Monthly Archives: September 2012

Properties And Usefulness Of Aggregates Of Synovial Mesenchymal Stem Cells As A Source For Cartilage Regeneration

Found from the website Arthritis Research & Therapy

[Note: Just read the very first beginning part where everything is summarized in the abstract, introduction, results, and conclusion. This article is very long ]

Me: So the study was mainly done to see how to grow articular cartilage, not the type found in the growth plates exactly. 

 

Properties and usefulness of aggregates of synovial mesenchymal stem cells as a source for cartilage regeneration

Shiro Suzuki1, Takeshi Muneta1,2, Kunikazu Tsuji2, Shizuko Ichinose3, Hatsune Makino4, Akihiro Umezawa4 and Ichiro Sekiya5*

  • *
    Corresponding author: Ichiro Sekiya sekiya.orj@tmd.ac.jp

Author Affiliations

1Section of Orthopedic Surgery, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan

Global Center of Excellence Program, International Research Center for Molecular Science in Tooth and Bone Diseases, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan

Instrumental Analysis Research Center, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan

Department of Reproductive Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan

5 Section of Cartilage Regeneration, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan

For all author emails, please log on.

Arthritis Research & Therapy 2012, 14:R136 doi:10.1186/ar3869

The electronic version of this article is the complete one and can be found online at: http://arthritis-research.com/content/14/3/R136

Received: 24 November 2011
Revisions received: 30 April 2012
Accepted: 7 June 2012
Published: 7 June 2012

© 2012 Suzuki et al.; licensee BioMed Central Ltd.

This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Introduction

Transplantation of mesenchymal stem cells (MSCs) derived from synovium is a promising therapy for cartilage regeneration. For clinical application, improvement of handling operation, enhancement of chondrogenic potential, and increase of MSCs adhesion efficiency are needed to achieve a more successful cartilage regeneration with a limited number of MSCs without scaffold. The use of aggregated MSCs may be one of the solutions. Here, we investigated the handling, properties and effectiveness of aggregated MSCs for cartilage regeneration.

Methods

Human and rabbit synovial MSCs were aggregated using the hanging drop technique. The gene expression changes after aggregation of synovial MSCs were analyzed by microarray and real time RT-PCR analyses. In vitro and in vivo chondrogenic potential of aggregates of synovial MSCs was examined.

Results

Aggregates of MSCs cultured for three days became visible, approximately 1 mm in diameter and solid and durable by manipulation; most of the cells were viable. Microarray analysis revealed up-regulation of chondrogenesis-related, anti-inflammatory and anti-apoptotic genes in aggregates of MSCs. In vitro studies showed higher amounts of cartilage matrix synthesis in pellets derived from aggregates of MSCs compared to pellets derived from MSCs cultured in a monolayer. In in vivo studies in rabbits, aggregates of MSCs could adhere promptly on the osteochondral defects by surface tension, and stay without any loss. Transplantation of aggregates of MSCs at relatively low density achieved successful cartilage regeneration. Contrary to our expectation, transplantation of aggregates of MSCs at high density failed to regenerate cartilage due to cell death and nutrient deprivation of aggregates of MSCs.

Conclusions

Aggregated synovial MSCs were a useful source for cartilage regeneration considering such factors as easy preparation, higher chondrogenic potential and efficient attachment.

Introduction

Synovial mesenchymal stem cells (MSCs) are an attractive cell source for cartilage regeneration because of their high expansion and chondrogenic potentials [1-5]. We previously reported that more than 60% of synovial mesenchymal stem cells placed on osteochondral defects adhered to the defect within 10 minutes and promoted cartilage regeneration [6,7]. With this local adherent technique, we can transplant synovial MSCs without scaffold. One of the disadvantages in this method is that the cell component in the suspension is invisible to the naked eye.

One of the solutions for this problem is to make aggregates of synovial MSCs [8-10]. This could enable MSCs not only to be visible but also to be heavier. Consequently, aggregates of MSCs will sink faster in the suspension medium than dispersed MSCs. The use of aggregates of MSCs may help to avoid loss of MSCs from targeted cartilage defects and improve the procedures of transplantation of synovial MSCs. However, there are still concerns; properties of synovial MSCs will be altered when synovial MSCs are aggregated. We do not know whether aggregates of MSCs adhere on the cartilage defect as we expect it will, and the proper number of aggregates is unclear.

In this study, properties of aggregates of human synovial MSCs were analyzed from the standpoints of morphology, gene profile and in vitro chondrogenic potential. Also, the effect of transplantation of aggregates of synovial MSCs was investigated in a rabbit cartilage defect model in terms of aggregate number, cell behavior and influential factors in the in vivo chondrogenesis of aggregates of synovial MSCs. Finally, we demonstrated the usefulness of aggregates of synovial MSCs as a source for cartilage regeneration therapy.

Materials and methods

Isolation and culture of human synovial MSCs

This study was approved by an institutional review board of Tokyo Medical and Dental University (No.1030), and informed consent was obtained from all subjects. Human synovium was harvested from donors during anterior cruciate ligament reconstruction surgery for ligament injury and digested in a 3 mg/ml collagenase D solution (Roche Diagnostics, Mannheim, Germany) in α-minimal essential medium (αMEM) (Invitrogen, Carlsbad, CA, USA) at 37°C. After three hours, digested cells were filtered through a 70 μm nylon filter (Becton, Dickinson and Company, Franklin Lakes, NJ, USA), and the remaining tissues were discarded. The digested cells were plated in a 150 cm2 culture dish (Nalge Nunc International, Rochester, NY, USA) in complete culture medium (CCM): αMEM containing 10% fetal bovine serum (FBS; Invitrogen), 100 units/ml penicillin (Invitrogen), 100 μg/ml streptomycin (Invitrogen), and 250 ng/ml amphotericin B (Invitrogen) and incubated at 37°C with 5% humidified CO2. The medium was changed to remove nonadherent cells one day later and cultured for 14 days as passage 0, then replated at 100 cells/cm2 in a 150 cm2 culture dish, cultured for 14 days and cryopreserved as passage 1. To expand the cells, a frozen vial of the cells was thawed, plated in 60 cm2 culture dishes, and incubated for four days in the recovery plate. These cells were replated at 100 cells/cm2 in a 150 cm2 culture dish (passage 3), and cultured for an additional 14 days. These passage 3 cells were harvested and used in this study.

Isolation and culture of rabbit synovial MSCs

This study was approved by the Animal Experimentation Committee of Tokyo Medical and Dental University (No.0120296A). Wild type skeletally mature Japanese White Rabbit and GFP transgenic rabbits [11,12] were anesthetized with an intramuscular injection of 25 mg/kg ketamine hydrochloride and with an intravenous injection of 45 mg/kg sodium pentobarbital and 150 μg/kg medetomidine hydrochloride. Synovium was harvested aseptically from knee joints of the rabbits, and digested in a 3 mg/ml collagenase type V in aMEM for three hours at 37°C. The digested cells were plated at 5 × 104 cells/cm2 in a 150 cm2 culture dish in CCM and incubated at 37°C with 5% humidified CO2. The medium was changed to remove nonadherent cells one day later and cultured for seven days as passage 0. The cells were then trypsinized, harvested and resuspended to be used for further assays. The cells that were transplanted in animals to be sacrificed at Day 0 and Day 14 were labeled for cell tracking by the fluorescent lipophilic tracer DiI (Molecular Probes, Eugene, OR, USA). For labeling, synovial MSCs were resuspended at 1 × 106 cells/ml in αMEM without FBS and a DiI was added at a final concentration of 5 μl/ml. After incubation for 20 minutes at 37°C with 5% humidified CO2, the cells were centrifuged at 450 g for 5 minutes and washed twice with phosphate-buffered saline (PBS) and the cells were then resuspended in CCM and cultured in hanging drops. We already reported that these cells had characteristics of MSCs [3,6,7,11].

Preparation of aggregates of synovial MSCs

A total of 2.5 × 105 synovial MSCs were trypsinized, harvested and resuspended in 35 μl of CCM, plated on an inverted culture dish lid. The lid was inverted and placed on a culture dish containing PBS. The cells were cultured at 37°C with 5% humidified CO2 for three days in hanging drops.

Histology of aggregates of human synovial MSCs

Aggregates of human synovial MSCs were fixed with 2.5% glutaraldehyde in 0.1 M PBS for two hours. The aggregates were washed overnight at 4°C in the same buffer and post-fixed with 1% OsO4 buffered with 0.1 M PBS for two hours. The aggregates were dehydrated in a graded series of ethanol and embedded in Epon 812. Semi-thin (1 μm) sections for light microscopy were collected on glass slides and stained for 30 seconds with toluidine blue.

In vitro chondrogenic differentiation assay

A total of 2.5 × 105 human synovial MSCs cultured as a monolayer were pelleted by trypsinization and centrifugation. The pellets or aggregate of human synovial MSCs cultured for three days in hanging drops were cultured in 400 μl chondrogenic medium consisting of high-glucose Dulbecco’s modified Eagle’s medium (Invitrogen) supplemented with 1,000 ng/ml BMP-7 (Stryker Biotech, Boston, MA, USA), 10 ng/ml transforming growth factor-β3 (R&D Systems, Minneapolis, MN, USA), 100 nM dexamethasone (Sigma-Aldrich Corp., St. Louis, MO, USA), 50 μg/ml ascorbate-2-phosphate, 40 μg/ml proline, 100 μg/ml pyruvate, and 1:100 diluted ITS+Premix (6.25 μg/ml insulin, 6.25 μg/ml transferrin, 6.25 ng/ml selenious acid, 1.25 mg/ml bovine serum albumin, and 5.35 mg/ml linoleic acid; BD Biosciences Discovery Labware, Bedford, MA, USA). The medium was changed every 3 to 4 days for 21 days.

Histology of pellets of human synovial MSCs

The pellets were embedded in paraffin, cut into 5-μm sections and stained with 1% Toluidine Blue. For immunohistochemistry, sections were treated with 0.4 mg/ml proteinase K (DAKO, Carpinteria, CA, USA) in Tris-HCl and normal horse serum after deparaffinization. Primary antibodies for type II collagen (Daiichi Fine Chemical, Toyama, Japan) and a secondary antibody of biotinylated horse anti-mouse IgG (Vector Laboratories, Burlingame, CA, USA) were employed. Immunostaining was detected with VECTASTAIN ABC reagent (Vector Laboratories) followed by 3,3′-diaminobenzidine staining.

Real-time RT PCR analysis

Total RNA was extracted from human synovial MSCs in a monolayer culture, aggregates of human synovial MSCs cultured for 1, 2 and 3 days, and the pellets cultured for 7, 14 and 21 days using QIAzol (Qiagen, Hiden, Germany) and the RNeasy mini kit (Qiagen). cDNA was synthesized with oligo-dT primer from total RNA using the Transcriptor High Fidelity cDNA Synthesis kit (Roche Diagnostics) according to the manufacturer’s protocol. Reverse transcription (RT) was performed by 30 minutes incubation at 55°C followed by 5 minutes incubation at 85°C. Real-time PCR was performed in a LightCycler 480 instrument (Roche Diagnostics). Primer sequences and TaqMan probes are listed in Table 1. After an initial denaturation step (95°C for 10 minutes), amplification was performed for 40 cycles (95°C for 15 seconds, 60°C for 60 seconds). Relative amounts of mRNA were calculated and standardized as previously described [13,14].

Table 1. Real time-RT PCR primer sequences

DNA microarray analysis

Total RNA was extracted from human synovial MSCs in a monolayer culture, aggregates of human synovial MSCs cultured for three days. Human Genome U133 Plus 2.0 Array (GeneChip; Affymetrix, Santa Clara, CA, USA) containing the oligonucleotide probe set for more than 47,000 transcripts was used. The fluorescence intensity of each probe was quantified by using the GeneChip Analysis Suite 5.0 (Affymetrix). Gene expression data were normalized in Robust MultiChip Analysis (RMA). To analyze the data, we used hierarchical clustering using TIGR MultiExperiment Viewer (MeV) [15]. The microarray data have been deposited to the public database (GEO accession# GSE 31980).

In vivo transplantation

Under anesthesia, the left knee joint was approached through a medial parapatellar incision, and the patella was dislocated laterally. Full-thickness osteochondral defects (5 mm × 5 mm wide, 1.5 mm deep) were created in the trochlear groove of the femur. A total of 5, 10, 20, 40 and 80 aggregates of autologous rabbit synovial MSCs (2.5 × 105 cells/aggregate) or 25 and 100 smaller aggregates of autologous rabbit synovial MSCs (1.0 × 105 cells/aggregate) suspended in PBS were transplanted to the defect. To trace the transplanted cells, DiI-labeled aggregates of autologous rabbit synovial MSCs and aggregates of allogenic synovial MSCs derived from GFP transgenic rabbit were transplanted to the defect. For the control group, the defect was left empty. All rabbits were returned to their cages after the operation and were allowed to move freely. Animals were sacrificed with an overdose of sodium pentobarbital at 1, 2, and 4 days and at 12 weeks after the operation (n = 5 at each time).

Macroscopic examination

The cartilage defects were examined macroscopically for color, integrity and smoothness. Osteoarthritic changes and synovitis of the knee were also investigated. Digital images were taken using an Olympus MVX10 (Olympus, Tokyo, Japan).

