failure-to-thrive at age six months, radiographs revealed severe rickets and serum 25(OH)D was 8 ng/mL (Nl, 30–100), yet low ALP activity 55 U/L (Nl, 124–341), normal Ca 9.3 mg/dL (Nl, 8.5–10.1) and Pi 6.4 mg/dL (Nl, 3.5–7.0), and low-normal parathyroid hormone 21 pg/mL (Nl, 14–72) were instead consistent with HPP. At age nine months, after 1000 IU of cholecalciferol orally each day for six weeks, serum 25(OH)D was 86 ng/mL, strength markedly better, and radiographs documented significant improvement of rickets. At age 18 months, with fully healed vitamin D deficiency rickets, findings of underlying HPP included a waddling gait and Gower sign, metaphyseal “tongues” of radiolucency, elevated serum pyridoxal 5′-phosphate 121 ng/mL (Nl, 2–33), and bi-allelic ALPL missense mutations. Then, nearly complete restoration of strength and radiographic healing of her remaining skeletal disease from HPP occurred during asfotase alfa enzyme replacement treatment. At no time, including presentation, were circulating Ca or Pi levels compromised. Instead, and in keeping with HPP, high-normal or elevated serum Ca and Pi concentrations were consistently documented. Thus, our findings suggest some role for vitamin D in musculoskeletal health beyond assuring circulating mineral sufficiency.
- Published
- 2020
10. Unique Variant ofNOD2Pediatric Granulomatous Arthritis With Severe 1,25‐Dihydroxyvitamin D‐Mediated Hypercalcemia and Generalized Osteosclerosis
- Author
-
Vinieth N. Bijanki, Lien Trinh, Steven Mumm, Michael P. Whyte, William H. McAlister, Gary S. Gottesman, Emilina Lim, Angela Nenninger, David Buchbinder, Matthew G Boden, and Deborah J. Veis
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,DNA Mutational Analysis ,Nod2 Signaling Adaptor Protein ,Arthritis ,Article ,Bone and Bones ,03 medical and health sciences ,Osteosclerosis ,0302 clinical medicine ,Bone Marrow ,Synovitis ,medicine ,Humans ,Orthopedics and Sports Medicine ,Amino Acid Sequence ,Vitamin D ,Blau syndrome ,030203 arthritis & rheumatology ,Granuloma ,Base Sequence ,business.industry ,Synovial Membrane ,Osteopetrosis ,medicine.disease ,030104 developmental biology ,Child, Preschool ,Mutation ,Hypercalcemia ,Female ,Sarcoidosis ,Nephrocalcinosis ,Generalized osteosclerosis ,business - Abstract
Pediatric granulomatous arthritis (PGA) refers to two formerly separate entities: autosomal dominant Blau syndrome (BS) and its sporadic phenocopy early-onset sarcoidosis (EOS). In 2001 BS and in 2005 EOS became explained by heterozygous mutations within the gene that encodes nucleotide-binding oligomerization domain-containing protein 2 (NOD2), also called caspase recruitment domain-containing protein 15 (CARD15). NOD2 is a microbe sensor in leukocyte cytosol that activates and regulates inflammation. PGA is characterized by a triad of autoinflammatory problems (dermatitis, uveitis, and arthritis) in early childhood, which suggests the causal NOD2/CARD15 mutations are activating defects. Additional complications of PGA were recognized especially when NOD2 mutation analysis became generally available. However, in PGA, hypercalcemia is only briefly mentioned, and generalized osteosclerosis is not reported, although NOD2 regulates NF-κB signaling essential for osteoclastogenesis and osteoclast function. Herein, we report a 4-year-old girl with PGA uniquely complicated by severe 1,25(OH)2 D-mediated hypercalcemia, nephrocalcinosis, and compromised renal function together with radiological and histopathological features of osteopetrosis (OPT). The classic triad of PGA complications was absent, although joint pain and an antalgic gait accompanied wrist, knee, and ankle swelling and soft non-tender masses over her hands, knees, and feet. MRI revealed tenosynovitis in her hands and suprapatellar effusions. Synovial biopsy demonstrated reactive synovitis without granulomas. Spontaneous resolution of metaphyseal osteosclerosis occurred while biochemical markers indicated active bone turnover. Anti-inflammatory medications suppressed circulating 1,25(OH)2 D, corrected the hypercalcemia, and improved her renal function, joint pain and swelling, and gait. Mutation analysis excluded idiopathic infantile hypercalcemia, type 1, and known forms of OPT, and identified a heterozygous germline missense mutation in NOD2 common in PGA (c.1001G>A, p.Arg334Gln). Thus, radiological and histological findings of OPT and severe hypercalcemia from apparent extrarenal production of 1,25(OH)2 D can complicate NOD2-associated PGA. Although the skeletal findings seem inconsequential, treatment of the hypercalcemia is crucial to protect the kidneys. © 2018 American Society for Bone and Mineral Research.
- Published
- 2018
- Full Text
- View/download PDF
11. Validation of a Novel Scoring System for Changes in Skeletal Manifestations of Hypophosphatasia in Newborns, Infants, and Children: The Radiographic Global Impression of Change Scale
- Author
-
Scott Moseley, David D. Thompson, Kenji P Fujita, William H. McAlister, and Michael P. Whyte
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,business.industry ,Intraclass correlation ,Endocrinology, Diabetes and Metabolism ,Hypophosphatasia ,Repeated measures design ,030209 endocrinology & metabolism ,medicine.disease ,Osteopenia ,03 medical and health sciences ,Inter-rater reliability ,030104 developmental biology ,0302 clinical medicine ,Asfotase alfa ,Medicine ,Orthopedics and Sports Medicine ,Analysis of variance ,business ,Kappa - Abstract
Hypophosphatasia (HPP) is the heritable metabolic disease characterized by impaired skeletal mineralization due to low activity of the tissue-nonspecific isoenzyme of alkaline phosphatase. Although HPP during growth often manifests with distinctive radiographic skeletal features, no validated method was available to quantify them, including changes over time. We created the Radiographic Global Impression of Change (RGI-C) scale to assess changes in the skeletal burden of pediatric HPP. Site-specific pairs of radiographs of newborns, infants, and children with HPP from three clinical studies of asfotase alfa, an enzyme replacement therapy for HPP, were obtained at baseline and during treatment. Each pair was scored by three pediatric radiologists ("raters"), with nine raters across the three studies. Intrarater and interrater agreement was determined by weighted Kappa coefficients. Interrater reliability was assessed using intraclass correlation coefficients (ICCs) and by two-way random effects analysis of variance (ANOVA) and a mixed-model repeated measures ANOVA. Pearson correlation coefficients evaluated relationships of the RGI-C to the Rickets Severity Scale (RSS), Pediatric Outcomes Data Collection Instrument Global Function Parent Normative Score, Childhood Health Assessment Questionnaire Disability Index, 6-Minute Walk Test percent predicted, and Z-score for height in patients aged 6 to 12 years at baseline. Eighty-nine percent (8/9) of raters showed substantial or almost perfect intrarater agreement of sequential RGI-C scores (weighted Kappa coefficients, 0.72 to 0.93) and moderate or substantial interrater agreement (weighted Kappa coefficients, 0.53 to 0.71) in patients aged 0 to 12 years at baseline. Moderate-to-good interrater reliability was observed (ICC, 0.57 to 0.65). RGI-C scores were significantly (p ≤ 0.0065) correlated with the RSS and with measures of global function, disability, endurance, and growth in the patients aged 6 to 12 years at baseline. Thus, the RGI-C is valid and reliable for detecting clinically important changes in skeletal manifestations of severe HPP in newborns, infants, and children, including during asfotase alfa treatment. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
- Published
- 2018
- Full Text
- View/download PDF
12. Gnathodiaphyseal dysplasia: Severe atypical presentation with novel heterozygous mutation of the anoctamin gene (ANO5)
- Author
-
Ghada A. Otaify, Marina Stolina, Marwan Shinawi, Olivier Lichtarge, Abby S. Hollander, Wei-Shen Chen, Samir K. El-Mofty, J. Eric Gordon, Albert S. Woo, Gary S. Gottesman, Michael P. Whyte, Marisa V. Andrews, William H. McAlister, Panagiotis Katsonis, Fan Zhang, and Deborah V. Veis
- Subjects
Male ,0301 basic medicine ,Diaphyseal sclerosis ,Pathology ,medicine.medical_specialty ,Histology ,Physiology ,Gnathodiaphyseal dysplasia ,Endocrinology, Diabetes and Metabolism ,Mutation, Missense ,Anoctamins ,Article ,03 medical and health sciences ,Exon ,medicine ,Humans ,Missense mutation ,Genetics ,business.industry ,Osteogenesis Imperfecta ,Debulking ,medicine.disease ,Cherubism ,Phenotype ,030104 developmental biology ,Child, Preschool ,Maxilla ,Mutation (genetic algorithm) ,business - Abstract
Gnathodiaphyseal dysplasia (GDD; OMIM #166260) is an ultra-rare autosomal dominant disorder caused by heterozygous mutation in the anoctamin 5 (ANO5) gene and features fibro-osseous lesions of the jawbones, bone fragility with recurrent fractures, and bowing/sclerosis of tubular bones. The physiologic role of ANO5 is unknown. We report a 5-year-old boy with a seemingly atypical and especially severe presentation of GDD and unique ANO5 mutation. Severe osteopenia was associated with prenatal femoral fractures, recurrent postnatal fractures, and progressive bilateral enlargement of his maxilla and mandible beginning at ~ 2 months-of-age that interfered with feeding and speech and required four debulking operations. Histopathological analysis revealed benign fibro-osseous lesions resembling cemento-ossifying fibromas of the jaw without psammomatoid bodies. A novel, de novo, heterozygous, missense mutation was identified in exon 15 of ANO5 (c.1553G>A; p.Gly518Glu). Our findings broaden the phenotypic and molecular spectra of GDD. Fractures early in life with progressive facial swelling are key features. We assessed his response to a total of 7 pamidronate infusions commencing at age 15 months. Additional reports must further elucidate the phenotype, explore any genotype-phenotype correlation, and evaluate treatments.
- Published
- 2018
- Full Text
- View/download PDF
13. Vitamin B6 deficiency with normal plasma levels of pyridoxal 5′-phosphate in perinatal hypophosphatasia
- Author
-
Michael P. Whyte, Samuel R. Cortez, Katherine Burgener, Raymond Kreienkamp, William H. McAlister, Stephen P. Coburn, Ana Solis Zavala, Olivia Castro, Gary S. Gottesman, Karen L. Ericson, Rachel Verzola, Jennifer D. May, Ana Maria Arbelaez, and Christopher McPherson
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Hypophosphatasia ,ALPL ,030209 endocrinology & metabolism ,Rickets ,Pyridoxine ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Asfotase alfa ,Internal medicine ,Lactation ,medicine ,Alkaline phosphatase ,Pyridoxal ,medicine.drug - Abstract
Pyridoxal 5′-phosphate (PLP), the principal circulating form of vitamin B6 (B6), is elevated in the plasma of individuals with hypophosphatasia (HPP). HPP is the inborn-error-of-metabolism caused by loss-of-function mutation(s) of ALPL, the gene that encodes the “tissue-nonspecific” isoenzyme of alkaline phosphatase (TNSALP). PLP accumulates extracellularly in HPP because it is a natural substrate of this cell-surface phosphomonoester phosphohydrolase. Even individuals mildly affected by HPP manifest this biochemical hallmark, which is used for diagnosis. Herein, an exclusively breast-fed newborn boy with life-threatening perinatal HPP had uniquely normal instead of markedly elevated plasma PLP levels before beginning asfotase alfa (AA) TNSALP-replacement therapy. These abnormal PLP levels were explained by B6 deficiency, confirmed by his low plasma level of 4-pyridoxic acid (PA), the B6 degradation product. His mother, a presumed carrier of one of his two ALPL missense mutations, had serum ALP activity of 50 U/L (Nl 40–130) while her plasma PLP level was 9 μg/L (Nl 5–50) and PA was 3 μg/L (Nl 3−30). Her dietary history and breast milk pyridoxal (PL) level indicated she too was B6 deficient. With B6 supplementation using a breast milk fortifier, the patient's plasma PA level corrected, while his PLP level remained in the normal range but now in keeping with AA treatment. Our experience reveals that elevated levels of PLP in the circulation in HPP require some degree of B6 sufficiency, and that anticipated increases in HPP can be negated by hypovitaminosis B6.
