Metabolic and Endocrine Bone Diseases






  • Chapter Outline



  • General Pathophysiology



  • Rickets



  • Renal Osteodystrophy



  • Parathyroid Disorders



  • Vitamin Disorders



  • Hypophosphatasia



  • Idiopathic Hyperphosphatasia



  • Growth Hormone Deficiency



  • Hypothyroidism



  • Idiopathic Juvenile Osteoporosis



  • Osteogenesis Imperfecta




General Pathophysiology


Maintenance of healthy bone in children is a multiorgan process that involves the coordination of various hormones and factors that affect calcium and phosphate metabolism. Parathyroid gland, kidney, liver, intestine, and bone are the key organs that secrete, regulate, or respond to these factors. Calcium, phosphate, vitamin D, parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), tissue-nonspecific alkaline phosphatase (TNSALP), and calcium-sensing receptor (CaSR) are some of the factors known to affect bone metabolism. Metabolic bone disease is caused by a dysfunction of one of the organs, such as renal failure, or abnormality of one of the factors involved with the bone metabolic process. Mutations of the genes that regulate calcium and phosphate metabolism have been described with a greater frequency because of the advancement of genetic research. The defect or deficiency of type I collagen chains, which are the major structural macromolecules of the bone matrix fiber network, can also have a detrimental effect on the function and metabolism of bone, as seen in osteogenesis imperfecta. To understand the pathophysiology and clinical manifestations of various metabolic bone disorders that affect children, it is important to have a clear understanding of how various organs, hormones, and factors involved in calcium and phosphate metabolism interact and affect the normal bone homeostatic process.


The body is extremely sensitive to serum calcium levels; a disturbance in calcium balance leads to abnormal irritability, conductivity, and contractility of the cardiovascular and neurologic systems. Only a very small portion of the body’s calcium is present in the bloodstream, and its level is tightly controlled. Almost all the body’s calcium is stored in bone as hydroxyapatite, which is composed of calcium and phosphate compounds; thus, if extra calcium is needed in the bloodstream to maintain cardiac or neurologic function, bone is the source of the required calcium, which is released via osteoclast-mediated bone resorption. Various factors, including PTH, regulate the activity of osteoclasts. For healthy bone formation, normal levels of calcium and phosphate are required, as demonstrated by hypophosphatemic rickets, in which renal phosphate wasting produces bone abnormalities. TNSALP activity is also important for bone formation, as evidenced by clinical features of hypophosphatasia.


Because of the liver and kidneys’ involvement in calcium and vitamin D metabolism, hepatic or renal diseases can lead to metabolic bone diseases. Serum calcium is under the regulation of vitamin D and PTH ( Fig. 42-1 ). PTH functions to increase serum calcium levels by increasing osteoclastic bone resorption, renal reabsorption of calcium in the proximal tubules, and stimulating the conversion of inactive 25-hydroxyvitamin D—25(OH)D—to its active form, 1,25(OH) 2 D (calcitriol). A low level of calcium (hypocalcemia) itself also stimulates the conversion of inactive form of vitamin D to its active form. The active form of vitamin D increases the intestinal absorption of calcium and increases renal reabsorption of phosphate. FGF23 is also an important negative regulator of vitamin D activation produced by bone cells. It inhibits 1α-hydroxylase activity, which is required for vitamin D activation. FGF23 also plays a key role in phosphate homeostasis by increasing renal phosphate excretion, thus decreasing the serum phosphate level (hypophosphatemia). An increase in FGF23 activity is an important pathophysiologic component of vitamin D–resistant rickets, which is associated with hypophosphatemia and low 1,25(OH) 2 D activity.




FIGURE 42-1


A, Metabolic control of calcium metabolism. B, Vitamin D metabolism. GI, Gastrointestinal; PTH, parathyroid hormone.


Vitamin D formation and activation is a complex process that involves the gut, skin, liver, and kidney. Ergosterol (provitamin D) is ingested and absorbed from the small intestine. These precursors of vitamin D must be absorbed from the gut and undergo conversion to vitamin D 2 (ergocalciferol) before undergoing hydroxylation to become active. Because these precursors are fat-soluble, gastrointestinal or hepatic diseases that produce steatorrhea result in an inability to absorb vitamin D. The skin is the site of conversion of 7-dehydrocholesterol to vitamin D 3 (cholecalciferol). This change occurs as a result of exposure to ultraviolet light.


Vitamins D 2 and D 3 are biologically inert and must undergo a series of hydroxylation reactions in their transformation to the active form, 1,25(OH) 2 D. The first hy­­droxylation takes place in the liver to produce 25(OH)D. The liver also produces 7-dehydrocholesterol (a provitamin D).The second hydroxylation occurs in the kidney via 1α-hydroxylase, which produces the active form, 1,25(OH) 2 D. Vitamin D activation is stimulated by hypocalcemia and high levels of PTH and inhibited by FGF23. An increase in 1,25(OH) 2 D in turn increases FGF23 production, which downregulates 1α-hydroxylase activity as a negative feedback mechanism. In renal failure, the conversion of inactive to active vitamin D is decreased because of the kidney damage itself as well as an increase in FGF23 production, which is stimulated by hyperphosphatemia secondary to the renal failure.




Rickets


Nutritional Rickets


Vitamin D deficiency in the diet leads to nutritional rickets. Although vitamin D deficiency is rare in developed countries because of vitamin D fortification, it is not absent from these areas. Some studies show a reemerging burden of rickets in developed countries. Certain populations are at risk, including premature infants, infants with prolonged breast feeding, children on a vegetarian diet, black children, and immigrant populations from the Indian subcontinent, Africa, and the Middle East. *



* References .

An increased frequency of nutritional rickets in the United States in children with dark skin pigmentation who are breast-fed past 6 months of age without vitamin D supplementation has been reported. In the developed countries, nutritional rickets is also seen in patients with celiac or hepatic disease, which affects vitamin D absorption from the gut. Children with nutritional rickets are usually first seen between the ages of 6 months and 3 years. Initial findings are listlessness, periarticular swelling, or angular deformities. Hypocalcemic seizure is also a common presentation during the first 2 years of life.


In the early twentieth century, Hess used a trial of cod liver oil to treat 65 infants with rickets and found that the rickets resolved in 92% of the infants during a 6-month course of treatment. This led to the development of the first rickets clinic in 1917. Mellanby and Park, in the mid-1920s, were the first to suggest that rickets could be prevented by adequate vitamin D intake. Since that time, milk and dairy products have been fortified with vitamin D. Thus it is only in cases of malnutrition and unusual dietary practices that vitamin D deficiency rickets is seen.


Pathology


In rickets, the primary disturbance is failure of mineralization of cartilage and osteoid tissue, which decreases longitudinal bone growth and weakens the mechanical properties of tubular bone. Thus the pathologic manifestations of the disease are most noticeable around growth plates and along long bones. Normal longitudinal growth of bone is the result of endochondral ossification. The key elements of the process are proliferation of chondrocytes in columns (zone of proliferation), cellular maturation to become hypertrophic chondrocytes, calcification of the cartilage matrix (zone of provisional calcification), vascular invasion of the terminal hypertrophic chondrocytes, and deposition of new bone. In rickets, failure of deposition of calcium along the matrix of the maturing chondrocyte columns is observed, along with a disorderly invasion of cartilage by blood vessels, lack of resorption at the zone of provisional calcification, and consequently increased thickness of the physis ( Fig. 42-2 ). The chondrocytes proliferate normally, but the normal process of endochondral ossification fails to take place.




FIGURE 42-2


Histologic appearance of rickets. A photomicrograph through the epiphyseal-metaphyseal junction shows uncalcified osteoid tissue, failure of deposition of calcium along the mature cartilage cell columns, and disorderly invasion of cartilage by blood vessels (×25, H & E).


Osteoblastic activity on the endosteal and periosteal surfaces of bone is normal, and abundant osteoid (nonmineralized bone matrix) is formed. With defective mineralization of osteoid, osteoclastic resorption of the osteoid does not take place. However, increased resorption of the mineralized bone because of secondary hyperparathyroidism occurs. Hence, the overly abundant osteoid produced by normal osteoblasts generates widened osteoid seams. Because of a lack of resorption of osteoids, the osteoid islets may even persist down into the diaphysis.


In rickets, an abnormality in the arrangement of bundles of collagen fibers in compact bone also exists. Instead of running parallel to the haversian canals, they course perpendicularly and are biomechanically inferior. Grossly, the rachitic bone is soft and becomes misshapen under the force of weight bearing. If the disease remains untreated, angular deformities of the lower extremities and deformities of the thoracic cage and pelvis may develop.


After treatment of rickets with vitamin D, calcium absorption increases and calcification of the cartilage columns and osteoid occurs. Osteoclasts resorb the calcified cartilage, and normal remodeling and improvement of bone follows.


Laboratory Findings


Vitamin D deficiency results in an inability to absorb calcium and phosphorus from the gut. Vitamin D status is best evaluated by measuring the level of serum 25(OH)D, which reflects the degree of deficiency. In contrast, the serum level of 1,25(OH) 2 D is not helpful because it may be normal in most patients with vitamin D deficiency. The PTH level is elevated in response to hypocalcemia (secondary hyperparathyroidism), which attempts to ameliorate the serum calcium level. Serum calcium levels are normal to mildly decreased, but phosphate levels are low (hypophosphatemia) and alkaline phosphatase levels are high ( Table 42-1 ). Urinary excretion of calcium is low because of enhanced renal tubular reabsorption.



Table 42-1

Biochemical Abnormalities in Rickets























































Biochemical Abnormality
Type of Rickets Calcium Phosphate Alkaline Phosphatase PTH 25-(OH) vitamin D 1,25-(OH) 2 vitamin D
Nutritional Nl Nl↓ ↓↓
Vitamin D–resistant (XLH, RTA, Fanconi, oncogenic) Nl Nl Nl Nl
Vitamin D–dependent type I (inability to hydroxylate) ↑↑ ↓↓
Vitamin D–dependent type II (receptor insensitivity) Nl↑↑ ↑↑↑↑
Renal osteodystrophy Nl↓ ↑↑ Nl ↓↓

Nl, Normal; PTH, parathyroid hormone; RTA, renal tubular acidosis; XLH, X-linked hypophosphatemia.


Clinical Features


The clinical features of nutritional rickets depend on the severity of the disease and may be subtle. Infants demonstrate generalized muscular weakness, lethargy, and irrita­bility. Sitting, standing, and walking are delayed. The abdomen may appear protuberant.


Early bone manifestations include a slight thickening of the ankles, knees, and wrists. Beading of the ribs, referred to as the rachitic rosary, is caused by enlargement of the costochondral junctions. As the disease continues, the pull of the diaphragm on the ribs produces a horizontal depression known as Harrison’s groove. Short stature results from insufficient longitudinal growth. Pectus carinatum is caused by forward projection of the sternum. Closure of the fontanelles is delayed and the sutures are thickened, which leads to a skull appearance described as resembling hot cross buns. The dentition is affected, with delays in appearance of the teeth and defects in the enamel.


As the child begins standing and walking, the softened long bones bow, and it is at this time that the child is usually brought to the orthopaedic surgeon for a diagnosis. In toddlers, bowleg, or genu varum, is one of the most common initial signs. In older children, genu valgum and coxa vara may be initial features. Stress fractures of the long bones may be present. Children may be seen acutely with unexplained fractures suggesting child abuse, tetany, and hypocalcemic seizures. Later, kyphoscoliosis may develop.


Radiographic Findings


Failure of the physeal cartilage to calcify and undergo normal endochondral ossification leads to an increased thickness of the physis and a hazy appearance of the provisional zone of calcification. The widened growth plate is particularly suspect for rickets, which differentiates this rare condition from the more common physiologic angular deformities of the lower extremities ( Fig. 42-3 ). The metaphysis abutting the physis is brushlike in appearance, with islands or columns of cartilage persisting well into the metaphysis ( Fig. 42-4 ). The metaphysis also appears cupped or flared. The bones have an osteopenic appearance overall, with thinning of the cortices. The bony trabeculae are indistinct. Looser’s lines, or radiolucent transverse bands (pseudofracture lines) that extend across the axes of the long bones, are evident on radiographs in 20% of patients with rickets.




FIGURE 42-3


Radiographs obtained in a 1-year-old black girl with nutritional rickets. A, Forearm. B, Knee. C, Pelvis. All physes are widened and the metaphyses are indistinct. Cupping is most prominent in the metaphysis of the distal radius and ulna and at the knee. See also Figure 42-6 .



FIGURE 42-4


A, Hazy metaphysis with cupping in a young boy with rickets. B, Accentuated genu varum is present. C, With vitamin D replacement therapy, the bony lesions healed in 6 months.


As the rickets continues, deformities of the long bones, ribs, pelvis, and spine develop. Thoracolumbar kyphosis—rachitic cat back—may be apparent on radiographs.


Although the diagnosis of nutritional rickets should be made on review of plain films, bone scintigraphy has been used in neonates to confirm the diagnosis and pick up areas of fractures. Increased uptake is seen. With treatment, mineralization occurs and radiographic abnormalities rapidly become normal. The physis thins, and bone density increases ( Fig. 42-5 ).




FIGURE 42-5


Radiographic appearance of the wrist of the girl whose radiographs (rickets) appear in Figure 42-3 after 4 months of treatment with vitamin D. The osteopenia has resolved and the physis is narrowed.


Treatment


Rickets is treated by the administration of vitamin D under the supervision of a pediatric specialist in metabolic bone disease. The usual course of treatment is 6 to 10 weeks. After 2 to 4 weeks, radiographs show improvement in mineralization. Tetracycline labeling shows response to therapy, with areas of new mineralization being labeled with the drug. Tetracycline should not be used in young children, however, because of staining of the teeth. If the child does not respond to vitamin D therapy, vitamin D–resistant rickets should be suspected. Because residual deformity is rare after medical treatment of nutritional rickets, there is no specific orthopaedic treatment of nutritional rickets.


Rickets of Prematurity


Very premature infants are particularly at risk for the development of nutritional rickets. Risk factors include hepatobiliary disease, total parenteral nutrition, diuretic therapy, physical therapy with passive motion, and chest percussion therapy. These infants are seen with pathologic fractures in the neonatal intensive care unit. With treatment of the rickets, the fractures heal readily, with minimal other treatment required. Resolution of the rachitic changes and fractures occurs as the infants gain weight.


Drug-Induced Rickets


Certain antiepileptic medications have been known to produce rachitic changes in children. Seizure medications that affect the liver may induce the cytochrome P-450 microsomal enzyme system and decrease levels of vitamin D. Hypocalcemia develops, which can aggravate the seizure disorder. Treatment with vitamin D is very helpful. The condition should be suspected in neurologic patients with seizures who begin sustaining frequent fractures.


Vitamin D–Resistant Rickets


Vitamin D–resistant rickets, also known as hereditary or familial hypophosphatemic rickets, encompasses a group of disorders in which normal dietary intake of vitamin D is insufficient to achieve normal mineralization of bone because of pathologic renal phosphate wasting. Hereditary hypophosphatemic rickets can be inherited as an X-linked dominant, autosomal dominant, or autosomal recessive form. X-linked dominant disease is the most frequent form of hereditary rickets, with an incidence of 1 in 20,000, and is considered the prototypic disorder of renal phosphate wasting. In 1995, a phosphate-regulating gene with homologies to endopeptidases on the X-chromosome (PHEX) was identified as the cause of X-linked dominant disease. The PHEX mutation produces elevated levels of FGF23 by a yet unidentified mechanism. FGF23 is a circulating hormone produced by osteoblasts and osteocytes that reduces renal phosphate reabsorption and the conversion of 25(OH)D to its active form, 1,25(OH) 2 D, by suppressing renal 25-hydroxy-1α-hydroxylase activity. Inhibition of 1α-hydroxylase prevents the normal compensatory increase in active vitamin D formation associated with hypophosphatemia. Thus increased FGF23 activity leads to increased renal phosphate excretion, hypophosphatemia, short stature, long bone bowing, and other radiographic features of rickets.


In autosomal dominant form of hereditary rickets, mutations in FGF23 have been identified that prevent the degradation of this hormone and produce renal phosphate wasting. In the autosomal recessive form of hereditary rickets, mutations in the dentin matrix protein 1 (DMP1) gene and ENPP1 have been reported. DMP1 is an important regulatory protein produced by osteoblasts and osteocytes that regulates the growth and development of dentin, bone, and cartilage and also plays a role in matrix mineralization. Thus DMP1 mutations impair osteocyte maturation and bone mineralization. DMP1 mutations also produce elevated levels of FGF23 through an undefined mechanism, leading to phosphaturia and hypophosphatemia. Other rare forms of hereditary rickets include hereditary hypophosphatemic rickets with hypercalciuria (autosomal recessive inheritance with mutation in SLC34A3) and autosomal recessive Fanconi syndrome caused by SLC34A1 mutation. In renal tubular acidosis, the kidney excretes fixed base and wastes bicarbonate, which also leads to wasting of calcium and sodium. The alkaline urine results in the precipitation of calcium and severe renal calcinosis.


Laboratory Findings


Laboratory studies reveal normal or almost normal levels of calcium. The serum phosphate concentration is significantly decreased, whereas the 1,25(OH) 2 D level may be inappropriately low in response to the hypophosphatemia. The PTH level is normal. Urine assays for phosphate demonstrate an increased concentration of phosphate in the urine. The serum alkaline phosphatase concentration is elevated but not to the levels seen with nutritional rickets (see Table 42-1 ).


Clinical Features


The disease usually becomes apparent at a slightly older age than nutritional rickets, with most patients becoming symptomatic between 1 and 2 years of age. Severe hypophosphatemic rickets can be recognized in early infancy, and when the disease is suspected because of the family history, laboratory determination of phosphorus concentrations can lead to the diagnosis in infants as young as 3 months. The usual initial complaints are delayed walking and angular deformities of the lower extremities. In contrast to what is seen in nutritional rickets, systemic manifestations such as irritability and apathy are minimal.


Physical findings in hypophosphatemic rickets include skeletal deformities, which resemble those seen in nutritional rickets but, because of the chronicity of the disease, become much more severe. Once affected children begin to walk, genu varum develops, although genu valgum may occur in some children ( Fig. 42-6 ). Periarticular enlargement is present as a result of widening of the physes and metaphyses. The rachitic rosary may also occur.




FIGURE 42-6


A, Unilateral genu varum in a 12-year-old child with poorly controlled vitamin D–resistant rickets. B, Same child at 15 years of age after right medial hemiepiphysiodesis of the femur. Left genu varum is now apparent.


Short stature is a feature of hypophosphatemic rickets. Height is usually 2 standard deviations (SDs) below the mean for age in these patients.


Radiographic Findings


The radiographic changes are the same as those seen in nutritional rickets and include physeal widening and indistinct osteopenic metaphyses. In the lower extremities, genu varum is obvious, and the distal femoral and proximal tibial physes are particularly widened medially ( Fig. 42-7 ). Coxa vara is present, and there may be general anterior and lateral bowing of the entire femur. The varus of the tibia is also generalized, not only present proximally but also producing varus angulation of the ankle.




FIGURE 42-7


A and B, Anteroposterior radiographs of the left and right lower extremities of a standing 7-year-old child with familial hypophosphatemic rickets. Severe genu varum and anterolateral bowing of the femur are evident. The distal femoral and proximal tibial physes are widened medially. See Figures 42-8 and 42-9 .


The upper extremities are involved as well, but to a lesser degree because of absence of the influence of weight bearing ( Fig. 42-8 ).




FIGURE 42-8


Physeal widening and metaphyseal cupping of the distal end of the radius and ulna in a 7-year-old child (same as in Fig. 42-7 ) with vitamin D–resistant rickets.


Treatment


Medical Treatment


The medical treatment of hypophosphatemic rickets is best managed by a pediatric nephrologist with expertise in metabolic bone disease. The usual treatment consists of oral replacement of phosphorus in large doses and the administration of an active form of vitamin D, calcitriol or alfacalcidol. Analogues of vitamin D 3 (1,25-dihydroxyvitamin D 3 ) are several hundred times more potent than the original form of vitamin D in treating hereditary rickets. The therapeutic target of medical therapy should not be to normalize the serum phosphate level because achieving normalization may not be a practical goal and may lead to overmedication and greater side effects. Focus should be on improving the skeletal deformity, height, and physeal function. In general, growth and skeletal deformities improve with medical therapy. Initiation of therapy in infancy has a greater impact on height but does not completely normalize skeletal development.


Nephrocalcinosis is a significant complication of medical treatment. In one study, renal calcinosis was present in 79% of treated children with hypophosphatemic rickets, and the severity of the calcinosis correlated with the dose of phosphorus. Because nephrocalcinosis is a significant complication, the decision whether to offer treatment to children with hypophosphatemic rickets has become controversial. Studies have shown that longitudinal growth is greater in children who undergo vitamin D treatment.


Treatment of children with hypophosphatemic rickets with growth hormone has been shown to increase height and have beneficial effects on bone density and phosphate retention. Preliminary studies reported that the administration of growth hormone with vitamin D increases the serum phosphate concentration and may reduce the incidence of nephrocalcinosis.


Orthopaedic Treatment


The orthotic management of vitamin D–resistant rickets has not been efficacious. If patients are experiencing increasing pain or difficulty walking, surgical correction of angular deformities should be performed. It is important to work closely with the nephrologist or endocrinologist who is managing the medical therapy because calcium levels can suddenly increase in a patient who is immobilized after surgery. Discontinuation of vitamin D before surgery should be discussed.


The deformity most commonly seen in patients with hypophosphatemic rickets is a gradual anterolateral bowing of the femur, combined with tibia vara. Multilevel osteotomy is generally required to satisfactorily correct the mechanical axis of the limb ( Fig. 42-9 ). The mechanical axis should be mildly overcorrected at surgery. The suggested fixation varies among reports. External fixation allows fine tuning of the alignment postoperatively, when the patient is able to stand ( Fig. 42-10 ). Others advocate the use of intramedullary fixation or plating ( Fig. 42-11 ). Regardless of the type of fixation used, careful preoperative planning of the surgical treatment of these multiplanar deformities is crucial to restoring alignment.




FIGURE 42-9


Postoperative radiographs of the child whose imaging findings are shown in Figures 42-7 and 42-8 . A, Appearance after distal femoral, proximal tibial, and distal tibial osteotomies for treatment of genu varum. B, Varus is recurring 1 year after surgery.



FIGURE 42-10


A, Clinical appearance of a 13-year-old girl with severe genu varum secondary to vitamin D–resistant rickets. Calluses on the knees were caused by crawling because of knee pain. B, Treatment consisted of distal femoral, proximal tibial, and distal tibial corticotomies and gradual correction with the Ilizarov device. Surgery on the two legs was staged because of the extent of the frame. C, Clinical appearance of the lower extremities at the end of treatment.



FIGURE 42-11


A, Coxa vara and genu varum in a 5-year-old boy with vitamin D–resistant rickets. B, Postoperative radiograph obtained after corrective osteotomy with plate fixation of the proximal femora and osteotomy with K-wire fixation of the proximal tibiae. C, Alignment remained satisfactory at 2-year follow-up.


Recurrent deformity is a common sequela of osteotomies in patients with hypophosphatemic rickets. Younger patients have a higher risk of recurrence. For this reason, milder deformities should not be corrected in early childhood. Some children have severe varus at a very young age that leads to thrust during gait. When gait is compromised or symptoms or pain is present, osteotomy should be performed and the alignment monitored for recurrent deformity.


Spinal deformity may be seen in patients with hypophosphatemic rickets. Kyphoscoliosis, Arnold-Chiari malformations, and spinal stenosis have all been described in patients with vitamin D–resistant rickets.


Adults with hypophosphatemic rickets are prone to the development of arthritis. Degradation of articular cartilage resembling osteochondritis dissecans has been described. Joint stiffness and bone pain are common complaints.


Tumor-Related Hypophosphatemic Rickets


An association between benign and malignant tumors and hypophosphatemic rickets has been described, termed oncogenic hypophosphatemic osteomalacia Conditions such as neurofibromatosis and fibrous dysplasia produce rickets on rare occasion. Osteoblastoma, hemangiopericytoma of bone, and skin tumors have produced rachitic changes in bone by disrupting the renal tubular resorption of phosphate. Certain tumors have been found to secrete phosphatonins, such as FGF23, that result in phosphaturia. Oncogenic rickets should be suspected in older children with hypophosphatemic rickets because the true genetic form is generally apparent by 2 years of age. The rachitic changes resolve with excision of the tumor.




