Fig 1 - uploaded by Giuseppe Musumeci
Content may be subject to copyright.
Telomere shortening. A telomere is a region of repetitive DNA (TTAGGG repeats) at the end of a chromosome, which protects the end of the chromosome from deterioration. Each time cells divide, telomeres shorten, and there is a limit to the number of times a given cell can go on dividing, the socalled "Hayflick limit". When the length of the telomeres is too short cell division stops. 

Telomere shortening. A telomere is a region of repetitive DNA (TTAGGG repeats) at the end of a chromosome, which protects the end of the chromosome from deterioration. Each time cells divide, telomeres shorten, and there is a limit to the number of times a given cell can go on dividing, the socalled "Hayflick limit". When the length of the telomeres is too short cell division stops. 

Source publication
Article
Full-text available
Background: Our previous studies demonstrated the expression of procollagen11A1 in fibroblasts of pancreatic cancer desmoplasia and the lack of expression in fibroblasts of pancreatitis by means of the polyclonal antibody (anti-proCOL11A1 pAb) we generated. In a similar way, we decided to compare the expression of procollagen11A1 in fibroblasts of...

Contexts in source publication

Context 1
... aging, or cell senescence, refers to the limited capacity of mitotic cells to further multiply in time (over 30-40 divisions). This limit is known as the "Hayflick limit" (Hayflick, 1984). This form of Age-related degeneration of articular cartilage is called "replicative senescence", also known as intrinsic senescence, which results from an arrest in cell-cycle progression. Some of the changes exhibited by cells, which have undergone replicative senescence can be found in cells in older adults, such as shortened telomeres, formation of senescence-associated (SA) heterochromatin (Muller, 2009) and changes of phenotype with an alteration in gene expression ( Bodnar et al., 1998). It has been hypothesized that the telomere length could be considered as a marker for replicative senescence. Telomeres cannot be completely replicated in primary cells and become shorter with each round of cell division. Telomeres are nucleoprotein structures (TTAGGG repeats) that cap the ends of the linear eukaryotic chromosomes and thereby protect their stability and integrity during replication by protecting chromosome ends against exonucleases (Fig. 1). Telomeres are replicated by a special reverse transcriptase called telomerase, in a complex mechanism that is coordinated with the genome's replication. Telomerase is an RNA-dependent DNA polymerase that synthesizes telomeric DNA sequences and comprises two essential components. One is the functional RNA component (in humans called hTERC), which serves as a template for telomeric DNA synthesis. The other is a catalytic protein (hTERT) with reverse transcriptase activity and the primary determinant for the enzyme activity ( Bryan and Cech, 1999;Kupiec, 2014). The level of telomerase in normal human somatic tissues is insufficient to prevent telomere shortening. Telomeres can be lengthened through increasing telomerase activity by exogenous expression of hTERT or hTR (the RNA template) (Greider, 1998). As proof of this concept the chondrocytes transduced with hTERT proved to be able to increase telomere length and therefore to prolong cell lifespan, increasing in this way the efficacy of cartilage repair (Martin and Buckwalter, 2003). Another type of senescence is "stress-induced senescence", also known as extrinsic senescence, which is independent of telomere length. In quiescent cells such as chondrocytes, this type of senescence may be more important than the replicative version, because progressive telomere shortening cannot completely explain senescence in these post-mitotic cells (Ben-Porath and Weinberg, 2005;Chen and Goligorsky, 2006). The various types of stress responsible for this kind of senescence include DNA damage, oxidative stress, oncogene activity, ultraviolet radiation and chronic inflammation ( Itahana et al., 2004;Campisi, 2005). Oxidative stress is thought to play a major role as a stressor. It results when the amount of reactive oxygen species (ROS) exceeds the anti-oxidant capacity of the cell. ROS are generated by intracellular enzymes such as nicotine amide adenine dinucleotide phosphate (NADPH) oxidase and 5- Age-related degeneration of articular cartilage lipoxygenase in response to activation of specific cell signaling pathways ( Kamata and Hirata, 1999;Finkel and Holbrook, 2000). A direct role for increased ROS levels in promoting cell senescence is a positive feedback activation of the ROS-protein kinase C delta (PKCδ) signaling pathway, which cooperates with the p16 INK4A -retinoblastoma protein (Rb) pathway, which plays an important role in the control of cell-cycle progression ( Takahashi et al., 2006). Telomere shortening is also observed in stress-induced senescence and it is due to oxidative damage to DNA caused by ROS. The ends of chromosomes are particularly sensitive to oxidative damage, which causes telomere erosion similar to that seen with replicative senescence ( Yudoh et al., 2005). Also, the ROS generated from excessive mechanical loading and stimulation of cytokines contribute to DNA damage, which subsequently results in telomere shortening ( Tomiyama et al., 2007;Davies et al., 2008). Cellular senescence, as well as apoptosis, can be viewed as a powerful tumor- suppressor mechanism that withdraws cells with irreparable DNA damage from the cell cycle (de Lange and Jacks, 1999;Artandi and DePinho, 2000;Puzzo et al., 2014) through the intrinsic or mitochondrial ( Loreto et al., 2011a;Caltabiano et al., 2013) and extrinsic apoptosis pathway ( Loreto et al., 2011b;Cardile et al., 2013). Several recent studies report that cartilage degeneration also coincides with increased apoptotic chondrocytes ( Musumeci et al., 2011a,b;Galanti et al., 2013). Therefore, the senescence signals, that is, a telomere-based one or a stress-based one, trigger a DNA damage response and this response shares a common signaling pathway that converges on either or both of the well-established two tumor-suppressor proteins, p53 (the p53-p21-pRb pathway) (Martin and Buckwalter, 2003;Herbig and Sedivy, 2006) and RB and pRb proteins (the p16-pRb pathway) ( Musumeci et al., 2010Musumeci et al., , 2011c). In the p53-p21-pRb pathway, senescence stimuli activate the p53, which then can induce senescence by activating pRb through p21, which is a transcriptional target of p53. This senescence can be reversed upon subsequent inactivation of p53. In the p16-pRb pathway, senescence stimuli induce p16, which activates pRb. Once the pRb pathway is engaged by p16, the senescence cannot be reversed by subsequent inactivation of p53, silencing of p16 or inactivation of pRb ( Beauséjour et al., 2003). The difference between these two pathways is that the p53-p21-pRb pathway mediates the senescence due to telomere shortening and the p16-pRB pathway is thought to mediate premature senescence ( Beauséjour et al., 2003). Once cells have entered senescence, they are arrested in the G1 phase of the cell cycle and display a characteristic morphology (vacuolated, flattened cells) and altered gene expression ( Cristofalo et al., 2004). The senescent cells exhibit the so-called "senescent secretory phenotype" (SSP), which Age-related degeneration of articular cartilage could be also correlated with the development of OA. It is interesting to note that the senescent cells, which are mitotically inactive, are biologically active (Campisi, 2005). These cells are able to increase the expression of genes that inhibit proliferation and to increase the secretion of several proteins, including inflammatory cytokines such as interleukin-6 (IL- Adda di Fagagna, 2007). Another important feature of senescent cells is represented by the epigenetic changes related to the formation of foci of heterochromatin, referred to as senescence-associated heterochromatin foci (SAHFs), which include histone variants such as the macro-H2A ( Zhang and Adams, ...
