Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892366

ABSTRACT

In order to overcome the resistance to radiotherapy in human chondrosarcoma cells, the prevention from efficient DNA repair with a combined treatment with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor AZD7648 was explored for carbon ion (C-ion) as well as reference photon (X-ray) irradiation (IR) using gene expression analysis, flow cytometry, protein phosphorylation, and telomere length shortening. Proliferation markers and cell cycle distribution changed significantly after combined treatment, revealing a prominent G2/M arrest. The expression of the G2/M checkpoint genes cyclin B, CDK1, and WEE1 was significantly reduced by IR alone and the combined treatment. While IR alone showed no effects, additional AZD7648 treatment resulted in a dose-dependent reduction in AKT phosphorylation and an increase in Chk2 phosphorylation. Twenty-four hours after IR, the key genes of DNA repair mechanisms were reduced by the combined treatment, which led to impaired DNA repair and increased radiosensitivity. A time-dependent shortening of telomere length was observed in both cell lines after combined treatment with AZD7648 and 8 Gy X-ray/C-ion IR. Our data suggest that the inhibition of DNA-PKcs may increase sensitivity to X-rays and C-ion IR by impairing its functional role in DNA repair mechanisms and telomere end protection.


Subject(s)
Chondrosarcoma , DNA-Activated Protein Kinase , Heavy Ion Radiotherapy , Telomere , Humans , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Cell Line, Tumor , Chondrosarcoma/metabolism , Chondrosarcoma/genetics , Chondrosarcoma/radiotherapy , Chondrosarcoma/drug therapy , Telomere/drug effects , Telomere/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , DNA Repair/drug effects , Radiation Tolerance/drug effects , Pyrazoles/pharmacology , Cell Proliferation/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects
2.
Int J Mol Sci ; 24(21)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37958891

ABSTRACT

Myxofibrosarcoma (MFS) is a subtype of soft tissue sarcoma of connective tissue, which is characterized by large intra-tumor heterogeneity. Therapy includes surgical resection. Additional chemotherapy is of limited effect. In this study, we demonstrated the potent anticancer activity of shikonin derivatives in our MFS cellular model of tumor heterogeneity for developing a new therapeutic approach. The impact of shikonin and ß,ß-dimethylacrylshikonin (DMAS) on viability, apoptotic induction, MAPK phosphorylation, and DNA damage response were analyzed by means of two human MFS cell lines, MUG-Myx2a and MUG-Myx2b, derived from a singular tumor tissue specimen. MFS cells showed a dose-dependent inhibition of cell viability and a significant induction of apoptosis. Treatment with shikonin derivatives caused an inhibition of pSTAT3 and an increase in pAKT, pERK, pJNK, and pp38. DMAS and shikonin inhibited the activation of the two master upstream regulators of the DNA damage response, ATR and ATM. MUG-Myx2b, which contains an additional PTEN mutation, was more sensitive in some targets. These data demonstrate the significant antitumorigenic effect of shikonin derivatives in MFS and highlight the importance of intra-tumor heterogeneity in treatment planning.


Subject(s)
Fibrosarcoma , Naphthoquinones , Humans , Adult , Signal Transduction , Cell Line, Tumor , Naphthoquinones/pharmacology , Apoptosis
3.
Cells ; 12(18)2023 09 18.
Article in English | MEDLINE | ID: mdl-37759523

ABSTRACT

Particle therapy (PT) that utilizes protons and carbon ions offers a promising way to reduce the side effects of radiation oncology, especially in pediatric patients. To investigate the influence of PT on growing bone, we exposed an organotypic rat ex vivo femur culture model to PT. After irradiation, histological staining, immunohistochemical staining, and gene expression analysis were conducted following 1 or 14 days of in vitro culture (DIV). Our data indicated a significant loss of proliferating chondrocytes at 1 DIV, which was followed by regeneration attempts through chondrocytic cluster formation at 14 DIV. Accelerated levels of mineralization were observed, which correlated with increased proteoglycan production and secretion into the pericellular matrix. Col2α1 expression, which increased during the cultivation period, was significantly inhibited by PT. Additionally, the decrease in ColX expression over time was more pronounced compared to the non-IR control. The chondrogenic markers BMP2, RUNX2, OPG, and the osteogenic marker ALPL, showed a significant reduction in the increase in expression after 14 DIV due to PT treatment. It was noted that carbon ions had a stronger influence than protons. Our bone model demonstrated the occurrence of pathological and regenerative processes induced by PT, thus building on the current understanding of the biological mechanisms of bone.


