Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Bone ; 110: 96-106, 2018 05.
Article in English | MEDLINE | ID: mdl-29360619

ABSTRACT

Hypophosphatasia (HPP) is the inborn-error-of-metabolism due to loss-of-function mutation(s) of the ALPL (TNSALP) gene that encodes the tissue non-specific isoenzyme of alkaline phosphatase (TNSALP). TNSALP represents a family of cell-surface phosphohydrolases differing by post-translational modification that is expressed especially in the skeleton, liver, kidney, and developing teeth. Thus, the natural substrates of TNSALP accumulate extracellularly in HPP including inorganic pyrophosphate (PPi), a potent inhibitor of mineralization, and pyridoxal 5'-phosphate (PLP), the principal circulating form of vitamin B6. The superabundance of extracellular PPi regularly causes tooth loss, and when sufficiently great can lead to rickets or osteomalacia. Sometimes diminished hydrolysis of PLP engenders vitamin B6-dependent seizures in profoundly affected babies. Autosomal dominant and autosomal recessive inheritance from among >340 ALPL mutations identified to date, typically missense and located throughout the gene, largely explains the remarkably wide-ranging severity of HPP, greatest of all skeletal diseases. In 2015, our demographic, clinical, and DXA findings acquired over 25 years from 173 children and adolescents with HPP validated and expanded the clinical nosology for pediatric patients to include according to increasing severity "odonto" HPP, "mild childhood" HPP, "severe childhood" HPP, "infantile" HPP, and "perinatal" HPP. Herein, we assessed this expanded nosology using biochemical hallmarks of HPP. We evaluated exclusively data from the 165 preteenage HPP patients in this cohort to exclude potential effects from physiological changes in TNSALP levels across puberty. All patients had subnormal serum total and bone-specific ALP and elevated plasma PLP, and nearly all had excessive urinary PPi excretion. Only the PLP levels were unchanged across puberty. Mean levels of all four biomarkers correlated with HPP severity ranked according to the HPP nosology, but the data overlapped among all four patient groups. Hence, these four biochemical hallmarks represent both a sensitive and reliable tool for diagnosing children with HPP. Furthermore, the hallmarks validate our expanded clinical nosology for pediatric HPP that, with limitations, is an improved framework for conceptualizing and working with this disorder's remarkably broad-ranging severity.


Subject(s)
Alkaline Phosphatase/metabolism , Hypophosphatasia/diagnosis , Adolescent , Biomarkers/metabolism , Bone and Bones/metabolism , Child , Child, Preschool , Cohort Studies , Female , Humans , Male , Mutation , Phosphates/chemistry , Puberty , Sexual Maturation , Young Adult
2.
Semin Arthritis Rheum ; 48(1): 9-11, 2018 08.
Article in English | MEDLINE | ID: mdl-29338885

ABSTRACT

OBJECTIVE: Calcium pyrophosphate deposition disease (CPDD) is arthritis caused by calcium pyrophosphate (CPP) crystal deposition in joints. It is commonly associated with aging as well as a handful of metabolic syndromes. Recent epidemiologic studies suggest a positive association of CPDD and rheumatoid arthritis (RA). Yet how these diseases are related remains unclear. We set out to describe 21 well-characterized patients with both diagnoses. METHODS: Medical records of patients with both RA and CPDD identified at a single academic practice site were reviewed for age, gender, age of CPDD and RA onset, disease duration, joint involvement, and lab values including rheumatoid factor (RF), cyclic citrullinated peptide antibody (CCP), iron studies, and parathyroid hormone and calcium levels. RESULTS: The mean age of CPDD onset was 69.5 ± 11.4 years, with a mean RA age onset of 53.9 ± 16 years, demonstrating a mean lag of 13.4 ± 10.9 years between diagnoses. The majority of RA patients were diagnosed with CPDD based on the presence of radiographic chondrocalcinosis (15/21). The most commonly involved joint was the knee, followed by the wrist, hip, and shoulder. CONCLUSIONS: These data show that the diagnosis of RA often precedes the diagnosis of CPDD. This asynchronous presentation taken together with the classic age of onset for CPDD and typical pattern of joint involvement supports the hypothesis that CPDD develops in RA patients through similar processes as those that cause the idiopathic forms of this disease.


Subject(s)
Arthritis, Rheumatoid/complications , Chondrocalcinosis/complications , Hand Joints/diagnostic imaging , Knee Joint/diagnostic imaging , Aged , Aged, 80 and over , Arthritis, Rheumatoid/diagnostic imaging , Chondrocalcinosis/diagnostic imaging , Female , Humans , Male , Middle Aged
4.
Rheum Dis Clin North Am ; 40(2): 343-56, 2014 May.
Article in English | MEDLINE | ID: mdl-24703351

ABSTRACT

Calcium crystal arthritis is often unrecognized, poorly managed, and few effective therapies are available. The most common types of calcium crystals causing musculoskeletal syndromes are calcium pyrophosphate (CPP) and basic calcium phosphate (BCP). Associated syndromes have different clinical presentations and divergent management strategies. Acute CPP arthritis is treated similarly to acute gouty arthritis, whereas chronic CPP and BCP arthropathy may respond to strategies used for osteoarthritis. Calcific tendonitis is treated with a variety of interventions designed to dissolve BCP crystals. A better understanding of the causes and larger well-planned trials of current therapies will lead to improved care.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antirheumatic Agents/therapeutic use , Chondrocalcinosis/therapy , Colchicine/therapeutic use , Gout Suppressants/therapeutic use , High-Energy Shock Waves/therapeutic use , Periarthritis/therapy , Tendinopathy/therapy , Anti-Inflammatory Agents/therapeutic use , Calcinosis/therapy , Calcium Phosphates , Durapatite , Humans , Interleukin 1 Receptor Antagonist Protein/therapeutic use
5.
J Rheumatol ; 41(1): 65-74, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24293574

ABSTRACT

OBJECTIVE: Accumulation of excess extracellular inorganic pyrophosphate leads to calcium pyrophosphate dihydrate (CPPD) crystal formation in articular cartilage. CPPD crystal formation occurs near morphologically abnormal chondrocytes resembling hypertrophic chondrocytes. The ANK protein was recently implicated as an important factor in the transport of intracellular inorganic pyrophosphate across the cell membrane. We characterized ANK in joint tissues from patients with and without CPPD deposition and correlated the presence of ANK with markers of chondrocyte hypertrophy. METHODS: Articular tissues were obtained from 24 patients with CPPD crystal deposition disease, 11 patients with osteoarthritis (OA) without crystals, and 6 controls. We determined the number of ANK-positive cells in joint tissues using immunohistochemistry and in situ hybridization, and correlated ANK positivity with markers of chondrocyte hypertrophy including Runx2, type X collagen, osteopontin (OPN), and osteocalcin (OCN). RESULTS: ANK was detected in synoviocytes, chondrocytes, osteoblasts, and osteocytes. ANK was seen extracellularly only in the matrix of cartilage and meniscus. The number of ANK-positive cells was significantly higher in CPPD than in OA or normal joint tissues. The amount and intensity of ANK immunoreactivity reached maximum levels in the large chondrocytes around crystal deposits. ANK was similarly distributed to and significantly correlated with Runx2, type X collagen, OPN, and OCN. CONCLUSION: ANK levels were higher in articular tissues from patients with CPPD deposition. ANK was concentrated around crystal deposits and correlated with markers of chondrocyte hypertrophy. These findings support a role for ANK in CPPD crystal formation in cartilage.


Subject(s)
Bone and Bones/metabolism , Cartilage, Articular/metabolism , Chondrocalcinosis/metabolism , Joints/metabolism , Phosphate Transport Proteins/metabolism , Up-Regulation , Adult , Aged , Bone and Bones/pathology , Cartilage, Articular/pathology , Chondrocalcinosis/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Female , Humans , Joints/pathology , Male , Middle Aged , Osteoarthritis/metabolism , Osteoarthritis/pathology
6.
Arthritis Res Ther ; 15(5): R154, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24286344

ABSTRACT

INTRODUCTION: Extracellular ATP (eATP) is released by articular chondrocytes under physiological and pathological conditions. High eATP levels cause pathologic calcification, damage cartilage, and mediate pain. We recently showed that stable over-expression of the progressive ankylosis gene product, ANK, increased chondrocyte eATP levels, but the mechanisms of this effect remained unexplored. The purpose of this work was to further investigate mechanisms of eATP efflux in primary articular chondrocytes and to better define the role of ANK in this process. METHODS: We measured eATP levels using a bioluminescence-based assay in adult porcine articular chondrocyte media with or without a 10 minute exposure to hypotonic stress. siRNAs for known ATP membrane transporters and pharmacologic inhibitors of ATP egress pathways were used to identify participants involved in chondrocyte eATP release. RESULTS: eATP levels increased after exposure to hypotonic media in a calcium-dependent manner in monolayer and 3-dimensional agarose gel cultures (p < 0.001). A potent transient receptor potential vanilloid 4 (TRPV4) agonist mimicked the effects of hypotonic media. ANK siRNA suppressed basal (p < 0.01) and hypotonically-stressed (p < 0.001) ATP levels. This effect was not mediated by altered extracellular pyrophosphate (ePPi) levels, and was mimicked by the ANK inhibitor, probenecid (p < 0.001). The P2X7/4 receptor inhibitor Brilliant Blue G also suppressed eATP efflux induced by hypotonic media (p < 0.001), while ivermectin, a P2X4 receptor stimulant, increased eATP levels (p < 0.001). Pharmacologic inhibitors of hemichannels, maxianion channels and other volume-sensitive eATP efflux pathways did not suppress eATP levels. CONCLUSIONS: These findings implicate ANK and P2X7/4 receptors in chondrocyte eATP efflux. Understanding the mechanisms of eATP efflux may result in novel therapies for calcium crystal arthritis and osteoarthritis.


Subject(s)
Adenosine Triphosphate/metabolism , Chondrocytes/metabolism , Extracellular Space/metabolism , Phosphate Transport Proteins/metabolism , Animals , Ankylosis/genetics , Ankylosis/metabolism , Ankylosis/pathology , Biological Transport/drug effects , Calcium/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Leucine/analogs & derivatives , Leucine/pharmacology , Phosphate Transport Proteins/antagonists & inhibitors , Phosphate Transport Proteins/genetics , Probenecid/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/metabolism , Receptors, Purinergic P2X7/metabolism , Sulfonamides/pharmacology , Swine , TRPV Cation Channels/agonists
8.
Clin Rheumatol ; 32(9): 1383-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23609408

ABSTRACT

Digital imaging combined with picture archiving and communication system (PACS) access allows detailed image retrieval and magnification. Calcium pyrophosphate dihydrate (CPPD) crystals preferentially deposit in fibrocartilages, the cartilage of the acromioclavicular (AC) joint being one such structure. We sought to determine if examination of the AC joints on magnified PACS imaging of chest films would be useful in identifying chondrocalcinosis (CC). Retrospective radiographic readings and chart reviews involving 1,920 patients aged 50 or more who had routine outpatient chest radiographs over a 4-month period were performed. Knee radiographs were available for comparison in 489 patients. Medical records were reviewed to abstract demographics, chest film reports, and diagnoses. AC joint CC was identified in 1.1 % (21/1,920) of consecutive chest films. Patients with AC joint CC were 75 years of age versus 65.4 in those without CC (p < 0.0002). Four hundred eighty-nine patients had knee films. Six of these patients had AC joint CC, and of these, five also had knee CC (83 %). Of the 483 without AC joint CC, 62 (12 %) had knee CC (p = 0.002). Patients with AC joint CC were more likely to have a recorded history of CPPD crystal deposition disease than those without AC joint CC (14 versus 1 %, p = 0.0017). The prevalence of AC joint CC increases with age and is associated with knee CC. A finding of AC joint CC should heighten suspicion of pseudogout or secondary osteoarthritis in appropriate clinical settings and, in a young patient, should alert the clinician to the possibility of an associated metabolic condition.


Subject(s)
Calcium Pyrophosphate/chemistry , Chondrocalcinosis/physiopathology , Clavicle/diagnostic imaging , Joints/physiopathology , Age Factors , Aged , Aged, 80 and over , Aging , Chondrocalcinosis/diagnosis , Chondrocalcinosis/epidemiology , Crystallization , Female , Humans , Knee Joint/diagnostic imaging , Male , Middle Aged , Osteoarthritis/diagnosis , Prevalence , Radiography, Thoracic , Retrospective Studies
9.
J Bone Miner Res ; 26(10): 2389-98, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21713987

ABSTRACT

Hypophosphatasia (HPP) is caused by deactivating mutation(s) within the gene that encodes the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP). Patients manifest rickets or osteomalacia and dental disease ranging from absence of skeletal mineralization in utero to only loss of adult dentition. Until recently, HPP skeletal disease in utero was thought to always predict a lethal outcome. However, several reports beginning in 1999 emphasized a benign prenatal form of HPP (BP-HPP) where skeletal disease detected in utero had a mild postnatal course. Here we describe prenatal and postnatal findings of 17 additional BP-HPP patients among our 178 pediatric HPP patients. Their findings are compared with those of their siblings with HPP, carrier parents, and others with identical TNSALP mutations. New information concerning 7 previously published BP-HPP patients accompanies a review of the HPP literature. Among our 17 BP-HPP patients, prenatal ultrasound showed normal chest or abdominal circumferences where recorded. Sometimes, poor skeletal mineralization, fetal crowding, and third-trimester improvement were observed. Postnatally, extremity bowing further improved (13 patients). BP-HPP severity postnatally spanned the "infantile" to "odonto" HPP phenotypes, resembling our patients who harbored identical TNSALP mutation(s). Eight had autosomal dominant (AD) and 9 had autosomal recessive (AR) BP-HPP. Fourteen of our 15 mothers were HPP carriers or affected. Of the 41 cumulative BP-HPP patients (24 literature cases meriting a BP-HPP diagnosis since 1996 plus our 17 patients), 63% had AR BP-HPP. Maternally transmitted HPP involved 11 of the 13 total AD BP-HPP probands (p = 0.01), supporting a maternal in utero effect on the baby. Fetal crowding, normal fetal mineralization and chest size, and TNSALP heterozygosity seem to identify BP-HPP. However, bowed fetal long bones with AR HPP, specific TNSALP mutations, or poor skeletal mineralization before the third trimester do not reliably diagnose HPP lethality.


Subject(s)
Bone and Bones/embryology , Hypophosphatasia/complications , Alkaline Phosphatase/genetics , Female , Humans , Infant , Mutation , Pregnancy , Pregnancy Complications , Ultrasonography, Prenatal
11.
Connect Tissue Res ; 52(2): 139-46, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20604715

ABSTRACT

OBJECTIVE: Extracellular inorganic pyrophosphate (ePPi) is a key regulator of pathologic mineralization in articular cartilage. Articular chondrocytes generate ePPi by the transportation of intracellular PPi (iPPi) through transport mechanisms such as ANK or by the degradation of extracellular adenosine triphosphate (eATP) by ectoenzymes. Although numerous modulators of ePPi have been characterized, little is known about eATP elaboration in cartilage. We sought to determine (1) whether eATP is coordinately regulated with ePPi and (2) whether ANK transports ATP. METHODS: Primary articular chondrocytes were treated with factors known to modulate ePPi levels including growth factors (TGFß1 and IGF-1), anion channel inhibitors, and chemicals that alter adenylyl cyclase and protein kinase C activities. Additional chondrocyte monolayers were infected with adenovirus containing functional (Ad-ANK) or mutated (Ad-ANK mutant) ANK sequences. eATP levels were measured with a bioluminescent assay. RESULTS: TGFß1 enhanced eATP accumulation by 33%, whereas IGF-1 decreased eATP accumulation by 63% and attenuated TGFß1-induced eATP release by 72%. Forskolin and probenecid diminished eATP accumulation by 55% and 89%. Phorbol-12-myristate-13-acetate increased eATP by 29%. Transfection of chondrocytes with Ad-ANK caused a 10-fold increase in eATP compared with control values. CONCLUSION: Modulation of eATP by various factors paralleled their effects on ePPi production, suggesting a shared pathway of ePPi and eATP production and implicating ANK in eATP transport. As eATP directly contributes to pathologic mineralization in articular cartilage, understanding eATP regulation may lead to effective therapies for crystal-associated arthritis.


Subject(s)
Adenosine Triphosphate/metabolism , Calcification, Physiologic/physiology , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Diphosphates/metabolism , Extracellular Space/metabolism , Membrane Proteins/metabolism , Adenoviridae , Adenylyl Cyclases/metabolism , Animals , Colforsin , Genetic Vectors , Humans , Hydrolysis , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Intercellular Signaling Peptides and Proteins , Phosphate Transport Proteins , Probenecid , Protein Kinase C/metabolism , Sus scrofa , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
12.
J Bone Miner Res ; 24(8): 1493-505, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19335222

ABSTRACT

Hypophosphatasia (HPP) is the inborn error of metabolism characterized by low serum alkaline phosphatase (ALP) activity caused by inactivating mutations within TNSALP, the gene that encodes the "tissue-nonspecific" isoenzyme of ALP (TNSALP). In HPP, extracellular accumulation of inorganic pyrophosphate, a TNSALP substrate, inhibits hydroxyapatite crystal growth leading to rickets or osteomalacia. Chronic recurrent multifocal osteomyelitis (CRMO) is the pediatric syndrome of periarticular pain and radiographic changes resembling infectious osteomyelitis but without lesional pathogens. Some consider CRMO to be an autoinflammatory disease. An unrelated boy and girl with the childhood form of HPP suffered chronic, multifocal, periarticular pain, and soft tissue swelling. To investigate this unusual complication, we evaluated their cumulative clinical, biochemical, radiological, and histopathological findings and performed mutation analysis of their TNSALP alleles. The earliest radiographic disturbances were typical of childhood HPP. Subsequently, changes consistent with CRMO developed at sites where there was pain, including lucencies, osteosclerosis, and marked expansion of the underlying metaphyses. Bone marrow edema was shown by MRI. Biopsies of affected bone showed nonspecific histopathological findings and no pathogens. The boy was heterozygous (c.1133A>T, p.D378V) and the girl compound heterozygous (c.350A>G, p.Y117C, c.400_401AC>CA, p.T134H) for different TNSALP missense mutations. Nonsteroidal anti-inflammatory drugs diminished their pain, which improved or resolved at maturity. HPP should be considered when CRMO is a diagnostic possibility. Metaphyseal radiographic changes and marrow edema associated with periarticular bone pain and soft tissue swelling suggestive of osteomyelitis can complicate childhood HPP.


Subject(s)
Hypophosphatasia/diagnosis , Osteomyelitis/diagnosis , Adolescent , Alkaline Phosphatase/genetics , Chronic Disease , Diagnosis, Differential , Female , Humans , Male , Mutation, Missense , Osteomyelitis/genetics , Osteomyelitis/physiopathology , Recurrence
13.
Arthritis Rheum ; 58(10): 3270-4, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18821676

ABSTRACT

OBJECTIVE: Basic calcium phosphate (BCP) crystals are common components of osteoarthritis (OA) synovial fluid. Progress in understanding the role of these bioactive particles in clinical OA has been hampered by difficulties in their identification. Tetracyclines stain calcium phosphate mineral in bone. The aim of this study was to investigate whether tetracycline staining might be an additional or alternative method for identifying BCP crystals in synovial fluid. METHODS: A drop of oxytetracycline was mixed with a drop of fluid containing synthetic or native BCP, calcium pyrophosphate dihydrate (CPPD), or monosodium urate (MSU) crystals and placed on a microscope slide. Stained and unstained crystals were examined by light microscopy, with and without a portable broad-spectrum ultraviolet (UV) pen light. A small set of characterized synovial fluid samples were compared by staining with alizarin red S and oxytetracycline. Synthetic BCP crystals in synovial fluid were quantified fluorimetrically using oxytetracycline. RESULTS: After oxytetracycline staining, synthetic and native BCP crystals appeared as fluorescent amorphous aggregates under UV light. Oxytetracycline did not stain CPPD or MSU crystals or other particulates. Oxytetracycline staining had fewer false-positive test results than did alizarin red S staining and could provide estimates of the quantities of synthetic BCP crystals in synovial fluid. CONCLUSION: With further validation, oxytetracycline staining may prove to be a useful adjunct or alternative to currently available methods for identifying BCP crystals in synovial fluid.


Subject(s)
Calcium Pyrophosphate/analysis , Oxytetracycline , Synovial Fluid/chemistry , Animals , Feasibility Studies , Histocytochemistry/methods , Humans , Microscopy, Ultraviolet/instrumentation , Sus scrofa
16.
Curr Opin Rheumatol ; 16(3): 268-72, 2004 May.
Article in English | MEDLINE | ID: mdl-15103256

ABSTRACT

PURPOSE OF REVIEW: Extracellular inorganic pyrophosphate (ePPi) both inhibits and promotes different forms of pathologic mineralization. Basic calcium phosphate (BCP) deposition results from depressed levels of ePPi while excess levels of ePPi leads to calcium pyrophosphate dihydrate crystal deposition (CPPD) disease. These crystals are also often identified in patients with osteoarthritis, the most prevalent form of arthritis causing significant morbidity. RECENT STUDIES: The two primary hypotheses for generation of ePPi, export of inorganic pyrophosphate through the multipass transmembrane protein ANK and generation of ePPi by ectoenzyme activity, continue to be supported and better understood through animal models and study of families with CPPD deposition disease. SUMMARY: As the pathophysiology of crystal formation in both articular cartilage and synovial fluid is better understood, the opportunity for prevention and treatment of pathologic mineralization increases. In particular, a more complex understanding of the ank gene, ectoenzyme PC-1, and the transglutaminase enzyme family may eventually translate into therapeutic application for both BCP deposition and CPPD deposition disease.


Subject(s)
Calcium Pyrophosphate/metabolism , Chondrocalcinosis/metabolism , Chondrocytes/metabolism , Diphosphates/metabolism , Calcium Phosphates/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/physiopathology , Crystallization , Extracellular Space/metabolism , Humans , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Synovial Fluid/metabolism , Transglutaminases/metabolism
17.
Arthritis Rheum ; 48(9): 2627-31, 2003 Sep.
Article in English | MEDLINE | ID: mdl-13130483

ABSTRACT

OBJECTIVE: To analyze ANKH in families with calcium pyrophosphate dihydrate crystal deposition disease (CPPD) for disease-causing mutations. METHODS: Two US families (one of British ancestry and the other of German/Swiss ancestry) with autosomal-dominant CPPD, whose disease phenotypes were found to be linked to chromosome 5p15.1 (locus symbol CCAL2), were screened by direct sequencing for mutations in ANKH, a gene in the CCAL2 candidate interval that has been shown to harbor mutations in other families with CPPD. Observed sequence variants were confirmed by antisense sequencing, and expression of the mutant allele was verified by reverse transcriptase-polymerase chain reaction amplification of messenger RNA followed by direct sequencing. RESULTS: The two US families displayed the same mutation at position 5 of the ANKH gene product (P5T). All affected members were heterozygous for the P-to-T variant, and the mutation was not seen in 204 control alleles. The two families displayed distinct disease haplotypes, suggesting that they were unrelated to each other. CONCLUSION: These observations represent the fourth and fifth families with heritable CPPD whose disease phenotypes are linked to the CCAL2 locus and who have missense mutations in the amino terminus of ANKH. This same position (P5) was the site of a missense mutation in an Argentine family of northern Italian ancestry; however, the sequence variant in that family generated a P5L mutation. The distinct disease haplotypes among the 3 families with P5 mutations suggest that the mutations arose independently and that the evolutionarily conserved P5 position of ANKH may represent a hot spot for mutation in families with autosomal-dominant CPPD.


Subject(s)
Chondrocalcinosis/genetics , Membrane Proteins/genetics , Mutation, Missense , DNA Mutational Analysis , Family Health , Female , Genetic Linkage , Haplotypes , Humans , Male , Pedigree , Phosphate Transport Proteins
18.
J Bone Miner Res ; 18(4): 624-36, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12674323

ABSTRACT

An 8-month-old girl who seemed certain to die from the infantile form of hypophosphatasia, an inborn error of metabolism characterized by deficient activity of the tissue-nonspecific isoenzyme of alkaline phosphatase (TNSALP), underwent the first trial of bone marrow cell transplantation for this heritable type of rickets. After cytoreduction, she was given T-cell-depleted, haplo-identical marrow from her healthy sister. Chimerism in peripheral blood and bone marrow became 100% donor. Three months later, she was clinically improved, with considerable healing of rickets and generalized skeletal remineralization. However, 6 months post-transplantation, worsening skeletal disease recurred, with partial return of host hematopoiesis. At the age of 21 months, without additional chemotherapy or immunosuppressive treatment, she received a boost of donor marrow cells expanded ex vivo to enrich for stromal cells. Significant, prolonged clinical and radiographic improvement followed soon after. Nevertheless, biochemical features of hypophosphatasia have remained unchanged to date. Skeletal biopsy specimens were not performed. Now, at 6 years of age, she is intelligent and ambulatory but remains small. Among several hypotheses for our patient's survival and progress, the most plausible seems to be the transient and long-term engraftment of sufficient numbers of donor marrow mesenchymal cells, forming functional osteoblasts and perhaps chondrocytes, to ameliorate her skeletal disease.


Subject(s)
Bone Marrow Transplantation , Hypophosphatasia/therapy , Alkaline Phosphatase/deficiency , Alkaline Phosphatase/genetics , Base Sequence , Bone and Bones/diagnostic imaging , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Hypophosphatasia/diagnostic imaging , Hypophosphatasia/genetics , Hypophosphatasia/metabolism , Infant , Mutation, Missense , Radiography , Stromal Cells/transplantation
19.
Curr Opin Rheumatol ; 15(3): 311-4, 2003 May.
Article in English | MEDLINE | ID: mdl-12707586

ABSTRACT

Accumulation of excess inorganic pyrophosphate in cartilage matrix leads to calcium pyrophosphate dihydrate crystal deposits. Recent animal and human studies now support a role for physiologic extracellular pyrophosphate levels in preventing ectopic apatite calcification in joints and extracellular tissues. Extracellular pyrophosphate is likely generated by ectoenzymes and/or is a consequence of transport of intracellular pyrophosphate to the extracellular space. Generation of pyrophosphate by chondrocytes is modulated by aging, several soluble growth factors and cytokines, and transglutaminase. The transduction mechanisms involved in regulating pyrophosphate metabolism include protein kinase C and adenylyl cyclase. It appears that regulation of extracellular pyrophosphate levels within a narrow range is complex and necessary for appropriate mineral homeostasis in articular and nonarticular tissues.


Subject(s)
Calcinosis/prevention & control , Calcium Pyrophosphate/metabolism , Cartilage, Articular/physiopathology , Chondrocalcinosis/physiopathology , Animals , Chondrocytes/metabolism , Chondrocytes/physiology , Extracellular Space , Female , Homeostasis/physiology , Humans , Male , Risk Factors , Sensitivity and Specificity
20.
Arthritis Rheum ; 46(12): 3218-29, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12483726

ABSTRACT

OBJECTIVE: Excess accumulation of extracellular inorganic pyrophosphate (ePPi) in aged human cartilage is crucial in calcium pyrophosphate dihydrate (CPPD) crystal formation in cartilage matrix. Two sources of ePPi are ePPi-generating ectoenzymes (NTPPPH) and extracellular transport of intracellular PPi by ANK. This study was undertaken to evaluate the role of NTPPPH and ANK in ePPi elaboration, by investigating expression of NTPPPH enzymes (cartilage intermediate-layer protein [CILP] and plasma cell membrane glycoprotein 1 [PC-1]) and ANK in human chondrocytes from osteoarthritic (OA) articular cartilage containing CPPD crystals and without crystals. METHODS: Chondrocytes were harvested from knee cartilage at the time of arthroplasty (OA with CPPD crystals [CPPD], n = 8; OA without crystals [OA], n = 10). Normal adult human chondrocytes (n = 1) were used as a control. Chondrocytes were cultured with transforming growth factor beta1 (TGFbeta1), which stimulates ePPi elaboration, and/or insulin-like growth factor 1 (IGF-1), which inhibits ePPi elaboration. NTPPPH and ePPi were measured in the media at 48 hours. Media CILP, PC-1, and ANK were determined by dot-immunoblot analysis. Chondrocyte messenger RNA (mRNA) was extracted for reverse transcriptase-polymerase chain reaction to study expression of mRNA for CILP, PC-1, and ANK. NTPPPH and ANK mRNA and protein were also studied in fresh frozen cartilage. RESULTS: Basal ePPi elaboration and NTPPPH activity in conditioned media from CPPD chondrocytes were elevated compared with normal chondrocytes, and tended to be higher compared with OA chondrocytes. Basal expression of mRNA for CILP (chondrocytes) and ANK (cartilage) was higher in both CPPD chondrocytes and CPPD cartilage extract than in OA or normal samples. PC-1 mRNA was less abundant in CPPD chondrocytes and cartilage extract than in OA chondrocytes and extract, although the difference was not significant. CILP, PC-1, and ANK protein levels were similar in CPPD, OA, and normal chondrocytes or cartilage extracts. Both CILP and ANK mRNA expression and ePPi elaboration were stimulated by TGFbeta1 and inhibited by IGF-1 in chondrocytes from all sources. CONCLUSION: CILP and ANK mRNA expression correlates with chondrocyte ePPi accumulation around CPPD and OA chondrocytes, and all respond similarly to growth factor stimulation. These findings suggest that up-regulated CILP and ANK expression contributes to higher ePPi accumulation from CPPD crystal-forming cartilage.


Subject(s)
Cartilage, Articular/metabolism , Chondrocalcinosis/metabolism , Extracellular Matrix Proteins/metabolism , Hyalin/metabolism , Membrane Proteins/metabolism , Pyrophosphatases/metabolism , Adult , Aged , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Diphosphates/metabolism , Extracellular Space/metabolism , Growth Substances/pharmacology , Humans , In Vitro Techniques , Middle Aged , Osteoarthritis/metabolism , Phosphate Transport Proteins , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...