Context 2
... from older adults exhibit many changes, typical of cell senescence, when compared with cells isolated from young adults. The most evident change is represented by telomere shortening, characteristic of replicative senescence. This evidence is Age-related degeneration of articular cartilage controversial as adult articular chondrocytes rarely, if ever, divide in normal tissue in vivo. The lack of cell division in normal adult articular cartilage suggests that the chondrocytes present in the cartilage of an older adult are likely to be the same cells that were present decades earlier. This fact makes these cells more susceptible to the accumulation of changes from both aging and extrinsic stress. In fact, it is most likely that chondrocyte senescence is the extrinsic type, induced by different stressors. The telomere shortening in adult chondrocytes could be due to DNA damage from ROS as discussed further above (Mankin, 1963;Martin and Buckwalter, 2001b;Martin et al., 2004b). The increased ROS levels could be both age-related (Del Carlo and Loeser, 2003) and generated from excessive mechanical loading and/or stimulation by cytokines ( Kurz et al., 2005;Davies et al., 2008). There is also evidence for reduced levels of antioxidant enzymes in cartilage with aging and in OA that would contribute to chondrocyte senescence and oxidative damage. In human articular chondrocytes, decreased levels of mitochondrial superoxide dismutase were found both with aging and in OA cells (Finkel and Holbrook, 2000). Moreover, it has been hypothesized that joint injury accelerates chondrocyte senescence and that this acceleration plays a role in the joint degeneration responsible for post- traumatic OA. Indeed, excessive loading of articular surfaces due to acute joint trauma or post-traumatic joint instability, incongruity or mal-alignment increases release of ROS, and the increased oxidative stress on chondrocytes accelerates chondrocyte senescence ( James et al., 2004). Other important features of chondrocyte senescence are the exhibition of SSP, which has important implications in development and progression of OA and the decline in the proliferative and anabolic response to growth factors, as well as their reduction in cartilage. It has been noted that senescent chondrocytes lose the ability in response to: IGF-I, which is known to be an important autocrine survival factor that stimulates cartilage matrix synthesis (Martin et al., 1997); TGF-β, an important cartilage anabolic factor ( Scharstuhl et al., 2002) and to bone morphogenetic protein 6 (BMP-6), known to stimulate proteoglycan synthesis ( Bobacz et al., 2003). Chondrocyte senescence also contributes to the decline in the cell number within the cartilaginous tissue, due to increased cell death. Several studies demonstrated the loss of cellular density in cartilage with aging or/and in OA ( Vignon et al., 1976;Adams and Horton, 1998;Horton et al., 1998;Aigner et al., 2004a;Kuhn et al., 2004). These findings provide evidence to support the concept that chondrocyte senescence may be involved in the progression of cartilage degeneration, Age-related degeneration of articular cartilage Fig. 5. Stress-induced senescence and Osteoarthritis. The telomere shortening process in senescent chondrocytes is more probably due to the stress-induced type of senescence. Oxidative stress and excessive mechanical loading are thought to be the major stressors that induce the increased production of ROS, which are responsible for DNA damage and for the subsequent senescence of the cells. Once cells have entered senescence, they are arrested in the G1 phase of the cell cycle and they display a characteristic gene expression called "senescent secretory phenotype", which is strongly correlated with the development of ...
Context 3
... body is made up of an incredibly large number of cells, around 100 billion, some of them have a rather short life, others such as chondrocytes remain for a lifetime, but at a certain point, the mechanism slows down, cell duplication starts to fail and the cells are no longer replaced by other ones. The cells arrest their cell cycle progression. Each cell has a limited number of possible divisions, which is fixed between 50 and 70. The reason lies in the structure of chromosomes that are duplicated at each division in order to obtain one copy for itself and another for the daughter cell that will play the same role in the body. The division process, however, is not perfect, the cellular mechanism fails to copy the ends of chromosomes, the so-called telomeres (Fig. 1). After each cell division, the telomeres become shorter and shorter and are eventually completely worn out and parts of chromosomes that contain essential genetic information begin to erode. At this point the cell is at the end of its life and it approaches death ...

Similar publications

Article
Full-text available
Immunoglobulin G4 (IgG4)-related sclerosing disease, an uncommon disease entity, is known to involve various organs. To our knowledge, few reports have been presented on IgG4-related sclerosing diseases involving the mediastinum, especially the posterior mediastinum. We present a case of IgG4-related sclerosing disease of the posterior mediastinum...
Article
Full-text available
Background: One of the earliest signs of breast cancer may be the presence of mammographically detected suspicious microcalcifications in the breast. The aim of the study was to present an open surgical biopsy of the mammographically detected suspicious microcalcifications in a breast, with preoperative wire marking of the lesions and intraoperativ...
Article
Full-text available
Rationale: Immunoglobulin (Ig) G4-related disease (IgG4-RD) is a chronic inflammatory disorder characterized by high levels of serum IgG4, swollen organs with fibrosis and abundant infiltration of IgG4-positive plasmacytes. Patient concerns: An 82-year-old male visited our hospital for an evaluation of a pancreatic enlargement and a bilateral su...

Citations

... Osteoarthritis (OA), the most common disease of joints in adults, is regarded as an age-related degenerative disease (Musumeci et al., 2015). Oxidative stress is involved in the pathogenesis of osteoarthritis, which mediates chondrocyte apoptosis and extracellular matrix (ECM) degeneration through the activation of ER stress (Zahan et al., 2020). ...
Article
Full-text available
The anticancer, antioxidant, and immunomodulatory properties of carotenoids from saffron or apocarotenoids (e.g., crocin, safranal, crocetin, and picrocrocin) have prompted research into their benefits. Apocarotenoids seem to be effective compound for the treatment of chronic diseases, such as neurodegenerative, cardiovascular, cancer, respiratory, and metabolic disorders. Endoplasmic reticulum (ER) is an essential organelle found in the cytoplasm of eukaryotic cells that participates in the biosynthesis of proteins, lipids, and steroid hormones. Given the role of the ER in the regulation of several fundamental biological processes, including metabolic pathways and immune responses, aberrant ER function can have a significant influence on these vital processes and result in serious pathological consequences. Exposure of cell to adverse environmental challenges, such as toxic agents, ischemia, and so on, causes accumulation of unfolded or misfolded proteins in the ER lumen, also called ER stress. There is a growing evidence to suggest that ER disturbance in the form of oxidative/nitrosative stress and subsequent apoptotic cell death plays major roles in the pathogenesis of many human diseases, including cardiovascular diseases, diabetes mellitus, neurodegenerative diseases, and liver diseases. Apocarotenoids with their unique properties can modulate ER stress through PERK/eIF2α/ATF4/CHOP (protein kinase R (PKR)‐like ER kinase/eukaryotic initiation factor 2α/activating transcription factor 4/C/EBP /homologous protein) and X‐Box Binding Protein 1/activating transcription factor 6 (XBP1/ATF6) pathways. In addition, they suppress apoptosis through inhibition of endoplasmic and mitochondrial‐dependent caspase cascade and can stimulate SIRT1 (silent information regulator 1) and Nrf2 (nuclear factor erythroid 2‐related factor 2) expression, thereby leading to protection against oxidative stress. This review summarizes the potential benefits of apocarotenoids in various ER‐stress‐related disorders.
... Meanwhile, ADSCs treatment-induced cartilage repair can be reduced by miR-7-5p through transfection miR-7-5p into rat chondrocytes. A recent study suggested that in the early stages of OA, autophagy could be protective for OA, autophagy levels of cartilage attenuated in the OA procession, especially in the middle and late stages of OA [33,[48][49][50]. ADSCs treatment could inverse an decreased cartilage autophagy levels, shown by the activation of autophagy markers (LC3A/B and p62) in OA rat [34], consistent with our findings. ...
Article
Background: Osteoarthritis (OA) is a highly degenerative joint disease, mainly companying with progressive destruction of articular cartilage. Adipose-derived stromal cells (ADSCs) therapy enhances articular cartilage repair, extracellular matrix (ECM) synthesis and attenuates joints inflammation, but specific mechanisms of therapeutic benefit remain poorly understood. This study aimed to clarify the therapeutic effects and mechanisms of ADSCs on cartilage damage in the keen joint of OA rat model. Methods: Destabilization of the medial meniscus (DMM) and anterior cruciate ligament transection (ACLT) surgery-induced OA rats were treated with allogeneic ADSCs by intra-articular injections for 6 weeks. The protective effect of ADSCs in vivo was measured using Safranin O and fast green staining, immunofluorescence and western blot analysis. Meanwhile, the miRNA-7-5p (miR-7-5p) expression was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The mechanism of increased autophagy with ADSCs addition through decreasing miR-7-5p was revealed using oligonucleotides, and adenovirus in rat chondrocytes. The luciferase reporter assay revealed the molecular role of miR-7-5p and autophagy related 4A (ATG4A). The substrate of mTORC1 pathway: (p-)p70S6 and (p-)S6 in OA models with ADSCs addition were detected by western blotting. Results: The ADSCs treatment repaired the articular cartilage and maintained chondrocytes ECM homeostasis through modulating chondrocytes autophagy in the OA model, indicators of the change of autophagic proteins expression and autophagic flux. Meanwhile, the increased autophagy induced by ADSCs treatment was closely related to the decreased expression of host-derived miR-7-5p, a negative modulator of OA progression. Functional genomics (overexpression of genes) in vitro studies demonstrate the inhibition of host-derived miR-7-5p in mediating the benefit of ADSCs administration in OA model. Then ATG4A was defined as a target gene of miR-7-5p, and the negative relation between miR-7-5p and ATG4A was investigated in the OA model treated with ADSCs. Furthermore, miR-7-5p mediated chondrocyte autophagy by targeting ATG4A in the OA model treated with ADSCs was confirmed with the rescue trial of ATG4A/miR-7-5p overexpression on rat chondrocyte. Finally, the mTORC1 signaling pathways mediated by host-derived miR-7-5p with ADSCs treatment were decreased in OA rats. Conclusions: ADSCs promote the chondrocytes autophagy by decreasing miR-7-5p in articular cartilage by targeting ATG4A and a potential role for ADSCs based therapeutics for preventing of articular cartilage destruction and extracellular matrix (ECM) degradation in OA.
... Oxidative stress also has an important role in the progression of age-induced osteoarthritis. As we age, chondrocytes undergo progressive senescence, leading to decreased responses to growth factors, mechanical stimuli, etc., increasing the risk of osteoarthritis [18,19]. It has been found that oxidative stress can contribute to chondrocyte senescence by shortening telomerase. ...
Article
Full-text available
Background Osteoarthritis, a common degenerative osteochondral disease, has a close relationship between its mechanism of occurrence and oxidative stress. However, there are relatively few relevant studies in this field, and a more mature research system has not yet been formed. Methods By searching the Web of Science (WOS) database, we obtained 1 412 publications in the field of osteoarthritis and oxidative stress. The search results were then analyzed bibliometrically using Citespace and VOSviewer, including a study of publication trends in the field, analysis of core authors, analysis of countries and institutions with high contributions, analysis of core journals, and to identify research trends and hot spots in the field, we performed keyword clustering. Results We collected 1 412 publications on the field of osteoarthritis and oxidative stress from 1998–2022. By analyzing the publication trends in the field, we noted an exponential increase in the number of publications per year since 2014. We then identified the core authors in the field (Blanco, Francisco J., Loeser, Richard F., Vaamonde-garcia, et.al) as well as the countries (China, USA, Italy et.al) and institutions (Xi An Jiao Tong Univ, Wenzhou Med Univ, Zhejiang Univ et.al). The OSTEOARTHRITIS AND CARTILAGE and INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES contain a large number of research papers in this field, and through keyword co-occurrence analysis, we counted 3 227 keywords appearing in the field of osteoarthritis and oxidative stress. These keywords were clustered into 9 groups, representing 9 different research hotspots. Conclusions Research in the field of osteoarthritis and oxidative stress has been developing since 1998 and is now maturing, but there is an urgent need to strengthen international academic exchanges and discuss the future focus of research development in the field of osteoarthritis and oxidative stress.
... Aging represents another leading risk factor for musculoskeletal disorders, considering also the metabolic alterations that occur with aging, that may lead to undernutrition, dysbiosis, and other digestive system-related disorders, which, in turn, can worsen agerelated variations, as in a vicious circle [18,19]. The impact of undernutrition on aged skeletal muscle has been explored by Barbé et al. [20] through the skeletal muscle proteome analysis in old rats. ...
Article
Full-text available
The intention of the present Special Issue is to focus on the latest research in the musculoskeletal system, with an emphasis on the molecular mechanisms underlying its pathophysiology, as well as innovative diagnostic tools and therapeutic perspectives [...]
... Moreover, the levels of matrix metallopeptidase (MMP)-2, 3, and 13 were also decreased in the ODN+BMP-7 group. The decreases in inflammatory cytokines and matrix-degrading enzymes suggest that the ODN intervention was able to successfully downregulate the NF-κB pathway associated with the production of inflammatory proteins, which has also been observed in previous studies [18,19]. Biomolecules 2023, 13, x FOR PEER REVIEW 7 of 14 ...
... Moreover, the levels of matrix metallopeptidase (MMP)-2, 3, and 13 were also decreased in the ODN+BMP-7 group. The decreases in inflammatory cytokines and matrix-degrading enzymes suggest that the ODN intervention was able to successfully downregulate the NF-κB pathway associated with the production of inflammatory proteins, which has also been observed in previous studies [18,19]. ...
... This antiinflammatory therapeutic is being heavily studied by members of our team for the treatment of periprosthetic osteolysis by total joint arthroplasty wear particles, which cause inflammation and the subsequent activation of the NF-κB pathway. They use in vivo murine models to study ODN efficacy as an anti-inflammatory intervention in young and old mice, which is essential for OA, as many individuals with OA are older than 65 years [19]. Their studies reported the ability of ODNs to bind to the NF-κB p65:IKb complex promoter region, suppressing the genetic activation of key inflammatory genes [15]. ...
Article
Full-text available
Osteoarthritis (OA) is a painful and disabling joint disease affecting millions worldwide. The lack of clinically relevant models limits our ability to predict therapeutic outcomes prior to clinical trials, where most drugs fail. Therefore, there is a need for a model that accurately recapitulates the whole-joint disease nature of OA in humans. Emerging microphysiological systems provide a new opportunity. We recently established a miniature knee joint system, known as the miniJoint, in which human bone-marrow-derived mesenchymal stem cells (hBMSCs) were used to create an osteochondral complex, synovial-like fibrous tissue, and adipose tissue analogs. In this study, we explored the potential of the miniJoint in developing novel treatments for OA by testing the hypothesis that co-treatment with anti-inflammation and chondroinducing agents can suppress joint inflammation and associated cartilage degradation. Specifically, we created a “synovitis”-relevant OA model in the miniJoint by treating synovial-like tissues with interleukin-1β (IL-1β), and then a combined treatment of oligodeoxynucleotides (ODNs) suppressing the nuclear factor kappa beta (NF-κB) genetic pathway and bone morphogenic protein-7 (BMP-7) was introduced. The combined treatment with BMP-7 and ODNs reduced inflammation in the synovial-like fibrous tissue and showed an increase in glycosaminoglycan formation in the cartilage portion of the osteochondral complex. For the first time, this study demonstrated the potential of the miniJoint in developing disease-modifying OA drugs. The therapeutic efficacy of co-treatment with NF-κB ODNs and BMP-7 can be further validated in future clinical studies.
... Cartilages represent the fundamental components of articular joints, and are functional units essential for human movement, allowing the transmission of the mechanical load between different mobile parts of the body with minimal friction [1,4]. Cartilage degeneration is associated with chondrocyte senescence, cellular oxidative stress, and cytokine production, all of which elicit a compounded detrimental effect that can ultimately lead to the development of osteoarthritis (OA) [5]. One of the key interventions to help prevent cartilage degeneration is reducing the constant load on joints and promoting their lubrication, two conditions that are usually not met in people that follow a sedentary lifestyle [6]. ...
... An increasing body of evidence suggests that the gut microbiome may be implicated in this process [91]. It is also necessary to consider that one factor that also contributes to cartilage degeneration is chondrocyte senescence, articular oxidative stress, and the production of pro-inflammatory cytokines [5]. The positive effects of physical activity on cartilage are determined both by the intensity and typology of the activity. ...
Article
Full-text available
Physiological aging triggers a cascade of negative effects on the human body and the human joint is only one of the several compartments affected by this irreversible and natural process. Osteoarthritis and cartilage degeneration can cause pain and disability; therefore, identifying the molecular processes underlying these phenomena and the biomarkers produced during physical activity is of critical importance. In the present review, the main goal was to identify and discuss the articular cartilage biomarkers analyzed in studies in which physical or sports activities were adopted and eventually to propose a standard operating procedure for the assessment. Articles collected from Pubmed, Web of Science, and Scopus were scrutinized to detect reliable cartilage biomarkers. The principal articular cartilage biomarkers detected in these studies were cartilage oligomeric matrix protein, matrix metalloproteinases, interleukins, and carboxy-terminal telopeptide. The articular cartilage biomarkers identified in this scoping review may aid in a better comprehension of where research on the topic is heading and offer a viable instrument for streamlining investigations on cartilage biomarker discovery.
... The natural process of aging has an impact on articular cartilage with chondrocyte loss and a decline in metabolic response, alterations to the matrix and synovial tissue composition, and impairing the ability to maintain and repair these tissues [1]. Chondrocyte senescence contributes to cartilage degeneration, characterized by oxidative stress and the production of cytokines causing the so-called stress-induced senescent state [2]. A sedentary lifestyle with the consequent absence of loading for the cartilage accelerates the progression of cartilage degeneration [3]. ...
Article
Full-text available
(1) Background: Cartilage degeneration with the natural aging process and the role of physical activity on cartilage wellness is still not clear. The objective of the present review was to understand how different physical activity interventions affect the cartilage and to propose a Standard Operating Procedure for an exercise program to maintain knee joint health; (2) Methods: Articles were collected on three different electronic databases and screened against the eligibility criteria. Results were collected in tables and the main outcomes were discussed narratively; (3) Results: A total of 24 studies have been included after the screening process and aerobic, strength, flexibility, postural balance, and mobility interventions were detected. Different protocols and types of interventions were adopted by the authors; (4) Conclusions: Physical activity interventions have mainly positive outcomes on cartilage structure, but the protocols adopted are different and various. A Standard Operating Procedure has been proposed for a physical intervention focalized on cartilage wellness that could be adopted as an intervention in the clinical setting. Furthermore, the creation of a standardized protocol wants to help scientific research to move in the same direction.
... At the same time, ADSCs treatment-induced cartilage repair can be reduced by miR-7-5p through transfection miR-7-5p into rat chondrocytes. Recent study suggested that in early stages of OA, autophagy could be protective for OA, autophagy levels of cartilage attenuated in the OA procession, especially in in middle and late stages of OA [33,[47][48][49]. ADSCs treatment could inverse an decreased cartilage autophagy levels, shown by the activation of autophagy markers (LC3A/B and p62) in OA rat [34], similar to our ndings. ...
Preprint
Full-text available
Background: Osteoarthritis (OA) is a highly degenerative joint disease, mainly companying with progressive destruction of articular cartilage. Adipose-derived stromal cells (ADSCs) therapy enhances articular cartilage repair, extracellular matrix (ECM) synthesis and attenuates joints inflammation, but specific mechanisms of therapeutic benefit remain poorly understood. This study aimed to clarify the therapeutic effects and mechanisms of ADSCs on cartilage damage in keen joint of OA rat model. Methods: In vivo study, destabilization of the medial meniscus (DMM) and anterior cruciate ligament transection (ACLT) surgery-induced OA rats were treated with allogeneic ADSCs by intra-articular injections for 6 weeks. The protective effect of ADSCs in vivo was measured using Safranin O and fast green staining, immunofluorescence and western blot analysis. Meanwhile, the miRNA-7-5p (miR-7-5p) expression was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The mechanism of increased autophagy with ADSCs addition through decreasing miR-7-5p was revealed using oligonucleotides, adenovirus in rat chondrocytes. The luciferase reporter assay were used to reveal the molecular role of miR-7-5p and autophagy related 4A (ATG4A). The substrate of mTORC1 pathway: (p-)p70S6 and (p-)S6 in OA models with ADSCs addition were detected by western blotting. Results: The ADSCs treatment repaired the articular cartilage and maintained chondrocytes ECM homeostasis through modulating chondrocytes autophagy in OA model, indicators of the change of autophagic proteins expression and autophagic flux. Meanwhile, the increased autophagy induced by ADSCs treatment was closely related to the decreased expression of host-derived miR-7-5p, negative modulator of OA progression. Functional genomics (overexpression of genes) in vitro studies demonstrates inhibition of host-derived miR-7-5p in mediating the benefit of ADSCs administration in OA model. Then ATG4A were defined as a target gene of miR-7-5p, and the negative relation between miR-7-5p and ATG4A were investigated in OA model treated with ADSCs. Furthermore, miR-7-5p mediated chondrocyte autophagy by targeting ATG4A in OA cell model treated with ADSCs was confirmed with the rescue trial of ATG4A/miR-7-5p overexpression on rat chondrocyte. Finally, the mTORC1 signaling pathways mediated by host-derived miR-7-5p with ADSCs treatment were decreased in OA rats. Conclusions: ADSCs promotes the chondrocytes autophagy through decreasing miR-7-5p in articular cartilage by targeting ATG4A and a potential role for ADSCs based therapeutics for prevention of articular cartilage destruction and extracellular matrix (ECM) degradation in OA.
... Dysfunction of chondrocyte autophagy is regarded as a pivotal pathogenesis of cartilage degradation in patients with OA. 7 Recent data suggest that enhanced autophagy flux could be protective during the early stages of OA, and is able to activate an adaptive response to cell stress to promote cell survival. [19][20][21] Inhibiting autophagy attenuates the chondroprotective and anti-inflammatory effects. 22 The autophagy regulatory network exerts different mechanisms to balance homeostasis in the joints. ...
Article
Full-text available
Objective Osteoarthritis (OA) is a chronic arthropathy that frequently occurs in the middle-aged and elderly population. The aim of this study was to investigate the molecular mechanism of OA based on autophagy theory. Design We downloaded the gene expression profile from the Gene Expression Omnibus repository. Differentially expressed genes (DEGs) related to the keyword “autophagy” were identified using the scanGEO online analysis tool. DEGs representing the same expression trend were screened using the MATCH function. Clinical synovial specimens were collected for identification, pathological diagnosis, hematoxylin and eosin staining, and real-time polymerase chain reaction analysis. Differential expression of mRNAs in the synovial membrane tissues and chondrocyte monolayer samples from OA patients was used to identify potential OA biomarkers. Protein-protein interactions were established by the STRING website and visualized with Cytoscape. Functional and pathway enrichment analyses were performed using the Metascape database. Results GABARAPL1, GABARAPL2, and ATG13 were obtained as co-expressed autogenes in the 3 data sets. They were all downregulated among OA synovial tissues compared with non-OA synovial tissues ( P < 0.01). A protein-protein interaction network was constructed based on these 3 genes and included 63 genes. A functional analysis revealed that these genes were associated with autophagy-related functions. The top hub genes in the protein-protein interaction network were presented. Furthermore, 3 key modules were extracted to be core control modules. Conclusions These results offer an important molecular understanding of the key transcriptional regulatory genes and modules based on the network of potential autophagy mechanisms in human OA.
... an enhanced understanding of the mechanisms relevant to oa development is critical for the investigation of novel treatment strategies. a well-established hypothesis is that chondrocyte degeneration causes premature aging due to excessive mechanical load or oxidative stress, leading to stress-induced aging, and ultimately, the onset of oa (19). Therefore, insights into the underlying mechanisms of chondrocyte degeneration are critical to researching oa development. ...
Article
Full-text available
Osteoarthritis (OA) is one of the most prevalent pain‑inducing and disabling diseases globally. Aging is a primary contributing factor to the progression of OA. Forkhead box protein O4 (FOXO4) is known to be involved in the cell cycle and apoptosis regulation. The aim of the present study was to investigate the association between FOXO4 expression and chondrocyte degeneration in rats. Chondrocytes were assigned to the control (4‑week‑old rats), natural degeneration (16‑week‑old rats) or induced degeneration (IL‑1β‑treated chondrocytes from 4‑week‑old rats) groups. Immunocytochemical analysis with β‑galactosidase staining revealed a greater number of stained cells present in the natural and induced degeneration groups than in the control group. PCR analysis indicated lower mRNA expression levels of collagen type II α1 chain (Col2α) and higher levels of FOXO4, and western blotting revealed reduced Col2α protein expression levels and significantly elevated FOXO4 levels in the natural and induced degeneration groups, compared with those in the control group. The results of the present study revealed that FOXO4 expression was altered in the natural and induced degeneration groups, and further research and exploration are needed to clarify the underlying mechanism.