Next Article in Journal
Exploring the Assembly of Resorc[4]arenes for the Construction of Supramolecular Nano-Aggregates
Previous Article in Journal
Novel Translational and Phosphorylation Modification Regulation Mechanisms of Tomato (Solanum lycopersicum) Fruit Ripening Revealed by Integrative Proteomics and Phosphoproteomics
Previous Article in Special Issue
Oxytocin and Bone: Review and Perspectives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Relationship between Oxytocin and Osteoarthritis: Hope or Despair?

by
Stephanie Ferrero
1,
Ez-Zoubir Amri
2 and
Christian Hubert Roux
1,2,*
1
Rheumatology Department, Hospital Pasteur 2 CHU, 06000 Nice, France
2
Inserm, CNRS, iBV, Université Côte d’Azur, 06000 Nice, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(21), 11784; https://doi.org/10.3390/ijms222111784
Submission received: 2 September 2021 / Revised: 21 October 2021 / Accepted: 29 October 2021 / Published: 29 October 2021
(This article belongs to the Special Issue Oxytocin: Involvement in Metabolic Disorders)

Abstract

:
Oxytocin (OT) is involved in breastfeeding and childbirth and appears to play a role in regulating the bone matrix. OT is synthesized in the supraoptic and paraventricular nuclei of the hypothalamus and is released in response to numerous stimuli. It also appears to be produced by osteoblasts in the bone marrow, acting as a paracrine–autocrine regulator of bone formation. Osteoarthritis (OA) is a disease of the whole joint. Different tissues involved in OA express OT receptors (OTRs), such as chondrocytes and osteoblasts. This hormone, which levels are reduced in patients with OA, appears to have a stimulatory effect on chondrogenesis. OT involvement in bone biology could occur at both the osteoblast and chondrocyte levels. The relationships between metabolic syndrome, body weight, and OA are well documented, and the possible effects of OT on different parameters of metabolic syndrome, such as diabetes and body weight, are important. In addition, the effects of OT on adipokines and inflammation are also discussed, especially since recent data have shown that low-grade inflammation is also associated with OA. Furthermore, OT also appears to mediate endogenous analgesia in animal and human studies. These observations provide support for the possible interest of OT in OA and its potential therapeutic treatment.

1. Introduction

Osteoarthritis (OA) is the most common form of arthritis and a leading cause of impaired mobility in the elderly population [1]. It is a major public health problem in an ageing and increasingly obese worldwide population, and the incidence of OA could increase in the coming years. Therapeutics for OA remain limited, with a constant increase in the cost of care, especially for lower limb localization [2], despite notable advances in understanding its physiopathology. However, recent advances in the epidemiologic and basic research on OA have permitted the differentiation of clinical phenotypes based on the following risk factors: aging, trauma, heredity, obesity, and metabolic syndrome [2,3]. The classification of OA into different phenotypes provides another lens to this complex disease and explains its different evolutionary trajectories depending on various etiological factors. This also suggests the use of specific therapies depending on the phenotype. These observations suggest that there is no longer one but several types of OA.
Alterations in the expression of different signaling pathway-related molecules, such as the TGF-β superfamily of proteins, Wnt/β-catenin, Notch, and Indian Hedgehog (Ihh) have been shown to contribute to the development and progression of OA by primarily inducing catabolic responses [3,4,5,6,7,8,9,10,11]. Such responses include the upregulation of inflammatory mediators that lead to cartilage extracellular matrix (ECM) degradation via increased expression of matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTs) [12,13,14,15,16]. OA was previously defined as a non-inflammatory arthropathy, but it is now well-recognized that it affects the joint as a whole, confirming an inflammatory component of this disease.
Oxytocin (OT), a neurohypophyseal hormone discovered in 1906, is synthesized in the paraventricular and supraoptic nuclei of the hypothalamus and is secreted by the posterior pituitary into the general circulation [17,18,19,20,21]. It plays an essential role in reproduction, acting on the smooth muscles of the uterus and the muscles of the mammary glands, particularly during and after childbirth. OT exerts its pharmacological properties by activating specific receptors present in multiple tissues of the body [18]. A growing body of evidence has shown that OT contributes to modulating several functions, such as social recognition, trust, anti-nociception, anti-inflammation, stress reduction, sexuality, and generosity [21,22,23,24,25]. It has also been shown to be produced by peripheral tissues, such as adipocytes, osteoblasts, muscles, testes, ovaries, heart, lung, and vascular tissue [17,18,19,26,27].
In addition, OT receptors (OTRs) have been shown to be expressed on human chondrocytes [27,28], osteoblasts [29], adipocytes, vasculo-endothelial cells [20], monocytes and macrophages [30], and brain cells [31]. Together, the inflammatory status and the expression of OTRs in previous studies have suggested that OT may play a role in OA. Furthermore, multiple studies have shown that OT may also have anti-inflammatory and antioxidant properties and regulate immune and anti-inflammatory responses. For example, OT has been shown to inhibit the secretion of pro-inflammatory cytokines, such as TNF-α, interleukin (IL)-6, IL-1b, glutamate, nitric oxide (NO), and reactive oxygen species (ROS) [32,33].
Interestingly, these parameters show that there are different inflammatory factors implicated in the pathophysiology of OA. OA is now known to be a low-grade inflammatory disorder of the joint as a whole, with inflammation driving several pathological changes [34]. As a progressive joint degenerative disorder, OA is characterized by cartilage damage, changes in the subchondral bone, osteophyte formation, and muscle weakness.
Inflammatory cytokines (TNF-α, IL-6, etc.) play an important role in chondrocyte catabolism at the cartilage level. Matrix-degrading enzymes are considered to play key roles in the degradation of cartilage matrix proteins and MMPs [35,36,37]. MMP expression is regulated by transcription factors, such as activator protein-1 (AP-1), PEA3, RUNX2, and nuclear factor (NF)-κB [38]. Mutations in the AP-1 site completely inhibited the induction of MMP expression. AP-1 contains members of the Fos and Jun families of proteins, where c-Fos/AP-1 directly controls MMP expression by binding to the AP-1 site on the promoters of inflammatory cytokines and MMP genes [39,40,41]. OT activates the MAPK cascade via different pathways, including the trans-activation of receptor tyrosine kinases and possibly different G protein-linked pathways, leading to c-Fos and c-Jun induction [42].
Moreover, Wu et al., showed that OTR expression in primary chondrocytes from patients with OA was reduced compared to that in chondrocytes from control patients [28]. That study showed a reduced expression of the OTR in response to TNF-α treatment, the ability of OT to reverse TNF-α-induced Col II degradation in a dose-dependent manner, and that OT attenuated TNF-α-induced MMP-1 and MMP-13 expression, as well as JAK2/STAT1 activation. Importantly, the inhibitory effect of OT on TNF-α-induced Col II degradation was found to be dependent on OTR. These results highlight the potential of OT in maintaining cartilage integrity. In a previous study, we showed that OT stimulates chondrogenesis in vitro [43]. We observed an increase in glycosaminoglycan content in the extracellular environment following OT treatment. OT induced an increase in the expression of aggrecan, cartilage oligomeric matrix protein (COMP), and SRT-related HMG-box gene 9 (Sox9). In contrast, the expression of the fibrous tissue marker Col A1 was downregulated. Moreover, we found that administration of OT attenuated the effects of IL-1β, as shown by the reduction in ADAMTS-4 mRNA transcript levels; thus, OT may modulate chondrogenesis. A recent study also showed that OT controls chondrocyte matrix degradation through the downregulation of metalloproteinases mRNA expression, strongly supporting the role of OT in the physiopathology of OA [28].