- Published
- 2021
- Full Text
- View/download PDF
14. Melorheostosis: Exome sequencing of an associated dermatosis implicates postzygotic mosaicism of mutated KRAS
- Author
-
Steven Mumm, Michael P. Whyte, Gary S. Gottesman, Elaine R. Mardis, Kilannin Krysiak, Lee Trani, Susan J. Bayliss, Robert Lesurf, Angela Nenninger, Vinieth N. Bijanki, Brian A. Van Tine, Obi L. Griffith, Katie M. Campbell, Malachi Griffith, Ilana S. Rosman, William H. McAlister, and Zachary L. Skidmore
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Histology ,Adolescent ,Melorheostosis ,Physiology ,Endocrinology, Diabetes and Metabolism ,Biology ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,Osteosclerosis ,0302 clinical medicine ,Germline mutation ,hemic and lymphatic diseases ,medicine ,PIK3CA Gene Mutation ,Humans ,Exome ,Genetic Predisposition to Disease ,Nevus ,neoplasms ,Osteopoikilosis ,Exome sequencing ,Mosaicism ,medicine.disease ,carbohydrates (lipids) ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,KRAS ,Haploinsufficiency - Abstract
Melorheostosis (MEL) is the rare sporadic dysostosis characterized by monostotic or polyostotic osteosclerosis and hyperostosis often distributed in a sclerotomal pattern. The prevailing hypothesis for MEL invokes postzygotic mosaicism. Sometimes scleroderma-like skin changes, considered a representation of the pathogenetic process of MEL, overlie the bony changes, and sometimes MEL becomes malignant. Osteopoikilosis (OPK) is the autosomal dominant skeletal dysplasia that features symmetrically distributed punctate osteosclerosis due to heterozygous loss-of-function mutation within LEMD3. Rarely, radiographic findings of MEL occur in OPK. However, germline mutation of LEMD3 does not explain sporadic MEL. To explore if mosaicism underlies MEL, we studied a boy with polyostotic MEL and characteristic overlying scleroderma-like skin, a few bony lesions consistent with OPK, and a large epidermal nevus known to usually harbor a HRAS, FGFR3, or PIK3CA gene mutation. Exome sequencing was performed to ~100× average read depth for his two dermatoses, two areas of normal skin, and peripheral blood leukocytes. As expected for non-malignant tissues, the patient's mutation burden in his normal skin and leukocytes was low. He, his mother, and his maternal grandfather carried a heterozygous, germline, in-frame, 24-base-pair deletion in LEMD3. Radiographs of the patient and his mother revealed bony foci consistent with OPK, but she showed no MEL. For the patient, somatic variant analysis, using four algorithms to compare all 20 possible pairwise combinations of his five DNA samples, identified only one high-confidence mutation, heterozygous KRAS Q61H (NM_033360.3:c.183A>C, NP_203524.1:p.Gln61His), in both his dermatoses but absent in his normal skin and blood. Thus, sparing our patient biopsy of his MEL bone, we identified a heterozygous somatic KRAS mutation in his scleroderma-like dermatosis considered a surrogate for MEL. This implicates postzygotic mosaicism of mutated KRAS, perhaps facilitated by germline LEMD3 haploinsufficiency, causing his MEL.
- Published
- 2017
- Full Text
- View/download PDF
15. SAT-LB085 First Report of Burosumab (Anti-FGF23 Monoclonal Antibody) for Rickets Complicating HRAS-Associated Cutaneous Skeletal Hypophosphatemia Syndrome
- Author
-
Gary S. Gottesman, Ana Maria Arbelaez, William H. McAlister, Susan J. Bayliss, Michael P. Whyte, Marwan Shinawi, Pamela S Smith, and Jeffrey L Sugarman
- Subjects
business.industry ,medicine.drug_class ,Bone and Mineral Metabolism ,Endocrinology, Diabetes and Metabolism ,Immunology ,medicine ,Rickets ,HRAS ,Hypercalcemia and Complications of Treatment ,medicine.disease ,business ,Monoclonal antibody ,Hypophosphatemia - Abstract
Background: Cutaneous skeletal hypophosphatemia syndrome (CSHS) is caused by somatic mosaicism of a RAS family gene (ie. HRAS, NRAS, KRAS). CSHS features: 1) congenital epidermal, melanocytic, or sebaceous nevi, 2) elevated circulating FGF23 levels that cause renal phosphate wasting and skeletal hypomineralization, and 3) focal bone lesions ipsilateral to the nevi as a possible source of excess FGF23. Conventional therapy for rickets in CSHS (phosphate salts and bioactive vitamin D) mirrors treatment for X-linked hypophosphatemia (XLH) with elevated circulating FGF23. Current therapy aims to treat signs and symptoms rather than the etiology. Clinical Case: A 21-month-old boy with left, widespread, unilateral nevus sebaceous developed bilateral lower extremity bowing with a wide-based gait. His serum phosphorus level was low at 2.1 mg/dl (Nl: 3-6) and FGF23 was elevated at 279 RU/mL (Nl: < 230). We prescribed conventional therapy for hypophosphatemic rickets and diagnosed linear sebaceous nevus syndrome, confirmed by mutation analysis of affected skin harboring HRAS p.Q61R (c.182A>G). Adherence to medical therapy was problematic, and he sustained 3 major long-bone fractures. By age 6 years, a walker was required to ambulate short distances with mostly wheelchair-dependence from pain and muscle weakness. Based on his elevated FGF23 level and clinical decline, burosumab (anti-FGF23 fully human monoclonal antibody), FDA-approved in 2018 for XLH, was initiated off-label. After weaning conventional therapy, burosumab was administered subcutaneously as a single dose of 0.8 mg/kg (20 mg) and decreased to 0.3 mg/kg (6 mg) two weeks later. Burosumab was then given every 2 weeks and adjusted by 0.1 mg/kg to maintain fasting serum phosphorus in the low-normal range for age (3.5 - 4.5 mg/dL). Laboratory and clinic follow-up occurred just prior to each injection for 3 months until serum phosphorus was stable as he received approximately 0.4 mg/kg (8 mg per dose). Biochemical testing and clinic visits will occur monthly for 3 months and are planned for quarterly afterwards, with repeat radiological imaging every 3-6 months for the first year of therapy. Duration of burosumab treatment has been 5 months. After his initial burosumab dose, serum phosphorus increased from 1.7 to 3.7 mg/dL and remained normal [Nl: 3.0 - 6.0 mg/dL] thereafter. Radiographs of his wrists and knees 3 months into therapy revealed striking improvement of his rickets but persisting focal bone lesions, characteristic of CSHS, on the left side. He now walks independently with endurance, strength, and resolving pain. Quality-of-life is markedly improved. Burosumab treatment is being tolerated well without side effects. Conclusion: Burosumab seems to be an effective therapy for FGF23-mediated rickets in pediatric CSHS. Unless otherwise noted, all abstracts presented at ENDO are embargoed until the date and time of presentation. For oral presentations, the abstracts are embargoed until the session begins. Abstracts presented at a news conference are embargoed until the date and time of the news conference. The Endocrine Society reserves the right to lift the embargo on specific abstracts that are selected for promotion prior to or during ENDO.
- Published
- 2019
- Full Text
- View/download PDF
16. New explanation for autosomal dominant high bone mass: Mutation of low-density lipoprotein receptor-related protein 6
- Author
-
Steven Mumm, Fan Zhang, Michael P. Whyte, Margaret Huskey, Vinieth N. Bijanki, Shenghui Duan, William H. McAlister, Gary S. Gottesman, Kathryn Dahir, Angela Nenninger, and Elizabeth L. Lin
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Frizzled ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,DNA Mutational Analysis ,030209 endocrinology & metabolism ,Biology ,medicine.disease_cause ,Bone and Bones ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Absorptiometry, Photon ,Skeletal disorder ,Bone Density ,Internal medicine ,medicine ,Missense mutation ,Humans ,Amino Acid Sequence ,Genes, Dominant ,Mutation ,Hip ,Base Sequence ,Wnt signaling pathway ,Age Factors ,LRP6 ,LRP5 ,Organ Size ,Body Height ,Spine ,Pedigree ,030104 developmental biology ,Endocrinology ,Low Density Lipoprotein Receptor-Related Protein-5 ,chemistry ,Low Density Lipoprotein Receptor-Related Protein-6 ,Sclerostin ,Female ,Tomography, X-Ray Computed - Abstract
LRP5 encodes low-density lipoprotein receptor-related protein 5 (LRP5). When LRP5 with a Frizzled receptor join on the surface of an osteoblast and bind a member of the Wnt family of ligands, canonical Wnt/β-catenin signaling occurs and increases bone formation. Eleven heterozygous gain-of-function missense mutations within LRP5 are known to prevent the LRP5 inhibitory ligands sclerostin and dickkopf1 from attaching to LRP5's first β-propeller, and thereby explain the rare autosomal dominant (AD) skeletal disorder "high bone mass" (HBM). LRP6 is a cognate co-receptor of LRP5 and similarly controls Wnt signaling in osteoblasts, yet the consequences of increased LRP6-mediated signaling remain unknown. We investigated two multi-generational American families manifesting the clinical and routine laboratory features of LRP5 HBM but without an LRP5 defect and instead carrying a heterozygous LRP6 missense mutation that would alter the first β-propeller of LRP6. In Family 1 LRP6 c.602CT, p.A201V was homologous to LRP5 HBM mutation c.641CT, p.A214V, and in Family 2 LRP6 c.553AC, p.N185H was homologous to LRP5 HBM mutation c.593AG, p.N198S but predicting a different residue at the identical amino acid position. In both families the LRP6 mutation co-segregated with striking generalized osteosclerosis and hyperostosis. Clinical features shared by the seven LRP6 HBM family members and ten LRP5 HBM patients included a broad jaw, torus palatinus, teeth encased in bone and, reportedly, resistance to fracturing and inability to float in water. For both HBM disorders, all affected individuals were taller than average for Americans (Ps 0.005), but with similar mean height Z-scores (P = 0.7606) and indistinguishable radiographic skeletal features. Absence of adult maxillary lateral incisors was reported by some LRP6 HBM individuals. In contrast, our 16 patients with AD osteopetrosis [i.e., Albers-Schönberg disease (A-SD)] had an unremarkable mean height Z-score (P = 0.9401) lower than for either HBM group (Ps 0.05). DXA mean BMD Z-scores in LRP6 HBM versus LRP5 HBM were somewhat higher at the lumbar spine (+7.8 vs +6.5, respectively; P = 0.0403), but no different at the total hip (+7.9 vs +7.7, respectively; P = 0.7905). Among the three diagnostic groups, only the LRP6 HBM DXA BMD values at the spine seemed to increase with subject age (R = +0.7183, P = 0.0448). Total hip BMD Z-scores were not significantly different among the three disorders (Ps 0.05), and showed no age effect (Ps 0.1). HR-pQCT available only for LRP6 HBM revealed indistinct corticomedullary boundaries, high distal forearm and tibial total volumetric BMD, and finite element analysis predicted marked fracture resistance. Hence, we have discovered mutations of LRP6 that cause a dento-osseous disorder indistinguishable without mutation analysis from LRP5 HBM. LRP6 HBM seems associated with generally good health, providing some reassurance for the development of anabolic treatments aimed to enhance LRP5/LRP6-mediated osteogenesis.
- Published
- 2019
17. Non-endemic skeletal fluorosis: Causes and associated secondary hyperparathyroidism (case report and literature review)
- Author
-
Vinieth N. Bijanki, Maighan Seagrove-Guffey, Fiona J. Cook, Steven Mumm, Michael P. Whyte, Deborah Wenkert, William H. McAlister, and Deborah J. Veis
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,Parathyroid hormone ,030209 endocrinology & metabolism ,Article ,Bone remodeling ,03 medical and health sciences ,Osteosclerosis ,Skeletal fluorosis ,0302 clinical medicine ,Bone Density ,Internal medicine ,medicine ,Humans ,Hyperparathyroidism ,Osteoid ,business.industry ,Middle Aged ,medicine.disease ,Spine ,030104 developmental biology ,Endocrinology ,Parathyroid Hormone ,Hyperparathyroidism, Secondary ,Secondary hyperparathyroidism ,Bone Diseases ,business - Abstract
Skeletal fluorosis (SF) is endemic primarily in regions with fluoride (F)-contaminated well water, but can reflect other types of chronic F exposure. Calcium (Ca) and vitamin D (D) deficiency can exacerbate SF. A 51-year-old man with years of musculoskeletal pain and opiate use was hypocalcemic with secondary hyperparathyroidism upon manifesting recurrent long bone fractures. He smoked cigarettes, drank large amounts of cola beverage, and consumed little dietary Ca. Then, after 5 months of Ca and D(3) supplementation, serum 25(OH)D was 21 ng/mL (Nl, 30–100), corrected serum Ca had normalized from 7.8 to 9.4 mg/dL (Nl, 8.5–10.1), alkaline phosphatase (ALP) had decreased from 1080 to 539 U/L (Nl, 46–116), yet parathyroid hormone (PTH) had increased from 133 to 327 pg/mL (Nl, 8.7–77.1). Radiographs revealed generalized osteosclerosis and a cystic osteopenic area in the left femoral neck and intertrochanteric region. DXA BMD Z-scores were +7.4 and +0.4 at the lumbar spine and “1/3” radius, respectively. Bone scintigraphy showed increased uptake in two ribs, periarticular areas, and proximal left femur at the site of a subsequent atraumatic fracture. Elevated serum collagen type I C-telopeptide 2513 pg/mL (Nl, 87–345) and osteocalcin >300 ng/mL (Nl, 9–38) indicated rapid bone turnover. Negative studies included hepatitis C Ab, prostate-specific antigen, serum and urine electrophoresis, and Ion Torrent mutation analysis for dense or high-turnover skeletal diseases. After discovering markedly elevated F concentrations in his plasma [4.84 mg/L (Nl, 0.02–0.08)] and spot urine [42.6 mg/L (Nl, 0.2–3.2)], a two-year history emerged of “huffing” computer cleaner containing difluoroethane. Non-decalcified histology of a subsequent right femur fracture showed increased osteoblasts and osteoclasts and excessive osteoid. A 24-hour urine collection contained 27 mg/L F (Nl, 0.2–3.2) and < 2 mg/dL Ca. Then, 19 months after “huffing” cessation and improved Ca and D(3) intake, yet with persisting bone pain, serum PTH was normal (52 pg/mL) and serum ALP and urine F had decreased to 248 U/L and 3.3 mg/L, respectively. Our experience combined with 15 publications in PubMed concerning unusual causes of non-endemic SF where the F source became known (19 cases in all) revealed: 11 instances from high consumption of black tea and/or F-containing toothpaste, 1 due to geophagia of F-rich soil, and 7 due to “recreational” inhalation of F-containing vapors. Circulating PTH measured in 13 was substantially elevated in 2 (including ours) and mildly increased in 2. Their SF severity, including bone turnover rate, seemed to reflect cumulative F exposure, renal function, and Ca and D status. Several factors appeared to condition our patient’s skeletal disease: i) direct anabolic effects of toxic amounts of F on his skeleton, ii) secondary hyperparathyroidism from degradation-resistant fluorapatite bone crystals and low dietary Ca, and iii) impaired mineralization of excessive osteoid due to hypocalcemia.
- Published
- 2021
- Full Text
- View/download PDF
18. Raine Syndrome (OMIM #259775), Caused ByFAM20CMutation, Is Congenital Sclerosing Osteomalacia With Cerebral Calcification (OMIM 259660)
- Author
-
Vinieth N. Bijanki, Steven Mumm, Michael P. Whyte, William H. McAlister, Ghada A. Otaify, M. Fallon, Shenghui Duan, Mary Ella M Pierpont, and William S. Sly
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Osteomalacia ,Cerebral calcification ,business.industry ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Osteopetrosis ,Trigonocephaly ,Raine syndrome ,Compound heterozygosity ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Internal medicine ,OMIM : Online Mendelian Inheritance in Man ,Medicine ,Missense mutation ,Orthopedics and Sports Medicine ,business - Abstract
In 1985, we briefly reported infant sisters with a unique, lethal, autosomal recessive disorder designated congenital sclerosing osteomalacia with cerebral calcification. In 1986, this condition was entered into Mendelian Inheritance In Man (MIM) as osteomalacia, sclerosing, with cerebral calcification (MIM 259660). However, no attestations followed. Instead, in 1989 Raine and colleagues published an affected neonate considering unprecedented the striking clinical and radiographic features. In 1992, “Raine syndrome” entered MIM formally as osteosclerotic bone dysplasia, lethal (MIM #259775). In 2007, the etiology emerged as loss-of-function mutation of FAM20C that encodes family with sequence similarity 20, member C. FAM20C is highly expressed in embryonic calcified tissues and encodes a kinase (dentin matrix protein 4) for most of the secreted phosphoproteome including FGF23, osteopontin, and other regulators of skeletal mineralization. Herein, we detail the clinical, radiological, biochemical, histopathological, and FAM20C findings of our patients. Following premortem tetracycline labeling, the proposita's non-decalcified skeletal histopathology after autopsy indicated no rickets but documented severe osteomalacia. Archival DNA revealed the sisters were compound heterozygotes for a unique missense mutation and a novel deletion in FAM20C. Individuals heterozygous for the missense mutation seemed to prematurely fuse their metopic suture and develop a metopic ridge sometimes including trigonocephaly. Our findings clarify FAM20C's role in hard tissue formation and mineralization, and show that Raine syndrome is congenital sclerosing osteomalacia with cerebral calcification. © 2016 American Society for Bone and Mineral Research.
- Published
- 2016
- Full Text
- View/download PDF
19. Auricular ossification: A newly recognized feature of osteoprotegerin-deficiency juvenile Paget disease
- Author
-
Steven Mumm, Michael P. Whyte, Angela Nenninger, Deborah Wenkert, William H. McAlister, Katherine L Madson, and Gary S. Gottesman
- Subjects
0301 basic medicine ,Ossification ,business.industry ,Osteitis Deformans ,030209 endocrinology & metabolism ,Elastic cartilage ,Anatomy ,medicine.disease ,Bone remodeling ,03 medical and health sciences ,Ectopic calcification ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Osteoprotegerin ,Genetics ,medicine ,Deformity ,medicine.symptom ,business ,Genetics (clinical) ,Calcification - Abstract
We report auricular ossification (AO) affecting the elastic cartilage of the ear as a newly recognized feature of osteoprotegerin (OPG)-deficiency juvenile Paget disease (JPD). AO and auricular calcification refer interchangeably to rigid pinnae, sparing the ear lobe, from various etiologies. JPD is a rare Mendelian disorder characterized by elevated serum alkaline phosphatase activity accompanied by skeletal pain and deformity from rapid bone turnover. Autosomal recessive transmission of loss-of-function mutations within TNFRSF11B encoding OPG accounts for most JPD (JPD1). JPD2 results from heterozygous constitutive activation of TNFRSF11A encoding RANK. Other causes of JPD remain unknown. In 2007, we reported a 60-year-old man with JPD1 who described hardening of his external ears at age 45 years, after 4 years of treatment with bisphosphonates (BPs). Subsequently, we noted rigid pinnae in a 17-year-old boy and 14-year-old girl, yet pliable pinnae in a 12-year-old boy, each with JPD1 and several years of BP treatment. Cranial imaging indicated cortical bone within the pinnae of both teenagers. Radiologic studies of our three JPD patients without mutations in TNFRSF11B showed normal auricles. Review of the JPD literature revealed possible AO in several reports. Two of our JPD1 patients had experienced difficult tracheal intubation, raising concern for mineralization of laryngeal elastic cartilage. Thus, AO is a newly recognized feature of JPD1, possibly exacerbated by BP treatment. Elastic cartilage at other sites in JPD1 might also ossify, and warrants investigation.
- Published
- 2016
- Full Text
- View/download PDF
20. Hypophosphatemic osteosclerosis, hyperostosis, and enthesopathy associated with novel homozygous mutations of DMP1 encoding dentin matrix protein 1 and SPP1 encoding osteopontin: The first digenic SIBLING protein osteopathy?
- Author
-
Michael P. Whyte, Vinieth N. Bijanki, Deborah J. Veis, Shenghui Duan, Margaret Huskey, William H. McAlister, Suhas Alur, S. Deepak Amalnath, Marc D. McKee, and Steven Mumm
- Subjects
0301 basic medicine ,Bone sialoprotein ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,India ,030209 endocrinology & metabolism ,Enthesopathy ,Biology ,Article ,03 medical and health sciences ,Osteosclerosis ,0302 clinical medicine ,stomatognathic system ,Dentin sialophosphoprotein ,medicine ,Humans ,Missense mutation ,Osteopontin ,Genetics ,Extracellular Matrix Proteins ,Hyperostosis ,Middle Aged ,Phosphoproteins ,Pseudoxanthoma elasticum ,medicine.disease ,DMP1 ,3. Good health ,Fibroblast Growth Factor-23 ,030104 developmental biology ,Dentin ,Mutation ,MEPE ,biology.protein ,Female - Abstract
The SIBLINGs are a subfamily of the secreted calcium-binding phosphoproteins and comprise five small integrin-binding ligand N-linked glycoproteins [dentin matrix protein-1 (DMP1), secreted phosphoprotein-1 (SPP1) also called osteopontin (OPN), integrin-binding sialoprotein (IBSP) also called bone sialoprotein (BPS), matrix extracellular phosphoglycoprotein (MEPE), and dentin sialophosphoprotein (DSPP)]. Each SIBLING has at least one “acidic, serine- and aspartic acid-rich motif’ (ASARM) and multiple Ser-x-Glu/pSer sequences that when phosphorylated promote binding of the protein to hydroxyapatite for regulation of biomineralization. Mendelian disorders from loss-of-function mutation(s) of the genes that encode the SIBLINGs thus far involve DSPP causing various autosomal dominant dysplasias of dentin but without skeletal disease, and DMP1 causing autosomal recessive hypophosphatemic rickets, type 1 (ARHR1). No diseases have been reported from gain-of-function mutation(s) of DSPP or DMP1 or from alterations of SPP1, IBSP, or MEPE. Herein, we describe severe hypophosphatemic osteosclerosis and hyperostosis associated with skeletal deformity, short stature, enthesopathy, tooth loss, and high circulating FGF23 levels in a middle-aged man and young woman from an endogamous family living in southern India. Both shared novel homozygous mutations within two genes that encode a SIBLING protein: stop-gain (“non-sense”) DMP1 (c.556G>T, p.Glu186Ter) and missense SPP1 (c.769C>T, p.Leu266Phe). The man alone also carried novel heterozygous missense variants within two additional genes that condition mineral homeostasis and are the basis for autosomal recessive disorders: CYP27B1 underlying vitamin D dependent rickets, type 1, and ABCC6 underlying both generalized arterial calcification of infancy, type 2 and pseudoxanthoma elasticum (PXE). By immunochemistry, his bone contained high amounts of OPN, particularly striking surrounding osteocytes. We review how our patients’ disorder may represent the first digenic SIBLING protein osteopathy.
- Published
- 2020
- Full Text
- View/download PDF
21. Phenotype and response to growth hormone therapy in siblings with B4GALT7 deficiency
- Author
-
Marwan Shinawi, William H. McAlister, Jennifer A. Wambach, F. Sessions Cole, Carla Sandler-Wilson, Daniel J. Wegner, and Bess A. Marshall
- Subjects
0301 basic medicine ,Adult ,Male ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Growth hormone ,Article ,Glycosaminoglycan ,03 medical and health sciences ,0302 clinical medicine ,Exome Sequencing ,Humans ,B4GALT7 ,Child ,Exome sequencing ,Genetics ,biology ,Siblings ,Galactosyltransferases ,Phenotype ,Growth hormone treatment ,030104 developmental biology ,Proteoglycan ,Child, Preschool ,Growth Hormone ,biology.protein ,Female ,Biomarkers - Abstract
B4GALT7 encodes beta-1,4-galactosyltransferase which links glycosaminoglycans to proteoglycans in connective tissues. Rare, biallelic variants in B4GALT7 have been associated with spondylodysplastic Ehlers-Danlos and Larsen of Reunion Island syndromes. Thirty patients with B4GALT7-related disorders have been reported to date with phenotypic variability. Using whole exome sequencing, we identified male and female siblings with biallelic, pathogenic B4GALT7 variants and phenotypic features of spondylodysplastic Ehlers-Danlos syndrome as well as previously unreported skeletal characteristics. We also provide detailed radiological characterization and describe the siblings’ responses to growth hormone treatment. Our report extends the phenotypic spectrum of B4GALT7-associated spondylodysplastic Ehlers- Danlos syndrome and reports results of growth hormone treatment for patients with this rare disorder.
- Published
- 2018
22. No vascular calcification on cardiac computed tomography spanning asfotase alfa treatment for an elderly woman with hypophosphatasia
- Author
-
Andrew J. Bierhals, Steven Mumm, Michael P. Whyte, and William H. McAlister
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Recombinant Fusion Proteins ,Hypophosphatasia ,030209 endocrinology & metabolism ,Rickets ,Generalized arterial calcification ,03 medical and health sciences ,Ectopic calcification ,0302 clinical medicine ,Absorptiometry, Photon ,Internal medicine ,medicine ,Humans ,Vascular Calcification ,Aorta ,Aged ,Osteomalacia ,business.industry ,Myocardium ,ALPL ,medicine.disease ,Pseudoxanthoma elasticum ,Alkaline Phosphatase ,030104 developmental biology ,Endocrinology ,Asfotase alfa ,Immunoglobulin G ,Mutation ,Calcium ,Female ,business ,Tomography, X-Ray Computed - Abstract
Hypophosphatasia (HPP) is the inborn-error-of-metabolism characterized enzymatically by insufficient activity of the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP) and caused by either mono- or bi-allelic loss-of-function mutation(s) of the gene ALPL that encodes this cell surface phosphomonoester phosphohydrolase. In HPP, the natural substrates of TNSALP accumulate extracellularly and include inorganic pyrophosphate (PPi), a potent inhibitor of biomineralization. This PPi excess leads to rickets or osteomalacia in all but the most mild “odonto” form of the disease. Adults with HPP understandably often also manifest calcium PPi dihydrate deposition, whereas enthesopathy and calcific periarthritis from hydroxyapatite (HA) crystal deposition can seem paradoxical in face of the defective skeletal mineralization. In 2015, asfotase alfa (AA), a HA-targeted TNSALP, was approved multinationally as an enzyme replacement therapy for HPP. AA hydrolyzes extracellular PPi (ePPi) and in HPP enables HA crystals to grow and mineralize skeletal matrix. In direct contrast to HPP, deficiency of ePPi characterizes the inborn-errors-of-metabolism generalized arterial calcification of infancy (GACI) and pseudoxanthoma elasticum (PXE). In GACI and PXE, deficiency of ePPi leads to ectopic mineralization including vascular calcification (VC). Therefore, in HPP, ectopic mineralization including VC could hypothetically result from, or be exacerbated by, the persistently high circulating TNSALP activity that occurs during AA treatment. Herein, using a routine computed tomography (CT) method to quantitate coronary artery calcium, we found no ectopic mineralization in the heart of an elderly woman with HPP before or after 8 months of AA treatment. Subsequently, investigational high-resolution peripheral quantitative CT and dual-energy X-ray absorptiometry showed absence of peripheral artery and aortic calcium after further AA treatment. Investigation of additional adults with HPP could reveal if the superabundance of ePPi protects against VC, and whether long-term AA therapy causes or exacerbates any ectopic mineralization.
- Published
- 2018
23. Absence of an osteopetrosis phenotype in IKBKG (NEMO) mutation-positive women: A case-control study
- Author
-
Weimin Qui, Christina Møller Andreasen, Deborah J. Veis, Allahdad Zarei, Michaela Tencerova, Moustapha Kassem, Dea Svaneby, Michael P. Whyte, William H. McAlister, Morten Frost, Charlotte Ejersted, Thomas Levin Andersen, and Anja Lisbeth Frederiksen
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,IKBKG ,030209 endocrinology & metabolism ,NF-κB ,Article ,Bone remodeling ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Incontinentia pigmenti ,NEMO ,Internal medicine ,Biopsy ,medicine ,Humans ,Immunodeficiency ,Aged ,medicine.diagnostic_test ,business.industry ,X-chromosome inactivation ,Osteopetrosis ,Middle Aged ,medicine.disease ,I-kappa B Kinase ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cross-Sectional Studies ,chemistry ,Case-Control Studies ,Mutation ,Female ,Bone marrow ,business - Abstract
Background: NF-κB essential modulator (NEMO), encoded by IKBKG, is necessary for activation of the ubiquitous transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Animal studies suggest NEMO is required for NF-κB mediated bone homeostasis, but this has not been thoroughly studied in humans. IKBKG loss-of-function mutation causes incontinentia pigmenti (IP), a rare X-linked disease featuring linear hypopigmentation, alopecia, hypodontia, and immunodeficiency. Single case reports describe osteopetrosis (OPT) in boys carrying hypomorphic IKBKG mutations. Method: We studied the bone phenotype in women with IP with evaluation of radiographs of the spine and non-dominant arm and leg; lumbar spine and femoral neck aBMD using DXA; μ-CT and histomorphometry of trans-iliac crest biopsy specimens; bone turnover markers; and cellular phenotype in bone marrow skeletal (stromal) stem cells (BM-MSCs) in a cross-sectional, age-, sex-, and BMI-matched case-control study. X-chromosome inactivation was measured in blood leucocytes and BM-MSCs using a PCR method with methylation of HpaII sites. NF-κB activity was quantitated in BM-MSCs using a luciferase NF-κB reporter assay. Results: Seven Caucasian women with IP (age: 24–67 years and BMI: 20.0–35.2 kg/m 2 ) and IKBKG mutation (del exon 4–10 (n = 4); c.460C>T (n = 3)) were compared to matched controls. The IKBKG mutation carriers had extremely skewed X-inactivation (>90:10%) in blood, but not in BM-MSCs. NF-κB activity was lower in BM-MSCs from IKBKG mutation carriers (n = 5) compared to controls (3094 ± 679 vs. 5422 ± 1038/μg protein, p < 0.01). However, no differences were identified on skeletal radiographics, aBMD, μ-architecture of the iliac crest, or bone turnover markers. The IKBKG mutation carriers had a 1.7–fold greater extent of eroded surfaces relative to osteoid surfaces (p < 0.01), and a 2.0–fold greater proportion of arrested reversal surface relative to active reversal surface (p < 0.01). Conclusion: Unlike mutation-positive males, the IKBKG mutation-positive women did not manifest OPT.
- Published
- 2018
- Full Text
- View/download PDF
24. Sclerosteosis: Report of type 1 or 2 in three Indian Tamil families and literature review
- Author
-
Margaret Huskey, Radhakrishna Pedapati, Vinieth N. Bijanki, Steven Mumm, Michael P. Whyte, Vivekanandan Muthupillai, Shenghui Duan, William H. McAlister, and S. Deepak Amalnath
- Subjects
0301 basic medicine ,Genetic Markers ,Male ,Histology ,Adolescent ,Physiology ,Endocrinology, Diabetes and Metabolism ,Population ,DNA Mutational Analysis ,India ,Consanguinity ,Biology ,medicine.disease_cause ,Bone and Bones ,03 medical and health sciences ,chemistry.chemical_compound ,Osteosclerosis ,medicine ,Missense mutation ,Humans ,Family ,Syndactyly ,education ,LDL-Receptor Related Proteins ,Adaptor Proteins, Signal Transducing ,Genetics ,education.field_of_study ,Mutation ,Minerals ,Base Sequence ,Osteopetrosis ,Hyperostosis ,Middle Aged ,medicine.disease ,Pedigree ,030104 developmental biology ,chemistry ,Bone Morphogenetic Proteins ,Sclerostin ,Female - Abstract
Sclerosteosis (SOST) refers to two extremely rare yet similar skeletal dysplasias featuring a diffusely radiodense skeleton together with congenital syndactyly. SOST1 is transmitted as an autosomal recessive (AR) trait and to date caused by ten homozygous loss-of-function mutations within the gene SOST that encodes the inhibitor of Wnt-mediated bone formation, sclerostin. SOST2 is transmitted as an autosomal dominant (AD) or AR trait and to date caused by one heterozygous or two homozygous loss-of-function mutation(s), respectively, within the gene LRP4 that encodes the sclerostin interaction protein, low-density lipoprotein receptor-related protein 4 (LRP4). Herein, we investigated two teenagers and one middle-aged man with SOST in three families living in the state of Tamil Nadu in southern India. Next generation sequencing of their genomic DNA using our high bone density gene panel revealed SOST1 in the teenagers caused by a unique homozygous nonsense SOST mutation (c.129C > G, p.Tyr43X) and SOST2 in the man caused by homozygosity for one of the two known homozygous missense LRP4 mutations (c.3508C > T, p.Arg1170Trp). He becomes the fourth individual and the first non-European recognized with SOST2. His clinical course was milder than the life-threatening SOST1 demonstrated by the teenagers who suffered blindness, deafness, and raised intracranial pressure, yet his congenital syndactyly was more striking by featuring bony fusion of digits. All three patients were from consanguineous families and heterozygosity for the SOST mutation was documented in the mothers of both teenagers. Thus, on the endogamous genetic background of Indian Tamils, SOST1 from sclerostin deficiency compared to SOST2 from LRP4 deactivation is a more severe and life-threatening disorder featuring complications due to osteosclerosis of especially the skull. In contrast, the syndactyly of SOST2 is particularly striking by involving bony fusion of some digits. Both the SOST and LRP4 mutations in this ethnic population likely reflect genetic founders.
- Published
- 2018
25. Neonatal High Bone Mass With First Mutation of the NF-κB Complex: Heterozygous De Novo Missense (p.Asp512Ser)RELA(Rela/p65)
- Author
-
Steven Mumm, Michael P. Whyte, Deborah V. Novack, William H. McAlister, Moustapha Kassem, Klaus Brusgaard, Peter Juel Thiis Knudsen, Christina Eckhardt, Henrik Daa Schrøder, Martin Jakob Larsen, Anja Lisbeth Frederiksen, Weimin Qiu, and Morten Frost
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Mutation ,Endocrinology, Diabetes and Metabolism ,Osteopetrosis ,Biology ,medicine.disease ,medicine.disease_cause ,Bone resorption ,Bone remodeling ,03 medical and health sciences ,Osteosclerosis ,030104 developmental biology ,Endocrinology ,Internal medicine ,medicine ,Mutation testing ,Cancer research ,Missense mutation ,Orthopedics and Sports Medicine ,Exome sequencing - Abstract
Heritable disorders that feature high bone mass (HBM) are rare. The etiology is typically a mutation(s) within a gene that regulates the differentiation and function of osteoblasts (OBs) or osteoclasts (OCs). Nevertheless, the molecular basis is unknown for approximately one-fifth of such entities. NF-κB signaling is a key regulator of bone remodeling and acts by enhancing OC survival while impairing OB maturation and function. The NF-κB transcription complex comprises five subunits. In mice, deletion of the p50 and p52 subunits together causes osteopetrosis (OPT). In humans, however, mutations within the genes that encode the NF-κB complex, including the Rela/p65 subunit, have not been reported. We describe a neonate who died suddenly and unexpectedly and was found at postmortem to have HBM documented radiographically and by skeletal histopathology. Serum was not available for study. Radiographic changes resembled malignant OPT, but histopathological investigation showed morphologically normal OCs and evidence of intact bone resorption excluding OPT. Furthermore, mutation analysis was negative for eight genes associated with OPT or HBM. Instead, accelerated bone formation appeared to account for the HBM. Subsequently, trio-based whole exome sequencing revealed a heterozygous de novo missense mutation (c.1534_1535delinsAG, p.Asp512Ser) in exon 11 of RELA encoding Rela/p65. The mutation was then verified using bidirectional Sanger sequencing. Lipopolysaccharide stimulation of patient fibroblasts elicited impaired NF-κB responses compared with healthy control fibroblasts. Five unrelated patients with unexplained HBM did not show a RELA defect. Ours is apparently the first report of a mutation within the NF-κB complex in humans. The missense change is associated with neonatal osteosclerosis from in utero increased OB function rather than failed OC action. These findings demonstrate the importance of the Rela/p65 subunit within the NF-κB pathway for human skeletal homeostasis and represent a new genetic cause of HBM. © 2015 American Society for Bone and Mineral Research.
- Published
- 2015
- Full Text
- View/download PDF
26. Lenz-Majewski Hyperostotic Dwarfism with Hyperphosphoserinuria from a Novel Mutation inPTDSS1Encoding Phosphatidylserine Synthase 1
- Author
-
Steven Mumm, Michael P. Whyte, Vinieth N. Bijanki, Angela Nenninger, William H. McAlister, and Amanda Blythe
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Dysostosis ,Dwarfism ,Osteopetrosis ,Phosphatidylserine ,Biology ,medicine.disease ,chemistry.chemical_compound ,Osteosclerosis ,Endocrinology ,chemistry ,Internal medicine ,Phosphoserine ,medicine ,Missense mutation ,Orthopedics and Sports Medicine ,Mesenchymal stem cell differentiation - Abstract
Lenz-Majewski hyperostotic dwarfism (LMHD) is an ultra-rare Mendelian craniotubular dysostosis that causes skeletal dysmorphism and widely distributed osteosclerosis. Biochemical and histopathological characterization of the bone disease is incomplete and nonexistent, respectively. In 2014, a publication concerning five unrelated patients with LMHD disclosed that all carried one of three heterozygous missense mutations in PTDSS1 encoding phosphatidylserine synthase 1 (PSS1). PSS1 promotes the biosynthesis of phosphatidylserine (PTDS), which is a functional constituent of lipid bilayers. In vitro, these PTDSS1 mutations were gain-of-function and increased PTDS production. Notably, PTDS binds calcium within matrix vesicles to engender hydroxyapatite crystal formation, and may enhance mesenchymal stem cell differentiation leading to osteogenesis. We report an infant girl with LMHD and a novel heterozygous missense mutation (c.829T>C, p.Trp277Arg) within PTDSS1. Bone turnover markers suggested that her osteosclerosis resulted from accelerated formation with an unremarkable rate of resorption. Urinary amino acid quantitation revealed a greater than sixfold elevation of phosphoserine. Our findings affirm that PTDSS1 defects cause LMHD and support enhanced biosynthesis of PTDS in the pathogenesis of LMHD.