Histological examination and fluorescent microscopic examination

The dissected distal femurs were immediately fixed in a 4% paraformaldehyde (PFA) solution. The specimens were decalcified in 4% ethylenediamine tetraacetic acid solution, dehydrated with a gradient ethanol series and embedded in paraffin blocks. Sagittal sections 5 μm thick were obtained from the center of each defect and were stained with toluidine blue and Safranin O. For fluorescent microscopic examination and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, the fixed specimens were incubated at 4°C for three hours in 5%, 10%, 15% and 20% sucrose solution, respectively. After incubation, the fixed specimens were mounted on a holder. Then 30% optimal cutting temperature (OCT) (Sakura Finetek, Tokyo, Japan) in sucrose solution was added gently into the holder. The holder was frozen in hexan chilled by dry ice and stored at -80°C. Cryosections (10 μm) were prepared with an ultracut S microtome (Reichert, Wien, Austria) and a Microm HM560 cryostat.

Histological score

Histological sections of the repaired tissue were analyzed using a grading system consisting of five categories (cell morphology, morphology, matrix staining, surface regularity, cartilage thickness and integration of donor with host), which were modified from the repaired cartilage score described by Wakitani and colleagues [16], so that overly thick, regenerated cartilage could not be overestimated [6]. The scoring was performed in a blinded manner by two observers and there was no significant interobserver difference. The ratio of the safranin-O positive area over the defect was evaluated. Zeiss AxioVison software (Carl Zeiss, Oberkochen, Germany) was used for measurement of defects and safranin-O positive areas.

In vitro viability assay

Aggregates of rabbit synovial MSCs were plated at 1 or 40 aggregates/well in 96-well plates (Nunc) in CCM, and incubated at 37°C with 5% humidified CO2 for seven days without medium change. Aggregates were fixed in 4% PFA for TUNEL staining.

TUNEL staining

For TUNEL staining, an apoptosis in situ detection kit (Wako Pure Chemical Industries, Ltd, Osaka, Japan) was used. The frozen semi-thin sections were incubated with terminal deoxynucleotidyl transferase for 10 minutes at 37°C in a moist chamber. The sections were washed with 0.1 M PBS for 15 minutes. Peroxidase-conjugated antibody was then applied to the specimens at 37°C for 10 minutes in a moist chamber. The sections were developed with 3,3-diaminobenizidine and counterstained with methyl green.

Statistical analysis

Comparisons between two groups were analyzed using the Mann-Whitney U test. Comparisons between multi groups were analyzed using the Kruskal-Wallis test and the Steel test. A P-value of < 0.05 was considered statistically significant.

Results

Appearance of aggregates of human synovial MSCs

Human synovial MSCs were aggregated using the hanging drop technique (Figure 1A). Three days after being cultured in the drop (Figure 1B), the aggregate, consisting of 250,000 MSCs, became approximately 1 mm in diameter (Figure 1C). The aggregate was not easily broken by manipulation. Sagittal sections of the aggregates showed heart-shape as a whole (Figure 1Da). The superficial layer was composed of spindle cells parallel to the surface, whereas the deep layer was comprised of round cells both at top and bottom of the aggregate (Figure 1Db, c). Though cells positive for TUNEL staining were observed, the number was only approximately under 5% (Figure 1Dd).

 

Figure 1. Preparation and appearance of aggregates of human synovial MSCs. (A): Scheme of preparation of aggregates using hanging drop technique. (B): Drops hanging on the cover of 15 cm dish. (C): Macroscopic image of aggregate consisted of 250,000 MSCs, three days after cultured in hanging drop. (D): Sagittal sections of aggregates stained with toluidine blue (a, b, c) and TUNEL (d). TUNEL positive cells are indicated with arrows.

Transcriptome profile of aggregates of human synovial MSCs

To examine the sequential changes of gene expression profiles during aggregation of human synovial MSCs, microarray analyses were performed. The differences of gene profile between before and after aggregation exceeded those among donor variances (Figure 2A). The number of genes up-regulated more than five-fold was 621. The number of genes up-regulated more than 100-fold was 10, and these genes were related to hypoxia (integrin, alpha 2 (ITGA2), stanniocalcin 1 (STC1), chemokine (C-X-C motif) receptor 4 (CXCR4)), nutrient (BMP2, proprotein convertase subtilisin/kexin type 1 (PCSK1), secreted phosphoprotein 1 (SPP1), ITGA2, STC1), extracellular region (MMP1, MMP3), and cell adhesion (SPP1, ITGA2) (Table 2). The most up-regulated gene was BMP2, increased to 273 folds (Table 2). STC1 was also highly up-regulated in aggregates of synovial MSCs. The number of genes down-regulated less than one-fifth was 409, and the ontology for the genes was related to cell cycle. The microarray data are available at the public database (GEO accession# GSE 31980).

 

Figure 2. Transcriptome changes after aggregation of human synovial MSCs. (A): Hierarchical clustering analysis for gene expression profile of aggregates of MSCs. The color code for the signal strength in the classification scheme is shown in the box left. High expression genes are indicated by shades of red and low expression genes are indicated by shades of green. (B): Expressions of chondrogenesis-related genes (SOX5, SOX6, SOX9, BMP2) and anti-inflammatory genes (TSG-6, STC-1) in aggregates of MSCs at Days 0 to 3 by real time RT-PCR analysis. The results are shown in four individual donors respectively.

Table 2. The top 10 upregulated genes in aggregates of MSCs

To further investigate gene expressions during aggregation of human synovial MSCs, real time RT-PCR analyses were additionally used for chondrogenesis-related genes (SRY (sex determining region Y)-box (SOX)5, -6, -9, and BMP2) and anti-inflammatory genes (TNFα inducible gene 6 (TSG-6), and STC-1) in four donors. In most cases, expressions for these genes increased sequentially (Figure 2B).

In vitro chondrogenesis of aggregates of human synovial MSCs

In vitro chondrogenic ability of human synovial MSCs after hanging drop culture was compared to that of MSCs after monolayer culture (Figure 3A). Aggregates of MSCs differentiated into chondrocytes as well (Figure 3B). The wet weight of pellets derived from MSCs after hanging drop culture was heavier than that of pellets derived from MSCs after monolayer culture in all four donors at 14 or 21 days (Figure 3C). Real time RT-PCR analysis showed higher expression levels of collagen (COL)2A1, aggrecan and SOX9 for pellets derived from MSCs after hanging drop culture compared to MSCs after monolayer culture at 14 and 21 days (Figure 3D). Cartilage extracellular matrix synthesis and accumulation of type II collagen were confirmed by histological analysis stained with toluidine blue and immunohistochemical analysis (Figure 3E).

 

Figure 3. In vitro chondrogenic ability of human synovial MSCs after hanging drop culture (A): Scheme for the analyses. (B): Macroscopic images of pellets derived from aggregates of MSCs and those of pellets derived from MSCs in a monolayer culture. (C): Wet weight in four individual donors. Values are the means with standard deviation (SD) (P < 0.05 by the Mann-Whitney U test). (D): Expressions of chondrogenesis-related genes by RT-PCR analyses. Values are the means with SD among four donors. The fold changes of SOX9 and AGGRECAN expression levels were shown when the gene expression levels at Day 0 were normalized as 1. The fold changes of COL2A1 expression levels were shown when the gene expression levels in MSCs in monolayer at Day 7 were normalized as 1 because COL2A1 expression level at Day 0 was undetectable. (E): Histological sections of pellets stained with toluidine blue and immunohistochemical analysis for type II collagen.

In vivo analysis for cartilage regeneration by transplantation of aggregates of synovial MSCs in rabbits

To examine whether transplantation of aggregates of synovial MSCs promotes cartilage regeneration, in vivo study was performed in rabbits. To further investigate the optimal number of aggregates consisting of 250,000 MSCs, 0 to 80 aggregates were transplanted into the defect.

At 0 days, in the case of 40 and 80 transplanted aggregates, the osteochondral defects were filled with aggregates labeled with DiI macroscopically (Figure 4A).

 

Figure 4. Cartilage regeneration by transplantation of aggregates of synovial MSCs in rabbits. (A): Macroscopic observation of osteochondral defects one minute after transplantation of indicated number of aggregates of MSCs. The aggregate consisted of 250,000 MSCs, labeled with DiI for visualization. (B): Macroscopic and histological observation. For histologies, sagittal sections were stained with safranin-O (SO) and toluidine blue (TB). (C, D): Magnified histology of the indicated area. (E): Histological score. Values are the means with SD. (n = 5; P < 0.05 by the Kruskal-Wallis test and the Steel test). (F): Ratio of the safranin-O positive area to the defect area. Values are the means with SD. (n = 5; P < 0.05 by the Kruskal-Wallis test and the Steel test).

At four weeks, in the case of 5 and 10 transplanted aggregates, the osteochondral defect was mostly covered with a thick cartilage matrix (Figure 4B, C). In the case of 20 and 40 transplanted aggregates, the defect was partially covered with cartilage matrix. In the case of 80 transplanted aggregates, the defect was filled with only fibrous tissue, which appeared to be similar to the control (Figure 4B).

At 12 weeks, in the case of 10 transplanted aggregates, the border between cartilage and bone moved up, and thickness of the regenerated cartilage became similar to the neighboring cartilage (Figure 4B, D). In the case of 5 and 20 transplanted aggregates, the bone defect was repaired, but the cartilage defect was filled partially with cartilage matrix. In the case of 40 and 80 transplanted aggregates, the osteochondral defect was poorly repaired, similar to the control (Figure 4B). Histological score was the best and the safranin-O positive area ratio was highest in the case of 10 transplanted aggregates both at 4 and 12 weeks (Figure 4E, F).

To trace MSCs, 10 aggregates of GFP positive MSCs were transplanted into the defect. At Day 1, no GFP positive aggregates could be observed in the knee joint except the defects with a fluorescent stereomicroscope. Histologically, aggregates changed their forms but have not fused yet (Figure 5A). At four weeks, the defect was filled with cartilage matrix and the GFP positive cells were still observed both at the bottom and the center of the regenerated cartilage (Figure 5B). Regenerated cartilage consisted of both GFP positive cells and GFP negative cells.

 

Figure 5. Transplantation of 10 aggregates of synovial MSCs derived from a GFP rabbit. (A): Sagittal sections of osteochondral defect under fluorescence for GFP at one day. (B): Macroscopic and histological observation four weeks after transplantation. Nuclei were shown as blue in higher magnified pictures.

Influences of cell number per aggregate and of aggregate number for transplantation

Cell number per aggregate as well as aggregate number may be a factor affecting properties of the aggregates. To answer this question, 25 or 100 aggregates, in which an aggregate consisted of 100,000 MSCs, were transplanted into the osteochondral defect.

At four weeks, in the case of 25 transplanted aggregates, the defect was fully filled with cartilage matrix (Figure 6A), in which the result was different from the case of 20 or more aggregates, in which an aggregate consisted of 250,000 MSCs. In the case of 100 transplanted aggregates, the defect was filled with fibrous tissue, and the histological score was inferior and the safranin-O positive area ratio was smaller. (Figure 6B, C).

 

Figure 6. Influences of cell number per an aggregate and of aggregate number for transplantation. (A): Macroscopic and histological observation four weeks after transplantation of 25 or 100 aggregates in which an aggregate consisted of 100,000 MSCs. (B): Histological score. Values are the means with SD (n = 4; P < 0.05 by the Mann-Whitney U test). (C): Ratio of the safranin-O positive area to the defect area. Values are the means with SD. (n = 4; P < 0.05 by the Mann-Whitney U test). (D): Histological observation two weeks after transplantation of 10 and 80 aggregates in which an aggregate consisted of 250,000 MSCs labeled with DiI. Sagittal sections under fluorescence and the serial sections stained with TUNEL were shown. (E): In vitro analyses of aggregates of rabbit synovial MSCs. One or 40 aggregates, in which an aggregate consisted of 250,000 MSCs, were cultured in a well of 96-well plates. Macroscopic images for the wells and sagittal sections of the aggregates stained with TUNEL were shown.

Influences of aggregate number on viability of MSCs

To clarify why transplantation of aggregates over a certain number resulted in poor outcome, viability of cells was first examined by TUNEL staining. Compared to the case of 10 transplanted aggregates, much more TUNEL positive cells could be observed in the case of 80 transplanted aggregates (Figure 6D).

Another factor might be a nutrient deprivation and in vitro analyses using aggregates of rabbit synovial MSCs were performed. Seven days after 1 or 40 aggregates were cultured in a well of 96-well plates, the medium color changed to yellow in the case of 40 aggregates, while the color remained red in the case of only 1 aggregate (Figure 6E). TUNEL positive cells were much higher in the case of 40 aggregates than in the case of only 1 aggregate.

Discussion

In this study, to form aggregates of synovial MSCs, the hanging drop technique was used [8-10]. This is a simple method; expensive or specific tools are not required. Three days after cultured in the drop, the aggregate, consisting of 250,000 MSCs, became approximately 1 mm in diameter, large enough to be visible and solid enough to aspirate with a pipette. Aggregates of MSCs sank faster in the suspension medium than dispersed MSCs and helped to avoid loss of MSCs from targeted cartilage defect. The use of aggregates was practically convenient for transplantation of MSCs.

In the previous report, the number of apoptotic or necrotic cells was greater in aggregates prepared with 100,000 or 250,000 human bone marrow MSCs, which was examined by flow cytometry, measuring propidium iodide uptake and annexin V labeling [10]. We examined the viability of aggregates of MSCs by TUNEL staining and confirmed that cells positive for TUNEL staining were observed; the number was small compared to the previous report. This difference may have been due to the difference of methods. Microarray analysis showed up-regulation of genes with ontology for regulation of cell death. The microarray data are available at the public database (GEO accession# GSE 31980). These results suggest that aggregation of 250,000 MSCs affect the viability of cells. However, we thought that aggregates of MSCs could be used as a source for cartilage regeneration because most cells which are cultured in drops for three days are viable.

Aggregation of synovial MSCs changed the gene expression profile dramatically without any special tools or chemical factors. This is possibly due to environmental changes, including cell-to-cell contact, hypoxic condition and low nutrient condition. Aggregation of human synovial MSCs increased expressions of several chondrogenesis-related genes and the most up-regulated gene was BMP2, which was also up-regulated in bone marrow MSCs [8,10].

In this study, we compared in vitro chondrogenesis potential of synovial MSCs after hanging drop culture with that of MSCs after monolayer culture. We used 1,000 ng/ml BMP7 for in vitro chondrogenic differentiation assay. We previously examined the dose effect of BMP6 between 0 to 500 ng/ml for in vitro chondrogenesis of bone marrow MSCs. Cartilage pellets increased in size along with the concentration of BMP6, and a maximal effect was at 500 ng/ml [17]. Our preliminary experiments showed that 1,000 ng/ml BMP6 induced larger cartilage pellets than 500 ng/ml BMP6 in bone marrow and synovial MSCs. We obtained similar results with BMP7. Real time RT-PCR analysis showed higher expression levels of COL2A1, aggrecan and SOX9 for pellets derived from MSC-aggregates after hanging drop culture compared to those of MSCs in a monolayer culture. Furthermore, the wet weight of pellets derived from MSC-aggregates after hanging drop culture was heavier than that of pellets derived MSCs in a monolayer culture. These indicate that chondrogenic potential increased in aggregates of MSCs after hanging drop culture.

In this study, we used an osteochondral defect model of rabbits, which have a higher, self-renewal capacity than bigger animals and humans. Therefore, the results obtained here should be critically evaluated. However, we prepared negative controls, which healed poorly at 4 and 12 weeks. We previously confirmed that the osteochondral defect created in the trochlear groove of the femur, similar to this study, was not repaired without any treatments 24 weeks after surgery [6]. These findings indicate that this rabbit model is useful to evaluate the effects of the treatments for cartilage regeneration.

For in vivo analysis of cartilage regeneration by transplantation of aggregates of synovial MSCs in rabbits, successful cartilage regeneration was observed in the cases of a relatively small number of transplanted aggregates of MSCs, and the worst results were observed when the highest number of aggregates of MSCs was transplanted. These results were not what we expected, because we previously reported that better cartilage regeneration was obtained when higher cell densities of MSCs were embedded in collagen gel [3].

Why were poor results obtained when more than a certain number of aggregates were transplanted? We listed three possible reasons. First, nutrition to maintain transplanted MSCs was depleted and the environment around transplanted MSCs worsened when too many aggregates were transplanted. As shown in Figure 6E, in the case of 40 aggregates that were cultured for seven days in a well of 96-well plates, medium color changed to yellow. This means that adjustment of pH could not be controlled. Second, TUNEL positive cells increased when too many aggregates were transplanted. The number of TUNEL positive cells was higher when too many aggregates were transplanted (Figure 6D) than before transplantation (Figure 1D) and after a suitable number of aggregates were transplanted (Figure 6D). Third, transplantation of too many aggregates prevented chondro-progenitor cells from moving to the osteochondral defect from bone marrow and from synovial fluid.

We confirmed that transplanted aggregates of synovial MSCs were directly differentiated into chondrocytes by transplanting MSCs derived from GFP transgenic rabbit. This result suggests that aggregates of synovial MSCs were involved in the reparative process. However, as shown in Figure 5B, in the case of aggregates of GFP positive MSCs being transplanted, regenerated cartilage consisted of both GFP positive cells and GFP negative cells. MSCs existed in synovial fluid [18] and these MSCs contributed to the repair of cartilage injury [6,19]. These results suggest that some host MSCs were also involved in the reparative process. In addition, host MSCs may have been involved in the anti-inflammatory process. In our rabbit osteochondral defect model, inflammation like a synovitis was not severe even in the control group. Therefore, we could not confirm the anti-inflammatory effect of MSCs. It would be interesting to investigate the anti-inflammatory effect of transplantation of aggregates of synovial MSCs and host MSCs in other arthritis models.

As previously reported, in bone marrow MSCs [10], aggregates of human synovial MSCs expressed anti-inflammatory genes TSG6 and STC1. TSG6 is secreted by synoviocytes, mononuclear cells and chondrocytes under inflammatory conditions and has an anti-inflammatory effect. Overexpression of TSG6 or administration of recombinant TSG6 inhibited inflammation and joint destruction in a murine collagen induced arthritis model [20-23]. STC1 is reported to have an anti-apoptotic effect as well as an anti-inflammatory effect [24,25]. However, their roles in joint homeostasis are unknown.

In this study, transplantation of low numbers of aggregates, in other words, low density of aggregates to the volume of the cartilage defect, showed better regeneration (Figures 4 and 6). This is favorable for clinical application. We have performed clinical trials of autologous human synovial MSCs transplantation for cartilage defects. In the experiences of 12 patients, approximately 50 million synovial MSCs at passage 0 were transplanted for approximately 280 mm2 cartilage defects (unpublished data). In a rabbit model, we transplanted synovial MSC-aggregates into the osteochondral defects without any loss of cells, and 10 MSC-aggregates (2.5 × 106 cells) per 25 mm2 defects were needed for better cartilage regeneration. According to these data, we can prepare a sufficient amount of human synovial MSCs at passage 0.

In this study, we did not use scaffolds for transplantation of aggregates of synovial MSCs. We were able to adhere aggregates of synovial MSCs on the osteochondral defect without scaffolds; however, the use of scaffolds or materials to improve survival of transplanted cells is attractive. One of the methods is the use of a fibrin glue, which has an effect of improving survival of transplanted cells [26]. In addition, cell transplantation of MSCs with a fibrin glue can probably be performed under arthroscopic surgery. Further studies are needed to improve cell transplantation procedures.

Conclusion

Aggregated synovial MSCs were a useful source for cartilage regeneration considering such factors as easy preparation, higher chondrogenic potential and efficient attachment.

Abbreviations

αMEM: α-minimal essential medium; BMP: bone morphogenetic protein; CCM: complete culture medium; COL: collagen; CXCR4: chemokine (C-X-C motif) receptor 4; EDTA: ethylenediaminetetraacetate; FBS: fetal bovine serum; GFP: green fluorescent protein; GJB2: gap junction protein, beta 2; ITGA2: integrin, alpha 2; MeV: MultiExperiment Viewer; MMP: matrix metalloproteinase; MSC: mesenchymal stem cell; OCT: optimal cutting temperature; PBS: phosphate-buffered saline; PCSK1: proprotein convertase subtilisin/kexin type 1; PFA: paraformaldehyde; RMA: Robust MultiChip Analysis; RT: reverse transcription; SD: standard deviation; SO: safranin-O; SOX: SRY (sex determining region Y)-box; SPP1: secreted phosphoprotein 1; STC1: stanniocalcin 1; TB: toluidine blue; TFPI2: tissue factor pathway inhibitor 2; TNF: tumor necrosis factor; TSG6: TNFα inducible gene 6; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SS participated in the design of the study, carried out the animal experiments, analyzed the results and drafted the manuscript. TM participated in the design of the study and provided the administrative and financial support. KT participated in the design of the study. SI helped with histological analysis. HM and AU carried out the microarray analysis and participated in the evaluation of the results. IS participated in the design of the study, provided the financial support and completed the final manuscript. All authors read and approved the final manuscript.

Acknowledgements

This study was supported by “the Project for Realization of Regenerative Medicine” and “the Global Center of Excellence (GCOE) Program” by the Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan to IS, grants from the Japanese Ministry of Education Global Center of Excellence Program, International Research Center for Molecular Science in Tooth and Bone Diseases to TM, and from a Health and Labor Sciences Research Grant, Research on Regenerative Medicine for Clinical Application to IS. Recombinant human BMP-7 was distributed by Stryker Biotech. We thank Miyoko Ojima for her expert help with histology and Izumi Nakagawa for management of our laboratory.

References

  1. Sakaguchi Y, Sekiya I, Yagishita K, Muneta T: Comparison of human stem cells derived from various mesenchymal tissues: superiority of synovium as a cell source.
    Arthritis Rheum 2005, 52:2521-2529. PubMed Abstract | Publisher Full Text
  2. Shirasawa S, Sekiya I, Sakaguchi Y, Yagishita K, Ichinose S, Muneta T: In vitro chondrogenesis of human synovium-derived mesenchymal stem cells: optimal condition and comparison with bone marrow-derived cells.
    J Cell Biochem 2006, 97:84-97. PubMed Abstract | Publisher Full Text
  3. Koga H, Muneta T, Nagase T, Nimura A, Ju YJ, Mochizuki T, Sekiya I: Comparison of mesenchymal tissues-derived stem cells for in vivo chondrogenesis: suitable conditions for cell therapy of cartilage defects in rabbit.
    Cell Tissue Res 2008, 333:207-215. PubMed Abstract | Publisher Full Text
  4. Nimura A, Muneta T, Koga H, Mochizuki T, Suzuki K, Makino H, Umezawa A, Sekiya I: Increased proliferation of human synovial mesenchymal stem cells with autologous human serum: comparisons with bone marrow mesenchymal stem cells and with fetal bovine serum.
    Arthritis Rheum 2008, 58:501-510. PubMed Abstract | Publisher Full Text
  5. Koga H, Engebretsen L, Brinchmann JE, Muneta T, Sekiya I: Mesenchymal stem cell-based therapy for cartilage repair: a review.
    Knee Surg Sports Traumatol Arthrosc 2009, 17:1289-1297. PubMed Abstract | Publisher Full Text
  6. Koga H, Shimaya M, Muneta T, Nimura A, Morito T, Hayashi M, Suzuki S, Ju YJ, Mochizuki T, Sekiya I: Local adherent technique for transplanting mesenchymal stem cells as a potential treatment of cartilage defect.
    Arthritis Res Ther 2008, 10:R84. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text
  7. Shimaya M, Muneta T, Ichinose S, Tsuji K, Sekiya I: Magnesium enhances adherence and cartilage formation of synovial mesenchymal stem cells through integrins.
    Osteoarthritis Cartilage 2010, 18:1300-1309. PubMed Abstract | Publisher Full Text
  8. Potapova IA, Gaudette GR, Brink PR, Robinson RB, Rosen MR, Cohen IS, Doronin SV: Mesenchymal stem cells support migration, extracellular matrix invasion, proliferation, and survival of endothelial cells in vitro.
    Stem Cells 2007, 25:1761-1768. PubMed Abstract | Publisher Full Text
  9. Potapova IA, Brink PR, Cohen IS, Doronin SV: Culturing of human mesenchymal stem cells as three-dimensional aggregates induces functional expression of CXCR4 that regulates adhesion to endothelial cells.
    J Biol Chem 2008, 283:13100-13107. PubMed Abstract | Publisher Full Text | PubMed Central Full Text
  10. Bartosh TJ, Ylostalo JH, Mohammadipoor A, Bazhanov N, Coble K, Claypool K, Lee RH, Choi H, Prockop DJ: Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties.
    Proc Natl Acad Sci USA 2010, 107:13724-13729. PubMed Abstract | Publisher Full Text | PubMed Central Full Text
  11. Miyamoto T, Muneta T, Tabuchi T, Matsumoto K, Saito H, Tsuji K, Sekiya I: Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits.
    Arthritis Res Ther 2010, 12:R206. PubMed Abstract | BioMed Central Full Text | PubMed Central Full Text
  12. Takahashi R, Kuramochi T, Aoyagi K, Hashimoto S, Miyoshi I, Kasai N, Hakamata Y, Kobayashi E, Ueda M: Establishment and characterization of CAG/EGFP transgenic rabbit line.
    Transgenic Res 2007, 16:115-120. PubMed Abstract | Publisher Full Text
  13. Niikura T, Hak DJ, Reddi AH: Global gene profiling reveals a downregulation of BMP gene expression in experimental atrophic nonunions compared to standard healing fractures.
    J Orthop Res 2006, 24:1463-1471. PubMed Abstract | Publisher Full Text
  14. Willems E, Leyns L, Vandesompele J: Standardization of real-time PCR gene expression data from independent biological replicates.
    Anal Biochem 2008, 379:127-129. PubMed Abstract | Publisher Full Text
  15. Saeed AI, Bhagabati NK, Braisted JC, Liang W, Sharov V, Howe EA, Li J, Thiagarajan M, White JA, Quackenbush J: TM4 microarray software suite.
    Methods Enzymol 2006, 411:134-193. PubMed Abstract
  16. Wakitani S, Goto T, Pineda SJ, Young RG, Mansour JM, Caplan AI, Goldberg VM: Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage.
    J Bone Joint Surg Am 1994, 76:579-592. PubMed Abstract | Publisher Full Text
  17. Sekiya I, Colter DC, Prockop DJ: BMP-6 enhances chondrogenesis in a subpopulation of human marrow stromal cells.
    Biochem Biophys Res Commun 2001, 284:411-418. PubMed Abstract | Publisher Full Text
  18. Morito T, Muneta T, Hara K, Ju YJ, Mochizuki T, Makino H, Umezawa A, Sekiya I: Synovial fluid-derived mesenchymal stem cells increase after intra-articular ligament injury in humans.
    Rheumatology (Oxford) 2008, 47:1137-1143. Publisher Full Text
  19. Koga H, Muneta T, Ju YJ, Nagase T, Nimura A, Mochizuki T, Ichinose S, von der Mark K, Sekiya I: Synovial stem cells are regionally specified according to local microenvironments after implantation for cartilage regeneration.
    Stem Cells 2007, 25:689-696. PubMed Abstract | Publisher Full Text
  20. Wisniewski HG, Hua JC, Poppers DM, Naime D, Vilcek J, Cronstein BN: TNF/IL-1-inducible protein TSG-6 potentiates plasmin inhibition by inter-alpha-inhibitor and exerts a strong anti-inflammatory effect in vivo.
    J Immunol 1996, 156:1609-1615. PubMed Abstract | Publisher Full Text
  21. Maier R, Wisniewski HG, Vilcek J, Lotz M: TSG-6 expression in human articular chondrocytes. Possible implications in joint inflammation and cartilage degradation.
    Arthritis Rheum 1996, 39:552-559. PubMed Abstract | Publisher Full Text
  22. Mindrescu C, Dias AA, Olszewski RJ, Klein MJ, Reis LF, Wisniewski HG: Reduced susceptibility to collagen-induced arthritis in DBA/1J mice expressing the TSG-6 transgene.
    Arthritis Rheum 2002, 46:2453-2464. PubMed Abstract | Publisher Full Text
  23. Mindrescu C, Thorbecke GJ, Klein MJ, Vilcek J, Wisniewski HG: Amelioration of collagen-induced arthritis in DBA/1J mice by recombinant TSG-6, a tumor necrosis factor/interleukin-1-inducible protein.
    Arthritis Rheum 2000, 43:2668-2677. PubMed Abstract | Publisher Full Text
  24. Block GJ, Ohkouchi S, Fung F, Frenkel J, Gregory C, Pochampally R, DiMattia G, Sullivan DE, Prockop DJ: Multipotent stromal cells are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1.
    Stem Cells 2009, 27:670-681. PubMed Abstract | Publisher Full Text
  25. Huang L, Garcia G, Lou Y, Zhou Q, Truong LD, DiMattia G, Lan XR, Lan HY, Wang Y, Sheikh-Hamad D: Anti-inflammatory and renal protective actions of stanniocalcin-1 in a model of anti-glomerular basement membrane glomerulonephritis.
    Am J Pathol 2009, 174:1368-1378. PubMed Abstract | Publisher Full Text | PubMed Central Full Text
  26. Guo HD, Wang HJ, Tan YZ, Wu JH: Transplantation of marrow-derived cardiac stem cells carried in fibrin improves cardiac function after myocardial infarction.
    Tissue Eng Part A 2011, 17:45-58. PubMed Abstract | Publisher Full Text

Invention Patent: Composition For Increasing Body Height – No FGFR3 Abnormality BY Activating Guanyl Cyclase

Me: I found a link to this patent from going through a discussion on the Make Me Taller boards. Can’t seem to be able to remember the link though. The patent application is long, very long. I woudl suggest you don’t read it except the abstract and a few things I will highlight in the document. 

This is sort of a big step forward. Apparently activating Guanyl Cyclase got the long bones of mice to grow thicker and longer.


This invention provides a composition for increasing a body height of a patient with short stature or an individual other than patients with short stature. More specifically, the invention provides: a composition for increasing the body height of an individual comprising a guanyl cyclase B (GC-B) activator as an active ingredient, the composition being to be administered to an individual free from FGFR3 abnormality; a method for increasing the body height of an individual free from FGFR3 abnormality which comprises activating GC-B; a method for screening an agent for increasing the body height of an individual which comprises selecting an agent for increasing the body height using GC-B activity as an indication; and a method for extending a cartilage bone free from FGFR3 abnormality which comprises activating GC-B in an individual.

Inventors:
Nakao, Kazuwa (4-1-2, Kitakutsukake-cho,Ohe,Nishikyo-ku, Kyoto-shi, Kyoto 6101101, JP)
Yasoda, Akihiro (6-47, Kamigaki-cho, Nishinomiya-shi, Hyogo, 6620865, JP)
Kitamura, Hidetomo C/o Chugai Seiyaku K. K. (135, Komakado 1-chome, Gotenba-shi, Shizuoka 4128513, JP)
Application Number:
EP20050728903
Publication Date:
01/17/2007
Filing Date:
03/31/2005
Assignee:
Nakao, Kazuwa (4-1-2, Kitakutsukake-cho, Ohe Nishikyo-ku, Kyoto-shi, Kyoto 610-1101, JP)
International Classes:
A61K45/00A61K38/00A61K38/22A61P19/00A61P43/00C07K14/47C12N15/00C12N15/09C12Q1/02C12Q1/527G01N33/15;G01N33/50G01N33/68
European Classes:
A61K38/22F; C12Q1/527; G01N33/68R
View Patent Images:
Foreign References:
JP2003113116A
JP2004107871A
WO/2002/074234A METHOD AND COMPOSITION FOR TREATMENT OF SKELETAL DYSPLASIAS
WO/1991/016342A NOVEL PHYSIOLOGICALLY ACTIVE PEPTIDE ORIGINATING IN HOG
JP4074198A
JP4139199A
JP4121190A
JP4120094A
JP4120095A
JP6009688A
Attorney, Agent or Firm:
Vossius & Partner (Siebertstrasse 4, 81675 München, DE)
Claims:
1. A composition for increasing a body height of an individual, comprising a guanyl cyclase B (GC-B) activator as an active ingredient, the composition being to be administered to an individual free from FGFR3 abnormality.

2. The composition of claim 1, for use in a patient with short stature.

3. The composition of claim 1, for use in an individual other than patients with short stature.

4. The composition of claim 1, wherein the increase in body height is extension of cartilage bones.

5. The composition of claim 1, wherein the increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.

6. The composition of claim 1, wherein the activator is a peptide.

7. The composition of claim 6, wherein the peptide is type C natriuretic peptide (CNP) or a derivative thereof.

8. The composition of claim 7, wherein the CNP is CNP-22 or CNP-53 from mammals including human, or birds.

9. The composition of claim 7, wherein the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.

10. The composition of claim 7, wherein the derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or 2, while possessing a CNP activity. 

11. A method for increasing a body height of an individual, comprising activating GC-B to increase the body height in an individual free from FGFR3 abnormality. 
12. The method of claim 11, wherein the increase in body height is extension of cartilage bones.

13. The method of claim 11, wherein the increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.

14. The method of claim 11, wherein the GC-B is activated by CNP or a derivative thereof.

15. The method of claim 14, wherein the CNP is CNP-22 or CNP-53 from mammals, including human, or birds.

16. The method of claim 14, wherein the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.

17. The method of claim 14, wherein the derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or 2 , while possessing a CNP activity.

18. A method for screening an agent for increasing the body height of an individual, comprising screening candidate agents for an agent for increasing the body height using the activity of GC-B as an indication.

19. The method of claim 18, which comprises preparing cultured cells that express GC-B or cells from articular chondrocytes, culturing the cells in the presence of a candidate agent, and screening candidate agents for an agent for increasing the body height of an individual using the activity of GC-B in the cells as an indication.

20. The method of claim 18, wherein the activity of GC-B is determined as an amount of produced intracellular cGMP.

21. The method of claim 18, wherein it comprises preparing a cultured cell line that has been forced to express GC-B, culturing the cell line in the presence or absence of a test substance, determining an amount of intracellular cGMP produced in the cell line, and screening candidate agents for an agent for increasing body heights using the difference, as an indication, in amounts of intracellular cGMP produced in the presence and absence of the test substance. 

22. A method for extending a cartilage bone free from FGFR3 abnormality in an individual, comprising activating GC-B in the individual. 

Description:
Technical Field

The present invention relates to a composition for increasing the body height of an individual, comprising, a guanyl cyclase B (GC-B) activator as an active ingredient. More specifically, the composition of the present invention can be used for treatment of a patient with short stature free from FGFR3 abnormality, or for increasing the body height in an individual other than patients with short stature disease.

The present invention also relates to a method for increasing a body height of an individual by activation of GC-B.

The present invention further relates to a method for screening for an agent for increasing a body height of an individual using the activity of GC-B as an indication, and to a method for extending cartilage bones free from FGFR3 abnormality by the activation of GC-B.

Background of Invention

The term “short stature” is medically defined as height more than two standard deviation (-2SD) below the mean height of the population of individuals of the same sex and the same age. When this criterion is fulfilled with respect to an individual, such an individual is diagnosed as a short-statured syndrome or dwarfism. The short stature is roughly divided into: short stature caused by endocrine abnormalities such as hyposecretion of growth hormones or insulin-like growth factor-I (IGF-I); short stature caused by non-endocrine abnormalities, including familial short stature, fetal hypoplastic short stature, or chromosomal abnormality-caused short stature; and secondary short stature caused by chemotherapy or radiation therapy.

The short stature or dwarfism has been treated so far by administration of growth hormones or by orthopedic surgeries, such as replacement of a hip joint with an artificial joint or limb lengthening. In the case of limb lengthening, the bone is surgically cut at age 10 or older and the body height is gradually extended using a special machine (a limb lengthener) over a period of around half a year. This operation, however, imposes severe pain on the patient. In the case of growth hormone therapy, height growth can be improved via periodical injection of growth hormones from early childhood; however, growth would be terminated upon discontinuation of injection. Such treatment techniques are not intended to treat diseases, and are not considered to be ideal from the viewpoint of the quality of life (QOL) of patients (American Journal of Medical Genetics 1997, 72: 71-76; European Journal of Endocrinology 1998; 138: 275-280). The short stature caused by endocrine abnormalities is a disease capable of treating with drugs such as recombinant growth hormones or IGF-I. In contrast, the cause of a nonendocrine abnormality-caused short stature like familial short stature or fetal hypoplastic short stature has not yet been elucidated. Since the effect of growth hormones on nonendocrine abnormality-caused short stature has not been approved, there are no effective therapeutic agents against such short stature (the Merck Manual, 17th edition, 1999, Nikkei Business Publications, Inc./Nikkei BP Publishing Center, Inc., Japan). Under these circumstances, development of therapeutic agents based on new mechanisms has been demanded.

Guanyl cyclase (GC) is a membrane protein belonging to the enzyme family that catalyzes the synthesis of the second messenger cGMP from GTP, and its examples include GC-A, GC-B, …, and GC-F. GC-B is found mainly in vascular endothelial cells, and thought to be involved in relaxation of the smooth muscle.

Natriuretic peptides (NPs) are divided into ANP (atrial sodium peptide), BNP (brain natriuretic peptide) and CNP (type c natriuretic peptide), and they are thought to elevate an intracellular cGMP level through two guanyl cyclase conjugated receptors (NPR-A for ANP and BNP, and NPR-B for CNP) and to perform intracellular signal transduction mediated by a plurality of cGMP effecter molecules (Ann Rev Biochem 1991; 60: 229-255). NPs have been reported to play an important role in the control of humoral homeostasis and blood pressure (J Clin Invest 1987; 93:1911-1921, J Clin Invest 1994; 87: 1402-1412), and their expression and biological activity in various tissues other than the cardiovascular system are known (Endocrinol 1991; 129:1104-1106, Ann Rev Biochem 1991; 60: 553-575). Concerning cartilage bones, effectiveness of overexpression of BNP (Proc. Natl. Acad. Sci., U.S.A., 1998, 95: 2337-2342) or CNP in the joints on the treatment of achondrogenesis resulting from mutation of a fibroblast growth factor receptor 3 (FGFR3) gene has been reported (Nat. Med., 2004, 10 (1): 80-86;

Japanese Patent Publication No. 2003-113116 A).

An object of the present invention is to provide a composition for increasing a body height of a patient with short stature or an individual other than patiens with short stature, who is free from FGFR3 abnormality, for therapeutic, cosmetic, or other purposes.

It is another object of the present invention to provide a method for increasing a body height in a patient with short stature or an individual other than patients with short stature by the activation of GC-B, wherein said patient and individual are both free from FGFR3 abnormality.

A further object of the present invention is to provide a method for screening for an agent for increasing a body height using the activity of GC-B as an indication.

A still further object of the present invention is to provide a method for extending a cartilage bone free from FGFR3 abnormality by the activation of GC-B.

Summary of the Invention

We have prepared a C-type natriuretic peptide (CNP) transgenic mouse, which expresses CNP, a guanyl cyclase B (GC-B) activator, systemically with elevated blood level of CNP, and then studied the effect of CNP on body height or on growth cartilage. As a result, we have now found that in the CNP transgenic mouse the increase in body height is accelerated, that the femoral growth plate cartilage becomes significantly thickened, and that, through the property analyses of such CNP transgenic mice, the increase in body height is accelerated by the effect of CNP on hematogenously in the absence of an abnormality in FGFR3.

Accordingly, the present invention comprises the following:

According to the first aspect, the present invention provides a composition for increasing a body height of an individual, comprising a GC-B activator as an active ingredient, the composition being to be administered to an individual free from FGFR3 abnormality.

In an embodiment of the invention, said composition is used for patients with short stature free from FGFR3 abnormality.

In another embodiment of the invention, said composition is used for individuals other than patients free from FGFR3 abnormality.

In another embodiment of the invention, said increase in body height is extension of cartilage bones.

In another embodiment of the invention, said increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.

In another embodiment of the invention, said activator is a peptide.

In another embodiment of the invention, the peptide is CNP or a derivative thereof.

In another embodiment of the invention, the CNP is CNP-22 or CNP-53 from mammals, including human, or birds.

According to another embodiment of the present invention, the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.

In another embodiment of the invention, said derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or 2 , while possessing a CNP activity.

According to the second aspect, the present invention provides a method for increasing a body height of an individual, comprising activating GC-B to increase the body height in an individual free from FGFR3 abnormality.

In an embodiment of the invention, said increase in body height is extension of cartilage bones.

In another embodiment of the invention, said increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.

In another embodiment of the invention, the GC-B is activated by CNP or a derivative thereof.

In another embodiment of the invention, the CNP is CNP-22 or CNP-53 from mammals, including human, or birds.

In another embodiment of the invention, the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.

In another embodiment of the invention, said derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or 2 , while possessing a CNP activity.

According to the third aspect, the present invention provides a method for screening of an agent for increasing the body height of an individual, comprising screening of candidate agents for an agent for increasing the body height using the activity of GC-B as an indication.

In an embodiment of the invention, the activity of GC-B is determined as an amount of produced intracellular cGMP.

In another embodiment of the invention, said method comprises, preparing a cultured cell line that has been forced to express GC-B, culturing the cell line in the presence or absence of a test substance, determining an amount of intracellular cGMP produced in the cell line, and screening candidate agents for an agent for increasing the body height of an individual using the difference, as an indication, in amounts of intracellular cGMP produced in the presence and absence of the test substance.

The present invention further provides a method for extending a cartilage bone free from FGFR3 abnormality in an individual comprising activating GC-B in the individual.

The specification of this application encompasses the contents as disclosed in the specification and/or drawings of

Japanese Patent Application No. 2004-107871, which is claimed as a priority of the application.

Brief Description of the Drawings

  • Fig. 1 shows the construction of a vector for preparing a CNP transgenic mouse. Fig. 1A: cDNA of the mouse CNP, which has been incorporated into pGEM-T Easy vector, was cut out with Pst I and blunt-ended at each end. Fig. 1B: pSG1 was treated with EcoR I and blunt-ended. Fig. 1C: The mouse CNP cDNA prepared in Fig. 1A was incorporated into the pSG1 obtained in Fig. 1B.
  • Fig. 2 shows a DNA fragment for injection. A fragment (about 2.3 kb) containing the CNP gene was cut out from pSG1-CNP prepared in Fig. 1C by digesting with Hind III and Xho I, and it was used as a fragment for injection.
  • Fig. 3 shows the results of a genotypical analysis of a CNP transgenic mouse. In the wild type mouse (WT) 3 signals (indicated as “Wild type CNP gene”) were detected, while in the transgenic mouse (Tgm) 2 signals (indicated as “Transgene”) derived from the transgene were detected in addition to the wild-type CNP gene.
  • Fig. 4 shows the growth curve of CNP transgenic mice on time. The naso-anal lengths of female CNP transgenic mice (TG) were significantly greater than those of a normal litter of female mice (WT) at 2 weeks old and thereafter (Fig. 4A). The naso-anal lengths of male CNP transgenic mice were significantly greater than those of a normal litter of male mice (WT) at 4 weeks old and thereafter (Fig. 4B). (*: p < 0.05; **: p < 0.01 vs. WT; unpaired Student’s t-test)
  • Fig. 5 shows thickening of growth cartilage in femora of CNP transgenic mice. Each thickness of the resting layer, proliferating layer and hypertrophic layer, and the total thickness of these layers in the CNP transgenic mice (CNP Tgm) were significantly greater than those of the normal littermates (Wild type). (*: p < 0.05; **: p < 0.01 vs. Wild type; unpaired Student’s t-test)

Detailed Description of the Invention
The present invention is further described with reference to the figures.

We analyzed the genotype of a CNP-transgenic mouse (CNP Tgm) produced as described later in Example 2 using Southern blotting. As a result, we detected 3 signals (“Wild type CNP gene”) in the wild type mouse, while detecting 2 signals (“Transgene”) derived from the transgene in the CNP Tgm in addition to the wild-type CNP gene, as shown in Fig. 3. The CNP levels in the liver, an organ expected to highly express said transgene, and in blood plasma were determined in order to study the expression of CNP in the CNP Tgm. As a result, it was found that the CNP Tgm showed about 10 fold and about 24 fold higher CNP levels in the liver and blood plasma, respectively, than the wild type, demonstrating statistically significant overexpression of CNP peptides (Table 1 in Example 4).

The naso-anal lengths of female and male CNP Tgms and normal litter were measured on time over a period of 2 to 9 weeks. As a result, the naso-anal lengths of the female and male CNP Tgms were greater than those of the normal litter, and the body heights of the CNP Tgms were more increased as well than the normal litter (Fig. 4A: female; Fig. 4B: male). Thus, it was confirmed that elevating a CNP level in blood resulted in acceleration of the increase in body height.

The thickness of the growth cartilage of CNP Tgm was histologically analyzed using the mean thickness of the resting layer, proliferating layer and hypertrophy layer of the growth cartilage on the patellar surface femur, and the total of the three layers (as the thickness of growth cartilage). As a result, it was confirmed that each thickness of the resting layer, proliferating layer and hypertrophy layer, and the total thickness thereof for CNP Tgm were greater with statistical significance than those of the wild type (Fig. 5). It was also demonstrated that CNP accelerates the increase in body height in animals by increasing each thickness of the resting layer, proliferating layers and hypertrophy layer of other cartilage bones, such as the tibiae, radiuses or ulnae, in addition to those of the cartilage bone of femora.

Thus, the present invention provides a composition for increasing a body height of an individual, comprising a GC-B activator as an active ingredient, the composition being to be administered to an individual free from FGFR3 abnormality.

In the present invention, the term “FGFR3 abnormality” refers to achondrogenesis or achondroplasia, which is caused by growth inhibition of cartilage bones resulting from mutations in the fibroblast growth factor receptor 3 (.FGFR3) gene, or achondrogenesis or achondroplasia caused by function control failure of FGFR3 or overexpression of FGFR3 gene resulting from mutations in the FGFR3 gene (

Japanese Patent Publication No. 2003-113116A; Nat. Med., 2004, 10(1): 80-86; and International Publication No.

WO 02/074234).

According to an embodiment of the invention, the composition is used for a patient with short stature free from FGFR3 abnormality. In the present invention, the term “short stature” refers to any short statured symptom or dwarfism which is not caused by FGFR3 abnormality, including for example (1) short stature caused by endocrine abnormalities, such as short stature caused by growth hormone hyposecretion (pituitary dwarfism) or short stature caused by hypothyreosis or adrenocortical hyperfunction; (2) short stature caused by non-endocrine abnormalities, such as familial short stature, fetal hypoplastic short stature, or short stature caused by chromosome abnormalities (e.g., Turner’s syndrome and Prader-Willi syndrome); and (3) secondary short stature caused by chemotherapy or radiation therapy.

According to another embodiment of the invention, the composition can be used for individuals free from FGFR3 abnormality other than patients with short stature. The present invention may be used for individuals free from FGFR3 abnormality other than patients with short stature, in the fields of cosmetics, medicine, and sports. Use for humans who have demands to increase their body heights is also within the scope of the invention.

Examples of the individuals who use of the present invention is intended include, but are not limited to, mammals including human, such as human, pig, and bovine. Preferred individual is a human.

According to another embodiment of the invention, the increase in body height is the extension of cartilage bones.

According to still another embodiment of the invention, the increase in body height is the extension of femora, tibiae, radiuses, and/or ulnae.

As used in the invention, the term “guanyl cyclase B (GC-B)” has the same meaning as natriuretic peptide receptor B (NPR-B).

As used in the invention, the term “activity of GC-B” has the same meaning as guanyl cyclase activity.

In the present invention, a guanyl cyclase B (GC-B) activator or GC-B activator is a peptide or a nonpeptidic low-molecular-weight compound, preferably a CNP peptide or a derivative thereof, that can bind to and activate GC-B, which is known as a CNP receptor. Peptides as used herein refer to a substance consisting of amide bond linkages of a plurality of (L-, D- and/or modified) amino acids, and include polypeptides and proteins. A GC-B activator can be identified, for example, by expressing a GC-B receptor in a cultured cell line such as COS-7, adding a candidate agent to the medium, culturing the cell line for a certain time period at a certain temperature (for example, 37°C, 5 minutes), and measuring the amount of intracellular cGMP produced (Science 1991, 252: 120-123). Using such an assay system, and using the amount of intracellular cGMP production as an indication, a GC-B activator may be identified and used in the present invention.

According to one embodiment of the invention, the GC-B activator is a peptide, and preferably CNP or a derivative thereof. Preferred CNP is selected from CNP-22 and CNP-53 from mammals, including human, or birds, and more preferably CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.

According to another embodiment of the invention, the CNP derivative as described above has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2, while possessing a CNP activity. Alternatively, the CNP derivative comprises a sequence having about 70% or more, about 80% or more, about 85% or more, about 90% or more, about 95% or more, about 97% or more, about 98% or more, or about 99% or more identity with the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2 and retains CNP activity.

The term “one or several” as used herein generally represents any integer between 1 and 10, preferably between 1 and 5, more preferably between 1 and 3. The “% identity” between two amino acid sequences may be determined using techniques well known to those skilled in the art, such as BLAST protein search (Altschul, S.F., Gish, W., Miller, W., Myers, E.W. & Lipman, D.J. (1990) “Basic Local Alignment Search Tool” J. Mol. Biol. 215:403-410).

Examples of CNPs usable in the present invention include CNPs from mammals including human (CNP-22: Biochem. Biophys. Res. Commun. 1990; 168: 863-870,

International Publication No. WO 91/16342, CNP-53: Biochem. Biophys. Res. Commun. 1990; 170:973-979,

Japanese Patent Publication No. 4-74198A (1992),

Japanese Patent Publication No. 4-139199A (1992),

Japanese Patent Publication No. 4-121190A (1992)), CNPs from birds (

Japanese Patent Publication No. 4-120094A (1992)), CNPs from amphibians (

Japanese Patent Publication No. 4-120095A (1992)), and CNP derivatives such as CNP analogous peptides disclosed in

Japanese Patent Publication No. 6-9688A (1994) and International Publication No.

WO 02/074234.

CNP-22 and CNP-53, which consist of 22 and 53 amino residues respectively, are known as naturally occurring CNPs. Because CNPs have a high homology in their sequences between birds and mammals including human, i.e. regardless of the kind of animals, CNPs from birds and mammals including human, preferably CNPs from mammals including human, and more preferably CNPs from human, can be used in the present invention. The amino acid sequence of human CNP-22 or CNP-53 has the sequence shown in SEQ ID NO: 1 or SEQ ID NO: 2 respectively, represented by:

  • Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Met Ser Gly Leu Gly Cys (human CNP-22; SEQ ID NO: 1); or
  • Asp Leu Arg Val Asp Thr Lys Ser Arg Ala Ala Trp Ala Arg Leu Leu Gln Glu His Pro Asn Ala Arg Lys Tyr Lys Gly Ala Asn Lys Lys Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Met Ser Gly Leu Gly Cys (human CNP-53; SEQ ID NO: 2),
  • each of which has an intramolecular disulfide bond, i.e. between 6-Cys and 22-Cys in human CNP-22 or between 37-Cys and 53-Cys in human CNP-53, forming a cyclic peptide structure.

Pig CNP-22 and rat CNP-22 have the same amino acid sequence as human CNP-22, whereas the amino acid residues at positions 17 and 28 are His and Gly, respectively, in pig CNP-53 and rat CNP-53, and they are Gln and Ala in human CNP-53, i.e., two amino acids are different in CNP-53 between human and pig or rat (

Japanese Patent Publication No. 4-139199A (1992),

Japanese Patent Publication No. 4-121190A (1992), and

Japanese Patent Publication No. 4-74198A (1992)). In addition, chicken CNP-22 has the same primary structure as human CNP-22, with the exception that the amino acid residue at position 9 is Val (

Japanese Patent Publication No. 4-120094A (1992)).

The CNPs usable in the invention include CNPs purified from natural sources, recombinant CNPs produced by known genetic engineering techniques, and CNPs produced by known chemical syntheses (for example, a solid phase synthesis using peptide synthesizer), preferably human CNP-22 and human CNP-53 produced by genetic engineering techniques. Production of human CNPs by genetic engineering techniques comprises, for example, the steps of incorporating the DNA sequence of human CNP-22 or CNP-53 (

Japanese Patent Publication No. 4-139199A (1992)) into a vector such as plasmid or phage, transforming the vector into a procaryotic or eucaryotic host cell, such as E. coli or yeast, and expressing the DNA in suitable culture medium, preferably allowing the cells to secrete the CNP peptide extracellularly, and collecting and purifying the CNP peptide produced. Polymerase chain reaction (PCR) technique can also be used to amplify target DNA.

Basic techniques such as genetic recombination, site-directed mutagenesis and PCR techniques are well-known to those skilled in the art, which are described, for example, in J. Sambrook et al., Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1990); Ausubel et al., Current Protocols In Molecular Biology, John Wiley & Sons (1998), and said techniques as disclosed therein may be used for the present invention. As the vectors, commercially available vectors or vectors as disclosed in publications may also be used.

CNP derivatives that may be used in the present invention have the CNP activity and have a cyclic peptide structure having a disulfide bond between two cysteine residues as seen in human CNP-22 or CNP-53. Examples of the CNP derivatives include: fragments of the CNPs as described above; peptides having a substitution of at least one amino acid by another amino acid in the CNPs above or fragments thereof; peptides having a deletion of at least one amino acid in the CNPs above or partial peptides thereof; and peptides having an addition of at least one amino acid in the CNPs above or partial peptides thereof. As used herein, the substitution, deletion or addition of amino acids means that a certain number of amino acids are substituted, deleted or added by a well-known method such as site-directed mutagenesis, with the proviso that the CNP activity is not lost. For example, the CNP-22 or CNP-53 derivatives have a substitution, deletion or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2, possessing the CNP activity.

In general, the substitution of amino acids is preferably a substitution between conservative amino acids. Conservative amino acids may be classified according to, for example, polarity (or hydrophobicity) or types of electric charges. Examples of nonpolar, uncharged amino acids include glycine, alanine, valine, leucine, isoleucine, proline, etc.; aromatic amino acids include phenylalanine, tyrosine and tryptophan; polar, uncharged amino acids include serine, threonine, cysteine, methionine, asparagine, glutamine, etc.; negatively charged amino acids include aspartic acid and glutamic acid; and positively charged amino acids include lysine, arginine and histidine.

The term “CNP activity” as used herein refers to the activity to act on GC-B to increase guanyl cyclase activity or the activity to significantly increase the body height of an individual. The CNP activity can be determined by measuring cellular guanyl cyclase activity, for example by measuring the amount of intracellular cGMP produced, or alternatively by administering a GC-B activator for a certain period to an animal such as mouse or rat and subsequently measuring the naso-anal length as described in Example 5 later.

Examples of CNP-22 analogous peptides include the following cyclic peptides as described in

Japanese Patent Publication No. 6-9688A (1994) and International Publication No.

WO 02/074234 (where underlines represent variations from human CNP-22).

  • Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ala Met Ser Gly Leu Gly Cys (SEQ ID NO: 3)
  • Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Gln Ser Gly Leu Gly Cys (SEQ ID NO: 4)
  • Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Ala Ser Gly Leu Gly Cys (SEQ ID NO: 5)
  • Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Met Ser Gly Leu Gly Cys (SEQ ID NO: 6) Ser Leu Arg Arg Ser Ser Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Met Ser Gly Leu Gly Cys (SEQ ID NO: 7)
  • Gly Leu Ser Lys Gly Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Met Ser Gly Leu Gly Cys Asn Ser Phe Arg Tyr (SEQ ID NO: 8)
  • Cys Phe Gly Leu Lys Leu Asp Arg Ile Gly Ser Gln Ser Gly Leu Gly Cys Asn Ser Phe Arg Tyr (SEQ ID NO: 9)
  • Cys Phe Gly Xaa Xbb XccAsp Arg Ile Gly Xdd Xee Ser Xff Xgg Gly Cys

(wherein Xaa = Leu, Ile, Val; Xbb = Lys, Leu, Met; Xcc = Leu, Ile, Ala, Val; Xdd = Ser, Ala, Gly, Thr, Asn; Xee = Met, Ala, Trp, His, Lys, Ser, Gly; Xff = Gly, Lys, Ala, Leu; Xgg = Leu, Met) (SEQ ID NO: 10).Examples of CNP-53 analogous peptides include cyclic peptides comprising amino acid variations similar to those of the CNP-22 analogous peptides.

The present invention also provides a method for increasing a body height in an individual, comprising activating GC-B to increase the body height in an individual free from FGFR3 abnormality. This invention is based on the finding that an GC-B activator can increase the body height of an individual free from FGFR3 abnormality. Specifically, the increase in body height is the extension of cartilage bones. More specifically, the increase in body height is the extension of femora, tibiae, radiuses, and/or ulnae. Specific examples of the GC-B activator are the CNPs or derivatives thereof as defined above. The CNP is preferably CNP-22 or CNP-53 from mammals, including human, or birds, and more preferably CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2. The CNP derivatives have a substitution, deletion or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2, while possessing a CNP activity. Other GC-B activators can be identified, for example, by expressing a GC-B receptor in a cultured cell line such as COS-7, adding a candidate agent to the medium, culturing the cell line for a certain time period at a certain temperature (for example, 37°C, 5 minutes), and measuring the amount of intracellular cGMP produced (Science 1991, 252: 120-123). Thus, using such an assay system and using the amount of produced intracellular cGMP as an indication, a GC-B activator can be identified and used for the present invention.

The present invention further provides a method for screening an agent for increasing the body height of an individual, comprising screening candidate agents for an agent for increasing the body height using the activity of GC-B as an indication. According to an embodiment of this invention, the GC-B can be activated by the CNPs as defined above or derivatives thereof. Because the GC-B is known to catalyze the synthesis of the second messenger cGMP from GTP through guanyl cyclase activity, the GC-B activity can be determined as an amount of produced intracellular cGMP.

According to another embodiment of this invention, the method comprises preparing cultured cells that express GC-B or cells from articular chondrocytes, culturing the cells in the presence of a candidate agent, and screening candidate agents for an agent for increasing body heights using the activity of GC-B in the cells as an indication.

According to preferred embodiment of this invention, the method comprises preparing a cultured cell line that has been forced to express GC-B, culturing the cell line in the presence or absence of a test substance, determining an amount of intracellular cGMP produced in the cell line, and screening of candidate agents for an agent for increasing the body height of an individual using the difference, as an indication, in amounts of intracellular cGMP produced in the presence and absence of the test substance.

In the screening method of the present invention, it may comprise screening for an agent for increasing the body height by, for example, expressing a GC-B receptor in a cultured cell line such as COS-7, adding a candidate agent to the medium, culturing the cell line for a certain time period at a certain temperature (for example, 37°C, 5 minutes), and measuring the amount of intracellular cGMP produced (Science 1991, 252: 120-123).

Furthermore, the present invention provides a method for extending a cartilage bone free from FGFR3 abnormality in an individual, comprising activating GC-B in the individual. According to an embodiment of this invention, the extension of cartilage bones can be accelerated in vivo, ex vivo or in vitro through the activation of GC-B. According to a referred embodiment of the invention, the method comprises accelerating the extension of cartilage bones free from FGFR3 abnormality by adding a GC-B activator, when culturing bone or cartilage.

The composition of the present invention is formulated into preparations for oral or parenteral administration by combining the GC-B activator defined above as an active ingredient with a pharmaceutically acceptable carrier, excipient, additives, or the like.

The composition of the present invention comprises the GC-B activator defined above as an active ingredient, and further comprises a carrier, excipient, and other additives that are used in conventional manufactures of medicaments.

Examples of the carriers and excipients for preparation include lactose, magnesium stearate, starch, talc, gelatin, agar, pectin, gum arabic, olive oil, sesame oil, cacao butter, ethylene glycol, and others conventionally used.

Examples of solid compositions for oral administration include tablets, pills, capsules, powders, granules, and the like. In such solid compositions, at least one active ingredient is mixed with at least one inert diluent, such as lactose, mannitol, glucose, hydroxypropylcellulose, microcrystal cellulose, starch, polyvinylpyrrolidone, magnesium aluminometasilicate, or the like. The composition may, according to a conventional method, also contain additives other than inert diluents, for example, a lubricant such as magnesium stearate, a disintegrating agent such as fibrous calcium glycolate, and a dissolution auxiliary agent such as glutamic acid or aspartic acid. Tablets or pills may, as required, be coated with a glycocalyx, such as sucrose, gelatin or hydroxypropyl methylcellulose phthalate, or with a gastro- or enteric-film, or with two or more layers. Capsules of an absorbable material, such as gelatine, are also included.

Liquid compositions for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs, and may also contain conventional inert diluents, such as purified water and ethanol. The composition may contain, other than the inert diluent, an adjuvant, such as wetting and suspending agents, a sweetening agent, a flavor, an aromatic ,and a preservative.

Examples of parenteral injections include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of aqueous solutions and suspensions include water for injection and physiological saline for injection. Examples of non-aqueous solutions and suspensions include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, alcohols such as ethanol, and polysorbate 80®. These compositions may further contain adjuvants, such as preservatives, wetting agents, emulsifiers, dispersants, stabilizers (for example, lactose), and dissolution auxiliary agents (e.g., glutamic acid and aspartic acid). The above-described materials may be sterilized by conventional sterilization methods, such as filter sterilization with a microfiltration membrane, heat sterilization such as autoclaving, or incorporation of disinfectants. Injections may be liquid preparations, or freeze-dried preparations that may be reconstituted before use. Examples of excipients for freeze-drying include sugar alcohols and sugars, such as mannitol and glucose.

The therapeutic or prophylactic agent of the present invention is administered by either oral or parenteral administration methods commonly used for pharmaceuticals. Preferred are parenteral administration methods, for example, injection (e,g., subcutaneous, intravenous, intramuscular and intraperitoneal injections), percutaneous administration, trans-mucosal administration (e.g., transnasal and transrectal), and trans-pulmonary administration. Oral administration may also be used.

The dosage of a GC-B activator, preferably a CNP as defined above or a derivative thereof, which is an active ingredient contained in the composition of the present invention, may be determined depending on the type of disease to be treated, the severity of the disease, patient’s age, and the like, and may generally range from 0.005 µg/kg to 100 mg/kg, preferably from 0.02 µg/kg to 5 mg/kg. However, the dosage of the pharmaceutical composition containing a CNP activator according to the present invention is not limited thereto.

The present invention includes, but is not limited to, the following.

  • (1) A composition for increasing a body height of an individual, comprising a guanyl cyclase B (GC-B) activator as an active ingredient, the composition being to be administered to an individual free from FGFR3 abnormality.
  • (2) The composition of (1) above, for use in a patient with short stature.
  • (3) The composition of (1) above, for use in an individual other than patients with short stature.
  • (4) The composition of (1) above, wherein the increase in body height is extension of cartilage bones.
  • (5) The composition of (1) above, wherein the increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.
  • (6) The composition of (1) above, wherein the activator is a peptide.
  • (7) The composition of (6) above, wherein the peptide is C-type natriuretic peptide (CNP) or a derivative thereof.
  • (8) The composition of (7) above, wherein the CNP is CNP-22 or CNP-53 from mammals including human, or birds.
  • (9) The composition of (7) above, wherein the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.
  • (10) The composition of (7) above, wherein the derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: I or 2, while possessing a CNP activity.
  • (11) A method for increasing a body height of an individual, comprising activating GC-B to increase the body height in an individual free from FGFR3 abnormality.
  • (13) The method of (11) above, wherein the increase in body height is extension of femora, tibiae, radiuses, and/or ulnae.
  • (14) The method of (11) above, wherein the GC-B is activated by CNP or a derivative thereof.
  • (15) The method of (14) above, wherein the CNP is CNP-22 or CNP-53 from mammals, including human, or birds.
  • (16) The method of (14) above, wherein the CNP is CNP-22 of SEQ ID NO: 1 or CNP-53 of SEQ ID NO: 2.
  • (17) The method of (14) above, wherein the derivative has a deletion, substitution or addition of one or several amino acids in the amino acid sequence of SEQ ID NO: 1 or 2 , while possessing a CNP activity.
  • (18) A method for screening an agent for increasing the body height of an individual, comprising screening candidate agents for an agent for increasing the body height using the activity of GC-B as an indication.
  • (19) The method of (18) above, which comprises preparing cultured cells that express GC-B or cells from articular chondrocytes, culturing the cells in the presence of a candidate agent, and screening candidate agents for an agent for increasing body heights using the activity of GC-B in the cells as an indication.
  • (20) The method of (18) above, wherein the activity of GC-B is determined as an amount of produced intracellular cGMP.
  • (21) The method of (18) above, wherein it comprises preparing a cultured cell line that has been forced to express GC-B, culturing the cell line in the presence or absence of a test substance, determining an amount of intracellular cGMP produced in the cell line, and screening candidate agents for an agent for increasing the body height of an individual using the difference, as an indication, in amounts of intracellular cGMP produced in the presence and absence of the test substance.
  • (22) A method for extending a cartilage bone free from FGFR3 abnormality in an individual, comprising activating GC-B in the individual.

The present invention will be described in more detail by the following examples, which are for illustrative purposes only and are not intended to limit the scope of the invention. Thus, the present invention is not limited to those examples.

Examples

Example 1: Construction of vector for preparing CNP transgenic mouse

As shown in Fig. 1A, the murine CNP cDNA (526 bp; FEBS Lett. 276:209-213, 1990) was subcloned into pGEM-T easy vector (Promega), and was then cut with Pst I and blunt-ended to prepare a mouse CNP cDNA. The vector PSG 1 (Promega; Fig. 1B) was cut with EcoRI, blunt-ended and ligated with the murine CNP cDNA, as shown in Fig. 1C, to prepare a SAP-mCNP vector (pSG1-CNP).

Example 2: Production of CNP transgenic mouse

A DNA fragment for injection was prepared as follows. The SAP-mCNP vector (pSG1-CNP; Fig. 1C) with an inserted CNP gene was first treated with Hind III and Xho I to cut out a fragment (about 2.3 kb) containing the CNP gene. The fragment was then collected using Gel Extraction Kit (QIAGEN), and was diluted with PBS at a concentration of 3 ng/µl, thereby obtaining the DNA fragment for injection (Fig. 2).

The mouse egg at pronucleus stage into which the DNA fragment was injected was collected as follows. First, a C57BL/6 female mouse (Clea Japan, Inc.) was injected intraperitoneally with 5 i.u pregnant mare serum gonadotropin (PMSG), and 48 hours later, with 5 i.u human chorionic gonadotropin (hCG), in order to induce superovulation. This female mouse was crossed with a congeneric male mouse. In the next morning of the crossing, in the female mouse the presence of a plug was confirmed and subsequently the oviduct was perfused to collect a mouse egg at pronucleus stage.

The DNA fragment for injection was injected into the pronucleus egg using a micromanipulator (Latest Technology in Gene Targeting (Yodosha, Japan), 190-207, 2000). Specifically, the DNA fragment was injected into 660 C57BL/6J embryos, and on the following day, 561 embryos at 2-cell stage were transplanted into the oviducts of recipient females on day 1 of false pregnancy at about 10 per each side of the oviduct (about 20/animal).

Recipient females, which had not been delivered of offsprings by the expected date of delivery, were subjected to cesarean section, resulting in the birth of offsprings which were raised by a foster mother. Total 136 offsprings were obtained, 5 of which were transgenic mice with an introduced CNP gene (hereafter referred to as “Tgm”). Hereinafter, the mouse initially obtained is referred to as the Founder.

All Founder mice were male, and the subsequent generation of offsprings (i.e., F1 mice) were obtained from four of the five lines.

Example 3: Genotype analysis of CNP transgenic mouse

Genotype analysis was performed by Southern blotting according to procedures as described below.

The tail (about 15 mm) was taken from the 3-week old mouse and treated with proteinase K (at 55°C, with shaking at 100 rpm over day and night) to obtain a lysis solution. The obtained solution was then subjected to an automated nucleic acid separator (KURABO NA-1000; Kurabo, Japan) to prepare genomic DNA. The genomic DNA (15 µg) was treated with Pvu II (200 U), then with phenol-chloroform to remove the restriction enzyme, and was precipitated with ethanol to collect the DNA. The obtained DNA was dissolved in 25 µL of TE and subjected to electrophoresis on 0.7% agarose gel (at 50V constant voltage), then the gel was treated with 0.25M HCl solution for 15 minutes to cleave the DNA, washed with water, and blotted overnight onto a nylon membrane in 0.4M NaOH solution. Thereafter, the DNA on the membrane was fixed by the UV crosslink method. The membrane was treated (at 42°C for 2 hours) with a hybridization solution (50% formamide, 0.5x Denhardt’s, 0.5% SDS, 5x SSPE), and a 32P labeled probe, which has been prepared with BcaBEST Labeling Kit (TaKaRa, Japan) using the CNP cDNA (about 0.5 kb) as a template, was added to the membrane for effecting hybridization at 42°C overnight. After treatment with a detergent solution (2x SSC, 0.1% SDS) at 55°C for 20 minutes, the membrane was exposed to Imaging Plate (Fuji Film) overnight to detect signals of the transgene using BAS2000 (Fuji Film, Japan) (Fig. 3). In the wild-type mouse (WT) 3 signals (wild-type CNP gene) were detected, while in the transgenic mouse (Tgm) 2 signals (transgene) derived from the transgene were detected in addition to the wild-type CNP gene.

Example 4: CNP expression in CNP transgenic mouse

A CNP-22 EIA measuring kit (PHOENIX PHARMACEUTICALS INC.) was used for the determination of a CNP level.

Three each of 7-week old male and female CNP transgenic mice, as well as 3 each of male and female normal litter of mice, were euthanized by exsanguination from the postcava under ether anesthesia.

The liver, which is an organ expected to exhibit high expression of the transgene, was removed, and the EIA assay buffer from the measuring kit as above was added at 1 ml per 0.1g of liver weight, followed by cooling on ice. The liver was homogenized in a Waring blender (Physcotron), and after centrifugation (at 2,000 rpm for 5 minutes), the supernatant was used as a sample for the determination of CNP-22 levels.

One mg of ethylenediaminetetraacetate-4Na (Junsei Chemical Co., Ltd., Japan) and 2 trypsin-inhibition units of aprotinin (Sigma) were added to the drawn blood and agitated to separate blood plasma, which was used as a sample for the determination of CNP-22 levels.

The results are shown in Table 1.

Table 1: CNP expression in CNP transgenic mouse
Liver (ng/g tissue) mean±SD Plasma (ng/mL) mean±SD
Wild type No.1 38.8 29.3±20.5 0.3 0.3±0.06
No.2 5.9 0.4
No.3 43.3 0.3
CNP tgm No.1 293.3 290±81.7** 10.3 8.0±4.7#
No.2 370.0 11.1
No.3 206.7 2.6
** : p<0.01 (unpaired Student’s t-test)
# : p<0.05 (Wilcoxon rank sum test)

 

The CNP transgenic mouse showed about 10 fold and about 24 fold higher CNP-22 level in the liver and blood plasma respectively, than the wild type when the mean ± SD values were compared between them. In each case the difference was statistically significant. It was confirmed, from the results, that the CNP peptide was overexpressed in the CNP transgenic mouse.

Example 5: Growth curve of CNP transgenic mouse

The naso-anal lengths (in cm) of female and male CNP transgenic mice, as well as those of female and male normal littermates, were measured on time over a period of 2 to 9 weeks (Fig. 4). As a result, the naso-anal lengths of the female and male CNP transgenic mice were both greater than those of the normal littermates, demonstrating that the increase in body height has been accelerated. This also demonstrates that the increase of a CNP level in the blood results in the acceleration of increasing the body height.

Example 6: Histological analysis of the growth cartilage of CNP transgenic mouse

For the purpose of analyzing the thickness of the growth cartilage histlogically, 5 each of 9-week old female CNP transgenic mice and female normal litter of mice were euthanized by exsanguination from the postcava under ether anesthesia, and the thigh bone was fixed in 20% formalin for a week. After soaking in a 20% aqueous solution of EDTA-4Na (pH 7.4) (Junsei Chemical Co., Ltd., Japan) for decalcification, the patellar surface femur was subjected to a midline sagittal section and embedded in paraffin by conventional method to prepare a paraffin block. A 4 µm-thick section was further sectioned with a microtome to prepare paraffin sections, which were stained with hematoxylin and eosin. For the thickness of the growth cartilage, one microscopic field observed using an objective lens (x 10) was incorporated into an image analysis software (IPAP, Sumika Technoservice, Japan), and each thickness of the resting layer, proliferating layer and hypertrophic layer was measured at 5 points in the microscopic field using the same software, and the calculated mean value thereof was taken as the thickness of each layer of the individual. The total of the three layers was regarded as the thickness of the growth cartilage of the individual. Mean values and standard deviations for these items were calculated between the CNP transgenic mice and the normal littermates (using Microsoft Excel 2000, Microsoft), and statistical analysis was performed using the unpaired Student’s t-test (SAS ver. 6.12; SAS Institute Japan, Japan).

This statistical analysis revealed that, for each thickness of the resting layer, proliferating layer and hypertrophic layer, and the total thickness thereof in CNP transgenic mice (CNP Tgm), the thicknesses of the growth cartilage layers were greater with statistical significance than those of the normal mice (wild type) (Fig. 5). From the results, it was also demonstrated that the CNP, a GC-B activator, promotes the increase of the body height in mammals by increasing the thickness of each layer of growth cartilage.

Industrial Applicability

The composition of the present invention, whcih comprises a GC-B activator as the active ingredient, enables the treatment of short statured symptoms, such as endocrine abnormality-caused short stature, non-endocrine abnormality-caused short stature, and secondary short stature, in an individual free from FGFR3 abnormality. The composition of the invention imposes less burden and pain on a patient, when compared with injection of growth hormones or insulin-like growth factor-I (IGF-I) or with orthopedic surgeries such as ostectomy. So, the composition can be an excellent therapeutic agent, which is beneficial for patient’s QOL. It can also be used for increasing the body height in an individual free from FGFR3 abnormality and other than patients with short stature. Additionally, the present invention enables the extension of cartilage bones free from FGFR3 abnormality in vivo, ex vivo, or in vitro through the activation of GC-B.

All publications, patents, and patent applications cited herein are incorporated herein by reference in their entirety.

Free text of Sequence Listing

  • Description in SEQ ID NO: 1: A disulfide bond is formed between 6-Cys and 22-Cys.
  • Description in SEQ ID NO: 2: A disulfide bond is formed between 37-Cys and 53-Cys.
  • Description of artificial sequence in SEQ ID NO: 3: CNP-22 derivative, where a disulfide bond is formed between 6-Cys and 22-Cys.
  • Description of artificial sequence in SEQ ID NO: 4: CNP-22 derivative, where a disulfide bond is formed between 6-Cys and 22-Cys.
  • Description of artificial sequence in SEQ ID NO: 5: CNP-22 derivative, where a disulfide bond is formed between 6-Cys and 22-Cys.
  • Description of artificial sequence in SEQ ID NO: 6: CNP-22 derivative, where a disulfide bond is formed between 1-Cys and 17-Cys.
  • Description of artificial sequence in SEQ ID NO: 7: CNP-22 derivative, where a disulfide bond is formed between 7-Cys and 23-Cys.
  • Description of artificial sequence in SEQ ID NO: 8: CNP-22 derivative, where a disulfide bond is formed between 6-Cys and 22-Cys.
  • Description of artificial sequence in SEQ ID NO: 9: CNP-22 derivative, where a disulfide bond is formed between 1-Cys and 17-Cys.
  • Description of artificial sequence in SEQ ID NO: 10: CNP-22 derivative, where 4-Xaa=Leu, Ile, Val; 5-Xaa=Lys, Leu, Met; 6-Xaa=Leu, Ile, Ala, Val; 11-Xaa=Ser, Ala, Gly, Thr, Asn; 12-Xaa=Met, Ala, Trp, His, Lys, Ser, Gly;14-Xaa=Gly, Lys, Ala, Leu; 15-Xaa=Leu, Met and where a disulfide bond is formed between 1-Cys and 17-Cys.        

Controlling a Stem Cell’s Form Can Determine Its Fate

From Science Daily

Shaping Up: Controlling a Stem Cell’s Form Can Determine Its Fate

ScienceDaily (Sep. 20, 2011) — “Form follows function!” was the credo of early 20th century architects making design choices based on the intended use of the structure. Cell biologists may be turning that on its head. New research by a team working at the National Institute of Standards and Technology (NIST) reinforces the idea that stem cells can be induced to develop into specific types of cells solely by controlling their shape. The results may be important to the design of materials to induce the regeneration of lost or damaged tissues in the body.

Tissue engineering seeks to repair or re-grow damaged body tissues, often using some form of stem cells. Stem cells are basic repair units in the body that have the ability to develop into any of several different forms. The NIST experiments looked at primary human bone marrow stromal cells, adult stem cells that can be isolated from bone marrow and can “differentiate” into bone, fat or cartilage cells, depending.

“Depending on what?” is one of the key questions in tissue engineering. How do you ensure that the stem cells turn into the type you need? Chemical cues have been known to work in cases where researchers have identified the proper additives — a hormone in the case of bone cells. Other research has suggested that cell differentiation on flat surfaces can be controlled by patterning the surface to restrict the locations where growing cells can attach themselves.

The experiments at NIST are believed to be the first head-to-head comparison of five popular tissue scaffold designs to examine the effect of architecture alone on bone marrow cells without adding any biochemical supplements other than cell growth medium. The scaffolds, made of a biocompatible polymer, are meant to provide a temporary implant that gives cells a firm structure on which to grow and ultimately rebuild tissue. The experiment included structures made by leaching and foaming processes (resulting in microscopic structures looking like clumps of insect-eaten lettuce), freeform fabrication (like microscopic rods stacked in a crisscross pattern) and electrospun nanofibers (a random nest of thin fibers). Bone marrow stromal cells were cultured on each, then analyzed to see which were most effective at creating deposits of calcium — a telltale of bone cell activity. Microarray analysis also was used to determine patterns of gene expression for the cultured cells.

The results show that the stem cells will differentiate quite efficiently on the nanofiber scaffolds — even without any hormone additives — but not so on the other architectures. The distinction, says NIST biologist Carl Simon, Jr., seems to be shape. Mature bone cells are characteristically long and stringy with several extended branches. Of the five different scaffolds, only the nanofiber one, in effect, forces the cells to a similar shape, long and branched, as they try to find anchor points. Being in the shape of a bone cell seems to induce the cells to activate the genes that ultimately produce bone tissue.

“This suggests that a good strategy to design future scaffolds would be to take into account what shape you want to put the cells in,” says Simon, adding, “That’s kind of a tall order though, you’d have to understand a lot of stuff: how cell morphology influences cell behavior, and then how the three-dimensional structure can be used to control it.” Despite the research still to be done on this method, the ability to physically direct cell differentiation by shape alone potentially would be simpler, cheaper and possibly safer than using biochemical supplements, he says.

The work was supported in part by the National Institute of Dental and Craniofacial Research, National Institutes of Health.

Me: So apparently you can get the right type of stem cell differentiation by using scaffolds of certain designs. You manage to control the form it goes into, you can control the type the stem cells turns into.

CartiHeal And Agili-CTM, a Single Stage Arthroscopic Cartilage Regeneration Implant

I found this article post that says that the company CartiHeal from Israel has gotten funding to continue their need in developing an innovative cell-free technology for regenerating hyaline cartilage. Found HERE

I do realize that the technology is mainly going to be used for the articular cartilage at the end of long bones, but I don’t see how this type of technology can’t be used as a form of implant since it is suppose to regenerate hyaline cartilage, .


CartiHeal Receives Additional Funding to Prepare for Market Launch of Agili-CTM, a Single Stage Arthroscopic Cartilage Regeneration Implant

TEL AVIV, Israel–(BUSINESS WIRE)–CartiHeal (2009) Ltd, a privately held medical device company developing an innovative cell-free technology for regenerating hyaline cartilage, raised up to $10 million, including $5 million in cash and $5 million in options. Half of the sum was raised from new investor, Elron, and the remainder from previous investors, Accelmed and Access Medical Ventures. The financing will be used to accelerate the development of CartiHeal’s technology portfolio, including its leading product, the Agili-CTM, as well as support ongoing clinical studies.

“In our post-marketing studies, initial clinical outcomes are very promising. Biopsies and MRIs show the regeneration of hyaline cartilage as early as 6-12 months following implantation. This is nothing short of a technological breakthrough”

CartiHeal’s Agili-CTM is a single step arthroscopically-placed implant, indicated for repairing cartilage and osteochondral defects. Agili-CTM has demonstrated, in both animal and human studies, an unprecedented ability to regenerate true hyaline cartilage—confirmed by the presence of Type II collagen—without the use of growth factors, stem cells or cell expansion techniques. These results have been confirmed by histological analysis performed by NAMSA, an independent research laboratory. The Agili-CTM is CE marked and CartiHeal is currently conducting post-approval clinical studies at leading centers in Europe.

Research has shown that injuries to the articular cartilage, if left untreated, lead to progressive joint changes and early onset of osteoarthritis. The most common treatments for articular cartilage injuries (e.g. microfracture and osteochondral grafting) fall short, since they do not result in regeneration of hyaline cartilage. Agili-CTM offers the 1.2 million patients undergoing cartilage repair annually worldwide, the potential for healing the defect with hyaline rather than “hyaline-like” fibrocartilage, possibly preventing the need for future joint replacement surgery in these patients.

“In our post-marketing studies, initial clinical outcomes are very promising. Biopsies and MRIs show the regeneration of hyaline cartilage as early as 6-12 months following implantation. This is nothing short of a technological breakthrough,” reports Nir Altschuler, Founder and CEO of CartiHeal. “We welcome our new investor and believe Elron’s extensive experience will assist us to become a leading player in the field of cartilage regeneration.”

Ari Bronshtein, CEO of Elron, commented, “The investment in CartiHeal attests to our commitment to continue investing in medical device companies based on revolutionary technology with significant potential. Elron’s know-how and experience in the biomed field, together with existing investors Accelmed and Access Medical and the Company’s management as well as its scientific advisory board, will contribute to CartiHeal’s advancement and help it achieve its goals.

About CartiHeal

Founded in 2009, CartiHeal is a privately held medical device company that develops proprietary implants for the regeneration of cartilage and bone disorders following sports injuries, trauma or degenerative joint changes. CartiHeal is a spin-out of the Department of Biotechnology Engineering at Ben-Gurion University, Israel.

About Elron

Elron is a leading holding company dedicated to building technology companies, primarily in the field of medical devices. Investments in med-tech companies during the last five years totaled over $150 million.

Elron’s portfolio companies include: Given Imaging, a world leader in developing and marketing diagnostic products for visualizing and detecting disorders of the GI tract; Pocared Diagnostics, which is developing a real-time and automated system for infectious diseases diagnosis using optical technology; BrainsGate, which is developing a system for treating ischemic stroke; Kyma, which is developing a remote patient monitoring system for chronic heart failure patients, and; SmartWave, which is developing a fully automatic implantable atrial defibrillator.

Contacts

CartiHeal
Nir Altschuler, CEO, +972-3-9085000
info@cartiheal.com

 

Comparing Pilates, Alexander Technique, And Chiropractor. Which Method Gives The Most Height Increase?

I found this article from the Daily Mail UK about a woman who had decided to take the three paths of pilates, alexander technique, and chiropractor and see which one could give her the most extra height.

It is from HERE. Again, I highlighted the most interesting parts . Hope you like it.


Can you grow an inch in a week?

by ALICE ROBINSON, Daily Mail

Modern life is making us lose height. This is the disturbing conclusion of health experts who have the unenviable task of righting the damage we do to our backs in the course of our everyday lives. ‘We spend far too much time slumped over computers, wearing high heels and idling around on sofas watching TV,’ says Sue Wakefield, executive director of the British Chiropractic Association. As a direct result, most of us may well be standing up to 2in shorter than we need to be.

Back problems are costing British firms 10 million working days a year and making us an unhappy and short nation in constant pain. Having woken every morning for two years with an aching neck and feeling permanently irritable, Alice Robinson, 5ft 4in, set out to find a cure. She tried three methods of realignment to see if she could gain a healthy spine – and extra inches. Her results may surprise you.

Pilates

Developed last century by Joseph H. Pilates – a German fitness enthusiast – and now fashionable among celebrities such as Sharon Stone, Madonna and Julia Roberts, Pilates was originally used to help injured dancers and athletes.

It is used to rectify poor posture by means of exercises that strengthen the spine, and stretch the vertebrae, which increases height, improves the circulation and opens up the joints.

This was initially only practised in studios, using beds to which pupils were loosely strapped, so that specific areas could be isolated and manipulated without moving other parts of the body.

However, ex-dancer and Pilates tutor Glenda Taylor has developed exercises that can be done for 10 to 15 minutes a day at home.

According to Taylor, Pilates is, in addition, like ironing out the spine, and a marvellous way of improving the way you look.

‘If your body is concertina-ed down, you necessarily look fatter. When you stand correctly, because you are standing taller, you are stretching your fat over a larger surface area – which gives the impression of being slimmer.’

Glenda agreed to give me three lessons to see how much difference we could make to my height.

During our first session, we concentrated on stretching out the spine while facing upwards. As I followed her instructions, trying to co-ordinate my arms and legs, I realised just how little control I have over virtually every part of my body.

We began by sitting in a yoga-like position, gradually dipping my head further and further towards my legs. Then Glenda took me through a variety of exercises such as sitting with one leg tucked up to my bottom and one stretched out, then curving my arms towards my toes to elongate the spine.

During the second session, Glenda taught me exercises that concentrated on lying on my stomach and arching the spine. The third session was devoted to stretching and limbering up to give suppleness, using household objects to perform the exercises.

Verdict: 1/2 inches (so that is 0.5 inches with 3 sessions)

After just three sessions, I had grown half an inch, and also found it easier to pull in my stomach muscles – an added bonus. However, with Pilates and the Alexander Technique, the effects can only be maintained with regular practice.

Alexander Technique

The Alexander Techinque has 800 practitioners in Britain, and the numbers are growing.

It was developed by Frederick Matthias Alexander, a 19th century actor, who lost full use of his voice through chronic laryngitis. He realised that muscular tension was causing this problem and developed a method of releasing it and allowing the spine to lengthen.

Practitioners claim some people grow as much as 2in through the technique. I put their claims to the test at Noel Kingsley’s London practice, with three sessions over the course of a week.

‘We all become accustomed to standing incorrectly,’ explains Noel. ‘Children are born with naturally perfect posture, but as we get older, we develop unconscious movement habits – such as slouching, or carrying heavy bags.

‘These affect our sense of wellbeing as well as causing physical problems, and the Alexander Technique is a way of releasing unwanted muscular tension that has accumulated over years of stressful living.

I was thrown by Noel’s first comment. He explained that when you stand correctly the head will feel as if it is tipped slightly forward with your chin pointing down, so you are not resting the weight of your head on the spine.

I then had to relax and Noel gently pushed down my shoulders and ran his hands up my neck. As I forced myself to stop resisting the action it is incredibly difficult to relinquish total control of your limbs to someone else – I could feel the muscles stretching and elongating.

The release of blood and oxygen made my whole body tingle, and after the first session I almost floated down Oxford Street and wafted onto the Tube.

Having been shown how to balance my head in the right way, I found it easy not to slip back into my former rigid stance. Over the next two sessions Noel showed me how I should stand (balanced), sitting (straight) and walking (in a much more relaxed fashion).

Having analysed my movement pattern, he used touch to guide me into the right positions, using his hands to gently push my shoulders down, my bottom in and my head forward. Ideally, the technique is taught to you over a series of lessons – Noel recommends 15.

The verdict: one inch   (wow, 3 sessions lead to 1.0 extra inch!!)

After my third and final session, I measured myself and was astounded to discover that I had grown an inch and could gaze down at the world from the lofty height of 5ft 5in. Not quite Kate Moss, but almost.

Chiropractor

Chiropractic involves diagnosing and treating disorders of the joints, muscles and bones. Minor displacements of the spinal bones can cause stress to the whole body as it compensates for misalignments.

One of the main treatments used is manipulation – where the joints are mobilised or stretched using a gentle motion to improve or restore normal function.

Although I gained just over a quarter of an inch from my sessions with Antoni Jakobowski – a chiropractor for more than ten years who sorts out the back problems of the U.S.

Having never contemplated whether my spine was in good condition, during my initial consultation I realised that I had spent two years with a constant nagging pain in my neck and right shoulder.

Bones and discs need motion to keep them healthy, otherwise calcium deposits form on the joints, making them harder to move.

The muscles surrounding the joints start to tense in order to try to stabilise the weak joint, and you start to feel pain.

Jakobowski explains that how much height you gain depends on where the problem is. ‘If you need work on your lower back, you will gain more height because the curve of your spine changes.’ When a bone is out of position the body leans forward to compensate.

Having worked out where the problem areas might be, he uses a nervoscope – a heat sensitive thermometer that identifies areas of inflammation.

Because blood is rushing to these areas, they are hotter. Running the nervoscope up and down my spine, it transpired that I had two vertebrae functioning improperly in my neck – from clutching the phone between my neck and shoulder.

Jakobowski placed his hands on my chin and neck and twisted it quickly to realign my neck vertebrae. There was a disconcerting popping sound, caused apparently by trapped gas being emitted from the joint, and the pain in my neck and shoulder disappeared instantly.

The verdict: one-quarter of an inch    (so 1 session with a chiropractor leads to 0.25 inches)

Not only was I standing a quarter of an inch taller, but I felt much more energetic and even-tempered. I wouldn’t hesitate to visit a chiropractor again. Depending on factors such as weight and age, the effect should be longer lasting.

Me: It looks like from the three methods, The alexander method lead to the most change of 1 inch (2.54 cms). However, I would be willing to bet that if the women kept on doing the pilates exercises or the chiropractor sessions, her height gain could have been more than just a fraction of an inch. Overall, they all seem to work in fixing standing and posture issues. 

Exercise has an affect on articular cartilage

The chondrogenic response to exercise in the proximal femur of normal and mdx mice.

“Submaximal exercise is used in the management of muscular dystrophy. The effects of mechanical stimulation on skeletal development are well understood, although its effects on cartilage growth have yet to be investigated in the dystrophic condition. The objective of this study was to investigate the chondrogenic response to voluntary exercise in dystrophin-deficient mice.
Control and dystrophin-deficient (mdx) mice were divided into sedentary and exercise-treated groups and tested for chondral histomorphometric differences at the proximal femur.
Control mice ran 7 km/week further than mdx mice on average, but this difference was not statistically significant. However, exercised control mice exhibited significantly enlarged femur head diameter, articular cartilage thickness, articular cartilage tissue area, and area of calcified cartilage relative to sedentary controls and exercised mdx mic{the enlarged femur head is consistent with articular cartilage endohcondral ossification [and] especially considering the area of calcified cartilage increased as well]. No differences were found between other treatment groups.
Mdx mice exhibit a reduced chondrogenic response to increased mechanical stimulation relative to controls. However, no significant reduction in articular dimensions was found, indicating loss of chondral tissue may not be a clinical concern with dystrophinopathy.”

“Exercise treatment consisted of voluntary access to a running wheel that lasted four weeks.”

“it is conceivable that joint forces in mdx mice failed to provide sufficient mechanical pressure to yield significant chondral expansion. Skeletal muscle of mdx mice of similar age to the ones used in this study have been shown to contain extensive fibrosis and degenerative myocytes with diminished muscle force production”<-maybe being able to generate enough force is a similar limiting factor for our purposes and if we could generate more muscular force we could induce longitudinal bone growth.

“a reduction in joint forces resulting from weaker muscle force production and slower running speeds may explain the diminished chondrogenic response to voluntary running exercise in the proximal femurs of mdx mice.”

The response of bone, articular cartilage and tendon to exercise in the horse.

” For bone, alterations in bone mineral content, mineral density and the morphology of the mineralized tissue are the most common end-points. Apparent bone density increases slightly after athletic training in the cortex, but substantially in the major load paths of the epiphyses and cuboidal bones, despite the lower material density of the new bone, which is deposited subperiosteally and on internal surfaces without prior osteoclastic resorption. With training of greater intensity, adaptive change is supervened by patho-anatomical change in the form of microdamage and frank lesions. In tendon, collagen fibril diameter distribution changes significantly during growth, but not after early training. The exact amount and type of protracted training that does cause reduction in mass average diameter (an early sign of progressive microdamage) have not been defined. Training is associated with an increase in the cross-sectional area of some tendons, possibly owing to slightly greater water content of non-collagenous or newly synthesized matrix. Early training may be associated with greater thickness of hyaline but not calcified articular cartilage, at least in some sites. The age at which adaptation of cartilage to biomechanical influences can occur may thus extend beyond very early life. However, cartilage appears to be the most susceptible of the three tissues to pathological alteration. The effect of training exercise on the anatomical or patho-anatomical features of connective tissue structures is affected by the timing, type and amount of natural or imposed exercise during growth and development which precedes the training.”

“Equine joint cartilage has similar appearance and properties to human articular cartilage”

“At both [phalanx] sites, water, DNA and GAG decreased during maturation whereas collagen content, hydroxylysine content and HP cross-links increased. Postnatal adaptation, resulting in biochemical and therefore biomechanical heterogeneity, which is important for future tissue strength and resistance to injury, occurs early and rapidly, possibly because collagen turnover is extremely low at older ages”

“Exercise influenced calcium content and levels of HP and lysylpyridinoline cross-links at the intermittently loaded site but not at the constantly loaded site of the proximal phalangeal bone, levels of lysyl-hydroxylation and lysylpyridinoline cross-linking being lower at the former than the latter site”

“The epiphyseal bone alters dramatically with exercise and is only arbitrarily separable from the subchondral bone.”  the subchondral bone is the bone that is at the end of the articular cartilage and the beginning of epiphyseal bone.