2. The Subchondral Bone

The subchondral bone is an intricate structure consisting of two distinct anatomic entities: the subchondral bone plate and subchondral trabecular bone, both of which have a close biomechanical and biochemical relationship with the overlying cartilage [44]. The subchondral bone plate has a marked porosity. It is filled by channels that directly link the articular cartilage and subchondral trabecular bone, allowing crossover communications by prostaglandins, leukotrienes, various growth factors, and inflammatory parameters. A surprisingly high number of arterial and veinous vessels, as well as nerves, penetrate through these channels and send tiny branches directly into calcified cartilage. The subchondral trabecular bone exerts important shock-absorbing and supportive functions in normal joints and may also be important for cartilage nutrient supply and metabolism [45].
The subchondral bone plays a vital role in the pathogenesis of OA. Strong evidence associates subchondral bone alterations with cartilage damage and loss in OA. Subchondral sclerosis is widely considered to be a prominent feature of late-stage OA. At the same time, early-stage OA is characterized by a thinning subchondral plate with increased porosity and deteriorated subchondral trabeculae with decreased bone density [45]. Thus, bone-altering diseases such as osteoporosis could have consequences in the cartilage. For many years, a possible link between osteoporosis and osteoarthritis has been discussed. They are indeed two entities related to age and genetic and environmental factors, including estrogen deficiency [46,47]. They both involve similar cell populations: osteoblasts and chondrocytes, derived from a common progenitor, the mesenchymal stem cell [48,49,50].
It is now widely recognized that the pituitary gland-bone axis plays an essential role in the endocrine regulation of the skeleton. In particular, different groups elucidated the role of pituitary hormones, such as the FSH, GH, prolactin, and OT in the regulation of bone homeostasis [51,52,53,54]. In addition, experiments performed in mice haploinsufficient for pituitary hormones showed decreased hormone levels on their receptors, seriously affecting the skeleton. However, the primary target organ could remain intact, indicating that the bone is more sensitive to the control of pituitary hormones [51,55,56,57]. Indeed, in a study in lactating mice, the bone was more sensitive to the action of OT than the breast because haploinsufficient OT+/− or OTR+/− mice showed profound osteopenia [58]. Consequently, mice deficient in OT or OTR showed profoundly impaired bone formation.
Given that calcium is mobilized from the maternal skeleton during late pregnancy and lactation, the hypothesis that the hormone regulating these functions might also control skeletal homeostasis led to the discovery of a direct anabolic action of OT on the skeleton [51]. A hypothesis on the central action of OT has already been discussed, and one study performed intra-cerebro-ventricular OT injections in mice to demonstrate improvement in bone anabolism. The failure of this study to prove the above hypothesis suggests that the action of OT on the skeleton is concentrated on the more peripheral regions [51]. This result could also be explained by the location of the receptors in the peripheral tissues. It has been shown that OT acts on the subchondral bone, an important structure involved in the pathophysiology of osteoarthritis [58]. Changes in the subchondral bone and the actions of osteoblasts during OA are also well-known [59].
Indeed, studies have shown that OT can promote osteoblastogenesis in human adipocyte and bone marrow mesenchymal (hMADS and hBMS) cells [29,60,61]. In addition, osteoblasts and osteoclasts express OT receptors (OTRs), the stimulation of which increases bone mass [29,30,31]. Indeed, in a study on rats published by our team [30], we have shown that intraperitoneal injection of OT could decrease serum calcium levels and NF-κB expression and increase osteoprotegerin levels and an increase in bone remodeling. Another study also showed that subcutaneous injection of OT in ovariectomized mice could play a role in bone microarchitecture, strengthening it through the control of osteoblast-adipocyte balance. In the same study, a significant decrease in OT levels was observed in osteoporotic postmenopausal women than non-osteoporotic postmenopausal women, proving the strong link between OT and bone microarchitecture [29]. These different elements, showing the action of OT on bone, reinforce the idea of the potential role of this hormone in osteoarthritis, given the important synergies between subchondral bone and cartilage.

3. The Muscle

Another joint structure that plays a role in OA is the muscle. The progressive loss of periarticular muscle mass and function has consequences on joint stability and health. Muscle wasting is inevitably associated with aging, and notable weakness in the quadriceps muscle was observed early in the disease process of OA and may even precede disease onset [62,63]. Alterations in the cellular and molecular properties of the quadriceps muscle regarding myofiber atrophy, reduction in muscle quality, and defective muscle regeneration in adults with moderate knee OA have been reported [64]. It seems that patients with OA had fewer Type I fibers and more hybrid IIa/x fibers compared to healthy controls.
Myoblasts express OTRs. Elabd et al., showed in their study on mice that OT, a key age-specific systemic regulator, supports the productive repair and maintenance of skeletal muscle and acts directly on muscle stem cells in vitro and in vivo. The pro-myogenic effect of OT was mediated by MAPK/ERK signaling [60], a pathway implicated in the pathophysiology of OA. A deficiency in skeletal muscle regeneration observed in aged mice was associated with increased fibrotic tissue formation [65,66,67]. Elabd et al., also showed that the fibrotic index was higher in older mice than in younger mice. Ectopic OT treatment significantly decreased the fibrotic index in older mice, whereas administration of OT antagonists increased fibrosis in young mice. Moreover, subcutaneous injection of OT improved muscle regeneration in old mice to a level comparable to that in young mice [60]. These data suggest that the age-related decline in OT is associated with a reduced regeneration of muscle fibers, paving the way for an interest in OT treatment to counteract muscle degradation.
Further intracellular signaling studies in human and mouse myoblasts associated the observations in the OT-mediated promotion of myogenic cell proliferation with extracellular signal-regulated kinase 1/2 phosphorylation in the MAPK/extracellular signal-related kinase pathway [68]. The binding of OT and OTR-selective agonists (Thr, Gly) to the OTRs in human myoblast cultures was also shown to promote myoblast fusion [68], substantiating prior observations of OT-mediated myogenic differentiation in murine L6-C5 skeletal muscle cells [69].

4. The Metabolic Syndrome and Adipocytes

Recent evidence indicates that OT enhances glucose uptake and lipid utilization in adipose tissue and skeletal muscle, suggesting that a dysfunction in the OT system could underlie insulin resistance and dyslipidemia pathogenesis. The relationship between OA and metabolic syndrome has also been extensively studied. Metabolic syndrome refers to a cluster of metabolic abnormalities, including obesity, insulin resistance, dyslipidemia, hyperglycemia, and hypertension. OA and diabetes frequently co-exist in patients by chance due to their high prevalence and shared risk factors, such as obesity and aging. Prolonged hyperglycemia, both in fasting and post-prandial states, leads to the production of advanced glycated end products (AGEs), oxidative stress, and low-grade inflammation, eventually resulting in damage to the vasculature, mainly in the heart, kidneys, eyes, nerves, and other tissues [70]. Meta-analyses confirmed an association between osteoarthritis and cardiovascular diseases [71]. To explain the link between OA and metabolic syndrome, in vivo models of obesity, diabetes, and dyslipidemia provided evidence of a systemic effect on joints associated with low-grade inflammation and oxidative stress. Obesity-associated inflammation is associated with osteoarthritis severity and may modulate osteoarthritis progression in mouse models. In addition, osteoarthritis synovium from patients with type 2 diabetes showed insulin-resistant features, suggesting that type 2 diabetes may participate in joint catabolism.
Despite evidence of the mechanical impact of being overweight/obese in elucidating lower limb OA in obese subjects with diabetes, their association in hand OA supports the role of diabetes in OA pathogenesis through two major pathways: oxidative stress resulting from chronic hyperglycemia, leading to the overproduction of pro-inflammatory cytokines and AGEs in joint tissue; insulin resistance, which may impact both local and systemic low-grade chronic inflammation. Dysglycemia, including diabetes, is only one factor of metabolic syndrome. Whether other components of metabolic syndrome, including high blood pressure and atherogenic dyslipidemia, together or independently affect OA pathophysiology remains to be explored [72].
Despite conflicting data in humans, metabolic syndrome—the clustering of cardiometabolic risk factors, including central obesity, insulin resistance, dyslipidemia, and hypertension—has been associated with reduced fasting serum OT levels, as reported in large-scale mixed-gender studies, such as that performed by Yuan et al. [73]. In addition, fasting serum OT concentration has been negatively associated with body mass index (BMI), waist circumference, levels of glycosylated hemoglobin (HbA1c), fasting glucose and post-prandial glucose, fasting and post-prandial insulin, TG, the homeostasis model assessment of insulin resistance (HOMA-IR) score, and C-reactive protein, as reported in a study of subjects with normal body weight and those with overweight and/or obesity [73,74].
The relationship between adipose tissue and OA has also been widely studied [75]. Among systemic adipose tissues, subcutaneous adipose tissue is significantly negatively associated with muscle mass and forces and could be related to the presence and progression of OA in the knee. The amount of visceral adipose tissue is associated with an increased cartilage loss and production of pro-inflammatory cytokines, both of which seem to play a role in the pathogenesis of knee OA [75]. Knee local adipose tissue, such as the infrapatellar fat pad, can interact with neighboring tissues and may have a biphasic effect in knee OA [76]. The underlying mechanisms for the role of systemic and local fat in knee OA could be related to biomechanical, metabolic, and inflammatory factors and fat fibrosis, which may have a separate or combined effect in OA.
OT has previously been shown to inhibit adipocyte differentiation and stimulate osteoblast differentiation [77]. Adipose tissue synthesizes and releases adipokines that modulate bone metabolism by directly or indirectly regulating bone formation and resorption. Adipokines, including leptin, visfatin, adiponectin, and resistin, have been demonstrated to have metabolic implications in the pathogenesis and progression of obesity-induced OA by modulating pro- and anti-inflammatory and anabolic/catabolic balance, apoptosis, matrix remodeling, and subchondral bone ossification [78]. Adipokines appear to play an important role in the pathogenesis of metabolic syndromes. Leptin is an adipokine that exerts its action by activating OB-Rb long-form isoform receptors. It primarily regulates food intake, body weight, and energy homeostasis through neuroendocrine functions and influences insulin sensitivity and lipid metabolism. Subjects with metabolic syndrome had higher leptin levels than those without metabolic syndrome [79].
Dong et al., investigated the differential expression of adipokines in patients with knee OA. They found that leptin expression was higher in patients with knee OA and metabolic syndrome than in those with knee OA without metabolic syndrome [80]. These data correlated with the results of the study of Pelletier et al., who demonstrated that serum levels of leptin predict higher cartilage volume loss in the lateral and medial compartments over time [81]. However, several studies have highlighted the difficulty in interpreting this relationship, given the close links between leptin and certain confounding factors such as BMI, age, and sex. Thus, some authors have found an association between leptin levels and the severity of knee OA or pain, but this was not found after adjustment for potential confounding factors [82,83].
Adipocytes also express OT receptors and signaling through these receptors induces lipolysis [84]. OT levels have been shown to be significantly negatively correlated with leptin levels, and to overcome leptin resistance as it decreases body weight and fat mass with an improvement in glucose metabolism [85]. OT administration has also been shown to successfully treat obesity in animal models of leptin deficiency or reduced leptin receptor signaling [86,87]. Two weeks of subcutaneous OT treatment on obese mice resulted in a reduction in body fat, which translated into a reduction in body weight. This effect was mainly observed during the first week of treatment. However, the effect was more prolonged in db/db mice, wherein the weight loss associated with the loss of fat mass was observed over 12 weeks, with the absence of loss of appetite, which could have been observed in ob/ob mice. A better glucose tolerance and insulin sensitivity was found in db/db mice compared to ob/ob mice [88,89]. However, this phenomenon has conflicting results, especially in some ex vivo or in vivo studies.

5. OT as a Potential Therapeutic Treatment

In view of these differences, the therapeutic role of OT in human clinical trials is all the more relevant. In a previous study, we showed that the rate of OT was significantly lower in patients with OA than in patients without OA [43].
In this review, we show the potential relationship between OT and osteoarthritis. The presence of OTR in tissues plays an important role in the physiopathology of OA. The potential beneficial action of OT on the cartilage, subchondral bone, muscle, and inflammation, and its link with other risk factors, including metabolic syndrome, are different points of discussion when looking at the applications of OT treatment in OA. Obesity is a major risk factor for OA, and one of our recommendations for the management of OA is weight loss. The anorexigenic effect of OT has been extensively documented in animals [88,90,91,92,93,94]. Systemic administration of OT has been shown to affect appetite, weight gain, glucose homeostasis, and lipid metabolism in animal models [84,85,86,87,88].
In a previous study, we showed that OT administration in rats limited weight gain compared to the controls [43]. However, definitive studies on the anorexigenic effects of OT in humans still lack due to contradictory results. In a pilot study, Zhang et al. [94] showed that the four times daily administration of intranasal OT (24 IU) in nine Asian men and women with obesity (BMI ≥ 28 kg/m2) for eight weeks led to a mean BMI reduction of 3.2 ± 1.9 kg/m2. The magnitude of weight loss was also greater in subjects with a higher degree of obesity. This pilot study raised more questions than answers.
Most of the available data on the use of OT for OA came from animal studies. In humans, the data available still cannot convincingly suggest the use of OT for OA. However, while OT injections produced an increase in non-esterified fatty acid (NEFA) levels a few days after delivery [95], it seems that administration of 5 IU of OT via quick infusion resulted in decreased NEFA levels in non-pregnant healthy subjects [96]. A more recent study using chronic treatment of obese humans with nasal OT delivery did not significantly modify triglyceride (TG) levels, although there was a trend toward a decrease in this parameter [92]. This pilot study [94] should be analyzed with caution because of the small number of subjects (eleven placebo-treated vs. nine treated with OT), despite randomization. As this is a study presenting results in both animals and humans, the results in mice showed an interesting effect on carbohydrate metabolism and a reversal of insulin resistance. This is very interesting because obesity is often associated with type 2 diabetes. Authors found that while OT treatment did not affect fasting blood glucose or insulin levels, it tended to reduce post-prandial glucose and insulin levels towards the normal ranges. Modifications independent of weight variations suggested that OT could employ a body-weight-independent mechanism to improve glucose and insulin homeostasis. OT treatment significantly reduced serum LDL and cholesterol levels, while it increased serum HDL levels (ref). The effect of 4-week treatment with OT in humans resulted in a body weight reduction of 4.6 ± 3.2 kg. This therapeutic effect continued to improve when the duration of OT treatment increased to 8 weeks, showing that compared to the baseline levels, the body weight of patients dropped 8.9 ± 5.4 kg (p < 0.001) [94].
The advantages of using OT in treating obesity can also be further increased, given that OT has social-neuropsychiatric benefits that can aid in controlling metabolic disease and promoting physical activity. OA is one of the most frequently occurring, painful conditions humans can experience. Pain is a major symptom of OA, involving both peripheral and central neurological mechanisms. OA pain is initiated from free axonal endings in the synovium, periosteum bone, and tendons but not in the cartilage.
Nociceptive messages involve neuromediators and regulating factors, such as neuronal growth factor (NGF) and central modifications of pain pathways [97]. In an animal model, OT release suppresses nociception and induces analgesia by affecting inflammatory pain pathways [98]. OT modulates pain by reaching the spinal cord through fast neuronal projections and slow peripheral pathways. Nersessian et al., showed that OT directly targets the pain receptor TRPV1, a non-selective Ca2+-permeable cation channel [99]. The analgesic actions of OT have been documented in patients with migraine administered with intranasal OT [100]. OT-promoted pain regulation involves endogenous analgesia [101].

6. Conclusions

All these data led us to postulate the role of OT treatment in osteoarthritis despite conflicting results in the management of obesity. Many reasons may explain the discrepant results in the use of OT in obesity treatment, such as the use of different models, doses, and duration of OT treatments. These observations should help us to think about how OT can be used in OA pathology. However, because of the complexity of the physiopathology of this disease, taking into account the action of OT on multiple tissues that play other important roles in osteoarthritis (cartilage, subchondral bone, muscle, adipocytes, etc.) and its anti-inflammatory action, OT may have different effects depending on the osteoarthritis phenotype. It is certain that the metabolic phenotype seems the most attractive. An evaluation of the effect of OT on osteoarthritis pain will probably be one of the more interesting points that need to be considered, as its peripheral and central origin are favorable aspects for its effectiveness. Only clinical studies on humans will allow us to answer these questions. However, unresolved questions will have to be answered, such as the mode of administration of OT, the rate of administration, as well as the dosage to be administered. There are many hopes, with the development of new technologies, that OT can be delivered directly to the target tissues to avoid side effects as well as to circumvent the very short half lifetime of OT.

Author Contributions

S.F. and C.H.R.: writing—original draft preparation; E.-Z.A. and C.H.R.: review, editing and supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This study did not report data.

Acknowledgments

This work was supported by CHU de Nice, CNRS and Société Française de Rhumatologie.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. O’Neill, T.W.; McCabe, P.S.; McBeth, J. Update on the epidemiology, risk factors and disease outcomes of osteoarthritis. Best Pract. Res. Clin. Rheumatol. 2018, 32, 312–326. [Google Scholar] [CrossRef]
  2. Salmon, J.H.; Rat, A.C.; Sellam, J.; Michel, M.; Eschard, J.P.; Guillemin, F.; Jolly, D.; Fautrel, B. Economic impact of lower-limb osteoarthritis worldwide: A systematic review of cost-of-illness studies. Osteoarthr. Cartil. 2016, 24, 1500–1508. [Google Scholar] [CrossRef] [Green Version]
  3. Wang, M.; Shen, J.; Jin, H.; Im, H.J.; Sandy, J.; Chen, D. Recent progress in understanding molecular mechanisms of cartilage degeneration during osteoarthritis. Ann. N. Y. Acad. Sci. 2011, 1240, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Serra, R.; Johnson, M.; Filvaroff, E.H.; LaBorde, J.; Sheehan, D.M.; Derynck, R.; Moses, H.L. Expression of a truncated, kinase-defective TGF-beta type II receptor in mouse skeletal tissue promotes terminal chondrocyte differentiation and osteoarthritis. J. Cell Biol. 1997, 139, 541–552. [Google Scholar] [CrossRef] [PubMed]
  5. Yang, X.; Chen, L.; Xu, X.; Li, C.; Huang, C.; Deng, C.X. TGF-beta/Smad3 signals repress chondrocyte hypertrophic differentiation and are required for maintaining articular cartilage. J. Cell Biol. 2001, 153, 35–46. [Google Scholar] [CrossRef] [Green Version]
  6. Shen, J.; Li, J.; Wang, B.; Jin, H.; Wang, M.; Zhang, Y.; Yang, Y.; Im, H.J.; O’Keefe, R.J.; Chen, D. Deletion of the transforming growth factor beta receptor type II gene in articular chondrocytes leads to a progressive osteoarthritis-like phenotype in mice. Arthritis Rheum. 2013, 65, 3107–3119. [Google Scholar] [CrossRef] [Green Version]
  7. Wang, M.; Tang, D.; Shu, B.; Wang, B.; Jin, H.; Hao, S.; Dresser, K.A.; Shen, J.; Im, H.J.; Sampson, E.R.; et al. Conditional activation of beta-catenin signaling in mice leads to severe defects in intervertebral disc tissue. Arthritis Rheum. 2012, 64, 2611–2623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Mirando, A.J.; Liu, Z.; Moore, T.; Lang, A.; Kohn, A.; Osinski, A.M.; O’Keefe, R.J.; Mooney, R.A.; Zuscik, M.J.; Hilton, M.J. RBP-Jkappa-dependent Notch signaling is required for murine articular cartilage and joint maintenance. Arthritis Rheum. 2013, 65, 2623–2633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Lin, A.C.; Seeto, B.L.; Bartoszko, J.M.; Khoury, M.A.; Whetstone, H.; Ho, L.; Hsu, C.; Ali, S.A.; Alman, B.A. Modulating hedge hog signaling can attenuate the severity of osteoarthritis. Nat. Med. 2009, 15, 1421–1425. [Google Scholar] [CrossRef] [PubMed]
  10. Lories, R.J.; Corr, M.; Lane, N.E. To Wnt or not to Wnt: The bone and joint health dilemma. Nat. Rev. Rheumatol. 2013, 9, 328–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Sassi, N.; Laadhar, L.; Allouche, M.; Achek, A.; Kallel-Sellami, M.; Makni, S.; Sellami, S. WNT signaling and chondrocytes: From cell fate determination to osteoarthritis physiopathology. J. Recept. Signal Transduct. Res. 2014, 34, 73–80. [Google Scholar] [CrossRef] [PubMed]
  12. Little, C.B.; Barai, A.; Burkhardt, D.; Smith, S.M.; Fosang, A.J.; Werb, Z.; Shah, M.; Thompson, E.W. Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum. 2009, 60, 3723–3733. [Google Scholar] [CrossRef] [PubMed]
  13. Glasson, S.S.; Askew, R.; Sheppard, B.; Carito, B.; Blanchet, T.; Ma, H.L.; Flannery, C.R.; Peluso, D.; Kanki, K.; Yang, Z.; et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 2005, 434, 644–648. [Google Scholar] [CrossRef] [PubMed]
  14. Kobayashi, H.; Hirata, M.; Saito, T.; Itoh, S.; Chung, U.I.; Kawaguchi, H. Transcriptional induction of ADAMTS5 protein by nuclear factor-κB (NF-κB) family member RelA/p65 in chondrocytes during osteoarthritis development. J. Biol. Chem. 2013, 288, 28620–28629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Stanton, H.; Rogerson, F.M.; East, C.J.; Golub, S.B.; Lawlor, K.E.; Meeker, C.T.; Little, C.B.; Last, K.; Farmer, P.J.; Campbell, I.K.; et al. ADAMTS5 is the major aggrecanase in mouse cartilage in vivo and in vitro. Nature 2005, 434, 648–652. [Google Scholar] [CrossRef]
  16. Goldring, M.B.; Otero, M.; Plumb, D.A.; Dragomir, C.; Favero, M.; El Hachem, K.; Hashimoto, K.; Roach, H.I.; Olivotto, E.; Borzì, R.M.; et al. Roles of inflammatory and anabolic cytokines in cartilage metabolism: Signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur. Cell. Mater. 2011, 21, 202–220. [Google Scholar] [CrossRef]
  17. Jankowski, M.; Danalache, B.; Wang, D.; Bhat, P.; Hajjar, F.; Marcinkiewicz, M.; Paquin, J.; McCann, S.M.; Gutkowska, J. Oxytocin in cardiac ontogeny. Proc. Natl. Acad. Sci. USA 2004, 101, 13074–13079. [Google Scholar] [CrossRef] [Green Version]
  18. Zingg, H.H.; Laporte, S.A. The oxytocin receptor. Trends Endocrinol. Metab. 2003, 14, 222–227. [Google Scholar] [CrossRef]
  19. Thibonnier, M.; Conarty, D.M.; Preston, J.A.; Plesnicher, C.L.; Dweik, R.A.; Erzurum, S.C. Human vascular endothelial cells express oxytocin receptors. Endocrinology 1999, 140, 1301–1309. [Google Scholar] [CrossRef]
  20. Gutkowska, J.; Jankowski, M.; Lambert, C.; Mukaddam-Daher, S.; Zingg, H.H.; McCann, S.M. Oxytocin releases atrial natriuretic peptide by combining with oxytocin receptors in the heart. Proc. Natl. Acad. Sci. USA 1997, 94, 11704–11709. [Google Scholar] [CrossRef] [Green Version]
  21. Russell, J.A.; Leng, G.; Douglas, A.J. The magnocellular oxytocin system, the fount of maternity: Adaptations in pregnancy. Front. Neuroendocrinol. 2003, 24, 27–61. [Google Scholar] [CrossRef]
  22. Neumann, I.D. Oxytocin: The neuropeptide of love reveals some of its secrets. Cell Metab. 2007, 5, 231–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kosfeld, M.; Heinrichs, M.; Zak, P.J.; Fischbacher, U.; Fehr, E. Oxytocin increases trust in humans. Nature 2005, 435, 673–676. [Google Scholar] [CrossRef] [Green Version]
  24. Kirsch, P.; Esslinger, C.; Chen, Q.; Mier, D.; Lis, S.; Siddhanti, S.; Gruppe, H.; Mattay, V.S.; Gallhofer, B.; Meyer-Lindenberg, A. Oxytocin modulates neural circuitry for social cognition and fear in humans. J. Neurosci. 2005, 25, 11489–11493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Andari, E.; Duhamel, J.R.; Zalla, T.; Herbrecht, E.; Leboyer, M.; Sirigu, A. Promoting social behavior with oxytocin in high-functioning autism spectrum disorders. Proc. Natl. Acad. Sci. USA 2010, 107, 4389–4394. [Google Scholar] [CrossRef] [Green Version]
  26. Assinder, S.J.; Carey, M.; Parkinson, T.; Nicholson, H.D. Oxytocin and vasopressin expression in the ovine testis and epididymis: Changes with the onset of Spermatogenesis. Biol. Reprod. 2000, 63, 448–456. [Google Scholar] [CrossRef] [PubMed]
  27. Péqueux, C.; Breton, C.; Hagelstein, M.T.; Geenen, V.; Legros, J.J. Oxytocin receptor pattern of expression in primary lung cancer and in normal human lung. Lung Cancer 2005, 50, 177–188. [Google Scholar] [CrossRef] [Green Version]
  28. Wu, Y.; Wu, T.; Xu, B.; Xu, X.; Chen, H.; Li, X. Oxytocin prevents cartilage matrix destruction via regulating matrix metalloproteinases. Biochem. Biophys. Res. Commun. 2017, 486, 601–606. [Google Scholar] [CrossRef] [PubMed]
  29. Elabd, C.; Basillais, A.; Beaupied, H.; Breuil, V.; Wagner, N.; Scheideler, M.; Zaragosi, L.E.; Massiéra, F.; Lemichez, E.; Trajanoski, Z.; et al. Oxytocin controls differentiation of human mesenchymal stem cells and reverses osteoporosis. Stem Cells 2008, 26, 2399–2407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Roux, C.; Pisani, D.F.; Yahia, H.B.; Djedaini, M.; Beranger, G.E.; Chambard, J.C.; Ambrosetti, D.; Michiels, J.F.; Breuil, V.; Ailhaud, G.; et al. Chondrogenic potential of stem cells derived from adipose tissue: A powerful pharmacological tool. Biochem. Biophys. Res. Commun. 2013, 440, 786–791. [Google Scholar] [CrossRef] [PubMed]
  31. Vaidyanathan, R.; Hammock, E.A.D. Oxytocin receptor dynamics in the brain across development and species. Dev. Neurobiol. 2017, 77, 143–157. [Google Scholar] [CrossRef] [PubMed]
  32. Szeto, A.; Nation, D.A.; Mendez, A.J.; Dominguez-Bendala, J.; Brooks, L.G.; Schneiderman, N.; McCabe, P.M. Oxytocin attenuates NADPH-dependent superoxide activity and IL-6 secretion in macrophages and vascular cells. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E1495–E1501. [Google Scholar] [CrossRef] [Green Version]
  33. Yuan, L.; Liu, S.; Bai, X.; Gao, Y.; Liu, G.; Wang, X.; Liu, D.; Li, T.; Hao, A.; Wang, Z. Oxytocin inhibits lipopolysaccharide-induced inflammation in microglial cells and attenuates microglial activation in lipopolysaccharide-treated mice. J. Neuroinflamm. 2016, 13, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592. [Google Scholar] [CrossRef] [PubMed]
  35. Tetlow, L.C.; Adlam, D.J.; Woolley, D.E. Matrix metalloproteinase and pro-inflammatory cytokine production by chondrocytes of human osteoarthritic cartilage: Associations with degenerative changes. Arthritis Rheum. 2001, 44, 585–594. [Google Scholar] [CrossRef]
  36. Freemont, A.J.; Hampson, V.; Tilman, R.; Goupille, P.; Taiwo, Y.; Hoyland, J.A. Gene expression of matrix metalloproteinases 1, 3, and 9 by chondrocytes in osteoarthritic human knee articular cartilage is zone and grade specific. Ann. Rheum. Dis. 1997, 56, 542–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Mehraban, F.; Kuo, S.Y.; Riera, H.; Chang, C.; Moskowitz, R.W. Prostromelysin and procollagenase genes are differentially up-regulated in chondrocytes from the knees of rabbits with experimental osteoarthritis. Arthritis Rheum. 1994, 37, 1189–1197. [Google Scholar] [CrossRef] [PubMed]
  38. Yan, C.; Boyd, D.D. Regulation of matrix metalloproteinase gene expression. J. Cell. Physiol. 2007, 211, 19–26. [Google Scholar] [CrossRef]
  39. Chakraborti, S.; Mandal, M.; Das, S.; Mandal, A.; Chakraborti, T. Regulation of matrix metalloproteinases: An overview. Mol. Cell. Biochem. 2003, 253, 269–285. [Google Scholar] [CrossRef] [PubMed]
  40. Gaire, M.; Magbanua, Z.; McDonnell, S.; McNeil, L.; Lovett, D.H.; Matrisian, L.M. Structure and expression of the human gene for the matrix metalloproteinase matrilysin. J. Biol. Chem. 1994, 269, 2032–2040. [Google Scholar] [CrossRef]
  41. Saez, E.; Rutberg, S.E.; Mueller, E.; Oppenheim, H.; Smoluk, J.; Yuspa, S.H.; Spiegelman, B.M. c-fos is required for malignant progression of skin tumors. Cell 1995, 82, 721–732. [Google Scholar] [CrossRef] [Green Version]
  42. Rimoldi, V.; Reversi, A.; Taverna, E.; Rosa, P.; Francolini, M.; Cassoni, P.; Parenti, M.; Chini, B. Oxytocin receptor elicits different EGFR/MAPK activation patterns depending on its localization in caveolin-1 enriched domains. Oncogene 2003, 22, 6054–6060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Roux, C.H.; Pisani, D.F.; Gillet, P.; Fontas, E.; Yahia, H.B.; Djedaini, M.; Ambrosetti, D.; Michiels, J.F.; Panaia-Ferrari, P.; Breuil, V.; et al. Oxytocin Controls Chondrogenesis and Correlates with Osteoarthritis. Int. J. Mol. Sci. 2020, 21, 3966. [Google Scholar] [CrossRef] [PubMed]
  44. Goldring, M.B.; Goldring, S.R. Articular cartilage and subchondral bone in the pathogenesis of osteoarthritis. Ann. N. Y. Acad. Sci. 2010, 1192, 230–237. [Google Scholar] [CrossRef] [PubMed]
  45. Li, G.; Yin, J.; Gao, J.; Cheng, T.S.; Pavlos, N.J.; Zhang, C.; Zheng, M.H. Subchondral bone in osteoarthritis: Insight into risk factors and microstructural changes. Arthritis Res. Ther. 2013, 15, 223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Armas, L.A.; Recker, R.R. Pathophysiology of osteoporosis: New mechanistic insights. Endocrinol. Metab. Clin. N. Am. 2012, 41, 475–486. [Google Scholar] [CrossRef]
  47. Sniekers, Y.H.; Weinans, H.; van Osch, G.J.; van Leeuwen, J.P. Oestrogen is important for maintenance of cartilage and subchondral bone in a murine model of knee osteoarthritis. Arthritis Res. Ther. 2010, 12, R182. [Google Scholar] [CrossRef] [Green Version]
  48. Zhao, L.J.; Jiang, H.; Papasian, C.J.; Maulik, D.; Drees, B.; Hamilton, J.; Deng, H.W. Correlation of obesity and osteoporosis: Effect of fat mass on the determination of osteoporosis. J. Bone Miner. Res. 2008, 23, 17–29. [Google Scholar] [CrossRef] [Green Version]
  49. Reid, I.R. Relationships between fat and bone. Osteoporos. Int. 2008, 19, 595–606. [Google Scholar] [CrossRef]
  50. Rosen, C.J.; Bouxsein, M.L. Mechanisms of Disease: Is osteoporosis the obesity of bone? Nat. Clin. Pract. Rheumatol. 2006, 2, 35–43. [Google Scholar] [CrossRef]
  51. Tamma, R.; Colaianni, G.; Zhu, L.L.; DiBenedetto, A.; Greco, G.; Montemurro, G.; Patano, N.; Strippoli, M.; Vergari, R.; Mancini, L.; et al. Oxytocin is an anabolicbone hormone. Proc. Natl. Acad. Sci. USA 2009, 106, 7149–7154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Sun, L.; Peng, Y.; Sharrow, A.C.; Iqbal, J.; Zhang, Z.; Papachristou, D.J.; Zaidi, S.; Zhu, L.L.; Yaroslavskiy, B.B.; Zhou, H.; et al. FSH directly regulates bone mass. Cell 2006, 125, 247–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Seriwatanachai, D.; Thongchote, K.; Charoenphandhu, N.; Pandaranandaka, J.; Tudpor, K.; Teerapornpuntakit, J.; Suthiphongchai, T.; Krishnamra, N. Prolactin directly enhances bone turnover by raising osteoblast-expressed receptor activator of nuclear factor kappaB ligand/osteoprotegerin ratio. Bone 2008, 42, 535–546. [Google Scholar] [CrossRef] [PubMed]
  54. Menagh, P.J.; Turner, R.T.; Jump, D.B.; Wong, C.P.; Lowry, M.B.; Yakar, S.; Rosen, C.J.; Iwaniec, U.T. Growth hormone regulates the balance between bone formation and bone marrow adiposity. J. Bone Miner. Res. 2010, 25, 757–768. [Google Scholar] [CrossRef] [PubMed]
  55. Colaianni, G.; Cuscito, C.; Colucci, S. FSH and TSH in the regulation of bone mass: The pituitary/immune/bone axis. Clin. Dev. Immunol. 2013, 2013, 382698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Colaianni, G.; Tamma, R.; Di Benedetto, A.; Yuen, T.; Sun, L.; Zaidi, M.; Zallone, A. The oxytocin-bone axis. J. Neuroendocrinol. 2014, 26, 53–57. [Google Scholar] [CrossRef] [Green Version]
  57. Tamma, R.; Sun, L.; Cuscito, C.; Lu, P.; Corcelli, M.; Li, J.; Colaianni, G.; Moonga, S.S.; Di Benedetto, A.; Grano, M.; et al. Regulation of bone remodeling by vasopressin explains the bone loss in hyponatremia. Proc. Natl. Acad. Sci. USA 2013, 110, 18644–18649. [Google Scholar] [CrossRef] [Green Version]
  58. Colaianni, G.; Sun, L.; Zaidi, M.; Zallone, A. Oxytocin and bone. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R970–R977. [Google Scholar] [CrossRef]
  59. Martel-Pelletier, J.; Boileau, C.; Pelletier, J.P.; Roughley, P.J. Cartilage in normal and osteoarthritis conditions. Best Pract. Res. Clin. Rheumatol. 2008, 22, 351–384. [Google Scholar] [CrossRef]
  60. Elabd, C.; Cousin, W.; Upadhyayula, P.; Chen, R.Y.; Chooljian, M.S.; Li, J.; Kung, S.; Jiang, K.P.; Conboy, I.M. Oxytocin is an age-specific circulating hormone that is necessary for muscle maintenance and regeneration. Nat. Commun. 2014, 5, 4082. [Google Scholar] [CrossRef] [Green Version]
  61. Beranger, G.E.; Djedaini, M.; Battaglia, S.; Roux, C.H.; Scheideler, M.; Heymann, D.; Amri, E.Z.; Pisani, D.F. Oxytocin reverses osteoporosis in a sex-dependent manner. Front. Endocrinol. 2015, 6, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Slemenda, C.; Brandt, K.D.; Heilman, D.K.; Mazzuca, S.; Braunstein, E.M.; Katz, B.P.; Wolinsky, F.D. Quadriceps weakness and osteoarthritis of the knee. Ann. Intern. Med. 1997, 127, 97–104. [Google Scholar] [CrossRef]
  63. Bennell, K.L.; Hunt, M.A.; Wrigley, T.V.; Lim, B.W.; Hinman, R.S. Role of muscle in the genesis and management of knee osteoarthritis. Rheum. Dis. Clin. North Am. 2008, 34, 731–754. [Google Scholar] [CrossRef] [PubMed]
  64. Noehren, B.; Kosmac, K.; Walton, R.G.; Murach, K.A.; Lyles, M.F.; Loeser, R.F.; Peterson, C.A.; Messier, S.P. Alterations in quadriceps muscle cellular and molecular properties in adults with moderate knee osteoarthritis. Osteoarthr. Cartil. 2018, 26, 1359–1368. [Google Scholar] [CrossRef] [Green Version]
  65. Brack, A.S.; Conboy, M.J.; Roy, S.; Lee, M.; Kuo, C.J.; Keller, C.; Rando, T.A. Increased Wnt signaling during aging alters muscle stem cell fate and increases fibrosis. Science 2007, 317, 807–810. [Google Scholar] [CrossRef] [PubMed]
  66. Goldspink, G.; Fernandes, K.; Williams, P.E.; Wells, D.J. Age-related changes in collagen expression in the muscles of mdx dystrophic and normal mice. Neuromuscul. Disord. 1994, 4, 183–191. [Google Scholar] [CrossRef]
  67. Liu, Y.; Conboy, I. Unexpected evolutionarily conserved rapid effects of viral infection on oxytocin receptor and TGF-β /pSmad3. Skelet. Muscle 2017, 7, 7. [Google Scholar] [CrossRef] [Green Version]
  68. Breton, C.; Haenggeli, C.; Barberis, C.; Heitz, F.; Bader, C.R.; Bernheim, L.; Tribollet, E. Presence of functional oxytocin receptors in cultured human myoblasts. J. Clin. Endocrinol. Metab. 2002, 87, 1415–1418. [Google Scholar] [CrossRef]
  69. Nervi, C.; Benedetti, L.; Minasi, A.; Molinaro, M.; Adamo, S. Arginine-vasopressin induces differentiation of skeletal myogenic cells and up-regulation of myogenin and Myf-5. Cell Growth Differ. 1995, 6, 81–89. [Google Scholar]
  70. King, K.B.; Rosenthal, A.K. The adverse effects of diabetes on osteoarthritis: Update on clinical evidence and molecular mechanisms. Osteoarthr. Cartil. 2015, 23, 841–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Louati, K.; Vidal, C.; Berenbaum, F.; Sellam, J. Association between diabetes mellitus and osteoarthritis: Systematic literature review and meta-analysis. RMD Open 2015, 1, e000077. [Google Scholar] [CrossRef]
  72. Veronese, N.; Cooper, C.; Reginster, J.Y.; Hochberg, M.; Branco, J.; Bruyère, O.; Chapurlat, R.; Al-Daghri, N.; Dennison, E.; Herrero-Beaumont, G.; et al. Type 2 diabetes mellitus and osteoarthritis. Semin. Arthritis Rheum. 2019, 49, 9–19. [Google Scholar] [CrossRef]
  73. Yuan, G.; Qian, W.; Pan, R.; Jia, J.; Jiang, D.; Yang, Q.; Wang, S.; Liu, Y.; Yu, S.; Hu, H.; et al. Reduced circulating oxytocin and high-molecular-weight adiponectin are risk factors for metabolic syndrome. Endocr. J. 2016, 63, 655–662. [Google Scholar] [CrossRef] [Green Version]
  74. Qian, W.; Zhu, T.; Tang, B.; Yu, S.; Hu, H.; Sun, W.; Pan, R.; Wang, J.; Wang, D.; Yang, L.; et al. Decreased circulating levels of oxytocin in obesity and newly diagnosed type 2 diabetic patients. J. Clin. Endocrinol. Metab. 2014, 99, 4683–4689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chang, J.; Liao, Z.; Lu, M.; Meng, T.; Han, W.; Ding, C. Systemic and local adipose tissue in knee osteoarthritis. Osteoarthr. Cartil. 2018, 26, 864–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Eymard, F.; Pigenet, A.; Citadelle, D.; Tordjman, J.; Foucher, L.; Rose, C.; Flouzat Lachaniette, C.H.; Rouault, C.; Clément, K.; Berenbaum, F.; et al. Knee and hip intra-articular adipose tissues (IAATs) compared with autologous subcutaneous adipose tissue: Aspecific phenotype for a central player in osteoarthritis. Ann. Rheum. Dis. 2017, 76, 1142–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Amri, E.Z.; Pisani, D.F. Control of bone and fat mass by oxytocin. Horm. Mol. Biol. Clin. Investig. 2016, 28, 95–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Tu, C.; He, J.; Wu, B.; Wang, W.; Li, Z. An extensive review regarding the adipokines in the pathogenesis and progression of osteoarthritis. Cytokine 2019, 113, 1–12. [Google Scholar] [CrossRef] [PubMed]
  79. Ghadge, A.A.; Khaire, A.A. Leptin as a predictive marker for metabolic syndrome. Cytokine 2019, 121, 154735. [Google Scholar] [CrossRef]
  80. Dong, N.; Gao, Y.H.; Liu, B.; Zhao, C.W.; Yang, C.; Li, S.Q.; Liu, J.G.; Qi, X. Differential expression of adipokines in knee osteoarthritis patients with and without metabolic syndrome. Int. Orthop. 2018, 42, 1283–1289. [Google Scholar] [CrossRef] [PubMed]
  81. Martel-Pelletier, J.; Raynauld, J.P.; Dorais, M.; Abram, F.; Pelletier, J.P. The levels of the adipokines adipsin and leptin are associated with knee osteoarthritis progression as assessed by MRI and incidence of total knee replacement in symptomatic osteoarthritis patients: A post hoc analysis. Rheumatology 2016, 55, 680–688. [Google Scholar] [CrossRef] [Green Version]
  82. Calvet, J.; Orellana, C.; Gratacós, J.; Berenguer-Llergo, A.; Caixàs, A.; Chillarón, J.J.; Pedro-Botet, J.; García-Manrique, M.; Navarro, N.; Larrosa, M. Synovial fluid adipokines are associated with clinical severity in knee osteoarthritis: A cross-sectional study in female patients with joint effusion. Arthritis Res. Ther. 2016, 18, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Calvet, J.; Orellana, C.; Albiñana Giménez, N.; Berenguer-Llergo, A.; Caixàs, A.; García-Manrique, M.; Galisteo Lencastre, C.; Navarro, N.; Larrosa, M.; Gratacós, J. Differential involvement of synovial adipokines in pain and physical function in female patients with knee osteoarthritis. A cross-sectional study. Osteoarthr. Cartil. 2018, 26, 276–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Yi, K.J.; So, K.H.; Hata, Y.; Suzuki, Y.; Kato, D.; Watanabe, K.; Aso, H.; Kasahara, Y.; Nishimori, K.; Chen, C.; et al. The regulation of oxytocin receptor gene expression during adipogenesis. J. Neuroendocrinol. 2015, 27, 335–342. [Google Scholar] [CrossRef] [PubMed]
  85. Chang, H.H.; Chang, W.H.; Chi, M.H.; Peng, Y.C.; Huang, C.C.; Yang, Y.K.; Chen, P.S. The OXTR polymorphism stratified the correlation of oxytocin and glucose homeostasis in non-diabetic subjects. Diabetes Metab. Syndr. Obes. 2019, 12, 2707–2713. [Google Scholar] [CrossRef] [Green Version]
  86. Maejima, Y.; Sedbazar, U.; Suyama, S.; Kohno, D.; Onaka, T.; Takano, E.; Yoshida, N.; Koike, M.; Uchiyama, Y.; Fujiwara, K.; et al. Nesfatin-1-regulated oxytocin ergic signaling in the paraventricular nucleus causes anorexia through a leptin-independent melanocortin pathway. Cell Metab. 2009, 10, 355–365. [Google Scholar] [CrossRef] [Green Version]
  87. Iwasaki, Y.; Maejima, Y.; Suyama, S.; Yoshida, M.; Arai, T.; Katsurada, K.; Kumari, P.; Nakabayashi, H.; Kakei, M.; Yada, T. Peripheral oxytocin activates vagal afferent neurons to suppress feeding in normal and leptin-resistant mice: A route for ameliorating hyperphagia and obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 308, R360–R369. [Google Scholar] [CrossRef] [Green Version]
  88. Altirriba, J.; Poher, A.L.; Caillon, A.; Arsenijevic, D.; Veyrat-Durebex, C.; Lyautey, J.; Dulloo, A.; Rohner-Jeanrenaud, F. Divergent effects of oxytocin treatment of obese diabetic mice on adiposity and diabetes. Endocrinology 2014, 155, 4189–4201. [Google Scholar] [CrossRef] [Green Version]
  89. Plante, E.; Menaouar, A.; Danalache, B.A.; Yip, D.; Broderick, T.L.; Chiasson, J.L.; Jankowski, M.; Gutkowska, J. Oxytocin treatment prevents the cardiomyopathy observed in obese diabetic male db/dbmice. Endocrinology 2015, 156, 1416–1428. [Google Scholar] [CrossRef]
  90. Arletti, R.; Benelli, A.; Bertolini, A. Influence of oxytocin on feeding behavior in the rat. Peptides 1989, 10, 89–93. [Google Scholar] [CrossRef]
  91. Maejima, Y.; Iwasaki, Y.; Yamahara, Y.; Kodaira, M.; Sedbazar, U.; Yada, T. Peripheral oxytocin treatment ameliorates obesity by reducing food intake and visceral fat mass. Aging 2011, 3, 1169–1177. [Google Scholar] [CrossRef]
  92. Deblon, N.; Veyrat-Durebex, C.; Bourgoin, L.; Caillon, A.; Bussier, A.L.; Petrosino, S.; Piscitelli, F.; Legros, J.J.; Geenen, V.; Foti, M.; et al. Mechanisms of the anti-obesity effects of oxytocin in diet-induced obese rats. PLoS ONE 2011, 6, e25565. [Google Scholar] [CrossRef] [Green Version]
  93. Mohan, S.; Khan, D.; Moffett, R.C.; Irwin, N.; Flatt, P.R. Oxytocin is present in islets and plays a role in beta-cell function and survival. Peptides 2018, 100, 260–268. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, H.; Wu, C.; Chen, Q.; Chen, X.; Xu, Z.; Wu, J.; Cai, D. Treatment of obesity and diabetes using oxytocin or analogs in patients and mouse models. PLoS ONE 2013, 8, e61477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Burt, R.L.; Leake, N.H.; Dannenburg, W.N. Metabolic activity of oxytocin in the puerperium. Nature 1963, 198, 293. [Google Scholar] [CrossRef]
  96. Fabian, E.; Stork, A.; Kobilková, J.; Fabianová, J. Inhibition of lipolysis by oxytocin and vasopressin. Acta Univ. Carol. Med. Monogr. 1977, 77 Pt 1, 141–145. [Google Scholar]
  97. Perrot, S. Osteoarthritis pain. Best Pract. Res. Clin. Rheumatol. 2015, 29, 90–97. [Google Scholar] [CrossRef]
  98. Eliava, M.; Melchior, M.; Knobloch-Bollmann, H.S.; Wahis, J.; da Silva Gouveia, M.; Tang, Y.; Ciobanu, A.C.; Triana Del Rio, R.; Roth, L.C.; Althammer, F.; et al. A New Population of parvocellular oxytocin Neurons Controlling Magno cellular Neuron Activity and inflammatory Pain processing. Neuron 2016, 89, 1291–1304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Nersesyan, Y.; Demirkhanyan, L.; Cabezas-Bratesco, D.; Oakes, V.; Kusuda, R.; Dawson, T.; Sun, X.; Cao, C.; Cohen, A.M.; Chelluboina, B.; et al. Oxytocin modulates nociception as an agonist of pain-sensing TRPV1. Cell Rep. 2017, 21, 1681–1691. [Google Scholar] [CrossRef] [Green Version]
  100. Wang, Y.L.; Yuan, Y.; Yang, J.; Wang, C.H.; Pan, Y.J.; Lu, L.; Wu, Y.Q.; Wang, D.X.; Lv, L.X.; Li, R.R.; et al. The interaction between the oxytocin and pain modulation in headache patients. Neuropeptides 2013, 47, 93–97. [Google Scholar] [CrossRef] [PubMed]
  101. González-Hernández, A.; Rojas-Piloni, G.; Condés-Lara, M. Oxytocin and analgesia: Future trends. Trends Pharmacol. Sci. 2014, 35, 549–551. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ferrero, S.; Amri, E.-Z.; Roux, C.H. Relationship between Oxytocin and Osteoarthritis: Hope or Despair? Int. J. Mol. Sci. 2021, 22, 11784. https://doi.org/10.3390/ijms222111784

AMA Style

Ferrero S, Amri E-Z, Roux CH. Relationship between Oxytocin and Osteoarthritis: Hope or Despair? International Journal of Molecular Sciences. 2021; 22(21):11784. https://doi.org/10.3390/ijms222111784

Chicago/Turabian Style

Ferrero, Stephanie, Ez-Zoubir Amri, and Christian Hubert Roux. 2021. "Relationship between Oxytocin and Osteoarthritis: Hope or Despair?" International Journal of Molecular Sciences 22, no. 21: 11784. https://doi.org/10.3390/ijms222111784

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop