• Aucun résultat trouvé

Alteration of the bone tissue material properties in type 1 diabetes mellitus: A Fourier transform infrared microspectroscopy study

N/A
N/A
Protected

Academic year: 2022

Partager "Alteration of the bone tissue material properties in type 1 diabetes mellitus: A Fourier transform infrared microspectroscopy study"

Copied!
22
0
0

Texte intégral

(1)

Alteration of the bone tissue material properties in type 1 diabetes mellitus: a Fourier-transform infrared microspectroscopy study

Aleksandra Mieczkowska1, Sity Aishah Mansur3,4, Nigel Irwin3,Peter R. Flatt3, Daniel Chappard1,2, Guillaume Mabilleau1,2

1: LUNAM Université, GEROM-LHEA, Institut de Biologie en Santé, Angers, France ; 2: LUNAM Université, SCIAM, Institut de Biologie en Santé, Angers, France ; 3: School of Biomedical Sciences, University of Ulster, Coleraine, United Kingdom ; 4: University Tun Hussein Onn Malaysia, Johor, Malaysia

Corresponding author: G. Mabilleau, guillaume.mabilleau@univ-angers.fr

Type 1 diabetes mellitus (T1DM) is a severe disorder characterized by hyperglycemia and hypoinsulinemia. A higher occurrence of bone fractures has been reported in T1DM and although bone mineral density is reduced in this disorder, it is also thought that bone quality may be altered in this chronic pathology. Vibrational microscopies such as Fourier-transform infrared microspectroscopy (FTIRM) represent an interesting approach to study bone quality as they allow investigation of the collagen and mineral compartment of the extracellular matrix in a specific bone location. However, as spectral feature arising from the mineral may overlap with those of the organic component, demineralization of bone sections should be performed for a full investigation of the organic matrix. The aims of the present study were to (i) develop a new approach, based on demineralization of thin bone tissue section to allow a better characterization of the bone organic component by FTIRM, (ii) validate collagen glycation and collagen integrity in bone tissue and (iii) better understand what alterations of tissue material properties in newly-forming bone occur in T1DM.

The streptozotocin-injected mouse (150 mg/kg body weight, injected at 8 weeks old) was used as T1DM model. Animals were randomly allocated to control (n=8) or diabetic (n=10) groups and were sacrificed 4 weeks post-STZ injection. Bones were collected at necropsy, embedded in polymethylmethacrylate and sectioned prior to examination by FTIRM. FTIRM collagen parameters were collagen maturity (area ratio between 1660 and 1690 cm-1 subbands), collagen glycation (area ratio between the 1032 cm-1 subband and amide I) and collagen integrity (area ratio between the 1338 cm-1 subband and amide II).

No significant differences in the mineral compartment of the bone matrix could be observed between controls and STZ-injected animals. On the other hand, as compared with controls, STZ-injected animals presented with significant higher value for collagen maturity (17%, p=0.0048) and collagen glycation (99%, p=0.0121) whilst collagen integrity was significantly lower by 170% (p=0.0121).

This study demonstrated the profound effect of early T1DM on the organic compartment of the bone matrix in newly-forming bone. Further studies in humans are required to ascertain whether T1DM also lead to similar effect on the quality of the bone matrix.

(2)

INTRODUCTION

Type 1 diabetes mellitus (T1DM) is a severe disorder that persists through the entire life of affected individuals. T1DM is characterized by the inability of pancreatic - cells to secrete insulin and therefore results in hyperglycemia. Several complications including bone loss and elevated fracture risk have been reported in T1DM [1, 2]. Increased fracture risk could be partially related to diabetic complications such as retinopathy and neuropathy that increase the incidence of fall [3, 4]. Another more likely explanation could reside in a direct effect of diabetes on bone tissue. Bone mineral density (BMD), measured by dual energy X-ray absorptiometry, has been reported to be lower in T1DM patients as compared to age-matched control subjects [5, 6]. However, elevated fracture risk among T1DM patients still persists even after adjusting for hip BMD [7]. Furthermore, the low BMD observed in T1DM does not fully explain the high fracture risk in this population [8].

Thus, there is a supposition that diabetic bone is more fragile than non-diabetic bone for a given BMD and that T1DM not only affects bone microarchitecture, but also bone quality.

The role of glucose in the formation of advanced glycation end products (AGE) is well known, and poorly controlled diabetes is suspected to increase the rate of collagen glycation. As AGE have been associated with a decrease in the toughness of trabecular and cortical bone [9, 10], it is plausible that a similar mechanism occurs in T1DM.

Rodent models of T1DM are often generated by injection of streptozotocin (STZ), which is specific toxic to insulin-producing pancreatic beta-cells, and leads to hypoinsulinemia and hyperglycemia [11].

Several studies using STZ-injected rodent models have been published in the last decade and reported inconsistent data on the effect of T1DM on bone tissue material properties, i.e.

the mineral and organic component of the bone matrix [12-15]. Previous studies seem suggest that bone microarchitecture deterioration appears early, whilst modification of intrinsic material, as determined by bone toughness, occurs later [16]. Furthermore, an inherent problem with the STZ model is that this compound is often injected around 10 weeks of age, and the general duration of studies is around 10-12 weeks. As such, most of the bone tissue has been formed before the onset of diabetes. To date, the gold standard method to assess the collagen component of the bone matrix is represented by high- performance liquid chromatography (HPLC) that requires prior tissue grinding, reduction and hydrolysis with acid/alkali. The downside of this approach is that HPLC does not account for variable turnover rates and tissue age that can modify the outcome of collagen analysis.

