• Keine Ergebnisse gefunden

No Adaptations in Bone of Leptin- Deficient ob/ob Mice in Response to Loading

N/A
N/A
Protected

Academic year: 2022

Aktie "No Adaptations in Bone of Leptin- Deficient ob/ob Mice in Response to Loading"

Copied!
8
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Introduction

Leptin is a peptide hormone produced predominantly by whi- te fat cells [1,47]. The mature protein, encoded by the obesity (ob) gene localized in human and mouse 7 and 6 chromosomes, respectively, is a 16 kDa non-glycosylated protein. Leptin le- vels in the blood are proportional to adipose tissue mass. In- itially discovered as a central regulator of appetite and energy expenditure, leptin levels can be considered as a signal to the body regarding its energy reserves. Although elevated leptin le- vels are present in obesity, it is likely that the physiological importance of leptin is that low levels indicate a state of star- vation [2].

Recent data suggest that leptin may regulate a variety of other physiological processes, such as insulin action [7], hemopoie- sis [5], immune function [28], reproduction [40], angiogenesis [26], and bone development and remodeling [19].

However, it is not clear whether leptin is a stimulator or an inhibitor of bone growth in humans. Some investigators noted

a positive relationship between serum leptin levels and bone mineral density (BMD). Iwamoto et al. described a weakly cor- relation of serum leptin level with bone mineral density of pelvis and left leg in the premenopausal women and Odebasi et al. found that there was no correlation between plasma lep- tin concentrations and BMD values in healthy postmenopau- sal woman but a weak correlation was observed in postmeno- pausal woman with osteoporosis [23,35]. Whereas others ob- served a negative relationship. Blum et al. suggest that for a given body weight, a higher proportion of fat and a higher se- rum leptin concentration have negative associations with bo- ne mass in premenopausal women [6]. Sato et al. suggest that an increase in serum leptin reduces bone formation and de- creases BMD in adult men [42].

Furthermore, no associations between serum leptin levels and BMD have been reported, which further confounds the inter- pretation of leptin’s effect on bone mass. Martini et al. conclude that serum leptin have no direct effect on bone mass and bo- ne turnover in healthy postmenopausal women [32] and Rauch et al. appear that leptin has less influence on the mature than on the growing skeleton [41].

Only a few studies in humans have examined the direct effect of leptin administration on BMD. Ogueh et al. [36] noted a Autor:Hansjoerg

Heep1, Christian Wede- meyer1, Jie Xu2, Sebas- tian Hofmeister1, Marius von Knoch1

Schlagworte:Leptin, trabekulärer und korti- kaler Knochen, MikroCT, biomechanische Belas- tung

Keywords:leptin, corti- cal und trabecular bone, microCT, biomechanical loading

Zitierweise dieses Bei- trages: BIOmaterialien 2008; 9 (1/2): S. 18-25

Leptin was found to be involved in regulation of bone development and remodeling. This study was performed to evaluate the effects of biomechanical loading due to body weight in leptin-defient ob/ob mice at the same pubertal stage. No significant difference was found between the two groups for all the parameters (p>0.05) suggesting that the presence of leptin may be an important pre-condition for positive correlation between loading and bone mass. Furthermore, concordance of bone formation was found among appendicular re- gions, but was not found between the axial and appendicular re- gions.

No Adaptations in Bone of Leptin- Deficient ob/ob Mice in Response to Loading

Keine Anpassung des Knochens von Leptin-defizienten ob/ob-Mäusen bei biomechanischer Belastung

1 Department of Orthopaedics, University of Duisburg-Essen, Pattbergstrasse 1-3, 45239 Essen, Germany

2 Department of Orthopaedics, The second affiliated hospital of Sun Yat-Sen University Guangzhou, China

(2)

significant negative correlation between fetal blood levels of leptin and cross-linked carboxyl-terminal telopeptide of Ty- pe 1 collagen (a marker of bone resorption). A modest negati- ve correlation was also noted between leptin levels and the concentration of carboxyl-terminal pro-peptide of Type 1 col- lagen (a marker of bone formation), but the researchers spe- culated that the overall effect of leptin on fetal bone metabo- lism was to increase bone mass by decreasing bone resorption [36]. One study reported an increase in bone mass and decre- ase in bodyweight after long-term leptin therapy in an obese 9-year-old girl with congenital leptin deficiency [12]. That might show the positive effect on bone formation in early de- velopment. Simha et al. concluded that the effect of leptin on bone metabolism may depend on the stage of life in humans because there were no effects of leptin on osteoblastic or oste- oclastic activity [43]. Larger clinical studies were therefore ne- cessary to clarify this hypothesis. In few recent reports the an- imal model was a useful surrogate in investigating the me- chanism of leptin. Both leptin and its receptors were found in murine fetal cartilage and bone template, as well as in the growth plate [27]. In addition, leptin increased both prolife- ration and differentiation of the chondrocyte population of skeletal growth centers in organ cultures. The results of Ma- or et al. indicate that leptin acts as a skeletal growth factor with a direct peripheral effect on skeletal growth centers and they speculate that the high circulating levels of leptin in ob- ese children might contribute to their growth [31]. Nakajima et al. show that the growth plate chondrocytes possess leptin re- ceptors (Ob-Rs), and leptin enhance chondrocyte proliferation and subsequent cell differentiation [34].

All these strongly supported the theory that leptin could sti- mulate bone growth in the early stages of life.

Weight-bearing exercise has been shown to have an advanta- geous effect on BMD. Chen et al. demonstrate results of the histomorphometric analysis after 9 weeks of exercise. They showed that the periosteal labeled surface, mineral apposition rate, and bone formation rate were profoundly increased in aged female rats [9]. Iwamoto et al. findings suggested that in the mature osteopenic rat, there was a beneficial effect of mo- derate running exercise with adequate calcium intake on bo- ne mass only in a weight-bearing long bone, the tibia. The mechanism for increased bone mass appeared to be both de- creased bone resorption and increased bone formation in can- cellous bone and increased bone formation in cortical bone [24]. Yeh et al. found that in the aged rat, by 9 weeks, exerci- se increases BMD in the tibia and in the vertebrae [46].

The phenomena of bone loss with disuse [45] and bone accru- al with in vivo loading [17] certainly emphasizes the impor- tance of mechanical loading for bone formation and mainte- nance in normal subjects without the knock-out ob gene. Me- chanical loading of bones via ground reaction forces and muscle pulling on the bones are generally accepted as neces- sary stimuli to attain peak BMD and maintain bone homeos- tasis. In general, mechanical loading stress on bones causes tissue deformation within the bone (due to strain) that stimu- lates the bone to adapt by remodeling to accommodate these demands, ultimately improving resistance to osteoporosis.

While previous studies have shown that bone parameters im- proved with weight-bearing exercise in normal subjects, we were interested in the co-influence of biomechanical loading on body weight and bone metabolism in subjects with leptin de- ficiency. The aim of this study was to investigate the differen- tial effects of different degrees of weight-bearing on trabecular and cortical bone formation in the mice without the stimulation of leptin. We chose the obese gene in mice model because this gene is known to effect both bone bone mass and body mass.

We hypothesized that biomechanical loading due to body weight might be an ignored factor which has lead to the cont- radictory results regarding BMD in studies with mice which were not known to be deficient in leptin or had inactive lep- tin receptors.

Materials and Methods

Animals

C57BL/6J-Lepob(ob/ob) female mice with an autosomal re- cessive mutation on chromosome 6 were received from the Janvier Laboratory (Le Genest St Isle, France) at five weeks of age. The animal experiment was appoved by the Univer- sity`s ethic committee and the local authorities according to the official guidelines. The animals were housed one per ca- ge with access to water ad libitum and a standard rodent diet (8640 Harlan Teklad 22/5[W]; Harlan Teklad, Madison, WI, USA) containing 1.13% calcium and 0.94% phosphorus.

The animals were maintained under conditions of a twelve- hour light and dark cycle with the light switched on at 6.00 a.m.. Food intake and body weight were recorded daily. The- re were two experimental groups divided according to the provision of food. Group A included 20 ob/ob mice with access to food ad libitum, the other 20 ob/ob mice in Group B received a limited amount of food (6 gr. each day). All the animals were euthanized by mechanical procedure at the age of twenty weeks.

Micro-CT

A high resolution micro-CT (SkyScan 1072, Aartselaar, Bel- gium) was used to perform qualitative and quantitative analysis of the limb and the vertebral body. All samples were scanned by a commercially available microcomputed tomographic scan- ner at the Department of Cardiology of the West German He- art Centre at the University of Duisburg-Essen.

Technical specifications of the Micro-CT

The micro-CT system is based on a scanner developed for high- resolution imaging (up to 4 µm cubic voxels) of sample sizes up to 2 cm3. The scanner uses a field x-ray tube with an 8 µm spot-seize and expected lifetime of > 10,000 hours. The tube is operated at between 20 and 100 kiloelectron volts and a cur- rent of up to 100 micro amperes. For scanning, the samples are placed between the microfocus X-ray source and a Char- ge-Coupled-Device (CCD) detector (matrix size: 1024x1024 pi- xels, field of view: 25 mm2). In order to prevent samples from moving during scanning, the imbs and vertebral bodies are placed in a tightly fitting rigid plastic tube. In the scanner’s chamber the specimens are placed on a stack of computer- controlled precision stages which are rotated in equiangular steps of 0.9° around an angle of 180°. When the object is pla- ced between the x-ray source and the CCD-detector, the cone- beam of X-rays passes the object and then hits the CCD-dete- ctor producing 2D-X-ray images. A personal computer is used to control the scanner and store the CCD image data recorded at each angle of view during the scanning process.

Tomographic Image Reconstruction

The X-ray projection data of the scanned samples are then submitted to the resident reconstruction program (Cone-beam Reconstruction, Skyscan, Aartselaar, Belgium), which is based on a Feldkamp filtered back projection algorithm [48] resulting in a volume image of up to 10,243 voxels, each cubic voxel being 4–19 µm on one side, depending on how much of the specimen had to be imaged.

(3)

Cancellous bone assessment by micro-CT

Three regions of interest (ROIs) from the vertebrae, proximal fe- mur and tibia were selected for structural analysis of the can- cellous bone. The ROI for the vertebrae was 0.5×0.5×0.5mm3 in the middle of Lumbar 3. A cubic region of 0.5×0.5×0.5mm3 in the metaphysis of the tibia was adjacent to (1mm away from) the growth plate and femoral head. Bone volume ratio (BV/TV) was calculated by adding the number of voxels re- presenting mineralized trabecular bone divided by the total volume, and expressed as a percentage. The bone surface ra- tio (BS/BV) was calculated by adding the surface components of the mineralized bone phase directly from the 3-D data and dividing by the total bone phase volume. Trabecular thickness (Tb. Th), trabecular separation (Tb.Sp) and trabecular number (Tb.N) were also based directly on the 3-D data using medial axis transformation and distance transformation.

Cortical bone assessment by micro-CT

In the femur, a cubic region of 2.5×2.5×2.5mm3in the midd- le of the shaft adjacent to the femoral crest was selected. This region, which could include the whole cycle of the cortical bo- ne, contained no trabecular bone. Bone volume (BV) and bo- ne surface (BS) were calculated by adding the number of vo- xels and the surface components of the mineralized bone from the 3-D data respectively. Cortical thickness (C.Th) was calcu- lated by the formula:

C.TH = 2x ,

introduced by Bagi CM et al [3], which can be explained by:

area of a ring = thickness of ring × length of middle line =

thickness × .

Determining the thickness via 3D parameters allowed a more robust and precise determination than calculating them di- rectly on individual slices only.

Statistical analysis

Data were analyzed and assessed using SPSS software (ver- sion 12.0; SPSS Institute Inc, Chicago). Descriptive statistics of all variables were determined including the mean and stan- dard deviation of each group. The difference of all parameters between the two groups was assessed by the Student`s t-Test because all parameters (were normal distributed which was tested with the Kolmogorov Smirnov test). Pearson’s correla- tion coefficient was used to assess the relationship between all the trabecular bone parameters of femur, vertebra and tibia.

A value of p≤0.05 was considered to be statistically significant.

Results

No death or health deterioration occurred during this study.

The body weight at each time point in the two groups is shown in Figure 1.

Animals with an ad libitum diet (Group A) were found to in- crease body weight significantly at the age of six weeks in com- parison with the lean mice (Group B). From this time point on the difference constantly increased. At the age of twenty weeks the obese mice were almost twice as heavy as the lean mice.

Micro-CT measurements were obtained for assessment of the morphological changes in the two groups (Fig. 2). Examination of trabecular bone in the limbs and spine revealed a statistically insignificant difference between the two groups. High weight- bearing insignificantly improved all trabecular bone parame- ters in the obese mice (Table 1).

Weight-bearing showed no correlation with the cortical bone parameters, including BV, BS and thickness of femoral shaft.

One-way ANOVA did not reveal any statistically significant difference between the parameters in the two groups (Table 1).

Figure 3 provided scatter plots of each of the morphological pa- Fig. 1: The curves show the change in body-weight in the two groups. In Group A the animals had an ad libitum diet, the animals in Group B a con- trolled diet. Inter-group difference was already significant at the age of six weeks (p<0.05).

Fig. 2. Three-dimensional images of all the ROIs of the mice in Groups A and B at the age of 20 weeks. The ROI for the vertebrae was 0.5x0.5x0.5mm3 in the middle of Lumbar 3. A cubic region of 0.5x0.5x0.5mm3in the metaphysis of the tibia was adjacent to (1mm away from) the growth plate and femoral head.

BV BS

outercircumference + innercircumference 2

(4)

rameters of the trabecular bone in the limbs and vertebrae.

The related regression results are summarized in Table 3. All the femoral parameters exhibited linear relations with those of the tibia, but non-linear relations with the vertebra.

Discussion

A great deal of research has confirmed that increased biome- chanical loading due to increased body weight contributes to the increased bone dimensions and mass observed in not on- ly our animal model but also obese humans. Frost et al. des- cribe that an increased loading of long bones produces the greatest mechanical stresses on the subperiosteal surface and stimulates bone formation by subperiosteal expansion [14].

For example, loading in the playing arm in racquet sports in- duces significant increases in bone dimensions and mass [18].

The effect of weight gain on bone loading has not only been examined in adults but also in postmenopausal women. Beck et al. show a significant increase in hip cortical section modulus due to periosteal expansion [4]. A qCT (quantitative compu- tertomography) study in healthy children suggested that weight-bearing and mechanical stresses are important deter- minants of cortical bone mass, whereas trabecular bone den- sity is influenced by hormonal factors associated with sexual development [33].

A study of bone biomechanics in adult rats with diet-induced obesity showed significantly greater bone strength in the ob- ese rats than in the controls. The cross-sectional geometry and ultimate fracture load of the femur were higher in the obese rats than in the controls [8].

In our study, however, there were no significant differences in trabecular and cortical bone mass between the leptin-defi- cient obese mice and the leptin-deficient control mice. In ot- her words, it seemed that increased biomechanical loading due

to increased body weight did not contribute to increased bone dimensions and mass in the leptin-deficient subjects.

We considered that our results did not negate the conclusion of the other research mentioned above regarding the positive effect of biomechanical loading on bone mass, as these stu- dies did not focus on leptin-deficient subjects. Numerous types of obesity were studied: one of them was congenital leptin de- ficiency, which is described in the human obesity gene map [38]. Oral et al. reported on a group of nine patients whose obesity was attributed to low serum leptin levels, and whose condition improved after leptin replacement [37]. Another stu- dy investigated subjects with a higher serum leptin level. Much research has concentrated on the positive effect of leptin in not only decreasing food intake but also increasing bone for- mation by modulating the sympathetic nervous system [10].

During the same phase, the increased biomechanical loading due to increased body weight contributed to the increased bo- ne mass as a co-influence. Hla et al. and the Early Postmeno- pausal Intervention Cohort (EPIC) Study Group show that the increased bone mineral content (BMC) in childhood obesity was revealed both in the weight-bearing limbs and the unlo- aded arms [22]. The new finding of our study, which focused on the co-influence of biomechanical loading in subjects with leptin-deficiency, is that it confirmed the presence of leptin as the pre-condition for a positive correlation between loa- ding and bone mass.

We could, of course, not eliminate the possible effect of other factors on bone mass, such as estrogen. Estrogen was confir- med to be present in postmenopausal obese females at a rela- tively higher serum level because adipocytes were a major si- te of estrogen production in these subjects. Heshmati et al. de- monstrate that endogenous residual estrogen levels determine bone resorption even in the late postmenopausal women [21].

Position Group BV/TV (%)

Mean S.D.

BS/BV (1/mm) Mean S.D.

Tb.Th (mm) Mean S.D.

Tb.Sp (mm) Mean S.D.

Tb.N(1/mm) Mean S.D.

Femur Group A

(diet ad libitum) Group B (control diet)

P-value

60.66 6.13

62.69 6.53

0.317

36.10 3.81

36.70 3.92

0.837

0.094 0.0069

0.093 0.0075

0.905

0.098 0.012

0.092 0.011

0.088

6.42 0.45

6.74 0.55

0.051

Tibia Group A

(diet ad libitum) Group B (control diet)

P-value

53.31 4.16

54.64 7.48

0.494

37.89 2.65

38.56 3.74

0.520

0.094 0.0063

0.093 0.0084

0.505

0.13 0.015

0.13 0.022

0.526

5.67 0.36

5.90 0.52

0.118 Vertebrae Group A

(diet ad libitum) Group B (control diet)

P-value

31.79 6.06

34.97 8.17

0.171

45.46 4.09

43.90 4.92

0.283

0.085 0.0067

0.085 0.0062

0.925

0.17 0.034

0.16 0.028

0.358

4.09 0.78

3.75 0.59

0.134 Table 1: Cross-section structural geometric properties of the femur, tibia and vertebrae were evaluated using micro-CT. Note: Summary of morphome- tric characteristics in the two groups which were different in body weight-bearing. No statistically significant difference was detected between the two groups (p>0.05).

(5)

Khosla et al. suggest that in contrast to traditional belief, age- related bone loss may be the result of estrogen deficiency not just in postmenopausal women, but also in men [25]. But the mice in our study were no more than twenty weeks old (in the pubertal stage) and without ovariotomy. Thus, estrogen as a confounding factor in our study is not very likely. There is so far no agreement regarding the positive effect of leptin in the early stages of life. The data gathered during some research studies appear conflicting or even contradictory. Ducy et al.

observed that three- and six- month old ob/ob mice, which were deficient in leptin, and db/db mice, which have a muta- ted and inactive leptin receptor, had higher trabecular bone mass associated with a higher mineral apposition rate when compared with their wild-type litter-mates [11]. On the other hand, Foldes et al. showed that obese fa/fa rats, which also had a mutated leptin receptor, had a lower bone mass with shorter and lighter femurs when compared with their normal litter-mates [13]. Lower femoral BMD and osteocalcin (a mar- ker of bone formation) serum were described in fa/fa rats, as well as decreased trabecular bone volume, trabecular thick- ness and trabecular number, as measured both by micro-CT and histomorphometry. Picherit et al. have compared femoral bone density and biochemical markers of bone metabolism in male and female fatty (leptin-resistant) Zucker rats and their le- an homozygous controls at 3 and 6 months of age [39]. Tamasi et al. used female leptin receptor-deficient Zucker (fa/fa) rats and their homozygous (Fa/Fa) and heterozygous (Fa/fa) lean controls at 9 and 15 weeks of age. They suggest that leptin exerts a positive effect on bone, based on the observed skele- tal phenotype of the Zucker (fa/fa) rat [44]. The reasons for these discrepancies remain unclear.

In our study, concordance of bone formation was found among appendicular regions, but not between the axial and appen- dicular regions. Hamrick et al [20] found that ob/ob mice show increased bone content, density and trabecular bone volume in the lumbar spine which was consistent with the results of Du- cy et al [11]. Due to the limitation of the design this study, we did not assess the change from each time point of growth.

Even so, the discrepancy of bone formation between limb and spine indicated the different effect of leptin on bone forma- tion in axis and appendicular regions, or the bone formation of the spine took place earlier than that of the limb and was stable at the time of our assessment. Large clinical studies are necessary to consolidate leptin’s role in the physiology of hu- man bone.

Abstract

Problemstellung: Das Hormon Leptin reguliert die Reifung und Entwicklung des Knochens. Jedoch nicht bekannt ist der Ein- fluß von Leptin auf das wachsende Skelett. Unsere Studie untersucht die Auswirkung der biomechanischen Belastung des Körpergewichtes von Leptin-defizienten ob/ob-Mäusen in der Pubertät.

Methodik: Beginnend mit einem Alter von 5 Wochen wurden zwei Gruppen von je 20 weiblichen ob/ob-Mäusen untersucht.

Mit einem Alter von 20 Wochen war das Körpergewicht der Gruppe A, die keine Einschränkung des Essverhaltens erfuhren, signifikant schwerer als in der Gruppe B unter kontrollierter Diät (p<0.05). Die trabekulären und kortikalen Strukturen des Knochens wurden mittels MikroCT untersucht.

Ergebnisse: Unter Berücksichtigung, daß Leptin einen wichti- Fig. 3: Plots of the correlation between the femoral, tibial and vertebral regions, respectively for trabecular parameters. The correlation between the femoral and tibial regions was represented by an open triangle (LL), between the femoral and vertebral regions by a filled square (IZ.

(6)

gen Faktor für eine positive Korrelation zwischen der Belastung und der Knochenmasse darstellt, zeigt sich keine signifikante Unterscheidung der beiden Gruppen in allen Parametern (p>0.05). Jedoch zeigen sich Übereinstimmungen der knö- chernen Strukturen entlang der Extremitäten, die sich nicht im Vergleich zwischen den Extremitäten und des Achsenske- letts nachweisen lassen.

References

[1] Ahima RS, Flier JS. Leptin.Annu Rev Physiol. 2000;62:413- 37.

[2] Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, Flier JS. Role of leptin in the neuroendocrine response to fasting. Nature. 1996 Jul 18;382(6588):250-2.

[3] Bagi CM, Hanson N, Andresen C, Pero R, Lariviere R, Tur- ner CH, Laib A. The use of micro-CT to evaluate cortical bone geometry and strength in nude rats: correlation with mecha- nical testing, pQCT and DXA. Bone, 2006 38:136-44.

[4] Beck TJ,Stone KL,Oreskovic TL,et al.Effects of current and discontinued estrogen replacement therapy on hip structural geometry: the study of osteoporotic fractures. J Bone Miner Res2001;16:2103–10.

[5] Bhatt R, Bhatt S, Rameshwar P, Siegel A. Long-term kind- led seizures induce alterations in hematopoietic functions: ro- le of serum leptin. Epilepsy Res. 2005;65(3):169-78.

[6] Blum M, Harris SS, Must A, Naumova EN, Phillips SM, Rand WM, Dawson-Hughes B. Leptin, body composition and bone mineral density in premenopausal women. Calcif Tissue Int. 2003;73(1):27-32.

[7] Boghossian S, Dube MG, Torto R, Kalra PS, Kalra SP. Hy- pothalamic clamp on insulin release by leptin-transgene ex- pression.Peptides. 2006;27(12):3245-54.

[8] Brahmabhatt V, Rho J, Bemardis L, Gillespie R, Ziv I. The ef- fects of dietary-induced obesity on the biomechanical proper- ties of femora in male rats. Int J Obes Relat Metab Disord 1998;22:813-18.

[9] Chen MM, Yeh JK, Aloia JF, Tierney JM, Sprintz S. Effect of treadmill exercise on tibial cortical bone in aged female rats: a histomorphometry and dual energy X-ray absorptio- metry study. Bone. 1994;15:313-19.

[10] Cock TA, Auwerx J. Leptin: cutting the fat off the bone.

Lancet, 2003 362:1572-74.

[11] Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, Shen J, Vinson C, Rueger JM, Karsenty G. Leptin in- hibits bone formation through a hypothalamic relay: a cen- tral control of bone mass. Cell. 2000;100(2):197-07.

[12] Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O’Rahilly S. Ef- fects of recombinant leptin therapy in a child with congenital leptin deficiency.N Engl J Med. 1999 ; 16;341(12):879-84.

[13] Foldes J, Shih MS, Levy J. Bone structure and calcium metabolism in obese Zucker rats. Int J Obes Relat Metab Disord.

1992;16(2):95-102.

[14] Frost HM. The mechanostat: a proposed pathogenic me- chanism of osteoporoses and the bone mass effects of mecha- nical and nonmechanical agents. BoneMiner1987;2:73–85.

[15] Goulding A, Cannan R, Williams SM, Gold EJ, Taylor RW, Lewis-Barned NJ. Bone mineral density in girls with forearm fractures. J Bone Miner Res 1998;13:143–8.

[16] Goulding A, Jones IE, Taylor RW, Williams SM, Manning PJ. Bone mineral density and body composition in boys with distal fore arm fractures: a dual-energy x-ray ab- sorptiometry study. J Pediatr 2001;139:509–15.

[17] Gross TS, Srinivasan S, Liu CC, Clemens TL, Bain SD. Noninvasive loading of the murine tibia: an in vi- vo model for the study of mechanotransduction. J Bo- ne Miner Res. 2002; 17:493-01.

[18] Haapasalo H, Kontulainen S,Sievanen H, Kannus P, Jarvinen M,Vuori I.Exercise-induced bone gain is due to enlargement in bone size without a change in volume tricbone density:a peripheral quantitative com- puted tomography study of the upper arms of mal eten- nisplayers.Bone2000; 27:351–7.

Group BV(mm3)

Mean S.D.

BS(mm2) Mean S.D.

Thickness(mm) Mean S.D.

Group A

(diet ad libitum) 0.28 0.025 5.274 0.300 0.108 0.005

Group B

(control diet) 0.277 0.015 5.553 1.608 0.103 0.014

P-value 0.176 0.337 0.120

Table 2: Cross-sectional structural geometric properties in the femoral shaft were evaluated using micro-CT.

Parameter r P-value Equation

Femur vs tibia BV/TV (%) BS/BV (1/mm)

Tb.Th (mm) Tb.Sp (mm)

0.525 0.569 0.512 0.320

<0.05

<0.05

<0.05

<0.05

y= 31.857 + 0.552 x y= 20.801 + 0.479 x y= 0.0437 + 0.529 x y= 0.0772 + 0.521 x Femur vs vertebra

BV/TV (%) BS/BV (1/mm)

Tb.Th (mm) Tb.Sp (mm)

0.136 -0.014

0.111 0.179

0.403 * 0.932 * 0.496 * 0.268 *

y= 23.752 + 0.156 x y= 45.286 - 0.0166 x y= 0.0754 + 0.0994 x y= 0.119 + 0.481x

Table 3: The pairs of BV/TV BS/BV, Tb.Th, Tb. N , Tb. Sp of the fe- moral and tibial ROIs with positive correlation coefficients and P va- lues below 0.05 tend to increase together. No correlation was found between the femoral and vertebral regions.

(7)

[19] Hamrick MW, Ding KH, Pennington C, Chao YJ, Wu YD, Howard B, Immel D, Borlongan C, McNeil PL, Bollag WB, Curl WW, Yu J, Isales CM. Age-related loss of muscle mass and bo- ne strength in mice is associated with a decline in physical activity and serum leptin. Bone. 2006;39(4):845-53.

[20] Hamrick MW, Pennington C, Newton D, Xie D, Isales C.

Leptin deficiency produces contrasting phenotypes in bones of the limb and spin. Bone, 2004 34:376-83.

[21] Heshmati HM, Khosla S, Robins SP, Geller N, McAlister CA, Riggs BL. Endogenous residual estrogen levels determine bone resorption even in the late postmenopausal women. J Bone Miner Res. 1997 12:S1;S121.

[22] Hla MM, Davis JW, Ross PD, Wasnick RD, Yates AJ, Ravn P, Hosking DJ, McClung MR, and the Early Postmenopausal Intervention Cohort (EPIC) Study Group. A multicenter study of the influence of fat and lean mass on bone mineral con- tent: Evidence for difference in their relative influence at ma- jor fracture sites. Am J Clin Nutr, 1996 64:354-60.

[23] Iwamoto I, Douchi T, Kosha S, Murakami M, Fujino T, Nagata Y. Relationships between serum leptin level and re- gional bone mineral density, bone metabolic markers in he- althy women. Acta Obstet Gynecol Scand. 2000;79(12):1060- 4.

[24] Iwamoto J, Takeda T, Ichimura S. Effect of exercise on ti- bial and lumbar vertebral bone mass in mature osteopenic rats:

bone histomorphometry study. J Orthop Sci.1998; 3:257-63.

[25] Khosla S, Melton LJ ?, Atkinson EJ, O’Fallon WM, Klee GG, Riggs BL. Relationship of serum sex steroid levels and bo- ne turnover markers with bone mineral density in men and women: A key role for bioavailable estrogen. J Clin Endocrinl Metab. 1998 83: 2266-74.

[26] Kitade M, Yoshiji H, Kojima H, Ikenaka Y, Noguchi R, Ka- ji K, Yoshii J, Yanase K, Namisaki T, Asada K, Yamazaki M, Tsujimoto T, Akahane T, Uemura M, Fukui H. Leptin-mediated neovascularization is a prerequisite for progression of nonal- coholic steatohepatitis in rats. Hepatology. 2006;44(4):983- 91.

[27] Kume K, Satomura K, Nishisho S, Kitaoka E, Yamanouchi K, Tobiume S, Nagayama M. Potential role of leptin in end- ochondral ossification. J Histochem Cytochem. 2002;50(2):159- 69.

[28] Lam QL, Lu LW. Role of leptin in immunity.Cell Mol Im- munol. 2007;4(1):1-13.

[29] Leonard MB, Shults J, Wilson BA, Tershakoves AM, Zemel BS. Obesity during childhood and adolescence augments bone mass and bone dimensions. Am J Clin Nutr. 2004 80:514-23.

[30] Ma D, Jones G. The association between bone mineral density, meta-carpal morphometry, and upper limb fractures in children:apopulation-based case-control study. J Clin End- ocrinol Metab 2003;88:1486–91.

[31] Maor G, Rochwerger M, Segev Y, Phillip M. Leptin acts as a growth factor on the chondrocytes of skeletal growth cen- ters. J Bone Miner Res. 2002;17(6):1034-43.

[32] Martini G, Valenti R, Giovani S, Franci B, Campagna S, Nuti R. Influence of insulin-like growth factor-1 and leptin on bone mass in healthy postmenopausal women. Bone.

2001;28(1):113-7.

[33] Mora S, Goodman WG, Loro ML, Roe TF, Sayre J, Gilsanz V. Age-related changes in cortical and cancellous vertebral bone density in girls: assessment with quantitative CT. AJR Am J Roentgenol 1994;162:405–9.

[34] Nakajima R, Inada H, Koike T, Yamano T. Effects of lep- tin to cultured growth plate chondrocytes.Horm Res.

2003;60(2):91-8.

[35] Odabasi E, Ozata M, Turan M, Bingol N, Yonem A, Cakir B, Kutlu M, Ozdemir IC. Plasma leptin concentrations in post-

menopausal women with osteoporosis. Eur J Endocrinol.

2000;142(2):170-3.

[36] Ogueh O, Sooranna S, Nicolaides KH, Johnson MR 2000 The relationship between leptin concentration and bone meta- bolism in the human fetus. J Clin Endocrinol Metab 85:1997–

99.

[37] Oral EA, Simha V, Ruiz E et al. Leptin-replacement therapy for lipodystrophy. N Engl J Med 2002; 346:570-8.

[38] Perusse L, Tuomo R, Aamir Z et al. The human obesity gene map: the 2004 update. Obes Res. 2005;13:381-490.

[39] Picherit C, Horcajada MN, Mathey J, Chanteranne B, Puel C, Lebecque P, Davicco MJ, Coxam V, Barlet JP. Isoflavone consumption does not increase the bone mass in osteopenic obese female zucker rats. Ann Nutr Metab. 2003;47(2):70-7.

[40] Popovic V, Casanueva FF. Leptin, nutrition and reprodu- ction: new insights. Hormones (Athens). 2002;1(4):204-17.

[41] Rauch F, Blum WF, Klein K, Allolio B, Schonau E. Does leptin have an effect on bone in adult women? Calcif Tissue Int.

1998;63(6):453-5.

[42] Sato M, Takeda N, Sarui H, Takami R, Takami K, Hayas- hi M, Sasaki A, Kawachi S, Yoshino K, Yasuda K. Association between serum leptin concentrations and bone mineral densi- ty, and biochemical markers of bone turnover in adult men. J Clin Endocrinol Metab. 2001;86(11):5273-6.

[43] Simha V, Zerwekh JE, Sakhaee K, Garg A. Effect of sub- cutaneous leptin replacement therapy on bone metabolism in patients with generalized lipodystrophy. J Clin Endocrinol Metab. 2002 Nov;87(11):4942-5.

Dr. med.

Hansjoerg Heep

Korrespondenzadresse:

Department of Orthopaedics, University of Duisburg-Essen Pattbergstr. 1-3

D-45329 Essen

hansjoerg.heep@uni-due.de Phone.: 0201/4089-2147 Fax: 0201/4089-2722 Akademischer Lebenslauf

Oberarzt, Orthopädische Klinik der Universität Duisburg- Essen im Evangel. Krankenhaus Essen-Werden

1986-1988 Studium der Chemie Universität Heidel- berg

1988-1994 Studium der Humanmedizin Heidelberg und München (LMU, Sommersemester 1988)

1997 Dissertation „Quantifizierung der RES- Clearance nach Reperfusion bei Leber- transplantation unter klinischen Bedin- gungen“

2001 Facharzt Chirurgie

2003 Schwerpunkt Unfallchirurgie

2006 Facharzt Orthopädie und Unfallchirurgie 2007 Zusatzbezeichnung Spezielle Orthopädi- sche Chirurgie, Physikalische Therapie 2003 Oberarzt Unfallchir., Universität Düssel-

dorf

2006 Orthopädie Universität Duisburg-Essen

(8)

[44] Tamasi JA, Arey BJ, Bertolini DR, Feyen JH. Characteri- zation of bone structure in leptin receptor-deficient Zucker (fa/fa) rats. J Bone Miner Res. 2003 Sep;18(9):1605-11.

[45] Warner SE, Sanford DA, Becker BA, Bain SD, Sriniva- sanS, Gross TS. Botox induced muscle paralysis rapidly de- grades bone. Bone. 2006; 38:257-64.

[46] Yeh JK, Aloia JF, Tierney JM, Sprintz S. Effect of treadmill exercise on vertebral and tibial bone mineral content and bo- ne mineral density in the aged adult rat: determined by dual energy X-ray absorptiometry. Calcif Tissue Int. 1993; 52:234- 38.

[47] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372(6505):425-32.

[48] Zhao S, Yu H, Wang G. A unified framework for exact cone-beam reconstruction formulas. Med Phys 2005; 32 (6):

1712-21.

Abbildung

Fig. 2. Three-dimensional images of all the ROIs of the mice in Groups A and B at the age of 20 weeks
Table 2: Cross-sectional structural geometric properties in the femoral shaft were evaluated using micro-CT.

Referenzen

ÄHNLICHE DOKUMENTE

There was statistically significant positive correlation with regard to serum leptin levels with MS disability (EDSS), severity (MSSS) and progression (PI).. Moreo- ver, a

In response to siRNA-mediated HDAC6 knockdown, the ChIP results revealed a sig- nificant decrease in HDAC6 binding (Fig. 5A) and a sig- nificant increase in acetylated H3K9/K14

Phenotype (body mass index, body fat, leptin levels and metabolic abnormalities) of human mono‑allelic likely pathogenic variants of the leptin receptor gene (LEPR wt/-) in

total MdBAr and MdBDn, as well as periosteal and endosteal MdBDn, compared with the wild-type mice at week 3. These outcomes, in accordance with the radiological

After defining the effect of different doses of E 2 on bone and the uterus in general the next aim is to test the hypothesis that due to the presence of both estrogen receptor (ER)

Leptin concentrations in the interscapular brown adipose tissue (IBAT) of the common shrew (Sorex araneus) were measured in different seasons.. The leptin concentrations in IBAT

Unterstützung, Geduld und Veto als ich wieder spät Nachts arbeitete. 우리 애기 감사합니다 Mijn ouders wil ik vooral bedanken voor alle kansen die ze mij gegeven hebben om mijn

While the PBS injected hearts showed low cellular content within the compacted non-transmural scar (Figure 6 A, B), the immediately injected WBM cells led to