Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nucleic Acids Res ; 51(8): 3590-3617, 2023 05 08.
Article in English | MEDLINE | ID: mdl-36987858

ABSTRACT

Chondrogenesis is a multistep process, in which cartilage progenitor cells generate a tissue with distinct structural and functional properties. Although several approaches to cartilage regeneration rely on the differentiation of implanted progenitor cells, the temporal transcriptomic landscape of in vitro chondrogenesis in different models has not been reported. Using RNA sequencing, we examined differences in gene expression patterns during cartilage formation in micromass cultures of embryonic limb bud-derived progenitors. Principal component and trajectory analyses revealed a progressively different and distinct transcriptome during chondrogenesis. Differentially expressed genes (DEGs), based on pairwise comparisons of samples from consecutive days were classified into clusters and analysed. We confirmed the involvement of the top DEGs in chondrogenic differentiation using pathway analysis and identified several chondrogenesis-associated transcription factors and collagen subtypes that were not previously linked to cartilage formation. Transient gene silencing of ATOH8 or EBF1 on day 0 attenuated chondrogenesis by deregulating the expression of key osteochondrogenic marker genes in micromass cultures. These results provide detailed insight into the molecular mechanism of chondrogenesis in primary micromass cultures and present a comprehensive dataset of the temporal transcriptomic landscape of chondrogenesis, which may serve as a platform for new molecular approaches in cartilage tissue engineering.


Subject(s)
Chondrogenesis , Transcriptome , Chondrogenesis/genetics , Cartilage/metabolism , Cell Differentiation/genetics , Stem Cells/metabolism , Cells, Cultured , Chondrocytes/metabolism
2.
J Pineal Res ; 73(4): e12827, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36030553

ABSTRACT

The biomechanical environment plays a key role in regulating cartilage formation, but the current understanding of mechanotransduction pathways in chondrogenic cells is incomplete. Among the combination of external factors that control chondrogenesis are temporal cues that are governed by the cell-autonomous circadian clock. However, mechanical stimulation has not yet directly been proven to modulate chondrogenesis via entraining the circadian clock in chondroprogenitor cells. The purpose of this study was to establish whether mechanical stimuli entrain the core clock in chondrogenic cells, and whether augmented chondrogenesis caused by mechanical loading was at least partially mediated by the synchronised, rhythmic expression of the core circadian clock genes, chondrogenic transcription factors, and cartilage matrix constituents at both transcript and protein levels. We report here, for the first time, that cyclic uniaxial mechanical load applied for 1 h for a period of 6 days entrains the molecular clockwork in chondroprogenitor cells during chondrogenesis in limb bud-derived micromass cultures. In addition to the several core clock genes and proteins, the chondrogenic markers SOX9 and ACAN also followed a robust sinusoidal rhythmic expression pattern. These rhythmic conditions significantly enhanced cartilage matrix production and upregulated marker gene expression. The observed chondrogenesis-promoting effect of the mechanical environment was at least partially attributable to its entraining effect on the molecular clockwork, as co-application of the small molecule clock modulator longdaysin attenuated the stimulatory effects of mechanical load. This study suggests that an optimal biomechanical environment enhances tissue homoeostasis and histogenesis during chondrogenesis at least partially through entraining the molecular clockwork.


Subject(s)
Circadian Clocks , Melatonin , Chondrogenesis , Mechanotransduction, Cellular , Melatonin/pharmacology , Transcription Factors/metabolism , Chondrocytes/metabolism , Cells, Cultured , Cell Differentiation
3.
Differentiation ; 107: 24-34, 2019.
Article in English | MEDLINE | ID: mdl-31152959