In this regard, vibrational spectroscopy in the mid-infrared (IR) region can provide molecular structure information about mineralized and non-mineralized connective tissue [17-19]. In addition, Fourier-transform IR microspectroscopy (FTIRM), in which the spectrometer is coupled to a light microscope, enables investigators to record spectra at discrete points within thin tissue section. This methodology has several advantages as compared with other approaches, such as X- ray diffraction, nuclear magnetic resonance or HPLC, in (i) allowing the use of the same sample for other investigations (histomorphometry, quantitative backscattered electron imaging, nanoindentation etc...) and

(3)

(ii) enabling the investigator to precisely determine tissue material properties in a specific location of a bone sample (between double tetracycline labels as for example).

Based on IR analyses of single crystals and model compounds, the origin of vibrations observed in the IR have been tabulated for the mineral and protein component of the bone matrix. Several calculated parameters have been validated for the analyses of bone tissue and are reviewed in depth elsewhere [20]. As for example, the 900-1200 cm-1 region, assigned to 13 phosphate vibrations, is often used to derive information regarding the maturity and size of the bone mineral. The 960-1140 cm-1 region also contains information regarding carbohydrate moieties [21]. But the presence of the strong 13 phosphate at the same wavenumber hampers the accurate determination of carbohydrate vibrations. As such, we hypothesized that investigation of the same bone section in a mineralized state (native state) followed by demineralization and re-analysis at the same location may be interesting to ascertain whether tissue material properties are affected in T1DM. Such an approach necessitates the validation of several IR parameters used in other connective tissue but bone, especially for the determination of collagen glycation and collagen integrity.

Therefore, the aims of the present study were to (i) develop a new approach, based on demineralization of thin bone tissue section to more accurately characterize the organic component of the bone matrix by FTIRM, (ii) validate collagen glycation and collagen integrity in bone tissue and (iii) better understand what alterations of tissue material properties occur in T1DM.

MATERIAL AND METHODS

Reagents

All chemicals were purchased from Sigma- Aldrich (Lyon, France) unless otherwise stated.

In vitro glycation of type I collagen

Type I collagen gels were prepared as reported elsewhere [22]. Type I collagen solution (4 mg/ml in 20 mM acetic acid) was diluted, on ice, with 0.1% acetic acid and neutralized with 0.4 M NaOH (pH 10.2) in PBS buffer to form 1.5 mg/ml gels. A 750 µl aliquot of the neutralized collagen solution was pipetted into wells of a 24-well plate and incubated at 37°C overnight to allow for fibril formation. All gels were washed extensively with distilled water prior to a 60-day period of incubation with glucose solution. At the end of the incubation period, collagen gels were dialyzed against milliQ water for a week, and half were deposited onto BaF2 window and allowed to dry to form a uniform film prior to FTIRM measurement. Eight gels per conditions have been analyzed.

In order to assess the glycation level, AGE- specific fluorescence, in the remaining half of collagen solutions, was determined using a M2 microplate reader (Molecular Devices, St Gregoire, France) with excitation and emission wavelengths set at 380 and 440 nm, respectively. The amount of fluorescence has been calibrated using several quinine sulfate concentrations (ranged 0-10 µg/ml). The amount of collagen in each solution was determined by the BCA method as described elsewhere [23]. AGE-specific fluorescence was then expressed as fluorescence intensity, in arbitrary unit/g collagen.

(4)

Animals

A total of 18 young male Swiss TO mice (8 weeks old) were randomly allocated to either the saline-injected control (n=8) or streptozotocin-injected diabetic (STZ-150 mg/kg body weight, n=10) groups. Six days post-STZ injection, diabetes was confirmed by measurement of blood glucose with an automated glucose oxidase procedure using a Beckman glucose analyzer II (Beckman Instruments, Galway, Ireland). Animals were injected intraperitoneally with calcein (10 mg/kg) ten and two days before necropsy.

Sacrifice was then performed 4 weeks post- STZ injection. All experiments were carried out according to UK home office regulations (UK Animals Scientific Procedures act 1986).

Bone tissue processing

At necropsy, femurs were harvested, cleaned of soft tissue and fixed with 70% ethanol for 24 hrs at 4°C and embedded in polymethylmethacrylate at 4°C. This fixation/embedding protocol has previously been successfully employed for FTIRM measurements [24]. Cross-sectional sections (4 µm-thickness) of the femur mid-diaphysis were cut dry on a heavy duty microtome equipped with tungsten carbide knives (Leica Polycut S). Four bone sections were analyzed for each animal. Each section was separated by 15 microns. Sections were first observed with a fluorescence Olympus AX-60 microscope (Olympus France, Rungis, France) with a 40x magnification with either bright field or fluorescence light for the visualization of calcein double labelling (excitation and emission filters set at 490 nm and 520 nm, respectively) and digital photographs were taken with both illuminations. White and

fluorescent pictures were overimposed using Adobe Photoshop CS5 (Adobe, Paris, France) and were used with demineralized bone section only to ensure proper locations of the two calcein lines. Sections were then sandwiched between BaF2 windows and ready for FTIRM data acquisition as detailed below.

FTIRM data acquisition

Spectral acquisitions were obtained on a Bruker Vertex 70 spectrometer (Bruker optics, Ettlingen, Germany) interfaced with a Bruker Hyperion 3000 infrared microscope equipped with a standard single element Mercury Cadmium Telluride (MCT) detector in the range 750-4000cm-1. The Bruker Hyperion 3000 was also equipped with a mercury lamp, a 455 nm excitation filter and a 520 nm emission filter (Bruker). The FTIR spectrometer was continuously purged with dry air from air purifiers. Using the fluorescence set up and a 36X IR Schwarzchild objective (NA=0.5), acquisition of spectrum was conducted with the video measurement mode with a pixel size of 1.1m. In this mode, an image is recorded and markers are manually positioned at the locations were spectra should be acquired and then due to a motorized stage, spectra are recorded at marked locations with an IR beam size of ~7 m at 1650 cm-1. Markers were positioned manually in the center of double calcein labels and were spaced by 20 m. The number of markers per double calcein regions was variable depending on calcein labeling length. At least one spectrum was acquired in each double labeled surface. At the meantime, marker locations were also added on the photoshop-processed images taken above. A minimum of 40 IR spectra was recorded for each sample at a resolution of 4 cm-1, with an

(5)

average of 32 scans in transmission mode with the Opus software (Bruker, release 6.5).

Background spectral images were collected under identical conditions from the same BaF2

windows at the beginning and end of each experiment to ensure instrument stability.

FTIRM data processing

The spectral region 850-1800 cm-1 was used for analysis. Each spectrum, obtained from a pMMA section, was first corrected for polymethylmethacrylate (pMMA) contribution by subtraction of pMMA background after normalization of the peak at 1730 cm-1. The 40 spectra of each sample were baseline corrected with an elastic correction method and vector normalized on the range 1495-1720 cm-1. Spectra were subjected to curve fitting using a commercial available software package (Grams/AI 8.0, Thermofisher scientific, Villebon sur Yvette, France). Briefly, the second derivative spectrum was used to determine the number and the position of the bands constituting every spectral interval.

Spectral curve fitting quality was assessed by a root mean square noise (RMSnoise) set at 1%. RMSnoise represent the variance between the smoothed trace and the original sample spectrum and is calculated as:

𝑅𝑀𝑆𝑛𝑜𝑖𝑠𝑒 = √1

𝑛∑(𝑎𝑖− 𝑝𝑖

𝑛

𝑖=1

where n is the number of data point in the fitted region, a is the true absorbance value and p is the predicted absorbance value determined by the Levenberg-Maquardt (LM) algorithm. Initial trace and second derivative were overimposed

and sub-band were positioned, with a gaussian shape, at minimum intensities observed on the second derivative spectrum. The LM algorithm was run and the position and area of sub- bands were obtained. Evaluated IR spectral parameters are presented Table 1.

Demineralization protocol

Sections were demineralized by immersion in a solution of 0.5 M ethylenediaminetetraacetic acid (EDTA) in Tris buffer pH 7.4 for 7 days.

The EDTA solution was replenished at day 3.

At the end of the 7-day period, sections were rinsed 8 times with Tris solution followed by 6 washes with milliQ water. Sections were then allowed to dry at 37°C for 48 hrs. Sections were then sandwiched between BaF2 windows and reexamined by FTIRM. As calcein labels were no longer present after demineralization, we used the photoshop-processed image to accurately determine the location of FTIR markers in the video measurement mode.

Measurements were then performed as described above.

Enzymatic treatment

Demineralized sections of control animals, already examined by FTIRM, were incubated in 1 mg/ml type IA-S collagenase or 0.25 mg/ml trypsin (Lonza, Verviers, Belgium) at 37°C for 10 min. Sections were then rinsed 6 times in milliQ water and allowed to dry at 37°C for 48 hrs. prior to FTIRM reexamination.

Quantitative backscattered electron imaging (qBEI)

Quantitative backscattered electron imaging was employed to determine the bone mineral density distribution (BMDD) of the section before and after demineralization [25].

Sections were observed with a scanning

(6)

electron microscope (EVO LS10, Carl Zeiss Ltd, Nanterre, France) equipped with a five quadrants semi-conductor backscattered electron detector and operated at 20 keV with a probe current of 250 pA and a working distance of 15 mm. The backscattered signal was calibrated using pure carbon (Z=6, mean grey level = 25), pure aluminum (Z=13, mean grey level =225) and pure silicon (Z=14, mean grey level =253) standards (Micro-analysis Consultants Ltd, St Ives, UK). Analysis was performed as previously reported [26]. Camean, representing the mean calcium concentration, was determined. The composition of the bone section was investigated by energy-dispersive X ray analysis (EDX) using an Inca Xmax device (Oxford Instruments, Oxford, UK). The detection limit of this EDX system was of 0.01% atomic percentage.

Statistical analysis

Results were expressed as mean ± standard error of the mean (SEM). Student's t-test with Levene’s test of homogeneity was used to compare the differences between the CTRL and STZ groups using the Systat statistical software release 13.0 (Systat software Inc., San Jose, CA). Differences observed in in vitro assays were assessed using Levene's test followed by ANOVA and bonferroni post- hoc test. Differences at p<0.05 were considered significant.

RESULTS

Demineralization efficiency

In order to validate our demineralization protocol, we first looked at the mineral presence by qBEI. As shown in Figure 2A, EDTA treatment led to a dramatic reduction of signal, suggesting demineralization. Indeed,

EDX analysis confirmed that calcium concentration was drastically reduced and phosphorus below the detection limit.

Furthermore, Camean, that represents the mean mineral concentration, decreased to 0 after EDTA treatment suggesting a mineralization state equivalent to osteoid tissue and as such the absence of mineral (Fig. 2B). FTIRM spectra recorded before and after EDTA treatment revealed dramatic changes (Fig.

2C). Firstly, the large band observed at 900- 1200 cm-1, due to phosphate 1,3 band, disappeared as a consequence of EDTA treatment, confirming demineralization.

Secondly, the band located at ~870 cm-1, representing 2 carbonate vibration, is no longer present in the demineralized bone section. Thirdly, the band located at 1350-1500 cm-1, representative of 3 carbonate and CH2 wagging, was dramatically modified by demineralization, to reveal two peaks located respectively at 1451 cm-1 and 1400 cm-1 and corresponding to CH2 and CH3 wagging of collagen side chain. The band centered at 1338 cm-1, representing the vibration of amino acid side chain, was better resolved in demineralized sections.