Subject(s)
Osteogenesis , Protons , Animals , Rats , Humans , Child , Microphysiological Systems , Femur , Carbon
4.
Int J Mol Sci ; 24(3)2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36768638

ABSTRACT

To overcome the resistance to radiotherapy in chondrosarcomas, the prevention of efficient DNA repair with an additional treatment was explored for particle beams as well as reference X-ray irradiation. The combined treatment with DNA repair inhibitors-with a focus on ATRi VE-821-and proton or carbon ions irradiation was investigated regarding cell viability, proliferation, cell cycle distribution, MAPK phosphorylation, and the expression of key DNA repair genes in two human chondrosarcoma cell lines. Pre-treatment with the PARPis Olaparib or Veliparib, the ATMi Ku-55933, and the ATRi VE-821 resulted in a dose-dependent reduction in viability, whereas VE-821 has the most efficient response. Quantification of γH2AX phosphorylation and protein expression of the DNA repair pathways showed a reduced regenerative capacity after irradiation. Furthermore, combined treatment with VE-821 and particle irradiation increased MAPK phosphorylation and the expression of apoptosis markers. At the gene expression and at the protein expression/phosphorylation level, we were able to demonstrate the preservation of DNA damage after combined treatment. The present data showed that the combined treatment with ATMi VE-821 increases the radiosensitivity of human chondrosarcoma cells in vitro and significantly suppresses efficient DNA repair mechanisms, thus improving the efficiency of radiotherapy.


Subject(s)
DNA Repair , Radiation Tolerance , Humans , Radiation Tolerance/genetics , Pyrazines/pharmacology , Sulfones/pharmacology , Protein Kinase Inhibitors/pharmacology , DNA Damage , Cell Line, Tumor , Ataxia Telangiectasia Mutated Proteins
5.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232764

ABSTRACT

Chondrosarcomas are particularly difficult to treat due to their resistance to chemotherapy and radiotherapy. However, particle therapy can enhance local control and patient survival rates. To improve our understanding of the basic cellular radiation response, as a function of dose and linear energy transfer (LET), we developed a novel water phantom-based setup for cell culture experiments and characterized it dosimetrically. In a direct comparison, human chondrosarcoma cell lines were analyzed with regard to their viability, cell proliferation, cell cycle, and DNA repair behavior after irradiation with X-ray, proton, and carbon ions. Our results clearly showed that cell viability and proliferation were inhibited according to the increasing ionization density, i.e., LET, of the irradiation modes. Furthermore, a prominent G2/M arrest was shown. Gene expression profiling proved the upregulation of the senescence genes CDKN1A (p21), CDKN2A (p16NK4a), BMI1, and FOXO4 after particle irradiation. Both proton or C-ion irradiation caused a positive regulation of the repair genes ATM, NBN, ATXR, and XPC, and a highly significant increase in XRCC1/2/3, ERCC1, XPC, and PCNA expression, with C-ions appearing to activate DNA repair mechanisms more effectively. The link between the physical data and the cellular responses is an important contribution to the improvement of the treatment system.


Subject(s)
Chondrosarcoma , Protons , Carbon , Chondrosarcoma/genetics , Chondrosarcoma/radiotherapy , Humans , Physics , Proliferating Cell Nuclear Antigen , Water , X-ray Repair Cross Complementing Protein 1
6.
BMC Cancer ; 22(1): 758, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35820864

ABSTRACT

BACKGROUND: Although chondrosarcoma is the second most common primary malignant bone tumor, treatment options are limited due to its extensive resistance to a chemo- and radiation therapy. Since shikonin has shown potent anticancer activity in various types of cancer cells, it represents a promising compound for the development of a new therapeutic approach. METHODS: The dose-relationships of shikonin and its derivatives acetylshikonin and cyclopropylshikonin on two human chondrosarcoma cell lines were measured using the CellTiter-Glo®. The changes in the cell cycle were presented by flow cytometry. Protein phosphorylation and expression apoptotic markers, MAPKs and their downstream targets were analyzed using western blotting and gene expression were evaluated using RT-qPCR. RESULTS: Chondrosarcoma cells showed a dose-dependent inhibition of cell viability after treatment with shikonin and its derivatives, with the strongest effect for shikonin and IC50 values of 1.3 ± 0.2 µM. Flow cytometric measurements revealed a G2/M arrest of the cells after treatment. Protein and gene expression analysis demonstrated a dose-dependent downregulation of survivin and XIAP, and an upregulation of Noxa, γH2AX, cleaved caspase-8, -9, -3, and -PARP. Furthermore, the expression of various death receptors was modulated. As MAPK signaling pathways play a key role in tumor biology, their phosphorylation pattern and their corresponding downstream gene regulation were analyzed. Treatment with shikonin derivatives caused an inhibition of pSTAT3 and an increase of pAKT and the MAPKs pERK, pJNK, and pp38 in a dose-dependent manner. CONCLUSIONS: These data demonstrated the significant anti-tumorigenic effect of shikonin derivatives in chondrosarcoma and encourage further research.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Mitogen-Activated Protein Kinases , Naphthoquinones , Receptors, Death Domain , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Humans , Naphthoquinones/pharmacology , Receptors, Death Domain/metabolism
7.
Int J Mol Sci ; 23(6)2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35328817

ABSTRACT

Osteoarthritis (OA) is the most common joint disorder and is characterized by the degeneration of articular cartilage. To develop new therapeutic approaches, we investigated the effect of shikonin derivatives on inflammation, MMP expression, and the regulation of MAPK signaling in human healthy (HC) and OA chondrocytes (pCH-OA). Viability was analyzed using the CellTiter-Glo® Assay. Inflammatory processes were investigated using a proteome profiler™ assay. Furthermore, we analyzed the effects of the shikonin derivatives by protein expression analysis of the phosphorylation pattern and the corresponding downstream gene regulation using RT-qPCR. Both HC and pCH-OA showed a dose-dependent decrease in viability after treatment. The strongest effects were found for shikonin with IC50 values of 1.2 ± 0.1 µM. Shikonin counteracts the inflammatory response by massively reducing the expression of the pro-inflammatory mediators. The phosphorylation level of ERK changed slightly. pJNK and pp38 showed a significant increase, and the downstream targets c/EBPs and MEF2c may play a role in the cartilage homeostasis. STAT3 phosphorylation decreased significantly and has a chondroprotective function through the regulation of cyclin D1 and Sox9. Our results demonstrate for the first time that shikonin derivatives have extensive effects on the inflammatory processes, MAPKs, and IL6/STAT3 downstream regulation in healthy and OA chondrocytes.


Subject(s)
Cartilage, Articular , Naphthoquinones , Osteoarthritis , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Naphthoquinones/metabolism , Naphthoquinones/pharmacology , Osteoarthritis/metabolism
8.
Front Cell Infect Microbiol ; 12: 837124, 2022.
Article in English | MEDLINE | ID: mdl-35300379

ABSTRACT

Periprosthetic infections are an eminent factor in patient care and also having significant economic implications. The number of biofilm-infection related replacement surgeries is increasing and will continue to do so in the following decades. To reduce both the health burden of the patients and the costs to the healthcare sector, new solutions for implant materials resistant to such infections are necessary. This study researches different surface modifications of cobalt-chromium-molybdenum (CoCrMo) based implant materials and their influence on the development of biofilms. Three smooth surfaces (CoCrMo, CoCrMo TiN, and CoCrMo polished) and three rough surfaces (CoCrMo porous coated, CoCrMo cpTi, and CoCrMo TCP) are compared. The most common infectious agents in periprosthetic infections are Staphylococcus aureus and Coagulase-negative staphylococci (e.g., Staphylococcus epidermidis), therefore strains of these two species have been chosen as model organisms. Biofilms were grown on material disks for 48 h and cell number, polysaccharide content, and protein contend of the biofilms were measured. Additionally, regulation of genes involved in early biofilm development (S. aureus icaA, icaC, fnbA, fnbB, clfB, atl; S. epidermidis atlE, aap) was detected using RT-q-PCR. All results were compared to the base alloy without modifications. The results show a correlation between the surface roughness and the protein and polysaccharide content of biofilm structures and also the gene expression of the biofilms grown on the different surface modifications. This is supported by the significantly different protein and polysaccharide contents of the biofilms associated with rough and smooth surface types. Additionally, early phase biofilm genes (particularly icaA, icaC, and aap) are statistically significantly downregulated compared to the control at 48 h on rough surfaces. CoCrMo TiN and polished CoCrMo were the two smooth surface modifications which performed best on the basis of low biofilm content.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Biofilms , Chromium/pharmacology , Cobalt/pharmacology , Humans , Molybdenum/pharmacology , Staphylococcus aureus/genetics , Staphylococcus epidermidis
9.
Sci Rep ; 11(1): 24116, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916568

ABSTRACT

Although particle therapy with protons has proven to be beneficial in the treatment of chondrosarcoma compared to photon-based (X-ray) radiation therapy, the cellular and molecular mechanisms have not yet been sufficiently investigated. Cell viability and colony forming ability were analyzed after X-ray and proton irradiation (IR). Cell cycle was analyzed using flow cytometry and corresponding regulator genes and key players of the DNA repair mechanisms were measured using next generation sequencing, protein expression and immunofluorescence staining. Changes in metabolic phenotypes were determined with nuclear magnetic resonance spectroscopy. Both X-ray and proton IR resulted in reduced cell survival and a G2/M phase arrest of the cell cycle. Especially 1 h after IR, a significant dose-dependent increase of phosphorylated γH2AX foci was observed. This was accompanied with a reprogramming in cellular metabolism. Interestingly, within 24 h the majority of clearly visible DNA damages were repaired and the metabolic phenotype restored. Involved DNA repair mechanisms are, besides the homology directed repair (HDR) and the non-homologous end-joining (NHEJ), especially the mismatch mediated repair (MMR) pathway with the key players EXO1, MSH3, and PCNA. Chondrosarcoma cells regenerates the majority of DNA damages within 24 h. These molecular mechanisms represent an important basis for an improved therapy.


Subject(s)
Cell Cycle/radiation effects , Chondrosarcoma/genetics , Chondrosarcoma/radiotherapy , DNA Repair/radiation effects , Photons/therapeutic use , Proton Therapy , Cell Survival/radiation effects , Chondrosarcoma/pathology , DNA Breaks, Double-Stranded/radiation effects , Dose-Response Relationship, Radiation , Humans , Radiotherapy Dosage , Time Factors , Tumor Cells, Cultured
10.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073090

ABSTRACT

Nuclear magnetic resonance therapy (NMRT) is discussed as a participant in repair processes regarding cartilage and as an influence in pain signaling. To substantiate the application of NMRT, the underlying mechanisms at the cellular level were studied. In this study microRNA (miR) was extracted from human primary healthy and osteoarthritis (OA) chondrocytes after NMR treatment and was sequenced by the Ion PI Hi-Q™ Sequencing 200 system. In addition, T/C-28a2 chondrocytes grown under hypoxic conditions were studied for IL-1ß induced changes in expression on RNA and protein level. HDAC activity an NAD(+)/NADH was measured by luminescence detection. In OA chondrocytes miR-106a, miR-27a, miR-34b, miR-365a and miR-424 were downregulated. This downregulation was reversed by NMRT. miR-365a-5p is known to directly target HDAC and NF-ĸB, and a decrease in HDAC activity by NMRT was detected. NAD+/NADH was reduced by NMR treatment in OA chondrocytes. Under hypoxic conditions NMRT changed the expression profile of HIF1, HIF2, IGF2, MMP3, MMP13, and RUNX1. We conclude that NMRT changes the miR profile and modulates the HDAC and the NAD(+)/NADH signaling in human chondrocytes. These findings underline once more that NMRT counteracts IL-1ß induced changes by reducing catabolic effects, thereby decreasing inflammatory mechanisms under OA by changing NF-ĸB signaling.


Subject(s)
Chondrocytes , Magnetic Resonance Spectroscopy/methods , MicroRNAs/metabolism , Osteoarthritis , Cell Line , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Osteoarthritis/metabolism , Osteoarthritis/therapy , Primary Cell Culture
11.
J Funct Biomater ; 12(2)2021 May 18.
Article in English | MEDLINE | ID: mdl-34069837

ABSTRACT

One of the most serious complications following joint replacement surgeries are periprosthetic infections (PIs) arising from the adhesion of bacteria to the artificial joint. Various types of titanium-aluminum-vanadium (TiAl6V4) alloy surface modifications (coatings with silver (Ag), titanium nitride (TiN), pure titanium (cpTi), combinations of cpTi and hydroxyapatite (HA), combinations of cpTi and tricalcium phosphate (TCP), and a rough-blasted surface of TiAl6V4) have been investigated to assess their effects on biofilm development. Biofilms were grown, collected, and analyzed after 48 h to measure their protein and glucose content and the cell viability. Biofilm-associated genes were also monitored after 48 h of development. There was a distinct difference in the development of staphylococcal biofilms on the surfaces of the different types of alloy. According to the findings of this study, the base alloy TiAl6V4 and the TiN-coated surface are the most promising materials for biofilm reduction. Rough surfaces are most favorable when it comes to bacterial infections because they allow an easy attachment of pathogenic organisms. Of all rough surfaces tested, rough-blasted TiAl6V4 was the most favorable as an implantation material; all the other rough surfaces showed more distinct signs of inducing the development of biofilms which displayed higher protein and polysaccharide contents. These results are supported by RT-qPCR measurements of biofilm associated genes for Staphylococcus aureus (icaA, icaC, fnbA, fnbB, clfB, atl) and Staphylococcus epidermidis (atle, aap).

12.
Materials (Basel) ; 14(6)2021 Mar 23.
Article in English | MEDLINE | ID: mdl-33807039

ABSTRACT

Osteogenic cells are strongly influenced in their behaviour by the surface properties of orthopaedic implant materials. Mesenchymal stem and progenitor cells (MSPCs) migrate to the bone-implant interface, adhere to the material surface, proliferate and subsequently differentiate into osteoblasts, which are responsible for the formation of the bone matrix. Five surface topographies on titanium aluminium vanadium (TiAl6V4) were engineered to investigate biocompatibility and adhesion potential of human osteoblasts and the changes in osteogenic differentiation of MSPCs. Elemental analysis of TiAl6V4 discs coated with titanium nitride (TiN), silver (Ag), roughened surface, and pure titanium (cpTi) surface was analysed using energy-dispersive X-ray spectroscopy and scanning electron microscopy. In vitro cell viability, cytotoxicity, adhesion behaviour, and osteogenic differentiation potential were measured via CellTiter-Glo, CytoTox, ELISA, Luminex® technology, and RT-PCR respectively. The Ag coating reduced the growth of osteoblasts, whereas the viability of MSPCs increased significantly. The roughened and the cpTi surface improved the viability of all cell types. The additive coatings of the TiAl6V4 alloy improved the adhesion of osteoblasts and MSPCs. With regard to the osteogenic differentiation potential, an enhanced effect has been demonstrated, especially in the case of roughened and cpTi coatings.

13.
Oncol Lett ; 21(6): 428, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33868466

ABSTRACT

Chondrosarcomas represent a heterogeneous group of primary bone cancers that are characterized by hyaline cartilaginous neoplastic tissue and are predominantly resistant to radiation and chemotherapy. However, adjuvant radiotherapy is often recommended in inoperable cases or after incomplete resections. To improve the efficiency of treatment, the present study tested a combination therapy with ionizing radiation (IR) and the proteasome inhibitor bortezomib. Using a three-dimensional (3D) spheroid model, 0-20 Gy of IR was applied to chondrosarcoma cells and healthy human chondrocytes. Following combined treatment with IR and bortezomib, the cell cycle distribution, apoptotic induction, the survivin pathway, autophagy and DNA damage were evaluated. Both cell types exhibited a slight decrease in viability following increasing doses of IR; the chondrosarcoma cells demonstrated a significant dose-dependent increase in the expression levels of the DNA damage marker histone H2AX phosphorylation at serine 139 (γH2AX). The combination treatment with bortezomib significantly decreased the cell viability after 48 h compared with that in irradiated cells. High-dose IR induced a G2/M phase arrest, which was accompanied by a decrease in the number of cells at the G1 and S phase. Co-treatment with bortezomib changed the distribution of the cell cycle phases. The mRNA expression levels of the proapoptotic genes Bcl-2-associated X protein (Bax) and Bak were significantly increased by bortezomib treatment and combination therapy with IR. In addition, the combination therapy resulted in a synergistic decrease of the expression levels of survivin and its corresponding downstream pathway molecules, including heat shock protein 90, X-linked inhibitor of apoptosis protein, smad 2 and smad 3. Comparative analyses of γH2AX at 1 and 24 h post-IR revealed efficient DNA repair in human chondrosarcoma cells. Therefore, additional bortezomib treatment may only temporarily improve the radiation sensitivity of chondrosarcoma cells. However, the inhibition of the survivin pathway by the combined treatment with IR and bortezomib, observed in the present study, revealed a novel aspect in the tumor biology of chondrosarcoma 3D spheroid cultures and may represent a potential target for therapy.

14.
Materials (Basel) ; 13(19)2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32992906

ABSTRACT

Surface roughness on orthopedic implant materials has been shown to be highly influential on the behavior of osteogenic cells. Mesenchymal stem and progenitor cells (MSPCs) migrate to the interface, adhere, proliferate, and differentiate into osteoblasts, which subsequently form bone matrix. Modifications of the implant surfaces should accelerate this process and improve biocompatibility. In this study, five surface topographies on cobalt chromium molybdenum (CoCrMo) were engineered to examine the influence on MSPCs. Scanning electron microscopy revealed significant differences in the morphology of untreated CoCrMo discs in comparison with CoCrMo with a titanium nitride (TiN) coating, polished and porous coated CoCrMo surfaces, and CoCrMo with a pure titanium (cpTi) coating. Elemental analysis was performed using energy-dispersive X-ray spectroscopy (EDX). Human primary MSPCs were expanded from tissue samples of spongiosa bone and characterized according to the criteria of the International Society for Cellular Therapy. The characteristic phenotype of MSPC was confirmed by flow cytometry and multilineage differentiation. Alcaline phosphatase and osteopontin expression increased significantly in all groups about 5-fold and 10-fold, respectively, in comparison to the undifferentiated controls. The porous coated surface showed a reduced expression of osteogenic markers. Due to the osteogenic differentiation, the expression of integrin α5ß1, which is particularly important for cell-material contact, increased 4-7-fold. In the dynamic process of bone biology, MSPCs cultured and differentiated on cpTi, showed significant upregulation of IL6 and leptin.

SELECTION OF CITATIONS
SEARCH DETAIL
...