ABSTRACT

Bone graft substitutes and bone void fillers are predominantly used to treat bone defects and bone fusion in orthopaedic surgery. Some aragonite-based scaffolds of coralline exoskeleton origin exhibit osteoconductive properties and are described as useful bone repair scaffolds. The purpose of this study was to evaluate the in vitro osteogenic potential of the bone phase of a novel aragonite-based bi-phasic osteochondral scaffold (Agili-C™, CartiHeal Ltd.) using adult human bone marrow-derived mesenchymal stem cells (MSCs). Analyses were performed at several time intervals: 3, 7, 14, 21, 28 and 42 days post-seeding. Osteogenic differentiation was assessed by morphological characterisation using light microscopy after Alizarin red and von Kossa staining, and scanning electron microscopy. The transcript levels of alkaline phosphatase (ALP), runt-related transcription factor 2 (RUNX2), bone gamma-carboxyglutamate (BGLAP), osteonectin (SPARC) and osteopontin (SPP1) were determined by quantitative PCR. Proliferation was assessed by a thymidine incorporation assay and proliferating cell nuclear antigen (PCNA) immunocytochemistry. Our results demonstrate that the bone phase of the bi-phasic aragonite-based scaffold supports osteogenic differentiation and enhanced proliferation of bone marrow-derived MSCs at both the molecular and histological levels. The scaffold was colonized by differentiating MSCs, suggesting its suitability for incorporation into bone voids to accelerate bone healing, remodelling and regeneration. The mechanism of osteogenic differentiation involves scaffold surface modification with de novo production of calcium phosphate deposits, as revealed by energy dispersive spectroscopy (EDS) analyses. This novel coral-based scaffold may promote the rapid formation of high quality bone during the repair of osteochondral lesions.


Subject(s)
Calcium Carbonate , Mesenchymal Stem Cells/cytology , Osteogenesis , Tissue Scaffolds , Bone Substitutes/chemistry , Calcium Carbonate/chemistry , Calcium Phosphates/metabolism , Cells, Cultured , Humans , Tissue Engineering
4.
Int J Mol Sci ; 21(16)2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32785075

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with protective functions in the central nervous system and various peripheral organs. PACAP has the highest expression level in the testes, among the peripheral organs, and has a positive regulative role in spermatogenesis and in sperm motility. In the present study, we explored testicular degenerative alterations in a mouse model of Alzheimer's disease (AD) (B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J) and demonstrated changes in PACAP-regulated signaling pathways. In addition, the effects of increased physical activity of AD (trained AD (TAD)) mice on testis were also followed. Reduced cell number and decreased thickness of basement membrane were detected in AD samples. These changes were compensated by physical activity. Expression of PACAP receptors and canonical signaling elements such as PKA, P-PKA, PP2A significantly decreased in AD mice, and altered Sox transcription factor expression was also detected. Via this signaling mechanism, physical activity compensated the negative effects of AD on the expression of type IV collagen. Our findings suggest that the testes of AD mice can be a good model of testis degeneration. Moreover, it can be an appropriate organ to follow the effects of various interventions such as physical activity on tissue regeneration and signaling alterations.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Signal Transduction/genetics , Testicular Diseases/metabolism , Testis/metabolism , Animals , Cell Count , Collagen Type IV/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , Physical Conditioning, Animal , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , SOX9 Transcription Factor/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis/pathology
5.
Cell Commun Signal ; 17(1): 166, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31842918

ABSTRACT

BACKGROUND: In vitro chondrogenesis depends on the concerted action of numerous signalling pathways, many of which are sensitive to the changes of intracellular Ca2+ concentration. N-methyl-D-aspartate (NMDA) glutamate receptor is a cation channel with high permeability for Ca2+. Whilst there is now accumulating evidence for the expression and function of NMDA receptors in non-neural tissues including mature cartilage and bone, the contribution of glutamate signalling to the regulation of chondrogenesis is yet to be elucidated. METHODS: We studied the role of glutamatergic signalling during the course of in vitro chondrogenesis in high density chondrifying cell cultures using single cell fluorescent calcium imaging, patch clamp, transient gene silencing, and western blotting. RESULTS: Here we show that key components of the glutamatergic signalling pathways are functional during in vitro chondrogenesis in a primary chicken chondrogenic model system. We also present the full glutamate receptor subunit mRNA and protein expression profile of these cultures. This is the first study to report that NMDA-mediated signalling may act as a key factor in embryonic limb bud-derived chondrogenic cultures as it evokes intracellular Ca2+ transients, which are abolished by the GluN2B subunit-specific inhibitor ifenprodil. The function of NMDARs is essential for chondrogenesis as their functional knock-down using either ifenprodil or GRIN1 siRNA temporarily blocks the differentiation of chondroprogenitor cells. Cartilage formation was fully restored with the re-expression of the GluN1 protein. CONCLUSIONS: We propose a key role for NMDARs during the transition of chondroprogenitor cells to cartilage matrix-producing chondroblasts.


