1
|
Noda Y, Kido J, Sawada T, Tanaka K, Kumeda K, Yoshida S, Sugawara K, Nakamura K. Newborn screening for hypophosphatasia: development of a high-throughput tissue nonspecific alkaline phosphatase activity assay using dried blood spots. JBMR Plus 2025; 9:ziae172. [PMID: 39925621 PMCID: PMC11803525 DOI: 10.1093/jbmrpl/ziae172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/11/2024] [Accepted: 12/24/2024] [Indexed: 02/11/2025] Open
Abstract
Hypophosphatasia (HPP) is an inherited metabolic disease caused by deficiency of tissue nonspecific alkaline phosphatase (TNAP) caused by pathogenic variants of the ALPL gene (MIM 171760). The clinical manifestations of HPP vary, ranging from a lethal perinatal-onset type to a moderate late-onset type presenting with nonspecific symptoms, such as arthropathy and musculoskeletal pain. HPP is characterized by low TNAP activity and defective bone mineralization, leading to bone deformity and skeletal abnormalities. Moreover, this disease can cause systemic complications, such as muscle weakness, seizures, pain, and respiratory failure, leading to premature death in infants. This study aimed to evaluate whether measuring TNAP activity in dried blood spots (DBSs) can identify patients with HPP. We developed an assay to assess TNAP activity using DBSs and screened 45 632 newborns born between February 2019 and March 2022 in Kumamoto Prefecture in Japan for HPP. We detected a single heterozygous variant of the ALPL gene in 5 newborns. During the clinical course follow-up, one newborn presented with HPP-related clinical manifestations. This is the first study on newborn screening (NBS) for HPP worldwide. NBS for HPP using DBSs may be practical and beneficial, as it is a high-throughput method. Moreover, the DBSs used for the TNAP assay are the same as those used for public-funded NBS worldwide. In the future, this system may be implemented as standard NBS for HPP.
Collapse
Affiliation(s)
- Yusuke Noda
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | - Jun Kido
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Takaaki Sawada
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kenichi Tanaka
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
| | | | | | - Keishin Sugawara
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Kimitoshi Nakamura
- Department of Pediatrics, Kumamoto University Hospital, Kumamoto, Japan
- Department of Pediatrics, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
2
|
Kishnani PS, Seefried L, Dahir KM, Martos-Moreno GÁ, Linglart A, Petryk A, Mowrey WR, Fang S, Ozono K, Högler W, Rockman-Greenberg C. New insights into the landscape of ALPL gene variants in patients with hypophosphatasia from the Global HPP Registry. Am J Med Genet A 2024; 194:e63781. [PMID: 38884565 DOI: 10.1002/ajmg.a.63781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
Hypophosphatasia (HPP) is a rare, inherited metabolic disease characterized by low tissue-nonspecific alkaline phosphatase activity due to ALPL gene variants. We describe ALPL variants from the observational, prospective, multinational Global HPP Registry. Inclusion in the analysis required a diagnosis of HPP, low serum ALP activity, and ≥1 ALPL variant. Of 1176 patients enrolled as of September 2022, 814 met inclusion criteria in Europe (48.9%), North America (36.7%), Japan (10.2%), Australia (2.6%), and elsewhere (1.6%). Most patients (74.7%) had 1 ALPL variant; 25.3% had ≥2 variants. Nearly all patients (95.6%) had known disease-causing variants; 4.4% had variants of uncertain significance. Disease-causing variants were predominantly missense (770/1556 alleles). The most common variants were c.571G>A (102/1628 alleles), c.1250A>G (66/1628 alleles), and c.1559del (61/1628 alleles). Variant profiles were generally consistent, except in Japan, where a higher proportion of patients (68.7%) had ≥2 ALPL variants, likely because more had disease onset before age 6 months (53.0% vs. 10.1%-23.1% elsewhere). Frameshift mutations (61/164 alleles) and inframe deletions (7/164 alleles) were more common in Japan. Twenty-three novel variants were discovered, each in a single geographic region, predominantly Europe. Analyses confirmed previously known ALPL variants, identified novel variants, and characterized geographic variation in frequency and type of ALPL variants in a large population.
Collapse
Affiliation(s)
| | | | - Kathryn M Dahir
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gabriel Ángel Martos-Moreno
- Hospital Infantil Universitario Niño Jesús, IIS La Princesa, Universidad Autónoma de Madrid, CIBERobn, ISCIII, Madrid, Spain
| | - Agnès Linglart
- Paris-Saclay University, AP-HP and INSERM, Paris, France
| | - Anna Petryk
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | | | - Shona Fang
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Keiichi Ozono
- Iseikai International General Hospital, Kita Ward, Osaka, Japan
| | | | | |
Collapse
|
3
|
Lee D, Park SY, Kim HS, Kang S. Short stature with low serum alkaline phosphatase activity: a case report of hypophosphatasia. Ann Pediatr Endocrinol Metab 2023; 28:312-317. [PMID: 38173385 PMCID: PMC10765032 DOI: 10.6065/apem.2244294.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/06/2023] [Accepted: 04/20/2023] [Indexed: 01/05/2024] Open
Abstract
Hypophosphatasia (HPP) is a rare condition characterized by abnormal bone mineralization. The manifestations of HPP vary from no symptoms to intrauterine fetal death; short stature is another indication of HPP. A 3 ½-year-old boy presented with short stature, transient hypercalcemia, and mild gait disturbance without definite bony deformity. Laboratory examination revealed transient hypercalcemia, normal phosphorous and 25-hydroxy vitamin D levels, and mildly low alkaline phosphatase levels. A targeted next-generation sequencing panel associated with inborn errors of metabolism revealed a pathogenic heterozygous mutation in the ALPL gene, c.979T>C (p.Phe327Leu). When a child visits a hospital with short stature, decreased height velocity, and low alkaline phosphatase level, clinicians should consider the possibility of HPP even if definite skeletal dysplasia is not evident.
Collapse
Affiliation(s)
- Donghyun Lee
- Department of Pediatrics, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
| | - So Yun Park
- Department of Pediatrics, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
| | - Heung Sik Kim
- Department of Pediatrics, Keimyung University Daegu Dongsan Hospital, Daegu, Korea
| | - Seokjin Kang
- Department of Pediatrics, Keimyung University Dongsan Hospital, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
4
|
Martos-Moreno GÁ, Rockman-Greenberg C, Ozono K, Petryk A, Kishnani PS, Dahir KM, Seefried L, Fang S, Högler W, Linglart A. Clinical Profiles of Children with Hypophosphatasia prior to Treatment with Enzyme Replacement Therapy: An Observational Analysis from the Global HPP Registry. Horm Res Paediatr 2023; 97:233-242. [PMID: 37442110 PMCID: PMC11078328 DOI: 10.1159/000531865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
INTRODUCTION The objective of this study was to better understand the clinical profiles of children with hypophosphatasia (HPP) prior to treatment with enzyme replacement therapy (ERT). METHODS Pretreatment demographics and medical histories of ERT-treated children (aged <18 years) enrolled in the Global HPP Registry (2015-2020) were analyzed overall, by age at first HPP manifestation (<6 months vs. 6 months to 18 years), and by geographic region (USA/Canada, Europe, and Japan). RESULTS Data from 151 children with HPP were analyzed. Sex distribution was balanced overall (52.3% female; 47.7% male) but differed in Japan (63.0% female; 37.0% male). Prior to ERT initiation, common manifestations were skeletal (67.5%) and extraskeletal, with the foremost types being muscular (48.3%), constitutional/metabolic (47.0%), and neurologic (39.7%). A high proportion of children who first presented at <6 months of age (perinatal/infantile period) had a history of bone deformity (59.3%) and respiratory failure (38.3%), while those aged 6 months to 18 years at first manifestation had a predominance of early loss of primary teeth (62.3%) and gross motor delay (41.0%). Those from Japan were reported to have a younger median age overall, the highest proportion of skeletal manifestations (80.4%) and growth impairment, while European data reported the highest proportion of muscular manifestations (70.7%). In the USA/Canada, skeletal and muscular manifestations were reported at the same frequency (57.4%). CONCLUSION Prior to ERT, skeletal and extraskeletal manifestations were commonly reported in children with HPP, with differences by age at first HPP manifestation and geographical region. Comprehensive assessments of children with HPP are warranted prior to ERT initiation.
Collapse
Affiliation(s)
- Gabriel Ángel Martos-Moreno
- Departments of Pediatrics and Pediatric Endocrinology Hospital Infantil Universitario Niño Jesús, IIS La Princesa, Universidad Autónoma de Madrid, CIBERobn, ISCIII, Madrid, Spain
| | | | - Keiichi Ozono
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Anna Petryk
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Kathryn M. Dahir
- Division of Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lothar Seefried
- Orthopedic Department, University of Würzburg, Würzburg, Germany
| | - Shona Fang
- Alexion, AstraZeneca Rare Disease, Boston, MA, USA
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Agnès Linglart
- AP-HP, Paris-Saclay University, service d’endocrinologie et diabète de l’enfant, DMU 3 SEA, Centre de Référence des Maladies Rares du Métabolisme du Calcium et du Phosphate, Filière OSCAR; Paris-Saclay University, INSERM U1185, Bicêtre Paris-Saclay Hospital, Le Kremlin-Bicêtre, Paris, France
| |
Collapse
|
5
|
Kim I, Noh ES, Kim MS, Jang JH, Jeon TY, Choi HW, Cho SY. Six-year clinical outcomes of enzyme replacement therapy for perinatal lethal and infantile hypophosphatasia in Korea: Two case reports. Medicine (Baltimore) 2023; 102:e32800. [PMID: 36820543 PMCID: PMC9907957 DOI: 10.1097/md.0000000000032800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
INTRODUCTION Hypophosphatasia (HPP) is a genetic disease caused by loss-of-function mutations in ALPL, which encodes tissue-nonspecific alkaline phosphatase (ALP). Early diagnosis and treatment of perinatal and infantile HPP are important because of their high mortality rates. Enzyme replacement therapy (ERT) using human recombinant tissue-nonspecific ALP asfotase alfa was introduced in Korea in 2016. We report the first experience of ERT over 6 years for perinatal lethal and infantile HPP in Korea. PATIENT CONCERNS The first patient was a 6-week-old Korean boy with a failure to thrive. The second patient was an 8-day-old Korean-Uzbek body with generalized tonic-clonic seizure with cyanosis. DIAGNOSES HPP was suspected in both patients because of the very low level of ALP activity and rachitic findings on radiographs, and the disease was confirmed by Sanger sequencing of the ALPL gene. INTERVENTION The first patient with infantile HPP started ERT at 21 months of age and the second patient with perinatal HPP started ERT at 30 days of age. Both patients received asfotase alfa (2 mg/kg 3 times per week subcutaneously, adjusted to 3 mg/kg 3 times per week if required) for 6 years. OUTCOMES After 6 years of ERT, radiographic findings and growth standard deviation scores improved in both patients. The second patient showed no evidence of rickets after 3 years of ERT. Mechanical respiratory support and supplemental oxygen were not required after 4.5 years of treatment in the first patient and at 2 months after treatment in the second patient. CONCLUSION Among the 2 patients, the patient who started ERT early had a much better prognosis despite a more severe initial clinical presentation. Our results suggest that early diagnosis and prompt treatment play an important role in improving long-term prognosis and avoiding morbidity and premature mortality in patients with perinatal and infantile HPP.
Collapse
Affiliation(s)
- Insung Kim
- Department of Public Health Administration, Asan City Health Center, Asan, Korea
| | - Eu-Seon Noh
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min-Sun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ja-Hyun Jang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Yeon Jeon
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae Won Choi
- Department of Orthodontics, The Institute of Oral Health Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Yoon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- * Correspondence: Sung Yoon Cho, Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea (e-mail: )
| |
Collapse
|
6
|
Okawa R, Nakano K. Dental manifestation and management of hypophosphatasia. JAPANESE DENTAL SCIENCE REVIEW 2022; 58:208-216. [PMID: 35814738 PMCID: PMC9260292 DOI: 10.1016/j.jdsr.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/22/2022] [Accepted: 06/26/2022] [Indexed: 11/24/2022] Open
Abstract
Hypophosphatasia is an inherited metabolic disorder characterized by defective mineralization of bones and teeth with a wide variety of manifestations, ranging from stillbirth to dental symptoms alone. Recently, the prognosis of severe hypophosphatasia patients has been greatly improved by the introduction of enzyme replacement therapy. The typical dental manifestation is early exfoliation of primary teeth due to disturbed cementum formation, so dentures are recommended to ensure that important oral functions are acquired. Some studies have shown that enzyme replacement therapy improves dental mineralization, resulting in the stabilization of periodontal tissues and better growth of tooth roots. A nationwide Japanese survey revealed the common genetic and dental manifestations of patients with mild hypophosphatasia, which markedly differ from those of the severe forms. There may be many undiagnosed mild patients, so dentists should contribute to the early diagnosis by screening possible cases based on the typical finding of early exfoliation of primary teeth. Early diagnosis is important for patients to receive early intervention in both medical and dental fields. The establishment of fundamental dental therapy to solve the dental problems is still underway and is eagerly anticipated.
Collapse
|
7
|
Two children with hypophosphatasia with a heterozygous c.1559delT variant in the ALPL gene, the most common variant in Japanese populations. Bone Rep 2022; 17:101626. [PMID: 36217348 PMCID: PMC9547180 DOI: 10.1016/j.bonr.2022.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/07/2022] Open
Abstract
Hypophosphatasia (HPP), a genetic disorder characterized by decreased tissue-nonspecific alkaline phosphatase (TNSALP) activity, is caused by loss-of-function mutations in the ALPL gene, which encodes TNSALP. The most frequent pathogenic variant in Japanese patients with HPP is a frameshift mutation in the ALPL gene, c.1559delT, and its carrier frequency is reported to be one in 480 in the Japanese population. We report the cases of two Japanese children with HPP who had a heterozygous c.1559delT variant in the ALPL gene. One case (involving a neonate) exhibited respiratory insufficiency associated with vitamin B6 dependent convulsions, significant defective mineralization similar to the severe form of HPP, and extremely low ALP activity. Enzyme replacement therapy (ERT) using asfotase alfa promptly improved her respiratory insufficiency, bone mineralization, and maintained her motor development during infancy. The second case involved a 10-year-old boy who demonstrated diffuse musculoskeletal pain and weakness that progressively disturbed mobility. Although he showed no bony lesions, the clinical symptoms and biochemical abnormalities were compatible with childhood HPP. ERT successfully relieved the severe generalized pain and significantly improved motor function. A heterozygous c.1559delT in ALPL is the most frequent variant in Japanese populations, with aprevalence of 1/480. Two Japanese children with this variant developed hypophosphatasia associated with a low serum alkaline phosphatase. One neonate showed respiratory insufficiency and defective bone mineralization similar to the severe form of HPP. The other 10-year-old boy demonstrated diffuse musculoskeletal pain and weakness that progressively disturbed mobility. Enzyme replacement therapy successfully improved pathological symptoms in both cases.
Collapse
|
8
|
Kato H, Hidaka N, Koga M, Kinoshita Y, Makita N, Nangaku M, Ito N. Radiological evaluation of pseudofracture after the administration of asfotase alfa in an adult with benign prenatal hypophosphatasia: A case report. Bone Rep 2022; 16:101163. [PMID: 35024386 PMCID: PMC8728307 DOI: 10.1016/j.bonr.2021.101163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/28/2022] Open
Abstract
Hypophosphatasia (HPP) is a congenital disorder with decreased activity of tissue-nonspecific alkaline phosphatase. Asfotase alfa is the only treatment approved for HPP and improves the impairment of bone mineralization. Although several previous studies have reported the efficacy of asfotase alfa to treat fractures and pseudofractures in patients with HPP, there are only a few reports with a detailed description of the healing process. In this case report, we present an 18-year-old female patient with benign prenatal HPP who received asfotase alfa to treat her pseudofracture. At the age of 17, a pseudofracture developed in her left tibia after repetitive gymnastic exercise for months. Following observation over a year, she was referred to our department. X-ray images indicated a narrow radiolucent band in the mid-diaphysis of her left tibia, and bone scintigraphy showed nuclide accumulation in the same region. Replacement therapy with asfotase alfa was started, resulting in pain relief in two months, and the disappearance of nuclide accumulation on bone scintigraphy and union of the pseudofracture on X-ray after two years. This is the first case report describing the detailed pseudofracture healing process in a patient with benign prenatal HPP initiating asfotase alfa.
Collapse
Affiliation(s)
- Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Minae Koga
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Yuka Kinoshita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Noriko Makita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| | - Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Japan
| |
Collapse
|
9
|
Status Epilepticus due to Asfotase Alfa Interruption in Perinatal Severe Hypophosphatasia. Pediatr Neurol 2022; 130:4-6. [PMID: 35303588 DOI: 10.1016/j.pediatrneurol.2021.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Hypophosphatasia (HPP), an inherited, metabolic disorder caused by loss-of-function mutations in the ALPL gene, affects not only bone and tooth mineralization but also central nervous system (CNS) function, resulting in vitamin B6/pyridoxine-responsive seizures. Asfotase alfa treatment mainly improves the skeletal manifestations of HPP. As of yet, there are no reports demonstrating seizure exacerbation caused by asfotase alfa interruption. CASE The patient was a 2-year and 8-month-old female with clinical and genetic diagnosis of perinatal severe HPP. Genetic analysis of ALPL identified compound heterozygous variants. Asfotase alfa and pyridoxine administration begun on postnatal day 2 restored normal development and suppressed seizures except for simple febrile seizures. From age 2 years when her asfotase alfa injections became irregular, she began experiencing seizure exacerbation, including status epilepticus, leading to acute encephalopathy and severe sequelae. The seizure exacerbations always coincided with low alkaline phosphatase (ALP) activity caused by the interruption of asfotase alfa administration. DISCUSSION The clinical course of the present case demonstrated the effect of asfotase alfa on CNS symptoms and a clear correlation between low serum ALP activity and seizure exacerbation. Serum ALP activity measurements were useful as a therapeutic marker in the present case. Furthermore, the risk of seizure exacerbation in the patient could have been predicted, given the genotype-phenotype correlation related to the ALPL gene in the Japanese population. CONCLUSION Regular asfotase alfa injections are needed to prevent seizure exacerbation in patients with HPP. Educating patients and their family about the need for regular asfotase alfa treatment is crucial to preventing disease exacerbation.
Collapse
|
10
|
Sugiyama Y, Watanabe T, Tajika M, Matsuhashi T, Shimura M, Fushimi T, Ichimoto K, Matsunaga A, Ebihara T, Tsuruoka T, Akiyama T, Murayama K. A Japanese single-center experience of the efficacy and safety of asfotase alfa in pediatric-onset hypophosphatasia. Orphanet J Rare Dis 2022; 17:78. [PMID: 35197081 PMCID: PMC8867653 DOI: 10.1186/s13023-022-02230-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/06/2022] [Indexed: 11/10/2022] Open
Abstract
Background Hypophosphatasia (HPP) is a rare inherited metabolic disorder caused by mutations in the ALPL gene, which encodes tissue nonspecific alkaline phosphatase. The severity of HPP is widely diverse from the perinatal form to the adult mild form. The former represents the most severe form and was earlier associated with high mortality due to pneumonia which was caused by severe hypomineralization of the bones—such as chest deformity and fractured ribs—and muscle weakness. Enzyme replacement therapy using asfotase alfa (AA) was approved in 2015 in Japan for treating patients with HPP and has improved their pulmonary function and life prognosis. There are several practical and ethical challenges related to using orphan drugs for a rare disorder in a publicly funded healthcare system. Sharing experiences about their application is essential towards formulating guidelines to assist clinicians with decisions about their initiation and withdrawal. We report the details of AA experience in ten cases of pediatric-onset HPP in nine families from January 2015 to November 2019 (median [interquartile range] age 11.0 [7.6–12.5] years; 60% male). This is a study of a single-center cohort describing the clinical course of patients with HPP, mainly consisting of the mild childhood form of HPP, treated with AA in Japan. Results One case of perinatal form of HPP, two cases of benign prenatal form, and seven cases of childhood form were observed. The most common symptom at onset was pain. All patients had low serum alkaline phosphatase levels as compared to the age-matched reference range before the commencement of AA. All HPP patients seem to have responded to AA treatment, as evidenced by pain alleviation, increased height standard deviation, improvement in respiratory condition and 6-min walk test result improvement, disappearance of kidney calcification, alleviation of fatigue, and/or increases in bone mineralization. There were no serious adverse events, but all patients had an injection site reaction and skin changes at the injection sites. Genetic analysis showed that eight out of ten patients had compound heterozygosity. Conclusions AA may be effective in patients with mild to severe pediatric-onset forms of HPP. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02230-y.
Collapse
Affiliation(s)
- Yohei Sugiyama
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan.,Department of Neonatology, Chiba Children's Hospital, Chiba, Japan
| | - Taijiro Watanabe
- Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Makiko Tajika
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Tetsuro Matsuhashi
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Masaru Shimura
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Takuya Fushimi
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Keiko Ichimoto
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Ayako Matsunaga
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
| | - Tomohiro Ebihara
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Neonatology, Chiba Children's Hospital, Chiba, Japan
| | - Tomoko Tsuruoka
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan.,Department of Neonatology, Chiba Children's Hospital, Chiba, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama city, Okayama Prefecture, 700-8558, Japan
| | - Kei Murayama
- Center for Medical Genetics, Chiba Children's Hospital, 579-1 Heta-cho, Midori-ku, Chiba City, Chiba Prefecture, 266-0007, Japan. .,Department of Metabolism, Chiba Children's Hospital, Chiba, Japan.
| |
Collapse
|
11
|
Tang H, Zhang Q, Xiang J, Yin L, Wang J, Wang T. Whole Exome Sequencing Aids the Diagnosis of Fetal Skeletal Dysplasia. Front Genet 2021; 12:599863. [PMID: 33777089 PMCID: PMC7987927 DOI: 10.3389/fgene.2021.599863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/27/2021] [Indexed: 11/22/2022] Open
Abstract
Skeletal dysplasia is a complex group of bone and cartilage disorders with strong clinical and genetic heterogeneity. Several types have prenatal phenotypes, and it is difficult to make a molecular diagnosis rapidly. In this study, the genetic cause of 16 Chinese fetuses with skeletal dysplasia were analyzed, and 12 cases yielded positive results including one deletion in DMD gene detected by SNP-array and 14 variants in other 6 genes detected by whole exome sequencing (WES). In addition, somatic mosaicism was observed. Our study expanded the pathogenic variant spectrum and elucidated the utilization of WES in improving the diagnosis yield of skeletal dysplasia.
Collapse
Affiliation(s)
- Hui Tang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Qin Zhang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Jingjing Xiang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Linliang Yin
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Jing Wang
- Suzhou Guangji Hospital, Suzhou, China
| | - Ting Wang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
12
|
Estimation of the carrier frequencies and proportions of potential patients by detecting causative gene variants associated with autosomal recessive bone dysplasia using a whole-genome reference panel of Japanese individuals. Hum Genome Var 2021; 8:2. [PMID: 33452237 PMCID: PMC7810679 DOI: 10.1038/s41439-020-00133-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/04/2022] Open
Abstract
Bone dysplasias are a group of rare hereditary diseases, with up to 436 disease types. Perinatal diagnosis is clinically important for adequate personalized management and counseling. There are no reports focused on pathogenic variants of bone dysplasias in the general population. In this study, we focused on autosomal recessive bone dysplasias. We identified pathogenic variants using whole-genome reference panel data from 3552 Japanese individuals. For the first time, we were able to estimate the carrier frequencies and the proportions of potential patients. For autosomal recessive bone dysplasias, we detected 198 pathogenic variants of 54 causative genes. We estimated the variant carrier frequencies and the proportions of potential patients with variants associated with four clinically important bone dysplasias: osteogenesis imperfecta (OI), hypophosphatasia (HPP), asphyxiating thoracic dysplasia (ATD), and Ellis–van Creveld syndrome (EvC). The proportions of potential patients with OI, ATD, and EvC based on pathogenic variants classified as “pathogenic” and “likely pathogenic” by InterVar were closer to the reported incidence rates in Japanese subjects. Furthermore, the proportions of potential patients with HPP variants classified as “pathogenic” and “likely pathogenic” in InterVar and “pathogenic” in ClinVar were closer to the reported incidence rates. For bone dysplasia, the findings of this study will provide a better understanding of the variant types and frequencies in the Japanese general population, and should be useful for clinical diagnosis, genetic counseling, and personalized medicine. A bioinformatics approach helps estimate carrier frequency of a rare inherited bone disease which causes abnormalities in skeletal shape and structure. Autosomal recessive bone dysplasias affect bone and cartilage development and result from inheriting two mutated genes, one from each parent. Junichi Sugawara, Tohoku University, Sendai, Japan, and colleagues used mutation databases and a bioinformatics tool for variant interpretation to detect 198 pathogenic variants in 54 genes associated with autosomal recessive bone dysplasia in a whole-genome reference panel of 3,552 general Japanese individuals (3.5KJPNv2). They then estimated the frequency of people in the sample carrying bone dysplasia mutations and the expected proportion in whom the disorder could manifest, which compared well with reported incidence rates in the general population. These findings could prove useful for calculating the risk of bone dysplasia in the future children of carrier parents.
Collapse
|
13
|
Matsushita M, Mishima K, Nagata T, Kamiya Y, Imagama S, Kitoh H. Asfotase alfa has a limited effect in improving the bowed limbs in perinatal benign hypophosphatasia: A case report. Clin Pediatr Endocrinol 2021; 30:53-56. [PMID: 33446953 PMCID: PMC7783131 DOI: 10.1297/cpe.30.53] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/27/2020] [Indexed: 01/04/2023] Open
Abstract
Hypophosphatasia (HPP) is a rare skeletal dysplasia characterized by impaired bone
mineralization, caused by loss-of-function mutations in the tissue-nonspecific alkaline
phosphatase (TNSALP) gene. Enzyme replacement therapy (ERT) by
administration of asfotase alfa was reported to improve the survival rate, bone
mineralization, and short stature in the severe form of HPP. However, the effect of
asfotase alfa in improving the skeletal phenotypes for the mild form of HPP has not been
elucidated. We report a case with perinatal benign HPP who had compound heterozygous
mutations of p.F327L and p.R30X in the TNSALP gene. No hypomineralization
was seen in the radiographs from the neonatal period, but bowing of the femurs and ulnares
bilaterally was persistent. ERT was administered during the age of 7.8 to 10.8 yr,
although there was an interruption in the treatment for one year. The bowed femurs and
ulnares were not improved by the treatment with asfotase alfa at the age of 10.8 yr. Bone
mineral density of the lumbar spine was between –0.5 and –1.0 of the z-score, and the
patient’s height was about –2.0 SD during the treatment. Asfotase alfa might have a
limited effect in improving the bowed limbs in perinatal benign hypophosphatasia.
Collapse
Affiliation(s)
- Masaki Matsushita
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenichi Mishima
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Nagata
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasunari Kamiya
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kitoh
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Orthopaedic Surgery, Aichi Children's Health and Medical Center, Aichi, Japan
| |
Collapse
|
14
|
Kato H, Hidaka N, Koga M, Kinoshita Y, Nangaku M, Makita N, Ito N. Altered Thyroid Function Tests Observed in Hypophosphatasia Patients Treated with Asfotase Alfa. Int J Endocrinol 2021; 2021:5492267. [PMID: 34745256 PMCID: PMC8568557 DOI: 10.1155/2021/5492267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Asfotase alfa is the only approved treatment that can normalize mineralization in patients with hypophosphatasia (HPP). Its interference in alkaline phosphatase (ALP) dependent immunoassays has been reported. OBJECTIVE To describe thyroid function tests interfered with by asfotase alfa and elucidate the underlying mechanism. Patients and Methods. Three patients with HPP treated with asfotase alfa were included. Thyroid hormone levels measured using five different immunoassays with or without ALP as a labeling enzyme during asfotase alfa treatment were evaluated. RESULTS After the initiation of asfotase alfa, three HPP patients showed low free triiodothyronine (FT3) and free thyroxine (FT4) measured with AIA-2000 (Tosoh, Tokyo, Japan), an enzyme immunoassay system that uses ALP as a labeling enzyme, but their thyroid-stimulating hormone (TSH) levels were within the normal range. The other CLEIA system using ALP as a label, AIA-CL2400 (Tosoh, Tokyo, Japan), and ALP-independent immunoassay systems demonstrated normal FT3 and FT4 levels. These data suggested that although the thyroid function of these three patients was normal, asfotase alfa interfered with the thyroid hormone measurements made with AIA-2000. AIA-2000 and AIA-CL2400 adopted one-step and delayed one-step measurements, respectively, and the same antibody was used for both immunoassays. However, asfotase alfa may be absorbed on the magnetic beads used in the AIA reagent with the AIA-2000 system but not absorbed on the microparticles used in AIA-CL2400. CONCLUSION Clinicians should be aware of the possible interference in thyroid function measurements by adopting specific types of immunoassays in asfotase alfa-treated HPP patients.
Collapse
Affiliation(s)
- Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Minae Koga
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yuka Kinoshita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Noriko Makita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Nobuaki Ito
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
15
|
Takagi M, Kato S, Muto T, Sano Y, Akiyama T, Takagi J, Okumura A, Iwayama H. Odontohypophosphatasia treated with asfotase alfa enzyme replacement therapy in a toddler: a case report. Clin Pediatr Endocrinol 2020; 29:115-118. [PMID: 32694888 PMCID: PMC7348629 DOI: 10.1297/cpe.29.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 11/12/2022] Open
Abstract
Hypophosphatasia (HPP) is a rare skeletal disorder caused by loss-of-function mutations
in Alkaline Phosphatase, Biomineralization associated (ALPL) gene that
encodes tissue-nonspecific alkaline phosphatase. Odontohypophosphatasia (odonto-HPP), a
mild form of HPP, is characterized only by oral manifestations including premature
exfoliation of deciduous teeth. Enzyme replacement therapy (ERT) is effective in severe
HPP cases; however, information about its efficacy for odonto-HPP is limited. A 2-yr-old
girl was referred to our hospital for mobility of her deciduous teeth with low serum
alkaline phosphatase (ALP) level of 253 U/L (reference range: 410–1,150 U/L) and high
urine phosphoethanolamine level of 1,419.9 µmol/g·Cre (7–70 µmol/g·Cre). She had no
history of bone fractures; however, several members of her family had low serum ALP levels
with a history of pathological fractures. She had a novel heterozygous missense mutation
(c.1183A>T, p.Ile395Phe) in ALPL, and therefore, was diagnosed with
odonto-HPP. After she was provided ERT to prevent premature exfoliation, no tooth mobility
was observed. However, two deciduous teeth exfoliated two months after starting ERT, which
was possibly triggered by a bout of common cold. Starting ERT following tooth mobility
might be relatively late. Previous studies on experimental mice showed that starting ERT
at birth may be effective in preventing premature exfoliation of deciduous teeth.
Collapse
Affiliation(s)
- Mizuki Takagi
- Postgraduate Clinical Training Center, Aichi Medical University Hospital, Aichi, Japan
| | - Shunsuke Kato
- Department of Pediatrics, Kasugai Municipal Hospital, Aichi, Japan
| | - Taichiro Muto
- Department of Pediatrics, Aichi Medical University, School of Medicine, Aichi, Japan
| | - Yoshimi Sano
- Division of Pediatric Dentistry and Orthodontics, Department of Plastic and Reconstructive Surgery, Fujita Health University Department of Plastic Surgery, Aichi, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Hospital, Okayama, Japan
| | - Junko Takagi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | - Akihisa Okumura
- Department of Pediatrics, Aichi Medical University, School of Medicine, Aichi, Japan
| | - Hideyuki Iwayama
- Department of Pediatrics, Aichi Medical University, School of Medicine, Aichi, Japan
| |
Collapse
|
16
|
Nakamura-Takahashi A, Tanase T, Matsunaga S, Shintani S, Abe S, Nitahara-Kasahara Y, Watanabe A, Hirai Y, Okada T, Yamaguchi A, Kasahara M. High-Level Expression of Alkaline Phosphatase by Adeno-Associated Virus Vector Ameliorates Pathological Bone Structure in a Hypophosphatasia Mouse Model. Calcif Tissue Int 2020; 106:665-677. [PMID: 32076747 DOI: 10.1007/s00223-020-00676-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/09/2020] [Indexed: 01/01/2023]
Abstract
Hypophosphatasia (HPP) is a systemic skeletal disease caused by mutations in the gene encoding tissue-nonspecific alkaline phosphatase (TNALP). We recently reported that survival of HPP model mice can be prolonged using an adeno-associated virus (AAV) vector expressing bone-targeted TNALP with deca-aspartate at the C terminus (TNALP-D10); however, abnormal bone structure and hypomineralization remained in the treated mice. Here, to develop a more effective and clinically applicable approach, we assessed whether transfection with TNALP-D10 expressing virus vector at a higher dose than previously used would ameliorate bone structure defects. We constructed a self-complementary AAV8 vector expressing TNALP driven by the chicken beta-actin (CBA) promoter (scAAV8-CB-TNALP-D10). The vector was injected into both quadriceps femoris muscles of newborn HPP mice at a dose of 4.5 × 1012 vector genome (v.g.)/body, resulting in 20 U/mL of serum ALP activity. The 4.5 × 1012 v.g./body-treated HPP mice grew normally and displayed improved bone structure at the knee joints in X-ray images. Micro-CT analysis showed normal trabecular bone structure and mineralization. The mechanical properties of the femur were also recovered. Histological analysis of the femurs demonstrated that ALP replacement levels were sufficient to promote normal, growth plate cartilage arrangement. These results suggest that AAV vector-mediated high-dose TNALP-D10 therapy is a promising option for improving the quality of life (QOL) of patients with the infantile form of HPP.
Collapse
Affiliation(s)
- Aki Nakamura-Takahashi
- Department of Pharmacology, Tokyo Dental College, 2-9-18, Kandamisaki-cho, Chiyoda-ku, Tokyo, 101-0061, Japan.
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan.
| | - Toshiki Tanase
- Department of Pediatric Dentistry, Tokyo Dental College, Tokyo, Japan
| | - Satoru Matsunaga
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Seikou Shintani
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Department of Pediatric Dentistry, Tokyo Dental College, Tokyo, Japan
| | - Shinichi Abe
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Department of Anatomy, Tokyo Dental College, Tokyo, Japan
| | - Yuko Nitahara-Kasahara
- Department of Molecular Therapy, National Center of Neurology and Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| | - Atsushi Watanabe
- Division of Clinical Genetics, Kanazawa University Hospital, Ishikawa, Japan
| | - Yukihiko Hirai
- Division of Molecular and Medical Genetics, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Akira Yamaguchi
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| | - Masataka Kasahara
- Department of Pharmacology, Tokyo Dental College, 2-9-18, Kandamisaki-cho, Chiyoda-ku, Tokyo, 101-0061, Japan.
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan.
| |
Collapse
|
17
|
Michigami T, Tachikawa K, Yamazaki M, Kawai M, Kubota T, Ozono K. Hypophosphatasia in Japan: ALPL Mutation Analysis in 98 Unrelated Patients. Calcif Tissue Int 2020; 106:221-231. [PMID: 31707452 DOI: 10.1007/s00223-019-00626-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022]
Abstract
Hypophosphatasia (HPP) is highly variable in clinical expression and is generally classified into six subtypes. Although it would be beneficial to be able to predict the clinical course from the ALPL genotype, studies on this issue are limited. Here, we aimed to clarify the features of Japanese HPP and the relationships between genotype and clinical manifestations. We analyzed 98 unrelated Japanese patients to investigate the percentage of each clinical form, frequently detected mutations, and the relationship between the genotype and phenotype. Some of the identified mutants were characterized by transfection experiments. Perinatal severe form was the most frequent (45.9%), followed by perinatal benign form (22.4%). Among the 196 alleles, p.Leu520ArgfsX86 (c.1559delT) was detected in 89 alleles, and p.Phe327Leu (c.979T>C) was identified in 23 alleles. All of the homozygotes for p.Leu520ArgfsX86 were classified into perinatal severe form, and patients carrying p.Phe327Leu in one of the alleles were classified into perinatal benign or odonto HPP. Twenty of the 22 patients with perinatal benign HPP were compound heterozygous for p.Phe327Leu and another mutation. Most patients with odonto HPP were found to be monoallelic heterozygotes for dominant-negative mutations or compound heterozygotes with mutants having residual activity. The high prevalence of p.Leu520ArgfsX86 and p.Phe327Leu mutations might underlie the high rate of perinatal severe and perinatal benign forms, respectively, in Japanese HPP. Although ALPL genotyping would be beneficial for predicting the clinical course to an extent, the observed phenotypical variability among patients sharing the same genotypes suggests the presence of modifiers.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, 594-1101, Osaka, Japan.
| | - Kanako Tachikawa
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, 594-1101, Osaka, Japan
| | - Miwa Yamazaki
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, 594-1101, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, 594-1101, Osaka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, 565-0871, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, 565-0871, Osaka, Japan
| |
Collapse
|
18
|
Lee SJ, Lee DW, Kim WD. Case Report of Lethal Perinatal Hypophosphatasia with Seizure and Respiratory Failure Diagnosed by ALPL Gene Mutation. NEONATAL MEDICINE 2020. [DOI: 10.5385/nm.2020.27.1.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
19
|
Michigami T, Ohata Y, Fujiwara M, Mochizuki H, Adachi M, Kitaoka T, Kubota T, Sawai H, Namba N, Hasegawa K, Fujiwara I, Ozono K. Clinical Practice Guidelines for Hypophosphatasia. Clin Pediatr Endocrinol 2020; 29:9-24. [PMID: 32029969 PMCID: PMC6958520 DOI: 10.1297/cpe.29.9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Hypophosphatasia (HPP) is a rare bone disease caused by inactivating mutations in the
ALPL gene, which encodes tissue-nonspecific alkaline phosphatase
(TNSALP). Patients with HPP have varied clinical manifestations and are classified based
on the age of onset and severity. Recently, enzyme replacement therapy using bone-targeted
recombinant alkaline phosphatase (ALP) has been developed, leading to improvement in the
prognosis of patients with life-threatening HPP. Considering these recent advances,
clinical practice guidelines have been generated to provide physicians with guides for
standard medical care for HPP and to support their clinical decisions. A task force was
convened for this purpose, and twenty-one clinical questions (CQs) were formulated,
addressing the issues of clinical manifestations and diagnosis (7 CQs) and those of
management and treatment (14 CQs). A systematic literature search was conducted using
PubMed/MEDLINE, and evidence-based recommendations were developed. The guidelines have
been modified according to the evaluations and suggestions from the Clinical Guideline
Committee of The Japanese Society for Pediatric Endocrinology (JSPE) and public comments
obtained from the members of the JSPE and a Japanese HPP patient group, and then approved
by the Board of Councils of the JSPE. We anticipate that the guidelines will be revised
regularly and updated.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Yasuhisa Ohata
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Hiroshi Mochizuki
- Division of Endocrinology and Metabolism, Saitama Children's Medical Center, Saitama, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Masanori Adachi
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Kanagawa, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Taichi Kitaoka
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Hideaki Sawai
- Department of Obstetrics and Gynecology, Hyogo College of Medicine, Hyogo, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Noriyuki Namba
- Division of Pediatrics and Perinatology, Tottori University Faculty of Medicine, Tottori, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Kosei Hasegawa
- Department of Pediatrics, Okayama University Hospital, Okayama, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Ikuma Fujiwara
- Department of Pediatrics, Sendai City Hospital, Miyagi, Japan.,Task Force for Hypophosphatasia Guidelines
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.,Task Force for Hypophosphatasia Guidelines
| |
Collapse
|
20
|
Fujisawa Y, Kitaoka T, Ono H, Nakashima S, Ozono K, Ogata T. Case Report: Efficacy of Reduced Doses of Asfotase Alfa Replacement Therapy in an Infant With Hypophosphatasia Who Lacked Severe Clinical Symptoms. Front Endocrinol (Lausanne) 2020; 11:590455. [PMID: 33391183 PMCID: PMC7775725 DOI: 10.3389/fendo.2020.590455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 11/12/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Hypophosphatasia is a rare bone disease characterized by impaired bone mineralization and low alkaline phosphatase activity. Here, we describe the course of bone-targeted enzyme replacement therapy with asfotase alpha for a female infant patient with hypophosphatasia who lacked apparent severe clinical symptoms. CASE PRESENTATION The patient exhibited low serum alkaline phosphatase (60 U/L; age-matched reference range, 520-1,580) in a routine laboratory test at birth. Further examinations revealed skeletal demineralization and rachitic changes, as well as elevated levels of serum calcium (2.80 mmol/L; reference range, 2.25-2.75 mmol/L) and ionic phosphate (3.17 mmol/L; reference range, 1.62-2.48 mmol/L), which are typical features in patients with hypophosphatasia. Sequencing analysis of the tissue-nonspecific alkaline phosphatase (TNSALP) gene identified two pathogenic mutations: c.406C>T, p.Arg136Cys and c.979T>C, p.Phe327Leu. Thus, the patient was diagnosed with hypophosphatasia. At the age of 37 days, she began enzyme replacement therapy using asfotase alpha at the standard dose of 6 mg/kg/week. Initial therapy from the age of 37 days to the age of 58 days substantially improved rickets signs in the patient; it also provided immediate normalization of serum calcium and ionic phosphate levels. However, serum ionic phosphate returned to a high level (2.72 mmol/L), which was presumed to be a side effect of asfotase alpha. Thus, the patient's asfotase alfa treatment was reduced to 2 mg/kg/week, which allowed her to maintain normal or near normal skeletal features thereafter, along with lowered serum ionic phosphate levels. Because the patient exhibited slight distal metaphyseal demineralization in the knee at the age of 2 years and 6 months, her asfotase alfa treatment was increased to 2.4 mg/kg/week. No signs of deterioration in bone mineralization were observed thereafter. At the age of 3 years, the patient's motor and psychological development both appeared normal, compared with children of similar age. CONCLUSION This is the first report in which reduced doses of asfotase alfa were administered to an infant patient with hypophosphatasia who lacked apparent severe clinical symptoms. The results demonstrate the potential feasibility of a tailored therapeutic option based on clinical severity in patients with hypophosphatasia.
Collapse
Affiliation(s)
- Yasuko Fujisawa
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
- *Correspondence: Yasuko Fujisawa,
| | - Taichi Kitaoka
- Department of Pediatrics, Faculty of Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroyuki Ono
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichi Nakashima
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Faculty of Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
21
|
Association of ALPL variants with serum alkaline phosphatase and bone traits in the general Japanese population: The Nagahama Study. J Hum Genet 2019; 65:337-343. [PMID: 31857675 DOI: 10.1038/s10038-019-0712-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/28/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022]
Abstract
Although alkaline phosphatase (ALP) activity is relatively low in carriers of recessive type hypophosphatasia (HPP), most are asymptomatic and therefore do not undergo medical evaluations. We analyzed the association of ALP-encoding ALPL variants with serum ALP and bone traits in the general Japanese population. Study participants (n = 9671) were from the Nagahama Study, which was a longitudinal cohort study of an apparently healthy general Japanese population. ALPL variants were analyzed by whole-genome sequencing or TaqMan probe assays using DNA extracted from peripheral blood samples. The speed of sound in calcaneal bone was assessed by quantitative ultrasound (QUS) and used as surrogate measures of bone mineral density. We identified 13 ALPL variants. Minor allele frequencies of three variants were higher than expected. Variant c.529G > A has been reported as a possible pathogenic variant for adult type HPP. Variants c.979C > T and c.1559delT are reported as pathogenic variants for perinatal severe HPP or infantile HPP. The allele frequencies of c.529G > A, c.979C > T, and c.1559delT were 0.0107, 0.0040, and 0.0014, respectively. Serum ALP activity was significantly lower and differed among the three variants (P < 0.001), as well as between individuals with and without any of the three variants (P < 0.001). Serum ALP activity was inversely associated with QUS values, although no direct association was observed between the ALPL variants and QUS values. An association between serum ALP activity and QUS was confirmed; however, we failed to detect an association between ALPL variants and bone traits in the general Japanese population.
Collapse
|
22
|
Michigami T. Skeletal mineralization: mechanisms and diseases. Ann Pediatr Endocrinol Metab 2019; 24:213-219. [PMID: 31905439 PMCID: PMC6944863 DOI: 10.6065/apem.2019.24.4.213] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Skeletal mineralization is initiated in matrix vesicles (MVs), the small extracellular vesicles derived from osteoblasts and chondrocytes. Calcium and inorganic phosphate (Pi) taken up by MVs form hydroxyapatite crystals, which propagate on collagen fibrils to mineralize the extracellular matrix. Insufficient calcium or phosphate impairs skeletal mineralization. Because active vitamin D is necessary for intestinal calcium absorption, vitamin D deficiency is a significant cause of rickets/osteomalacia. Chronic hypophosphatemia also results in rickets/osteomalacia. Excessive action of fibroblast growth factor 23 (FGF23), a key regulator of Pi metabolism, leads to renal Pi wasting and impairs vitamin D activation. X-linked hypophosphatemic rickets (XLH) is the most common form of hereditary FGF23-related hypophosphatemia, and enhanced FGF receptor (FGFR) signaling in osteocytes may be involved in the pathogenesis of this disease. Increased extracellular Pi triggers signal transduction via FGFR to regulate gene expression, implying a close relationship between Pi metabolism and FGFR. An anti-FGF23 antibody, burosumab, has recently been developed as a new treatment for XLH. In addition to various forms of rickets/osteomalacia, hypophosphatasia (HPP) is characterized by impaired skeletal mineralization. HPP is caused by inactivating mutations in tissue-nonspecific alkaline phosphatase, an enzyme rich in MVs. The recent development of enzyme replacement therapy using bone-targeting recombinant alkaline phosphatase has improved the prognosis, motor function, and quality of life in patients with HPP. This links impaired skeletal mineralization with various conditions, and unraveling its pathogenesis will lead to more precise diagnoses and effective treatments.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Japan,Address for correspondence: Toshimi Michigami, MD, PhD Department of Bone and Mineral Research, Research I nstitute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan Tel: +81-725-56-1220 Fax: +81-725-57-3021 E-mail:
| |
Collapse
|
23
|
Mao X, Liu S, Lin Y, Chen Z, Shao Y, Yu Q, Liu H, Lu Z, Sheng H, Lu X, Huang Y, Liu L, Zeng C. Two novel mutations in the ALPL gene of unrelated Chinese children with Hypophosphatasia: case reports and literature review. BMC Pediatr 2019; 19:456. [PMID: 31760938 PMCID: PMC6876108 DOI: 10.1186/s12887-019-1800-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Hypophosphatasia (HPP) is an inherited disorder of defective skeletal mineralization caused by mutations in the ALPL gene that encodes the Tissue Non-specific Alkaline Phosphatase (TNSALP). It is subdivided into six forms depending on the age of onset: perinatal lethal, prenatal benign, infantile, childhood, adult, and odonto HPP. Among these, infantile HPP is characterized by early onset and high frequency of lethal outcome. Few studies have reported the phenotype and genetic characteristics of HPP in Chinese children. CASE PRESENTATION Three forms of HPP were identified in four unrelated patients from four different Chinese families, including one lethal infantile (patient 1), two childhood (patient 2 and 3) and one odonto HPP (patient 4). Six variants in the ALPL gene were identified, including five missense mutations and one frameshift mutation. Of which, none were reported previously in the Chinese population, and two were novel (c.359G > C: p.G120A and c.1017dupG: p.H340AfsX3). Patient 1 carrying a novel homozygous (c.359G > C) mutation showed respiratory distress and pneumonia at first day of his life. He presented nearly negligible level of serum ALP activity, overall skeletal hypominaralization and died at 3 months old. Patient 2, 3 and 4 were compound heterozygotes with decreased serum ALP activity. Patient 2 and 3 presented premature loss of deciduous teeth, muscle weakness and bone pain, whereas patient 4 had early loss of deciduous teeth only. All four pedigrees exhibited autosomal recessive pattern of inheritance. CONCLUSIONS In this study, six mutations in the ALPL gene were found in four Chinese HPP patients, two of which were novel: c.359G > C in exon 5 and c.1017dupG in exon 10. Our results strongly indicated that the novel mutation c.359G > C might be disease-causing and associated with severe infantile form of HPP.
Collapse
Affiliation(s)
- Xiaojian Mao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Sichi Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Zhen Chen
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd., Guangzhou, 510623, China
| | - Yongxian Shao
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Qiaoli Yu
- Department of Dentistry, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd., Guangzhou, 510623, China
| | - Haiying Liu
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd., Guangzhou, 510623, China
| | - Zhikun Lu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Huiyin Sheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Xinshuo Lu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Yonglan Huang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China.
| | - Chunhua Zeng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Rd, Guangzhou, 510623, China.
| |
Collapse
|
24
|
Yokoi K, Nakajima Y, Shinkai Y, Sano Y, Imamura M, Akiyama T, Yoshikawa T, Ito T, Kurahashi H. Clinical and genetic aspects of mild hypophosphatasia in Japanese patients. Mol Genet Metab Rep 2019; 21:100515. [PMID: 31641588 PMCID: PMC6796780 DOI: 10.1016/j.ymgmr.2019.100515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/05/2019] [Accepted: 09/02/2019] [Indexed: 11/29/2022] Open
Abstract
Background Hypophosphatasia (HPP) is a rare inborn error of metabolism that results from a dysfunctional tissue non-specific alkaline phosphatase enzyme (TNSALP). Although genotype-phenotype correlations have been described in HPP patients, only sparse information is currently available on the genetics of mild type HPP. Methods We investigated 5 Japanese patients from 3 families with mild HPP (patients 1 and 2 are siblings; patient 4 is a daughter of patient 5) who were referred to Fujita Health University due to the premature loss of deciduous teeth. Physical and dental examinations, and blood, urine and bone density tests were conducted. Genetic analysis of the ALPL gene was performed in all patients with their informed consent. Results After a detailed interview and examination, we found characteristic symptoms of HPP in some of the study cases. Mobile teeth or the loss of permanent teeth were observed in 2 patients, and 3 out of 5 patients had a history of asthma. The serum ALP levels of all patients were 30% below the lower limit of the age equivalent normal range. ALPL gene analysis revealed compound heterozygous mutations, including Ile395Val and Leu520Argfs in family 1, Val95Met and Gly491Arg in family 2, and a dominant missense mutation (Gly456Arg) in family 3. The 3D-modeling of human TNSALP revealed three mutations (Val95Met, Ile395Val and Gly456Arg) at the homodimer interface. Severe collisions between the side chains were predicted for the Gly456Arg variant. Discussion One of the characteristic findings of this present study was a high prevalence of coexisting asthma and a high level serum IgE level. These characteristics may account for the fragility of tracheal tissues and a predisposition to asthma in patients with mild HPP. The genotypes of the five mild HPP patients in our present study series included 1) compound heterozygous for severe and hypomorphic mutations, and 2) dominant-negative mutations. All of these mutations were at the homodimer interface, but only the dominant-negative mutation was predicted to cause a severe collision effect between the side chains. This may account for varying mechanisms leading to different effects on TNSALP function.
Collapse
Affiliation(s)
- Katsuyuki Yokoi
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan.,Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| | - Yoko Nakajima
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Yasuko Shinkai
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| | - Yoshimi Sano
- Department of Plastic Surgery, Division of Pediatric Dentistry & Orthodontics, Fujita Health University of Medicine, Toyoake 470-1192, Japan
| | - Mototaka Imamura
- Department of Plastic Surgery, Division of Pediatric Dentistry & Orthodontics, Fujita Health University of Medicine, Toyoake 470-1192, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Tetsuya Ito
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Hiroki Kurahashi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake 470-1192, Japan
| |
Collapse
|
25
|
Okawa R, Kokomoto K, Kitaoka T, Kubota T, Watanabe A, Taketani T, Michigami T, Ozono K, Nakano K. Japanese nationwide survey of hypophosphatasia reveals prominent differences in genetic and dental findings between odonto and non-odonto types. PLoS One 2019; 14:e0222931. [PMID: 31600233 PMCID: PMC6786601 DOI: 10.1371/journal.pone.0222931] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 09/10/2019] [Indexed: 12/03/2022] Open
Abstract
Hypophosphatasia (HPP) is a rare and intractable metabolic bone disease caused by mutations in the ALPL gene. Here, we undertook a nationwide survey of HPP in Japan, specifically regarding the prominent genetic and dental manifestations of odonto (n = 16 cases) and other (termed “non-odonto”) (n = 36 cases) types. Mean serum alkaline phosphatase (ALP) values in odonto-type patients were significantly greater than those of non-odonto-type patients (P<0.05). Autosomal dominant and autosomal recessive inheritance patterns were detected, respectively, in 89% of odonto-type and 96% of non-odonto-type patients. The ALPL “c.1559delT” mutation, associated with extremely low ALP activity, was found in approximately 70% of cases. Regarding dental manifestations, all patients classified as odonto-type showed early exfoliation of the primary teeth significantly more frequently than patients classified as non-odonto-type (100% vs. 56%; P<0.05). Tooth hypomineralisation was detected in 42% of non-odonto-type patients, but not in any odonto-type patients (0%; P<0.05). Collectively, these results suggest that genetic and dental manifestations of patients with odonto-type and non-odonto-type HPP are significantly different, and these differences should be considered during clinical treatment of patients with HPP.
Collapse
Affiliation(s)
- Rena Okawa
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
- * E-mail:
| | - Kazuma Kokomoto
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Taichi Kitaoka
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Watanabe
- Division of Clinical Genetics, Kanazawa University Hospital, Ishikawa, Japan
| | - Takeshi Taketani
- Department of Pediatrics, Shimane University Faculty of Medicine, Shimane, Japan
| | - Toshimi Michigami
- Department of Pediatric Nephrology and Metabolism, and Department of Bone and Mineral Research, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
26
|
Ishiguro T, Sugiyama Y, Ueda K, Muramatsu Y, Tsuda H, Kotani T, Michigami T, Tachikawa K, Akiyama T, Hayakawa M. Findings of amplitude-integrated electroencephalogram recordings and serum vitamin B6 metabolites in perinatal lethal hypophosphatasia during enzyme replacement therapy. Brain Dev 2019; 41:721-725. [PMID: 31000369 DOI: 10.1016/j.braindev.2019.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/17/2019] [Accepted: 03/29/2019] [Indexed: 11/24/2022]
Abstract
Hypophosphatasia (HPP) is a rare disorder caused by low serum tissue non-specific alkaline phosphatase (ALP) activity due to hypomorphic mutations in the ALPL gene. HPP is characterized by defective bone mineralization. It frequently accompanies pyridoxine-responsive seizures. Because alkaline phosphatase change pyridoxal 5' phosphate (PLP) into pyridoxal (PL), which can cross the blood brain barrier and regulates inhibitory neurotransmitter gamma-aminobutyric acid. The female patient was born at a gestational age of 37 weeks 2 days. She presented severe respiratory disorder due to extreme thoracic hypoplasia. With the extremely low serum ALP value (14 IU/L), she was clinically diagnosed as HPP. The diagnosis was confirmed with genetic testing. On day1, the subclinical seizures were detected by aEEG. Together with enzyme replacement therapy by asfotase alfa, pyridoxine hydrochloride was administered, then the seizures were rapidly controlled. While confirming that there was no seizure by aEEG monitoring, pyridoxine hydrochloride was gradually discontinued after 1 month. Before administration of pyridoxine hydrochloride, PL was extremely low (4.7 nM) and PLP was increased (1083 nM). After the withdrawal, PL was increased to 84.9 nM only by enzyme replacement. Monitoring with aEEG enabled early intervention for pyridoxine responsive seizures. Confirming increased serum PL concentration is a prudent step in determining when to reduce or discontinue pyridoxine hydrochloride during enzyme replacement therapy.
Collapse
Affiliation(s)
- Tomonori Ishiguro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichiro Sugiyama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.
| | - Kazuto Ueda
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Yukako Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Tsuda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Kanako Tachikawa
- Department of Bone and Mineral Research, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
27
|
Ishijima Y, Iizuka T, Kagami K, Masumoto S, Nakade K, Mitani Y, Niida Y, Watanabe A, Yamazaki R, Ono M, Fujiwara H. Prenatal diagnosis facilitated prompt enzyme replacement therapy for prenatal benign hypophosphatasia. J OBSTET GYNAECOL 2019; 40:132-134. [PMID: 31335231 DOI: 10.1080/01443615.2019.1606177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Yuka Ishijima
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Sakiko Masumoto
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kyohei Nakade
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Mitani
- Department of Pediatrics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, Uchinada, Japan
| | - Atsushi Watanabe
- Clinical Genetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Rena Yamazaki
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynaecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
28
|
Nakano C, Kitabatake Y, Takeyari S, Ohata Y, Kubota T, Taketani K, Kogo M, Ozono K. Genetic correction of induced pluripotent stem cells mediated by transcription activator-like effector nucleases targeting ALPL recovers enzyme activity and calcification in vitro. Mol Genet Metab 2019; 127:158-165. [PMID: 31178256 DOI: 10.1016/j.ymgme.2019.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/25/2019] [Accepted: 05/25/2019] [Indexed: 01/22/2023]
Abstract
Hypophosphatasia (HPP) is an inheritable disease affecting both skeletal systems and extra-skeletal organs due to mutations of the gene ALPL, which encodes tissue-nonspecific alkaline phosphatase. Recently, an enzyme replacement therapy using asfotase alfa was developed to ameliorate the complications of HPP. However, it requires frequent injections and is expensive to maintain. As an alternative, cell and gene therapy using human induced pluripotent stem cells (iPSCs) after precise correction of the mutation is feasible due to advances in genome-editing technology. In the study, we examined the alkaline phosphatase (ALP) activity and calcification in vitro of two childhood HPP patient-derived iPSCs after the correction of the c.1559delT mutation, which is the most frequent mutation in Japanese patients with HPP, using transcription activator-like effector nucleases (TALENs). The gene correction targeting vector was designed for site-directed mutagenesis using TALEN. After selection with antibiotics, some clones with the selection cassette were obtained. Gene correction was confirmed by Sanger sequencing. The mutation was corrected in one allele of ALPL in homozygous patients and compound heterozygous patients. The correction of ALPL did not result in an increase in ALP when the selection cassette remained. Conversely, iPSCs exhibited ALP activity after the elimination of the cassette using Cre/LoxP. The quantitative analysis showed the half ALP activity in corrected iPSCs of that of control iPSCs, corresponding to heterozygous correction of the mutation. In addition, osteoblasts differentiated from the corrected iPSCs exhibited high ALP activity and some calcification in vitro. Moreover, the osteoblast-like phenotype was confirmed by increased expression of osteoblast-specific genes such as COL1A1 and osteocalcin. These results suggest that gene correction in iPSCs may be a candidate treatment for HPP patients.
Collapse
Affiliation(s)
- Chiho Nakano
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan; Unit of Dentistry, Osaka University Hospital, Osaka, Japan
| | - Yasuji Kitabatake
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinji Takeyari
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasuhisa Ohata
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ken Taketani
- Department of Pediatrics, Shimane University, Osaka, Japan
| | - Mikihiko Kogo
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
29
|
Fukushima K, Kawai-Kowase K, Yonemoto Y, Fujiwara M, Sato H, Sato M, Kubota T, Ozono K, Tamura J. Adult hypophosphatasia with compound heterozygous p.Phe327Leu missense and c.1559delT frameshift mutations in tissue-nonspecific alkaline phosphatase gene: a case report. J Med Case Rep 2019; 13:101. [PMID: 31014398 PMCID: PMC6480864 DOI: 10.1186/s13256-019-2045-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 03/08/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Hypophosphatasia is an inherited bone disease characterized by low alkaline phosphatase activity encoded by ALPL. Clinically, hypophosphatasia can be categorized as perinatal, infantile, childhood, and adult forms, as well as odonto-hypophosphatasia, according to the age at first sign or dental manifestations. Adult hypophosphatasia typically presents in middle-aged patients who appear to be in good health in early adulthood and manifests as painful feet caused by recurrent, slow-healing stress fractures of the lower limb. Because the symptoms of adult hypophosphatasia vary and are common, many patients with hypophosphatasia might be not diagnosed accurately and thus may receive inappropriate treatment. CASE PRESENTATION We report a case of a 35-year-old Japanese woman with low serum alkaline phosphatase detected at a routine medical checkup. She had mild muscle/bone pain but no history of rickets, fractures, or dental problems. Measurement of bone mineral density of the lumbar spine and the femoral neck revealed osteopenia below the expected range for age in a young adult. Abdominal ultrasonography revealed numerous microcalcifications in both kidneys. Analysis of amino acids in urine revealed that phosphoethanolamine was elevated. Low serum alkaline phosphatase activity, elevation of phosphoethanolamine, and low bone mineral density supported the diagnosis of hypophosphatasia. ALPL mutation analysis revealed two mutations: p.Phe327Leu and c.1559delT. These genetic abnormalities were previously reported in perinatal, infantile, and childhood but not adult hypophosphatasia. On the basis of the clinical presentation, laboratory and imaging findings, and genetic analyses, the patient was definitively diagnosed with adult hypophosphatasia. To the best of our knowledge, this is the first case report of adult hypophosphatasia with the compound heterozygous mutations p.Phe327Leu and c.1559delT. CONCLUSIONS Although the risk of bone fracture was high in this case, treatment approaches differ between osteoporosis and hypophosphatasia. Because adult hypophosphatasia diagnosis is often difficult because of their varied symptoms, hypophosphatasia should be considered in the differential diagnosis of low serum alkaline phosphatase. Early diagnosis is important so that appropriate treatment can be initiated.
Collapse
Affiliation(s)
- Kazunori Fukushima
- Department of Emergency Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Keiko Kawai-Kowase
- Department of General Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Yukio Yonemoto
- Department of Orthopedic Surgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroko Sato
- Department of General Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Mahito Sato
- Department of General Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Junich Tamura
- Department of General Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
30
|
Abstract
Hypophosphatasia (HPP) is a rare inherited disorder primarily affecting bone and dental mineralization. Although there is a continuum in the severity of the disease, clinical forms may be arbitrarily distinguished on the basis of age at onset and the presence or absence of bone symptoms: perinatal, infantile, juvenile, adult, prenatal benign, and odontological. Severe forms (perinatal and infantile) are autosomally recessively inherited while less severe forms may be autosomally recessively or dominantly inherited. Genetic counseling is complicated by the coexistence of the two modes of inheritance, the incomplete penetrance of the dominant forms, the markedly variable expression of the disease, including intra-familial expression, and the existence of a benign prenatal form that may sometimes be difficult to distinguish from the severe prenatal form. The disease is due to loss-of-function mutations in the Alkaline Phosphatase-Liver (ALPL) gene encoding the tissue nonspecific alkaline phosphatase (TNSALP). The great variety of missence mutations and the dominant negative effect of some mutations largely explain the clinical heterogeneity. Directed mutagenesis studies allowed further elucidation of the cellular pathophysiology of HPP, classification of the alleles in terms of their severity and dominant negative effect, and molecular explanations of the dominant inheritance mode. Genetics significantly contributed to show that there are in fact two HPPs, rare, severe and recessive HPP, and mild recessive or mild dominant HPP, which is markedly more frequent and probably under-diagnosed. The prevalence of the severe forms of HPP has been estimated to be 1/300,000 in France and Northern Europe while the prevalence of the moderate forms of HPP may reach 1/6,370.
Collapse
Affiliation(s)
- E Mornet
- Service de biologie, unité de génétique constitutionnelle, centre hospitalier de Versailles, Le Chesnay, France.
| |
Collapse
|
31
|
Misawa A, Orimo H. lncRNA HOTAIR Inhibits Mineralization in Osteoblastic Osteosarcoma Cells by Epigenetically Repressing ALPL. Calcif Tissue Int 2018; 103:422-430. [PMID: 29846771 DOI: 10.1007/s00223-018-0434-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/24/2018] [Indexed: 12/23/2022]
Abstract
HOTAIR is a lncRNA that plays critical role in gene regulation and chromatin dynamics through epigenetic mechanisms. In this work we studied the physiological role of HOTAIR during the process of mineralization using osteoblastic osteosarcoma cells focusing in ALPL (Tissue Non-Specific Alkaline Phosphatase), a pivotal gene that controls bone formation. HOTAIR knockdown resulted in upregulation of ALPL, increase of alkaline phosphatase (ALP) activity, and enhanced mineralization in osteoblastic SaOS-2 cells cultured in mineralizing medium. Luciferase assays using reporter vectors containing ALPL promoter showed that HOTAIR repression increases ALPL promoter activity. Furthermore, HOTAIR knockdown increased histone H3K4 methylation levels at ALPL promoter region, suggesting that ALPL repression by HOTAIR is regulated by epigenetic mechanisms. This work supports that physiological bone formation is epigenetically regulated by a lncRNA.
Collapse
Affiliation(s)
- Aya Misawa
- Division of Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Hideo Orimo
- Division of Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
32
|
Akiyama T, Kubota T, Ozono K, Michigami T, Kobayashi D, Takeyari S, Sugiyama Y, Noda M, Harada D, Namba N, Suzuki A, Utoyama M, Kitanaka S, Uematsu M, Mitani Y, Matsunami K, Takishima S, Ogawa E, Kobayashi K. Pyridoxal 5'-phosphate and related metabolites in hypophosphatasia: Effects of enzyme replacement therapy. Mol Genet Metab 2018; 125:174-180. [PMID: 30049651 DOI: 10.1016/j.ymgme.2018.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/12/2018] [Accepted: 07/12/2018] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To investigate the utility of serum pyridoxal 5'-phosphate (PLP), pyridoxal (PL), and 4-pyridoxic acid (PA) as a diagnostic marker of hypophosphatasia (HPP) and an indicator of the effect of, and patient compliance with, enzyme replacement therapy (ERT), we measured PLP, PL, and PA concentrations in serum samples from HPP patients with and without ERT. METHODS Blood samples were collected from HPP patients and serum was frozen as soon as possible (mostly within one hour). PLP, PL, and PA concentrations were analyzed using high-performance liquid chromatography with fluorescence detection after pre-column derivatization by semicarbazide. We investigated which metabolites are associated with clinical phenotypes and how these metabolites change with ERT. RESULTS Serum samples from 20 HPP patients were analyzed. The PLP-to-PL ratio and PLP concentration were elevated in all HPP patients. They correlated negatively with serum alkaline phosphatase (ALP) activity and showed higher values in more severe phenotypes (perinatal severe and infantile HPP) compared with other phenotypes. PL concentration was reduced only in perinatal severe HPP. ERT reduced the PLP-to-PL ratio to mildly reduced or low-normal levels and the PLP concentration was reduced to normal or mildly elevated levels. Urine phosphoethanolamine (PEA) concentration did not return to normal levels with ERT in most patients. CONCLUSIONS The serum PLP-to-PL ratio is a better indicator of the effect of ERT for HPP than serum PLP and urine PEA concentrations, and a PLP-to-PL ratio of <4.0 is a good indicator of the effect of, and patient compliance with, ERT.
Collapse
Affiliation(s)
- Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Daisuke Kobayashi
- Department of Food and Chemical Toxicology, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Hokkaido, Japan
| | - Shinji Takeyari
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuichiro Sugiyama
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Masahiro Noda
- Department of Pediatrics, Showa General Hospital, Tokyo, Japan
| | - Daisuke Harada
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Noriyuki Namba
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Atsushi Suzuki
- Department of Neonatology and Pediatrics, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Maiko Utoyama
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Sachiko Kitanaka
- Department of Pediatrics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Mitsugu Uematsu
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yusuke Mitani
- Department of Pediatrics, Kanazawa University Hospital, Ishikawa, Japan
| | - Kunihiro Matsunami
- Department of Pediatrics, Gifu Prefectural General Medical Center, Gifu, Japan
| | | | - Erika Ogawa
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Katsuhiro Kobayashi
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
33
|
Four novel mutations in the ALPL gene in Chinese patients with odonto, childhood, and adult hypophosphatasia. Biosci Rep 2018; 38:BSR20171377. [PMID: 29724887 PMCID: PMC6131208 DOI: 10.1042/bsr20171377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 01/17/2023] Open
Abstract
Hypophosphatasia (HPP) is a rare inherited disorder characterized by defective bone and/or dental mineralization, and decreased serum alkaline phosphatase (ALP) activity. ALPL, the only gene related with HPP, encodes tissue non-specific ALP (TNSALP). Few studies were carried out in ALPL gene mutations in the Chinese population with HPP. The purpose of the present study is to elucidate the clinical and genetic characteristics of HPP in five unrelated Chinese families and two sporadic patients. Ten clinically diagnosed HPP patients from five unrelated Chinese families and two sporadic patients and fifty healthy controls were genetically investigated. All 12 exons and exon–intron boundaries of the ALPL gene were amplified by PCR and directly sequenced. The laboratory and radiological investigations were conducted simultaneously in these HPP ten patients. A 3D model of the TNSALP was used to predict the dominant negative effect of identified missense mutations. Three odonto, three childhood, and four adult types of HPP were clinically diagnosed. Ten mutations were identified in five unrelated Chinese families and two sporadic patients, including eight missense mutations and two frameshift mutations. Of which, four were novel: one frameshift mutation (p.R138Pfsx45); three missense mutations (p.C201R, p.V459A, p.C497S). No identical mutations and any other new ALPL mutations were found in unrelated 50 healthy controls. Our study demonstrated that the ALPL gene mutations are responsible for HPP in these Chinese families. These findings will be useful for clinicians to improve understanding of this heritable bone disorder.
Collapse
|
34
|
Oyachi M, Harada D, Sakamoto N, Ueyama K, Kondo K, Kishimoto K, Izui M, Nagamatsu Y, Kashiwagi H, Yamamuro M, Tamura M, Kikuchi S, Akiyama T, Michigami T, Seino Y, Namba N. A case of perinatal hypophosphatasia with a novel mutation in the ALPL gene: clinical course and review of the literature. Clin Pediatr Endocrinol 2018; 27:179-186. [PMID: 30083035 PMCID: PMC6073057 DOI: 10.1297/cpe.27.179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/24/2018] [Indexed: 11/28/2022] Open
Abstract
Hypophosphatasia (HPP) is a metabolic bone disease characterized by failure of bone
calcification and vitamin B6 dependent seizures. It is caused by loss-of-function
mutations in the ALPL gene. A newborn girl required respiratory support
by nasal-directional positive airway pressure at birth, and pyridoxine hydrochloride
administration for vitamin B6-dependent seizures observed from day two. Umbilical cord
blood showed low alkaline phosphatase (ALP) activity and high pyridoxal phosphate levels.
Radiographs showed severe rickets-like appearance of the bones. Genetic analysis of the
ALPL gene revealed compound heterozygous mutations,
c.1559delT/p.Ser188Pro. We diagnosed her with perinatal severe HPP, and started the
patient on asfotase alfa from day six. Following enzyme replacement therapy (ERT),
skeletal mineralization and respiratory insufficiency improved with no remarkable
side-effects. Crying vital capacity (CVC) was used to evaluate respiratory status, which
continuously improved from 13.3 mL/kg (day 22) to 20.6 mL/kg (day 113). Since no seizures
occurred, pyridoxine hydrochloride was tapered off at one year of age. Strategies to
manage perinatal severe HPP cases following ERT have not been established till date. A
review of the literature shows that CVC may be a good indicator for weaning from
ventilatory support. In addition, ERT will most likely enable withdrawal of pyridoxine
treatment.
Collapse
Affiliation(s)
- Maki Oyachi
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Daisuke Harada
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Natsuko Sakamoto
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Kaoru Ueyama
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Kawai Kondo
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Kanako Kishimoto
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Masafumi Izui
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Yuiko Nagamatsu
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Hiroko Kashiwagi
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Miho Yamamuro
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Makoto Tamura
- Department of Pediatrics and Neonatology, Takatsuki General Hospital, Osaka, Japan
| | - Shin Kikuchi
- Department of Pediatrics and Neonatology, Takatsuki General Hospital, Osaka, Japan
| | - Tomoyuki Akiyama
- Department of Child Neurology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute of Osaka Women's and Children's Hospital, Osaka, Japan
| | - Yoshiki Seino
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| | - Noriyuki Namba
- Department of Pediatrics, Osaka Hospital, Japan Community Healthcare Organization (JCHO), Osaka, Japan
| |
Collapse
|
35
|
Abstract
We review here clinical, pathophysiological, diagnostic, genetic and molecular aspects of Hypophosphatasia (HPP), a rare inherited metabolic disorder. The clinical presentation is a continuum ranging from a prenatal lethal form with no skeletal mineralization to a mild form with late adult onset presenting with nonpathognomonic symptoms. The prevalence of severe forms is low, whereas less severe forms are more frequently observed. The disease is caused by loss-of-function mutations in the ALPL gene encoding the Tissue Nonspecific Alkaline Phosphatase (TNSALP), a central regulator of mineralization. Severe forms are recessively inherited, whereas moderate forms are either recessively or dominantly inherited, and the more severe the disease is, the more often it is subject to recessive inheritance. The diagnosis is based on a constantly low alkaline phosphatase (AP) activity in serum and genetic testing that identifies ALPL mutations. More than 340 mutations have been identified and are responsible for the extraordinary clinical heterogeneity. A clear but imperfect genotype-phenotype correlation has been observed, suggesting that other genetic or environmental factors modulate the phenotype. Enzyme replacement therapy is now available for HPP, and other approaches, such as gene therapy, are currently being investigated.
Collapse
Affiliation(s)
- Etienne Mornet
- Unité de Génétique Constitutionnelle, Service de Biologie, Centre Hospitalier de Versailles, 177 rue de Versailles, 78150 Le Chesnay, France.
| |
Collapse
|
36
|
Ikenoue S, Miyakoshi K, Ishii T, Sato Y, Otani T, Akiba Y, Kasuga Y, Ochiai D, Matsumoto T, Ichihashi Y, Matsuzaki Y, Tachikawa K, Michigami T, Nishimura G, Ikeda K, Hasegawa T, Tanaka M. Discordant fetal phenotype of hypophosphatasia in two siblings. Am J Med Genet A 2017; 176:171-174. [PMID: 29160033 DOI: 10.1002/ajmg.a.38531] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/12/2017] [Accepted: 10/15/2017] [Indexed: 11/11/2022]
Abstract
Hypophosphatasia (HPP) is an autosomal recessive metabolic disorder with impaired bone mineralization due to mutations in the ALPL gene. The genotype-phenotype correlation of this disorder has been widely described. Here, we present two affected siblings, whose fetal phenotypes were discordant. A 31-year-old Japanese woman, G0P0, was referred to our institution because of fetal micromelia. After obstetric counseling, the pregnancy was terminated at 21 weeks' gestation. Post-mortem radiographs demonstrated severely defective mineralization of the skeleton. The calvarial, spinal, and tubular bones were mostly missing. Only the occipital bones, mandible, clavicles, ribs, one thoracic vertebra, ilia, and tibia were relatively well ossified. The radiological findings suggested lethal HPP. Genetic testing for genomic DNA extracted from the umbilical cord identified compound heterozygous mutations in the ALPL gene (c.532T>C, p.Y178H; c.1559delT, p.Leu520Argfs*86). c.532T>C was a novel variant showing no residual activity of the protein by the functional analysis. The parents were heterozygous carriers. In the next pregnancy, biometric values on fetal ultrasonography at 20 and 26 weeks' gestation were normal. At 34 weeks, however, a small chest and shortening of distal long bones came to attention. The neonate delivered at 41 weeks showed serum ALP of <5U/L. Radiological examination showed only mild thoracic hypoplasia and metaphyseal mineralization defects of the long bones. ALP replacement therapy was introduced shortly after birth, and the neonate was discharged at day 22 without respiratory distress. Awareness of discordant fetal phenotypes in siblings with HPP precludes a diagnostic error, and enables early medical intervention to mildly affected neonates.
Collapse
Affiliation(s)
- Satoru Ikenoue
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kei Miyakoshi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tomohiro Ishii
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Yu Sato
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Toshimitsu Otani
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Akiba
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshifumi Kasuga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daigo Ochiai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tadashi Matsumoto
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yosuke Ichihashi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Matsuzaki
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Kanako Tachikawa
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women's and Children's Hospital, Osaka, Japan
| | - Gen Nishimura
- Department of Pediatric Imaging, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Kazushige Ikeda
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.,Division of Neonatology, Perinatal Center for Maternal and Child Health, Saitama City Hospital, Saitama, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
37
|
Kitaoka T, Tajima T, Nagasaki K, Kikuchi T, Yamamoto K, Michigami T, Okada S, Fujiwara I, Kokaji M, Mochizuki H, Ogata T, Tatebayashi K, Watanabe A, Yatsuga S, Kubota T, Ozono K. Safety and efficacy of treatment with asfotase alfa in patients with hypophosphatasia: Results from a Japanese clinical trial. Clin Endocrinol (Oxf) 2017; 87:10-19. [PMID: 28374482 DOI: 10.1111/cen.13343] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Hypophosphatasia (HPP) is a rare skeletal disease characterized by hypomineralization and low alkaline phosphatase activity. Asfotase alfa (AA) has been recently developed to treat HPP complications. This study evaluated its safety and efficacy in Japan. DESIGN Open-label, multicentre, prospective trial. Patients were enrolled in 11 hospitals from June 2014 to July 2015. PATIENTS Thirteen patients (9 females, 4 males) ages 0 days to 34 years at baseline were enrolled and treated with AA (2 mg/kg three times weekly subcutaneously in all but one patient). All had ALPL gene mutations. HPP forms were perinatal (n=6), infantile (n=5), childhood (n=1) and adult (n=1). MEASUREMENTS Safety determined from adverse events (AEs) and laboratory data was the primary outcome measure. Efficacy was assessed as a secondary outcome measure from overall survival, respiratory status, rickets severity and gross motor development. RESULTS Injection site reactions were the most frequent AEs. Serious AEs possibly related to treatment were convulsion and hypocalcaemia observed in a patient with the perinatal form. In addition, hypercalcaemia and/or hyperphosphatemia was observed in three patients with the infantile form and a low-calcium and/or low-phosphate formula was given to these patients. With respect to efficacy, all patients survived and the radiographic findings, developmental milestones and respiratory function improved. CONCLUSION Asfotase alfa therapy improved skeletal, respiratory and physical symptoms with a few serious AEs in patients with HPP. Our results add support to the safety and efficacy of AA therapy for HPP patients.
Collapse
Affiliation(s)
- Taichi Kitaoka
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshihiro Tajima
- Department of Pediatrics, Hokkaido University School of Medicine, Sapporo, Japan
| | - Keisuke Nagasaki
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toru Kikuchi
- Division of Pediatrics, Department of Homeostatic Regulation and Development, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Katsusuke Yamamoto
- Department of Pediatric Nephrology and Metabolism, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Toshimi Michigami
- Department of Bone and Mineral Research, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Ikuma Fujiwara
- Department of Pediatrics, Tohoku University School of Medicine, Miyagi, Japan
| | - Masayuki Kokaji
- Department of Pediatrics, Showa General Hospital, Tokyo, Japan
| | - Hiroshi Mochizuki
- Division of Endocrinology and Metabolism, Saitama Children's Medical Center, Saitama, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Atsushi Watanabe
- Division of Clinical Genetics, Nippon Medical School Hospital, Tokyo, Japan
| | - Shuichi Yatsuga
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Fukuoka, Japan
| | - Takuo Kubota
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
38
|
Tenorio J, Álvarez I, Riancho-Zarrabeitia L, Martos-Moreno GÁ, Mandrile G, de la Flor Crespo M, Sukchev M, Sherif M, Kramer I, Darnaude-Ortiz MT, Arias P, Gordo G, Dapía I, Martinez-Villanueva J, Gómez R, Iturzaeta JM, Otaify G, García-Unzueta M, Rubinacci A, Riancho JA, Aglan M, Temtamy S, Hamid MA, Argente J, Ruiz-Pérez VL, Heath KE, Lapunzina P. Molecular and clinical analysis ofALPLin a cohort of patients with suspicion of Hypophosphatasia. Am J Med Genet A 2017; 173:601-610. [DOI: 10.1002/ajmg.a.37991] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/18/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Jair Tenorio
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
| | | | | | - Gabriel Á. Martos-Moreno
- Department of Endocrinology; Hospital Universitario Niño Jesús, IIS La Princesa; Madrid Spain
- Department of Pediatrics; Universidad Autónoma de Madrid; Madrid Spain
- CIBEROBN, Centro de Investigación Biomédica en Red sobre Fisiopatología de la obesidad y nutrición; Instituto de Salud Carlos III; Madrid Spain
| | - Giorgia Mandrile
- Department of Medical Genetics; San Luigi University Hospital; Orbassano Italy
- Department Clinical and Biological Sciences; University of Torino; Torino Italy
| | | | - Mikhail Sukchev
- Diagnostic Specialist; Alexion Pharmaceuticals; Moscow Russia
| | - Mostafa Sherif
- Medical Division; Alexion Pharma Middle East; Dubai Media City United Arab Emirates
| | - Iza Kramer
- Department of Pediatrics; Privat Hospitalet Denmark; Charlottenlund Denmark
| | | | - Pedro Arias
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
| | - Gema Gordo
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
| | - Irene Dapía
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
| | | | - Rubén Gómez
- Department of Biochemistry; Hospital Universitario La Paz, IdiPaz; Madrid Spain
| | | | - Ghada Otaify
- Division of Human Genetics and Genome Research; Department of Clinical Genetics; National Research Centre, Cairo; Egypt
- Centre of Excellence for Human Genetics; National Research Centre; Cairo Egypt
| | - Mayte García-Unzueta
- Department of Clinical Biochemistry; Hospital Universitario Marqués Valdecilla, IDIVAL; Santander Spain
| | | | - José A. Riancho
- Department of Internal Medicine; Hospital Universitario Marqués Valdecilla, IDIVAL; University of Cantabria, RETICEF; Santander Spain
| | - Mona Aglan
- Division of Human Genetics and Genome Research; Department of Clinical Genetics; National Research Centre, Cairo; Egypt
- Centre of Excellence for Human Genetics; National Research Centre; Cairo Egypt
| | - Samia Temtamy
- Division of Human Genetics and Genome Research; Department of Clinical Genetics; National Research Centre, Cairo; Egypt
- Centre of Excellence for Human Genetics; National Research Centre; Cairo Egypt
| | - Mohamed Abdel Hamid
- Centre of Excellence for Human Genetics; National Research Centre; Cairo Egypt
- Division of Human Genetics and Genome Research; Department of Medical Molecular Genetics; National Research Centre; El Cairo Egypt
| | - Jesús Argente
- Department of Endocrinology; Hospital Universitario Niño Jesús, IIS La Princesa; Madrid Spain
- Department of Pediatrics; Universidad Autónoma de Madrid; Madrid Spain
- CIBEROBN, Centro de Investigación Biomédica en Red sobre Fisiopatología de la obesidad y nutrición; Instituto de Salud Carlos III; Madrid Spain
| | - Víctor L. Ruiz-Pérez
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE); Hospital Universitario La Paz; Madrid Spain
- Instituto de Investigaciones Biológicas (IB); Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid; Madrid Spain
| | - Karen E. Heath
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE); Hospital Universitario La Paz; Madrid Spain
| | - Pablo Lapunzina
- Institute of Medical and Molecular Genetics (INGEMM); Hospital Universitario La Paz, IdiPAZ; Universidad Autónoma de Madrid; Madrid Spain
- CIBERER; Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII; Madrid Spain
- Skeletal Dysplasia Multidisciplinary Unit (UMDE); Hospital Universitario La Paz; Madrid Spain
| |
Collapse
|
39
|
Abstract
Hypophosphatasia (HPP) is an inherited systemic bone disease that is characterized by bone hypomineralization. HPP is classified into six forms according to the age of onset and severity as perinatal (lethal), perinatal benign, infantile, childhood, adult, and odontohypophosphatasia. The causative gene of the disease is the ALPL gene that encodes tissue-nonspecific alkaline phosphatase (TNAP). TNAP is expressed ubiquitously, and its physiological role is apparent in bone mineralization. A defect in bone mineralization can manifest in several ways, including rickets or osteomalacia in HPP patients. Patients with severe forms suffer from respiratory failure because of hypoplastic chest, which is the main cause of death. They sometimes present with seizures due to a defect in vitamin B6 metabolism resulting from the lack of alkaline phosphatase activity in neuronal cells, which is also lethal. Patients with a mild form of the disease exhibit rickets or osteomalacia and a functional defect of exercise. Odontohypophosphatasia shows only dental manifestations. To date, 302 mutations in the ALPL gene have been reported, mainly single-nucleotide substitutions, and the relationships between phenotype and genotype have been partially elucidated. An established treatment for HPP was not available until the recent development of enzyme replacement therapy. The first successful enzyme replacement therapy in model mice using a modified human TNAP protein (asfotase alfa) was reported in 2008, and subsequently success in patients with severe form of the disease was reported in 2012. In 2015, asfotase alfa was approved in Japan in July, followed by in the EU and Canada in August, and then by the US Food and Drug Administration in the USA in October. It is expected that therapy with asfotase alfa will drastically change treatments and prognosis of HPP.
Collapse
Affiliation(s)
- Hideo Orimo
- Division of Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
40
|
Abstract
Hypophosphatasia (HPP) is due to deficient activity of the tissue-nonspecific isoenzyme of alkaline phosphatase (TNAP). This enzyme cleaves extracellular substrates inorganic pyrophosphates (PPi), pyridoxal-5'-phosphate (PLP), phosphoethanolamine (PEA) and nucleotides, and probably other substrates not yet identified. During the last 15 years the role of TNAP in mineralization, and to a less degree in brain, has been investigated, providing hypotheses and explanations for both bone and neuronal HPP phenotypes. ALPL, the gene encoding TNAP, is subject to many mutations, mostly missense mutations. A few number of mutations are recurrently found and may be quite frequent in particular populations. This reflects founder effects. The great variety of mutations results in a great number of compound heterozygous genotypes and in highly variable clinical expressivity. A good correlation was observed between the severity of the disease and in vitro enzymatic activity of the mutant protein measured after site-directed mutagenesis. Many missense mutations found in severe hypophosphatasia produced a mutant protein that failed to reach the cell membrane , was accumulated in the cis-Golgi and was subsequently degraded in the proteasome. Missense mutations located in the catalytic site or in the homodimer interface were often shown by site-directed mutagenesis to have a dominant negative effect. Currently molecular diagnosis of HPP is based on the sequencing of the coding sequence of ALPL that allows detection of approximately 95 % of mutations in severe cases. In addition, other genes, especially genes encoding proteins involved in the regulation of extracellular PPi concentration, could modify the phenotype (modifier genes).
Collapse
|
41
|
Whyte MP, Rockman-Greenberg C, Ozono K, Riese R, Moseley S, Melian A, Thompson DD, Bishop N, Hofmann C. Asfotase Alfa Treatment Improves Survival for Perinatal and Infantile Hypophosphatasia. J Clin Endocrinol Metab 2016; 101:334-42. [PMID: 26529632 PMCID: PMC4701846 DOI: 10.1210/jc.2015-3462] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Hypophosphatasia (HPP) is an inborn error of metabolism that, in its most severe perinatal and infantile forms, results in 50-100% mortality, typically from respiratory complications. OBJECTIVES Our objective was to better understand the effect of treatment with asfotase alfa, a first-in-class enzyme replacement therapy, on mortality in neonates and infants with severe HPP. DESIGN/SETTING Data from patients with the perinatal and infantile forms of HPP in two ongoing, multicenter, multinational, open-label, phase 2 interventional studies of asfotase alfa treatment were compared with data from similar patients from a retrospective natural history study. PATIENTS Thirty-seven treated patients (median treatment duration, 2.7 years) and 48 historical controls of similar chronological age and HPP characteristics. INTERVENTIONS Treated patients received asfotase alfa as sc injections either 1 mg/kg six times per week or 2 mg/kg thrice weekly. MAIN OUTCOME MEASURES Survival, skeletal health quantified radiographically on treatment, and ventilatory status were the main outcome measures for this study. RESULTS Asfotase alfa was associated with improved survival in treated patients vs historical controls: 95% vs 42% at age 1 year and 84% vs 27% at age 5 years, respectively (P < .0001, Kaplan-Meier log-rank test). Whereas 5% (1/20) of the historical controls who required ventilatory assistance survived, 76% (16/21) of the ventilated and treated patients survived, among whom 75% (12/16) were weaned from ventilatory support. This better respiratory outcome accompanied radiographic improvements in skeletal mineralization and health. CONCLUSIONS Asfotase alfa mineralizes the HPP skeleton, including the ribs, and improves respiratory function and survival in life-threatening perinatal and infantile HPP.
Collapse
Affiliation(s)
- Michael P Whyte
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Cheryl Rockman-Greenberg
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Keiichi Ozono
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Richard Riese
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Scott Moseley
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Agustin Melian
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - David D Thompson
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Nicholas Bishop
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| | - Christine Hofmann
- Shriners Hospital for Children (M.P.W.) and Division of Bone and Mineral Diseases at Washington University School of Medicine (M.P.W.), St Louis, Missouri 63110; University of Manitoba and Children's Hospital Research Institute of Manitoba (C.R.-G.), Winnipeg, MB R3T 2N2 Canada; Graduate School of Medicine (K.O.), Osaka University, Osaka, 565-0871 Japan; Alexion Pharmaceuticals, Inc. (R.R., S.M., A.M., D.D.T.), Cheshire, Connecticut 06410; Department of Human Metabolism (N.B.), University of Sheffield, Sheffield, S10 2TN United Kingdom; Sheffield Children's Hospital (N.B.), Sheffield, S10 2TH United Kingdom; University Children's Hospital (C.H.), University of Würzburg, Würzburg, D-97080 Germany
| |
Collapse
|
42
|
Taillandier A, Domingues C, De Cazanove C, Porquet-Bordes V, Monnot S, Kiffer-Moreira T, Rothenbuhler A, Guggenbuhl P, Cormier C, Baujat G, Debiais F, Capri Y, Cohen-Solal M, Parent P, Chiesa J, Dieux A, Petit F, Roume J, Isnard M, Cormier-Daire V, Linglart A, Millán JL, Salles JP, Muti C, Simon-Bouy B, Mornet E. Molecular diagnosis of hypophosphatasia and differential diagnosis by targeted Next Generation Sequencing. Mol Genet Metab 2015; 116:215-20. [PMID: 26432670 PMCID: PMC5257278 DOI: 10.1016/j.ymgme.2015.09.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 09/26/2015] [Accepted: 09/26/2015] [Indexed: 11/24/2022]
Abstract
Hypophosphatasia (HPP) is a rare inherited skeletal dysplasia due to loss of function mutations in the ALPL gene. The disease is subject to an extremely high clinical heterogeneity ranging from a perinatal lethal form to odontohypophosphatasia affecting only teeth. Up to now genetic diagnosis of HPP is performed by sequencing the ALPL gene by Sanger methodology. Osteogenesis imperfecta (OI) and campomelic dysplasia (CD) are the main differential diagnoses of severe HPP, so that in case of negative result for ALPL mutations, OI and CD genes had often to be analyzed, lengthening the time before diagnosis. We report here our 18-month experience in testing 46 patients for HPP and differential diagnosis by targeted NGS and show that this strategy is efficient and useful. We used an array including ALPL gene, genes of differential diagnosis COL1A1 and COL1A2 that represent 90% of OI cases, SOX9, responsible for CD, and 8 potentially modifier genes of HPP. Seventeen patients were found to carry a mutation in one of these genes. Among them, only 10 out of 15 cases referred for HPP carried a mutation in ALPL and 5 carried a mutation in COL1A1 or COL1A2. Interestingly, three of these patients were adults with fractures and/or low BMD. Our results indicate that HPP and OI may be easily misdiagnosed in the prenatal stage but also in adults with mild symptoms for these diseases.
Collapse
Affiliation(s)
- Agnès Taillandier
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| | - Christelle Domingues
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| | - Clémence De Cazanove
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| | - Valérie Porquet-Bordes
- Endocrinologie, Maladies Osseuses, Génétique et Gynécologie Médicale, Hôpital des Enfants, CHU de Toulouse, Toulouse Cedex 9, France.
| | - Sophie Monnot
- Université Paris-Descartes, Sorbonne Paris Cité, Institut Imagine and INSERM U1163, Hôpital Necker-Enfants Malades, Paris, France.
| | - Tina Kiffer-Moreira
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Agnès Rothenbuhler
- APHP, Bicêtre Paris Sud, Department of Pediatric Endocrinology and Diabetology for Children, Le Kremlin Bicêtre 94270, France; APHP, Reference Center for Rare Disorders of the Mineral Metabolism and Plateforme D'expertise Paris Sud, Le Kremlin Bicêtre 94270, France.
| | - Pascal Guggenbuhl
- Service de Rhumatologie, Hôpital Sud, CHU de Rennes, 16, Boulevard de Bulgarie, BP90347, 35203 Rennes Cedex 2, France.
| | - Catherine Cormier
- Rheumatology Department, Cochin University Hospital, 75015 Paris, France.
| | - Geneviève Baujat
- Centres de Référence Maladies Osseuses Constitutionnelles (MOC), Hôpital Universitaire Necker-Enfants Malades et Institut Imagine (AP-HP), 75015 Paris, France.
| | - Françoise Debiais
- Service de rhumatologie, CHU de Poitiers, 86021 Poitiers Cedex, France.
| | - Yline Capri
- Department of Genetics, APHP-Robert Debré University Hospital, Paris, France.
| | - Martine Cohen-Solal
- Department of Rheumatology, INSERM UMR-1132, Lariboisière Hospital and University, Paris Diderot Sorbonne, Paris, France.
| | - Philippe Parent
- Service de Génétique Clinique, CHU Brest, Brest F-29200, France.
| | - Jean Chiesa
- Department of Genetics, University Hospital, Nîmes, France.
| | - Anne Dieux
- Service de Génétique Clinique, CHU, Lille, France.
| | | | - Joelle Roume
- Unité de Génétique Médicale, Centre Intercommunal Poissy-St-Germain en Laye, Poissy, France.
| | - Monica Isnard
- Gynécologie Obstétrique, Centre Hospitalier de Mulhouse, 68051 Mulhouse Cedex, France
| | - Valérie Cormier-Daire
- Université Paris-Descartes, Sorbonne Paris Cité, Institut Imagine and INSERM U1163, Hôpital Necker-Enfants Malades, Paris, France.
| | - Agnès Linglart
- APHP, Bicêtre Paris Sud, Department of Pediatric Endocrinology and Diabetology for Children, Le Kremlin Bicêtre 94270, France; APHP, Reference Center for Rare Disorders of the Mineral Metabolism and Plateforme D'expertise Paris Sud, Le Kremlin Bicêtre 94270, France.
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Jean-Pierre Salles
- Endocrinologie, Maladies Osseuses, Génétique et Gynécologie Médicale, Hôpital des Enfants, CHU de Toulouse, Toulouse Cedex 9, France.
| | - Christine Muti
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| | - Brigitte Simon-Bouy
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| | - Etienne Mornet
- Unité de Génétique Constitutionnelle, Centre Hospitalier de Versailles, 78150 Le Chesnay, France.
| |
Collapse
|
43
|
Taketani T, Oyama C, Mihara A, Tanabe Y, Abe M, Hirade T, Yamamoto S, Bo R, Kanai R, Tadenuma T, Michibata Y, Yamamoto S, Hattori M, Katsube Y, Ohnishi H, Sasao M, Oda Y, Hattori K, Yuba S, Ohgushi H, Yamaguchi S. Ex Vivo Expanded Allogeneic Mesenchymal Stem Cells with Bone Marrow Transplantation Improved Osteogenesis in Infants with Severe Hypophosphatasia. Cell Transplant 2015; 24:1931-43. [DOI: 10.3727/096368914x685410] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Patients with severe hypophosphatasia (HPP) develop osteogenic impairment with extremely low alkaline phosphatase (ALP) activity, resulting in a fatal course during infancy. Mesenchymal stem cells (MSCs) differentiate into various mesenchymal lineages, including bone and cartilage. The efficacy of allogeneic hematopoietic stem cell transplantation for congenital skeletal and storage disorders is limited, and therefore we focused on MSCs for the treatment of HPP. To determine the effect of MSCs on osteogenesis, we performed multiple infusions of ex vivo expanded allogeneic MSCs for two patients with severe HPP who had undergone bone marrow transplantation (BMT) from asymptomatic relatives harboring the heterozygous mutation. There were improvements in not only bone mineralization but also muscle mass, respiratory function, and mental development, resulting in the patients being alive at the age of 3. After the infusion of MSCs, chimerism analysis of the mesenchymal cell fraction isolated from bone marrow in the patients demonstrated that donor-derived DNA sequences existed. Adverse events of BMT were tolerated, whereas those of MSC infusion did not occur. However, restoration of ALP activity was limited, and normal bony architecture could not be achieved. Our data suggest that multiple MSC infusions, following BMT, were effective and brought about clinical benefits for patients with lethal HPP. Allogeneic MSC-based therapy would be useful for patients with other congenital bone diseases and tissue disorders if the curative strategy to restore clinically normal features, including bony architecture, can be established.
