Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Jun 26, 2023; 15(6): 502-513
Published online Jun 26, 2023. doi: 10.4252/wjsc.v15.i6.502
Adipokines regulate mesenchymal stem cell osteogenic differentiation
Zhong-Hua Xu, Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou 213200, Jiangsu Province, China
Chen-Wei Xiong, Kai-Song Miao, Zhen-Tang Yu, Jun-Jie Zhang, Chang-Lin Yu, Yong Huang, Xin-Die Zhou, Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
Chen-Wei Xiong, Kai-Song Miao, Zhen-Tang Yu, Jun-Jie Zhang, Chang-Lin Yu, Yong Huang, Xin-Die Zhou, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
Xin-Die Zhou, Department of Orthopedics, Gonghe County Hospital of Traditional Chinese Medicine, Hainan Tibetan Autonomous Prefecture 811800, Qinghai Province, China
ORCID number: Jun-Jie Zhang (0000-0003-1062-3407); Xin-Die Zhou (0000-0003-4948-0191).
Author contributions: Xu ZH and Xiong CW contributed equally to this work; Xu ZH contributed to the conceptualization, methodology, and software; Xiong CW contributed to the data curation, and writing-original draft preparation; Miao KS and Yu ZT contributed to the visualization and investigation; Zhang JJ and Yu CL contributed to the supervision, software, and validation; Huang Y contributed to the writing-reviewing and editing; Zhou XD contributed to the data collection.
Supported by the Changzhou Science & Technology Program, No. CJ20210104, CJ20220120, and CJ20210005; Qinghai Province Health System Guidance Plan Project, No. 2022-wjzdx-106; Young Talent Development Plan of Changzhou Health commission, No. CZQM2020059; and Top Talent of Changzhou “The 14th Five-Year Plan” High-Level Health Talents Training Project, No. 2022CZBJ059 and 2022CZBJ061.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xin-Die Zhou, MD, Department of Orthopedics, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, No. 29 Xinglong Lane, Tianning District, Changzhou 213000, Jiangsu Province, China. zhouxindie@njmu.edu.cn
Received: November 14, 2022
Peer-review started: November 14, 2022
First decision: February 21, 2023
Revised: February 26, 2023
Accepted: April 24, 2023
Article in press: April 24, 2023
Published online: June 26, 2023

Abstract

Mesenchymal stem cells (MSCs) can differentiate into various tissue cell types including bone, adipose, cartilage, and muscle. Among those, osteogenic differentiation of MSCs has been widely explored in many bone tissue engineering studies. Moreover, the conditions and methods of inducing osteogenic differentiation of MSCs are continuously advancing. Recently, with the gradual recognition of adipokines, the research on their involvement in different pathophysiological processes of the body is also deepening including lipid metabolism, inflammation, immune regulation, energy disorders, and bone homeostasis. At the same time, the role of adipokines in the osteogenic differentiation of MSCs has been gradually described more completely. Therefore, this paper reviewed the evidence of the role of adipokines in the osteogenic differentiation of MSCs, emphasizing bone formation and bone regeneration.

Key Words: Mesenchymal stem cells, Adipokines, Adipose tissue, Osteogenic differentiation, Osteogenesis, Bone regeneration

Core Tip: Bone tissue supports and protects the organs of the human body. There is a close relationship between the immune system and bone homeostasis. Adipose tissue is an essential accessory tissue around bone tissue, which regulates bone homeostasis through the secretion of adipocytokines. There are many types of adipokines, but only some have been studied in detail. Different adipokines affect the behavior and differentiation of mesenchymal stem cells under different local microenvironments and surrounding inflammation, thus coordinating and participating in the regulation of bone homeostasis.



INTRODUCTION

Adipose tissue is currently considered an endocrine organ[1] and comprises adipose cells, endothelial cells, fibroblasts, and immune cells[2]. Adipokines are factors secreted by adipose tissue and have multiple functions[3] involving various biological processes including immune responses, inflammation, glucose metabolism, insulin secretion, sensitivity regulation, regulation of blood pressure and myocardial contractility, blood vessel growth, and lipid metabolism[3,4]. Therefore, adipokines regulate different biological processes in different organs including the brain, liver, muscles, blood vessels, heart, and pancreas[5]. The function, characterization, molecular targets, and potential clinical disease correlation of adipokines are still unclear and the main focus of future adipokine research.

Mesenchymal stem cells (MSCs), pluripotent stem cells derived from the mesoderm, were identified by surface markers such as CD29, CD37, CD44, CD90, CD105, and CD166[6]. MSCs can be readily extracted from many tissues including bone marrow, umbilical cord, placenta, fat, liver, and skin[7]. However, the most well-studied source is bone marrow. MSCs have been shown to differentiate into mature cells of various tissues including cartilage, bone, tendon, ligament, and adipose tissue[8]. Due to its multipotential nature, MSCs have been used to treat many diseases including tumors, central nervous system disease, liver disease, graft-versus-host disease, inflammation, immune system disease, and bone regeneration[9-12]. In this review, we focus on the osteogenic differentiation of MSCs.

Bone is a rigid organ that supports and protects the other vital organs in the body. In adults, bones are renewed approximately every 7 years[13], and bone formation by osteoblasts and bone resorption by osteoclasts play a significant role. Osteoclasts originate from hematopoietic stem cell precursors, and osteoblasts originate from MSCs[14]. The dynamic balance of the two processes maintains the stability of bone metabolism, whereas the destruction of balance leads to various diseases including osteoporosis[15], osteopenia[16], and bone nonunion[17]. Osteoblasts promote the deposition of calcium salts in the bone matrix and stimulate bone remodeling and osteoblast differentiation of MSCs. It can be verified by the detection of runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), and osteopontin (OPN). Therefore, the biological characteristics of MSC osteogenic differentiation have been widely used in bone tissue engineering to treat bone defects caused by trauma, infection, and tumor surgery[18-20]. As a common progenitor of both adipocytes and osteoblasts, MSCs are in a delicate equilibrium state during differentiation, whereas adipose-inducing factors inhibit the osteogenic differentiation of MSCs. In contrast, bone-inducing factors inhibit the adipogenic differentiation of MSCs[6]. As an important active secretion of fat, the position and role of adipokine in the osteogenic differentiation of MSCs are worth further consideration. Therefore, we reviewed the role of adipokines in the osteogenic differentiation of MSCs.

CYTOKINE AND CYTOKINE-LIKE PROTEINS

Interleukin (IL) is an essential inflammatory adipokine that plays a vital role in the differentiation of MSCs in the early stage of bone reconstruction[21]. Lacey et al[22] found that low-dose IL-1β (0.001-1 ng/mL) inhibited ALP activity, reduced RUNX2 and procollagen expression, and inhibited the degree of mineralization of MSCs in mice. IL-6 is a multifunctional lymphoid factor with pro-inflammatory and anti-inflammatory effects[23]. At the same time, it can be secreted by osteoblasts to stimulate the secretion of osteoclasts and participate in bone homeostasis. IL-6 induces osteogenic differentiation in human bone marrow-derived MSCs (BMSCs) via mitogen-activated protein kinase signaling[24]. IL-10 can reduce the synthesis of pro-inflammatory cytokines and chemokines and inhibit the expression of IL-1 and tumor necrosis factor alpha (TNF-α)[25]. In mice, IL-10 inhibits the osteogenic differentiation of MSCs prior to ALP expression[26]. IL-17 cytokines act by binding to the IL-17 receptor family[27]. In the early stage of bone injury, IL-17 secretion increases, promoting the transformation of MSCs into bone progenitor cells or osteoblasts. In some cases, IL-17 can also act as an anti-osteoblast factor, leading to bone loss[28,29].

TNF-α is a pro-inflammatory cytokine that can bind to the TNF receptor superfamily and participate in the regulation of a variety of biological processes. Different doses of TNF-α showed different osteogenic differentiation activity of MSCs. Wang et al[30] showed that a high dose of TNF (50 ng/mL) could stimulate the upregulation of some osteogenic factors in MSCs, including vascular endothelial growth factor and insulin growth factor. Lacey et al[22] cultured BMSCs with different doses of TNF-α and found that low-dose TNF-α (0.1-10 ng/mL) inhibited the mineralization and activation of ALP and OPN in cultured MSCs.

Monocyte chemotactic protein 1 (MCP-1), also known as C-C motif chemotactic factor ligand 2, can influence monocyte migration and subsequent macrophage polarization[31]. Xie et al[32] showed that in the process of osteogenic differentiation, MSCs from patients with ankylosing spondylitis secreted more MCP-1 than MSCs from healthy people. Enhanced MCP-1 secretion promoted monocyte migration, increased classical macrophage polarization, and enhanced TNF-α secretion[32]. Other adipokine-related cytokines, such as progranulin and resistin, have not been reported to correlate with MSC osteogenic differentiation.

Transforming growth factor β (TGF-β) has a unique correlation with the differentiation of adult MSCs[33]. Through the precise matching of ligands, receptors, and cell signaling molecules, TGF-β is involved in the lineage transformation process of the differentiation of various stem cells such as lipids, osteoblasts, chondrogenic and myogenic cells[34]. Tang et al[35] confirmed that TGF-β1 induced the migration of MSCs to the bone resorption site of mice by activating the activin receptor-like kinase 5-Smad2/3-Smad4 pathway and restricted the further recruitment of osteoclasts but did not induce osteogenic differentiation. However, other studies have reported that TGF-β inhibits osteogenic differentiation through Wnt signaling interactions and inhibits RUNX2 through the activation of Smad3[36,37]. However, TGF-β has also been reported to promote the osteogenic differentiation of MSCs[38,39]. However, further research needs to be carried out in the future.

Chemerin is a secreted protein derived from adipocytes and liver cells involved in physiological processes including inflammation, angiogenesis, and calcium mobilization[40,41]. Epidemiological studies have reported that patients with osteoporosis have higher circulating chemerin[42], and the knockout of chemerin or its receptor CMKLR1 inhibits lipogenesis and promotes the osteogenic differentiation of MSCs[43]. Li et al[41] showed that chemerin promoted the osteogenic differentiation of C3H10T1/2 cells and MSCs through Akt/Gsk3β/β-catenin signaling. However, Akt inhibitors (MK2206) inhibited chemerin’s promotion of osteogenic differentiation and active β-catenin.

PROTEINS OF THE FIBRINOLYTIC SYSTEM

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor of the fibrinolytic system[44]. Adipose-derived PAI-1 is associated with various pathologic conditions including inflammation, diabetes, cancer, and obesity[44]. Takafuji et al[45] studied the role of PAI-1 in the osteogenic differentiation of MSCs using wild-type and PAI-1-deficient mice and found that the loss of PAI-1 significantly weakened the expression of BMSC osteogenic genes, such as bone morphogenetic protein 2 (BMP-2) and ALP.

Tissue factor, another adipokine that plays a crucial role in the clotting process[46], whose overexpression in the body can lead to multiple forms of thrombosis[47]. In a study aimed at improving coagulation activity, Rangasami et al[48] found that pluronic micelle-mediated tissue factor silencing could effectively induce the higher differentiation of MSCs in osteogenic and lipid-forming media.

COMPLEMENT AND COMPLEMENT-RELATED PROTEINS

Adipsin was the first adipocyte-secreted protein to be identified[49] and is currently named complement factor D[50]. Fat cells produce it through the activation of peroxisome proliferator-activated receptor gamma[51]. More recently, adipsin was shown to promote insulin secretion by pancreatic β cells and prevent β-cell death[52]. By activating Wnt signaling, adipsin initiates adipogenesis from BMSCs[53]. Experiments on BMSCs of adipsin knockout mice showed the increased expression of mineralized nodules and osteoblast markers including RUNX2, COL1A1, and osteocalcin compared with MSCs of normal origin[53].

Complement and complement-related proteins from adipose tissue include complement component 1q and TNF-related protein family, complement factor B, and acylating simulation protein[3,54]. However, it has not been reported whether they induce or inhibit the osteogenic differentiation of MSCs.

ADIPOKINES

Leptin, a hormone derived from adipose tissue, is involved in pathophysiological processes such as food absorption, energy metabolism, inflammation, immunity, and bone homeostasis[55-58]. Leptin binds to its leptin receptor, a marker specific to BMSCs[59]. Leptin has been shown to cross-regulate BMP-9 signaling through the JAK/STAT signaling pathway in MSCs, thereby enhancing BMP-9-induced osteogenesis[60].

Adiponectin plays a vital role in anti-inflammation, glucolipid metabolism, and insulin resistance regulation[61,62]. Wang et al[63] reported that adiponectin regulates BMSC osteogenic differentiation and osteogenesis through the Wnt/β-catenin pathway. Similar results have also been reported in other studies[64-66].

Visfatin is commonly produced by visceral adipose tissue and is also known as nicotinamide phosphoribosyltransferase (Nampt) or pre-B cell cluster enhancer. It is strongly expressed in osteogenic differentiation[67] and promotes the proliferation and mineralization activity of osteoblasts[68]. Visfatin induces the secretion of IL-6, IL-8, and MCP-1 during the osteogenic differentiation of MSCs and significantly increases matrix mineralization during osteogenic differentiation, while the expression of type I collagen is decreased[69].

Nicotinamide adenine dinucleotide (NAD) is involved in energy metabolism and protein modification[70]. Nampt has recently been identified as a novel adipokine[71]. Nampt is a rate-limiting enzyme and participates in all-around MC3T3 E1-osteogenesis prior to the cell differentiation process of NAD salvage pathways. Knocking out Nampt, or adding its specific inhibitor, Fk866, resulted in decreased intracellular NAD concentration and decreased osteogenic ability[67]. Thus, Nampt can be used as a specific marker for the osteogenic differentiation of MSCs[72].

Visceral adipose tissue-derived serine protease inhibitor (vaspin), an adipose-derived hormone, attenuates osteogenic differentiation of the preosteoblast cell line MC3T3-E1[73] and antagonizes the osteogenic differentiation of rat osteoblasts. However, the role of vaspin in the osteogenic differentiation of MSCs has not been reported[74].

BMPs, the largest component of the TGF-β ligand family, regulate multiple organogenetic pathways, fat formation, and energy metabolism[75,76]. BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and BMP-7 all strongly promote osteogenesis. Short-term addition of BMP-2 increases osteocalcin expression[77], and BMP-7 induces the increased expression of ALP, a marker of osteoblast differentiation, and accelerates calcification[78]. The absence of BMP-2 and BMP-4 results in severely impaired osteogenic function, but the limb skeleton still develops normally without BMP-4[79]. BMP-3 regulates adult bone mass by limiting the differentiation of bone progenitor cells into mature osteoblasts[80]. It is important to note that BMP-7 has been marketed and used in surgery to aid fracture healing, with no reported local or systemic adverse events[81]. The effects of BMP-5[82,83] and BMP-6[84,85] on the osteogenic differentiation of MSCs have also been reported.

Nesfatin-1 is a novel anorexia polypeptide that has a wide range of biological effects including energy metabolism, gastrointestinal function, anxiety and depression, and the regulation of cardiovascular and reproductive function[86-88]. The role of nesfatin-1 in the osteogenic differentiation of MSCs has not been reported, but it can promote the expression of osteogenic genes such as ALP and RUNX2 in newly derived rat stem cells[89]. Therefore, we speculate that Nesfatin-1 has a similar role in the osteogenic differentiation of MSCs, but this conclusion still needs to be confirmed by further studies.

Cathepsins are an important category of enzymes located within the lysosomes[90]. Cathepsins are produced by various tissues, which also include adipose tissue[91]. Cathepsin K is a crucial enzyme in the degradation of the organic bone matrix, and its expression in bone formation-related cells, including fibroblasts, osteoblasts, and MSCs, has also been confirmed[92,93]. Zhang et al[94] showed that knockout or inhibition of cathepsin K can promote the regeneration of BMSCs of jaw bone through glycolysis, thus promoting alveolar bone regeneration. Similarly, cathepsin S deficiency alters the balance between adipocyte and osteoblast differentiation, increases bone turnover, and alters bone microstructure[95].

Apelin is an endogenous ligand of the G protein-coupled apelin receptor[96]. Besides being an adipokine, apelin is also expressed in skeletal muscle, the central nervous system, the heart, and other tissues, and is involved in lipolysis, glucose metabolism, cell proliferation, and angiogenesis[97]. Exogenous addition of apelin protein or overexpression of apelin promotes postpositional MSC osteoblast differentiation by activating the Wnt/β-catenin signaling pathway[98].

Omentin-1 is the adipokine most commonly expressed in omental adipose tissue and is also abundant in plasma[99]. Omentin-1 is involved in the physiological processes of inflammation, insulin, and cardiovascular functions[99,100]. For bone effects, a study of postmenopausal women found a negative correlation between omentin-1 levels and lumbar bone density[101]. Tang et al[102] found that omentin-1 has a dose-dependent effect on the viability of MC3T3-E1 cells, which can significantly increase the expression of members of the TGF-β/Smad signaling pathway, and also significantly increase the expression levels of BMP-2, RUNX2, OPN, osteocalcin, and other proteins, thus promoting osteogenesis.

Lipocalin 2 (LCN2) is a protein involved in host defense, autoimmunity, insulin resistance, skin healing, tumor, and infection[103,104]. LCN2 disrupts osteoclast formation in bone tissue by negatively regulating the proliferation and differentiation of osteoclast precursors[105]. As a secretory bone factor, LCN2 positively affects the osteogenic differentiation and in vivo osteogenesis of MC3T3-E1[106].

Melatonin is an indoleamine that is synthesized and secreted primarily by the pineal gland in mammals but is also secreted by adipose tissue[107]. Melatonin mainly affects the circadian rhythm and sleep-wake cycle and is also involved in immune regulation and inhibition of tumor growth[108,109]. Melatonin is also involved in MSC differentiation, which is involved in developing and regenerating bone, muscle, and fat tissues. In BMSCs, melatonin enhances osteogenesis and inhibits lipogenesis. Melatonin also differentiates bone marrow progenitors from adipocytes to osteoblasts[110,111].

Gremlin-1 is a highly conserved glycoprotein, mainly distributed in the extracellular matrix, with a small amount in the endoplasmic reticulum[112]. As an adipokine, gremlin-1 plays an important role in adipose tissue homeostasis[113]. At the same time, studies have shown that gremlin-1 is a BMP protein inhibitor, which can inhibit their binding to BMP receptors on the cell membrane by binding to BMP-2, BMP-4, and BMP-7[112]. Specific overexpression of gremlin-1 in mouse bone tissue results in severe osteoporosis; however, conditional knockout of gremlin-1 increases trabecular volume and bone formation[114]. Gremlin-1 has also been shown to inhibit the viability and osteogenic differentiation of human BMSCs[115].

LIPID TRANSPORT

Apolipoprotein E (ApoE), one of the main components of plasma very low-density lipoprotein[116], regulates lipid homeostasis by regulating lipid transport between tissues and cells. ApoE4 is associated with hyperlipidemia and hypercholesterolemia, leading to coronary heart disease, stroke, and atherosclerosis[117-119]. BMP-2 can upregulate the ApoE level of the mouse mesenchymal progenitor cell line (C3H10T1/2), leading to enhanced osteogenic differentiation. At the same time, ApoE is also expressed in vitro in mouse cranial primary osteoblasts with advanced osteoblast sequences[120].

ENZYMES

Dipeptidyl peptidase 4 (DPP-4) is a protein secreted in the salivary glands, prostate, seminal vesicles, endometrium, small intestine, and decidual membrane, and has recently been identified in adipose tissue as well[121]. DPP-4 is an important drug target in type 2 diabetes and directly induces insulin resistance in adipocytes and skeletal muscle[121]. DPP-4 not only reflects but also promotes adipose tissue dysfunction. Choi et al[122] found that DPP-4, when overexpressed, could restrict the induction of osteogenic differentiation of heart artery flap-derived mesenchymal cells by the autocrine insulin-like growth factor-1 signaling pathway, but this result has not been verified on MSCs.

Tissue inhibitors of metalloproteinases (TIMPs) have four main members, TIMP-1, TIMP-2, TIMP-3, and TIMP-4, and are primarily responsible for degrading most proteins in the extracellular matrix[123,124]. TIMPs are generally considered to be inhibitors of matrix metalloproteinases (MMPs) through the action of their terminal N-domain[125]. Meanwhile, TIMPs exist in the extracellular matrix in a soluble form and preemptively bind to the extracellular matrix, thus inhibiting the effect of MMPs[126]. TIMPs can selectively inhibit different MMPs, metalloproteinase and a disintegrin and metalloproteinase with thrombospondin motifs[125,126]. Inhibition of endogenous TIMP-1 can inhibit the proliferation, metabolic activity, and osteogenic differentiation ability of MSCs by activating Wnt/β-catenin signaling[127]. However, Liang et al[128] found in the process of MSC osteogenic differentiation that TIMP-1 knockdown increased the deposition of calcium nodules, ALP activity, and the expression of osteocalcin protein by activating Wnt/β-catenin signaling. The conclusions here are contradictory and need further confirmation by other studies. Studies targeting TIMP-3 have shown that increased expression of TIMP-3 can significantly promote osteogenic differentiation of MSCs in the fracture model of diabetic rats[129].

CONCLUSION

The formation and regeneration of bone tissue usually require regulation of the local microenvironment. The balance between bone resorption and bone regeneration is essential for bone tissue regeneration. Adipokines are exogenous immune regulatory substances secreted by adipose tissue, and are widely involved in pathophysiological processes of surrounding tissues, including bone homeostasis and bone regeneration. Not all human adipokines have been identified, but the current literature has revealed that the surface adipose tissue secretes more than 600 factors or proteins involving many processes of human pathophysiology[130]. There are many types of adipokines, including cytokines[22], fibrinolysin[44], complement and related proteins[49], enzymes[121], lipid transport systems[116], endocannabinoids[131], and angiotensinogen[132] (Table 1).

Table 1 Key activities of factors released by adipose tissue.
Classification
Appellation
Mechanism
Ref.
Cytokine and cytokine-like proteinsInterleukinIL-6 induces osteogenic differentiation in human bone marrow-derived MSCs via MAPK signaling. IL-10 inhibits osteogenic differentiation of MSCs prior to ALP expression. IL-17 promoting the transformation of MSC into bone progenitor cells or osteoblasts[24,26,28]
TNF-αHigh dose of TNF could stimulate the upregulation of some osteogenic factors in MSCs, including VEGF and insulin-like growth factor. Low-dose TNF-α inhibited the mineralization and activation of ALP and OPN in cultured MSCs[22,30]
MCP-1Influencing monocyte migration and subsequent macrophage polarization[31]
TGF-βThrough the precise matching of ligands, receptors, and cell signaling molecules, TGF-β is involved in the lineage transformation process of the differentiation of various stem cells, such as lipid, osteoblast, chondrogenic, and myogenic[34]
ChemerinChemerin promotes lipogenesis and inhibits osteogenic differentiation of MSCs[42]
Proteins of the fibrinolytic systemPAI-1 Loss of PAI-1 significantly weakened the expression of bone marrow-derived MSC osteogenic genes, such as BMP-2 and ALP[45]
Tissue factorTissue factor silencing could effectively induce higher differentiation of MSCs in osteogenic and lipid-forming media[48]
Complement and complement-related proteinsAdipsinAdipsin initiates adipogenesis from bone marrow MSCs by activating Wnt signaling[53]
AdipokinesLeptinLeptin has been shown to cross-regulate BMP-9 signaling through the JAK/STAT signaling pathway in MSCs, thereby enhancing BMP-9-induced osteogenesis[60]
Adiponectinadiponectin regulates BMSC osteogenic differentiation and osteogenesis through the Wnt/β-catenin pathway[63]
VisfatinPromoting the proliferation and mineralization activity of osteoblasts[68]
NicotinamideNampt is a speed-limit enzyme and participates in the all-around MC3T3-E1. Osteogenesis prior to the cell differentiation process of NAD salvage pathways[67]
VisceralAttenuating the osteogenic differentiation of preosteoblast cell line MC3T3-E1[73]
Bone morphogenetic proteinsBMP-7 induced increased expression of ALP, a marker of osteoblast differentiation, and accelerated calcification. The absence of BMP-2 and BMP-4 resulted in severely impaired osteogenic function. BMP-3 regulates adult bone mass by limiting the differentiation of bone progenitor cells into mature osteoblasts[78-80]
Nesfatin-1Promoting the expression of osteogenic genes such as ALP and RUNX2 in rats’ newly derived stem cells[89]
CathepsinsKnockout or inhibition of cathepsin K could promote the regeneration of bone marrow MSCs of jaw bone through glycolysis. Cathepsin S deficiency alters the balance between adipocyte and osteoblast differentiation, increases bone turnover, and alters bone microstructure[94,95]
ApelinPromoting postpositional MSC osteoblast differentiation by activating the Wnt/β-catenin signaling pathway[98]
Omentin-1Increasing the expression of BMP2, RUNX2, OPN, and osteocalcin[102]
Lipocalin 2Disrupting osteoclast formation in bone tissue by negatively regulating the proliferation and differentiation of osteoclast precursors[105]
MelatoninDifferentiating bone marrow progenitors from adipocytes to osteoblasts[111]
Gremlin-1BMP protein inhibitor[112]
Lipid transportApoE Enhancing osteogenic differentiation of the mouse mesenchymal progenitor cell line[120]
EnzymesDPP-4Restricting the induction of osteogenic differentiation of heart artery flap-derived mesenchymal cells by the autocrine insulin-like growth factor-1 signaling pathway[122]
Tissue inhibitors of metalloproteinasesInhibition of endogenous TIMP-1 can inhibit the proliferation, metabolic activity, and osteogenic differentiation ability of MSCs by activating the Wnt/β-catenin signal[127]

In summary, this paper reviewed the current research on the regulation and influence of adipokine in the osteoblast differentiation of MSCs. However, this review did not include all currently discovered adipokines but only included published studies involving osteogenic differentiation of MSCs. Most of the included studies were conducted in BMSCs, with a small number involving osteoblast precursor cells, progenitor cells, and a small number of other tissue-derived stem cells. Our review suggests that different adipokines have different effects on the outcome of osteogenic differentiation, bone regeneration, and bone remodeling of MSCs. The progress of related research provides a good reference for subsequent preclinical and clinical studies and a new reference for treating osteogenic disorders and diseases of osteoblastic homeostasis.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell biology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Scarfì S, Italy; Stogov MV, Russia S-Editor: Zhang H L-Editor: Filipodia P-Editor: Liu JH

References
1.  Klöting N, Blüher M. Adipocyte dysfunction, inflammation and metabolic syndrome. Rev Endocr Metab Disord. 2014;15:277-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 322]  [Article Influence: 32.2]  [Reference Citation Analysis (0)]
2.  Blüher M. Adipose tissue dysfunction contributes to obesity related metabolic diseases. Best Pract Res Clin Endocrinol Metab. 2013;27:163-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 239]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
3.  Fasshauer M, Blüher M. Adipokines in health and disease. Trends Pharmacol Sci. 2015;36:461-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 656]  [Article Influence: 72.9]  [Reference Citation Analysis (0)]
4.  Blüher M. Adipokines – removing road blocks to obesity and diabetes therapy. Mol Metab. 2014;3:230-240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 161]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
5.  Blüher M, Mantzoros CS. From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century. Metabolism. 2015;64:131-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 270]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
6.  Chen Q, Shou P, Zheng C, Jiang M, Cao G, Yang Q, Cao J, Xie N, Velletri T, Zhang X, Xu C, Zhang L, Yang H, Hou J, Wang Y, Shi Y. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 2016;23:1128-1139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 606]  [Cited by in F6Publishing: 744]  [Article Influence: 93.0]  [Reference Citation Analysis (0)]
7.  Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279-4295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4817]  [Cited by in F6Publishing: 4876]  [Article Influence: 221.6]  [Reference Citation Analysis (0)]
8.  Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15372]  [Cited by in F6Publishing: 14786]  [Article Influence: 591.4]  [Reference Citation Analysis (0)]
9.  Li T, Xia M, Gao Y, Chen Y, Xu Y. Human umbilical cord mesenchymal stem cells: an overview of their potential in cell-based therapy. Expert Opin Biol Ther. 2015;15:1293-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 148]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
10.  Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci. 2019;30:839-855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 65]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
11.  Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol. 2021;12:728190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
12.  Ding DC, Chang YH, Shyu WC, Lin SZ. Human umbilical cord mesenchymal stem cells: a new era for stem cell therapy. Cell Transplant. 2015;24:339-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 326]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
13.  Teitelbaum SL. Bone resorption by osteoclasts. Science. 2000;289:1504-1508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2794]  [Cited by in F6Publishing: 2670]  [Article Influence: 111.3]  [Reference Citation Analysis (0)]
14.  Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3539]  [Cited by in F6Publishing: 3167]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
15.  Jiang Y, Zhang P, Zhang X, Lv L, Zhou Y. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Prolif. 2021;54:e12956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 103]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
16.  Akbar MA, Lu Y, Elshikha AS, Chen MJ, Yuan Y, Whitley EM, Holliday LS, Chang LJ, Song S. Transplantation of Adipose Tissue-Derived Mesenchymal Stem Cell (ATMSC) Expressing Alpha-1 Antitrypsin Reduces Bone Loss in Ovariectomized Osteoporosis Mice. Hum Gene Ther. 2017;28:179-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Loi F, Córdova LA, Pajarinen J, Lin TH, Yao Z, Goodman SB. Inflammation, fracture and bone repair. Bone. 2016;86:119-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 644]  [Cited by in F6Publishing: 689]  [Article Influence: 86.1]  [Reference Citation Analysis (0)]
18.  Fu X, Liu G, Halim A, Ju Y, Luo Q, Song AG. Mesenchymal Stem Cell Migration and Tissue Repair. Cells. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 269]  [Cited by in F6Publishing: 467]  [Article Influence: 93.4]  [Reference Citation Analysis (0)]
19.  Brown C, McKee C, Bakshi S, Walker K, Hakman E, Halassy S, Svinarich D, Dodds R, Govind CK, Chaudhry GR. Mesenchymal stem cells: Cell therapy and regeneration potential. J Tissue Eng Regen Med. 2019;13:1738-1755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 298]  [Article Influence: 59.6]  [Reference Citation Analysis (0)]
20.  Grayson WL, Bunnell BA, Martin E, Frazier T, Hung BP, Gimble JM. Stromal cells and stem cells in clinical bone regeneration. Nat Rev Endocrinol. 2015;11:140-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 271]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
21.  Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci. 2022;79:272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 34]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
22.  Lacey DC, Simmons PJ, Graves SE, Hamilton JA. Proinflammatory cytokines inhibit osteogenic differentiation from stem cells: implications for bone repair during inflammation. Osteoarthritis Cartilage. 2009;17:735-742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 215]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
23.  Majumdar MK, Thiede MA, Haynesworth SE, Bruder SP, Gerson SL. Human marrow-derived mesenchymal stem cells (MSCs) express hematopoietic cytokines and support long-term hematopoiesis when differentiated toward stromal and osteogenic lineages. J Hematother Stem Cell Res. 2000;9:841-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 338]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
24.  Rezaee F, Rellick SL, Piedimonte G, Akers SM, O’Leary HA, Martin K, Craig MD, Gibson LF. Neurotrophins regulate bone marrow stromal cell IL-6 expression through the MAPK pathway. PloS One. 2010;5:e9690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
25.  Carmody EE, Schwarz EM, Puzas JE, Rosier RN, O’Keefe RJ. Viral interleukin-10 gene inhibition of inflammation, osteoclastogenesis, and bone resorption in response to titanium particles. Arthritis Rheum. 2002;46:1298-1308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 66]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
26.  Van Vlasselaer P, Borremans B, Van Den Heuvel R, Van Gorp U, de Waal Malefyt R. Interleukin-10 inhibits the osteogenic activity of mouse bone marrow. Blood. 1993;82:2361-2370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Krstić J, Mojsilović S, Mojsilović SS, Santibanez JF. Regulation of the mesenchymal stem cell fate by interleukin-17: Implications in osteogenic differentiation. World J Stem Cells. 2021;13:1696-1713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
28.  Ling L, Nurcombe V, Cool SM. Wnt signaling controls the fate of mesenchymal stem cells. Gene. 2009;433:1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 314]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
29.  Chang J, Wang Z, Tang E, Fan Z, McCauley L, Franceschi R, Guan K, Krebsbach PH, Wang CY. Inhibition of osteoblastic bone formation by nuclear factor-kappaB. Nat Med. 2009;15:682-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 364]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
30.  Wang M, Crisostomo PR, Herring C, Meldrum KK, Meldrum DR. Human progenitor cells from bone marrow or adipose tissue produce VEGF, HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am J Physiol Regul Integr Comp Physiol. 2006;291:R880-R884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 214]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
31.  Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res. 2009;29:313-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2327]  [Cited by in F6Publishing: 2630]  [Article Influence: 175.3]  [Reference Citation Analysis (0)]
32.  Xie Z, Wang P, Li J, Li Y, Wang S, Wu X, Sun S, Cen S, Su H, Deng W, Liu Z, Ouyang Y, Wu Y, Shen H. MCP1 triggers monocyte dysfunctions during abnormal osteogenic differentiation of mesenchymal stem cells in ankylosing spondylitis. J Mol Med (Berl). 2017;95:143-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
33.  Crane JL, Cao X. Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling. J Clin Invest. 2014;124:466-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 295]  [Article Influence: 29.5]  [Reference Citation Analysis (0)]
34.  Li SN, Wu JF. TGF-β/SMAD signaling regulation of mesenchymal stem cells in adipocyte commitment. Stem Cell Res Ther. 2020;11:41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 78]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
35.  Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, Zhao L, Nagy TR, Peng X, Hu J, Feng X, Van Hul W, Wan M, Cao X. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med. 2009;15:757-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 911]  [Cited by in F6Publishing: 846]  [Article Influence: 56.4]  [Reference Citation Analysis (0)]
36.  Zhou S. TGF-β regulates β-catenin signaling and osteoblast differentiation in human mesenchymal stem cells. J Cell Biochem. 2011;112:1651-1660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 97]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
37.  Jian H, Shen X, Liu I, Semenov M, He X, Wang XF. Smad3-dependent nuclear translocation of beta-catenin is required for TGF-beta1-induced proliferation of bone marrow-derived adult human mesenchymal stem cells. Genes Dev. 2006;20:666-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 227]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
38.  Du G, Cheng X, Zhang Z, Han L, Wu K, Li Y, Lin X. TGF-Beta Induced Key Genes of Osteogenic and Adipogenic Differentiation in Human Mesenchymal Stem Cells and MiRNA-mRNA Regulatory Networks. Front Genet. 2021;12:759596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
39.  Igarashi Y, Chosa N, Sawada S, Kondo H, Yaegashi T, Ishisaki A. VEGF-C and TGF-β reciprocally regulate mesenchymal stem cell commitment to differentiation into lymphatic endothelial or osteoblastic phenotypes. Int J Mol Med. 2016;37:1005-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
40.  Bozaoglu K, Bolton K, McMillan J, Zimmet P, Jowett J, Collier G, Walder K, Segal D. Chemerin is a novel adipokine associated with obesity and metabolic syndrome. Endocrinology. 2007;148:4687-4694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 576]  [Cited by in F6Publishing: 602]  [Article Influence: 35.4]  [Reference Citation Analysis (0)]
41.  Li J, Zhang T, Huang C, Xu M, Xie W, Pei Q, Xie X, Wang B, Li X. Chemerin located in bone marrow promotes osteogenic differentiation and bone formation via Akt/Gsk3β/β-catenin axis in mice. J Cell Physiol. 2021;236:6042-6054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
42.  Kadric L, Zylla S, Nauck M, Völzke H, Friedrich N, Hannemann A. Associations Between Plasma Chemerin Concentrations and Bone Quality in Adults From the General Population. Endocrinology. 2018;159:2378-2385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
43.  Muruganandan S, Roman AA, Sinal CJ. Role of chemerin/CMKLR1 signaling in adipogenesis and osteoblastogenesis of bone marrow stem cells. J Bone Miner Res. 2010;25:222-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 121]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
44.  Kaji H. Adipose Tissue-Derived Plasminogen Activator Inhibitor-1 Function and Regulation. Compr Physiol. 2016;6:1873-1896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
45.  Takafuji Y, Tatsumi K, Ishida M, Kawao N, Okada K, Matsuo O, Kaji H. Plasminogen activator inhibitor-1 deficiency suppresses osteoblastic differentiation of mesenchymal stem cells in mice. J Cell Physiol. 2019;234:9687-9697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
46.  Drake TA, Morrissey JH, Edgington TS. Selective cellular expression of tissue factor in human tissues. Implications for disorders of hemostasis and thrombosis. Am J Pathol. 1989;134:1087-1097.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Nitori N, Ino Y, Nakanishi Y, Yamada T, Honda K, Yanagihara K, Kosuge T, Kanai Y, Kitajima M, Hirohashi S. Prognostic significance of tissue factor in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2005;11:2531-2539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 125]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
48.  Rangasami VK, Nawale G, Asawa K, Kadekar S, Samanta S, Nilsson B, Ekdahl KN, Miettinen S, Hilborn J, Teramura Y, Varghese OP, Oommen OP. Pluronic Micelle-Mediated Tissue Factor Silencing Enhances Hemocompatibility, Stemness, Differentiation Potential, and Paracrine Signaling of Mesenchymal Stem Cells. Biomacromolecules. 2021;22:1980-1989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
49.  Cook KS, Min HY, Johnson D, Chaplinsky RJ, Flier JS, Hunt CR, Spiegelman BM. Adipsin: a circulating serine protease homolog secreted by adipose tissue and sciatic nerve. Science. 1987;237:402-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in F6Publishing: 273]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
50.  Rosen BS, Cook KS, Yaglom J, Groves DL, Volanakis JE, Damm D, White T, Spiegelman BM. Adipsin and complement factor D activity: an immune-related defect in obesity. Science. 1989;244:1483-1487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 214]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
51.  Choy LN, Rosen BS, Spiegelman BM. Adipsin and an endogenous pathway of complement from adipose cells. J Biol Chem. 1992;267:12736-12741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 75]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
52.  Gómez-Banoy N, Guseh JS, Li G, Rubio-Navarro A, Chen T, Poirier B, Putzel G, Rosselot C, Pabón MA, Camporez JP, Bhambhani V, Hwang SJ, Yao C, Perry RJ, Mukherjee S, Larson MG, Levy D, Dow LE, Shulman GI, Dephoure N, Garcia-Ocana A, Hao M, Spiegelman BM, Ho JE, Lo JC. Adipsin preserves beta cells in diabetic mice and associates with protection from type 2 diabetes in humans. Nat Med. 2019;25:1739-1747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
53.  Aaron N, Kraakman MJ, Zhou Q, Liu Q, Costa S, Yang J, Liu L, Yu L, Wang L, He Y, Fan L, Hirakawa H, Ding L, Lo J, Wang W, Zhao B, Guo E, Sun L, Rosen CJ, Qiang L. Adipsin promotes bone marrow adiposity by priming mesenchymal stem cells. Elife. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
54.  Bienertova-Vasku J, Vinciguerra M, Buzga M, Villaroya F. Adipokines as Biomarkers in Health and Disease. Dis Markers. 2018;2018:5696815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
55.  Reid IR, Baldock PA, Cornish J. Effects of Leptin on the Skeleton. Endocr Rev. 2018;39:938-959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 92]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
56.  Park HK, Ahima RS. Leptin signaling. F1000Prime Rep. 2014;6:73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 116]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
57.  Yue R, Zhou BO, Shimada IS, Zhao Z, Morrison SJ. Leptin Receptor Promotes Adipogenesis and Reduces Osteogenesis by Regulating Mesenchymal Stromal Cells in Adult Bone Marrow. Cell Stem Cell. 2016;18:782-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 299]  [Article Influence: 37.4]  [Reference Citation Analysis (0)]
58.  Kelesidis T, Kelesidis I, Chou S, Mantzoros CS. Narrative review: the role of leptin in human physiology: emerging clinical applications. Ann Intern Med. 2010;152:93-100.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 450]  [Cited by in F6Publishing: 410]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
59.  Zhou BO, Yue R, Murphy MM, Peyer JG, Morrison SJ. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014;15:154-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 807]  [Cited by in F6Publishing: 925]  [Article Influence: 92.5]  [Reference Citation Analysis (0)]
60.  Zhang B, Yang L, Zeng Z, Feng Y, Wang X, Wu X, Luo H, Zhang J, Zhang M, Pakvasa M, Wagstaff W, He F, Mao Y, Qin K, Ding H, Zhang Y, Niu C, Wu M, Zhao X, Wang H, Huang L, Shi D, Liu Q, Ni N, Fu K, Athiviraham A, Moriatis Wolf J, Lee MJ, Hynes K, Strelzow J, El Dafrawy M, Xia Y, He TC. Leptin Potentiates BMP9-Induced Osteogenic Differentiation of Mesenchymal Stem Cells Through the Activation of JAK/STAT Signaling. Stem Cells Dev. 2020;29:498-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 34]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
61.  Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med. 2001;7:947-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1868]  [Cited by in F6Publishing: 1797]  [Article Influence: 78.1]  [Reference Citation Analysis (0)]
62.  Padmalayam I, Suto M. Role of adiponectin in the metabolic syndrome: current perspectives on its modulation as a treatment strategy. Curr Pharm Des. 2013;19:5755-5763.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
63.  Wang Y, Zhang X, Shao J, Liu H, Liu X, Luo E. Adiponectin regulates BMSC osteogenic differentiation and osteogenesis through the Wnt/β-catenin pathway. Sci Rep. 2017;7:3652.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
64.  Chen T, Wu YW, Lu H, Guo Y, Tang ZH. Adiponectin enhances osteogenic differentiation in human adipose-derived stem cells by activating the APPL1-AMPK signaling pathway. Biochem Biophys Res Commun. 2015;461:237-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
65.  Pu Y, Wu H, Lu S, Hu H, Li D, Wu Y, Tang Z. Adiponectin Promotes Human Jaw Bone Marrow Stem Cell Osteogenesis. J Dent Res. 2016;95:769-775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
66.  Wang Y, Du Y, Yuan H, Pan Y, Wu J, Du X, Hao S, Yan Z, Li X, Liu K, Xu F. Human amnion-derived mesenchymal stem cells enhance the osteogenic differentiation of human adipose-derived stem cells by promoting adiponectin excretion via the APPL1-ERK1/2 signaling pathway. IUBMB Life. 2020;72:296-304.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
67.  Li Y, He J, He X, Li Y, Lindgren U. Nampt expression increases during osteogenic differentiation of multi- and omnipotent progenitors. Biochem Biophys Res Commun. 2013;434:117-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
68.  Otero M, Lago R, Gomez R, Lago F, Dieguez C, Gómez-Reino JJ, Gualillo O. Changes in plasma levels of fat-derived hormones adiponectin, leptin, resistin and visfatin in patients with rheumatoid arthritis. Ann Rheum Dis. 2006;65:1198-1201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 382]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
69.  Tsiklauri L, Werner J, Kampschulte M, Frommer KW, Berninger L, Irrgang M, Glenske K, Hose D, El Khassawna T, Pons-Kühnemann J, Rehart S, Wenisch S, Müller-Ladner U, Neumann E. Visfatin alters the cytokine and matrix-degrading enzyme profile during osteogenic and adipogenic MSC differentiation. Osteoarthritis Cartilage. 2018;26:1225-1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
70.  Braidy N, Berg J, Clement J, Khorshidi F, Poljak A, Jayasena T, Grant R, Sachdev P. Role of Nicotinamide Adenine Dinucleotide and Related Precursors as Therapeutic Targets for Age-Related Degenerative Diseases: Rationale, Biochemistry, Pharmacokinetics, and Outcomes. Antioxid Redox Signal. 2019;30:251-294.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 128]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
71.  Sommer G, Garten A, Petzold S, Beck-Sickinger AG, Blüher M, Stumvoll M, Fasshauer M. Visfatin/PBEF/Nampt: structure, regulation and potential function of a novel adipokine. Clin Sci (Lond). 2008;115:13-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 93]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
72.  He X, He J, Shi Y, Pi C, Yang Y, Sun Y, Ma C, Lin L, Zhang L, Li Y. Nicotinamide phosphoribosyltransferase (Nampt) may serve as the marker for osteoblast differentiation of bone marrow-derived mesenchymal stem cells. Exp Cell Res. 2017;352:45-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
73.  Liu Y, Xu F, Pei HX, Zhu X, Lin X, Song CY, Liang QH, Liao EY, Yuan LQ. Vaspin regulates the osteogenic differentiation of MC3T3-E1 through the PI3K-Akt/miR-34c loop. Sci Rep. 2016;6:25578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
74.  Wang H, Chen F, Li J, Wang Y, Jiang C, Zhang M, Xu J. Vaspin antagonizes high fat-induced bone loss in rats and promotes osteoblastic differentiation in primary rat osteoblasts through Smad-Runx2 signaling pathway. Nutr Metab (Lond). 2020;17:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
75.  Schulz TJ, Huang P, Huang TL, Xue R, McDougall LE, Townsend KL, Cypess AM, Mishina Y, Gussoni E, Tseng YH. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature. 2013;495:379-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 303]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
76.  Schulz TJ, Tseng YH. Emerging role of bone morphogenetic proteins in adipogenesis and energy metabolism. Cytokine Growth Factor Rev. 2009;20:523-531.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 118]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
77.  Huang Z, Ren PG, Ma T, Smith RL, Goodman SB. Modulating osteogenesis of mesenchymal stem cells by modifying growth factor availability. Cytokine. 2010;51:305-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 108]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
78.  Gu K, Zhang L, Jin T, Rutherford RB. Identification of potential modifiers of Runx2/Cbfa1 activity in C2C12 cells in response to bone morphogenetic protein-7. Cells Tissues Organs. 2004;176:28-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
79.  Tsuji K, Cox K, Bandyopadhyay A, Harfe BD, Tabin CJ, Rosen V. BMP4 is dispensable for skeletogenesis and fracture-healing in the limb. J Bone Joint Surg Am. 2008;90 Suppl 1:14-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
80.  Kokabu S, Gamer L, Cox K, Lowery J, Tsuji K, Raz R, Economides A, Katagiri T, Rosen V. BMP3 suppresses osteoblast differentiation of bone marrow stromal cells via interaction with Acvr2b. Mol Endocrinol. 2012;26:87-94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
81.  Vaccaro AR, Whang PG, Patel T, Phillips FM, Anderson DG, Albert TJ, Hilibrand AS, Brower RS, Kurd MF, Appannagari A, Patel M, Fischgrund JS. The safety and efficacy of OP-1 (rhBMP-7) as a replacement for iliac crest autograft for posterolateral lumbar arthrodesis: minimum 4-year follow-up of a pilot study. Spine J. 2008;8:457-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 128]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
82.  Rivera JC, Strohbach CA, Wenke JC, Rathbone CR. Beyond osteogenesis: an in vitro comparison of the potentials of six bone morphogenetic proteins. Front Pharmacol. 2013;4:125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
83.  Tiaden AN, Breiden M, Mirsaidi A, Weber FA, Bahrenberg G, Glanz S, Cinelli P, Ehrmann M, Richards PJ. Human serine protease HTRA1 positively regulates osteogenesis of human bone marrow-derived mesenchymal stem cells and mineralization of differentiating bone-forming cells through the modulation of extracellular matrix protein. Stem Cells. 2012;30:2271-2282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
84.  Zachos TA, Shields KM, Bertone AL. Gene-mediated osteogenic differentiation of stem cells by bone morphogenetic proteins-2 or -6. J Orthop Res. 2006;24:1279-1291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 52]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
85.  Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J Cell Biochem. 2006;98:538-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 156]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
86.  Goebel-Stengel M, Stengel A. Role of Brain NUCB2/nesfatin-1 in the Stress-induced Modulation of Gastrointestinal Functions. Curr Neuropharmacol. 2016;14:882-891.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
87.  Levata L, Dore R, Jöhren O, Schwaninger M, Schulz C, Lehnert H. Nesfatin-1 Acts Centrally to Induce Sympathetic Activation of Brown Adipose Tissue and Non-Shivering Thermogenesis. Horm Metab Res. 2019;51:678-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
88.  Ranjan A, Choubey M, Yada T, Krishna A. Nesfatin-1 ameliorates type-2 diabetes-associated reproductive dysfunction in male mice. J Endocrinol Invest. 2020;43:515-528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
89.  Xu K, Zhang Z, Chen M, Moqbel SAA, He Y, Ma C, Jiang L, Xiong Y, Wu L. Nesfatin-1 Promotes the Osteogenic Differentiation of Tendon-Derived Stem Cells and the Pathogenesis of Heterotopic Ossification in Rat Tendons via the mTOR Pathway. Front Cell Dev Biol. 2020;8:547342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
90.  Lecaille F, Chazeirat T, Saidi A, Lalmanach G. Cathepsin V: Molecular characteristics and significance in health and disease. Mol Aspects Med. 2022;88:101086.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
91.  Fain JN. Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm. 2006;74:443-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 445]  [Cited by in F6Publishing: 448]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
92.  Liu F, Zhou ZF, An Y, Yu Y, Wu RX, Yin Y, Xue Y, Chen FM. Effects of cathepsin K on Emdogain-induced hard tissue formation by human periodontal ligament stem cells. J Tissue Eng Regen Med. 2017;11:2922-2934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
93.  Whitty C, Wardale RJ, Henson FMD. The regulation of sclerostin by cathepsin K in periodontal ligament cells. Biochem Biophys Res Commun. 2018;503:550-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
94.  Zhang W, Dong Z, Li D, Li B, Liu Y, Zheng X, Liu H, Zhou H, Hu K, Xue Y. Cathepsin K deficiency promotes alveolar bone regeneration by promoting jaw bone marrow mesenchymal stem cells proliferation and differentiation via glycolysis pathway. Cell Prolif. 2021;54:e13058.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
95.  Rauner M, Föger-Samwald U, Kurz MF, Brünner-Kubath C, Schamall D, Kapfenberger A, Varga P, Kudlacek S, Wutzl A, Höger H, Zysset PK, Shi GP, Hofbauer LC, Sipos W, Pietschmann P. Cathepsin S controls adipocytic and osteoblastic differentiation, bone turnover, and bone microarchitecture. Bone. 2014;64:281-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
96.  Fasshauer M, Blüher M, Stumvoll M. Adipokines in gestational diabetes. Lancet Diabetes Endocrinol. 2014;2:488-499.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 147]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
97.  Castan-Laurell I, Dray C, Attané C, Duparc T, Knauf C, Valet P. Apelin, diabetes, and obesity. Endocrine. 2011;40:1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
98.  Hang K, Ye C, Xu J, Chen E, Wang C, Zhang W, Ni L, Kuang Z, Ying L, Xue D, Pan Z. Apelin enhances the osteogenic differentiation of human bone marrow mesenchymal stem cells partly through Wnt/β-catenin signaling pathway. Stem Cell Res Ther. 2019;10:189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
99.  Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, Shuldiner AR, Fried SK, McLenithan JC, Gong DW. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab. 2006;290:E1253-E1261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 583]  [Article Influence: 32.4]  [Reference Citation Analysis (0)]
100.  Yamawaki H, Tsubaki N, Mukohda M, Okada M, Hara Y. Omentin, a novel adipokine, induces vasodilation in rat isolated blood vessels. Biochem Biophys Res Commun. 2010;393:668-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 161]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
101.  Tohidi M, Akbarzadeh S, Larijani B, Kalantarhormozi M, Ostovar A, Assadi M, Vahdat K, Farrokhnia M, Sanjdideh Z, Amirinejad R, Nabipour I. Omentin-1, visfatin and adiponectin levels in relation to bone mineral density in Iranian postmenopausal women. Bone. 2012;51:876-881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
102.  Tang C, Liang D, Qiu Y, Zhu J, Tang G. Omentin1 induces osteoblast viability and differentiation via the TGFβ/Smad signaling pathway in osteoporosis. Mol Med Rep. 2022;25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
103.  Yan QW, Yang Q, Mody N, Graham TE, Hsu CH, Xu Z, Houstis NE, Kahn BB, Rosen ED. The adipokine lipocalin 2 is regulated by obesity and promotes insulin resistance. Diabetes. 2007;56:2533-2540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 328]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
104.  Jung M, Weigert A, Mertens C, Rehwald C, Brüne B. Iron Handling in Tumor-Associated Macrophages-Is There a New Role for Lipocalin-2? Front Immunol. 2017;8:1171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
105.  Kim HJ, Yoon HJ, Yoon KA, Gwon MR, Jin Seong S, Suk K, Kim SY, Yoon YR. Lipocalin-2 inhibits osteoclast formation by suppressing the proliferation and differentiation of osteoclast lineage cells. Exp Cell Res. 2015;334:301-309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
106.  Yin C, Jia X, Zhao Q, Zhao Z, Wang J, Zhang Y, Li Z, Sun H. Transcription factor 7-like 2 promotes osteogenic differentiation and boron-induced bone repair via lipocalin 2. Mater Sci Eng C Mater Biol Appl. 2020;110:110671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
107.  Luchetti F, Canonico B, Bartolini D, Arcangeletti M, Ciffolilli S, Murdolo G, Piroddi M, Papa S, Reiter RJ, Galli F. Melatonin regulates mesenchymal stem cell differentiation: a review. J Pineal Res. 2014;56:382-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 124]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
108.  Cardinali DP, Srinivasan V, Brzezinski A, Brown GM. Melatonin and its analogs in insomnia and depression. J Pineal Res. 2012;52:365-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 207]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
109.  Rodriguez-Garcia A, Mayo JC, Hevia D, Quiros-Gonzalez I, Navarro M, Sainz RM. Phenotypic changes caused by melatonin increased sensitivity of prostate cancer cells to cytokine-induced apoptosis. J Pineal Res. 2013;54:33-45.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
110.  Murdolo G, Piroddi M, Luchetti F, Tortoioli C, Canonico B, Zerbinati C, Galli F, Iuliano L. Oxidative stress and lipid peroxidation by-products at the crossroad between adipose organ dysregulation and obesity-linked insulin resistance. Biochimie. 2013;95:585-594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 82]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
111.  Zhang L, Zhang J, Ling Y, Chen C, Liang A, Peng Y, Chang H, Su P, Huang D. Sustained release of melatonin from poly (lactic-co-glycolic acid) (PLGA) microspheres to induce osteogenesis of human mesenchymal stem cells in vitro. J Pineal Res. 2013;54:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
112.  Topol LZ, Bardot B, Zhang Q, Resau J, Huillard E, Marx M, Calothy G, Blair DG. Biosynthesis, post-translation modification, and functional characterization of Drm/Gremlin. J Biol Chem. 2000;275:8785-8793.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 108]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
113.  Grillo E, Ravelli C, Colleluori G, D’Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev. 2023;69:51-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
114.  Gazzerro E, Smerdel-Ramoya A, Zanotti S, Stadmeyer L, Durant D, Economides AN, Canalis E. Conditional deletion of gremlin causes a transient increase in bone formation and bone mass. J Biol Chem. 2007;282:31549-31557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 86]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
115.  Hu K, Sun H, Gui B, Sui C. Gremlin-1 suppression increases BMP-2-induced osteogenesis of human mesenchymal stem cells. Mol Med Rep. 2017;15:2186-2194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
116.  Shore VG, Shore B. Heterogeneity of human plasma very low density lipoproteins. Separation of species differing in protein components. Biochemistry. 1973;12:502-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 256]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
117.  Marais AD. Apolipoprotein E and Atherosclerosis. Curr Atheroscler Rep. 2021;23:34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
118.  Marais AD. Apolipoprotein E in lipoprotein metabolism, health and cardiovascular disease. Pathology. 2019;51:165-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 176]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
119.  Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol Dis. 2014;72 Pt A:3-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 478]  [Cited by in F6Publishing: 437]  [Article Influence: 43.7]  [Reference Citation Analysis (0)]
120.  Bächner D, Schröder D, Betat N, Ahrens M, Gross G. Apolipoprotein E (ApoE), a Bmp-2 (bone morphogenetic protein) upregulated gene in mesenchymal progenitors (C3H10T1/2), is highly expressed in murine embryonic development. Biofactors. 1999;9:11-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
121.  Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr S, Ouwens DM, Eckardt K, Kaufman JM, Ryden M, Müller S, Hanisch FG, Ruige J, Arner P, Sell H, Eckel J. Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome. Diabetes. 2011;60:1917-1925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 431]  [Cited by in F6Publishing: 444]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
122.  Choi B, Lee S, Kim SM, Lee EJ, Lee SR, Kim DH, Jang JY, Kang SW, Lee KU, Chang EJ, Song JK. Dipeptidyl Peptidase-4 Induces Aortic Valve Calcification by Inhibiting Insulin-Like Growth Factor-1 Signaling in Valvular Interstitial Cells. Circulation. 2017;135:1935-1950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
123.  Lambert E, Dassé E, Haye B, Petitfrère E. TIMPs as multifacial proteins. Crit Rev Oncol Hematol. 2004;49:187-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 364]  [Cited by in F6Publishing: 317]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
124.  Lu P, Takai K, Weaver VM, Werb Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol. 2011;3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1129]  [Cited by in F6Publishing: 1364]  [Article Influence: 104.9]  [Reference Citation Analysis (0)]
125.  Arpino V, Brock M, Gill SE. The role of TIMPs in regulation of extracellular matrix proteolysis. Matrix Biol. 2015;44-46:247-254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 382]  [Cited by in F6Publishing: 442]  [Article Influence: 49.1]  [Reference Citation Analysis (0)]
126.  Jackson HW, Defamie V, Waterhouse P, Khokha R. TIMPs: versatile extracellular regulators in cancer. Nat Rev Cancer. 2017;17:38-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 287]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
127.  Egea V, Zahler S, Rieth N, Neth P, Popp T, Kehe K, Jochum M, Ries C. Tissue inhibitor of metalloproteinase-1 (TIMP-1) regulates mesenchymal stem cells through let-7f microRNA and Wnt/β-catenin signaling. Proc Natl Acad Sci U S A. 2012;109:E309-E316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 110]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
128.  Liang T, Gao W, Zhu L, Ren J, Yao H, Wang K, Shi D. TIMP-1 inhibits proliferation and osteogenic differentiation of hBMSCs through Wnt/β-catenin signaling. Biosci Rep. 2019;39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
129.  Jiang C, Xia W, Wu T, Pan C, Shan H, Wang F, Zhou Z, Yu X. Inhibition of microRNA-222 up-regulates TIMP3 to promotes osteogenic differentiation of MSCs from fracture rats with type 2 diabetes mellitus. J Cell Mol Med. 2020;24:686-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
130.  Lehr S, Hartwig S, Sell H. Adipokines: a treasure trove for the discovery of biomarkers for metabolic disorders. Proteomics Clin Appl. 2012;6:91-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 221]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
131.  Kra G, Daddam JR, Moallem U, Kamer H, Kočvarová R, Nemirovski A, Contreras GA, Tam J, Zachut M. Effects of omega-3 supplementation on components of the endocannabinoid system and metabolic and inflammatory responses in adipose and liver of peripartum dairy cows. J Anim Sci Biotechnol. 2022;13:114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
132.  Cruz-López EO, Uijl E, Danser AHJ. Perivascular Adipose Tissue in Vascular Function: Does Locally Synthesized Angiotensinogen Play a Role? J Cardiovasc Pharmacol. 2021;78:S53-S62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]