Assessment of the extent of collagen glycation in collagen gel

Next, to study collagen glycation and identification of suitable peak to measure glycation, we used collagen gels that were incubated in glucose solution. As depicted in Figure 3A, the main consequence of incubation with 25 mM glucose was a large band appearing between 960-1140 cm-1 on the FTIR spectra and representative of C-O-C, C-OH and C-C ring vibrations. Curve fitting of this region, based on second derivative analysis,

(7)

revealed the presence of seven underlying bands located at 995 cm-1, 1012 cm-1, 1032 cm-1, 1049 cm-1, 1064 cm-1, 1082 cm-1 and 1101 cm-1 (Fig. 3B). In order to ascertain whether one of these bands could be used to monitor collagen glycation, we looked at their respective evolution in the presence of 0, 25 and 50 mM glucose. The subband located at 1032 cm-1 was the biggest (39.1%) in the region 960-1180 cm-1 at a glucose concentration of 50 mM (Table 2).

Furthermore, the 1032 cm-1 subband was the only subband that showed an increase over the three tested glucose concentrations (Table 2). As such, we decided to use this subband for determination of collagen glycation.

Validation of collagen glycation and collagen integrity parameters

Collagen glycation was determined by calculating the area ratio of the 1032 cm-1 subband over the Amide I region (Fig. 4A).

This ratio augmented significantly in a dose- dependent manner by 10- and 34-fold with 25 and 50 mM glucose incubations, respectively.

Collagen glycation was also determined by assessing the fluorescence emission at 440 nm (Fig. 4B). Similarly, the fluorescence intensity (arbitrary unit/µg collagen) was significantly and dose-dependently augmented by 5- and 12-fold with 25 and 50 mM glucose incubations, respectively. These two methodologies examine collagen glycation and were significantly correlated with a R2 value of 0.9992 (p=0.013). However, no significant modification of collagen secondary structure was evidenced after glycation (Fig. 4C) although a sub-band located at ~1674 cm-1 was present in 5 of the 8 collagen gels incubated with 50mM glucose.

In order to assess whether collagen integrity was affected by collagen glycation, we measured this parameter in the presence of 0, 25 and 50 mM glucose. Collagen integrity was unaffected by collagen glycation (Fig. 4D). We then assessed collagen integrity in demineralized bone section of control animals before and after enzymatic treatment. The ratio 1338 cm-1/Amide II was significantly reduced after treatment with collagenase but not with trypsin (p=0.008 and p=0.540 respectively, Fig.

4E). Secondary structure of bone tissue was dramatically altered after collagenase treatment with significant augmentations in - turn and -helix and significant reductions in unordered and triple helix (Fig. 4F). On the other hand, trypsin did not significantly alter tissue secondary structure.

T1DM did not alter the mineral component of the bone matrix but increased collagen maturity

Structural parameters were assessed by FTIR in mineralized sections of control and diabetic mice. No major difference was evidenced in FTIRM spectra (Fig. 5A). The degree of mineralization, carbonate substitution and mineral crystallinity were not significantly different between the two groups of animal (Fig. 5B-D). On the other hand, collagen maturity was significantly increased by 17%

(p=0.0048) in diabetic animals as compared to controls (Fig. 5E).

T1DM resulted in dramatic alterations of collagen structural feature

Structural collagen parameters were also investigated in demineralized bone sections of control and diabetic animals. Again, no major differences in spectral features were observed

(8)

between the two animal groups, except in the region 1000-1050 cm-1, with an increase in diabetic mice (Fig. 6A). Collagen glycation was significantly higher in diabetic mice by 99%

(p=0.0121, Fig. 6B). Collagen integrity was significantly lower in diabetic animals by almost 170% (p=0.0121, Fig. 6C). Interestingly, we observed significant modifications in tissue secondary structure in diabetic mice with a significant higher amount of -turn (2-fold, p<0.001) and significant reductions in unordered (3.8-fold, p=0.018) and triple helix (4.25-fold, p<0.001) (Fig. 6D).

DISCUSSION

Worldwide, the number of people who develop T1DM is increasing. Whilst living with T1DM is manageable with diet, insulin therapy, and exercise, diabetic complications still occur.

Among them, bone fracture represents a severe complication that greatly alters the quality of life. Despite the need to prevent fracture occurrence, there is a paucity of mechanistic information on how hypoinsulinemia and hyperglycemia associated with T1DM affect bone tissue, and especially tissue material properties. In the present study, an innovative FTIRM analysis of bone tissue in a mineralized state was used followed by analysis at the same location upon demineralization. We validated several FTIRM parameters (collagen glycation, collagen integrity and tissue secondary structure) that were useful in assessing structural modifications of the bone tissue. Furthermore, we demonstrated a significant augmentation of collagen glycation in diabetic bone tissue, and also collagen denaturation (reduction in

collagen integrity and breakdown of the triple helix) that could potentially alter bone toughness.

First of all, demineralization of bone sections led to modifications of spectral feature with biggest modifications observed in the 1500-1350 cm-1 and 1200-900 cm-1 regions. In mineralized section, the 1500-1350 cm-1 region was assigned to 3 carbonate and CH2 wagging. Demineralization led to removal of carbonate moieties as evidenced by the absence of the ~870 cm-1 2 carbonate band.

It is legitimate then to assume that the modifications of the 1500-1350 cm-1 observed in demineralized section also reflect the absence of the 3 carbonate vibration and allows a better resolution of underlying peak representative of CH2 and CH3 wagging of collagen side chains. Demineralization of bone sections also highlighted changes in the region 1200-900 cm-1 with the removal of 31 phosphate vibrations and the apparition of underlying peak assigned to carbohydrate moieties. However, prior to the assignment of any demineralized sub-bands to collagen properties, several validations were required.

In vitro glycation of type I collagen gel demonstrated that the relative area of the 1032 cm-1 subband of the carbohydrate region was dose-dependently augmented during glycation of collagen. This correlated with the fluorometric assessment of collagen glycation, a methodology validated for the assessment of AGE in collagen matrix. Taken together, these data suggest that the 1032 cm-1 subband might be useful in determining collagen glycation in demineralized bone tissue. However, before further validation is made to fully ascertain that the 1032/amide I ratio is a marker of AGE

(9)

formation, this parameter should be considered as only indicative of collagen glycation. One might wonder why we did not choose the entire carbohydrate region (960-1140 cm-1) or any of the other subbands as markers of collagen glycation. As evidenced in the present study, the carbohydrate region (960-1140 cm-1) is composed of seven underlying bands. Some of these subbands (995 cm-1, 1012 cm-1, 1049 cm-1, 1064 cm-1 and 1101 cm-1) were reduced after trypsin treatment (data not shown), suggesting that these subbands were generated by non-collagenous proteins.

Indeed, this region has been proposed in other connective tissue as a marker of proteoglycan content [27]. The 1064 cm-1 band was recently reported by Rieppo and coworkers as a marker of sulfated proteoglycan in cartilage [21].

These authors showed that enzymatic removal of proteoglycan dramatically affected this peak.

The extracellular bone matrix contains several proteoglycans and, as such, assignment of this peak in bone tissue is more difficult. The 1082 cm-1 subband can be assigned to the C-O stretching vibration of carbohydrate but also to the symmetric PO2 stretching vibration [28].

The extracellular matrix of the bone tissue contains several phosphorylated proteins (siblings, fibronectin, vitronectin, etc…) that could potentially influence the area of the 1082 cm-1 subband and although phosphorus was not identified with EDX analysis (detection limit of 0.01 % atomic percent), we cannot rule out the presence of phosphorus trace in the organic matrix that could have modified the 1082 cm-1 subband. Furthermore, Rieppo and co-workers did not observe dramatic alteration of the 1032 cm-1 peak upon enzymatic removal of proteoglycan, suggesting that this peak, although located in the carbohydrate region,

might be an indicator of collagen glycation rather than glycosaminoglycan [21]. In our study, no reduction of the 1032 cm-1 peak was observed upon trypsin digestion, suggesting that this peak could not be assigned to non- collagenous proteins and/or proteoglycans.

Recently, Guilbert et al., reported in an elegant spectroscopic study that the intensity ratio of the 1032 cm-1 peak over the amide I peak was increased upon collagen glycation, supporting the use of the 1032 cm-1 band as a marker of collagen glycation [29].

In the current study, we also introduced and validated a parameter called collagen integrity. In earlier FTIR studies, absorbance of the 1338 cm-1 band was shown to decrease in intensity as collagen denatures [28]. The 1338 cm-1 band is assigned to the CH2 wagging vibration of collagen amino acid side chains [28]. The 1338 cm-1/amide II ratio was first introduced by West et al. and these authors evidenced a smaller ratio in osteoarthritic joint as compared to normal [30].

Further work to elucidate the molecular origin of this change confirmed that reduction in the 1338 cm-1 absorbance was linked to collagen degradation [31]. In the present study, we reported similar finding in demineralized bone where the use of collagenase significantly reduced collagen integrity. On the other hand, neither glycation of type I collagen nor trypsin treatment altered this parameter. This suggests that collagen integrity in bone tissue is a marker of collagen degradation.

Protein secondary structure was also determined by second derivative and peak fitting of the amide I region as previously

(10)

reported [32, 33]. For collagen gels, the amide I region reflects the secondary structure of type I collagen. However, the bone matrix is a mixed between type I collagen (~90%) and non-collagenous proteins (~10%). As such, information obtained from the amide I region of bone tissue reflects the overall secondary structure of this tissue. Nevertheless, collagen is distinct from other proteins in that this protein comprises three polypeptide chains (- chains) which assemble together to form a triple helix with a characteristic band at ~1638 cm-1 [34]. In the present study, tissue secondary structure was dramatically altered by collagenase but not by trypsin. We failed to evidence any significant alteration of collagen secondary structure upon glycation or trypsin treatment although a sub-band located at

~1674 cm-1 was evidenced in 5 out of 8 collagen gels incubated with 50 mM glucose.

However, collagenase treatment led to a dramatic change in the bone tissue secondary structure with a significant alteration of the collagen matrix as represented by the decrease in the triple helix.

The mineral component of the bone matrix seemed unaffected in T1DM, despite previous reports of lower mineralization degree in STZ-induced T1DM [15, 35]. However, in these publications, the study period post-STZ injection as well as the STZ dose were different as compare to the present study, and it is plausible that mineralization default arises from this difference. Another possible explanation could reside in the mouse strain. In the current study, we used Swiss TO mice based on the fact that the extent of beta-cells destruction in the pancreas, in response to STZ injection, is similar to what is observed in

humans suffering of T1DM. However, the spectral signature of the phosphate band appeared peculiar and the absence of mineral modifications might only be valid in this animal model. On the other hand, collagen maturity, determined on mineralized bone sections, was significantly increased in the diabetic mice.

Previously, Saito et al, reported a significant reduction in immature but not in mature collagen crosslinks in the WBN/Kob diabetic rat model [36]. As collagen maturity is calculated as the ratio of the 1660 cm-1/1690 cm-1 subbands, respectively assigned to mature and immature collagen cross-links [37], one could expect an increase in collagen maturity in diabetic animals as observed in the present study. However, at the present we have no data that could suggest that collagen glycation and AGE formation could affect the FTIR-determined collagen maturity.

Nevertheless, the most notable difference in tissue material properties was observed for the collagen component. As expected, T1DM led to a dramatic and significant 2-fold increase in collagen glycation.

Previous studies have also reported a higher AGE accumulation in the bone matrix of diabetic animals supporting our results [14, 36]. Collagen integrity was significantly lower in diabetic animals and accompanied by profound changes in tissue secondary structure. As reported above, collagen integrity was not significantly affected by trypsin treatment, suggesting that the 1338 cm-1 vibration is strictly related to collagen amino acid side- chain. Reduction of collagen integrity and secondary structure (fall in triple helix) suggest a denaturation of type I collagen in T1DM.

These modifications cannot be explained by

(11)

the augmented collagen glycation but rather by enzymatic degradation of matrix collagen.

Interestingly, further reports have been made regarding higher matrix metalloproteinase (MMP) activities, mainly MMP-2, MMP-8, MMP-9, MMP-13 and MMP-14, in connective tissues, other than bone, in type 1 diabetes [38-42]. To the best of our knowledge, MMP activities have never been assessed in bone tissue of diabetic animals. However, it is plausible that MMP activities could be increased in T1DM due to the chronic inflammation state, and this could potentially participate to the reduced collagen integrity by cleaving the triple helix. Furthermore, all FTRIM measurements have been performed between double calcein labelling, suggesting that the observed alterations of collagen were not due to a difference in tissue age and that they appeared fairly quickly after the onset of diabetes. As such, it is reasonable to assume that the observed reduction in bone toughness seen in other rodent models of STZ-induced

T1DM could be due to modification of the collagen component of the bone matrix as evidenced in the present study.

In conclusion, in the present study, we highlighted that investigation of the bone matrix in a demineralized state brings important structural feature of the bone tissue. Indeed, several alterations of collagen quality (collagen glycation, integrity and secondary structure) in the newly-forming bone matrix of diabetic animals have been observed and could lead to the observed reduction in bone toughness.

Further studies are now required to demonstrate whether these findings are transposable to bone tissue of individuals affected by T1DM.

ACKNOWLEDGEMENTS

This work was supported by grants from the University of Angers and the University of Ulster Research Challenge Funding Programme.

REFERENCES

[1] Kemink SA, Hermus AR, Swinkels LM, Lutterman JA, Smals AG. Osteopenia in insulin- dependent diabetes mellitus; prevalence and aspects of pathophysiology. J Endocrinol Invest 2000; 23:295-303.

[2] Krakauer JC, McKenna MJ, Buderer NF, Rao DS, Whitehouse FW, Parfitt AM. Bone loss and bone turnover in diabetes. Diabetes 1995; 44:775-82.

[3] Forsen L, Meyer HE, Midthjell K, Edna TH. Diabetes mellitus and the incidence of hip fracture:

results from the Nord-Trondelag Health Survey. Diabetologia 1999; 42:920-5.

[4] Vestergaard P, Rejnmark L, Mosekilde L. Diabetes and its complications and their relationship with risk of fractures in type 1 and 2 diabetes. Calcif Tissue Int 2009; 84:45-55.

[5] Tuominen JT, Impivaara O, Puukka P, Ronnemaa T. Bone mineral density in patients with type 1 and type 2 diabetes. Diabetes Care 1999; 22:1196-200.

[6] Danielson KK, Elliott ME, LeCaire T, Binkley N, Palta M. Poor glycemic control is associated with low BMD detected in premenopausal women with type 1 diabetes. Osteoporos Int 2009;

20:923-33.

(12)

[7] Strotmeyer ES, Cauley JA, Schwartz AV, Nevitt MC, Resnick HE, Bauer DC, et al.

Nontraumatic fracture risk with diabetes mellitus and impaired fasting glucose in older white and black adults: the health, aging, and body composition study. Arch Intern Med 2005;

165:1612-7.

[8] Strotmeyer ES, Cauley JA. Diabetes mellitus, bone mineral density, and fracture risk. Curr Opin Endocrinol Diabetes Obes 2007; 14:429-35.

[9] Tang SY, Allen MR, Phipps R, Burr DB, Vashishth D. Changes in non-enzymatic glycation and its association with altered mechanical properties following 1-year treatment with risedronate or alendronate. Osteoporos Int 2009; 20:887-94.

[10] Tang SY, Zeenath U, Vashishth D. Effects of non-enzymatic glycation on cancellous bone fragility. Bone 2007; 40:1144-51.

[11] Szkudelski T. The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas. Physiol Res 2001; 50:537-46.

[12] Reddy GK, Stehno-Bittel L, Hamade S, Enwemeka CS. The biomechanical integrity of bone in experimental diabetes. Diabetes Res Clin Pract 2001; 54:1-8.

[13] Zhang SQ, Chen GH, Lu WL, Zhang Q. Effects on the bones of vanadyl acetylacetonate by oral administration: a comparison study in diabetic rats. J Bone Miner Metab 2007; 25:293- 301.

[14] Silva MJ, Brodt MD, Lynch MA, McKenzie JA, Tanouye KM, Nyman JS, et al. Type 1 diabetes in young rats leads to progressive trabecular bone loss, cessation of cortical bone growth, and diminished whole bone strength and fatigue life. J Bone Miner Res 2009; 24:1618-27.

[15] Facchini DM, Yuen VG, Battell ML, McNeill JH, Grynpas MD. The effects of vanadium treatment on bone in diabetic and non-diabetic rats. Bone 2006; 38:368-77.

[16] Nyman JS, Even JL, Jo CH, Herbert EG, Murry MR, Cockrell GE, et al. Increasing duration of type 1 diabetes perturbs the strength-structure relationship and increases brittleness of bone.

Bone 2011; 48:733-40.

[17] Boskey A, Mendelsohn R. Infrared analysis of bone in health and disease. J Biomed Opt 2005; 10:031102.

[18] Carden A, Morris MD. Application of vibrational spectroscopy to the study of mineralized tissues (review). J Biomed Opt 2000; 5:259-68.

[19] Miller LM, Dumas P. Chemical imaging of biological tissue with synchrotron infrared light.

Biochim Biophys Acta 2006; 1758:846-57.

[20] Boskey A, Pleshko Camacho N. FT-IR imaging of native and tissue-engineered bone and cartilage. Biomaterials 2007; 28:2465-78.

[21] Rieppo L, Saarakkala S, Narhi T, Helminen HJ, Jurvelin JS, Rieppo J. Application of second derivative spectroscopy for increasing molecular specificity of Fourier transform infrared spectroscopic imaging of articular cartilage. Osteoarthritis Cartilage 2012; 20:451-9.

[22] Roy R, Boskey A, Bonassar LJ. Processing of type I collagen gels using nonenzymatic glycation. J Biomed Mater Res A 2010; 93:843-51.

(13)

[23] Mabilleau G, Chappard D, Sabokbar A. Role of the A20-TRAF6 axis in lipopolysaccharide- mediated osteoclastogenesis. J Biol Chem 2011; 286:3242-9.

[24] Aparicio S, Doty SB, Camacho NP, Paschalis EP, Spevak L, Mendelsohn R, et al. Optimal methods for processing mineralized tissues for Fourier transform infrared microspectroscopy.

Calcif Tissue Int 2002; 70:422-9.

[25] Roschger P, Fratzl P, Eschberger J, Klaushofer K. Validation of quantitative backscattered electron imaging for the measurement of mineral density distribution in human bone biopsies.

Bone 1998; 23:319-26.

[26] Mabilleau G, Mieczkowska A, Irwin N, Flatt PR, Chappard D. Optimal bone mechanical and material properties require a functional glucagon-like peptide-1 receptor. J Endocrinol 2013;

219:59-68.

[27] Camacho NP, West P, Torzilli PA, Mendelsohn R. FTIR microscopic imaging of collagen and proteoglycan in bovine cartilage. Biopolymers 2001; 62:1-8.

[28] Jackson M, Choo LP, Watson PH, Halliday WC, Mantsch HH. Beware of connective tissue proteins: assignment and implications of collagen absorptions in infrared spectra of human tissues. Biochim Biophys Acta 1995; 1270:1-6.

[29] Guilbert M, Said G, Happillon T, Untereiner V, Garnotel R, Jeannesson P, et al. Probing non- enzymatic glycation of type I collagen: a novel approach using Raman and infrared biophotonic methods. Biochim Biophys Acta 2013; 1830:3525-31.

[30] West PA, Bostrom MP, Torzilli PA, Camacho NP. Fourier transform infrared spectral analysis of degenerative cartilage: an infrared fiber optic probe and imaging study. Appl Spectrosc 2004; 58:376-81.

[31] West PA, Torzilli PA, Chen C, Lin P, Camacho NP. Fourier transform infrared imaging spectroscopy analysis of collagenase-induced cartilage degradation. J Biomed Opt 2005;

10:14015.

[32] Byler DM, Susi H. Examination of the secondary structure of proteins by deconvolved FTIR spectra. Biopolymers 1986; 25:469-87.

[33] Pelton JT, McLean LR. Spectroscopic methods for analysis of protein secondary structure.

Anal Biochem 2000; 277:167-76.

[34] Brodsky B, Ramshaw JA. The collagen triple-helix structure. Matrix Biol 1997; 15:545-54.

[35] Hamada Y, Kitazawa S, Kitazawa R, Fujii H, Kasuga M, Fukagawa M. Histomorphometric analysis of diabetic osteopenia in streptozotocin-induced diabetic mice: a possible role of oxidative stress. Bone 2007; 40:1408-14.

[36] Saito M, Fujii K, Mori Y, Marumo K. Role of collagen enzymatic and glycation induced cross- links as a determinant of bone quality in spontaneously diabetic WBN/Kob rats. Osteoporos Int 2006; 17:1514-23.

[37] Paschalis EP, Verdelis K, Doty SB, Boskey AL, Mendelsohn R, Yamauchi M. Spectroscopic characterization of collagen cross-links in bone. J Bone Miner Res 2001; 16:1821-8.

[38] Ning R, Chopp M, Yan T, Zacharek A, Zhang C, Roberts C, et al. Tissue plasminogen activator treatment of stroke in type-1 diabetes rats. Neuroscience 2012; 222:326-32.

(14)

[39] Ryan ME, Ramamurthy NS, Sorsa T, Golub LM. MMP-mediated events in diabetes. Ann N Y Acad Sci 1999; 878:311-34.

[40] Silva JA, Ferrucci DL, Peroni LA, Abrahao PG, Salamene AF, Rossa-Junior C, et al.

Sequential IL-23 and IL-17 and increased Mmp8 and Mmp14 expression characterize the progression of an experimental model of periodontal disease in type 1 diabetes. J Cell Physiol 2012; 227:2441-50.

[41] Symeonidis C, Papakonstantinou E, Galli A, Tsinopoulos I, Mataftsi A, Batzios S, et al. Matrix metalloproteinase (MMP-2, -9) and tissue inhibitor (TIMP-1, -2) activity in tear samples of pediatric type 1 diabetic patients: MMPs in tear samples from type 1 diabetes. Graefes Arch Clin Exp Ophthalmol 2013; 251:741-9.

[42] Takahashi N, Takasu S. A close relationship between type 1 diabetes and vitamin A- deficiency and matrix metalloproteinase and hyaluronidase activities in skin tissues. Exp Dermatol 2011; 20:899-904.

(15)

TABLES

Table 1: FTIRM parameters

Parameter Definition

Mineral phase

Degree of mineralization Area of 900-1200 cm-1 phosphate band/area of amide I band (1585-1720 cm-1) Carbonate substitution Area of 850-890 cm-1 band/area of phosphate band

Mineral crystallinity Area of 1030 cm-1 subband/area of 1020 cm-1 subband Protein phase

Collagen maturity Area of 1660 cm-1 subband/area of 1690 cm-1 subband

Collagen integrity Area of 1338 cm-1 subband/area of amide II band (1585-1495 cm-1). Reduction in this parameter suggests collagen denaturation.

Tissue secondary structure Relative area of subbands located at 1694 cm-1 (-turn), 1681 cm-1 (-sheet), 1669 cm-1 (-turn), 1656 cm-1 (-helix), 1647 cm-1 (unordered), 1638 cm-1 (triple helix) and 1626 cm-1 (-sheet).

Collagen glycation Area of 1032 cm-1 subband/area of amide I band Parameters in italic can only be assessed after demineralization of the bone matrix.

Table 2: Relative contribution in % of underlying band in the 965-1180 cm-1 region. *: p<0.05 vs.

previous glucose concentration

Glucose concentration (mM)

Underlying band (cm-1) 0 25 50

995 2.6 ± 0.3 7.6 ± 1.2 * 5.0 ± 0.9

1012 13.8 ± 2.0 4.6 ± 1.9 * 4.8 ± 1.6

1032 13.3 ± 2.2 28.0 ± 0.8 * 39.1 ± 1.0 *

1049 5.6 ± 2.0 12.8 ± 4.3 * 9.6 ± 3.1

1064 16.9 ± 2.7 17.4 ± 2.0 12.8 ± 2.1

1082 40.4 ± 2.5 8.5 ± 0.7 * 12.2 ± 1.2

1101 7.4 ± 2.1 21.2 ± 7.3 * 16.5 ± 5.7

(16)

Figure 1: Schematic representation of the experimental design.

(17)

Figure 2: Control of bone section demineralization. (A) Bone sections were imaged by qBEI before and after EDTA treatment. The corresponding EDX analysis showed the absence of phosphorus and drastic reduction in calcium after treatment. (B) Camean, representative of the mean mineral content, is dropped to 0 after EDTA treatment. (C) FTIRM spectra of mineralized (black line) and demineralized (grey line) bone section. ***: p<0.001 vs. before treatment

(18)

Figure 3: In vitro glycation of type I collagen gels. (A) FTIRM spectra of control collagen gel (grey line) and after incubation with 25 mM glucose. (B) Curve fitting of the 960-1140 cm-1 region, revealing seven underlying band centered at 995 cm-1, 1012 cm-1, 1032 cm-1, 1049 cm-1, 1064 cm-1, 1082 cm-1 and 1101 cm-1.

(19)
(20)

Figure 4: Validation of demineralized parameters. Collagen glycation was determined by (A) FTIRM and (B) fluorescence readings at 440 nm. (C) Collagen structure was assessed by curve fitting of the amide I region in the presence of 0mM, 25mM and 50mM glucose. (D) Collagen integrity was determined in the presence of 0mM, 25mM and 50mM glucose. (E) Collagen integrity was computerized before and after enzymatic treatment. (F) Tissue secondary structure was tabulated after enzymatic treatment. *: p<0.05, **: p<0.01 and ***:

p<0.001 vs. no glucose. FTIRM parameters are defined Table 1.

(21)

Figure 5: Investigation of mineralized FTIRM parameters. (A) Example of FTIRM spectrum of control (black line) and diabetic (grey line) mice. (B) Degree of mineralization, (C) carbonate substitution and (D) mineral crystallinity were not significantly different between control (CTRL) and diabetic (STZ) mice. (E) Collagen maturity was significantly higher in diabetic mice. **: p<0.01 vs. CTRL mice.

(22)

Figure 6: Investigation of demineralized FTIRM parameters. (A) Example of FTIRM spectrum of demineralized bone section of control (black line) and diabetic (grey line) mice. (B) Collagen glycation and (C) collagen integrity were significantly different in diabetic mice.

(D) Tissue secondary structure was modified in diabetic mice as evidenced by higher value for -turn and lower value for unordered and triple helix. *: p<0.05 and ***: p<0.001 vs control mice.

Références

Documents relatifs

These novel 3D quantitative results regarding the osteocyte lacunae and micro-cracks properties in osteonal and interstitial tissue highlight the major role

Signi fi cant di ff erences were found between frontal and parietal bone for e ff ective failure stress and failure strain, and superior and inferior aspect of calvarium for bone

In this study, hydroxyapatite was developed using the wet precipitation method from hydrated calcium chloride (CaCl2,12H2O) as a source of calcium and di-sodium hydrogen

In this study, the mechanical and microstructural properties of the synthetic hydroxyapatite, with Ca/p stoichiometry equal to 1.67, were determined as a function of

Some GLP-1 receptor agonist (GLP-1RA) have been approved for the treatment of type 2 diabetes mellitus and although clinical trials may not have been de- signed to investigate

While no difference in the 1660/1690 cm 21 area ratio was found between control and lathyritic bones, a difference in this ratio was shown depending on the location in the bone, with

Figure 1: Successive steps of our multiscale modeling approach: (0) elementary nanoscale components; (1) wet collagen composite formed by a collagen molecules matrix containing

Based on this finding, two optimization approaches are devised to find the geometrical parameters of the artificial microstructure that better mimics the elastic response of