Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Zool Res ; 42(6): 746-760, 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34636194

ABSTRACT

Oxygen is an essential molecule for animal respiration, growth, and survival. Unlike in terrestrial environments, contamination and climate change have led to the frequent occurrence of hypoxia in aquatic environments, thus impacting aquatic animal survival. However, the adaptative mechanisms underlying fish responses to environmental hypoxia remain largely unknown. Here, we used large yellow croaker ( Larimichthys crocea) and large yellow croaker fry (LYCF) cells to investigate the roles of the Hif-1α/Hsf1/Hsp70 signaling pathway in the regulation of cellular redox homeostasis, and apoptosis. We confirmed that hypoxia induced the expression of Hif-1α, Hsf1, and Hsp70 in vivo and in vitro. Genetic Hsp70 knockdown/overexpression indicated that Hsp70 was required for maintaining redox homeostasis and resisting oxidative stress in LYCF cells under hypoxic stress. Hsp70 inhibited caspase-dependent intrinsic apoptosis by maintaining normal mitochondrial membrane potential, enhancing Bcl-2 mRNA and protein expression, inhibiting Bax and caspase3 mRNA expression, and suppressing caspase-3 and caspase-9 activation. Hsp70 suppressed caspase-independent intrinsic apoptosis by inhibiting nuclear translocation of apoptosis-inducing factor (AIF) and disturbed extrinsic apoptosis by inactivating caspase-8. Genetic knockdown/overexpression of Hif-1α and dual-luciferase reporter assay indicated that Hif-1α activated the Hsf1 DNA promoter and enhanced Hsf1 mRNA transcription. Hsf1 enhanced Hsp70 mRNA transcription in a similar manner. In summary, the Hif-1α/Hsf1/Hsp70 signaling pathway plays an important role in regulating redox homeostasis and anti-apoptosis in L. crocea under hypoxic stress.


Subject(s)
Heat Shock Transcription Factors/metabolism , Homeostasis/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/pharmacology , Perciformes/metabolism , Signal Transduction/physiology , Animals , Apoptosis , Cell Line , Cloning, Molecular , Computational Biology , Gene Expression Regulation/drug effects , Heat Shock Transcription Factors/genetics , Homeostasis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Oxidation-Reduction , Oxygen/chemistry , Perciformes/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Water/chemistry
2.
Zool Res ; 42(5): 592-605, 2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34387415

ABSTRACT

The large yellow croaker (Larimichthys crocea), which is an economically important mariculture fish in China, is often exposed to environmental hypoxia. Reactive oxygen species (ROS) homeostasis is essential for the maintenance of normal physiological conditions in an organism. Direct evidence that environmental hypoxia leads to ROS overproduction is scarce in marine fish. Furthermore, the sources of ROS overproduction in marine fish under hypoxic stress are poorly known. In this study, we investigated the effects of hypoxia on redox homeostasis in L. crocea and the impact of impaired redox homeostasis on fish. We first confirmed that hypoxia drove ROS production mainly via the mitochondrial electron transport chain and NADPH oxidase complex pathways in L. crocea and its cell line (large yellow croaker fry (LYCF) cells). We subsequently detected a marked increase in the antioxidant systems of the fish. However, imbalance between the pro-oxidation and antioxidation systems ultimately led to excessive ROS and oxidative stress. Cell viability showed a remarkable decrease while oxidative indicators, such as malondialdehyde, protein carbonylation, and 8-hydroxy-2 deoxyguanosine, showed a significant increase after hypoxia, accompanied by tissue damage. N-acetylcysteine (NAC) reduced ROS levels, alleviated oxidative damage, and improved cell viability in vitro. Appropriate uptake of ROS scavengers (e.g., NAC and elamipretide Szeto-Schiller-31) and inhibitors (e.g., apocynin, diphenylene iodonium, and 5-hydroxydecanoate) may be effective at overcoming hypoxic toxicity. Our findings highlight previously unstudied strategies of hypoxic toxicity resistance in marine fish.


Subject(s)
Antioxidants/metabolism , Fishes/metabolism , Oxidative Stress/physiology , Oxygen/chemistry , Oxygen/metabolism , Reactive Oxygen Species , Animals , Cell Line , Cell Survival , Environment , Homeostasis , NADP
3.
Cell Death Differ ; 28(3): 1110-1125, 2021 03.
Article in English | MEDLINE | ID: mdl-33116295

ABSTRACT

While the capacity to regenerate tissues or limbs is limited in mammals, including humans, axolotls are able to regrow entire limbs and major organs after incurring a wound. The wound blastema has been extensively studied in limb regeneration. However, due to the inadequate characterization of ECM and cell subpopulations involved in the regeneration process, the discovery of the key drivers for human limb regeneration remains unknown. In this study, we applied large-scale single-cell RNA sequencing to classify cells throughout the adult axolotl limb regeneration process, uncovering a novel regeneration-specific mitochondria-related cluster supporting regeneration through energy providing and the ECM secretion (COL2+) cluster contributing to regeneration through cell-cell interactions signals. We also discovered the dedifferentiation and re-differentiation of the COL1+/COL2+ cellular subpopulation and exposed a COL2-mitochondria subcluster supporting the musculoskeletal system regeneration. On the basis of these findings, we reconstructed the dynamic single-cell transcriptome of adult axolotl limb regenerative process, and identified the novel regenerative mitochondria-related musculoskeletal populations, which yielded deeper insights into the crucial interactions between cell clusters within the regenerative microenvironment.


Subject(s)
Ambystoma mexicanum/genetics , Ambystoma mexicanum/physiology , Mitochondria/genetics , Muscle, Skeletal/physiology , Regeneration/genetics , Amputation, Surgical , Animals , Cell Differentiation , Extremities/physiology , Extremities/surgery , Gene Expression Profiling , RNA-Seq , Single-Cell Analysis , Transcriptome
4.
Med Teach ; 42(11): 1243-1249, 2020 11.
Article in English | MEDLINE | ID: mdl-32772756

ABSTRACT

INTRODUCTION: Team-based learning (TBL) is gradually being integrated into Chinese medical education. This study reports its current application status in Chinese medical schools, as well as the underlying challenges and strategies to improve TBL application. METHOD: We screened publication databases and surveys to investigate TBL usage and concerns regarding TBL application by Chinese medical educators. Articles published by 79 Chinese medical schools include 163 articles among 20 topic areas of basic medicine and 226 articles among 16 clerkship disciplines. The opinions of 123 Chinese medical teachers were solicited from 46 medical schools in 26 provinces/municipalities. RESULTS: Approximately less than half of Chinese medical schools used TBL in basic medicine or clerkship disciplines. Among these, only 10% of schools reported TBL usage in both clerkship disciplines and basic medicine. Both quantitative and qualitative results revealed that public awareness of TBL, executive support, professional training, sharing of resources and integration of multiple disciplines are critical factors in facilitating TBL application, and in recruiting and developing TBL teachers. CONCLUSION: TBL application in Chinese medical education is limited. Executive/financial support and establishment of a platform to provide technical support, share resources and regulate TBL practice quality will facilitate TBL application in Chinese medical education.


Subject(s)
Education, Medical , Problem-Based Learning , Attitude , China , Group Processes , Humans , Schools, Medical
5.
Cell Death Differ ; 27(8): 2344-2362, 2020 08.
Article in English | MEDLINE | ID: mdl-32051546

ABSTRACT

Rab5 is a master regulator for endosome biogenesis and transport while its in vivo physiological function remains elusive. Here, we find that Rab5a is upregulated in several in vivo and in vitro myogenesis models. By generating myogenic Rab5a-deficient mice, we uncover the essential roles of Rab5a in regulating skeletal muscle regeneration. We further reveal that Rab5a promotes myoblast differentiation and directly interacts with insulin receptor substrate 1 (IRS1), an essential scaffold protein for propagating IGF signaling. Rab5a interacts with IRS1 in a GTP-dependent manner and this interaction is enhanced upon IGF-1 activation and myogenic differentiation. We subsequently identify that the arginine 207 and 222 of IRS1 and tyrosine 82, 89, and 90 of Rab5a are the critical amino acid residues for mediating the association. Mechanistically, Rab5a modulates IRS1 activation by coordinating the association between IRS1 and the IGF receptor (IGFR) and regulating the intracellular membrane targeting of IRS1. Both myogenesis-induced and IGF-evoked AKT-mTOR signaling are dependent on Rab5a. Myogenic deletion of Rab5a also reduces the activation of AKT-mTOR signaling during skeletal muscle regeneration. Taken together, our study uncovers the physiological function of Rab5a in regulating muscle regeneration and delineates the novel role of Rab5a as a critical switch controlling AKT-mTOR signaling by activating IRS1.


Subject(s)
Cell Differentiation , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/physiology , Myoblasts/cytology , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/physiology , rab5 GTP-Binding Proteins/metabolism , Animals , Cell Line , HEK293 Cells , Hindlimb/metabolism , Humans , Intracellular Membranes/metabolism , Mice, Inbred C57BL , Muscle Development/genetics , Myoblasts/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/genetics , rab5 GTP-Binding Proteins/genetics
6.
Mol Cell Biol ; 38(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30275345

ABSTRACT

The regenerative process of injured muscle is dependent on the fusion and differentiation of myoblasts derived from muscle stem cells. Hsp70 is important for maintaining skeletal muscle homeostasis and regeneration, but the precise cellular mechanism remains elusive. In this study, we found that Hsp70 was upregulated during myoblast differentiation. Depletion or inhibition of Hsp70/Hsc70 impaired myoblast differentiation. Importantly, overexpression of p38 mitogen-activated protein kinase α (p38MAPKα) but not AKT1 rescued the impairment of myogenic differentiation in Hsp70- or Hsc70-depleted myoblasts. Moreover, Hsp70 interacted with MK2, a substrate of p38MAPK, to regulate the stability of p38MAPK. Knockdown of Hsp70 also led to downregulation of both MK2 and p38MAPK in intact muscles and during cardiotoxin-induced muscle regeneration. Hsp70 bound MK2 to regulate MK2-p38MAPK interaction in myoblasts. We subsequently identified the essential regions required for Hsp70-MK2 interaction. Functional analyses showed that MK2 is essential for both myoblast differentiation and skeletal muscle regeneration. Taken together, our findings reveal a novel role of Hsp70 in regulating myoblast differentiation by interacting with MK2 to stabilize p38MAPK.


Subject(s)
Cell Differentiation/physiology , HSP70 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Down-Regulation/physiology , Mice , Mice, Inbred C57BL , Muscle Development/physiology , Muscle, Skeletal/physiology , Myoblasts/physiology , Up-Regulation/physiology
7.
Tissue Eng Part B Rev ; 24(6): 443-453, 2018 12.
Article in English | MEDLINE | ID: mdl-29724151

ABSTRACT

Tendon injuries are common musculoskeletal system disorders, but the tendons have poor regeneration ability. To address this issue, tendon tissue engineering provides potential strategies for future therapeutic treatment. Elements of the physical microenvironment, such as the mechanical force and surface topography, play a vital role in regulating stem cell fate, enhancing the differentiation efficiency of seed cells in tendon tissue engineering. Various inducible scaffolds have been widely explored for tendon regeneration, and scaffold-enhancing modifications have been extensively studied. In this review, we systematically summarize the effects of the physical microenvironment on stem cell differentiation and tendon regeneration; we also provide an overview of the inducible scaffolds for stem cell tenogenic differentiation. Finally, we suggest some potential scaffold-based therapies for tendon injuries, presenting an interesting perspective on tendon regenerative medicine.


Subject(s)
Cell Differentiation , Regeneration , Stem Cells/cytology , Tendons/cytology , Tissue Engineering/methods , Animals , Humans , Stem Cells/physiology , Tendons/physiology , Tissue Scaffolds
8.
Front Pharmacol ; 9: 210, 2018.
Article in English | MEDLINE | ID: mdl-29636680

ABSTRACT

Osteoporosis is a common health problem worldwide caused by an imbalance of bone formation vs. bone resorption. However, current therapeutic approaches aimed at enhancing bone formation or suppressing bone resorption still have some limitations. In this study, we demonstrated for the first time that cepharanthine (CEP, derived from Stephania cepharantha Hayata) exerted a protective effect on estrogen deficiency-induced bone loss. This protective effect was confirmed to be achieved through inhibition of bone resorption in vivo, rather than through enhancement of bone formation in vivo. Furthermore, the in vitro study revealed that CEP attenuated receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast formation, and suppressed bone resorption by impairing the c-Jun N-terminal kinase (JNK) and phosphatidylinositol 3-kinase (PI3K)-AKT signaling pathways. The inhibitory effect of CEP could be partly reversed by treatment with anisomycin (a JNK and p38 agonist) and/or SC79 (an AKT agonist) in vitro. Our results thus indicated that CEP could prevent estrogen deficiency-induced bone loss by inhibiting osteoclastogenesis. Hence, CEP might be a novel therapeutic agent for anti-osteoporosis therapy.

9.
Stem Cells ; 36(4): 527-539, 2018 04.
Article in English | MEDLINE | ID: mdl-29315990

ABSTRACT

Tendon repair is a clinical challenge because of the limited understanding on tenogenesis. The synthesis of type I collagen (Collagen I) and other extracellular matrix are essential for tendon differentiation and homeostasis. Current studies on tenogenesis focused mostly on the tenogenic transcriptional factors while the signaling controlling tenogenesis on translational level remains largely unknown. Here, we showed that mechanistic target of rapamycin (mTOR) signaling was activated by protenogenic growth factor, transforming growth factors beta1, and insulin-like growth factor-I. The expression of mTOR was upregulated during tenogenesis of mesenchymal stem cells (MSCs). Moreover, mTOR was downregulated in human tendinopathy tissues and was inactivated upon statin treatment. Both inhibition and depletion of AKT or mTOR significantly reduced type I collagen production and impaired tenogenesis of MSCs. Tendon specific-ablation of mTOR resulted in tendon defect and reduction of Collagen I. However, there is no evident downregulation of tendon associated collagens at the transcription level. Our study demonstrated that AKT-mTOR axis is a key mediator of tendon differentiation and provided a novel therapeutic target for tendinopathy and tendon injuries. Stem Cells 2018;36:527-539.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tendons/metabolism , Animals , Mesenchymal Stem Cells/cytology , Mice , Tendons/cytology , Transforming Growth Factor beta1/metabolism
10.
Br J Pharmacol ; 175(6): 859-876, 2018 03.
Article in English | MEDLINE | ID: mdl-29130485

ABSTRACT

BACKGROUND AND PURPOSE: Aseptic prosthesis loosening, caused by wear particles, is one of the most common causes of arthroplasty failure. Extensive and over-activated osteoclast formation and physiological functioning are regarded as the mechanism of prosthesis loosening. Therapeutic modalities based on inhibiting osteoclast formation and bone resorption have been confirmed to be an effective way of preventing aseptic prosthesis loosening. In this study, we have investigated the effects of sophocarpine (SPC, derived from Sophora flavescens) on preventing implant loosening and further explored the underlying mechanisms. EXPERIMENTAL APPROACH: The effects of SPC in inhibiting osteoclastogenesis and bone resorption were evaluated in osteoclast formation, induced in vitro by the receptor activator of NF-κB ligand (RANKL). A rat femoral particle-induced peri-implant osteolysis model was established. Subsequently, micro-CT, histology, mechanical testing and bone turnover were used to assess the effects of SPC in preventing implant loosening. KEY RESULTS: In vitro, we found that SPC suppressed osteoclast formation, bone resorption, F-actin ring formation and osteoclast-associated gene expression by inhibiting NF-κB signalling, specifically by targeting IκB kinases. Our in vivo study showed that SPC prevented particle-induced prosthesis loosening by inhibiting osteoclast formation, resulting in reduced periprosthetic bone loss, diminished pseudomembrane formation, improved bone-implant contact, reduced bone resorption-related turnover and enhanced stability of implants. Inhibition of NF-κB signalling by SPC was confirmed in vivo. CONCLUSION AND IMPLICATIONS: SPC can prevent implant loosening through inhibiting osteoclast formation and bone resorption. Thus, SPC might be a novel therapeutic agent to prevent prosthesis loosening and for osteolytic diseases.


Subject(s)
Alkaloids/pharmacology , Bone Resorption/prevention & control , Osteoclasts/drug effects , Osteogenesis/drug effects , Alkaloids/isolation & purification , Animals , Disease Models, Animal , Male , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteolysis/prevention & control , Prosthesis Failure , RANK Ligand/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sophora/chemistry
11.
Chemosphere ; 184: 907-915, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28651317

ABSTRACT

Land-based recirculating aquaculture systems (RAS) and cage culture are important methods of Larimichthys crocea production. The effects of environmental factors on physiological and biochemical aspects of L. crocea require clarification. Temperature and salinity are controlled in RAS and directly affect L. crocea growth and survival. To explore optimal parameters, the oxygen consumption rate (RO), ammonium excretion rate (RN), and O/N ratio at different temperatures (8, 14, 20, 26, and 32 °C) and salinities (5, 15, 25, and 35‰) were determined. RO, RN, and O/N first increased and then decreased with elevated temperature and salinity, peaking at 26 °C and 25‰, respectively. This suggests that the metabolism of L. crocea was maximal at 26 °C and 25‰ salinity, which promote its growth and survival. Additionally, hypoxia affects cage culture, and has significant effects on enzymatic activities and stress-inducible gene expression. To accelerate the selective breeding of hypoxia-tolerant L. crocea in cage culture, we measured adenosine triphosphatase (ATPase), lactate dehydrogenase (LDH), and succinate dehydrogenase (SDH) activities, and hypoxia-inducing factor 1 (HIF-1) mRNA expression in the myocardium under hypoxia (2.5, 3.5, and 4.5 mg L-1). ATPase and SDH activities first decreased and then increased under hypoxia, whereas LDH activity and HIF-1α expression first increased and then decreased. Thus, under hypoxia, the myocardial mitochondria switched from being susceptible to being resistant to injury induced by energy metabolism, and respiration in L. crocea likely converted from aerobic to anaerobic during adaptation. Furthermore, the upregulation of HIF-1α mRNA suggests it has an active role in protection against anoxic damage.


Subject(s)
Adaptation, Physiological , Perciformes/physiology , Acclimatization , Animals , Energy Metabolism , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Perciformes/metabolism , RNA, Messenger/metabolism , Salinity , Stress, Physiological
12.
PLoS One ; 12(5): e0178781, 2017.
Article in English | MEDLINE | ID: mdl-28562696

ABSTRACT

Steroid-associated osteonecrosis (SAON) might induce bone collapse and subsequently lead to joint arthroplasty. Core decompression (CD) is regarded as an effective therapy for early-stage SAON, but the prognosis is unsatisfactory due to incomplete bone repair. Parathyroid hormone[1-34] (PTH[1-34]) has demonstrated positive efficacy in promoting bone formation. We therefore evaluated the effects of PTH on improving the effects of CD in Early-Stage SAON. Distal femoral CD was performed two weeks after osteonecrosis induction or vehicle injection, with ten of the ON-induced rabbits being subjected to six-week PTH[1-34] treatment and the others, including ON-induced and non-induced rabbits, being treated with vehicle. MRI confirmed that intermittent PTH administration improved SAON after CD therapy. Micro-CT showed increased bone formation within the tunnel. Bone repair was enhanced with decreased empty osteocyte lacunae and necrosis foci area, resulting in enhanced peak load and stiffness of the tunnel. Additionally, PTH enlarged the mean diameter of vessels in the marrow and increased the number of vessels within the tunnels, as well as elevated the expression of BMP-2, RUNX2, IGF-1, bFGF and VEGF, together with serum OCN and VEGF levels. Therefore, PTH[1-34] enhances the efficacy of CD on osteogenesis and neovascularization, thus promoting bone and blood vessels repair in the SAON model.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Decompression, Surgical , Disease Models, Animal , Neovascularization, Physiologic/drug effects , Osteogenesis , Osteonecrosis/drug therapy , Parathyroid Hormone/pharmacology , Animals , Male , Osteonecrosis/chemically induced , Osteonecrosis/physiopathology , Rabbits
13.
Chemosphere ; 169: 418-427, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27889508

ABSTRACT

Organisms at all levels of evolutionary complexity react to hypoxic stress. To clarify the effects of acute hypoxia on physiological and biochemical responses of Larimichthys crocea, we measured the activity levels of the antioxidant enzymes superoxide dismutase and catalase, hemoglobin concentration, functional indices of the liver (aspartate transaminase, alanine transaminase), heart (phosphocreatine kinase), and immune system (alkaline phosphatase), as well as mRNA expression levels of the immunity-related genes Hsp70 and HIF-1α at different time points of hypoxic. In addition, liver, gill, and kidney samples were histologically analyzed. We found that hemoglobin concentration and all enzyme activities increased during hypoxia, although these effects were transient and most indices returned to basal levels thereafter. The extent of the increase in the parameter values was inversely proportional to the dissolved oxygen content. Hsp70 and HIF-1α mRNA expression levels increased significantly in the blood, liver, gills, and kidneys following exposure to hypoxia, which may play an important role in protecting fish against oxidative damage. However, we found histological evidence of hypoxia-induced injuries to the gills, liver, and kidneys, which are involved in breathing, detoxification, and osmotic balance maintenance, respectively. Thus, despite the upregulation of defensive mechanisms, acute hypoxia still caused irreversible damage of organs. In conclusion, we observed that, in response to acute hypoxic stress, L. crocea enhances immune defensive function and antioxidant capacity. A better understanding of the regulation of the molecular anti-hypoxia mechanisms can help speeding up the selective breeding of hypoxia-tolerant L. crocea.


Subject(s)
Catalase/metabolism , Gills/injuries , Hypoxia/pathology , Kidney/injuries , Liver/injuries , Perciformes/metabolism , Superoxide Dismutase/metabolism , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Creatine Kinase, MB Form/blood , Gene Expression , Gene Expression Regulation , Gills/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/metabolism , Liver/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/blood
14.
Stem Cells ; 34(4): 1083-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26851078

ABSTRACT

Calcification of soft tissues, such as heart valves and tendons, is a common clinical problem with limited therapeutics. Tissue specific stem/progenitor cells proliferate to repopulate injured tissues. But some of them become divergent to the direction of ossification in the local pathological microenvironment, thereby representing a cellular target for pharmacological approach. We observed that HIF-2alpha (encoded by EPAS1 inclined form) signaling is markedly activated within stem/progenitor cells recruited at calcified sites of diseased human tendons and heart valves. Proinflammatory microenvironment, rather than hypoxia, is correlated with HIF-2alpha activation and promoted osteochondrogenic differentiation of tendon stem/progenitor cells (TSPCs). Abnormal upregulation of HIF-2alpha served as a key switch to direct TSPCs differentiation into osteochondral-lineage rather than teno-lineage. Notably, Scleraxis (Scx), an essential tendon specific transcription factor, was suppressed on constitutive activation of HIF-2alpha and mediated the effect of HIF-2alpha on TSPCs fate decision. Moreover, pharmacological inhibition of HIF-2alpha with digoxin, which is a widely utilized drug, can efficiently inhibit calcification and enhance tenogenesis in vitro and in the Achilles's tendinopathy model. Taken together, these findings reveal the significant role of the tissue stem/progenitor cells fate decision and suggest that pharmacological regulation of HIF-2alpha function is a promising approach for soft tissue calcification treatment.


Subject(s)
Achilles Tendon/drug effects , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Calcinosis/drug therapy , Therapy, Soft Tissue , Achilles Tendon/growth & development , Achilles Tendon/pathology , Aged , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Calcinosis/genetics , Calcinosis/pathology , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cellular Microenvironment/drug effects , Chondrogenesis/genetics , Digoxin/administration & dosage , Humans , Male , Middle Aged , Rats , Rheumatic Heart Disease/genetics , Rheumatic Heart Disease/pathology , Stem Cells/drug effects , Stem Cells/pathology
15.
Sci Adv ; 2(11): e1600874, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28138519

ABSTRACT

The repair of injured tendons remains a formidable clinical challenge because of our limited understanding of tendon stem cells and the regulation of tenogenesis. With single-cell analysis to characterize the gene expression profiles of individual cells isolated from tendon tissue, a subpopulation of nestin+ tendon stem/progenitor cells (TSPCs) was identified within the tendon cell population. Using Gene Expression Omnibus datasets and immunofluorescence assays, we found that nestin expression was activated at specific stages of tendon development. Moreover, isolated nestin+ TSPCs exhibited superior tenogenic capacity compared to nestin- TSPCs. Knockdown of nestin expression in TSPCs suppressed their clonogenic capacity and reduced their tenogenic potential significantly both in vitro and in vivo. Hence, these findings provide new insights into the identification of subpopulations of TSPCs and illustrate the crucial roles of nestin in TSPC fate decisions and phenotype maintenance, which may assist in future therapeutic strategies to treat tendon disease.


Subject(s)
Databases, Nucleic Acid , Gene Expression Regulation/physiology , Nestin/metabolism , Stem Cells/metabolism , Tendons/metabolism , Animals , Mice , Mice, Transgenic , Nestin/genetics , Stem Cells/cytology , Tendons/cytology
16.
Biomaterials ; 53: 239-50, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25890723

ABSTRACT

Elucidating the regulatory mechanisms of osteogenesis of human mesenchymal stem cell (hMSC) is important for the development of cell therapies for bone loss and regeneration. Here we showed that hsa-miR-199a-5p modulated osteogenic differentiation of hMSCs at both early and late stages through HIF1a pathway. hsa-miR-199a expression was up-regulated during osteogenesis for both of two mature forms, miR-199a-5p and -3p. Over-expression of miR-199a-5p but not -3p enhanced differentiation of hMSCs in vitro, whereas inhibition of miR-199a-5p reduced the expression of osteoblast-specific genes, alkaline phosphatase (ALP) activity, and mineralization. Furthermore, over-expression of miR-199a enhanced ectopic bone formation in vivo. Chitosan nanoparticles were used for delivery of stable modified hsa-miR-199a-5p (agomir) both in vitro and in vivo, as a proof-of-concept for stable agomir delivery on bone regeneration. The hsa-mir199a-5p agomir were mixed with Chitosan nanoparticles to form nanoparticle/hsa-mir199a-5p agomir plasmid (nanoparticle/agomir) complexes, and nanoparticle/agomir complexes could improve the in vivo regeneration of bone. Further mechanism studies revealed that hypoxia enhanced osteogenesis at early stage and inhibited osteogenesis maturation at late stage through HIF1a-Twist1 pathway. At early stage of differentiation, hypoxia induced HIF1a-Twist1 pathway to enhance osteogenesis by up-regulating miR-199a-5p, while at late stage of differentiation, miR-199a-5p enhanced osteogenesis maturation by inhibiting HIF1α-Twist1 pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mesenchymal Stem Cells/drug effects , MicroRNAs/administration & dosage , Nanoparticles , Osteogenesis/drug effects , Animals , Cell Differentiation/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred NOD , MicroRNAs/pharmacology , Nuclear Proteins/metabolism , Rats , Rats, Sprague-Dawley , Twist-Related Protein 1/metabolism
17.
Biomaterials ; 44: 173-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25617136

ABSTRACT

Physical topographic cues from various substrata have been shown to exert profound effects on the growth and differentiation of stem cells due to their niche-mimicking features. However, the biological function of different topographic materials utilized as bio-scaffolds in vivo have not been rigorously characterized. This study investigated the divergent differentiation pathways of mesenchymal stem cells (MSCs) and neo-tissue formation trigged by aligned and randomly-oriented fibrous scaffolds, both in vitro and in vivo. The aligned group was observed to form more mature tendon-like tissue in the Achilles tendon injury model, as evidenced by histological scoring and collagen I immunohistochemical staining data. In contrast, the randomly-oriented group exhibited much chondrogenesis and subsequent bone tissue formation through ossification. Additionally, X-ray imaging and osteocalcin immunohistochemical staining also demonstrated that osteogenesis in vivo is driven by randomly oriented topography. Furthermore, MSCs on the aligned substrate exhibited tenocyte-like morphology and enhanced tenogenic differentiation compared to cells grown on randomly-oriented scaffold. qRT-PCR analysis of osteogenic marker genes and alkaline phosphatase (ALP) staining demonstrated that MSCs cultured on randomly-oriented fiber scaffolds displayed enhanced osteogenic differentiation compared with cells cultured on aligned fiber scaffolds. Finally, it was demonstrated that cytoskeletal tension release abrogated the divergent differentiation pathways on different substrate topography. Collectively, these findings illustrate the relationship between topographic cues of the scaffold and their inductive role in tissue regeneration; thus providing an insight into future development of smart functionalized bio-scaffold design and its application in tissue engineering.


Subject(s)
Cell Differentiation , Cell Lineage , Regeneration/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Achilles Tendon/diagnostic imaging , Achilles Tendon/physiology , Alkaline Phosphatase/metabolism , Animals , Biomarkers/metabolism , Biomechanical Phenomena , Cell Line , Cells, Cultured , Cytoskeleton/metabolism , Female , Gene Expression Regulation , Immunohistochemistry , Lactic Acid/chemistry , Mesenchymal Stem Cells , Mice , Nanofibers/chemistry , Nanofibers/ultrastructure , Osteogenesis , Polyesters , Polymers/chemistry , Radiography , Rats , Staining and Labeling , Wound Healing , X-Rays
18.
Sci Rep ; 4: 5515, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24992450

ABSTRACT

Injured adult tendons do not exhibit optimal healing through a regenerative process, whereas fetal tendons can heal in a regenerative fashion without scar formation. Hence, we compared FFs (mouse fetal fibroblasts) and AFs (mouse adult fibroblasts) as seed cells for the fabrication of scaffold-free engineered tendons. Our results demonstrated that FFs had more potential for tendon tissue engineering, as shown by higher levels of tendon-related gene expression. In the in situ AT injury model, the FFs group also demonstrated much better structural and functional properties after healing, with higher levels of collagen deposition and better microstructure repair. Moreover, fetal fibroblasts could increase the recruitment of fibroblast-like cells and reduce the infiltration of inflammatory cells to the injury site during the regeneration process. Our results suggest that the underlying mechanisms of better regeneration with FFs should be elucidated and be used to enhance adult tendon healing. This may assist in the development of future strategies to treat tendon injuries.


Subject(s)
Achilles Tendon/physiopathology , Fibroblasts/physiology , Regeneration , Achilles Tendon/metabolism , Achilles Tendon/pathology , Animals , Biomechanical Phenomena , Cell Survival , Cells, Cultured , Collagen Type III/genetics , Collagen Type III/metabolism , Female , Fetus/cytology , Fibroblasts/transplantation , Gene Expression , Mice , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Receptor, EphA4/genetics , Receptor, EphA4/metabolism , Tissue Engineering
19.
Tissue Eng Part A ; 20(13-14): 1815-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24410299

ABSTRACT

Although cell transplantation therapy can effectively promote functional tendon repair, occasional ectopic ossification during tendon regeneration undermines its efficacy. The effect of transplanted cell types on ectopic ossification has not yet been systematically evaluated. This study compared the rate of ectopic ossification during tendon repair upon transplantation with mouse fetal fibroblasts (FFs) and their adult counterparts (adult fibroblasts [AFs]). Alkaline phosphatase (ALP) staining, immunofluorescence, and gene expression analysis were used to compare the spontaneous osteogenic differentiation of FFs and AFs in vitro. X-ray, histology, and gene expression analysis were used to investigate the ectopic ossification in a mouse Achilles tendon repair model in vivo. ALP staining and immunofluorescence data in vitro showed that FFs had less spontaneous osteogenic differentiation capacity, and lower expression of runt-related transcription factor 2 (runx2). For the in vivo study, the FFs transplant group displayed reduced ectopic ossification (2/7 vs. 7/7, Mann-Whitney test p<0.01) at 14 weeks post-transplantation and enhanced tendon repair (general histological score at week 6, 7.53 vs. 10.56, p<0.05). More chondrocytes formed at 6 weeks, and all mice developed bone marrow at 14 weeks post-transplantation in the AFs transplant group. Gene expression analysis of the regenerated tissue showed significantly higher expression levels of transforming growth factor beta1 (TGF-ß1) and transforming growth factor beta3 (TGF-ß3) in the AFs group during the early stages of tendon repair. Our study demonstrates that transplantation of fetal instead of AFs is more promising for tendon repair, underscoring the importance of the origin of seed cells for tendon repair.


Subject(s)
Aging/physiology , Fetus/cytology , Fibroblasts/transplantation , Ossification, Heterotopic/pathology , Tendons/pathology , Wound Healing , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Cell Separation , Cell Shape , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Mice, Inbred ICR , Osteogenesis , Radiography , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Tendons/diagnostic imaging , Transforming Growth Factor beta/metabolism
20.
Tissue Eng Part A ; 20(11-12): 1583-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24328506

ABSTRACT

AIM: Despite our previous study that demonstrates that human embryonic stem cells (hESCs) can be used as seed cells for tendon tissue engineering after stepwise induction, suboptimal tendon regeneration implies that a new strategy needs to be developed for tendon repair. We investigated whether overexpression of the tendon-specific transcription factor scleraxis (SCX) in hESC-derived mesenchymal stem cells (hESC-MSCs) together with knitted silk-collagen sponge scaffold could promote tendon regeneration. METHODS AND RESULTS: hESCs were initially differentiated into MSCs and then engineered with scleraxis (SCX+hESC-MSCs). Engineered tendons were constructed with SCX+hESC-MSCs and a knitted silk-collagen sponge scaffold and then mechanical stress was applied. SCX elevated tendon gene expression in hESC-MSCs and concomitantly attenuated their adipogenic and chondrogenic potential. Mechanical stress further augmented the expression of tendon-specific genes in SCX+hESC-MSC-engineered tendon. Moreover, in vivo mechanical stimulation promoted the alignment of cells and increased the diameter of collagen fibers after ectopic transplantation. In the in vivo tendon repair model, the SCX+hESC-MSC-engineered tendon enhanced the regeneration process as shown by histological scores and superior mechanical performance compared with control cells, especially at early stages. CONCLUSION: Our study offers new evidence concerning the roles of SCX in tendon differentiation and regeneration. We demonstrated a novel strategy of combining hESCs, genetic engineering, and tissue-engineering principles for tendon regeneration, which are important for the future application of hESCs and silk scaffolds for tendon repair.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Collagen/pharmacology , Mesenchymal Stem Cells/cytology , Silk/pharmacology , Tendons/physiology , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Biomechanical Phenomena/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Implants, Experimental , Mesenchymal Stem Cell Transplantation , Mesoderm/cytology , Mice, Nude , Organ Specificity , Rats, Sprague-Dawley , Regeneration/drug effects , Tendons/drug effects , Tendons/pathology , Tendons/ultrastructure , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...