Renal Osteodystrophy


As the rate of successful treatment of renal failure in children with kidney transplants has increased, the prevalence of renal osteodystrophy has risen. Manifestations of renal osteodystrophy are present in 66% to 79% of children with renal failure. Children in whom renal disease develops in infancy or early childhood are more likely to have osteodystrophy than those who are older when the renal disease develops. Renal failure in children is caused by diseases such as chronic pyelonephritis, congenital abnormalities, and polycystic kidney disease. Renal osteodystrophy is more common in renal disease secondary to congenital or hereditary conditions than in acquired renal failure. Renal osteodystrophy is distinctly different from nutritional or hypophosphatemic rickets. It is often driven by the presence of secondary hyperparathyroidism, which leads to activation of osteoclasts and resorption of bone. This type of bone involvement is termed high-turnover disease.


With improved control of hyperparathyroidism, another form of osteodystrophy, termed low-turnover disease, has been recognized. This adynamic disorder has been attributed to the use of high doses of exogenous calcium as phosphate-binding agents or during dialysis and to aggressive calcitriol therapy. Parathyroidectomy may also contribute to this syndrome. In one report, 60% of patients with chronic renal disease had a low-turnover disorder, whereas 40% had high-turnover osteitis fibrosa cystica. Thus children with renal failure are at risk for both types of metabolic bone disorder. Patients with slowly progressive forms of renal disease such as tubulointerstitial disease are at risk for the hyperparathyroid, high-turnover form of disease. Those with more rapidly progressive diseases such as the glomerular syndromes are more likely to have an adynamic metabolism. It is important to note that the adynamic form may be present with a high PTH level.


Pathophysiology


The pathophysiology of high-turnover renal osteodystrophy begins with the inability of the damaged glomerulus to excrete phosphorus, which results in hyperphosphatemia. Furthermore, advancing loss of nephrons and increased FGF23 levels decrease the production of dihydroxyvitamin D from the kidney. Calcium absorption from the small intestine is diminished in the absence of vitamin D. The serum calcium level is also diminished by the physiochemical binding of calcium to phosphate. Resulting hypocalcemia triggers the release of PTH, which increases osteoclast-mediated bone resorption in an attempt to normalize the serum calcium level. The hyperphosphatemia worsens with the release of minerals from bones, thereby leading to a cycle of bone resorption. PTH acts directly to stimulate osteoclast activity, which worsens the bony changes and leads to osteitis fibrosa.


High levels of serum phosphate are universal in renal failure. In the setting of elevated phosphate levels, calcium may precipitate out and lead to ectopic calcification in tissues. The usual areas for ectopic calcification are the corneas and conjunctivae, skin, blood vessels, and periarticular soft tissues.


Pathology


Features of rickets and hyperparathyroidism are present in renal osteodystrophy ( Fig. 42-12 ). The rachitic changes consist of failure to replace physeal chondrocytes by endochondral ossification. Physeal cartilage persists into the metaphysis. The physis is widened, and the zone of provisional calcification is irregular as a result of the lack of endochondral ossification occurring at the physis. Bony trabeculae have abundant osteoid and widened osteoid seams.




FIGURE 42-12


Histologic findings of osteodystrophy secondary to chronic renal insufficiency. A, Photomicrograph of a section through the widened physis showing extension of cartilage cells into the metaphysis (×25, ••). B, Higher magnification of the same area (×100, H & E). Note the uncalcified osteoid tissue and replacement of normal fatty bone marrow by hyperplastic fibrous tissue.


The histologic features of hyperparathyroidism include osteoclastic resorption of bone. Marrow is replaced by hyperplastic fibrous tissue. Patchy formation of new bone leads to areas of osteosclerosis, present in 20% of patients with renal osteodystrophy.


Laboratory Findings


The blood urea nitrogen level is high, as is the serum creatinine concentration. Levels of serum phosphate, alkaline phosphatase, and PTH are elevated. The serum calcium concentration is almost always low, as is the albumin level. Acidosis is present, and vitamin D levels are decreased (see Table 42-1 ).


Bone biopsy may be necessary for accurate diagnosis and can help guide treatment.


Clinical Features


Children with renal osteodystrophy resemble those with rickets. They are short for their age, and their bones are fragile. Because of the effects of weight bearing, lower extremity involvement is more severe than upper extremity involvement. Patients may complain of bone pain, and fractures occur easily. Skeletal deformities may consist of genu valgum, periarticular enlargement of the long bones, and slipped capital femoral epiphysis (SCFE). Gait may be abnormal because of muscular weakness, and a Trendelenburg gait is present in patients with SCFE. Enlargement of the costochondral cartilage may produce a rachitic rosary, as in nutritional rickets.


Radiographic Findings


Generalized osteopenia is notable, with thinning of the cortices and indistinct bony trabeculae. Overall, the bone looks blurry, like ground glass. The skull takes on a salt and pepper appearance because of the coarse granular pattern. The physes are increased in thickness, and the provisional zone of calcification looks uncalcified and indistinct ( Fig. 42-13 ). Cupping of the physes is not present, unlike the case in nutritional rickets.




FIGURE 42-13


Osteodystrophy secondary to chronic renal insufficiency in a young girl with progressive slipped capital femoral epiphyses. Anteroposterior ( A ) and lateral ( B ) radiographs of both ankles at the age of 12 years showed increased thickness of the physes and irregularity of the metaphysis at the zone of provisional calcification. C, Lateral view of the skull showing a ground-glass appearance. Note the absence of lamina dura of the teeth.


Changes of hyperparathyroidism develop with time. SCFE may be seen, even in young patients ( Fig. 42-14 ). The terminal tufts of the distal phalanges of the fingers resorb, as do the lateral end of the clavicle and the symphysis pubis. Subperiosteal resorption also occurs in the metacarpals and ulna.




FIGURE 42-14


A, Anteroposterior (AP) radiograph of a 7-year-old boy with hip pain. Slipped capital femoral epiphysis is present, osteopenia is obvious, and the physes are wide. AP ( B ) and lateral ( C ) radiographs of the hips after treatment of renal failure with dialysis. The proximal femoral physes have narrowed.


Osteosclerosis, when present, is most common at the base of the skull and in the vertebrae. The horizontal striped appearance of the spine is called a rugger jersey spine.


In severe and prolonged renal failure, peculiar, aggressive-appearing lytic areas may develop within the long bones, termed brown tumors. The surrounding cortex is thinned and then expands. The margins of a brown tumor are not well defined. Pathologic fracture may result. Radiographically, these lesions mimic malignancy. They are well visualized by magnetic resonance imaging (MRI).


Because many patients with renal failure, and all patients who have undergone kidney transplantation, are treated with steroids, the typical skeletal abnormalities seen with chronic steroid use may develop. Corticosteroid-associated osteonecrosis is common and develops most frequently in the femoral heads ( Fig. 42-15 ).




FIGURE 42-15


Avascular necrosis of the left hip in a 7-year-old boy after renal transplantation and steroid therapy.


Treatment


Medical Treatment


Treatment of the underlying renal disease is of primary importance. Dialysis and transplantation are extending the life span of these patients. Medical treatment of osteodystrophy starts with the prescription of vitamin D. Because the abnormal kidney cannot participate in the hydroxylation of provitamin D, the active 1,25(OH) 2 D form is given. Serum calcium levels are closely monitored because too much calcium leads to ectopic calcification. The use of high-dose pulsed IV, intraperitoneal, and oral calcitriol therapy has significantly decreased serum PTH levels and retarded the progression of osteitis fibrosa. Treatment of acidosis with sodium bicarbonate is also important in improving the metabolic bone disease. Phosphate-binding agents have been administered for the management of hyperphosphatemia, but their use has been falling out of favor because of problems with aluminum toxicity, which can lead to encephalopathy and worsening of the osteomalacia and osteodystrophy. Bone biopsy is useful in monitoring the response to therapy and surveillance for such complications. Aluminum toxicity is treated by administering aluminum-chelating agents.


Decreased growth is a significant problem for a child with renal insufficiency, probably because of disturbances in the growth hormone–insulin-like growth factor I axis. Administration of recombinant human growth hormone can restore growth, thus making it possible to achieve normal or improved adult height. Growth hormone biomechanically weakens the physis, so vigilance on the part of the treating physician for the development of SCFE or physiolysis must be maintained.


In renal osteodystrophy that is recalcitrant to medical treatment, parathyroidectomy may play a role in control of the bony disease.


Orthopaedic Treatment


Patients with renal osteodystrophy are referred to the orthopaedic surgeon for the treatment of three problems: (1) angular deformity of the lower extremities; (2) SCFE; and (3) avascular necrosis. In the surgical correction of any of the orthopaedic deformities in renal osteodystrophy, the hazards and complications should be carefully weighed because of the increased risks in this patient population. Anemia, hypertension, bleeding tendencies, and electrolyte imbalances are all present in patients with renal failure. The risk for infection is also increased, particularly if the patient has received a transplant and is undergoing immunosuppressive therapy. Despite these potential risks, an osteotomy can be performed safely with careful coordination of surgery and perioperative management with the pediatric nephrologist.


Angular Deformity.


Angular deformity occurs in renal osteodystrophy because the bone is soft, undermineralized, and prone to bend with weight bearing. Genu valgum is the most common deformity, but genu varum may occur in some patients. It has been proposed that if the onset of renal osteodystrophy occurs before 4 years of age, varus deformity may develop because the normal alignment of the leg is in mild varus, which then is accentuated as the bone becomes weak. Similarly, older children are predisposed to the development of genu valgum because of the normal valgus alignment of the lower extremity. Valgus at the ankle may accompany the genu valgum.


Some milder deformities will correct with medical treatment of the renal osteodystrophy. Deformities do not respond well to bracing. If the patient is symptomatic and has had optimal medical management of the osteodystrophy without resolution of deformity, an osteotomy is performed. Preoperative assessment of the deformity with long-leg standing radiography will permit the surgeon to decide the location of the deformity and how many osteotomies will be needed to correct the mechanical axis most effectively. Usually the distal end of the femur is the site of greatest deformity, but some patients also need a proximal tibial osteotomy. Internal or external fixation may be used. Application of the Ilizarov device in metabolic bone disease has met with success, although healing was delayed. Recurrence is common in patients with continuing metabolic disease, so medical treatment should be optimized before osteotomy whenever possible. Elevation of the serum alkaline phosphatase concentration above 500 U/L is a good marker of ongoing metabolic bone disease. A bone biopsy may be needed to establish that the bone is metabolically healthy before osteotomy. Milder deformity may respond to physeal stapling.


A subset of patients with genu valgum shows evidence of a proximal tibial growth disturbance in the form of a physeal abnormality in the proximal lateral tibial physis. Oppenheim and associates compared the physeal widening of the lateral physis with that seen in the medial physis in Blount disease. These patients benefit from tibial osteotomy for realignment.


Slipped Capital Femoral Epiphysis.


SCFE is associated with renal osteodystrophy, but the clinical picture of a patient with renal slips differs from the usual clinical scenario. Often the patients are younger than those with idiopathic SCFE, and obesity is not commonly seen. Bilaterality is extremely common. Radiographs show more physeal widening than is usual in SCFE, and osteopenia and blurring of the metaphysis may be obvious. The orthopaedic surgeon should be aware of the radiographic appearance of renal SCFE because, on rare occasion, patients may seek treatment of hip or groin pain while being unaware of their renal disease. In such cases it is up to the orthopaedic surgeon to make the diagnosis of renal osteodystrophy, and promptly refer the child to a nephrologist for appropriate treatment.


There are inherent problems in the surgical treatment of SCFE in renal osteodystrophy. The goal of routine treatment of SCFE is to stop proximal femoral physeal growth and thus heal the slip. This may not be a desirable goal in a very young child with renal osteodystrophy. Also, physeal healing may be difficult to achieve in the presence of osteitis fibrosa and metabolic imbalance. Fortunately, in many patients, the hip pain resolves and the proximal femoral physis narrows with medical treatment of the renal osteodystrophy, so surgery is not necessary for every patient with renal SCFE. If the slip is displaced or if symptoms persist despite good medical control of the osteodystrophy, surgery may be needed. Fixation with special partially threaded screws to achieve stability by crossing the physis but not closing it has been performed in a small series of patients with renal slips. In an adolescent with SCFE secondary to renal disease, epiphyseal closure with in situ fixation is the treatment of choice once the metabolic bone disease is being treated appropriately ( Fig. 42-16 ).




FIGURE 42-16


A and B, Valgus slipped capital femoral epiphysis in an 11-year-old child after renal transplantation and hypothyroidism. C and D, In situ fixation was performed.


Physiolysis has been described in other physes in children with renal osteodystrophy. Sites at which physiolysis has occurred include the distal femur, proximal humerus, and distal radius and ulna ( Fig. 42-17 ). Treatment consists of medical management of the metabolic bone disease and cast immobilization.




FIGURE 42-17


Osteodystrophy secondary to chronic renal insufficiency in a young girl. An anteroposterior radiograph shows slipping of the humeral head.


Avascular Necrosis.


Another orthopaedic complication seen in patients with renal failure is avascular necrosis, usually of the femoral head. It may be unilateral or bilateral. Prolonged steroid use, commonly needed after renal transplantation, is the probable cause of avascular necrosis in most children, although it has been seen in the hips of some children with renal failure who were not taking steroids. Treatment is symptomatic.




Parathyroid Disorders


Primary Hyperparathyroidism


Primary hyperparathyroidism results from hyperplasia or adenoma of the parathyroid glands, which leads to increased secretion of PTH. The increased PTH stimulates osteoclastic resorption of bone, which produces hypercalcemia. The initial symptoms of hyperparathyroidism are lethargy, bone pain, and abdominal complaints. The diagnosis is usually made late in the course of disease, when the child has abdominal symptoms, toxicity, and a hypercalcemic crisis. The symptoms of hyperparathyroidism are nonspecific, so the diagnosis is frequently missed at initial evaluation. Prolonged hypercalcemia leads to ectopic calcification in tissues and the formation of renal calculi. Abdominal pain and constipation result from the decreased abdominal motility. Hypertension is commonly present. In severe cases, the patient may become obtunded.


In approximately two thirds of the patients the cause is an adenoma, and in the remainder hyperplasia of the gland is usually found. Patients who have undergone head and neck irradiation are particularly susceptible to the development of parathyroid adenomas. Hyperparathyroidism can also be a component of the multiple endocrine neoplasia syndromes, which are inherited and can present rarely in childhood. There are also very rare genetic forms of hyperparathyroidism, some of which are self-limiting with medical treatment, whereas others are life-threatening. Infants born to parents with familial hypocalciuric hypercalcemia are at risk for the development of severe neonatal hyperparathyroidism (a rare autosomal recessive disorder) as a result of homozygous mutations in the calcium-sensing receptor gene. The condition presents within days after birth with life-threatening hypercalcemia, which requires emergent resection of the parathyroid glands.


The radiographic findings resemble those of renal osteodystrophy. Bone resorption is seen in the terminal tufts of the phalanges and in the clavicle. The bone appears osteopenic. Angular deformities resembling those seen in rickets can occur.


Laboratory evaluation generally reveals hypercalcemia, hypophosphatemia, and an elevated alkaline phosphatase concentration. Rarely, the calcium and phosphate concentrations are normal. The PTH level is elevated on direct assays.


Treatment is directed toward correcting the cause of the hyperparathyroidism. In cases of adenoma, tumor resection is performed. Adenomas are imaged with radionuclide scans, and it is not uncommon for multiple glands to be involved. Hypercalcemic crisis is treated by hydration, calciuresis, inhibition of bone calcium resorption, and treatment of the parathyroid abnormality.


Hypoparathyroidism


Hypoparathyroidism can result from failure of the parathyroid glands to synthesize or secrete PTH or from tissue resistance to PTH. Parathyroid gland destruction from iatrogenic causes—surgery or radiation—or an infiltrative process is more relevant to the adult population, whereas genetic causes of hypoparathyroidism affecting parathyroid gland development and function are more relevant to the pediatric population. Aplasia or dysplasia of parathyroid gland can result from mutations of the GCMB (glial cell missing) gene. Autosomal dominant and recessive forms exist. Because the GCMB gene is almost exclusively expressed in the parathyroid gland, its mutation causes isolated hypoparathyroidism without other phenotypes. Mutation of the X chromosome region q26-q27 is another cause of isolated hypoparathyroidism. This form is inherited as an X-linked recessive trait in males.


Reduced synthesis or secretion of PTH can result from CaSR mutations (chromosome 3q13.3-21). The mutation causes a decrease in PTH secretion despite low serum calcium levels because of activation of CaSR at a lower extracellular calcium set point. Rare mutations in the PTH gene can also reduce synthesis and secretion of PTH.


Hypoparathyroidism is also associated with DiGeorge syndrome, which is caused by microdeletions in chromosome 22q11. Thymic aplasia or hypoplasia with immunodeficiency, cleft palate, dysmorphic facies, and cardiac defects are other features of DiGeorge syndrome. De novo mutation is much more common than the autosomal dominant form of DiGeorge syndrome. Another autosomal dominant syndrome consists of hypoparathyroidism, sensorineural deafness, and renal dysplasia. Other autosomal recessive types are associated with growth retardation, seizures, and severe mental retardation.


Hypoparathyroidism should be distinguished from pseudohypoparathyroidism, in which the production of PTH is increased but the end organs cannot respond to the hormone. The PTH infusion test is useful for differentiating the two. When a test dose of PTH is administered to a patient with hypoparathyroidism, urinary excretion of phosphate and cyclic adenosine monophosphate (cAMP) increases appropriately and dramatically in contrast to a patient with pseudohypoparathyroidism, whose response to the PTH test is blunted because of target tissue resistance to PTH.


The initial symptoms of hypoparathyroidism are those of hypocalcemia—tetany caused by increased neuromuscular irritability, muscle cramps, facial or distal extremity paresthesias (Chvostek and Trousseau signs), and lethargy. The skin is dry and the hair is brittle and scanty. The teeth erupt late and fall out early. Cataracts may be present, and papilledema may occur. Mental retardation is seen in very young children.


Laboratory evaluation reveals low serum PTH and calcium levels, high serum phosphate levels, and low urinary calcium concentration because of hypocalcemia. Hypoproteinemia should be considered because the serum calcium concentration is normally decreased in patients with decreased albumin concentrations. The serum phosphorus concentration is elevated. Radiographs may be normal or may reveal increased radiopacity of the cortices of the long bones. Soft tissue calcification can occur, including calcification of the basal ganglia.


Standard treatment is calcium and vitamin D therapy. Nephrocalcinosis is a known complication of vitamin D therapy, however. Administrating PTH to treat hypoparathyroidism is being investigated and promising results have been reported in small studies with follow-up of up to 3 years. PTH therapy, however, is not yet considered standard treatment for hypoparathyroidism. Infants with hypoparathyroidism complicated by tetany may need calcium infusion. There is no orthopaedic treatment specific to the disease.


Pseudohypoparathyroidism


Pseudohypoparathyroidism is similar to hypoparathyroidism in its clinical and radiographic manifestations but differs in that PTH levels are elevated and it does not respond to the administration of exogenous PTH. The parathyroid glands are hyperplastic and secrete large amounts of the hormone, but the kidneys are resistant to PTH. Bone changes consistent with hyperparathyroidism occur because the skeleton responds to the elevated PTH level. Thus, findings include hypocalcemia and hyperphosphatemia resembling hypoparathyroidism, as well as osteitis fibrosa cystica resembling hyperparathyroidism. The skeletal changes seen in pseudohypoparathyroidism are also termed Albright osteodystrophy because Albright and associates were the first to describe the disease.


The cause is usually genetic. There are four subtypes of pseudohypoparathyroidism—Ia, Ib, Ic, and II. The molecular genetics of type Ia disease is caused by mutations in the maternally inherited guanine nucleotide–binding protein α-subunit gene (GNAS), which produces a lack of GNAS signaling in the proximal renal tubules; this in turn affects the adenylyl cyclase activity. Many mutations in the GNAS gene have been identified. Patients with type Ia disease have been found to have resistance to other hormones as well and suffer from multiple endocrinopathies, such as hypothyroidism and growth hormone deficiency. These patients can present with clinical features of Albright hereditary osteodystrophy, which include a round face, mental retardation, frontal bossing, short stature, obesity, brachydactyly, and ectopic ossification.


In type Ib pseudohypoparathyroidism, the typical features of Albright’s osteodystrophy are not seen, and hormone resistance appears to be limited to PTH and thyroid-stimulating hormone (TSH). Abnormal methylation of the maternal GNAS allele has been reported.


Type Ic is a variant of type Ia, with clinical features of Albright osteodystrophy and resistance to multiple hormones. However, no deficiency in GNAS activity is detectable in a standard in vitro assay because of the location of GNAS mutation. The clinical appearance of affected infants is normal, with osteodystrophic features gradually becoming apparent at 2 to 4 years of age. There is a characteristic shortening of the metacarpals, especially the first, fourth, and fifth, termed brachydactyly ( Fig. 42-18 ). When the hands are clenched into a fist, dimples are present at the sites of the knuckles of the fourth and fifth digits, thus giving rise to the mnemonic “knuckle, knuckle, dimple, dimple.” Multiple exostoses may be present, and the radius may be bowed. Patients are very short and often obese, and the facies has been described as moon-shaped. Heterotopic calcifications occur, especially in the periarticular tissues. Intracerebral calcifications have also been described. Sensorineural hearing loss is common.




FIGURE 42-18


Pseudohypoparathyroidism. An anteroposterior radiograph of the hands shows shortening (brachydactyly) of the first, fourth, and fifth metacarpals.


As noted, certain types of pseudohypoparathyroidism may be associated with hypothyroidism, Turner syndrome, and diabetes. Brachydactyly may also be seen in Turner syndrome and in myositis ossificans progressiva.


The diagnosis is assisted by administering PTH. A patient with pseudohypoparathyroidism is unable to respond to the exogenous hormone, so there will be no increase in serum calcium or urinary phosphate levels, and plasma cAMP (a product of adenylyl cyclase) will also not increase.


Treatment has been with vitamin D, but this has led to problems with nephrocalcinosis.




Vitamin Disorders


Hypervitaminosis D


Hypervitaminosis D is a result of the ingestion of excessive doses of vitamin D. Patients at risk are those who are taking vitamin D for the treatment of metabolic bone diseases such as vitamin D–resistant rickets and hypoparathyroidism. The elevated vitamin D level promotes the intestinal absorption of calcium and thereby leads to hypercalcemia. The optimal nutritional requirements for vitamin D in newborns and infants have been established.


Pathology


Histologically, wide osteoid seams are found around the trabeculae, similar to what is seen in rickets. The physis, however, is well calcified and normal in width and length. Metastatic calcification may be found in the kidneys, arteries, thyroid, pancreas, lungs, stomach, and brain. Deposition of calcium salts in the kidneys and degenerative changes in the arteries may produce significant morbidity.


Laboratory Findings


The hypercalcemia can be severe. The serum phosphate concentration is normal, with a diminished alkaline phosphatase concentration.


Clinical Features


Symptoms and signs of hypercalcemia are seen. Anorexia, constipation, nausea and vomiting, polyuria, thirst, and symptoms of dehydration are the early manifestations. The child feels very tired. With progression of the intoxication, mental depression and stupor develop. Renal failure and hypertension are common.


Radiographic Findings


Dense metaphyseal bands are seen in the long bones and result from an increase in the proximal zone of calcification. The diaphyses show osteopenia as a result of demineralization. Osteosclerosis is visible at the base of the skull, and there may be premature closure of the sutures. The vertebral end-plates are dense. Metastatic calcifications are seen in soft tissues ( Fig. 42-19 ).




FIGURE 42-19


Hypervitaminosis D in a 5-year-old boy who had taken 50,000 IU of vitamin D/day for the past 14 months. A, Lateral view of the skull showing metastatic calcification of the cerebral and cerebellar falces. B and C, Anteroposterior view of both hips and lower limbs. Note the increased radiopacity of the metaphyses.


Treatment


Treatment is medical and consists of the immediate cessation of vitamin D supplements. Diuretics are given, with replacement of volume with saline. Because dehydration can be fatal, serum electrolyte levels must be carefully monitored. Steroids inhibit calcium absorption in the kidney and gut and are helpful in correcting the calcium level. Bisphosphonates inhibit bone resorption and have been useful in treating vitamin D intoxication. Sodium phosphate should not be given because its administration leads to ectopic calcification.


Scurvy


Scurvy is caused by a nutritional deficiency of vitamin C, ascorbic acid. The disease is rare and is now most commonly seen in patients following extreme diets, such as patients with anorexia nervosa. Historically, scurvy was described in sailors whose diets lacked vitamin C during long sea voyages.


Pathology


When vitamin C is deficient, collagen synthesis is impaired. Vitamin C is necessary for the hydroxylation of lysine and proline to hydroxylysine and hydroxyproline, two amino acids crucial to the proper cross-linking of the triple helix of collagen. The result is primitive collagen formation throughout the body, including the blood vessels, which predisposes to hemorrhage.


Osteoblasts become dysfunctional, with failure to produce osteoid tissue and form new bone. Chondroblasts, however, continue to function normally, and mineralization is unaffected. This leads to the persistence of cartilage cells, and calcified chondroid approaches the metaphysis. Radiographically, an opaque white line termed Frankel’s line is seen at the junction of the physis and metaphysis.


Generalized osteoporosis results from lack of osteoid and new bone. Osteoclasts are normal, but osteoblasts become flattened, with a resemblance to connective tissue fibroblasts. The bone trabeculae and cortices of the long bones are thin and fragile. Hemorrhage and fractures are common, but the body’s attempt to repair these injuries is disorderly. The provisional zone of calcification is weak, which leads to epiphyseal separations. In the teeth, dentin formation is abnormal because of the defective collagen.


Clinical Features


Scurvy develops after 6 to 12 months of dietary deprivation of vitamin C, so it is not seen in neonates. Early manifestations consist of loss of appetite, irritability, and failure to thrive. Hemorrhage of the gums is common, and they become bluish and swollen. Subperiosteal hemorrhage is a distinctive sign that usually occurs in the distal femur and tibia and proximal humerus. The limbs become exquisitely tender, so much so that the infant screams on movement of the affected areas. The child lies still in the frog-leg position to minimize pain, a posture called pseudoparalysis. The limbs are swollen and bruised. Beading of the ribs at the costochondral junctions may occur. Hemorrhage may also develop in the soft tissues, including the joints, kidneys, and gut, and petechiae may be seen. The hair takes on a coiled appearance. Anemia and impaired wound healing are common. Severe hypertension has been described.


Radiographic Findings


The changes of scurvy are best seen at the knees, wrists, and proximal humeri ( Fig. 42-20 ). Osteopenia is the first change seen, with thinning of the cortices. The zone of provisional calcification increases in width and opacity (Frankel’s line) because of failure of resorption of the calcified cartilaginous matrix, and it stands out in comparison to the severely osteopenic metaphyses. The margins of the epiphyses appear relatively sclerotic, a finding termed ringing of the epiphyses, or Wimberger sign. Lateral spur formation at the ends of the metaphysis is produced by outward projection of the zone of provisional calcification. The scurvy line or scorbutic zone is a radiolucent transverse band adjacent to the dense provisional zone. The corner or angle sign of scurvy is a peripheral metaphyseal cleft caused by a defect in the spongiosa and cortex adjacent to the provisional zone of calcification. Epiphyseal separation may occur.




FIGURE 42-20


Scurvy in a 10-month-old infant. A, Anteroposterior radiograph of both lower limbs demonstrates early changes in the scorbutic bones. Note the generalized osteoporosis with rarefaction of the spongiosa and atrophy of the cortex. There is relatively increased opacity of the provisional zones of calcification at the ends of the metaphyses and around the margins of the epiphyseal centers of ossification (ringing of the epiphyses). B, Two weeks after treatment with ascorbic acid, marked calcification of subperiosteal hematoma of the right femur has occurred. Such minimal calcification is also evident in the medial aspects of the distal left femoral shaft and proximal left tibia. Note the multiple metaphyseal spur formation. C, Three months later there are further radiographic signs of healing scurvy. The cortices have become thicker and the spongiosa has almost normal density. Note the persistence of rarefaction in the epiphyseal centers.


Subperiosteal hemorrhage usually occurs at the femur, tibia, or humerus and is initially seen as an increase in soft tissue density. The hemorrhage becomes radiodense as the scurvy is treated and the lesions calcify. The development of a physeal bar in a patient with scurvy has been described.


Differential Diagnosis


The most common entity for which scurvy is mistaken is osteomyelitis. The symptoms of pain, tenderness, subperiosteal soft tissue swelling, and pseudoparalysis resemble the symptoms of infection. Because infection is common and scurvy is extremely rare, the condition can be misdiagnosed initially. The sedimentation rate, C-reactive protein level, and white blood cell count are normal in scurvy, however. Other diagnoses to be considered for this clinical picture include polio, leukemia, and purpuric conditions, such as Henoch-Schönlein purpura and thrombocytopenic purpura. Syphilis may be suspected but usually occurs earlier.


Serum levels of vitamin C may be difficult to interpret in scurvy. A more reliable test is the absence of vitamin C in the buffy coat of centrifuged blood.


Treatment


Treatment is administration of vitamin C. Rapid recovery is usual, with the pain and tenderness resolving. Scurvy is prevented by adequate intake of vitamin C, defined as 25 mg/day for infants, 30 to 40 mg/day for children and 40 to 75 mg/day for adults. Intoxication does not occur.


Hypervitaminosis A


Vitamin A is a fat-soluble vitamin whose primary biologic functions are concerned with skeletal growth, maintenance and regeneration of epithelial tissues, and preservation of visual purple in the retina. It is also necessary for membrane stability. The normal plasma level of vitamin A is 80 to 100 IU/100 mL. Hypervitaminosis A is very rare and usually results from inappropriate use of vitamin supplements. Retinoids used for acne also contain vitamin A and can lead to toxicity.


Clinical Features


Clinically, the soft tissues overlying the hyperostotic bones are swollen and tender. Proliferation of basal cells and hyperkeratinization cause dry, itchy skin. Anorexia, vomiting, and lethargy are caused by increased intracranial pressure. The child fails to thrive. Hepatomegaly with cirrhosis-like liver damage or splenomegaly may be present.


Radiographic Findings


The development of bone changes in patients with hypervitaminosis A is slow, so radiographs are normal initially. For this reason, radiographs are normal in children younger than 1 year. Once changes do occur, there is periosteal hyperostosis and thickening of the cortex of the long bones. The ulna, radius, metacarpals, and metatarsals are particularly affected. The mandible is spared, a fact that distinguishes hypervitaminosis A from Caffey disease. Subperiosteal new bone formation is seen ( Fig. 42-21 ). Bone scintigraphy shows increased uptake. Premature partial or complete physeal closure may be present.




FIGURE 42-21


Hypervitaminosis A in a 2-year-old child. Note the subperiosteal new bone formation and cortical thickening of both tibiae and both ulnae. The mandible and other facial bones are not affected. A and B, Radiographs of the right and left forearms. C, Radiograph of both lower limbs.


Diagnosis


The diagnosis is made by determining the plasma level of vitamin A, which will be elevated 5 to 15 times the normal value. Hypercalcemia can be present. Hypervitaminosis A must be differentiated from infantile cortical hyperostosis (Caffey disease), scurvy, and congenital syphilis.


Treatment


Treatment entails total cessation of administration of vitamin A and eliminating all foods containing vitamin A from the diet. Because of the large body reserves of vitamin A, the hyperostosis will disappear only after a long period, although the systemic symptoms resolve quickly. Growth of the long bones should be monitored because premature physeal closure may not become apparent for years after the initial insult.




Hypophosphatasia


Hypophosphatasia is a rare inheritable disorder cause by a deficiency of TNSALP. TNSALP is an enzyme that generates inorganic phosphate by hydrolyzing inorganic pyrophosphate. Inorganic phosphate is required for hydroxyapatite formation, whereas inorganic pyrophosphate suppresses the formation and growth of hydroxyapatite. There is wide variation in the severity of the disease, with prognosis related to the age at onset. A number of forms of hypophosphatasia exist—perinatal lethal, perinatal benign, infantile, childhood, adult, and odontohypophospatasia.


Inheritance


The gene for hypophosphatasia is the TNSALP gene ( TNSALP ), and many different mutations have been described within this gene. The transmission of lethal forms is autosomal recessive, whereas milder forms may have autosomal dominant or recessive transmission. Heterozygous carriers of hypophosphatasia can be detected by abnormally diminished alkaline phosphatase concentrations in plasma.


Pathology


The pathology seen in hypophosphatasia closely resembles that seen in patients with rickets. Osteoid production proceeds unharmed, but without alkaline phosphatase, mineralization of osteoid cannot occur. This leads to widening of the physis, with persistence of the provisional zone of calcification, which cannot calcify, and islands of cartilage continuing down into the metaphysis. The normal columnar arrangement of the chondrocytes of the growth plate is disturbed. If hypercalcemia is present, heterotopic calcification can occur, especially in the kidney.


Laboratory Findings


The hallmark of hypophosphatasia is a decrease or lack of alkaline phosphatase. The enzyme is decreased not only in serum but also in tissues such as the kidneys, bones, leukocytes, and spleen. Serum phosphorus, vitamin D, and PTH levels are normal, but hypercalcemia may be present, especially in young children. Characteristic findings in urine are elevated levels of phosphoethanolamine, which may be elevated in other endocrinopathies, and inorganic pyrophosphate. Pyridoxal-5′-phosphate levels are also increased in hypophosphatasia in relation to disease severity. Disease carriers have been found to have decreased serum alkaline phosphatase levels and increased urinary pyrophosphate levels.


Clinical Features and Radiographic Findings


Perinatal Hypophosphatasia


The clinical findings vary with the age at which the disease is manifested. In the perinatal lethal form, absence of skeletal mineralization is detected by prenatal ultrasound, and the infants may be stillborn. If they survive birth, they usually die from respiratory complications in early infancy because of hypoplastic lungs and chest wall deformities. A nonlethal, benign, prenatal form also exists and has been recognized. These patients have congenital limb bowing with a variable degree of hypomineralization, which improves postnatally, along with their limb bowing.


Radiographs of infants with the severe or lethal form of perinatal hypophosphatasia reveal diffuse, severe demineralization of the entire skeleton ( Fig. 42-22 ). Ossification of the skull is incomplete, and the suture lines are very wide. The ribs are unossified at the ends and slender in the middle. The pelvis is small, soft, and poorly mineralized. The vertebral bodies are paper thin and the neural arches cannot be seen. The long bones have jagged rarefied defects extending into the metaphysis.




FIGURE 42-22


Typical radiographic changes of hypophosphatasia in a 4-month-old infant. A and B, Both upper limbs. C, Lower limbs. D, Spine (see text for discussion). E, Skull. F, Femur, obtained at autopsy (see text for discussion).


Infantile Hypophosphatasia


In the infantile form, patients appear normal at birth. The onset of symptoms presents later in infancy, usually at approximately 6 months of age. Affected children fail to thrive and experience anorexia, vomiting, dehydration, fever, hypotonia, and sometimes seizures.


Demineralization of the bones occurs but is not as marked as in the perinatal form. The bones look rachitic, with widened physes, bossing of the skull, bowing of the ribs, and flaring of the metaphyses of the long bones and costochondral junctions. Lucent streaks in the metaphyses represent nests of unossified physeal cartilage. Fractures and bowing of the extremities are common. The cranial sutures are initially wide but close prematurely, thereby leading to increased intracranial pressure. Some patients show spontaneous improvement with time whereas others show progressive skeletal deterioration, with fractures and flail chest leading to pneumonia and lethal outcome. Hypercalcemia and hypercalciuria may cause renal calcinosis. Renal failure and hypertension then follow.


Childhood Hypophosphatasia


This form is most heterogeneous. Skeletal deformities, enlarged joints, delayed walking, waddling gait, short stature caused by disturbance of normal endochondral ossification, dolichocephaly, failure to thrive, bone pain, fractures, and intracranial hypertension may be observed. Dentition problem is common, with premature loss of the primary teeth. Spontaneous improvement has been observed.


Adult Hypophosphatasia


A rare adult form of hypophosphatasia usually presents during middle age. Clinically, the disease usually becomes apparent with nonhealing metatarsal stress fractures or thigh pain secondary to pseudofractures of the femur. Osteomalacia is present. Chondrocalcinosis and severe osteoarthropathy may develop.


Diagnosis


Prenatal Diagnosis


Hypophosphatasia can be diagnosed in fetuses. Ultrasound shows deficient ossification of the fetal skull. A definitive diagnosis can be established by amniocentesis and molecular genetic testing to search for mutations in TNSALP in at-risk infants.


Differential Diagnosis


Hypophosphatasia is usually confused with severe type II osteogenesis imperfecta because of the presence of birth fractures and the severe demineralization. Thanatophoric dwarfism and achondrogenesis can also resemble the perinatal form of hypophosphatasia.


Less severe forms of hypophosphatasia should be differentiated from the various types of rickets. In rickets, the alkaline phosphatase concentration is generally increased, whereas in hypophosphatasia, by definition it is decreased or not measurable.


Treatment


Although there is no curative treatment for hypophosphatasia, a limited number of drugs have been reported to be effective in managing certain problems associated with hypophosphatasia. Nonsteroidal antiinflammatory drugs (NSAIDs) have been shown to improve pain in childhood hypophosphatasia. Recombinant human PTH 1-34 can improve and resolve metatarsal stress fractures in adult hypophosphatasia. A case report of bone marrow cell transplantation in a patient with severe infantile hypophosphatasia showing clinical and radiographic improvements has been reported. In another patient with infantile hypophosphatasia, successful transplantation of bone fragments and cultured osteoblasts, with improvement in the clinical severity of the disease, has also been reported. If the diagnosis of rickets is mistakenly made, treatment with vitamin D can worsen the heterotopic calcification and nephrocalcinosis. Enzyme replacement therapy is not yet available.


Fractures require orthopaedic referral. Healing of fractures is generally delayed in patients with hypophosphatasia. Occasionally, multiple osteotomies with intramedullary fixation, as would be done in cases of severe osteogenesis imperfecta, are needed to correct the bowing and lend structural support to the long bones.




Idiopathic Hyperphosphatasia


Idiopathic hyperphosphatasia is an extremely rare bone dysplasia characterized by Paget disease–like features, which include an increased bone turnover, with failure to replace immature woven bone with mature lamellar bone, skeletal deformity, bone expansion, and increased risk of pathologic fractures. It also known as juvenile Paget disease of bone or familial hyperphosphatasia. Biochemically, serum levels of alkaline phosphatase are increased—hence, the name hyperphosphatasia—as is urinary excretion of hydroxyproline. The disease is transmitted as an autosomal recessive trait, and mutations in the TNFRSFIIB gene, which encodes osteoprotegerin (OPG), have been identified. Depending on the location of the mutation, the severity of the disease can be mild to severe.


Clinically, the long bones are bowed and prone to stress fractures because of osteopenia and decreased biomechanical strength of the bone. Kyphosis and acetabular protrusion may also develop. The patient’s skull is enlarged and progressive sensorineural deafness occurs. Initial complaints are painful swelling of the limbs and bowing. Muscle mass appears diminished, and the limbs may be warm. Affected children are very short.


Radiographic findings include generalized diaphyseal expansion of the bones, with subperiosteal new bone deposition. Fractures are transverse and usually nondisplaced. The spine and pelvis show patchy areas of sclerosis. The base and vault of the skull are thickened.


Pathologic studies of bone tissue show extensive fibrosis of the marrow with cellular hyperactivity. There is evidence of increased bone resorption and bone formation resembling fibrous dysplasia. There may be a mosaic pattern of cement lines resembling what is seen in Paget disease.


Conditions from which hyperphosphatasia must be distinguished include Camurati-Engelmann disease, craniodiaphyseal dysplasia, and fibrous dysplasia. Hyperphosphatasia can be differentiated from all these conditions by the distinct elevation in the serum alkaline phosphatase level.


Treatment consists of antiresorptive drugs such as calcitonin and bisphosphonates. Successful treatment of the disease has been reported in case reports and case series with the use of pamidronate and ibandronate.




Growth Hormone Deficiency


Isolated growth hormone deficiency can have a congenital or acquired cause. However, the cause is unknown (idiopathic) in most cases. The hereditary or familial form of growth hormone deficiency has four types, which are based on the inheritance pattern and phenotype—autosomal recessive (types IA and IB), autosomal dominant (type II), and X-linked (type III). Various mutations in the genes for growth hormone ( GH1 ), receptor of growth hormone–releasing factor (GHRHR), and transcription factor SOX3 have been reported in the cases of familial isolated growth hormone deficiency. The GH1 mutation is the most common. In the vast majority of the familial cases, however, a mutation is not detected, suggesting that unknown genetic factors may also be involved in the pathogenesis. In type IA deficiency, various GH1 mutations (deletions, frameshift, or nonsense mutation) lead to a severely truncated or absent growth hormone, and serum growth hormone is undetectable. In type IB deficiency, mutations in the gene for GH1 or GHRHR have been described. In contrast to type IA, serum growth hormone level is low but detectable in type IB. In type II deficiency, GH1 mutations are also observed; however, the locations of the mutations mainly affect the splicing of mRNA for GH1, producing a smaller isoform of growth hormone. This has an altered protein structure that affects the secretion of growth hormone and other hormones and the proliferation and survival of the somatotrophic cells of the anterior pituitary. Thus, the patients with type II deficiency may have a hypoplastic anterior pituitary on MRI and may develop clinical features of other pituitary hormone deficiencies. Type III deficiency has an X-linked inheritance; affected males have an absent or severe deficiency of gamma globulins. Mutations in the transcription factor SOX3 and the gene for Bruton tyrosine kinase have been implicated with this condition. SOX3 plays an important role in the development of the pituitary gland, craniofacial structures, and central nervous system (CNS). Thus, type III deficiency may be associated with mental retardation, craniofacial dysmorphism, panhypopituitarism, anterior pituitary hypoplasia, and posterior pituitary abnormalities.


Clinical Features


In congenital forms the infant is of normal size, but diminished growth is noted within the first 6 months in type IA deficiency. The limbs are of normal proportion in relation to the head and trunk. Intelligence is normal in most cases. The phenotypic feature of type IB deficiency is less severe in terms of growth disturbance than type IA. The condition can be associated with hypogonadism and a delay in or absence of sexual maturation.


In the acquired form caused by a pituitary lesion, signs of neurologic deficit, such as impaired vision, ocular disturbances, and pathologic sleepiness, are present.


Radiographic Findings


In congenital hypopituitarism, skeletal maturation is delayed. The ossification centers are late in both appearance and closure. Osteoporosis of the long bones and the skull is present. The fontanelles close later than normal.


Where there is a lesion in the pituitary, radiographs reveal enlargement of the sella turcica, the home of the pituitary. Intrasellar or suprasellar calcification suggests craniopharyngioma. MRI is especially useful for viewing the pituitary. Enlargement, hypoplasia, or tumor can be seen.


Diagnosis


Serum levels of growth hormone will be low or absent. Because low levels are normal in healthy children, a stimulatory test is usually needed to confirm the lack of growth hormone. Insulin or l -arginine is administered to produce hypoglycemia, which stimulates the release of growth hormone. Growth hormone levels do not increase in patients with pituitary dwarfism after the administration of these agents.


Treatment


Pituitary dwarfism is treated by the administration of synthetic growth hormone. This treatment stimulates growth and should be monitored by a pediatric endocrinologist. Patients with growth hormone deficiency after resection of craniopharyngiomas rarely have an isolated deficiency in growth hormone, so additional hormone replacement therapy is necessary, under the guidance of the endocrinologist. In some children with growth hormone deficiency, panhypopituitarism has developed in adulthood, with hypothyroidism and abnormalities in antidiuretic hormone. On rare occasion, referral to an orthopaedic surgeon is needed for the treatment of SCFE.




Hypothyroidism


Thyroid hormone deficiency may be congenital or acquired. The degree of deficiency, age at onset, and duration of the deficiency are all factors that determine the severity of disease. Hypothyroidism is fairly common, with an incidence of 1/4000 newborns.


Congenital hypothyroidism, previously known as cretinism, is the most common endocrine disorder found in neonates. It leads to dwarfism and mental retardation if treatment is delayed for more than 3 months postnatally. Neonatal screening for congenital hypothyroidism has allowed early diagnosis and treatment, with prevention of mental retardation. It is more common in girls than in boys.


Causes


Congenital hypothyroidism can be caused by prenatal developmental defects of the thyroid gland producing a structural abnormality (thyroid agenesis or dysgenesis) or by defects in thyroid hormone biosynthesis (thyroid dyshormonogenesis). The former, structural causes of congenital hypothyroidism, are found in most cases (80%). Structural abnormalities range from aplasia of the thyroid, hypoplasia, and goiter to ectopic thyroid tissue. Defects in thyroid hormone synthesis are responsible for the remaining 20% of cases.


Clinical Features


Symptoms in early infancy include prolonged jaundice, lethargy, sleepiness, feeding difficulties, and constipation. Frequently these infants are overweight. Other features include dry skin, scanty coarse hair, an enlarged tongue, umbilical hernias, and an expressionless face ( Fig. 42-23 ). Developmental delay is noted. Associated congenital malformations, especially heart defects, are more likely to occur in children with congenital hypothyroidism.




FIGURE 42-23


Typical clinical appearance of congenital hypothyroidism.


In acquired hypothyroidism, which is manifested later in childhood, sluggishness, a slowdown in growth, and worsening school performance are noted. SCFE may occur and lead to groin, hip, or knee pain. Hypogonadism is present, and the children are often overweight.


Radiographic Findings


Thyroid hormone is very important in regulating bone growth and maturation. In patients with hypothyroidism, enchondral bone formation is disturbed. The skeleton is immature for the infant’s chronologic age. Appearance of the epiphyses is delayed, and they appear irregular and fragmented, which in childhood resembles what is seen in Perthes disease or multiple epiphyseal dysplasia. Epiphyseal dysgenesis was the term used by Reilly and Smyth to describe the ossific nuclei. The physis may be irregular and widened, similar to the radiographic picture in rickets.


The bone age of the patient is delayed. The long bones are abnormally widened as a result of normal intramembranous bone formation in the context of disturbed endochondral ossification.


The head appears large. Radiographically, ossification of the skull is retarded and the base of the skull is shortened. The sella turcica may be enlarged. Closure of the fontanelles is delayed. A delay in the development of normal dentition is common. Spinal radiographs show a tendency toward thoracolumbar kyphosis. The vertebral body of L2 is wedge-shaped and the anterior margin is beaked. L1 and L3 may have a similar appearance. The bony end-plates are convex. Osteosclerosis develops in some patients as a result of hypercalcemia. Radiographs in these patients show transverse radiopaque bands in the metaphyseal areas and thickened cortices. Metastatic calcification can occur. MRI has shown enlargement of the pituitary in children with congenital hypothyroidism.


Diagnosis


Great advances have been made in the diagnosis of congenital hypothyroidism. Screening programs are in place to measure TSH levels in the newborn nursery. An elevated TSH level is suggestive of congenital hypothyroidism. Further laboratory evaluation of thyroid hormone levels and further imaging studies consisting of radionuclide scintigraphy of the thyroid or thyroid ultrasound are then performed to ascertain the cause of the hormone deficiency.


Early diagnosis is mandatory because a delay in diagnosis can lead to irreversible mental retardation. The workup of a developmentally delayed child should include laboratory evaluation of thyroid function when the cause of the delay is unknown.


There is an association between Down syndrome and hypothyroidism. Studies have shown that 15% to 35% of infants with Down syndrome have congenital hypothyroidism, and annual screening of these children is recommended. Hypothyroidism should be especially considered in children with Down syndrome who have SCFE. In one study, six of eight patients with Down syndrome and slipped epiphyses had hypothyroidism.


Other laboratory findings in children with hypothyroidism may include high serum calcium levels. Patients with panhypopituitarism will have not only hypothyroidism but also the other hormonal deficiencies seen in this disorder, such as growth hormone deficiency.


Treatment


Treatment begins immediately on diagnosis. Hormone replacement therapy with thyroxine is initiated and carefully monitored. If treatment is begun by 24 months of age, subsequent growth has been shown to be normal by 5 years. With hormonal replacement therapy, the pubertal growth spurt is normal, and adult height is within normal limits. Long-term thyroxine replacement therapy has not been shown to decrease bone mass and lead to osteopenia. Prompt treatment results in normal intellectual development.


Prenatal diagnosis through cord blood sampling has been achieved, and prenatal treatment of hypothyroidism by means of thyroid hormone injected into amniotic fluid has been successful in experimental settings. If congenital hypothyroidism remains untreated, the mental retardation is progressive, and most children die early of respiratory infection.


Orthopaedic Considerations


In an older child, SCFE may be the first manifestation of hypothyroidism ( Fig. 42-24 ). Screening recommendations range from screening all patients with SCFE for thyroid disease to no routine screening whatsoever. We believe that any patient with SCFE who is younger than usual (<11 years), who has a family history of thyroid abnormalities, or who does not have the typical obese body habitus should be screened for hypothyroidism with a TSH test. Patients with Down syndrome and slips should be considered to have hypothyroidism until proven otherwise. In patients with SCFE secondary to hypothyroidism, contralateral prophylactic pinning should be performed because the incidence of bilateral SCFE in hypothyroidism is 61%.




FIGURE 42-24


A and B, Right slipped capital femoral epiphysis in a 13-year-old girl. Hypothyroidism was diagnosed on initial evaluation. Physeal widening is seen in the asymptomatic left hip. C and D, In situ fixation was performed bilaterally. E and F, At 2-year follow-up, the physes were healed.




Idiopathic Juvenile Osteoporosis


Idiopathic juvenile osteoporosis is a rare metabolic bone disease of childhood characterized by a profound reduction in bone mass of unknown cause. The cardinal features of idiopathic juvenile osteoporosis are as folllows: (1) onset before puberty; (2) compression fractures of the vertebrae and long bones; (3) formation of new but osteoporotic bone; and (4) spontaneous recovery after skeletal maturity.


Causes


The cause of idiopathic juvenile osteoporosis remains unknown. The disease is not genetically transmitted. The basic mechanism of disease is decreased bone remodeling activity and decreased bone formation. Bone histology generally shows a decreased cancellous bone volume and a very low bone formation rate on cancellous surfaces. Bone remodeling activity is less involved in the cortical bone surfaces than cancellous bone surfaces. In one study, secretion of synthesized collagen by cultured skin fibroblasts in some patients with idiopathic juvenile osteoporosis was reduced, whereas the range of collagen secretion in other patients with the disease overlapped the normal range. Another study found diminished levels of the carboxy-terminal propeptide of type I procollagen in patients with juvenile osteoporosis, again indicating abnormalities in collagen metabolism.


Biomechanical studies are conflicting. Serum calcium and phosphorus levels are normal in these patients. Calcium balance, however, is negative, with poor gastrointestinal absorption of calcium. Alkaline phosphatase and urinary hydroxyproline levels are usually normal as well. Although most studies reported normal vitamin D levels, two studies found low levels of calcitriol (1,25-dihydroxycholecalciferol). Therapy was then directed toward the vitamin deficiency, with improvement in the disease. It may be that different forms of the disease have different biochemical profiles.


Clinical Features


The mean age at onset is 7 years, with cases reported in children as young as 1 year. By definition, the disease is always manifested before puberty. There is no gender predilection.


The initial complaints in children with idiopathic juvenile osteoporosis are back and leg pain. Patients may refuse to walk or may have a slow gait or limp. The examining physician should always remember that a limp in children is a common orthopaedic problem, whereas idiopathic juvenile osteoporosis is an extremely rare condition.


Radiographic Findings


Diffuse generalized osteoporosis is seen on radiographs of the spine and limbs ( Fig. 42-25 ). The normal trabecular pattern is markedly decreased and the cortices of the bones are thinned. On lateral radiographs of the spine, a codfish appearance is seen. Increased thoracic or thoracolumbar kyphosis with anterior wedging of the vertebrae may develop. Vertebral compression fractures may be evident, and scoliosis may be present.




FIGURE 42-25


Idiopathic juvenile osteoporosis. Anteroposterior ( A ) and lateral ( B ) radiographs of the spine showing the severe osteoporosis. Note the compression fractures of the vertebrae in the lumbar region. C, AP radiograph of both tibiae. Note the plastic bowing of the fibulae and marked osteoporosis.


Another radiographic feature is the presence of long bone fractures in various stages of healing. The fractures are usually metaphyseal and tend to occur in areas of highest stress, such as the femoral neck. Other areas in which stress fractures are common are the distal femur and proximal tibia. The skull does not have a wormian appearance.


Diagnosis


The diagnosis is one of exclusion. The various known causes of osteoporosis in childhood are listed in Box 42-1 . The most difficult distinction to make is between idiopathic juvenile osteoporosis and mild osteogenesis imperfecta. Patients with a positive family history have osteogenesis imperfecta, yet individuals with no affected relatives may have either disease. Other distinguishing features of osteogenesis imperfecta that are not associated with idiopathic juvenile osteoporosis are blue sclerae, dentinogenesis imperfecta, ligamentous laxity, and easy bruising. Patients with juvenile osteoporosis do not sustain fractures in early infancy, which may help differentiate the two diseases in some patients. Finally, the fracture callus in juvenile osteoporosis is osteopenic.



Box 42-1

Causes of Osteoporosis in Childhood


Endocrine Disorders





  • Hyperthyroidism



  • Hyperparathyroidism



  • Hypogonadism



  • Glucocorticoid excess—Cushing syndrome, steroid therapy



Metabolic Disorders





  • Homocystinuria



  • Gastrointestinal malabsorption



  • Idiopathic hypoproteinemia



  • Vitamin C deficiency



  • Rickets of any cause



Liver disease


Renal Disease





  • Chronic tubular acidosis



  • Idiopathic hypercalciuria



  • Lowe syndrome



  • Uremia and regular hemodialysis



Bone Diseases





  • Osteogenesis imperfecta



  • Idiopathic juvenile osteoporosis



  • Idiopathic osteolysis



  • Turner syndrome (XO chromosome anomaly)



Malignant Diseases





  • Leukemia



  • Lymphoma



Miscellaneous Causes





  • Disuse osteoporosis of paralyzed limbs as in myelomeningocele



  • Generalized osteoporosis of Still disease, especially after steroid therapy



  • Heparin therapy



  • Anticonvulsant drug therapy




Fibroblast studies may be of some help in establishing the diagnosis of osteogenesis imperfecta, but overlap of results with normal ranges and with results in idiopathic juvenile osteoporosis may occur in some children. Bone biopsy is not generally necessary to diagnose osteogenesis imperfecta or idiopathic juvenile osteoporosis but, when it is performed, increased woven immature bone is seen in osteogenesis imperfecta, whereas increased osteoclastic resorption of bone is seen in idiopathic juvenile osteoporosis.


Another very important clinical distinction to make is that between leukemia and idiopathic juvenile osteoporosis. A child with leukemia may have osteopenia and compression fractures, so urgent referral to a pediatric hematologist is wise in the evaluation of a patient with osteoporosis. Usually, a bone marrow aspirate will be required to rule out leukemia definitively.


Treatment


Treatment of idiopathic juvenile osteoporosis is controversial. Isolated reports of successful medical treatment with calcitonin, calcitriol, bisphosphonates, and estrogen have been published. All reported improved bone mineralization and decreased fractures. It appears that when a demonstrable deficiency is found through laboratory testing, treatment aimed toward correcting that deficiency is warranted.


Orthopaedic treatment of the spine is usually conservative. Bracing may relieve back pain and treat the kyphotic deformity. The Milwaukee brace has been used for this purpose, with reported success. The role of the brace in accelerating osteoporosis by stress shielding the spine is unknown. Use of the brace should be discontinued gradually as the osteoporosis resolves. Similarly, scoliosis should be managed orthotically when possible. Spinal fusion has been performed in isolated cases, but continued progression of the deformity because of bending of the fusion mass has been described.


Long bone fractures should be managed by conventional means. Immobilization should be kept to a minimum because prolonged immobilization leads to worsening osteoporosis and may result in a cycle of fractures.




Osteogenesis Imperfecta


Osteogenesis imperfecta is a genetic disorder of connective tissue with the clinical trademark of bone fragility, as evidenced by long bone fractures. Other major clinical features may include skeletal deformity, blue sclerae, hearing loss, and fragile opalescent teeth (dentinogenesis imperfecta). In adults, valvular insufficiency and aortic root dilation can occur because of the collagen pathology. Less severe manifestations may include generalized ligamentous laxity, hernias, easy bruisability, and excessive sweating. The spectrum of age at presentation, severity of skeletal manifestations, and natural course of the disorder is very broad.



References .

The severity of the bone fragility is the best example of this wide spectrum; fragility can be so severe that the affected infant is born with crumpled ribs, a fragile cranium, and long bone fractures incompatible with life, whereas at the opposite end of the spectrum, an older child who otherwise appears normal may sustain only a few fractures after a reasonable amount of trauma.


The distinction between child abuse (nonaccidental injury) and excessive bone fragility may be difficult to make in these latter circumstances. It is now known that at least 90% of affected individuals have an identifiable genetic defect producing quantitative or qualitative abnormalities in type I collagen (or both types of defect).



References .

Type I collagen is the major structural protein found in bone and connective tissue of dentin, sclera, skin, ear bones, vessels, and heart valves. The disorder may be inherited from a parent in an autosomal dominant fashion, may occur as a spontaneous mutation or, rarely, may be inherited as a homozygous autosomal recessive trait from both parents. The autosomal dominant forms are caused by a quantitative or qualitative type I collagen defect, whereas autosomal recessive forms are caused by noncollagenous proteins that interact with type I collagen during posttranslational modification or during the folding of the triple helix.


Pathophysiology


The vast majority (at least 90%) of individuals with osteogenesis imperfecta have an identifiable genetic defect in one of the two chains that form the type I collagen. §



§ References .

Some understanding of normal collagen formation and errors in the metabolic process that are seen in osteogenesis imperfecta is essential for understanding the pathophysiology and variability of the disorder.


Normal Type I Collagen Metabolism


Type I collagen is the major structural collagen of the bone, skin, tendons, dentin, and sclera. It is a triple-helix molecule made of two α 1 chains and one α 2 chain. In a normal fibroblast, precursor subunits for these two types of strands (pro-α 1 [I] and pro-α 2 [I] polypeptide chains) are synthesized in the rough endoplasmic reticulum. These two procollagen polypeptide chains are encoded by two separate genes, COL1A1 (encoding for pro-α 1 [I]), located on the long arm of chromosome 17, and COL1A2 (encoding for pro-α 2 [I]), located on the long arm of chromosome 7. Combining these three chains into the triple helix begins at the carboxy-terminal end and propagates toward the amino-terminal end. The process of alignment and assembly of the triple helix is supported by endoplasmic reticulum–resident molecular chaperones such as GRP78, Serpin H1, and the prolyl 3-hydroxylation complex, which consists of CRTAP, P3H1, and PPIB. An essential feature of the pro-α chains required for proper folding of the triple helix is a recurrent pattern of glycine residues at every third peptide position in the chain (Glycine-X-Y sequence). It is at these residues that cross-linking of the three chains occurs. The type I procollagen molecules are secreted from the cell and are processed extracellularly to form the type I collagen molecules ( Fig. 42-26, A ).




FIGURE 42-26


Schematic representation of normal and abnormal collagen formation. A, Normal type I collagen formation. Two pro-α 1 (I) (encoded by COL1AI on chromosome 17) and one pro-α 2 (I) (encoded by COL1A2 on chromosome 7) polypeptide chains form a left-handed triple helix, beginning at the carboxy end and continuing to the amino end. Cross-linking occurs at glycine residues located at every third position in the chains. The procollagen molecule is then secreted from the endoplasmic reticulum into the extracellular matrix, where coalescence into the complete type I collagen fiber continues. B, Quantitative defect typified by Sillence type IA osteogenesis imperfecta. There is a stop codon for one of the COL1AI genes, which results in no mRNA from that gene. As a result, normal pro-α 1 polypeptide chains are produced in levels approximately 50% of normal, with the subsequent production of ≈50% of the normal amount of type I collagen. The collagen produced is electrophoretically normal, and no abnormal collagen is detectable. C, Formation of mutant type I collagen from some defect in COL1AI or COL1A2 . Skips or substitutions for glycine occur at some point along the polypeptide chains encoding for pro-α 1 or pro-α 2 . The mutant polypeptide chain results in poorer cross-linking. Defects closer to the carboxy terminal are potentially more serious because triple helix formation begins at this end. The mutant procollagen is usually produced in reduced amounts, so there is a qualitative and quantitative deficiency of type I collagen. This type of defect is typical of Sillence types II, III, and IV osteogenesis imperfecta.


Collagen Metabolism in Osteogenesis Imperfecta


In almost 90% of patients with osteogenesis imperfecta, the genetic defect is found in type I collagen α-chain genes. The mutations can produce a qualitative or quantitative abnormality of type I collagen formation. Type I collagen can be assayed by performing gel electrophoresis of samples from cultured dermal fibroblasts. The assay may show a quantitative decrease in the amount of structurally normal type I collagen because of a premature stop codon in the affected allele (one copy of the gene involved) or frameshift mutations. In this scenario, a patient who is heterozygous for the condition will secrete approximately half the normal amount of type I collagen (haploinsufficiency), with no abnormal type I collagen identifiable (see Fig. 42-26, B ). This is the type of defect most commonly identified in type IA osteogenesis imperfecta in Sillence’s classification (see later, “ Classification and Heredity ”). Rare types, with normal type I collagen but more severe reductions in quantity than the typical type I collagen (≤20%), have been identified.


Alternatively, there can be an error in substitution or deletion, usually involving a glycine peptide residue somewhere along the polypeptide chain. In such a case, the affected patient will produce a structurally or qualitatively abnormal, less effectual collagen, generally in reduced amounts. The severity of the disruption of function caused by the structural abnormality of the collagen is in part related to the location of the glycine residue error. Substitutions at the carboxy end of the polypeptide chains are potentially more serious because cross-linking of the triple helix begins at the carboxy terminal of the chains. In general, patients with mutations that affect glycine residues and the quality of a collagen α chain have more severe skeletal involvement than patients with haploinsufficiency mutations. This type of defect, which impairs the function of type I collagen, is the more commonly identified defect in Sillence’s types II, III, and IV (see later, also see Fig. 42-26, C ). Patients with the most severe or lethal varieties tend to have the coding defect at the carboxy end of the pro-α 1 (I) or pro-α 2 (I) chains.


In addition to type I collagen mutations affecting its quantity or quality, other gene mutations, which produce recessive osteogenesis imperfecta (types VI, VII, VIII, IX, X, XI), have been identified. These include genes that encode the components of the collagen 3-hydroxylation complex, which play a role in the assembly of the triple helix. Collectively, the recessive form probably accounts for less than 5% of cases of osteogenesis imperfecta.


Classification and Heredity


The identification of more than 280 specific locations of disruptions in genetic coding for type I collagen has improved our understanding of the nature and variability of the clinical manifestations but has not simplified the classification process. Variability in the natural history, time of onset, different patterns of inheritance, relatively high incidence of spontaneous mutations, and variability in clinical severity, even when the mode of inheritance is known, further compromise the effectiveness of classification schemes. Two classification schemes, those of Sillence and associates and Shapiro, are clinically useful despite their limitations.


In 1979, Sillence and Danks delineated four distinct types of osteogenesis imperfecta based on clinical and genetic characteristics ( Table 42-2 ). In their original description, four types were described and identified as autosomal dominant (types I and IV) or autosomal recessive (types II and III). More recent work on the nature of type I collagen disorders and the molecular genetic basis for these disorders has elucidated the nature of the genetic defect, and true autosomal recessive transmission is rare in this condition. Cole has recommended modification of the original Sillence classification based on an extensive review of the collagen defect in 200 patients with osteogenesis imperfecta. Discoveries of noncollagen gene defects causing osteogenesis imperfecta and histologic features dissimilar to those of the conventional types have led to the addition of types V to XI to osteogenesis imperfecta nosology. Type V has autosomal dominant transmission, whereas types VI to XI have recessive transmission. It is important to note that these latter types collectively account for approximately 5% of cases of osteogenesis imperfecta.



Table 42-2

Classification of Osteogenesis Imperfecta Syndromes






































































































Type Inheritance Teeth Bone Fragility Deformity of Long Bones Growth Retardation Presenile Hearing Loss (%) Prognosis Sclerae Spine Skull Other Incidence
IA Autosomal dominant Normal Variable, less severe than other types Moderate Short stature, 2%-3% below mean 40 Fair Distinctly blue throughout life Scoliosis and kyphosis in 20% Wormian bones on radiographs Premature arcus senilis 1/30,000
IB Autosomal dominant Dentinogenesis imperfecta Variable, less severe than other types Moderate Short, 2%-3% below mean 40 Fair Distinctly blue throughout life Scoliosis and kyphosis in 20% Wormian bones on radiographs Premature arcus senilis 1/30,000
II Autosomal recessive Unknown (because of perinatal death) Very extreme Crumbled bone (accordion femora) marked Unknown (because of perinatal death) Perinatal death Blue Marked absence of ossification 1/62,000 live births
III Autosomal recessive Dentinogenesis imperfecta Severe Progressive bowing of the long bones and spine Severe, smallest of all patients with osteogenesis imperfecta Nonambulatory, wheelchair- bound; may die in third decade Bluish at birth, become less blue with age, white in adult Kyphoscoliosis Hypoplastic, more ossified than in type II, wormian bones Very rare
IVA Autosomal dominant Normal Moderate Moderate Short stature Low frequency Fair Normal Kyphoscoliosis Hypoplastic, wormian bones Unknown
IVB Autosomal dominant Dentinogenesis imperfecta Moderate Moderate Short stature Low frequency Fair Normal Kyphoscoliosis Hypoplastic, wormian bones Unknown

Adapted from Sillence DO, Danks DM: The differentiation of genetically distinct varieties of osteogenesis imperfecta in the newborn period, Clin Res 26:178, 1978.


Osteogenesis Imperfecta Type I


Osteogenesis imperfecta type I is characterized by generalized osteoporosis, with abnormal bony fragility, distinct blue sclerae throughout life, and presenile conductive hearing loss. This is the most common type of osteogenesis imperfecta in most series, and type I patients are, in general, the least affected in terms of the incidence of fractures and bone deformity. Fractures generally occur during the ambulatory stage of child development and decrease after puberty. This type is inherited as an autosomal dominant condition, although spontaneous mutations occur. Molecular genetic studies have revealed that this type is characterized by a quantitative defect in type I collagen.



References .

Specifically, one of the inherited COL1A1 genes in affected patients will not produce effective mRNA for pro-α 1 collagen, so the amount of type I collagen is effectively reduced to approximately 50% of the normal amount. However, that 50% is structurally normal, and no mutant type I collagen is detectable by electrophoretic techniques. Dentinogenesis imperfecta is present in some of these patients. Those without dentinogenesis imperfecta are subclassified as having osteogenesis imperfecta type IA, and those with dentinogenesis imperfecta are classified as having type IB.


Osteogenesis Imperfecta Type II


Type II osteogenesis imperfecta is characterized by extreme bone fragility. Type II patients are almost invariably stillborn or die shortly after birth. The long bones are crumbled (accordion femora) from in utero fractures, and ossification of the skull is markedly delayed; on palpation, the cranial vault feels like numerous small plates of bone. Originally, this condition was thought to be inherited as an autosomal recessive trait. However, work on the nature of type I collagen disturbance has revealed that the defect in most cases is a severe disruption in the qualitative function of type I collagen.



References .

In most cases the condition is inherited as a dominant negative condition, often as the result of a spontaneous mutation; many different mutations have been described.


This pattern of inheritance is more in keeping with the risk of recurrence in subsequent pregnancies of couples with a previously affected fetus. If the condition were inherited as an autosomal recessive trait, the risk in subsequent pregnancies should be approximately 25% or, if caused by a spontaneous mutation, essentially 0%. The risk of having a subsequent affected fetus has been estimated at approximately 7%. In these situations, one of the parents was identified as being mosaic for the dominant negative gene, which accounts for the low but present risk. For a discussion of diagnostic evaluations for identifying an affected fetus, see later (“ Prognostication and Parental Counseling : Antenatal Diagnosis ”).


Osteogenesis Imperfecta Type III


This variety of osteogenesis imperfecta is characterized by severe bone fragility, multiple fractures and progressive marked deformity of the long bones, and severe growth retardation. Types III and IV patients are more severely affected than most type I patients and constitute most patients with severe deformity and frequent fractures; they may require extremity intramedullary rodding and have more difficulty ambulating or are unable to do so. Type III may have qualitative and quantitative changes in type I collagen and may be inherited as an autosomal recessive or dominant negative trait. These patients typically have triangular facies, frontal bossing, extremely short stature, and vertebral compression. They may also have basilar invagination and scoliosis. A higher prevalence of coxa vara is seen in this type.


The sclerae are bluish at birth but become less blue with age. In adolescents, the sclerae are of normal hue. The most severely affected surviving patients often have this type of disease.


Osteogenesis Imperfecta Type IV


Osteogenesis imperfecta type IV has heterogeneous severity that overlaps with types I and III. It is from this heterogeneous group that patients with type V, VI, or VII osteogenesis imperfecta have been identified over the past 2 decades. Type IV osteogenesis imperfecta is inherited as an autosomal dominant condition, and most patients, like those with types II and III, have qualitative and quantitative changes in type I collagen. At birth the sclerae are of normal hue; if they are bluish, they become progressively less so with maturation and are normal in adolescence. The osteoporosis, bone fragility, and long bone deformities are of variable severity. Dentinogenesis imperfecta also occurs in some affected individuals; those with normal dentition are classified as having type IVA disease and those with dentinogenesis imperfecta as having type IVB disease.


Osteogenesis Imperfecta Type V


A mutation in the 5′-untranslated region of a gene encoding interferon-induced transmembrane protein 5 (IFITM5) has been shown to cause type V disease. Hypertrophic callus after fracture, a radiodense band adjacent to the physis of long bone, and calcification of the interosseous membrane between the radius and ulna are the three key features of this type. A key histologic feature is the meshlike appearance of the lamellar bone. Patients have white sclera, absence of dentinogenesis imperfecta, and radial head dislocation. The severity of the disease in terms of bone deformation ranges from mild to moderate.


Osteogenesis Imperfecta Type VI


Type VI is distinguished from other types by its distinct histologic feature seen on bone biopsy samples. The lamellar bone has a fish scale pattern instead of a layered pattern when examined under polarized light microscope, and features of severe mineralization defect are found. Patients have white sclerae and do not have dentinogenesis imperfecta. The severity of skeletal deformity can be moderate to severe. SERPINF1, which encodes pigment epithelium-derived factor (PEDF), has been identified as the causative gene for this type.


Osteogenesis Imperfecta Types VII, VIII, and IX


These three types have in common a defect in the prolyl 3-hydroxylation complex in the endoplasmic reticulum, which plays a crucial role in the assembly of the triple helix by posttranslational modification of specific proline residues in unfolded collagen α-chains. Components of the complex include cartilage-associated protein ( CRTAP ), prolyl 3-hydroxylase 1 (encoded by LEPRE1 ), and peptidyl-prolyl cis-trans isomerase B ( PPIB ) which are the causative genes for types VII, VIII, and IX disease, respectively. All three types have autosomal recessive transmission. Clinically, type VII disease is associated with rhizomelia, moderate to severe skeletal involvement, and growth deficiency. Type VII was first described in the families of First Nations people in northern Quebec. Null mutations in CRTAP are lethal. Type VIII disease can also be severe or lethal and is associated with rhizomelia; it was first described in South African blacks. Type IX disease is similar in severity to types VII and VIII diseases but does not produce rhizomelia.


Osteogenesis Imperfecta Types X and XI


Types X and XI disease are caused by defects in SERPINH1 and FKBP10, respectively. These are so-called collagen chaperons that bind and accompany the procollagen molecule from the endoplasmic reticulum to the Golgi apparatus. Type X disease is associated with severe bone dysplasia, blue sclera, dentinogenesis imperfecta, transient skin bullae, pyloric stenosis, and renal stones. Type XI disease is associated with bone dysplasia, ligamentous laxity, platyspondyly, and scoliosis. Sclerae and teeth are normal.


One of the problems for the orthopaedic surgeon and the families of patients with osteogenesis imperfecta is the significant variability in the severity of long bone deformity and fracture frequency in the Sillence classification categories. Even within families, whose members presumably share the same genetic defect, one sibling may have only a few fractures whereas another may have repeated fractures, deformity requiring intramedullary rods, and difficulty ambulating without lower extremity bracing or an upper extremity aid. Because of this practical problem, clinical classifications based on the age at onset and severity of fractures still have prognostic relevance for orthopaedic surgeons and affected individuals.


Looser in 1906 classified osteogenesis imperfecta into two types, osteogenesis imperfecta congenita, characterized by the presence of numerous fractures at birth, and osteogenesis imperfecta tarda, in which the fracture(s) occur after the perinatal period. Shapiro recommended a further modification of Looser’s classification consisting of four categories—congenita A, congenita B, tarda A, and tarda B. This classification has excellent practical application for the orthopaedic surgeon and families of affected individuals in regard to prognosis for survival and ambulation. Shapiro classified patients as having osteogenesis imperfecta congenita if they had fractures in utero or at birth, whereas Looser and other authors used congenita only for in utero fractures. The distinction between the two congenita types is based on the timing of the fracture and radiographic features of the affected bones. Patients with congenita A sustain fractures in utero or at birth, with the additional radiographic features of crumpled long bones, crumpled ribs with rib cage deformity, and a fragile skull ( Fig. 42-27 ). These features are incompatible with life, and the patients are almost always stillborn or die shortly after birth from intracranial hemorrhage or respiratory insufficiency. Patients with congenita B have fractures at birth but are radiographically distinct from congenita A patients in that the long bones, as typified by the femur, are more tubular and have more normal funnelization in the metaphysis, the ribs are more normally formed (although there may be rib fractures), and there is no rib cage deformity. These patients are severely affected, but this type is compatible with survival. Patients with tarda A have an onset of fractures before walking. The age at onset of fractures was not prognostic for ambulation within this group in Shapiro’s study. Patients with tarda B suffer their first fracture after walking age; all these patients were ambulatory in Shapiro’s study.


May 25, 2019 | Posted by in ORTHOPEDIC | Comments Off on Metabolic and Endocrine Bone Diseases

Full access? Get Clinical Tree

Get Clinical Tree app for offline access