- Published
- 2015
- Full Text
- View/download PDF
27. Validation of a Novel Scoring System for Changes in Skeletal Manifestations of Hypophosphatasia in Newborns, Infants, and Children: The Radiographic Global Impression of Change Scale
- Author
-
Michael P, Whyte, Kenji P, Fujita, Scott, Moseley, David D, Thompson, and William H, McAlister
- Subjects
Male ,Recombinant Fusion Proteins ,Infant, Newborn ,Hypophosphatasia ,Infant ,Alkaline Phosphatase ,Bone and Bones ,Child, Preschool ,Immunoglobulin G ,Humans ,Enzyme Replacement Therapy ,Female ,Child ,Tomography, X-Ray Computed - Abstract
Hypophosphatasia (HPP) is the heritable metabolic disease characterized by impaired skeletal mineralization due to low activity of the tissue-nonspecific isoenzyme of alkaline phosphatase. Although HPP during growth often manifests with distinctive radiographic skeletal features, no validated method was available to quantify them, including changes over time. We created the Radiographic Global Impression of Change (RGI-C) scale to assess changes in the skeletal burden of pediatric HPP. Site-specific pairs of radiographs of newborns, infants, and children with HPP from three clinical studies of asfotase alfa, an enzyme replacement therapy for HPP, were obtained at baseline and during treatment. Each pair was scored by three pediatric radiologists ("raters"), with nine raters across the three studies. Intrarater and interrater agreement was determined by weighted Kappa coefficients. Interrater reliability was assessed using intraclass correlation coefficients (ICCs) and by two-way random effects analysis of variance (ANOVA) and a mixed-model repeated measures ANOVA. Pearson correlation coefficients evaluated relationships of the RGI-C to the Rickets Severity Scale (RSS), Pediatric Outcomes Data Collection Instrument Global Function Parent Normative Score, Childhood Health Assessment Questionnaire Disability Index, 6-Minute Walk Test percent predicted, and Z-score for height in patients aged 6 to 12 years at baseline. Eighty-nine percent (8/9) of raters showed substantial or almost perfect intrarater agreement of sequential RGI-C scores (weighted Kappa coefficients, 0.72 to 0.93) and moderate or substantial interrater agreement (weighted Kappa coefficients, 0.53 to 0.71) in patients aged 0 to 12 years at baseline. Moderate-to-good interrater reliability was observed (ICC, 0.57 to 0.65). RGI-C scores were significantly (p ≤ 0.0065) correlated with the RSS and with measures of global function, disability, endurance, and growth in the patients aged 6 to 12 years at baseline. Thus, the RGI-C is valid and reliable for detecting clinically important changes in skeletal manifestations of severe HPP in newborns, infants, and children, including during asfotase alfa treatment. © 2018 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
- Published
- 2017
28. Sustained radiographic and functional improvements with asfotase alfa treatment from up to 7 years in children with hypophosphatasia
- Author
-
Andrew Denker, Michael P. Whyte, Scott Moseley, Cheryl Rockman-Greenberg, and William H. McAlister
- Subjects
Pediatrics ,medicine.medical_specialty ,business.industry ,Radiography ,Asfotase alfa ,Hypophosphatasia ,Medicine ,General Medicine ,business ,medicine.disease - Published
- 2017
- Full Text
- View/download PDF
29. PHEX3′-UTR c.*231A>G Near The Polyadenylation Signal Is a Relatively Common, Mild, American Mutation That Masquerades as Sporadic or X-Linked Recessive Hypophosphatemic Rickets
- Author
-
Steven Mumm, Michael P. Whyte, Margaret Huskey, William H. McAlister, Fan Zhang, Gary S. Gottesman, Valerie Wollberg, Adela Cajic, Katherine L Madson, and Deborah Wenkert
- Subjects
Genetics ,medicine.medical_specialty ,Osteomalacia ,Mutation ,business.industry ,Three prime untranslated region ,Endocrinology, Diabetes and Metabolism ,PHEX ,Haplotype ,medicine.disease ,medicine.disease_cause ,Hypophosphatemic Rickets ,Endocrinology ,Internal medicine ,medicine ,Mutation testing ,Orthopedics and Sports Medicine ,business ,X-linked recessive inheritance - Abstract
Heritable forms of hypophosphatemic rickets (HR) include X-linked dominant (XLH), autosomal recessive, and autosomal dominant HR (from deactivating mutations in PHEX, DMP1 or ENPP1, and activating mutations in FGF23, respectively). Over 30 years, we have cared for 284 children with HR. For those 72 deemed sporadic XLH, we preliminarily reported mutation analysis for 30 subjects. Eleven had PHEX mutations. However, the remaining 19 lacked readily identifiable defects in PHEX, DMP1, or FGF23. In 2008, a novel single-base change near the polyadenylation (pA) signal in the 3′-UTR of PHEX was identified in XLH by other investigators. This c.*231A > G mutation is 3-bp upstream of the putative pA signal (AATAAA) in PHEX. Accordingly, we investigated whether this 3′-UTR defect accounted for HR in any of these 19 sporadic XLH patients. PCR amplification and sequencing of their 3′-UTR region showed the c.*231A > G mutation in four unrelated boys. Then, among an additional 22 of our 72 “sporadic” XLH patients, one boy and one girl were found to have the 3′-UTR defect, totaling six patients. Among these 52 sporadic XLH patients with PHEX analysis, 36 were girls and 16 were boys; ie, a ∼2:1 gender ratio consistent with XLH. However, finding five boys and only one girl with this 3′-UTR mutation presented an unexplained gender bias (p = 0.02). Haplotyping for the five boys, all reportedly unrelated, showed a common core haplotype suggesting a founder. Five of their six mothers had been studied clinically and biochemically (three radiologically). Remarkably, the seemingly unaffected mothers of four of these boys carried the 3′-UTR mutation. These healthy women had normal height, straight limbs, lacked the radiographic presentation of XLH, and showed normal or slight decreases in fasting serum Pi levels and/or TmP/GFR. Hence, PHEX c.*231A > G can masquerade as sporadic or X-linked recessive HR. © 2014 American Society for Bone and Mineral Research.
- Published
- 2014
- Full Text
- View/download PDF
30. Rapid Skeletal Turnover in a Radiographic Mimic of Osteopetrosis
- Author
-
Deborah V. Novack, Steven Mumm, Michael P. Whyte, William H. McAlister, Jo Blair, Marina Stolina, Nicholas Shaw, Laurence Abernethy, Timothy R. Helliwell, and Katherine L Madson
- Subjects
musculoskeletal diseases ,Bone growth ,Bone mineral ,medicine.medical_specialty ,Pathology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,Osteopetrosis ,medicine.disease ,Bone remodeling ,chemistry.chemical_compound ,Osteosclerosis ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Osteoclast ,Internal medicine ,medicine ,Sclerostin ,Orthopedics and Sports Medicine ,business - Abstract
Among the high bone mass disorders, the osteopetroses reflect osteoclast failure that prevents skeletal resorption and turnover, leading to reduced bone growth and modeling and characteristic histopathological and radiographic findings. We report an 11-year-old boy with a new syndrome that radiographically mimics osteopetrosis (OPT), but features rapid skeletal turnover. He presented at age 21 months with a parasellar, osteoclast-rich giant cell granuloma. Radiographs showed a dense skull, generalized osteosclerosis and cortical thickening, medullary cavity narrowing, and diminished modeling of tubular bones. His serum alkaline phosphatase was >5000 IU/L (normal
- Published
- 2014
- Full Text
- View/download PDF
31. Multicentric carpotarsal osteolysis syndrome is caused by only a few domain-specific mutations inMAFB, a negative regulator of RANKL-induced osteoclastogenesis
- Author
-
Katherine L Madson, Shenghui Duan, Gary S. Gottesman, Angela Nenninger, Steven Mumm, Deborah Wenkert, Margaret Huskey, Michael P. Whyte, William H. McAlister, and Ronald M. Laxer
- Subjects
Adult ,Male ,Proband ,Adolescent ,MafB Transcription Factor ,Molecular Sequence Data ,Osteoclasts ,Osteolysis ,Biology ,medicine.disease_cause ,Article ,Cohort Studies ,Pathogenesis ,Young Adult ,Transactivation ,Osteogenesis ,Genetics ,medicine ,Humans ,Missense mutation ,Child ,Gene ,Genetics (clinical) ,Mutation ,Base Sequence ,RANK Ligand ,Hand ,Penetrance ,Radiography ,MAFB ,Child, Preschool ,Female - Abstract
Multicentric carpotarsal osteolysis syndrome (MCTO), an autosomal dominant disorder that often presents sporadically, features carpal-tarsal lysis frequently followed by nephropathy and renal failure. In 2012, mutations in the single-exon gene MAFB were reported in 13 probands with MCTO. MAFB is a negative regulator of RANKL-mediated osteoclastogenesis. We studied nine MCTO patients (seven sporadic patients and one affected mother and son) for MAFB mutation. We PCR-amplified and selectively sequenced the MAFB region that contains the transactivation domain in this 323 amino acid protein, where mutations were previously reported for MCTO. We found five different heterozygous missense defects among eight probands: c.176CT, p.Pro59Leu; c.185CT, p.Thr62Ile; c.206CT, p.Ser69Leu (four had this defect); c.209CT, p.Ser70Leu; and c.211CT, p.Pro71Ser. All 5 mutations are within a 13 amino acid stretch of the transactivation domain. Four were identical to the previously reported mutations. Our unique mutation (c.185CT, p.Thr62Ile) involved the same domain. DNA available from seven parents of the seven sporadic patients did not show their child's MAFB mutation. The affected mother and son had an identical defect. Hence, the mutations for 7/8 probands were suspected to have arisen spontaneously as there was no history of features of MCTO in either parent. Penetrance of MCTO seemed complete. Lack of nonsense or other truncating mutations suggested a dominant-negative pathogenesis. Our findings indicate that only a few transactivation domain-specific mutations within MAFB cause MCTO.
- Published
- 2014
- Full Text
- View/download PDF
32. Panostotic Expansile Bone Disease With Massive Jaw Tumor Formation and a Novel Mutation in the Signal Peptide of RANK
- Author
-
Anne L. Schafer, Andrea M Tom, Andrew E. Horvai, William H. McAlister, Steven Mumm, Michael P. Whyte, Edward C. Hsiao, Mark S. Anderson, Frederick V. Schaefer, Ivan H. El-Sayed, Michael T. Collins, and Dolores M. Shoback
- Subjects
Pathology ,medicine.medical_specialty ,Bone disease ,Endocrinology, Diabetes and Metabolism ,Fibrous dysplasia ,RANK Ligand ,Anatomy ,Mandibular Neoplasms ,Biology ,medicine.disease ,Bone resorption ,Bone remodeling ,Osteoprotegerin ,medicine ,GNAS complex locus ,biology.protein ,Orthopedics and Sports Medicine - Abstract
Precise regulation of bone resorption is critical for skeletal homeostasis. We report a 32-year-old man with a panostotic expansile bone disease and a massive hemorrhagic mandibular tumor. Originally from Mexico, he was deaf at birth and became bow-legged during childhood. There was no family history of skeletal disease. Puberty occurred normally, but during adolescence he experienced difficulty straightening his limbs, sustained multiple fractures, and developed a bony tumor on his chin. By age 18 years, all limbs were misshapen. The mandibular mass grew and protruded from the oral cavity, extending to the level of the lower ribs. Other bony defects included a similar maxillary mass and serpentine limbs. Upon referral at age 27 years, biochemical studies showed serum alkaline phosphatase of 1760 U/L (Nl: 29-111) and other elevated bone turnover markers. Radiography of the limbs showed medullary expansion and cortical thinning with severe bowing. Although the jaw tumors were initially deemed inoperable, mandibular mass excision and staged partial maxillectomy were eventually performed. Tumor histopathology showed curvilinear trabeculae of woven bone on a background of hypocellular fibrous tissue. Fibrous dysplasia of bone was suspected, but there was no mutation in codon 201 of GNAS in samples from blood or tumor. His clinical and radiographic findings, elevated serum markers, and disorganized bone morphology suggested amplified receptor activator of NF-κB (RANK) signaling, even though his disorder differed from conditions with known constitutive activation of RANK signaling (eg, familial expansile osteolysis). We found a unique 12-base pair duplication in the signal peptide of TNFRSF11A, the gene that encodes RANK. No exon or splice site mutations were found in the genes encoding RANK ligand or osteoprotegerin. Alendronate followed by pamidronate therapies substantially decreased his serum alkaline phosphatase activity. This unique patient expands the phenotypes and genetic basis of the mendelian disorders of RANK signaling activation. © 2014 American Society for Bone and Mineral Research.
- Published
- 2014
- Full Text
- View/download PDF
33. Juvenile Paget's disease in an Iranian kindred with vitamin D deficiency and novel homozygousTNFRSF11Bmutation
- Author
-
William H. McAlister, Shiva Nasirabadi, Forough Saki, Steven Mumm, Michael P. Whyte, and Zohreh Karamizadeh
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Bone density ,biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,medicine.disease ,vitamin D deficiency ,Endocrinology ,Osteoprotegerin ,RANKL ,Internal medicine ,Vitamin D and neurology ,biology.protein ,Medicine ,Missense mutation ,Orthopedics and Sports Medicine ,medicine.symptom ,business ,Bone pain - Abstract
Juvenile Paget's disease (JPD) is a rare heritable osteopathy characterized biochemically by markedly increased serum alkaline phosphatase (ALP) activity emanating from generalized acceleration of skeletal turnover. Affected infants and children typically suffer bone pain and fractures and deformities, become deaf, and have macrocranium. Some who survive to young adult life develop blindness from retinopathy engendered by vascular microcalcification. Most cases of JPD are caused by osteoprotegerin (OPG) deficiency due to homozygous loss-of-function mutations within the TNFRSF11B gene that encodes OPG. We report a 3-year-old Iranian girl with JPD and craniosynostosis who had vitamin D deficiency in infancy. She presented with fractures during the first year-of-life followed by bone deformities, delayed development, failure-to-thrive, and pneumonias. At 1 year-of-age, biochemical studies of serum revealed marked hyperphosphatasemia together with low-normal calcium and low inorganic phosphate and 25-hydroxyvitamin D levels. Several family members in previous generations of this consanguineous kindred may also have had JPD and vitamin D deficiency. Mutation analysis showed homozygosity for a unique missense change (c.130T>C, p.Cys44Arg) in TNFRSF11B that would compromise the cysteine-rich domain of OPG that binds receptor activator of NF-κB ligand (RANKL). Both parents were heterozygous for this mutation. The patient's serum OPG level was extremely low and RANKL level markedly elevated. She responded well to rapid oral vitamin D repletion followed by pamidronate treatment given intravenously. Our patient is the first Iranian reported with JPD. Her novel mutation in TNFRSF11B plus vitamin D deficiency in infancy was associated with severe JPD uniquely complicated by craniosynostosis. Pamidronate treatment with vitamin D sufficiency can be effective therapy for the skeletal disease caused by the OPG deficiency form of JPD.
- Published
- 2013
- Full Text
- View/download PDF
34. Raine Syndrome (OMIM #259775), Caused By FAM20C Mutation, Is Congenital Sclerosing Osteomalacia With Cerebral Calcification (OMIM 259660)
- Author
-
Michael P, Whyte, William H, McAlister, Michael D, Fallon, Mary Ella, Pierpont, Vinieth N, Bijanki, Shenghui, Duan, Ghada A, Otaify, William S, Sly, and Steven, Mumm
- Subjects
Adult ,Male ,Extracellular Matrix Proteins ,Casein Kinase I ,Infant, Newborn ,Calcinosis ,Cleft Palate ,Fibroblast Growth Factor-23 ,Osteomalacia ,Microcephaly ,Exophthalmos ,Humans ,Abnormalities, Multiple ,Female ,Cerebrum ,Osteosclerosis - Abstract
In 1985, we briefly reported infant sisters with a unique, lethal, autosomal recessive disorder designated congenital sclerosing osteomalacia with cerebral calcification. In 1986, this condition was entered into Mendelian Inheritance In Man (MIM) as osteomalacia, sclerosing, with cerebral calcification (MIM 259660). However, no attestations followed. Instead, in 1989 Raine and colleagues published an affected neonate considering unprecedented the striking clinical and radiographic features. In 1992, "Raine syndrome" entered MIM formally as osteosclerotic bone dysplasia, lethal (MIM #259775). In 2007, the etiology emerged as loss-of-function mutation of FAM20C that encodes family with sequence similarity 20, member C. FAM20C is highly expressed in embryonic calcified tissues and encodes a kinase (dentin matrix protein 4) for most of the secreted phosphoproteome including FGF23, osteopontin, and other regulators of skeletal mineralization. Herein, we detail the clinical, radiological, biochemical, histopathological, and FAM20C findings of our patients. Following premortem tetracycline labeling, the proposita's non-decalcified skeletal histopathology after autopsy indicated no rickets but documented severe osteomalacia. Archival DNA revealed the sisters were compound heterozygotes for a unique missense mutation and a novel deletion in FAM20C. Individuals heterozygous for the missense mutation seemed to prematurely fuse their metopic suture and develop a metopic ridge sometimes including trigonocephaly. Our findings clarify FAM20C's role in hard tissue formation and mineralization, and show that Raine syndrome is congenital sclerosing osteomalacia with cerebral calcification. © 2016 American Society for Bone and Mineral Research.
- Published
- 2016
35. Asfotase alfa therapy for children with hypophosphatasia
- Author
-
Karen E. Mack, Kim Hamilton, Kori Kagan, Cheryl Rockman-Greenberg, David D. Thompson, Kenji P Fujita, Katherine L Madson, Scott Moseley, Amy Reeves, Amy Yakimoski, Michael P. Whyte, Dawn Phillips, Tatjana Odrljin, and William H. McAlister
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Recombinant Fusion Proteins ,Hypophosphatasia ,030209 endocrinology & metabolism ,Rickets ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Clinical endpoint ,Humans ,Medicine ,Child ,business.industry ,Muscle weakness ,Lipohypertrophy ,General Medicine ,Enzyme replacement therapy ,Alkaline Phosphatase ,medicine.disease ,Surgery ,Clinical trial ,030104 developmental biology ,Immunoglobulin G ,Asfotase alfa ,Quality of Life ,Female ,Clinical Medicine ,medicine.symptom ,business - Abstract
Background. Hypophosphatasia (HPP) is caused by loss-of-function mutation(s) of the gene that encodes the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP). Consequently, cell-surface deficiency of TNSALP phosphohydrolase activity leads to extracellular accumulation of inorganic pyrophosphate, a natural substrate of TNSALP and inhibitor of mineralization. Children with HPP can manifest rickets, skeletal pain, deformity, fracture, muscle weakness, and premature deciduous tooth loss. Asfotase alfa is a recombinant, bone-targeted, human TNSALP injected s.c. to treat HPP. In 2012, we detailed the 1-year efficacy of asfotase alfa therapy for the life-threatening perinatal and infantile forms of HPP. Methods. Here, we evaluated the efficacy and safety of asfotase alfa treatment administered to children 6-12 years of age at baseline who were substantially impaired by HPP. Two radiographic scales quantitated HPP skeletal disease, including comparisons to serial radiographs from similarly affected historical control patients. Results. Twelve children receiving treatment were studied for 5 years. The 6-month primary endpoint was met, showing significant radiographic improvement. Additional significant improvements included patient growth, strength, motor function, agility, and quality of life, which for most patients meant achieving normal values for age- and sex-matched peers that were sustained at 5 years of treatment. For most, pain and disability resolved. Mild to moderate injection-site reactions were common and were sometimes associated with lipohypertrophy. Low anti-asfotase alfa antibody titers were noted in all patients. No evidence emerged for clinically important ectopic calcification or treatment resistance. Conclusions. Asfotase alfa enzyme replacement therapy has substantial and sustained efficacy with a good safety profile for children suffering from HPP. Trial Registration. ClinicalTrials.gov NCT00952484 (https://clinicaltrials.gov/ct2/show/NCT00952484) and NCT01203826 (https://clinicaltrials.gov/ct2/show/NCT01203826). Funding. Alexion Pharmaceuticals Inc. and Shriners Hospitals for Children.
- Published
- 2016
- Full Text
- View/download PDF
36. Validation of a novel scoring system, the radiographic global impression of change (RGI-C) scale, for assessing skeletal manifestations of hypophosphatasia in infants and children
- Author
-
Michael P. Whyte, William H. McAlister, David D. Thompson, Kenji Fujita, and Scott Moseley
- Subjects
Orthodontics ,Scoring system ,Scale (ratio) ,business.industry ,Radiography ,Hypophosphatasia ,Medicine ,Dentistry ,General Medicine ,business ,medicine.disease - Published
- 2016
- Full Text
- View/download PDF
37. COL1 C-propeptide cleavage site mutations cause high bone mass osteogenesis imperfecta
- Author
-
Sergey Leikin, Joan C. Marini, Marina Brusel, Nadja Fratzl-Zelman, Theresa E. Hefferan, William H. McAlister, Michael J. Yaszemski, Carl-Johan Rubin, Aileen M. Barnes, Klaus Klaushofer, Katarina Lindahl, Andreas Kindmark, Paul Roschger, Östen Ljunggren, Elena Makareeva, Adele L. Boskey, Steven Mumm, Michael P. Whyte, and Efrat Kessler
- Subjects
Male ,musculoskeletal diseases ,medicine.medical_specialty ,Adolescent ,Bone density ,Molecular Sequence Data ,Bone Matrix ,macromolecular substances ,Biology ,Mineralization (biology) ,Bone and Bones ,Collagen Type I ,Mice ,Osteosclerosis ,Calcification, Physiologic ,Bone Density ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Child ,Genetics (clinical) ,Osteogenesis Imperfecta ,medicine.disease ,Phenotype ,Peptide Fragments ,Collagen Type I, alpha 1 Chain ,Osteopenia ,Procollagen peptidase ,Endocrinology ,Osteogenesis imperfecta ,Mutation ,Female ,Procollagen ,Calcification - Abstract
Osteogenesis imperfecta (OI) is most often caused by mutations in the type I procollagen genes (COL1A1/COL1A2). We identified two children with substitutions in the type I procollagen C-propeptide cleavage site, which disrupt a unique processing step in collagen maturation and define a novel phenotype within OI. The patients have mild OI caused by mutations in COL1A1 (Patient 1: p.Asp1219Asn) or COL1A2 (Patient 2: p.Ala1119Thr), respectively. Patient 1 L1-L4 DXA Z-score was +3.9 and pQCT vBMD was+3.1; Patient 2 had L1-L4 DXA Z-score of 0.0 and pQCT vBMD of -1.8. Patient BMD contrasts with radiographic osteopenia and histomorphometry without osteosclerosis. Mutant procollagen processing is impaired in pericellular and in vitro assays. Patient dermal collagen fibrils have irregular borders. Incorporation of pC-collagen into matrix leads to increased bone mineralization. FTIR imaging confirms elevated mineral/matrix ratios in both patients, along with increased collagen maturation in trabecular bone, compared to normal or OI controls. Bone mineralization density distribution revealed a marked shift toward increased mineralization density for both patients. Patient 1 has areas of higher and lower bone mineralization than controls; Patient 2's bone matrix has a mineral content exceeding even classical OI bone. These patients define a new phenotype of high BMD OI and demonstrate that procollagen C-propeptide cleavage is crucial to normal bone mineralization.
- Published
- 2011
- Full Text
- View/download PDF
38. Elevated serum lactate dehydrogenase isoenzymes and aspartate transaminase distinguish Albers-Schönberg disease (Chloride Channel 7 Deficiency Osteopetrosis) among the sclerosing bone disorders
- Author
-
William H. McAlister, Steven Mumm, Michael P. Whyte, Lydia G Kempa, Deborah Wenkert, and Fan Zhang
- Subjects
medicine.medical_specialty ,biology ,Endocrinology, Diabetes and Metabolism ,Aspartate transaminase ,Osteopetrosis ,medicine.disease ,Resorption ,TCIRG1 ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,Lactate dehydrogenase ,medicine ,biology.protein ,Chloride channel ,Orthopedics and Sports Medicine ,Creatine kinase ,CLCN7 - Abstract
Osteopetrosis (OPT) refers to the consequences of generalized failure of skeletal resorption during growth. Most cases are explained by loss-of-function mutation within the genes that encode either chloride channel 7 (CLCN7) or a vacuolar proton pump subunit (TCIRG1), each compromising acid secretion by osteoclasts. Patients suffer fractures and sometimes cranial nerve entrapment and insufficient medullary space for hematopoiesis. In 1996, we reported that a high serum level of the brain isoenzyme of creatine kinase (BB-CK), the CK of osteoclasts, characterizes OPT dueamong the sclerosing bone disorders (J Clin Endocrinol Metab. 1996;11:1438). Now, we show that elevation in serum of multiple lactate dehydrogenase (LDH) isoenzymes with aspartate transaminase (AST) distinguishes autosomal dominant OPT due to loss-of-function mutation in CLCN7 [Albers-Schonberg disease (A-SD)] among these conditions. Serum total LDH and AST levels as high as 3× and 2×, respectively, the upper limits of normal for age-appropriate controls, were persistent and essentially concordant in A-SD. Serum LDH was elevated in 7 of 9 children and in the 2 adults studied with A-SD. LDH isoenzyme quantitation showed excesses of LDH-2, -3, and -4. Neither total LDH nor AST increases were found in other forms of OPT, including bisphosphonate-induced OPT, or in 41 children and 6 adults representing 20 additional sclerosing bone disorders. Serum TRACP-5b and BB-CK also were markedly elevated in A-SD. Hence, high serum levels of several enzymes characterize A-SD. Elevated serum LDH isoenzymes and AST indicate a disturbance (of uncertain clinical significance) within multiple extraosseous tissues when there is CLCN7 deficiency.
- Published
- 2010
- Full Text
- View/download PDF
39. Dysosteosclerosis presents as an 'Osteoclast-Poor' form of osteopetrosis: Comprehensive investigation of a 3-year-old girl and literature review
- Author
-
Steven Mumm, Michael P. Whyte, Angie R Nenninger, Margaret Huskey, William H. McAlister, Xiafang Zhang, Deborah Wenkert, and Deborah V. Novack
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Bone disease ,business.industry ,Endocrinology, Diabetes and Metabolism ,Long bone ,Parathyroid hormone ,Osteopetrosis ,030206 dentistry ,Anatomy ,medicine.disease ,Short stature ,Osteopenia ,03 medical and health sciences ,Osteosclerosis ,0302 clinical medicine ,medicine.anatomical_structure ,Osteoclast ,Medicine ,Orthopedics and Sports Medicine ,medicine.symptom ,business ,030304 developmental biology - Abstract
Dysosteosclerosis (DSS), an extremely rare dense bone disease, features short stature and fractures and sometimes optic atrophy, cranial nerve palsy, developmental delay, and failure of tooth eruption in infancy or early childhood consistent with osteopetrosis (OPT). Bone histology during childhood shows unresorbed primary spongiosa from deficient osteoclast action. Additionally, there is remarkable progressive flattening of all vertebrae and, by adolescence, paradoxical metaphyseal osteopenia with thin cortical bone. Reports of consanguinity indicate autosomal recessive inheritance, yet more affected males than females suggest X-linked recessive inheritance. We investigated a nonconsanguineous girl with DSS. Osteosclerosis was discovered at age 7 months. Our studies, spanning ages 11 to 44 months, showed weight at approximately 50th percentile, and length diminishing from approximately 30th percentile to –2.3 SD. Head circumference was +4 SD. The patient had frontal bossing, blue sclera, normal teeth, genu valgum, and unremarkable joints. Radiographs showed orbital and facial sclerosis, basilar thickening, bone-in-bone appearance of the pelvis, sclerotic long bone ends, and fractures of ribs and extremities. Progressive metaphyseal widening occurred as vertebrae changed from ovoid to flattened and became beaked anteriorly. A hemogram was normal. Consistent with OPT, serum parathyroid hormone (PTH) concentrations reflected dietary calcium levels. Serum bone alkaline phosphatase, osteocalcin, and TRACP-5b were subnormal. The iliac crest contained excessive primary spongiosa and no osteoclasts. No mutations were identified in the splice sites or exons for the genes encoding chloride channel 7, T-cell immune regulator 1, OPT-associated transmembrane protein 1, and monocyte colony-stimulating factor (M-CSF) and its receptor C-FMS, ANKH, OPG, RANK, and RANKL. Genomic copy-number microarray was unrevealing. Hence, DSS is a distinctive OPT of unknown etiology featuring osteoclast deficiency during early childhood. How osteopenia follows is an enigma of human skeletal pathobiology. © 2010 American Society for Bone and Mineral Research.
- Published
- 2010
- Full Text
- View/download PDF
40. Bisphosphonate-Induced Osteopetrosis: Novel Bone Modeling Defects, Metaphyseal Osteopenia, and Osteosclerosis Fractures After Drug Exposure Ceases
- Author
-
Deborah V. Novack, Perry L. Schoenecker, Karen L. Clements, Deborah Wenkert, William H. McAlister, and Michael P. Whyte
- Subjects
Male ,Adolescent ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Spondylolysis ,Bone remodeling ,Osteosclerosis ,medicine ,Humans ,Orthopedics and Sports Medicine ,Child ,Bone pain ,Diphosphonates ,business.industry ,Ulna ,Osteopetrosis ,Anatomy ,Bisphosphonate ,medicine.disease ,Osteopenia ,Bone Diseases, Metabolic ,medicine.anatomical_structure ,Bone Remodeling ,medicine.symptom ,business - Abstract
In 2003, we reported on a 12-yr-old boy who had developed osteopetrosis (OPT) while receiving pamidronate (PMD) for idiopathic bone pain and enigmatic elevation in circulating bone alkaline phosphatase. Now 17 yr of age, he was re-evaluated 6.5 yr after PMD exposure stopped. Our patient described less bone pain but further limb fractures. His growth plates were fused, yet hyperphosphatasemia persisted. Radiographs documented interval fractures of a metacarpal, an osteosclerotic distal radius, and a dense diaphyseal segment of an ulna where a "chalkstick" break remained incompletely healed after 2 yr. There was new L(4) spondylolysis, and previous L(5) spondylolysis had caused spondylolisthesis. Modeling disturbances of OPT persisted, but partial recovery was shown by metaphyseal surfaces with a unique concave shape. Metaphyseal osteosclerosis had remodeled imperfectly to become focal areas of dense, diaphyseal bone. Newer metaphyseal bone was unexpectedly osteopenic, especially in his distal femurs where cortices were thin and a paucity of trabeculae was documented by CT. Femoral necks had become short and wide with an abnormal contour. A "bone-within-bone" configuration was now present throughout his skeleton. In vertebrae, endplates were thin, and trabecular osteopenia was present central and peripheral to the bands of osteosclerosis. BMD Z-scores assessed by DXA had decreased into the normal range in his spine, hip, and whole body. Iliac crest biopsy showed active bone formation, with much less accumulated primary spongiosa than during the PMD infusions. Osteoclasts that had been dysmorphic, round cells without polarization and off of bone surfaces were now unremarkable in number, location, and appearance. In conclusion, bisphosphonate toxicity during childhood can impair skeletal modeling and remodeling with structural changes that evolve and carry into adult life.
- Published
- 2008
- Full Text
- View/download PDF
41. Absence of MMP2 Mutation in Idiopathic Multicentric Osteolysis with Nephropathy
- Author
-
Steven Mumm, Michael P. Whyte, Deborah Wenkert, Stefanie M Wiegand, and William H. McAlister
- Subjects
Male ,Pathology ,medicine.medical_specialty ,MMP2 ,Osteolysis ,Adolescent ,DNA Mutational Analysis ,Polymerase Chain Reaction ,Nephropathy ,Arthropathy ,medicine ,Humans ,Orthopedics and Sports Medicine ,RNA, Messenger ,Child ,Kidney transplantation ,Proteinuria ,business.industry ,General Medicine ,medicine.disease ,body regions ,Mutation ,Mutation (genetic algorithm) ,Matrix Metalloproteinase 2 ,Kidney Diseases ,Osteolysis, Essential ,Surgery ,medicine.symptom ,business ,Kidney disease - Abstract
The genetic basis of idiopathic multicentric osteolysis with nephropathy is unknown. This disorder is typically a sporadic, but sometimes an autosomal dominant, condition featuring carpal-tarsal destruction and nephropathy causing renal failure. Loss-of-function mutation within the gene encoding matrix metalloproteinase 2 (MMP2) causes the autosomal recessive disorder nodulosis-arthropathy-osteolysis syndrome characterized by carpal-tarsal destruction, subcutaneous nodules, and generalized osteoporosis. We questioned whether sporadic idiopathic multicentric osteolysis with nephropathy is allelic with nodulosis-arthropathy osteolysis syndrome and undertook sequence analysis of the matrix metalloproteinase 2 gene in three unrelated affected boys. Although symptoms appeared by age 2 years, idiopathic multicentric osteolysis was diagnosed at ages 5, 5, and 12 years with flares of pain and limited motion or swelling of wrists, ankles, elbows, knees, and shoulders. Proteinuria was present on referral at ages 8, 7, and 12 years, respectively. Kidney transplantation was necessary for one boy at age 17 years. Coding exons and adjacent mRNA splice sites of the matrix metalloproteinase 2 gene were analyzed by polymerase chain reaction amplification and DNA sequencing. Matrix metalloproteinase 2 gene analysis was negative for mutation in the three patients. Sequence analysis of the matrix metalloproteinase 2 gene shows sporadic idiopathic multicentric osteolysis with nephropathy is not allelic to nodulosis-arthropathy-osteolysis syndrome. The genetic bases of idiopathic multicentric osteolysis disorders remain unknown.
- Published
- 2007
- Full Text
- View/download PDF
42. Infantile Hypophosphatasia: Transplantation Therapy Trial Using Bone Fragments and Cultured Osteoblasts
- Author
-
Sharon A. Perlman, Steven Mumm, Michael P. Whyte, Richard A. Cahill, William H. McAlister, Stephen P. Coburn, Ann Steele, and Deborah Wenkert
- Subjects
medicine.medical_specialty ,Pathology ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Hypophosphatasia ,Rickets ,Biochemistry ,Metabolic bone disease ,Cytogenetics ,Endocrinology ,Internal medicine ,medicine ,Humans ,Knee ,Child ,Cells, Cultured ,Bone Marrow Transplantation ,Cell Proliferation ,Bone Transplantation ,Osteoblasts ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Stem Cells ,Biochemistry (medical) ,Infant ,Cell Differentiation ,Osteoblast ,DNA ,Alkaline Phosphatase ,medicine.disease ,Metabolic Bone Disorder ,Radiography ,Transplantation ,medicine.anatomical_structure ,Asfotase alfa ,Hypercalcemia ,Alkaline phosphatase ,Female ,business - Abstract
Hypophosphatasia (HPP) is a rare, heritable, metabolic bone disease due to deficient activity of the tissue-nonspecific isoenzyme of alkaline phosphatase. The infantile form features severe rickets often causing death in the first year of life from respiratory complications. There is no established medical treatment. In 1997, an 8-month-old girl with worsening and life-threatening infantile HPP improved considerably after marrow cell transplantation.Our aim was to better understand and to advance these encouraging transplantation results.In 1999, based on emerging mouse transplantation models involving implanted donor bone fragments as well as osteoblast-like cells cultured from bone, we treated a 9-month-old girl suffering a similar course of infantile HPP.Four months later, radiographs demonstrated improved skeletal mineralization. Twenty months later, PCR analysis of adherent cells cultured from recipient bone suggested the presence of small amounts of paternal (donor) DNA despite the absence of hematopoietic engraftment. This patient, now 8 yr old (7 yr after transplantation), is active and growing, and has the clinical phenotype of the more mild, childhood form of HPP.Cumulative experience suggests that, after immune tolerance, donor bone fragments and marrow may provide precursor cells for distribution and engraftment in the skeletal microenvironment in HPP patients to form tissue-nonspecific isoenzyme of alkaline phosphatase-replete osteoblasts that can improve mineralization.
- Published
- 2007
- Full Text
- View/download PDF
43. Congenital insensitivity to pain: Fracturing without apparent skeletal pathobiology caused by an autosomal dominant, second mutation in SCN11A encoding voltage-gated sodium channel 1.9
- Author
-
Voraluck Phatarakijnirund, Deborah Wenkert, Steven Mumm, William H. McAlister, Michael P. Whyte, Deborah V. Novack, and Karen L. Clements
- Subjects
0301 basic medicine ,Joint hypermobility ,Male ,Pathology ,medicine.medical_specialty ,Histology ,Pain Insensitivity, Congenital ,Physiology ,Endocrinology, Diabetes and Metabolism ,DNA Mutational Analysis ,Molecular Sequence Data ,Electromyography ,Iliac crest ,Bone and Bones ,Article ,03 medical and health sciences ,Fractures, Bone ,Young Adult ,0302 clinical medicine ,Deformity ,medicine ,Missense mutation ,Humans ,Amino Acid Sequence ,Child ,NAV1.9 Voltage-Gated Sodium Channel ,Genes, Dominant ,medicine.diagnostic_test ,Base Sequence ,Hyperhidrosis ,business.industry ,Infant, Newborn ,Infant ,medicine.disease ,Pedigree ,030104 developmental biology ,medicine.anatomical_structure ,Peripheral neuropathy ,Child, Preschool ,Mutation ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Congenital insensitivity to pain - Abstract
Congenital insensitivity to pain (CIP) comprises the rare heritable disorders without peripheral neuropathy that feature inability to feel pain. Fracturing and joint destruction are common complications, but lack detailed studies of mineral and skeletal homeostasis and bone histology. In 2013, discovery of a heterozygous gain-of-function mutation in SCN11A encoding voltage-gated sodium channel 1.9 (Nav1.9) established a distinctive CIP in three unrelated patients who suffered multiple painless fractures, self-inflicted mutilation, chronic diarrhea, and hyperhidrosis. Here, we studied a mother and two children with CIP by physical examination, biochemical testing, radiological imaging including DXA, iliac crest histology, and mutation analysis. She suffered fractures primarily of her lower extremities beginning at age two years, and had Charcot deformity of both ankles and joint hypermobility. Nerve conduction velocity together with electromyography were normal. Her children had recurrent major fractures beginning in early childhood, joint hypermobility, and chronic diarrhea. She had an excoriated external nare, and both children had hypertrophic scars from scratching. Skin collagen studies were normal. Radiographs revealed fractures and deformities. However, lumbar spine and total hip BMD Z-scores, biochemical parameters of mineral and skeletal homeostasis, and iliac crest histology of the mother (after in vivo tetracycline labeling) were normal. Genomic DNA from the children revealed a unique heterozygous missense mutation in exon 23 (c.3904C>T, p.Leu1302Phe) of SCN11A that is absent in SNP databases and alters an evolutionarily conserved amino acid. This autosomal dominant CIP reflects the second gain-of-function mutation of SCN11A. Perhaps joint hypermobility is an unreported feature. How mutation of Nav1.9 causes fracturing remains unexplained. Lack of injury awareness is typically offered as the reason, and was supported by our unremarkable biochemical, radiological, and histological findings indicating no skeletal pathobiology. However, low-trauma fracturing in these patients suggests an uncharacterized defect in bone quality.
- Published
- 2015
44. Response to: A rapid skeletal turnover in radiographic mimic of osteopetrosis might be secondary to systemic mastocytosis
- Author
-
Katherine L Madson, William H. McAlister, Jo Blair, Nicholas Shaw, Deborah V. Novack, Steven Mumm, and Michael P. Whyte
- Subjects
Male ,Pathology ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Radiography ,Osteoporosis ,Osteopetrosis ,medicine.disease ,Bone remodeling ,Osteosclerosis ,medicine ,Humans ,Orthopedics and Sports Medicine ,Bone Remodeling ,Systemic mastocytosis ,business - Published
- 2015
45. Deactivating Germline Mutations in LEMD3 Cause Osteopoikilosis and Buschke-Ollendorff Syndrome, but Not Sporadic Melorheostosis
- Author
-
Steven Mumm, Michael P. Whyte, Xiafang Zhang, Richard J. Mier, Deborah Wenkert, and William H. McAlister
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Melorheostosis ,Endocrinology, Diabetes and Metabolism ,Nonsense mutation ,Osteopetrosis ,Syndrome ,Biology ,medicine.disease ,Osteochondrodysplasia ,humanities ,Germline ,Buschke–Ollendorff syndrome ,Germline mutation ,medicine ,Humans ,Female ,Orthopedics and Sports Medicine ,Osteopoikilosis ,Child ,Nevus ,Germ-Line Mutation - Abstract
Autosomal dominant OPK and BOS feature widespread foci of osteosclerotic trabeculae without or with skin lesions, respectively. Occasionally, a larger area of dense bone in OPK or BOS resembles MEL, a sporadic sclerosing disorder primarily involving cortical bone. Others, finding deactivating germline LEMD3 mutations in OPK or BOS, concluded such defects explain all three conditions. We found germline LEMD3 mutations in OPK and BOS but not in sporadic MEL. Introduction: In 2004, others discovered that heterozygous, loss-of-function, germline mutations in the LEMD3 gene (LEMD3 or MAN1) cause both osteopoikilosis (OPK) and Buschke-Ollendorff syndrome (BOS). OPK is an autosomal dominant, usually benign, skeletal dysplasia featuring multiple, small, especially metaphyseal, oval or round, dense trabecular foci distributed symmetrically throughout the skeleton. BOS combines OPK with connective tissue nevi comprised of collagen and elastin. In some OPK and BOS families, an individual may have relatively large, asymmetric areas of dense cortical bone interpreted as melorheostosis (MEL). MEL, however, classically refers to a sporadic, troublesome skeletal dysostosis featuring large, asymmetric, “flowing hyperostosis” of long bone cortices often with overlying, constricting soft tissue abnormalities. However, a heterozygous germline mutation in LEMD3 was offered to explain MEL. Materials and Methods: We studied 11 unrelated individuals with sclerosing bone disorders where LEMD3 mutation was a potential etiology: familial OPK (1), familial BOS (2), previously reported familial OPK with MEL (1), sporadic MEL (3), sporadic MEL with mixed-sclerosing-bone dystrophy (1), and patients with other unusual sclerosing bone disorders (3). All coding exons and adjacent mRNA splice sites for LEMD3 were amplified by PCR and sequenced using genomic DNA from leukocytes. We did not study lesional tissue from bone or skin. Results: In the OPK family, a heterozygous nonsense mutation (c.1433T>A, p.L478X) was discovered in exon 1. In the two BOS families, a heterozygous nonsense mutation (exon 1, c.1323C>A, p.Y441X) and a heterozygous frame-shift mutation (exon 1, c.332_333insTC) were identified. In the individual with MEL and familial OPK, a heterozygous nonsense mutation (c.1963C>T, p.R655X) was detected in exon 7. However, no LEMD3 mutation was found for any other patient, including all four with sporadic MEL. Conclusions: We confirm that OPK and BOS individuals, including those with MEL-like lesions, have heterozygous, deactivating, germline LEMD3 mutations. However, MEL remains of unknown etiology.
- Published
- 2006
- Full Text
- View/download PDF
46. Desmoplastic Nested Spindle Cell Tumor of Liver
- Author
-
Laura S. Finn, Paul E. Swanson, William H. McAlister, Jacob C. Langer, Robert K. Minkes, Antonio G. Nascimento, Louis P. Dehner, Michael Covinsky, Eduardo Ruchelli, Keith A Anderson, and D. Ashley Hill
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Hepatoblastoma ,Liver tumor ,Adolescent ,medicine.medical_treatment ,Vimentin ,Disease-Free Survival ,Pathology and Forensic Medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Hepatectomy ,Humans ,Stromal tumor ,Child ,biology ,Epithelioid Cells ,Liver Neoplasms ,Sarcoma ,Anatomy ,medicine.disease ,Immunohistochemistry ,Desmoplasia ,Child, Preschool ,biology.protein ,Female ,Surgery ,medicine.symptom ,Epithelioid cell - Abstract
We describe a distinctive tumor of the liver in four children composed of nested spindled and epithelioid cells with extensive desmoplasia that we have termed "desmoplastic nested spindle cell tumor of the liver." All four patients were previously healthy. One patient had a presumptive diagnosis of hepatic hemangioma 11 years prior to presentation. Grossly, the tumors were well circumscribed, lobular white masses, ranging from 2.8 to 15 cm in diameter. These tumors were characterized by the presence of cohesive nests of plump, bland spindle cells arranged in short fascicles with an accompanying desmoplastic stroma. Epithelioid areas ranging from palisading epithelioid cells at the periphery of some nests to pseudoglandular and polygonal cells with intercellular bridges were invariably present. Mitotic activity was low. Calcification and ossification were present. Non-neoplastic bile ducts and hepatic elements were seen both within and surrounding the tumor cell nests. Each tumor displayed cytoplasmic reactivity for vimentin, pan-cytokeratin, CD57, and nuclear staining for WT1. Neuroendocrine markers were negative. Ultrastructurally, the tumor cells showed focally well-developed cell junctions, basal lamina, and few cytoplasmic organelles. All tumors were confined to the liver and were resected without complication. Two patients received postoperative adjuvant therapy for presumed hepatoblastoma. The patients are doing well without recurrence at 7.5 years, 7 years, 5 years, and 8 months post-surgery. The morphologic appearance and immunohistochemical profile of these lesions are unique in our experience and represent a new category of pediatric liver tumor.
- Published
- 2005
- Full Text
- View/download PDF
47. Talo-patello-scaphoid osteolysis, synovitis, and short fourth metacarpals in sisters: A new syndrome?
- Author
-
Calvin B. Williams, Jasvinder A. Singh, and William H. McAlister
- Subjects
medicine.medical_specialty ,Osteolysis ,Adolescent ,Genes, Recessive ,Consanguinity ,Wrist pain ,Talus ,Synovitis ,medicine ,Humans ,Bone Resorption ,Genetics (clinical) ,Scaphoid Bone ,business.industry ,Siblings ,Brachydactyly ,Patella ,Syndrome ,Anatomy ,medicine.disease ,Dermatology ,Arthritis, Juvenile ,Radiography ,Tarsal Bone ,medicine.anatomical_structure ,Female ,Osteolysis, Essential ,Metacarpus ,medicine.symptom ,Ankle ,business ,Juvenile rheumatoid arthritis - Abstract
Osteolysis syndromes are characterized by resorption of affected bones with associated swelling and pain. Various forms of multicentric osteolysis syndromes including autosomal dominant and recessive carpal-tarsal osteolysis, Torg, François, Whyte-Hemingway, Hajdu-Cheney, Winchester, and other forms have been described. Most present in pre-school years with extensive involvement and destruction of multiple bones. We present a sister-pair, both of whom presented in early teenage, i.e., 13 and 15.5 years, respectively, with bilateral ankle, knee, and later, wrist pain. Radiological examination revealed bilateral osteolysis of tali, scaphoids, and patellae, and short fourth metacarpals in both sisters. Further investigation revealed absence of renal involvement, a normal excretion of amino acids, mucopolysaccharides and oligosaccharides, and presence of chronic synovitis in both sisters. Both parents and a younger brother were without radiographic or clinical evidence of the disease and there was no history of consanguinity. Thus, our sister-pair presented with the same carpal and tarsal bone involvement at a much later age, with evidence of chronic synovitis, along with short fourth metacarpals (brachydactyly type E changes) and without renal disease, suggesting a new syndrome with probable autosomal recessive inheritance.
- Published
- 2003
- Full Text
- View/download PDF
48. Brachytelephalangic chondrodysplasia punctata with marked cervical stenosis and cord compression: report of two cases
- Author
-
Benjamin C. P. Lee, William H. McAlister, and Thomas E. Herman
- Subjects
Male ,musculoskeletal diseases ,Chondrodysplasia Punctata ,Pathology ,medicine.medical_specialty ,Cord ,Foot Deformities, Congenital ,macromolecular substances ,Fingers ,Spinal Stenosis ,Humans ,Medicine ,Abnormalities, Multiple ,Radiology, Nuclear Medicine and imaging ,Chondrodysplasia punctata ,business.industry ,Brachydactyly ,Infant, Newborn ,Binder syndrome ,Anatomy ,musculoskeletal system ,medicine.disease ,Spinal cord ,Osteochondrodysplasia ,Radiography ,Stenosis ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Cervical Vertebrae ,business ,Spinal Cord Compression ,Cervical vertebrae - Abstract
Severe cervical spine stenosis with cord compression has not been well documented in brachytelephalangic chondrodysplasia punctata. We report two boys with phenotypic features of brachytelephalangic chondrodysplasia punctata who had severe cervical spine stenosis secondary to dysplastic cervical vertebrae, and discuss the significance of this association and its relation to the phenotypically similar Binder phenotype.
- Published
- 2002
- Full Text
- View/download PDF
49. Lenz-Majewski hyperostotic dwarfism with hyperphosphoserinuria from a novel mutation in PTDSS1 encoding phosphatidylserine synthase 1
- Author
-
Michael P, Whyte, Amanda, Blythe, William H, McAlister, Angela R, Nenninger, Vinieth N, Bijanki, and Steven, Mumm
- Subjects
Bone Diseases, Developmental ,Sequence Homology, Amino Acid ,Nitrogenous Group Transferases ,Molecular Sequence Data ,Infant ,Bone and Bones ,Radiography ,Phosphoserine ,Intellectual Disability ,Mutation ,Animals ,Homeostasis ,Humans ,Abnormalities, Multiple ,Female ,Amino Acid Sequence ,Amino Acids - Abstract
Lenz-Majewski hyperostotic dwarfism (LMHD) is an ultra-rare Mendelian craniotubular dysostosis that causes skeletal dysmorphism and widely distributed osteosclerosis. Biochemical and histopathological characterization of the bone disease is incomplete and nonexistent, respectively. In 2014, a publication concerning five unrelated patients with LMHD disclosed that all carried one of three heterozygous missense mutations in PTDSS1 encoding phosphatidylserine synthase 1 (PSS1). PSS1 promotes the biosynthesis of phosphatidylserine (PTDS), which is a functional constituent of lipid bilayers. In vitro, these PTDSS1 mutations were gain-of-function and increased PTDS production. Notably, PTDS binds calcium within matrix vesicles to engender hydroxyapatite crystal formation, and may enhance mesenchymal stem cell differentiation leading to osteogenesis. We report an infant girl with LMHD and a novel heterozygous missense mutation (c.829TC, p.Trp277Arg) within PTDSS1. Bone turnover markers suggested that her osteosclerosis resulted from accelerated formation with an unremarkable rate of resorption. Urinary amino acid quantitation revealed a greater than sixfold elevation of phosphoserine. Our findings affirm that PTDSS1 defects cause LMHD and support enhanced biosynthesis of PTDS in the pathogenesis of LMHD.
- Published
- 2014
50. Juvenile Paget's disease with heterozygous duplication within TNFRSF11A encoding RANK
- Author
-
Eduardo Santini-Araujo, Virginia Preliasco, William H. McAlister, Deborah V. Novack, Xiafang Zhang, Steven Mumm, Michael P. Whyte, and Cristina Tau
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Deoxypyridinoline ,Pathology ,Heterozygote ,Histology ,Osteolysis ,Bone disease ,Adolescent ,Physiology ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,DNA Mutational Analysis ,Molecular Sequence Data ,Pamidronate ,Biology ,Article ,Bone remodeling ,chemistry.chemical_compound ,Internal medicine ,Gene Duplication ,medicine ,Dentition ,Humans ,Amino Acid Sequence ,Femur ,Child ,Femur fracture ,Base Sequence ,Diphosphonates ,Receptor Activator of Nuclear Factor-kappa B ,Osteopetrosis ,medicine.disease ,Osteitis Deformans ,Endocrinology ,Paget's disease of bone ,chemistry ,Child, Preschool ,Mutation ,Female ,Tomography, X-Ray Computed - Abstract
Mendelian disorders of RANKL/OPG/RANK signaling feature the extremes of aberrant osteoclastogenesis and cause either osteopetrosis or rapid turnover skeletal disease. The patients with autosomal dominant accelerated bone remodeling have familial expansile osteolysis, early-onset Paget’s disease of bone, expansile skeletal hyperphosphatasia, or panostotic expansile bone disease due to heterozygous 18-, 27-, 15-, and 12-bp insertional duplications, respectively, within exon 1 of TNFRSF11A that encodes the signal peptide of RANK. Juvenile Paget’s disease (JPD), an autosomal recessive disorder, manifests extremely fast skeletal remodeling, and is usually caused by loss-of-function mutations within TNFRSF11B that encodes OPG. These disorders are ultra-rare. A 13-year-old Bolivian girl was referred at age 3 years. One femur was congenitally short and curved. Then, both bowed. Deafness at age 2 years involved missing ossicles and eroded cochleas. Teeth often had absorbed roots, broke, and were lost. Radiographs had revealed acquired tubular bone widening, cortical thickening, and coarse trabeculation. Biochemical markers indicated rapid skeletal turnover. Histopathology showed accelerated remodeling with abundant osteoclasts. JPD was diagnosed. Immobilization from a femur fracture caused severe hypercalcemia that responded rapidly to pamidronate treatment followed by bone turnover marker and radiographic improvement. No TNFRSF11B mutation was found. Instead, a unique heterozygous 15-bp insertional tandem duplication (87dup15) within exon 1 of TNFRSF11A predicted the same pentapeptide extension of RANK that causes expansile skeletal hyperphosphatasia (84dup15). Single nucleotide polymorphisms in TNFRSF11A and TNFRSF11B possibly impacted her phenotype. Our findings: i) reveal that JPD can be associated with an activating mutation within TNFRSF11A, ii) expand the range and overlap of phenotypes among the mendelian disorders of RANK activation, and iii) call for mutation analysis to improve diagnosis, prognostication, recurrence risk assessment, and perhaps treatment selection among the monogenic disorders of RANKL/OPG/RANK activation.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.