Collapse
Affiliation(s)
- Takeshi Taketani
- Division of Blood Transfusion, Shimane University Hospital, Izumo, Shimane, Japan
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Chigusa Oyama
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Aya Mihara
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Yuka Tanabe
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Mariko Abe
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Tomohiro Hirade
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Satoshi Yamamoto
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Ryosuke Bo
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Rie Kanai
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Taku Tadenuma
- Division of Rehabilitation, Shimane University Hospital, Izumo, Shimane, Japan
| | - Yuko Michibata
- Division of Rehabilitation, Shimane University Hospital, Izumo, Shimane, Japan
| | - Soichiro Yamamoto
- Department of Orthopedics, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Miho Hattori
- Division of Blood Transfusion, Shimane University Hospital, Izumo, Shimane, Japan
| | - Yoshihiro Katsube
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Hiroe Ohnishi
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Mari Sasao
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Yasuaki Oda
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Koji Hattori
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Shunsuke Yuba
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Hajime Ohgushi
- Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology, Amagasaki, Hyogo, Japan
| | - Seiji Yamaguchi
- Department of Pediatrics, Shimane University School of Medicine, Izumo, Shimane, Japan
| |
Collapse
|
44
|
Shum AMY, Fung DCY, Corley SM, McGill MC, Bentley NL, Tan TC, Wilkins MR, Polly P. Cardiac and skeletal muscles show molecularly distinct responses to cancer cachexia. Physiol Genomics 2015; 47:588-99. [PMID: 26395599 DOI: 10.1152/physiolgenomics.00128.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 09/18/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer cachexia is a systemic, paraneoplastic syndrome seen in patients with advanced cancer. There is growing interest in the altered muscle pathophysiology experienced by cachectic patients. This study reports the microarray analysis of gene expression in cardiac and skeletal muscle in the colon 26 (C26) carcinoma mouse model of cancer cachexia. A total of 268 genes were found to be differentially expressed in cardiac muscle tissue, compared with nontumor-bearing controls. This was fewer than the 1,533 genes that changed in cachectic skeletal muscle. In addition to different numbers of genes changing, different cellular functions were seen to change in each tissue. The cachectic heart showed signs of inflammation, similar to cachectic skeletal muscle, but did not show the upregulation of ubiquitin-dependent protein catabolic processes or downregulation of genes involved in cellular energetics and muscle regeneration that characterizes skeletal muscle cachexia. Quantitative PCR was used to investigate a subset of inflammatory genes in the cardiac and skeletal muscle of independent cachectic samples; this revealed that B4galt1, C1s, Serpina3n, and Vsig4 were significantly upregulated in cardiac tissue, whereas C1s and Serpina3n were significantly upregulated in skeletal tissue. Our skeletal muscle microarray results were also compared with those from three published microarray studies and found to be consistent in terms of the genes differentially expressed and the functional processes affected. Our study highlights that skeletal and cardiac muscles are affected differently in the C26 mouse model of cachexia and that therapeutic strategies cannot assume that both muscle types will show a similar response.
Collapse
Affiliation(s)
- Angie M Y Shum
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - David C Y Fung
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Susan M Corley
- New South Wales Systems Biology Initiative, University of New South Wales Australia, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Max C McGill
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - Nicholas L Bentley
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Mitochondrial Bioenergetics Group, Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia
| | - Timothy C Tan
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Blacktown Clinical School and Blacktown Hospital, Blacktown, New South Wales, Australia; and Cardiac Ultrasound Laboratory, Massachusetts General Hospital, Boston, Massachusetts
| | - Marc R Wilkins
- New South Wales Systems Biology Initiative, University of New South Wales Australia, New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Australia, New South Wales, Australia
| | - Patsie Polly
- Inflammation and Infection Research Centre, University of New South Wales Australia, New South Wales, Australia; Department of Pathology, School of Medical Sciences, Faculty of Medicine, University of New South Wales Australia, New South Wales, Australia;
| |
Collapse
|
45
|
Abstract
Hypophosphatasia (HPP) is a bone metabolic disorder caused by mutations in the liver/bone/kidney alkaline phosphatase gene (ALPL), which encodes tissue-nonspecific alkaline phosphatase (TNAP). This disease is characterized by disrupted bone and tooth mineralization, and reduced serum AP activity. Along with bone and tooth symptoms, many neurological symptoms, seizure, encephalopathy, intracranial hypertension, mental retardation, deafness, and growth hormone deficiency (GHD), are frequently found in HPP patients. Seizure occurs in severe HPP types soon after birth, and responds to pyridoxine, but is an indicator of lethal prognosis. Encephalopathy rarely presents in severe HPP types, but has severe sequelae. Intracranial hypertension complicated in mild HPP types develops after the age of 1 year and sometimes need neurosurgical intervention. Mental retardation, deafness and GHD are more frequently found in Japanese HPP patients. Mental retardation occurs in all HPP types. Deafness in perinatal lethal type is both conductive and sensorineural. GHD develops in all but perinatal lethal type and the diagnosis tends to delay. The pathogenesis of these neural features of HPP might be due to impairment of both vitamin B6 metabolism and central nervous system development by ALPL mutations.
Collapse
|
46
|
Matsushita M, Kitoh H, Michigami T, Tachikawa K, Ishiguro N. Benign prenatal hypophosphatasia: a treatable disease not to be missed. Pediatr Radiol 2014; 44:340-3. [PMID: 24145968 DOI: 10.1007/s00247-013-2805-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/30/2013] [Accepted: 09/15/2013] [Indexed: 10/26/2022]
Abstract
Prenatal bowing of the long bones is often associated with severe bone dysplasias. We report a child who presented marked bowing of the long bones at birth but showed a relatively benign postnatal course with spontaneous improvement of bowing. The fetal imaging showed normal skeletal mineralization and normal chest and abdominal circumferences despite the limb bowing and shortening. Decreased serum alkaline phosphatase activity and elevated urine phosphoethanolamine was biochemically evident, and compound heterozygous mutations in the tissue-nonspecific alkaline phosphatase (TNSALP) gene were identified, which confirmed the diagnosis of a benign form of prenatal hypophosphatasia. Benign prenatal hypophosphatasia should be considered in the differential diagnosis of congenital bowing of the long bones.
Collapse
Affiliation(s)
- Masaki Matsushita
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | | | | | | | | |
Collapse
|
47
|
Watanabe A, Satoh S, Fujita A, Naing BT, Orimo H, Shimada T. Perinatal hypophosphatasia caused by uniparental isodisomy. Bone 2014; 60:93-7. [PMID: 24334170 DOI: 10.1016/j.bone.2013.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/04/2013] [Accepted: 12/06/2013] [Indexed: 01/30/2023]
Abstract
Hypophosphatasia (HPP) is an inherited disorder characterized by defective bone mineralization caused by mutations in the alkaline phosphatase gene (ALPL). Clinically, the disease spans a great continuum of disease severity and six forms can be distinguished according to the age of onset. The most severe is the autosomal recessive perinatal form, a major prenatal skeletal dysplasia in Japan. The ALPL mutation c.1559delT causes perinatal HPP and occurs frequently in the Japanese. Most patients with perinatal HPP in Japan are homozygous for c.1559delT, and their parents are usually heterozygous with no evidence of consanguinity. Here we identified a fetus with perinatal HPP resulting from an unusual mechanism known as paternal uniparental isodisomy (UPD) of chromosome 1. Sequence analysis of ALPL in the patient revealed the presence of the homozygous mutation c.1559delT. We suspected UPD because the father and mother were heterozygous and wild type, respectively. Analysis of polymorphic microsatellite markers spanning chromosome 1 and whole-genome arrays revealed a uniparental inheritance from the father and excluded deletions or de novo mutations. This is the first description of perinatal HPP caused by UPD. This report also emphasizes the low recurrence risk of a non-Mendelian inheritance pattern in UPD and the value of determining parental genotypes with homozygous mutations in a patient to confirm whether the condition is caused by UPD or not, even when the mutation is detected as a hot spot, as described in the literature.
Collapse
Affiliation(s)
- Atsushi Watanabe
- Division of Clinical Genetics, Nippon Medical School Hospital, Tokyo, Japan; Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.
| | - Shuhei Satoh
- Aomori Prefectural Central Hospital, Aomori, Japan
| | - Atsushi Fujita
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Banyar Than Naing
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Hideo Orimo
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Takashi Shimada
- Division of Clinical Genetics, Nippon Medical School Hospital, Tokyo, Japan; Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
48
|
Taketani T, Onigata K, Kobayashi H, Mushimoto Y, Fukuda S, Yamaguchi S. Clinical and genetic aspects of hypophosphatasia in Japanese patients. Arch Dis Child 2014; 99:211-5. [PMID: 24276437 DOI: 10.1136/archdischild-2013-305037] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE We examined the clinical and genetic features of hypophosphatasia (HPP) in Japanese patients. HPP is a rare metabolic bone disorder of bone mineralisation caused by mutations in the liver/bone/kidney alkaline phosphatase (ALPL) gene, which encodes tissue-non-specific alkaline phosphatase isoenzyme. METHODS We retrospectively investigate the incidence and clinical features of 52 patients with paediatric HPP who were born between 1999 and 2010. Mutations of the ALPL gene were analysed in 31 patients. RESULTS The annual incidence of perinatal lethal HPP (PLH) was estimated to be 2-3/1 000 000 births. The most frequent clinical type was PLH followed by prenatal benign. In addition to bone symptoms, cerebral manifestations were frequently observed including convulsion, mental retardation, deafness and short stature with growth hormone deficiency. Respiratory failure was the most significant predictor of a poor prognosis for PLH. The first and second most frequent mutations in the ALPL gene were c.1559delT and c.T979C (p.F327L), respectively. The c.1559delT homozygous mutation was lethal with respiratory failure. Patients with the p.F327L compound heterozygous mutation had the different non-lethal type with short stature and a gradual improvement in ALP level and bone mineralisation. CONCLUSIONS The most frequent clinical type was the PLH type with prognosis related to respiratory failure, biochemical/radiological changes and ALPL mutations. Cerebral manifestations frequently occurred. Genotype-phenotype correlations were associated with specific outcomes in the PLH type, whereas different clinical features were associated with the same genotype in the non-lethal type.
Collapse
Affiliation(s)
- Takeshi Taketani
- Division of Blood Transfusion, Shimane University Hospital, , Shimane, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Matsuo K, Mukai T, Furuya A, Suzuki S, Tanahashi Y, Azuma H. A Case of Vitamin D Deficiency without Elevation of Serum Alkaline Phosphatase in a Carrier of Hypophosphatasia. Clin Pediatr Endocrinol 2013. [PMID: 24170964 PMCID: PMC3809733 DOI: 10.1297/cpe.22.73] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Elevated serum alkaline phosphatase (ALP) is a screening marker for the diagnosis of vitamin D deficiency, which may fail to be diagnosed if serum ALP is not elevated. Here, we describe a case of vitamin D deficiency without elevation of serum ALP. A 1-year-old Japanese girl was referred to our hospital for the evaluation of genu varum. Her serum intact PTH level was elevated, while her serum ALP level was normal. Furthermore, her serum 25-hydroxyvitamin D level was reduced, and her urine phosphoethanolamine (PEA) level was mildly elevated. ALPL gene analysis revealed she was a heterozygous carrier of hypophosphatasia (c.1559delT). Serum intact PTH and urine PEA evaluations were helpful for diagnosing vitamin D deficiency and hypophosphatasia carrier status, respectively. Therefore, the possibility of vitamin D deficiency without elevation of serum ALP should be considered.
Collapse
Affiliation(s)
- Kumihiro Matsuo
- Department of Pediatrics, Asahikawa Medical College, Asahikawa, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Haraikawa M, Tanabe R, Sogabe N, Sugimoto A, Kawamura Y, Michigami T, Hosoi T, Goseki-Sone M. A study of the association between serum bone-specific alkaline phosphatase and serum phosphorus concentration or dietary phosphorus intake. J Nutr Sci Vitaminol (Tokyo) 2013; 58:442-5. [PMID: 23419404 DOI: 10.3177/jnsv.58.442] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Alkaline phosphatase (ALP) hydrolyzes a variety of monophosphate esters into phosphoric acid and alcohol at a high optimum pH (pH 8-10). Human ALPs are classified into four types: tissue-non specific (TNSALP, liver/bone/kidney), intestinal, placental, and germ cell types. Based on studies of hypophosphatasia (HPP), which is a systemic bone disease caused by the presence of either one or two pathologic mutations in ALPL that encodes TNSALP, TNSALP was suggested to be indispensable for skeletal mineralization. In this study, we explored the possibility that dietary nutrients contribute to regulate serum bone-specific ALP (BAP) activity. Serum biochemical parameters, such as serum ALP, BAP, osteocalcin, and fibroblast growth factor 23 (FGF23), were measured in healthy young subjects (n=193). Dietary nutrient intakes were measured based on 3-d food records before the day of blood examinations. The presence of a carrier of the deletion of T at nucleotide 1559 (c.1559delT), which has been reported to be the most frequent in Japanese HPP, was not detected in any subject. By the analysis of BAP activity and other biochemical parameters or dietary nutrient intakes, we obtained significant correlations between BAP activity and serum phosphorus (r=-0.165, p=0.022), calcium intake (mg/1,000 kcal/d) (r=-0.186, p=0.010), or phosphorus intake (mg/1,000 kcal/d) (r=-0.226, p=0.002). Further study on the regulation of BAP activity and calcium and/or phosphorus homeostasis will provide useful data for improving skeletal health.
Collapse
Affiliation(s)
- Mayu Haraikawa
- Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|