Subject(s)
Chondrogenesis/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium/analysis , Calcium/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chickens , Chondrogenesis/drug effects , Glutamic Acid/analysis , N-Methylaspartate/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Signal Transduction/drug effects
6.
Int J Mol Sci ; 20(1)2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30621194

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is an endogenous neuropeptide also secreted by non-neural cells, including chondrocytes. PACAP signaling is involved in the regulation of chondrogenesis, but little is known about its connection to matrix turnover during cartilage formation and under cellular stress in developing cartilage. We found that the expression and activity of hyaluronidases (Hyals), matrix metalloproteinases (MMP), and aggrecanase were permanent during the course of chondrogenesis in primary chicken micromass cell cultures, although protein levels changed daily, along with moderate and relatively constant enzymatic activity. Next, we investigated whether PACAP influences matrix destructing enzyme activity during oxidative and mechanical stress in chondrogenic cells. Exogenous PACAP lowered Hyals and aggrecanase expression and activity during cellular stress. Expression and activation of the majority of cartilage matrix specific MMPs such as MMP1, MMP7, MMP8, and MMP13, were also decreased by PACAP addition upon oxidative and mechanical stress, while the activity of MMP9 seemed not to be influenced by the neuropeptide. These results suggest that application of PACAP can help to preserve the integrity of the newly synthetized cartilage matrix via signaling mechanisms, which ultimately inhibit the activity of matrix destroying enzymes under cellular stress. It implies the prospect that application of PACAP can ameliorate articular cartilage destruction in joint diseases.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Chondrocytes/drug effects , Oxidative Stress , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Stress, Mechanical , Animals , Apoptosis Regulatory Proteins/administration & dosage , Cartilage/drug effects , Cartilage/metabolism , Cell Culture Techniques , Chick Embryo , Chondrocytes/metabolism , Endopeptidases/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Hyaluronoglucosaminidase/metabolism , Hydrogen Peroxide/pharmacology , Matrix Metalloproteinases/metabolism , Oxidants/pharmacology
7.
J Neuroinflammation ; 15(1): 335, 2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30509328

ABSTRACT

OBJECTIVE: The regulatory role of capsaicin-sensitive peptidergic sensory nerves has been shown in acute inflammation, but little is known about their involvement in T/B-cell driven autoimmune arthritis. This study integratively characterized the function of these nerve endings in the proteoglycan-induced chronic arthritis (PGIA), a translational model of rheumatoid arthritis. METHODS: Peptidergic afferents were defunctionalized by resiniferatoxin (RTX) pretreatment in BALB/c mice, PGIA was induced by repeated antigen challenges. Hind paw volume, arthritis severity, grasping ability and the mechanonociceptive threshold were monitored during the 17-week experiment. Myeloperoxidase activity, vascular leakage and bone turnover were evaluated by in vivo optical imaging. Bone morphology was assessed using micro-CT, the intertarsal small joints were processed for histopathological analysis. RESULTS: Following desensitization of the capsaicin-sensitive afferents, ankle edema, arthritis severity and mechanical hyperalgesia were markedly diminished. Myeloperoxidase activity was lower in the early, but increased in the late phase, whilst plasma leakage and bone turnover were not altered. Desensitized mice displayed similar bone spurs and erosions, but increased trabecular thickness of the tibia and bony ankylosis of the spine. Intertarsal cartilage thickness was not altered in the model, but desensitization increased this parameter in both the non-arthritic and arthritic groups. CONCLUSION: This is the first integrative in vivo functional and morphological characterization of the PGIA mouse model, wherein peptidergic afferents have an important regulatory function. Their overall effect is proinflammatory by increasing acute inflammation, immune cell activity and pain. Meanwhile, their activation decreases spinal ankylosis, arthritis-induced altered trabecularity, and cartilage thickness in small joints.


Subject(s)
Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/pathology , Capsaicin/pharmacology , Proteoglycans/toxicity , Sensory System Agents/pharmacology , Sensory Thresholds/drug effects , Animals , Ankle/diagnostic imaging , Cartilage/pathology , Disease Models, Animal , Diterpenes/pharmacology , Female , Hindlimb/drug effects , Hindlimb/physiopathology , Mice , Mice, Inbred BALB C , Neurotoxins/pharmacology , Peptides/metabolism , Reactive Oxygen Species/metabolism , Severity of Illness Index , Spine/diagnostic imaging
8.
Int J Mol Sci ; 19(7)2018 06 30.
Article in English | MEDLINE | ID: mdl-29966365

ABSTRACT

Heterotetrameric N-methyl-d-aspartate type glutamate receptors (NMDAR) are cationic channels primarily permeable for Ca2+. NR1 and NR3 subunits bind glycine, while NR2 subunits bind glutamate for full activation. As NR1 may contain a nuclear localization signal (NLS) that is recognized by importin-α, our aim was to investigate if NMDARs are expressed in the nuclei of melanocytes and melanoma cells. A detailed NMDAR subunit expression pattern was examined by RT-PCRs (reverse transcription followed by polymerase chain reaction), fractionated western blots and immunocytochemistry in human epidermal melanocytes and in human melanoma cell lines A2058, HT199, HT168M1, MEL35/0 and WM35. All kind of NMDAR subunits are expressed as mRNAs in melanocytes, as well as in melanoma cells, while NR2B protein remained undetectable in any cell type. Western blots proved the exclusive presence of NR1 and NR3B in nuclear fractions and immunocytochemistry confirmed NR1-NR3B colocalization inside the nuclei of all melanoma cells. The same phenomenon was not observed in melanocytes. Moreover, protein database analysis revealed a putative NLS in NR3B subunit. Our results support that unusual, NR1-NR3B composed NMDAR complexes are present in the nuclei of melanoma cells. This may indicate a new malignancy-related histopathological feature of melanoma cells and raises the possibility of a glycine-driven, NMDA-related nuclear Ca2+-signalling in these cells.


Subject(s)
Melanoma/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Humans , Melanocytes/metabolism , Melanoma/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/genetics , Signal Transduction/physiology
9.
Int J Mol Sci ; 19(9)2018 Aug 27.
Article in English | MEDLINE | ID: mdl-30150589

ABSTRACT

: Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with diverse developmental roles, including differentiation of skeletal elements. It is a positive regulatory factor of chondrogenesis and osteogenic differentiation in vitro, but little is known about its in vivo role in bone formation. In our experiments, diaphyses of long bones from hind limbs of PACAP gene-deficient mice showed changes in thickness and increased staining intensity. Our main goal was to perform a detailed morphological and molecular biological analysis of femurs from PACAP knockout (KO) and wild type (WT) mice. Transverse diameter and anterior cortical bone thickness of KO femurs showed significant alterations with disturbed Ca2+ accumulation and collagen type I expression. Higher expression and activity of alkaline phosphatase were also observed, accompanied by increased fragility PACAP KO femurs. Increased expression of the elements of bone morphogenic protein (BMP) and hedgehog signalling was also observed, and are possibly responsible for the compensation mechanism accounting for the slight morphological changes. In summary, our results show that lack of PACAP influences molecular and biomechanical properties of bone matrix, activating various signalling cascade changes in a compensatory fashion. The increased fragility of PACAP KO femur further supports the role of endogenous PACAP in in vivo bone formation.


Subject(s)
Chondrogenesis/genetics , Osteogenesis/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Signal Transduction/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Calcium/metabolism , Cell Differentiation/genetics , Collagen Type I/genetics , Collagen Type I/metabolism , Femur/diagnostic imaging , Femur/metabolism , Gene Expression , Mice, Knockout , Pituitary Adenylate Cyclase-Activating Polypeptide/deficiency , X-Ray Microtomography
10.
Pflugers Arch ; 467(2): 429-42, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24841338

ABSTRACT

Osteoarthritis (OA) is the most common form of chronic musculoskeletal disorders. A migratory stem cell population termed chondrogenic progenitor cells (CPC) with in vitro chondrogenic potential was previously isolated from OA cartilage. Since intracellular Ca(2+) signalling is an important regulator of chondrogenesis, we aimed to provide a detailed understanding of the Ca(2+) homeostasis of CPCs. In this work, CPCs immortalised by lentiviral administration of the human telomerase reverse transcriptase (hTERT) and grown in monolayer cultures were studied. Expressions of all three IP3Rs were confirmed, but no RyR subtypes were detected. Ca(2+) oscillations observed in CPCs were predominantly dependent on Ca(2+) release and store replenishment via store-operated Ca(2+) entry; CPCs express both STIM1 and Orai1 proteins. Expressions of adenosine receptor mRNAs were verified, and adenosine elicited Ca(2+) transients. Various P2 receptor subtypes were identified; P2Y1 can bind ADP; P2Y4 is targeted by UTP; and ATP may evoke Ca(2+) transients via detected P2X subtypes, as well as P2Y1 and P2Y2. Enzymatic breakdown of extracellular nucleotides by apyrase completely abrogated Ca(2+) oscillations, suggesting that an autocrine/paracrine purinergic mechanism may drive Ca(2+) oscillations in these cells. As CPCs possess a broad spectrum of functional molecular elements of Ca(2+) signalling, Ca(2+)-dependent regulatory mechanisms can be supposed to influence their differentiation potential.


