Hypophosphatasia is a rare heritable disease caused by mutations that affect the activity of the tissue non-specific alkaline phosphatase (TNAP) isozyme. The clinical severity varies widely from complete absence of bone mineralization and stillbirth in the perinatal/infantile form to spontaneous fractures and loss of decidual teeth in adult and odontohypophosphatasia. While hypophosphatasia is usually inherited as an autosomal recessive trait, variable expressivity, incomplete penetrance and autosomal dominant inheritance have been reported. Several missense mutations in the TNAP gene have been reported in hypophosphatasia and compound heterozygosity appears frequently. No treatment is currently available for hypophosphatasia. Recently, we have produced and characterized a mouse model of infantile hypophosphatasia generated by introducing a null allele in the TNAP locus. In this grant application we propose to test the hypotheses that A) the apparent complex genetics of hypophosphatasia, results from mutations affecting different functional domains in the TNAP molecule and B) that autologous transplantation of ex vivo manipulated bone marrow osteoprogenitor stem cells may be a viable treatment modality for hypophosphatasia. Consequently, the specific aims are: 1) To study and compare the structural and functional disturbances in the TNAP molecule caused by those missense mutations that have been found associated with the perinatal/infantile, adult and odontohypophosphatasia. 2) To generate animal models of adult and odontohypophosphatasia in order to understand the pathogenesis and mode of inheritance in comparison to our perinatal/infantile hypophosphatasia model. 3) To introduce AP expression constructs into bone marrow osteoprogenitor stem cells derived from our hypophosphatasia mice and analyze the ability of these ex vivo manipulated stem cells to correct the hypophosphatasia phenotype. Our work will provide information about the molecular mechanism(s) of pathogenesis of hypophosphatasia and the molecular basis for the different genetic modes of, transmission. Hypophosphatasia will be used as a paradigm for genetic bone diseases as we test the feasibility of using genetically manipulated mesenchymal stem cells in the treatment of this disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Dental & Craniofacial Research (NIDCR)
Type
Research Project (R01)
Project #
5R01DE012889-02
Application #
6164425
Study Section
Orthopedics and Musculoskeletal Study Section (ORTH)
Program Officer
Sharrock, William J
Project Start
1999-03-01
Project End
2004-02-29
Budget Start
2000-03-01
Budget End
2001-02-28
Support Year
2
Fiscal Year
2000
Total Cost
$352,365
Indirect Cost
Name
Sanford-Burnham Medical Research Institute
Department
Type
DUNS #
009214214
City
La Jolla
State
CA
Country
United States
Zip Code
92037
Pettengill, Matthew; Matute, Juan D; Tresenriter, Megan et al. (2018) Correction: Human alkaline phosphatase dephosphorylates microbial products and is elevated in preterm neonates with a history of late-onset sepsis. PLoS One 13:e0197532
Li, Qiaoli; Huang, Jianhe; Pinkerton, Anthony B et al. (2018) Inhibition of Tissue-Nonspecific Alkaline Phosphatase Attenuates Ectopic Mineralization in the Abcc6-/- Mouse Model of PXE but Not in the Enpp1 Mutant Mouse Models of GACI. J Invest Dermatol :
Yang, Won Ho; Heithoff, Douglas M; Aziz, Peter V et al. (2018) Accelerated Aging and Clearance of Host Anti-inflammatory Enzymes by Discrete Pathogens Fuels Sepsis. Cell Host Microbe 24:500-513.e5
Patel, Jessal J; Zhu, Dongxing; Opdebeeck, Britt et al. (2018) Inhibition of arterial medial calcification and bone mineralization by extracellular nucleotides: The same functional effect mediated by different cellular mechanisms. J Cell Physiol 233:3230-3243
Simão, Ana Maria Sper; Bolean, Maytê; Favarin, Bruno Zoccaratto et al. (2018) Lipid microenvironment affects the ability of proteoliposomes harboring TNAP to induce mineralization without nucleators. J Bone Miner Metab :
Foster, B L; Ao, M; Salmon, C R et al. (2018) Osteopontin regulates dentin and alveolar bone development and mineralization. Bone 107:196-207
Brun, Lucas R; Lombarte, M; Roma, S et al. (2018) Increased calcium uptake and improved trabecular bone properties in intestinal alkaline phosphatase knockout mice. J Bone Miner Metab 36:661-667
Bottini, Massimo; Mebarek, Saida; Anderson, Karen L et al. (2018) Matrix vesicles from chondrocytes and osteoblasts: Their biogenesis, properties, functions and biomimetic models. Biochim Biophys Acta Gen Subj 1862:532-546
Morcos, M W; Al-Jallad, H; Li, J et al. (2018) PHOSPHO1 is essential for normal bone fracture healing: An Animal Study. Bone Joint Res 7:397-405
Huang, Nai-Jia; Lin, Ying-Cing; Lin, Chung-Yueh et al. (2018) Enhanced phosphocholine metabolism is essential for terminal erythropoiesis. Blood 131:2955-2966

Showing the most recent 10 out of 109 publications