Subject(s)
Adult Stem Cells/metabolism , Calcium Signaling , Cartilage/metabolism , Receptors, Purinergic/metabolism , Aged , Calcium Channels/genetics , Calcium Channels/metabolism , Cartilage/cytology , Cells, Cultured , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , Receptors, Purinergic/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Stromal Interaction Molecule 1
11.
Curr Rheumatol Rep ; 17(7): 43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25980668

ABSTRACT

Chondrocytes, the single cell type in adult articular cartilage, have conventionally been considered to be non-excitable cells. However, recent evidence suggests that their resting membrane potential (RMP) is less negative than that of excitable cells, and they are fully equipped with channels that control ion, water and osmolyte movement across the chondrocyte membrane. Amongst calcium-specific ion channels, members of the voltage-dependent calcium channel (VDCC) family are expressed in chondrocytes where they are functionally active. L-type VDCC inhibitors such as nifedipine and verapamil have contributed to our understanding of the roles of these ion channels in chondrogenesis, chondrocyte signalling and mechanotransduction. In this narrative review, we discuss published data indicating that VDCC function is vital for chondrocyte health, especially in regulating proliferation and maturation. We also highlight the fact that activation of VDCC function appears to accompany various inflammatory aspects of osteoarthritis (OA) and, based on in vitro data, the application of nifedipine and/or verapamil may be a promising approach for ameliorating OA severity. However, very few studies on clinical outcomes are available regarding the influence of calcium antagonists, which are used primarily for treating cardiovascular conditions in OA patients. This review is intended to stimulate further research on the chondrocyte 'channelome', contribute to the development of novel therapeutic strategies and facilitate the retargeting and repositioning of existing pharmacological agents currently used for other comorbidities for the treatment of OA.


Subject(s)
Calcium Channels/physiology , Chondrocytes/physiology , Osteoarthritis/physiopathology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cartilage/drug effects , Cartilage/physiology , Chondrocytes/drug effects , Humans , Inflammation , Osteoarthritis/drug therapy
12.
Int J Mol Sci ; 16(8): 18412-38, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26262612

ABSTRACT

Mature and developing chondrocytes exist in a microenvironment where mechanical load, changes of temperature, osmolarity and acidic pH may influence cellular metabolism. Polymodal Transient Receptor Potential Vanilloid (TRPV) receptors are environmental sensors mediating responses through activation of linked intracellular signalling pathways. In chondrogenic high density cultures established from limb buds of chicken and mouse embryos, we identified TRPV1, TRPV2, TRPV3, TRPV4 and TRPV6 mRNA expression with RT-PCR. In both cultures, a switch in the expression pattern of TRPVs was observed during cartilage formation. The inhibition of TRPVs with the non-selective calcium channel blocker ruthenium red diminished chondrogenesis and caused significant inhibition of proliferation. Incubating cell cultures at 41 °C elevated the expression of TRPV1, and increased cartilage matrix production. When chondrogenic cells were exposed to mechanical load at the time of their differentiation into matrix producing chondrocytes, we detected increased mRNA levels of TRPV3. Our results demonstrate that developing chondrocytes express a full palette of TRPV channels and the switch in the expression pattern suggests differentiation stage-dependent roles of TRPVs during cartilage formation. As TRPV1 and TRPV3 expression was altered by thermal and mechanical stimuli, respectively, these are candidate channels that contribute to the transduction of environmental stimuli in chondrogenic cells.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , TRPV Cation Channels/metabolism , Animals , Cartilage/cytology , Cartilage/physiology , Cell Culture Techniques , Cells, Cultured , Chick Embryo , Chondrocytes/cytology , Chondrogenesis/drug effects , Hot Temperature , Mice , RNA, Messenger/genetics , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Transcriptome , Weight-Bearing
13.
Int J Mol Sci ; 16(8): 17344-67, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26230691

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurohormone exerting protective function during various stress conditions either in mature or developing tissues. Previously we proved the presence of PACAP signaling elements in chicken limb bud-derived chondrogenic cells in micromass cell cultures. Since no data can be found if PACAP signaling is playing any role during mechanical stress in any tissues, we aimed to investigate its contribution in mechanotransduction during chondrogenesis. Expressions of the mRNAs of PACAP and its major receptor, PAC1 increased, while that of other receptors, VPAC1, VPAC2 decreased upon mechanical stimulus. Mechanical load enhanced the expression of collagen type X, a marker of hypertrophic differentiation of chondrocytes and PACAP addition attenuated this elevation. Moreover, exogenous PACAP also prevented the mechanical load evoked activation of hedgehog signaling: protein levels of Sonic and Indian Hedgehogs and Gli1 transcription factor were lowered while expressions of Gli2 and Gli3 were elevated by PACAP application during mechanical load. Our results suggest that mechanical load activates PACAP signaling and exogenous PACAP acts against the hypertrophy inducing effect of mechanical load.


Subject(s)
Chondrocytes/metabolism , Embryonic Stem Cells/metabolism , Hedgehog Proteins/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Stress, Mechanical , Animals , Cells, Cultured , Chick Embryo , Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Signal Transduction , Trans-Activators/metabolism , Zinc Finger Protein GLI1
14.
Curr Protoc ; 4(3): e1005, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38465381

ABSTRACT

Embryonic limb bud-derived micromass cultures are valuable tools for investigating cartilage development, tissue engineering, and therapeutic strategies for cartilage-related disorders. This collection of fine-tuned protocols used in our laboratories outlines step-by-step procedures for the isolation, expansion, and differentiation of primary mouse limb bud cells into chondrogenic micromass cultures. Key aspects covered in these protocols include synchronized fertilization of mice (Basic Protocol 1), tissue dissection, cell isolation, micromass formation, and culture optimization parameters, such as cell density and medium composition (Basic Protocol 2). We describe techniques for characterizing the chondrogenic differentiation process by histological analysis (Basic Protocol 3). The protocols also address common challenges encountered during the process and provide troubleshooting strategies. This fine-tuned comprehensive protocol serves as a valuable resource for scientists working in the fields of developmental biology, cartilage tissue engineering, and regenerative medicine, offering an updated methodology for the study of efficient chondrogenic differentiation and cartilage tissue regeneration. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Synchronized fertilization of mice Basic Protocol 2: Micromass culture of murine embryonic limb bud-derived cells Basic Protocol 3: Qualitative assessment of cartilage matrix production using Alcian blue staining.


Subject(s)
Cartilage , Chondrogenesis , Animals , Mice , Cells, Cultured , Cell Differentiation , Mammals
15.
Int J Mol Sci ; 14(8): 16141-67, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23921684

ABSTRACT

Murine micromass models have been extensively applied to study chondrogenesis and osteogenesis to elucidate pathways of endochondral bone formation. Here we provide a detailed comparative analysis of the differentiation potential of micromass cultures established from either BMP-2 overexpressing C3H10T1/2 cells or mouse embryonic limb bud-derived chondroprogenitor cells, using micromass cultures from untransfected C3H10T1/2 cells as controls. Although the BMP-2 overexpressing C3H10T1/2 cells failed to form chondrogenic nodules, cells of both models expressed mRNA transcripts for major cartilage-specific marker genes including Sox9, Acan, Col2a1, Snorc, and Hapln1 at similar temporal sequence, while notable lubricin expression was only detected in primary cultures. Furthermore, mRNA transcripts for markers of osteogenic differentiation including Runx2, Osterix, alkaline phosphatase, osteopontin and osteocalcin were detected in both models, along with matrix calcification. Although the adipogenic lineage-specific marker gene FABP4 was also expressed in micromass cultures, Oil Red O-positive cells along with PPARγ2 transcripts were only detected in C3H10T1/2-derived micromass cultures. Apart from lineage-specific marker genes, pluripotency factors (Nanog and Sox2) were also expressed in these models, reflecting on the presence of various mesenchymal lineages as well as undifferentiated cells. This cellular heterogeneity has to be taken into consideration for the interpretation of data obtained by using these models.


Subject(s)
Chondrogenesis/genetics , Limb Buds/embryology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Adipogenesis/genetics , Animals , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/genetics , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Genetic Markers/genetics , Mice , Mice, Inbred C3H , RNA, Messenger/biosynthesis , Signal Transduction/genetics
16.
Curr Protoc ; 3(7): e835, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37427867

ABSTRACT

Much of the skeletal system develops by endochondral ossification, a process that takes place in early fetal life. This makes the early stages of chondrogenesis, i.e., when chondroprogenitor mesenchymal cells differentiate to chondroblasts, challenging to study in vivo. In vitro methods for the study of chondrogenic differentiation have been available for some time. There is currently high interest in developing fine-tuned methodology that would allow chondrogenic cells to rebuild articular cartilage and restore joint functionality. The micromass culture system that relies on embryonic limb bud-derived chondroprogenitor cells is a popular method for the study of the signaling pathways that control the formation and maturation of cartilage. In this protocol, we describe a technique fine-tuned in our laboratory for culturing limb bud-derived mesenchymal cells from early-stage chick embryos in high density (Basic Protocol 1). We also provide a fine-tuned method for high-efficiency transient transfection of cells before plating using electroporation (Basic Protocol 2). In addition, protocols for histochemical detection of cartilage extracellular matrix using dimethyl methylene blue, Alcian blue, and safranin O are also provided (Basic Protocol 3 and Alternate Protocols 1 and 2, respectively). Finally, a step-by-step guide on a cell viability/proliferation assay using MTT reagent is also described (Basic Protocol 4). © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Micromass culture of chick embryonic limb bud-derived cells Basic Protocol 2: Transfection of cells with siRNA constructs using electroporation prior to micromass culturing Basic Protocol 3: Qualitative and quantitative assessment of cartilage matrix production using dimethyl methylene blue staining and image analysis Alternate Protocol 1: Qualitative assessment of cartilage matrix production using Alcian blue staining Alternate Protocol 2: Qualitative assessment of cartilage matrix production using safranin O staining Basic Protocol 4: Measurement of mitochondrial activity with the MTT assay.


Subject(s)
Chickens , Methylene Blue , Animals , Chick Embryo , Methylene Blue/metabolism , Alcian Blue/metabolism , Cells, Cultured , Cartilage/metabolism , Regeneration
17.
Cells ; 10(10)2021 10 06.
Article in English | MEDLINE | ID: mdl-34685658

ABSTRACT

We investigated the gene expression pattern of selected enzymes involved in DNA methylation and the effects of the DNA methylation inhibitor 5-azacytidine during in vitro and in vivo cartilage formation. Based on the data of a PCR array performed on chondrifying BMP2-overexpressing C3H10T1/2 cells, the relative expressions of Tet1 (tet methylcytosine dioxygenase 1), Dnmt3a (DNA methyltransferase 3), and Ogt (O-linked N-acetylglucosamine transferase) were further examined with RT-qPCR in murine cell line-based and primary chondrifying micromass cultures. We found very strong but gradually decreasing expression of Tet1 throughout the entire course of in vitro cartilage differentiation along with strong signals in the cartilaginous embryonic skeleton using specific RNA probes for in situ hybridization on frozen sections of 15-day-old mouse embryos. Dnmt3a and Ogt expressions did not show significant changes with RT-qPCR and gave weak in situ hybridization signals. The DNA methylation inhibitor 5-azacytidine reduced cartilage-specific gene expression and cartilage formation when applied during the early stages of chondrogenesis. In contrast, it had a stimulatory effect when added to differentiated chondrocytes, and quantitative methylation-specific PCR proved that the DNA methylation pattern of key chondrogenic marker genes was altered by the treatment. Our results indicate that the DNA demethylation inducing Tet1 plays a significant role during chondrogenesis, and inhibition of DNA methylation exerts distinct effects in different phases of in vitro cartilage formation.


Subject(s)
Chondrogenesis/genetics , DNA Methyltransferase 3A/genetics , DNA-Binding Proteins/genetics , Epigenesis, Genetic , N-Acetylglucosaminyltransferases/genetics , Proto-Oncogene Proteins/genetics , Animals , Azacitidine/pharmacology , Bone Morphogenetic Protein 2/metabolism , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Chondrogenesis/drug effects , DNA Methylation/genetics , DNA Methyltransferase 3A/metabolism , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation, Developmental/drug effects , Mice , Models, Biological , N-Acetylglucosaminyltransferases/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism
18.
Cartilage ; 13(2_suppl): 53S-67S, 2021 12.
Article in English | MEDLINE | ID: mdl-32059614

ABSTRACT

OBJECTIVE: Circadian rhythms in cartilage homeostasis are hypothesized to temporally segregate and synchronize the activities of chondrocytes to different times of the day, and thus may provide an efficient mechanism by which articular cartilage can recover following physical activity. While the circadian clock is clearly involved in chondrocyte homeostasis in health and disease, it is unclear as to what roles it may play during early chondrogenesis. DESIGN: The purpose of this study was to determine whether the rhythmic expression of the core circadian clock was detectable at the earliest stages of chondrocyte differentiation, and if so, whether a synchronized expression pattern of chondrogenic transcription factors and developing cartilage matrix constituents was present during cartilage formation. RESULTS: Following serum shock, embryonic limb bud-derived chondrifying micromass cultures exhibited synchronized temporal expression patterns of core clock genes involved in the molecular circadian clock. We also observed that chondrogenic marker genes followed a circadian oscillatory pattern. Clock synchronization significantly enhanced cartilage matrix production and elevated SOX9, ACAN, and COL2A1 gene expression. The observed chondrogenesis-promoting effect of the serum shock was likely attributable to its synchronizing effect on the molecular clockwork, as co-application of small molecule modulators (longdaysin and KL001) abolished the stimulating effects on extracellular matrix production and chondrogenic marker gene expression. CONCLUSIONS: Results from this study suggest that a functional molecular clockwork plays a positive role in tissue homeostasis and histogenesis during early chondrogenesis.


Subject(s)
Cartilage, Articular , Circadian Clocks , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Chondrogenesis , Circadian Clocks/genetics , Extracellular Matrix/metabolism
19.
J Alzheimers Dis ; 81(3): 1195-1209, 2021.
Article in English | MEDLINE | ID: mdl-33896841

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative illness, with several peripheral pathological signs such as accumulation of amyloid-ß (Aß) plaques in the kidney. Alterations of transforming growth factor ß (TGFß) signaling in the kidney can induce fibrosis, thus disturbing the elimination of Aß. OBJECTIVE: A protective role of increased physical activity has been proven in AD and in kidney fibrosis, but it is not clear whether TGFß signalization is involved in this effect. METHODS: The effects of long-term training on fibrosis were investigated in the kidneys of mice representing a model of AD (B6C3-Tg(APPswe,PSEN1dE9)85Dbo/J) by comparing wild type and AD organs. Alterations of canonical and non-canonical TGFß signaling pathways were followed with PCR, western blot, and immunohistochemistry. RESULTS: Accumulation of collagen type I and interstitial fibrosis were reduced in kidneys of AD mice after long-term training. AD induced the activation of canonical and non-canonical TGFß pathways in non-trained mice, while expression levels of signal molecules of both TGFß pathways became normalized in trained AD mice. Decreased amounts of phosphoproteins with molecular weight corresponding to that of tau and the cleaved C-terminal of AßPP were detected upon exercising, along with a significant increase of PP2A catalytic subunit expression. CONCLUSION: Our data suggest that physical training has beneficial effects on fibrosis formation in kidneys of AD mice and TGFß signaling plays a role in this phenomenon.


Subject(s)
Alzheimer Disease/pathology , Kidney/pathology , Physical Conditioning, Animal/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Fibrosis/metabolism , Fibrosis/pathology , Kidney/metabolism , Male , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
20.
J Mol Neurosci ; 71(8): 1543-1555, 2021 Aug.
Article in English | MEDLINE | ID: mdl-31808034

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a naturally secreted signaling peptide and has important regulatory roles in the differentiation of the central nervous system and its absence results in disorders in femur development. PACAP has an important function in prevention of oxidative stress or mechanical stress in chondrogenesis but little is known about its function in bone regeneration. A new callus formation model was set to investigate its role in bone remodeling. Fracturing was 5 mm distal from the proximal articular surface of the tibia and the depth was 0.5 mm. Reproducibility of callus formation was investigated with CT 3, 7, and 21 days after the operation. Absence of PACAP did not alter the alkaline phosphatase (ALP) activation in PACAP KO healing process. In developing callus, the expression of collagen type I increased in wild-type (WT) and PACAP KO mice decreased to the end of healing process. Expression of the elements of BMP signaling was disturbed in the callus formation of PACAP KO mice, as bone morphogenic protein 4 (BMP4) and 6 showed an early reduction in bone regeneration. However, elevated Smad1 expression was demonstrated in PACAP KO mice. Our results indicate that PACAP KO mice show various signs of disturbed bone healing and suggest PACAP compensatory and fine tuning effects in proper bone regeneration.


Subject(s)
Bone Regeneration , Bony Callus/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bony Callus/physiology , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL