Next Article in Journal
The Role of Circular RNAs in the Physiology and Pathology of the Mammalian Ovary
Next Article in Special Issue
Emerging RUNX2-Mediated Gene Regulatory Mechanisms Consisting of Multi-Layered Regulatory Networks in Skeletal Development
Previous Article in Journal
Association of B Cells with Idiopathic Recurrent Pregnancy Loss: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Progress and Current Status in Hajdu-Cheney Syndrome with Focus on Novel Genetic Research
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Kruppel-like Factors in Skeletal Physiology and Pathologies

1
Department of Oral Anatomy and Developmental Biology, Osaka University Graduate School of Dentistry, 1-8 Yamada-oka, Suita 565-0871, Osaka, Japan
2
Division of Special Care Dentistry, Osaka University Dental Hospital, 1-8 Yamada-oka, Suita 565-0871, Osaka, Japan
3
Department of Applied Prosthodontics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8521, Nagasaki, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15174; https://doi.org/10.3390/ijms232315174
Submission received: 4 November 2022 / Revised: 28 November 2022 / Accepted: 30 November 2022 / Published: 2 December 2022
(This article belongs to the Special Issue Genetics in Bone Diseases)

Abstract

:
Kruppel-like factors (KLFs) belong to a large group of zinc finger-containing transcription factors with amino acid sequences resembling the Drosophila gap gene Krüppel. Since the first report of molecular cloning of the KLF family gene, the number of KLFs has increased rapidly. Currently, 17 murine and human KLFs are known to play crucial roles in the regulation of transcription, cell proliferation, cellular differentiation, stem cell maintenance, and tissue and organ pathogenesis. Recent evidence has shown that many KLF family molecules affect skeletal cells and regulate their differentiation and function. This review summarizes the current understanding of the unique roles of each KLF in skeletal cells during normal development and skeletal pathologies.

1. Skeletal Development and Maintenance

Skeletons play a crucial role in the physical support of the body. Subsequent to gastrulation, the notochord is formed, which is a cartilaginous shaft in the growing fetus, providing physical support to the body. In the late stages of embryogenesis, numerous bones and cartilage develop to accommodate the acute expansion of body size. Several cell types, including osteoblasts, chondrocytes, osteoclasts, osteocytes, and vascular endothelial cells, cooperate to generate skeletal tissue, of which bone-forming osteoblasts and bone-resorbing osteoclasts are the two main cellular constituents [1].
Osteoprogenitor cells arise from multiple anatomical sites during embryogenesis. Osteoblasts in the craniofacial regions originate from neural crest cells derived from the dorsal neural ectoderm [2]. In contrast, axial and appendicular long bones originate from the paraxial mesoderm and lateral plate mesoderm, respectively [3]. Bones also have the unique ability to regenerate after injury and are restored to the pre-injury state in most cases. Postnatal skeletal stem cells play an important role in bone repair [4]. Skeletal stem cells can differentiate into bone, cartilage, and bone stroma, and also support hematopoiesis [5,6]. The differentiation of bone cells relies on the intrinsic activation of specific transcription factors at specific moments and sites. Crucial transcription factors, including SRY-box transcription factor 9 (SOX9), runt-related transcription factor 2 (RUNX2), osterix (SP7), nuclear factor of activated T-cells (NFATc1), nuclear factor-κB (NF-kB), and strawberry notch homolog 2 (SBNO2), have been identified in the timely differentiation of bone-forming and bone-resorbing cells [4]. Accumulating evidence has demonstrated that Kruppel-like factor (KLF), a vertebrate homologue of Drosophila Krüppel, plays vital roles in bone cell differentiation and skeletal maintenance. This review summarizes the current knowledge of KLFs in skeletal physiology and pathologies.

2. Krüppel in Drosophila

Kruppel-like factors were initially identified as C2H2 type zinc finger proteins that contain amino acid sequences resembling the Drosophila segmentation gap gene Krüppel (Figure 1) [7]. Kruppel-type zinc fingers contain a consensus amino acid sequence (Y/F)XCX2CX3FX5LX2HX3HTGEKP, in which X can be any amino acid (Cys and His residues conserved in the C2H2 class written in bold) [8,9,10]. The consensus features of the Kruppel family of zinc finger genes include an H-C link (amino acid sequence HTGEKP(Y/F)XC connecting the His of one finger to the Cys of the next) [7]. As conservation is observed in the zinc finger amino acid sequences, as well as in the interfinger spacer domain, this sequence has been called the Krüppel motif (Figure 1A,B). The Krüppel motif is also observed in molecules belonging to the Sp1 (Figure 1B) and GLI families [11,12,13,14]. In fly embryos, the Krüppel gene is intimately involved in the anterior–posterior axis and body segment formation [15]. Through the positive and negative transcriptional effects of maternally coordinated genes (consisting mainly of bicoid and nanos), gap genes are zygotically transcribed at specific spatial domains. In particular, the expression domain of Krüppel is determined by the maternally derived bicoid, which is expressed anteriorly, and nanos, which is expressed posteriorly. The interactions between these molecules coordinate to form a robustly local Krüppel expression domain. Significantly, remarkable alterations in gap gene expression, together with the secondary pair-rule genes, were observed in Krüppel mutant flies, indicating a central role played by Krüppel in normal embryonic fly morphogenesis [16].

3. Kruppel-like Factors in Vertebrates

The KLF family of proteins are transcription factors that play diverse biological roles in cell differentiation, cell cycle regulation, cellular metabolism, embryonic development, and apoptosis by regulating specific target genes [13,14,17,18,19,20]. KLF belongs to a large group of specificity protein 1 (Sp1)/KLF zinc finger-containing transcription factors (Figure 1A) [14]. These family molecules contain 81 amino acid triple-C2H2 zinc finger DNA-binding domains (DBD) separated by two highly conserved linker sequences that share high similarities to the Drosophila Krüppel protein (Figure 1B) [9,21,22]. As the zinc finger motifs existing in the Sp1 class function as a DBD, biochemical binding studies have shown that they have similar affinities for different guanine-cytosine-rich sequences [23,24]. Concurrently, depending on the specific amino acid residues positioned at the zinc finger DBD, these molecules also show a preference for recognizing the unique alignment of guanine-rich sequences [25,26,27]. After the identification of KLF1 (erythroid KLF; EKLF), which was the first zinc finger molecule to contain high similarities to Sp1, many KLFs have been identified in various vertebrate species [27,28]. Importantly, Sp1 clearly differed from KLFs in the presence of the buttonhead (Btd) box domain just 5′ of the KLF-DBD (Figure 1) [29,30,31]. The Btd box domain is a unique alignment present in the Drosophila buttonhead protein (Figure 1B) [30,32]. There are currently 17 KLF family molecules in humans and mice based on the following criteria: (1) the molecule contains a KLF-type DBD without the Btd motif and (2) the presence of only three zinc fingers (Figure 1). As described above, the KLF family plays diverse physiological roles, including skeletal development and maintenance, which are the main focus of this review. Accumulating evidence has also demonstrated that KLFs play crucial roles in the etiology and progression of many mammalian diseases [20]. These include hematopoietic disorders, cardiovascular diseases, gastrointestinal disorders, and bone-related disorders.

4. Roles of KLFs in Normal Bone Physiology (Table 1)

4.1. KLF1

KLF1 was initially called erythroid KLF (EKLF) because of its crucial role in red cell development and fetal-to-adult hemoglobin switching [33]. KLF1 has been shown to be specifically expressed in erythroblasts in the fetal liver and adult bone marrow [34,35]. Heterozygous KLF1 has been identified in patients with congenital dyserythropoietic anemia type IV [36,37]. These patients display growth defects; however, this is considered a secondary effect of malnutrition and not cell-autonomous effects of KLF1 in bone cells. It has also been reported that some patients with congenital dyserythropoietic anemia type I show congenital skeletal defects, such as missing digits; however, this condition is not caused by KLF1 mutation [38,39]. To date, there is no evidence of the cell-autonomous involvement of KLF1 in bone cell development or maintenance.

4.2. KLF2

KLF2 is closely related to KLF1 and is necessary for β-globin gene expression [40]. In contrast to KLF1, which is exclusively expressed in erythroid cells, KLF2 is dynamically expressed in various tissues, including skeletal cells, during embryogenesis [41]. KLF2 knockout animals were embryonic lethal around embryonic day 13 and showed severe abdominal hemorrhage accompanied by abnormal craniofacial morphogenesis [42].
In osteoblasts, KLF2 has been shown to enhance differentiation by upregulating Runx2, which is known as one of the master regulators of osteoblast differentiation [43,44]. Osteogenic MC3T3-E1 cell differentiation and mineralization were promoted by KLF2 by increasing Runx2 mRNA and protein levels. In contrast, KLF2 has been shown to negatively regulate RANKL-induced osteoclast differentiation of monocytes in vitro [45]. As conventional Klf2 knockout animals are embryonically lethal during the mid-developmental stage, the conclusive positive in vivo function of Klf2 in skeletal maintenance needs further investigation.

4.3. KLF3

KLF3 inhibition by mutated lncRNA Reg1cp results in human high bone mass syndrome [46]. Reg1cp binds directly to KLF3 and inhibits its activity in vitro. In vivo, mutant Reg1cp increased the formation of CD31hiEmcnhi endothelium in the bone marrow by repressing KLF3 transcriptional activity. This was strongly evidenced by the increased number of CD31hiEmcnhi vessels and increased bone formation in endothelial cell-specific Klf3 knockout mice [46]. These endothelial cells appear to couple angiogenesis and osteogenesis, leading to increased bone mass. Bone marrow stromal cell-derived exosomal miR-21-5p can enhance the proliferation and differentiation of osteoblasts [47]. miR-21-5p in exosomes can directly bind to the 3′-UTR of KLF3 and downregulate its expression, leading to increased osteoblast activity. Another miRNA, miR-20a-5p, has been shown to promote adipogenic differentiation of murine bone marrow stromal cells by targeting KLF3 [48]. Although the development of promising therapeutic strategies targeting miRNAs is still necessary, the regulation of KLF3 activity in bone marrow stromal cells might be one of the ways to increase bone mass while repressing adipogenesis. A number of cofactors have been identified in skeletal and nonskeletal cells for KLF3 functioning as a transcriptional repressor (Figure 2) [49,50,51].

4.4. KLF4

KLF4 is expressed in a tissue-restricted manner and is expressed in osteoblasts [52]. Klf4 conventional knockout mice are perinatally lethal [53]. Kim et al. used an osteoblast-specific Col1a1-Cre deleter strain [54] to generate conditional Klf4 knockout mice to examine bone phenotypes [55]. They found that osteoblast-specific KLF4 knockout mice showed markedly increased bone mass, despite increased osteoclast differentiation. Overexpression of KLF4 in osteoblasts attenuated osteoblast differentiation and mineralization in vitro [55,56]; at the same time, KLF4 in osteoblasts attenuated osteoclast differentiation in a coculture system of bone marrow cells and osteoblasts [55,57]. Repressed osteoclast maturation was also observed in transgenic mice expressing KLF4, specifically in osteoblasts [57]. KLF4 is physically and functionally associated with Runx2 (Figure 2), which implies that KLF4 functions in a bone-specific manner, although its wide expression has been observed in numerous tissues. KLF4 seems to regulate osteoblast function in numerous ways, including (1) physical and functional association with the master regulator of osteogenesis Runx2 [55,56], (2) blocking vitamin D receptor (VDR) transcriptional activity [55], (3) affecting ciliary-initiated Hedgehog signaling [58], and (4) modulating the expression of the xenobiotic nuclear receptor [59]. Although KLF4 is expressed in osteoclasts at a moderate level, KLF4 does not show cell-autonomous effects on osteoclast differentiation [55]. Significantly, when SP7-cre was used to target osteoprogenitors to delete KLF4 under conditional heterozygotic conditions, these mice showed fewer osteoblasts, reduced bone formation, and significantly smaller body size [60]. These observations indicate that KLF4 functions in skeletal cells in a stage-dependent manner to regulate the differentiation status of osteoblasts and indirectly regulates osteoclast differentiation in a non-cell-autonomous manner to fine-tune the developmental status of the entire skeleton. KLF4 physically and functionally associates with various molecules to play these context- and tissue-dependent roles for normal skeletal development (Figure 2) [61,62,63,64,65].
KLF4 regulates dentinogenesis, which is the formation of another hard tissue (dentin) in the body. KLF4 promotes odontoblastic differentiation of dental papilla mesenchymal cells through the regulation of dentin matrix protein 1 (DMP1) and modulation of transforming growth factor beta TGF-β signaling pathways [66,67,68].

4.5. KLF5

KLF5 is a ubiquitously expressed molecule that is predominantly expressed in the skin, digestive tract, kidneys, and urinary bladder [69]. Klf5 conventional knockout mice died very early in utero before E8.5, indicating that Klf5 plays an indispensable role in early embryogenesis [70]. In bone cells, KLF5 is expressed in developing chondrocytes and osteoblasts, but is not detected in osteoclasts [71]. Klf5+/− mice showed retarded skeletal growth during the perinatal period, but caught up during the early postnatal period. Cartilage matrix degradation was severely impaired, causing delayed primary ossification center formation in the long bones [71]. The possible role of KLF5 in osteoblasts has not been investigated in detail. There is a study suggesting non-cell-autonomous roles of KLF5 in osteoclastogenesis [72]. Numerous cofactors have been identified in a cell- and tissue-context manner that partly explain the indispensable roles played by KLF5 (Figure 2) [73,74,75,76,77,78].

4.6. KLF6

KLF6 is a ubiquitously expressed molecule that regulates growth arrest and functions as a tumor suppressor [79]. Gene knockout of Klf6 is embryonically lethal around E12 due to reduced hematopoiesis and poor yolk sac vascularization [80]. The physiological role of KLF6 in bone cells has not yet been investigated in detail.

4.7. KLF7

It has been reported that KLF7 is lowly expressed in people with high bone mineral density [81]. By comparing the gene expression profiles of normal and aged senescent human osteoblasts, Chen et al. identified miRNA targeting KLF7 as one of the genes significantly upregulated in aged osteoblasts [82]. However, the roles of KLF7 have not yet been determined. In contrast, osteoclast differentiation was enhanced by KLF7. Mechanistically, KLF7 directly represses heme oxygenase-1 (HO-1) expression, which is known as a negative regulator of osteoclast differentiation [81,83,84,85].

4.8. KLF8

KLF8 is a ubiquitously expressed molecule, but there have been no reports regarding its physiological effects on bone cells, both in vivo and in vitro. However, KLF8 plays a critical role in oncogenic and epithelial–mesenchymal transformation in various human tumors, including osteosarcoma, the most common and aggressive primary bone tumor [86,87,88,89,90]. This point is further discussed in a later section.

4.9. KLF9

Similar to KLF8, KLF9 has thus far not been reported to be involved in bone development and physiology. KLF9 has been shown to be intimately involved in bone pathology, including osteoarthritis and osteosarcoma [91,92,93,94,95]. This is further discussed in a later section.

4.10. KLF10

KLF10 was initially identified as an early-response gene in human osteoblasts following TGF-β treatment [96]. Based on this observation, KLF10 was initially named TGF beta inducible early-response gene 1 (TIEG1). KLF10 has important roles in bone biology by modulating a number of crucial signaling pathways, including TGF-β, bone morphogenetic protein (BMP), and estrogen-mediated pathways [97,98,99,100,101]. The indispensable roles played by KLF10 during skeletal development have been underscored by the skeletal phenotype observed in Klf10 knockout mice [102]. Klf10 knockout mice show growth defects in body length, delayed long bone growth, and delayed primary ossification center formation [102,103]. Chondrocyte maturation in the epiphyseal growth plate is also suppressed in long bones. Indian hedgehog (Ihh) expression in Klf10 knockout mice growth plates was severely reduced, which potentially altered the PTHrP-Ihh negative feedback loop [102]. Klf10 knockout mice show female-specific defects in bone strength and microarchitecture [103,104]. The osteopenic bone phenotype of Klf10 knockout mice was rescued by sclerostin antibody treatment [105]. In humans, single-nucleotide polymorphisms (SNPs) in KLF10 are associated with decreased volumetric bone mineral density (BMD) at the femoral neck in men [106]. In vitro studies have shown that KLF10 controls the expression and function of Runx2 and Sp7 (osterix) in osteoblasts [107,108]. In contrast, KLF10 also regulates osteoclast differentiation and survival by mediating NFATc1, Akt serine/threonine kinase (AKT), and extracellular signal-regulated kinases (ERK) pathways, and loss of KLF10 reduces NFATc1 activation, which slows the rate of osteoclast differentiation [109]. In summary, KLF10 displays pleiotropic cell- and non-cell-autonomous roles in skeletal cell differentiation, and numerous cofactors have been identified (Figure 2) [110,111].
Figure 2. Physical and functional association partner molecules of KLF proteins intimately involved in differentiation of skeletal cells. KLF3 [49,50,51], KLF4 [55,56,61,62,63,64], KLF5 [73,74,75,76,77], and KLF10 [110,111] interacting proteins are shown in the right box of each molecule.
Figure 2. Physical and functional association partner molecules of KLF proteins intimately involved in differentiation of skeletal cells. KLF3 [49,50,51], KLF4 [55,56,61,62,63,64], KLF5 [73,74,75,76,77], and KLF10 [110,111] interacting proteins are shown in the right box of each molecule.
Ijms 23 15174 g002

4.11. KLF11

Similar to KLF10, KLF11 was initially termed as TIEG2 because of its induction after TGF-β stimulation [112]. KLF11 promotes the effects of TGF-β on cell growth control in vitro by influencing the Smad-mediated pathway, which leads to the regulation of apoptosis and cell cycle arrest [113]. Klf11 knockout mice have been generated, but these mice showed no growth, developmental, or hematopoietic defects [114]. A few reports have shown the possible involvement of KLF11 in bone pathologies; these are discussed in a later section.
Table 1. In vivo skeletal phenotypes caused by loss (LOF) or gain (GOF) of function of KLFs.
Table 1. In vivo skeletal phenotypes caused by loss (LOF) or gain (GOF) of function of KLFs.
FactorsPhenotypesStudies
KLF1Growth defects (LOF in human; unlikely cell-autonomous actions)[36,37,38,39]
KLF2Abnormal craniofacial morphogenesis (LOF in mouse)[42]
KLF3High bone mass (LOF in human; by mutated lncRNA)[46]
KLF4High bone mass with increased osteoclast differentiation (osteoblast-specific LOF in mouse)
Repressed osteoclast maturation (osteoblast-specific GOF in mouse)
[55]
[57]
KLF5Retarded skeletal growth with delayed primary ossification center formation (LOF in mouse)[71]
KLF6Remain to be investigated
KLF7High bone mineral density associated with its low expression (in human)[81]
KLF8Remain to be investigated
KLF9Remain to be investigated
KLF10Growth defect with suppressed cartilage maturation and primary ossification center formation[102,103]
KLF11None[114]
KLF12Remain to be investigated (in vitro studies exist)
KLF13Remain to be investigated (in vitro studies exist)
KLF14Remain to be investigated (in vitro studies exist)
KLF15Remain to be investigated (in vitro studies exist)
KLF16Remain to be investigated (in vitro studies exist)
KLF17Remain to be investigated

4.12. KLF12

Only a few reports have shown that KLF12 affects skeletal cell differentiation. Human umbilical cord blood-derived mesenchymal stem cells (hUC-MSCs) can potentially become osteoblasts in vitro. Zhao et al. showed that the long intergenic non-protein-coding gene LINC02381 weakens the osteogenic differentiation of hUC-MSCs through the induction of KLF12 [115]. Significantly, KLF12 was able to repress transcription of Wnt4, which has been shown to prevent skeletal aging and pathological inflammation [116].

4.13. KLF13

KLF13 is ubiquitously expressed in many tissues, but a particularly strong expression is observed in the lymph nodes, thymus, and heart [117]. Lavallee et al. showed that KLF13 is indispensable for cardiac development and is functionally associated with cardiac GATA factors, which are cardinal molecules for normal cardiogenesis [118]. In bone cells, gene expression profiling after dexamethasone stimulation of osteoblastic MC3T3E1 cell line showed that KLF13 expression was significantly induced, but its functional significance has not been elucidated [119].

4.14. KLF14

KLF14 is a maternally expressed imprinted transcription factor [120], and is known to be located at the gene locus related to multiple metabolic disease phenotypes [121]. In human bone marrow stromal cells (hBMSCs), KLF14 is downregulated after osteogenic induction [122]. KLF14 overexpression in hBMSCs induced cell cycle arrest and inhibited osteogenic differentiation. In contrast, inhibiting KLF14 expression activated Wnt3a expression, which activates osteogenic gene expression and differentiation [122].

4.15. KLF15

Decreased bone formation is responsible for the pathogenesis of glucocorticoid-induced osteoporosis [123]. Recent evidence has shown that glucocorticoid administration enhances KLF15 expression, which leads to a reduction in osteoblast function [124]. In addition to the negative effect of KLF15 on bone cells, KLF15 enhances adipogenesis by regulating adipogenic gene expression, including peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT-enhancer-binding protein (C/EBPα) [125,126]. KLF15 has also been shown to enhance chondrogenesis of human MSCs in vitro by enhancing Sox9 expression [127].

4.16. KLF16

KLF16 is a ubiquitously expressed molecule, including in the bone marrow, but is known to be highly expressed in the brain tissue [128]. Only a limited number of reports regarding the effect of KLF16 on bone cells have been published. Jang et al. recently reported that KLF16 is downregulated during adipogenic differentiation and negatively regulates adipogenesis by targeting PPARγ [129].

4.17. KLF17

KLF17 is exclusively expressed in male testicular cells [130], and no studies on cell-autonomous effects on bone cells have been reported to date.

5. Role of KLFs in Osteoarthritis, Osteoporosis, and Osteosarcoma

5.1. Osteoarthritis

Osteoarthritis (OA) is a multifactorial, inflammatory, age-related disease characterized by cartilage destruction. OA is the most common form of joint disease, resulting in chronic pain and disability [131]. Progressive destruction of articular cartilage is observed due to disequilibrium between anabolism and catabolism of the cartilage extracellular matrix (ECM) [132]. Evidence showing the involvement of KLFs in OA pathologies is rapidly increasing (Table 2). In 2017, Yuan et al. showed that KLF2 is downregulated in human OA chondrocytes [133]. In an in vitro study using IL-1β to mimic OA cartilage, they showed that KLF2 represses matrix metalloproteinase 13 (MMP13) expression induced by IL-1β. Soon after, several KLFs were shown to be downregulated in human OA cartilage through genome-wide or candidate approaches [134,135,136,137,138]. These include KLF2, KLF4, KLF5, KLF9, KLF10, KLF11, and KLF15. KLF2 suppresses cartilage matrix destruction by decreasing the production of reactive oxygen species (ROS) [139]. KLF4 mediates the protective effects of simvastatin on articular chondrocytes [140]. KLF4 strongly induces cartilage collagen genes and proteoglycan-4 (PRG4) [140,141] and represses the expression of catabolic genes [141]. The efficacy of KLF2 and KLF4 induction was also demonstrated by the reduced severity of OA-associated symptoms in a mouse OA model [141]. Activation of the TGF-β pathway or inactivation of the Janus kinase 2 (JAK2)/signal transducer and activator of the transcription 3 (STAT3) pathway by KLF4 have also been suggested as mechanisms for the amelioration of OA symptoms [138,142].
KLF5 has been shown to be downregulated in human OA samples [136,141]; however, the exact role played by KLF5 in OA pathogenesis is still controversial. Shinoda et al. showed that KLF5 causes cartilage degradation through the strong induction of MMP9 [71]. The authors used the surgical OA model in Klf5+/− mice, which, however, did not show any difference when compared to the control mice [71]. Early growth response 1 (EGR1), which is highly expressed in OA cartilage and known to accelerate cartilage degradation, activates KLF5 activity by preventing KLF5 degradation [136]. Contrarily, KLF5 has been shown to protect against chondrocyte damage caused by IL-1b in certain contexts [143]. Kawata et al. also reported that KLF5 was downregulated in human OA samples [141].
The role of KLF9 in OA is still not well understood, but Zheng et al. reported that KLF9 may be related to the suppression of immune cell infiltration [95].
KLF10 and KLF11 are downregulated in human OA samples [134]. However, there are conflicting reports showing that KLF10 and KLF11 are increased in human OA samples. Zheng et al. showed that KLF10 was upregulated in cartilaginous tissue of OA patients as well as in the cartilage of an OA mouse model [144]. KLF10 overexpression in human chondrocytes inhibits proliferation and migration, suggesting possible negative effects on articular chondrocyte maintenance. Han et al. showed that KLF11 is highly expressed in the cartilage tissue of OA patients and in IL-1b-induced human chondrocytes [145].
KLF15 was significantly downregulated in chondrocytes from OA patients [134,135]. An in vitro study has shown that TNF-α acutely represses KLF15 expression in chondrocytes, and KLF15 attenuates the expression of MMP3 induced by TNF-α stimulation [135].

5.2. Osteoporosis

Osteoporosis is a systemic chronic pathology that requires long-term management after disease onset. Most osteoporotic patients are elderly; however, similar pathological conditions can be observed in patients undergoing long-term therapeutic glucocorticoid treatment (glucocorticoid-induced osteoporosis—GIO) [123]. The skeletons of patients show low bone mass and microstructural bone quality [146]. This deterioration causes osteoporosis patients to suffer from a high incidence of fragility fractures [147,148]. In severe cases, treatment with bisphosphonates, denosumab, teriparatide, or abaloparatide is essential to prevent femoral or vertebral fractures [146,149]. However, because of several limitations, as well as the high cost of these medicines [146], it is important to identify novel druggable molecular or cellular targets to tackle this difficult disease. Recent evidence has shown that several KLF family molecules are intimately involved in the onset or progression of osteoporosis, which is supported by both human and animal studies (Table 2).
The KLF1 E325R mutation is associated with congenital dyserythropoietic anemia, which causes severe hemolytic anemia in patients [150,151]. Patients with hemolytic anemia are known to have a significantly high risk of developing early onset osteopenia [152], and an increased risk of nonvertebral osteoporotic fractures in elderly men [153]. As the cell-autonomous roles played by KLF1 in skeletal cells are still unknown, these observations could be due to the nonskeletal cell-autonomous roles played by KLF1.
KLF3 inhibition by the mutated lncRNA Reg1cp has been shown to cause human high bone mass syndrome [46]. The mutant Reg1cp directly binds to KLF3 and inhibits its activity. Animal studies have shown that vascular endothelial-specific Klf3 knockout (KO) mice have increased bone mass, similar to that observed in humans [46]. An intimate relationship between angiogenesis and osteogenesis has been shown earlier [154,155,156] and dysregulation of vasculature is associated with many bone diseases, including osteoporosis [156]. A natural compound, ophiopogonin D, has been identified as a KLF3 inhibitor, and administration of this compound to wild-type animals increased bone mass, raising the intriguing possibility of KLF3 as a therapeutic target [46].
In vivo observations showed that KLF4 plays a crucial role in bone homeostasis in a stage-dependent manner. When KLF4 was conditionally deleted in osteoprogenitor cells using the Sp7-Cre deletion strain [157], mice displayed reduced osteoblastogenesis, resulting in low bone mass and reduced growth even under heterozygotic conditions [60]. However, when Klf4 was conditionally deleted in osteoblasts using the Col1a1-Cre strain [54], this strain showed the opposite phenotype, resulting in markedly high bone mass [55]. Klf4 is known as one of the factors for cellular reprogramming [158], but is considered a strong cell cycle repressor [159]. Klf4 has multifaceted functions, depending on numerous contexts. To further understand the stage-dependent functions of Klf4 in osteoblast-lineage cells, it is important to investigate skeletal phenotypes using other Cre deleter strains such as Ocn-Cre [160], Dmp1-Cre [161], Prrx1-Cre [162], Dermo1-Cre [163,164], and Lepr-Cre strains [165].
The KLF10 gene was found to be significantly reduced in osteoporotic patients [166]. Another quantitative trait locus (QTL) study showed that the KLF10 locus is associated with volumetric cortical bone mineral density [106]. In ovariectomized rats, KLF10 expression is reduced in the femoral tissue [167]. Consistent with these observations, KlfKO mice show an osteopenic phenotype, resulting in reduced bone strength [103]. However, these skeletal phenotypes are only observed in females [103,104]. The causes of the gender-specific phenotypes observed in Klf10 knockout mice are still unclear. KLF10 is known to be induced by estrogen receptor-mediated signals; this action may, at least partly, underlie the gender-specific phenotypes [168]. As described above, several genes in osteoblast-lineage cells have been proposed as targets of the effects of estrogens, in which KLF10 is included [101]. Previous reports have shown that the effects of estrogens and androgens on bones are largely caused by their influences on the generation and lifespan of osteoblasts, osteocytes, and osteoclasts. The gender-specific differences of the Klf10 knockout mice phenotypes could be examined further from these points.
KLF15 is one of the target genes of glucocorticoid receptor signaling and is intimately involved in tissue adipogenesis [169]. Recently, KLF15 was shown to induce GIO by enhancing marrow adipogenesis [126]. Tanshinol, an aqueous polyphenol isolated from Saliva miltorrhiza Bunge [170], exerts protective effects against GIO via unknown mechanisms [171,172]. Recent reports have identified that tanshinol prevents glucocorticoid-induced bone loss by blocking KLF15 activation [124,126].

5.3. Bone Tumors

KLF expression and function are altered in a large number of human cancers, and KLFs regulate cancer cell proliferation, apoptosis, metastasis, the tumor microenvironment, and cancer stem cells [173]. Individual KLFs can have either exclusive or redundant functions. Furthermore, several molecular switches that completely change the functions of KLFs in cancer have been identified [174,175,176,177,178]. The context-dependent functions of KLFs remain to be explored. In this section, we summarize the involvement of KLFs in osteosarcoma, which is a major primary bone malignant tumor with a poor prognosis (Table 2).
Osteosarcoma (OS) is the most common malignant bone tumor, and is thought to be derived from bone-forming mesenchymal stem cells. OS accounts for almost 20% of all primary malignant bone tumors worldwide [179,180]. Several KLFs have been reported to be intimately involved in OS proliferation, migration, invasion, and metastasis, as well as OS cancer stem cell regulation.
KLF2 expression is frequently diminished in various malignancies and is considered a tumor suppressor. KLF2 plays a role in growth inhibition and has proapoptotic and antiangiogenic roles. Silencing of KLF2 occurs in OS cells through transcriptional repression mediated by the polycomb group protein enhancer of zeste homolog 2 (EZH2) [181]. The recently discovered circular RNA CircLRP6 was also shown to promote OS progression through the inhibition of KLF2 [182].
KLF4 expression is highly associated with human OS cancer stemness [183]. Overexpression of KLF4 in OS cells increased sphere-forming potential, enhanced stemness gene expression, enhanced chemotherapy resistance, and increased metastatic potential [183]. Adriamycin treatment is known to cause chemotherapy resistance, owing to the enhanced stemness phenotype in OS cells [184]. OS cells expressing high levels of KLF4 accumulated after adriamycin treatment and were considered a key factor for adriamycin-induced stemness. Simvastatin administration significantly reduced KLF4 expression and prevented the adriamycin-induced enhancement of metastasis and cancer stemness in OS cells [184].
KLF8 and KLF11 have also been shown to be involved in OS cancer stem cell induction [185,186]. KLF8 is highly expressed in OS tissues and highly enriched in CD133-positive cancer stem cell populations [185]. This was shown to be caused by KLF8 activating Sox2 transcription via downregulation of miR-429 [185]. The oncogenic roles of KLF8 have been shown in other studies that showed the involvement of KLF8 in epithelial–mesenchymal transition (EMT) processes [86,88,89,187] and chemotherapy resistance [188,189]. KLF11 has previously been shown to play a crucial negative role in cancer stem cells in non-OS tumors [190]. Through genome-wide screening, Wang et al. recently identified KLF11 as a top-hit suppressor molecule in OS cancer stem cell induction [186]. Mechanistically, KLF11 was associated with yes-associated protein (YAP)/TEA domain family member 1 (TEAD) at adjacent DNA sites, where KLF11 suppressed YAP/TEAD functions by recruiting the SIN3 transcription regulator family member A (SIN3A)/histone deacetylase (HDAC) repressor complex [186]. KLF11 was epigenetically silenced in OS cancer stem cells (CSCs), causing sustained YAP/TEAD activation. KLF11 can be activated in OS cells by thiazolidinedione, a drug that is already used for the treatment of type II diabetes [191].
KLF5 is highly expressed in human OS cells [192]. KLF5 appears to promote OS cell proliferation, invasion, and metastasis via inducing miR-487a [192]. Significantly, a small molecule inhibitor of KLF5, ML264, could induce cell cycle arrest, migration, and invasive capabilities of OS cells [193].
KLF6 is considered a tumor suppressor in various tumors and acts by upregulating p21 and downregulating B-cell lymphoma 2 (BCL-2) [194]. KLF6 is downregulated in OS cells, and KLF6 represses OS cell proliferation and invasion in vitro [194].
KLF9 is downregulated in human OS tissues and is known to block progression of OS cells [94]. Repression of KLF9 in OS cells are induced by several microRNAs and CircRNAs [91,93].
Table 2. Studies of KLFs in bone pathologies.
Table 2. Studies of KLFs in bone pathologies.
FactorsConclusionStudies
OsteoarthritisKLF2


KLF4

KLF5
KLF15
Downregulated in OA patients
Represses IL1β-induced MMP13 expression
Suppresses ROS production
Downregulated in human OA samples
Induces PRG4 and collagen genes
Downregulated in human OA samples
Downregulated in human OA samples
[133,134,135,136,137,138,139]


[140,141]

[136,141]
[135,136]
OsteoporosisKLF3
KLF4

KLF10


Endothelial cell-specific KO mice show high bone mass
Osteoblast-specific KO mice show high bone mass
Osteoprogenitor-specific KO mice show low bone mass
Downregulated in osteoporotic patients
Downregulated in ovariectomy rats
Female Klf10 KO mice show reduced bone strength
[46]
[54,60]

[103,166,167]


OsteosarcomaKLF2
KLF4
KLF5

KLF6
KLF8

KLF9
KLF11

Downregulated in OS cells in vitro
Induces cancer stemness
Highly expressed in OS cells
ML264 (KLF5 inhibitor) induces OS cell activation
Downregulated in OS cells
Highly expressed in OS samples
Induces cancer stemness
Downregulated in OS cells
Downregulated in OS samples
Suppresses cancer stem cell induction
[181]
[183,184]
[192,193]

[194]
[185,186]

[194]
[186,191]

6. Future Perspectives

The understanding of the role of KLF family molecules has deepened during this decade. However, many aspects need to be investigated further to understand how each member of the KLF protein family regulates gene expression in a cell- and tissue-specific manner. In particular, less-studied KLF family molecules are yet to be fully explored. This goal could be partly achieved by expanding the investigation of cofactors of each KLF protein in various cell types (Figure 2). It is important to reveal the transcriptional complex in which each KLF participates to obtain the overall picture to classify it as a KLF protein complex. This information will enable researchers to predict whether family proteins possess synergistic or antagonistic properties. To introduce KLF as a therapeutic target for specific bone pathologies, we need to understand the functional redundancies of each KLF in disease onset or progression. Finally, as some KLF knockout animals are embryonic or perinatally lethal, the possible roles played by KLFs in human diseases need to be carefully analyzed.

Author Contributions

Conceptualization, M.A. and S.O.; writing—original draft preparation, M.A., N.S., Y.I. and S.O.; writing—review and editing, M.A. and S.O.; funding acquisition, M.A. and S.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Grants-in-Aid for Science Research from the Japan Society for the Promotion of Science (JSPS) (22K06295 to M.A. and 21H03142 to S.O.), Takeda Science Foundation, Koyanagi Foundation, and Uehara Memorial Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge Mariko Kasai for administrative support and Chizuko Inui-Yamamoto for technical support.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Long, F. Building strong bones: Molecular regulation of the osteoblast lineage. Nat. Rev. Mol. Cell Biol. 2011, 13, 27–38. [Google Scholar] [CrossRef] [PubMed]
  2. Le Douarin, N.M.; Smith, J. Development of the peripheral nervous system from the neural crest. Annu. Rev. Cell Biol. 1988, 4, 375–404. [Google Scholar] [CrossRef] [PubMed]
  3. Berendsen, A.D.; Olsen, B.R. Bone development. Bone 2015, 80, 14–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Salhotra, A.; Shah, H.N.; Levi, B.; Longaker, M.T. Mechanisms of bone development and repair. Nat. Rev. Mol. Cell Biol. 2020, 21, 696–711. [Google Scholar] [CrossRef]
  5. Chan, C.K.; Seo, E.Y.; Chen, J.Y.; Lo, D.; McArdle, A.; Sinha, R.; Tevlin, R.; Seita, J.; Vincent-Tompkins, J.; Wearda, T.; et al. Identification and specification of the mouse skeletal stem cell. Cell 2015, 160, 285–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chan, C.K.F.; Gulati, G.S.; Sinha, R.; Tompkins, J.V.; Lopez, M.; Carter, A.C.; Ransom, R.C.; Reinisch, A.; Wearda, T.; Murphy, M.; et al. Identification of the Human Skeletal Stem Cell. Cell 2018, 175, 43–56.e21. [Google Scholar] [CrossRef] [Green Version]
  7. Schuh, R.; Aicher, W.; Gaul, U.; Cote, S.; Preiss, A.; Maier, D.; Seifert, E.; Nauber, U.; Schroder, C.; Kemler, R.; et al. A conserved family of nuclear proteins containing structural elements of the finger protein encoded by Kruppel, a Drosophila segmentation gene. Cell 1986, 47, 1025–1032. [Google Scholar] [CrossRef] [Green Version]
  8. Knipple, D.C.; Seifert, E.; Rosenberg, U.B.; Preiss, A.; Jackle, H. Spatial and temporal patterns of Kruppel gene expression in early Drosophila embryos. Nature 1985, 317, 40–44. [Google Scholar] [CrossRef]
  9. Preiss, A.; Rosenberg, U.B.; Kienlin, A.; Seifert, E.; Jackle, H. Molecular genetics of Kruppel, a gene required for segmentation of the Drosophila embryo. Nature 1985, 313, 27–32. [Google Scholar] [CrossRef]
  10. Rosenberg, U.B.; Preiss, A.; Seifert, E.; Jackle, H.; Knipple, D.C. Production of phenocopies by Kruppel antisense RNA injection into Drosophila embryos. Nature 1985, 313, 703–706. [Google Scholar] [CrossRef]
  11. Kinzler, K.W.; Ruppert, J.M.; Bigner, S.H.; Vogelstein, B. The GLI gene is a member of the Kruppel family of zinc finger proteins. Nature 1988, 332, 371–374. [Google Scholar] [CrossRef] [PubMed]
  12. Ruppert, J.M.; Kinzler, K.W.; Wong, A.J.; Bigner, S.H.; Kao, F.T.; Law, M.L.; Seuanez, H.N.; O’Brien, S.J.; Vogelstein, B. The GLI-Kruppel family of human genes. Mol. Cell Biol. 1988, 8, 3104–3113. [Google Scholar] [CrossRef]
  13. Dang, D.T.; Pevsner, J.; Yang, V.W. The biology of the mammalian Kruppel-like family of transcription factors. Int. J. Biochem. Cell Biol. 2000, 32, 1103–1121. [Google Scholar] [CrossRef] [PubMed]
  14. Kaczynski, J.; Cook, T.; Urrutia, R. Sp1- and Kruppel-like transcription factors. Genome Biol. 2003, 4, 206. [Google Scholar] [CrossRef] [Green Version]
  15. Nusslein-Volhard, C.; Wieschaus, E. Mutations affecting segment number and polarity in Drosophila. Nature 1980, 287, 795–801. [Google Scholar] [CrossRef] [PubMed]
  16. Kozlov, K.; Surkova, S.; Myasnikova, E.; Reinitz, J.; Samsonova, M. Modeling of gap gene expression in Drosophila Kruppel mutants. PLoS Comput. Biol. 2012, 8, e1002635. [Google Scholar] [CrossRef]
  17. Bieker, J.J. Kruppel-like factors: Three fingers in many pies. J. Biol. Chem. 2001, 276, 34355–34358. [Google Scholar] [CrossRef] [Green Version]
  18. Lomberk, G.; Urrutia, R. The family feud: Turning off Sp1 by Sp1-like KLF proteins. Biochem. J. 2005, 392, 1–11. [Google Scholar] [CrossRef] [Green Version]
  19. McConnell, B.B.; Ghaleb, A.M.; Nandan, M.O.; Yang, V.W. The diverse functions of Kruppel-like factors 4 and 5 in epithelial biology and pathobiology. BioEssays News Rev. Mol. Cell Dev. Biol. 2007, 29, 549–557. [Google Scholar] [CrossRef] [Green Version]
  20. McConnell, B.B.; Yang, V.W. Mammalian Kruppel-like factors in health and diseases. Physiol. Rev. 2010, 90, 1337–1381. [Google Scholar] [CrossRef] [Green Version]
  21. Iuchi, S. Three classes of C2H2 zinc finger proteins. Cell Mol. Life Sci. CMLS 2001, 58, 625–635. [Google Scholar] [CrossRef] [PubMed]
  22. Presnell, J.S.; Schnitzler, C.E.; Browne, W.E. KLF/SP Transcription Factor Family Evolution: Expansion, Diversification, and Innovation in Eukaryotes. Genome Biol. Evol. 2015, 7, 2289–2309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Briggs, M.R.; Kadonaga, J.T.; Bell, S.P.; Tjian, R. Purification and biochemical characterization of the promoter-specific transcription factor, Sp1. Science 1986, 234, 47–52. [Google Scholar] [CrossRef]
  24. Kadonaga, J.T.; Carner, K.R.; Masiarz, F.R.; Tjian, R. Isolation of cDNA encoding transcription factor Sp1 and functional analysis of the DNA binding domain. Cell 1987, 51, 1079–1090. [Google Scholar] [CrossRef] [PubMed]
  25. Kingsley, C.; Winoto, A. Cloning of GT box-binding proteins: A novel Sp1 multigene family regulating T-cell receptor gene expression. Mol. Cell Biol. 1992, 12, 4251–4261. [Google Scholar] [CrossRef] [PubMed]
  26. Pesce, M.; Marin Gomez, M.; Philipsen, S.; Scholer, H.R. Binding of Sp1 and Sp3 transcription factors to the Oct-4 gene promoter. Cell Mol. Biol. 1999, 45, 709–716. [Google Scholar]
  27. Suske, G. The Sp-family of transcription factors. Gene 1999, 238, 291–300. [Google Scholar] [CrossRef]
  28. Cook, T.; Gebelein, B.; Urrutia, R. Sp1 and its likes: Biochemical and functional predictions for a growing family of zinc finger transcription factors. Ann. N. Y. Acad. Sci. 1999, 880, 94–102. [Google Scholar] [CrossRef]
  29. Hardeman, J. The First Permanent Molar and Its Early Management. Am. J. Dent. Sci. 1884, 18, 59–63. [Google Scholar]
  30. Wimmer, E.A.; Jackle, H.; Pfeifle, C.; Cohen, S.M. A Drosophila homologue of human Sp1 is a head-specific segmentation gene. Nature 1993, 366, 690–694. [Google Scholar] [CrossRef]
  31. Suske, G.; Bruford, E.; Philipsen, S. Mammalian SP/KLF transcription factors: Bring in the family. Genomics 2005, 85, 551–556. [Google Scholar] [CrossRef] [PubMed]
  32. Wimmer, E.A.; Frommer, G.; Purnell, B.A.; Jackle, H. buttonhead and D-Sp1: A novel Drosophila gene pair. Mech. Dev. 1996, 59, 53–62. [Google Scholar] [CrossRef] [PubMed]
  33. Borg, J.; Papadopoulos, P.; Georgitsi, M.; Gutierrez, L.; Grech, G.; Fanis, P.; Phylactides, M.; Verkerk, A.J.; van der Spek, P.J.; Scerri, C.A.; et al. Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin. Nat. Genet. 2010, 42, 801–805. [Google Scholar] [CrossRef] [PubMed]
  34. Nuez, B.; Michalovich, D.; Bygrave, A.; Ploemacher, R.; Grosveld, F. Defective haematopoiesis in fetal liver resulting from inactivation of the EKLF gene. Nature 1995, 375, 316–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Perkins, A.C.; Sharpe, A.H.; Orkin, S.H. Lethal beta-thalassaemia in mice lacking the erythroid CACCC-transcription factor EKLF. Nature 1995, 375, 318–322. [Google Scholar] [CrossRef]
  36. Wickramasinghe, S.N.; Illum, N.; Wimberley, P.D. Congenital dyserythropoietic anaemia with novel intra-erythroblastic and intra-erythrocytic inclusions. Br. J. Haematol. 1991, 79, 322–330. [Google Scholar] [CrossRef]
  37. Arnaud, L.; Saison, C.; Helias, V.; Lucien, N.; Steschenko, D.; Giarratana, M.C.; Prehu, C.; Foliguet, B.; Montout, L.; de Brevern, A.G.; et al. A dominant mutation in the gene encoding the erythroid transcription factor KLF1 causes a congenital dyserythropoietic anemia. Am. J. Hum. Genet. 2010, 87, 721–727. [Google Scholar] [CrossRef] [Green Version]
  38. Tamary, H.; Dgany, O.; Proust, A.; Krasnov, T.; Avidan, N.; Eidelitz-Markus, T.; Tchernia, G.; Genevieve, D.; Cormier-Daire, V.; Bader-Meunier, B.; et al. Clinical and molecular variability in congenital dyserythropoietic anaemia type I. Br. J. Haematol. 2005, 130, 628–634. [Google Scholar] [CrossRef]
  39. Heimpel, H.; Schwarz, K.; Ebnother, M.; Goede, J.S.; Heydrich, D.; Kamp, T.; Plaumann, L.; Rath, B.; Roessler, J.; Schildknecht, O.; et al. Congenital dyserythropoietic anemia type I (CDA I): Molecular genetics, clinical appearance, and prognosis based on long-term observation. Blood 2006, 107, 334–340. [Google Scholar] [CrossRef]
  40. Tallack, M.R.; Perkins, A.C. Three fingers on the switch: Kruppel-like factor 1 regulation of gamma-globin to beta-globin gene switching. Curr. Opin. Hematol. 2013, 20, 193–200. [Google Scholar] [CrossRef]
  41. Wani, M.A.; Conkright, M.D.; Jeffries, S.; Hughes, M.J.; Lingrel, J.B. cDNA isolation, genomic structure, regulation, and chromosomal localization of human lung Kruppel-like factor. Genomics 1999, 60, 78–86. [Google Scholar] [CrossRef] [PubMed]
  42. Wani, M.A.; Means, R.T., Jr.; Lingrel, J.B. Loss of LKLF function results in embryonic lethality in mice. Transgenic Res. 1998, 7, 229–238. [Google Scholar] [CrossRef] [PubMed]
  43. Hou, Z.; Wang, Z.; Tao, Y.; Bai, J.; Yu, B.; Shen, J.; Sun, H.; Xiao, L.; Xu, Y.; Zhou, J.; et al. KLF2 regulates osteoblast differentiation by targeting of Runx2. Lab. Investig. A J. Tech. Methods Pathol. 2019, 99, 271–280. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, I.; Kim, J.H.; Kim, K.; Seong, S.; Kim, N. The IRF2BP2-KLF2 axis regulates osteoclast and osteoblast differentiation. BMB Rep. 2019, 52, 469–474. [Google Scholar] [CrossRef]
  45. Das, H.; Kumar, A.; Lin, Z.; Patino, W.D.; Hwang, P.M.; Feinberg, M.W.; Majumder, P.K.; Jain, M.K. Kruppel-like factor 2 (KLF2) regulates proinflammatory activation of monocytes. Proc. Natl. Acad. Sci. USA 2006, 103, 6653–6658. [Google Scholar] [CrossRef] [Green Version]
  46. Yang, M.; Guo, Q.; Peng, H.; Xiao, Y.Z.; Xiao, Y.; Huang, Y.; Li, C.J.; Su, T.; Zhang, Y.L.; Lei, M.X.; et al. Kruppel-like factor 3 inhibition by mutated lncRNA Reg1cp results in human high bone mass syndrome. J. Exp. Med. 2019, 216, 1944–1964. [Google Scholar] [CrossRef] [PubMed]
  47. You, M.; Ai, Z.; Zeng, J.; Fu, Y.; Zhang, L.; Wu, X. Bone mesenchymal stem cells (BMSCs)-derived exosomal microRNA-21-5p regulates Kruppel-like factor 3 (KLF3) to promote osteoblast proliferation in vitro. Bioengineered 2022, 13, 11933–11944. [Google Scholar] [CrossRef]
  48. Zhu, E.; Zhang, J.; Zhou, J.; Yuan, H.; Zhao, W.; Wang, B. miR-20a-5p promotes adipogenic differentiation of murine bone marrow stromal cells via targeting Kruppel-like factor 3. J. Mol. Endocrinol. 2018, 60, 225–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Turner, J.; Crossley, M. Cloning and characterization of mCtBP2, a co-repressor that associates with basic Kruppel-like factor and other mammalian transcriptional regulators. EMBO J. 1998, 17, 5129–5140. [Google Scholar] [CrossRef] [Green Version]
  50. Shi, Y.; Sawada, J.; Sui, G.; Affar el, B.; Whetstine, J.R.; Lan, F.; Ogawa, H.; Luke, M.P.; Nakatani, Y.; Shi, Y. Coordinated histone modifications mediated by a CtBP co-repressor complex. Nature 2003, 422, 735–738. [Google Scholar] [CrossRef]
  51. Turner, J.; Nicholas, H.; Bishop, D.; Matthews, J.M.; Crossley, M. The LIM protein FHL3 binds basic Kruppel-like factor/Kruppel-like factor 3 and its co-repressor C-terminal-binding protein 2. J. Biol. Chem. 2003, 278, 12786–12795. [Google Scholar] [CrossRef] [Green Version]
  52. Shields, J.M.; Christy, R.J.; Yang, V.W. Identification and characterization of a gene encoding a gut-enriched Kruppel-like factor expressed during growth arrest. J. Biol. Chem. 1996, 271, 20009–20017. [Google Scholar] [CrossRef] [PubMed]
  53. Segre, J.A.; Bauer, C.; Fuchs, E. Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat. Genet. 1999, 22, 356–360. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, J.E.; Nakashima, K.; de Crombrugghe, B. Transgenic mice expressing a ligand-inducible cre recombinase in osteoblasts and odontoblasts: A new tool to examine physiology and disease of postnatal bone and tooth. Am. J. Pathol. 2004, 165, 1875–1882. [Google Scholar] [CrossRef] [PubMed]
  55. Kim, J.H.; Kim, K.; Youn, B.U.; Lee, J.; Kim, I.; Shin, H.I.; Akiyama, H.; Choi, Y.; Kim, N. Kruppel-like factor 4 attenuates osteoblast formation, function, and cross talk with osteoclasts. J. Cell Biol. 2014, 204, 1063–1074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Michikami, I.; Fukushi, T.; Tanaka, M.; Egusa, H.; Maeda, Y.; Ooshima, T.; Wakisaka, S.; Abe, M. Kruppel-like factor 4 regulates membranous and endochondral ossification. Exp. Cell Res. 2012, 318, 311–325. [Google Scholar] [CrossRef]
  57. Fujikawa, J.; Tanaka, M.; Itoh, S.; Fukushi, T.; Kurisu, K.; Takeuchi, Y.; Morisaki, I.; Wakisaka, S.; Abe, M. Kruppel-like factor 4 expression in osteoblasts represses osteoblast-dependent osteoclast maturation. Cell Tissue Res. 2014, 358, 177–187. [Google Scholar] [CrossRef]
  58. Takeuchi, Y.; Kito, A.; Itoh, S.; Naruse, H.; Fujikawa, J.; Sadek, K.M.; Akiyama, S.; Yamashiro, T.; Wakisaka, S.; Abe, M. Kruppel-Like Factor 4 represses osteoblast differentiation via ciliary Hedgehog signaling. Exp. Cell Res. 2018, 371, 417–425. [Google Scholar] [CrossRef]
  59. Saeki, N.; Itoh, Y.; Kanai, R.; Itoh, S.; Inububishi, T.; Akiyama, S.; Inui-Yamamoto, C.; Abe, M. Pregnane X receptor (PXR) represses osteoblast differentiation through repression of the Hedgehog signaling pathway. Exp. Cell Res. 2022, 416, 113156. [Google Scholar] [CrossRef]
  60. Yu, S.; Guo, J.; Sun, Z.; Lin, C.; Tao, H.; Zhang, Q.; Cui, Y.; Zuo, H.; Lin, Y.; Chen, S.; et al. BMP2-dependent gene regulatory network analysis reveals Klf4 as a novel transcription factor of osteoblast differentiation. Cell Death Dis. 2021, 12, 197. [Google Scholar] [CrossRef]
  61. Zhang, W.; Geiman, D.E.; Shields, J.M.; Dang, D.T.; Mahatan, C.S.; Kaestner, K.H.; Biggs, J.R.; Kraft, A.S.; Yang, V.W. The gut-enriched Kruppel-like factor (Kruppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J. Biol. Chem. 2000, 275, 18391–18398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Feinberg, M.W.; Cao, Z.; Wara, A.K.; Lebedeva, M.A.; Senbanerjee, S.; Jain, M.K. Kruppel-like factor 4 is a mediator of proinflammatory signaling in macrophages. J. Biol. Chem. 2005, 280, 38247–38258. [Google Scholar] [CrossRef]
  63. Liu, Y.; Sinha, S.; McDonald, O.G.; Shang, Y.; Hoofnagle, M.H.; Owens, G.K. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J. Biol. Chem. 2005, 280, 9719–9727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yoshida, T.; Gan, Q.; Owens, G.K. Kruppel-like factor 4, Elk-1, and histone deacetylases cooperatively suppress smooth muscle cell differentiation markers in response to oxidized phospholipids. Am. J. Physiol. Cell Physiol. 2008, 295, C1175–C1182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Yoshida, T.; Yamashita, M.; Horimai, C.; Hayashi, M. Deletion of Kruppel-like factor 4 in endothelial and hematopoietic cells enhances neointimal formation following vascular injury. J. Am. Heart Assoc. 2014, 3, e000622. [Google Scholar] [CrossRef]
  66. Lin, H.; Liu, H.; Sun, Q.; Yuan, G.; Zhang, L.; Chen, Z. KLF4 promoted odontoblastic differentiation of mouse dental papilla cells via regulation of DMP1. J. Cell. Physiol. 2013, 228, 2076–2085. [Google Scholar] [CrossRef]
  67. Liu, H.; Lin, H.; Zhang, L.; Sun, Q.; Yuan, G.; Zhang, L.; Chen, S.; Chen, Z. miR-145 and miR-143 regulate odontoblast differentiation through targeting Klf4 and Osx genes in a feedback loop. J. Biol. Chem. 2013, 288, 9261–9271. [Google Scholar] [CrossRef] [Green Version]
  68. Tao, H.; Lin, H.; Sun, Z.; Pei, F.; Zhang, J.; Chen, S.; Liu, H.; Chen, Z. Klf4 Promotes Dentinogenesis and Odontoblastic Differentiation via Modulation of TGF-beta Signaling Pathway and Interaction With Histone Acetylation. J. Bone Miner. Res. 2019, 34, 1502–1516. [Google Scholar] [CrossRef]
  69. Luo, Y.; Chen, C. The roles and regulation of the KLF5 transcription factor in cancers. Cancer Sci. 2021, 112, 2097–2117. [Google Scholar] [CrossRef]
  70. Shindo, T.; Manabe, I.; Fukushima, Y.; Tobe, K.; Aizawa, K.; Miyamoto, S.; Kawai-Kowase, K.; Moriyama, N.; Imai, Y.; Kawakami, H.; et al. Kruppel-like zinc-finger transcription factor KLF5/BTEB2 is a target for angiotensin II signaling and an essential regulator of cardiovascular remodeling. Nat. Med. 2002, 8, 856–863. [Google Scholar] [CrossRef]
  71. Shinoda, Y.; Ogata, N.; Higashikawa, A.; Manabe, I.; Shindo, T.; Yamada, T.; Kugimiya, F.; Ikeda, T.; Kawamura, N.; Kawasaki, Y.; et al. Kruppel-like factor 5 causes cartilage degradation through transactivation of matrix metalloproteinase 9. J. Biol. Chem. 2008, 283, 24682–24689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, B.; Li, Y.; Wu, Q.; Xie, L.; Barwick, B.; Fu, C.; Li, X.; Wu, D.; Xia, S.; Chen, J.; et al. Acetylation of KLF5 maintains EMT and tumorigenicity to cause chemoresistant bone metastasis in prostate cancer. Nat. Commun. 2021, 12, 1714. [Google Scholar] [CrossRef]
  73. Guo, P.; Dong, X.Y.; Zhao, K.; Sun, X.; Li, Q.; Dong, J.T. Opposing effects of KLF5 on the transcription of MYC in epithelial proliferation in the context of transforming growth factor beta. J. Biol. Chem. 2009, 284, 28243–28252. [Google Scholar] [CrossRef] [Green Version]
  74. Du, J.X.; Hagos, E.G.; Nandan, M.O.; Bialkowska, A.B.; Yu, B.; Yang, V.W. The E3 ubiquitin ligase SMAD ubiquitination regulatory factor 2 negatively regulates Kruppel-like factor 5 protein. J. Biol. Chem. 2011, 286, 40354–40364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nakajima, Y.; Akaogi, K.; Suzuki, T.; Osakabe, A.; Yamaguchi, C.; Sunahara, N.; Ishida, J.; Kako, K.; Ogawa, S.; Fujimura, T.; et al. Estrogen regulates tumor growth through a nonclassical pathway that includes the transcription factors ERbeta and KLF5. Sci. Signal. 2011, 4, ra22. [Google Scholar] [CrossRef] [PubMed]
  76. Gao, Y.; Shi, Q.; Xu, S.; Du, C.; Liang, L.; Wu, K.; Wang, K.; Wang, X.; Chang, L.S.; He, D.; et al. Curcumin promotes KLF5 proteasome degradation through downregulating YAP/TAZ in bladder cancer cells. Int. J. Mol. Sci. 2014, 15, 15173–15187. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, C.; Nie, Z.; Zhou, Z.; Zhang, H.; Liu, R.; Wu, J.; Qin, J.; Ma, Y.; Chen, L.; Li, S.; et al. The interplay between TEAD4 and KLF5 promotes breast cancer partially through inhibiting the transcription of p27Kip1. Oncotarget 2015, 6, 17685–17697. [Google Scholar] [CrossRef] [Green Version]
  78. Jia, J.; Zhang, H.B.; Shi, Q.; Yang, C.; Ma, J.B.; Jin, B.; Wang, X.; He, D.; Guo, P. KLF5 downregulation desensitizes castration-resistant prostate cancer cells to docetaxel by increasing BECN1 expression and inducing cell autophagy. Theranostics 2019, 9, 5464–5477. [Google Scholar] [CrossRef]
  79. Narla, G.; Heath, K.E.; Reeves, H.L.; Li, D.; Giono, L.E.; Kimmelman, A.C.; Glucksman, M.J.; Narla, J.; Eng, F.J.; Chan, A.M.; et al. KLF6, a candidate tumor suppressor gene mutated in prostate cancer. Science 2001, 294, 2563–2566. [Google Scholar] [CrossRef]
  80. Matsumoto, N.; Kubo, A.; Liu, H.; Akita, K.; Laub, F.; Ramirez, F.; Keller, G.; Friedman, S.L. Developmental regulation of yolk sac hematopoiesis by Kruppel-like factor 6. Blood 2006, 107, 1357–1365. [Google Scholar] [CrossRef] [Green Version]
  81. Chen, C.; Hu, F.; Miao, S.; Sun, L.; Jiao, Y.; Xu, M.; Huang, X.; Yang, Y.; Zhou, R. Transcription Factor KLF7 Promotes Osteoclast Differentiation by Suppressing HO-1. Front. Genet. 2022, 13, 798433. [Google Scholar] [CrossRef]
  82. Chen, Y.J.; Chang, W.A.; Huang, M.S.; Chen, C.H.; Wang, K.Y.; Hsu, Y.L.; Kuo, P.L. Identification of novel genes in aging osteoblasts using next-generation sequencing and bioinformatics. Oncotarget 2017, 8, 113598–113613. [Google Scholar] [CrossRef] [PubMed]
  83. Ke, K.; Safder, M.A.; Sul, O.J.; Kim, W.K.; Suh, J.H.; Joe, Y.; Chung, H.T.; Choi, H.S. Hemeoxygenase-1 maintains bone mass via attenuating a redox imbalance in osteoclast. Mol. Cell Endocrinol. 2015, 409, 11–20. [Google Scholar] [CrossRef]
  84. Florczyk-Soluch, U.; Jozefczuk, E.; Stepniewski, J.; Bukowska-Strakova, K.; Mendel, M.; Viscardi, M.; Nowak, W.N.; Jozkowicz, A.; Dulak, J. Various roles of heme oxygenase-1 in response of bone marrow macrophages to RANKL and in the early stage of osteoclastogenesis. Sci. Rep. 2018, 8, 10797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Han, D.; Gu, X.; Gao, J.; Wang, Z.; Liu, G.; Barkema, H.W.; Han, B. Chlorogenic acid promotes the Nrf2/HO-1 anti-oxidative pathway by activating p21(Waf1/Cip1) to resist dexamethasone-induced apoptosis in osteoblastic cells. Free Radic. Biol. Med. 2019, 137, 1–12. [Google Scholar] [CrossRef] [PubMed]
  86. Wang, X.; Zhao, J. KLF8 transcription factor participates in oncogenic transformation. Oncogene 2007, 26, 456–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lu, H.; Hu, L.; Li, T.; Lahiri, S.; Shen, C.; Wason, M.S.; Mukherjee, D.; Xie, H.; Yu, L.; Zhao, J. A novel role of Kruppel-like factor 8 in DNA repair in breast cancer cells. J. Biol. Chem. 2012, 287, 43720–43729. [Google Scholar] [CrossRef] [Green Version]
  88. Zhang, H.; Liu, L.; Wang, Y.; Zhao, G.; Xie, R.; Liu, C.; Xiao, X.; Wu, K.; Nie, Y.; Zhang, H.; et al. KLF8 involves in TGF-beta-induced EMT and promotes invasion and migration in gastric cancer cells. J. Cancer Res. Clin. Oncol. 2013, 139, 1033–1042. [Google Scholar] [CrossRef]
  89. Lu, H.; Hu, L.; Yu, L.; Wang, X.; Urvalek, A.M.; Li, T.; Shen, C.; Mukherjee, D.; Lahiri, S.K.; Wason, M.S.; et al. KLF8 and FAK cooperatively enrich the active MMP14 on the cell surface required for the metastatic progression of breast cancer. Oncogene 2014, 33, 2909–2917. [Google Scholar] [CrossRef] [Green Version]
  90. Kumar, S.; Behera, A.; Saha, P.; Kumar Srivastava, A. The role of Kruppel-like factor 8 in cancer biology: Current research and its clinical relevance. Biochem. Pharmacol. 2021, 183, 114351. [Google Scholar] [CrossRef]
  91. Jin, Y.; Yang, L.; Li, X. MicroRNA-652 promotes cell proliferation and osteosarcoma invasion by directly targeting KLF9. Exp. Ther. Med. 2020, 20, 2953–2960. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, Y.; Zhu, T.; He, F.; Chen, A.C.; Yang, H.; Zhu, X. Identification of Key Genes and Pathways in Osteoarthritis via Bioinformatic Tools: An Updated Analysis. Cartilage 2021, 13, 1457S–1464S. [Google Scholar] [CrossRef] [PubMed]
  93. Chen, Q.; Zhou, H.; Rong, W. Circular RNA_0078767 upregulates Kruppel-like factor 9 expression by targeting microRNA-889, thereby inhibiting the progression of osteosarcoma. Bioengineered 2022, 13, 14313–14328. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, Y.; Han, K.; Cao, Y.; Hu, Y.; Shao, Z.; Tong, W.; Han, Y.; Liu, Y. KLF9 regulates miR-338-3p/NRCAM axis to block the progression of osteosarcoma cells. J. Cancer 2022, 13, 2029–2039. [Google Scholar] [CrossRef]
  95. Zhang, J.; Zhang, S.; Zhou, Y.; Qu, Y.; Hou, T.; Ge, W.; Zhang, S. KLF9 and EPYC acting as feature genes for osteoarthritis and their association with immune infiltration. J. Orthop. Surg. Res. 2022, 17, 365. [Google Scholar] [CrossRef]
  96. Subramaniam, M.; Harris, S.A.; Oursler, M.J.; Rasmussen, K.; Riggs, B.L.; Spelsberg, T.C. Identification of a novel TGF-beta-regulated gene encoding a putative zinc finger protein in human osteoblasts. Nucleic Acids Res. 1995, 23, 4907–4912. [Google Scholar] [CrossRef] [Green Version]
  97. Hefferan, T.E.; Reinholz, G.G.; Rickard, D.J.; Johnsen, S.A.; Waters, K.M.; Subramaniam, M.; Spelsberg, T.C. Overexpression of a nuclear protein, TIEG, mimics transforming growth factor-beta action in human osteoblast cells. J. Biol. Chem. 2000, 275, 20255–20259. [Google Scholar] [CrossRef] [Green Version]
  98. Johnsen, S.A.; Subramaniam, M.; Janknecht, R.; Spelsberg, T.C. TGFbeta inducible early gene enhances TGFbeta/Smad-dependent transcriptional responses. Oncogene 2002, 21, 5783–5790. [Google Scholar] [CrossRef] [Green Version]
  99. Johnsen, S.A.; Subramaniam, M.; Katagiri, T.; Janknecht, R.; Spelsberg, T.C. Transcriptional regulation of Smad2 is required for enhancement of TGFbeta/Smad signaling by TGFbeta inducible early gene. J. Cell. Biochem. 2002, 87, 233–241. [Google Scholar] [CrossRef]
  100. Johnsen, S.A.; Subramaniam, M.; Monroe, D.G.; Janknecht, R.; Spelsberg, T.C. Modulation of transforming growth factor beta (TGFbeta)/Smad transcriptional responses through targeted degradation of TGFbeta-inducible early gene-1 by human seven in absentia homologue. J. Biol. Chem. 2002, 277, 30754–30759. [Google Scholar] [CrossRef] [Green Version]
  101. Hawse, J.R.; Subramaniam, M.; Monroe, D.G.; Hemmingsen, A.H.; Ingle, J.N.; Khosla, S.; Oursler, M.J.; Spelsberg, T.C. Estrogen receptor beta isoform-specific induction of transforming growth factor beta-inducible early gene-1 in human osteoblast cells: An essential role for the activation function 1 domain. Mol. Endocrinol. 2008, 22, 1579–1595. [Google Scholar] [CrossRef]
  102. Lee, J.M.; Ko, J.Y.; Park, J.W.; Lee, W.K.; Song, S.U.; Im, G.I. KLF10 is a modulatory factor of chondrocyte hypertrophy in developing skeleton. J. Orthop. Res. 2020, 38, 1987–1995. [Google Scholar] [CrossRef] [PubMed]
  103. Hawse, J.R.; Iwaniec, U.T.; Bensamoun, S.F.; Monroe, D.G.; Peters, K.D.; Ilharreborde, B.; Rajamannan, N.M.; Oursler, M.J.; Turner, R.T.; Spelsberg, T.C.; et al. TIEG-null mice display an osteopenic gender-specific phenotype. Bone 2008, 42, 1025–1031. [Google Scholar] [CrossRef] [Green Version]
  104. Bensamoun, S.F.; Hawse, J.R.; Subramaniam, M.; Ilharreborde, B.; Bassillais, A.; Benhamou, C.L.; Fraser, D.G.; Oursler, M.J.; Amadio, P.C.; An, K.N.; et al. TGFbeta inducible early gene-1 knockout mice display defects in bone strength and microarchitecture. Bone 2006, 39, 1244–1251. [Google Scholar] [CrossRef] [PubMed]
  105. Gingery, A.; Subramaniam, M.; Pitel, K.S.; Li, X.; Ke, H.Z.; Turner, R.T.; Iwaniec, U.T.; Hawse, J.R. Sclerostin antibody treatment rescues the osteopenic bone phenotype of TGFbeta inducible early gene-1 knockout female mice. J. Cell Physiol. 2020, 235, 5679–5688. [Google Scholar] [CrossRef]
  106. Yerges, L.M.; Klei, L.; Cauley, J.A.; Roeder, K.; Kammerer, C.M.; Ensrud, K.E.; Nestlerode, C.S.; Lewis, C.; Lang, T.F.; Barrett-Connor, E.; et al. Candidate gene analysis of femoral neck trabecular and cortical volumetric bone mineral density in older men. J. Bone Miner. Res. 2010, 25, 330–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Hawse, J.R.; Cicek, M.; Grygo, S.B.; Bruinsma, E.S.; Rajamannan, N.M.; van Wijnen, A.J.; Lian, J.B.; Stein, G.S.; Oursler, M.J.; Subramaniam, M.; et al. TIEG1/KLF10 modulates Runx2 expression and activity in osteoblasts. PLoS ONE 2011, 6, e19429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Subramaniam, M.; Pitel, K.S.; Withers, S.G.; Drissi, H.; Hawse, J.R. TIEG1 enhances Osterix expression and mediates its induction by TGFbeta and BMP2 in osteoblasts. Biochem. Biophys. Res. Commun. 2016, 470, 528–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Cicek, M.; Vrabel, A.; Sturchio, C.; Pederson, L.; Hawse, J.R.; Subramaniam, M.; Spelsberg, T.C.; Oursler, M.J. TGF-beta inducible early gene 1 regulates osteoclast differentiation and survival by mediating the NFATc1, AKT, and MEK/ERK signaling pathways. PLoS ONE 2011, 6, e17522. [Google Scholar] [CrossRef] [Green Version]
  110. Xiong, Y.; Svingen, P.A.; Sarmento, O.O.; Smyrk, T.C.; Dave, M.; Khanna, S.; Lomberk, G.A.; Urrutia, R.A.; Faubion, W.A., Jr. Differential coupling of KLF10 to Sin3-HDAC and PCAF regulates the inducibility of the FOXP3 gene. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2014, 307, R608–R620. [Google Scholar] [CrossRef] [Green Version]
  111. Lin, C.H.; Lin, S.Y.; Chang, H.W.; Ko, L.J.; Tseng, Y.S.; Chang, V.H.; Yu, W.C. CDK2 phosphorylation regulates the protein stability of KLF10 by interfering with binding of the E3 ligase SIAH1. Biochim. Et Biophys. Acta 2015, 1853, 1174–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Cook, T.; Gebelein, B.; Mesa, K.; Mladek, A.; Urrutia, R. Molecular cloning and characterization of TIEG2 reveals a new subfamily of transforming growth factor-beta-inducible Sp1-like zinc finger-encoding genes involved in the regulation of cell growth. J. Biol. Chem. 1998, 273, 25929–25936. [Google Scholar] [CrossRef] [PubMed]
  113. Lin, L.; Mahner, S.; Jeschke, U.; Hester, A. The Distinct Roles of Transcriptional Factor KLF11 in Normal Cell Growth Regulation and Cancer as a Mediator of TGF-beta Signaling Pathway. Int. J. Mol. Sci. 2020, 21, 2928. [Google Scholar] [CrossRef] [PubMed]
  114. Song, C.Z.; Gavriilidis, G.; Asano, H.; Stamatoyannopoulos, G. Functional study of transcription factor KLF11 by targeted gene inactivation. Blood Cells Mol. Dis. 2005, 34, 53–59. [Google Scholar] [CrossRef] [Green Version]
  115. Zhao, G.; Luo, W.D.; Yuan, Y.; Lin, F.; Guo, L.M.; Ma, J.J.; Chen, H.B.; Tang, H.; Shu, J. LINC02381, a sponge of miR-21, weakens osteogenic differentiation of hUC-MSCs through KLF12-mediated Wnt4 transcriptional repression. J. Bone Miner. Metab. 2022, 40, 66–80. [Google Scholar] [CrossRef]
  116. Yu, B.; Chang, J.; Liu, Y.; Li, J.; Kevork, K.; Al-Hezaimi, K.; Graves, D.T.; Park, N.H.; Wang, C.Y. Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-kappaB. Nat. Med. 2014, 20, 1009–1017. [Google Scholar] [CrossRef] [Green Version]
  117. Song, A.; Chen, Y.F.; Thamatrakoln, K.; Storm, T.A.; Krensky, A.M. RFLAT-1: A new zinc finger transcription factor that activates RANTES gene expression in T lymphocytes. Immunity 1999, 10, 93–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Lavallee, G.; Andelfinger, G.; Nadeau, M.; Lefebvre, C.; Nemer, G.; Horb, M.E.; Nemer, M. The Kruppel-like transcription factor KLF13 is a novel regulator of heart development. EMBO J. 2006, 25, 5201–5213. [Google Scholar] [CrossRef] [Green Version]
  119. Leclerc, N.; Luppen, C.A.; Ho, V.V.; Nagpal, S.; Hacia, J.G.; Smith, E.; Frenkel, B. Gene expression profiling of glucocorticoid-inhibited osteoblasts. J. Mol. Endocrinol. 2004, 33, 175–193. [Google Scholar] [CrossRef] [Green Version]
  120. Parker-Katiraee, L.; Carson, A.R.; Yamada, T.; Arnaud, P.; Feil, R.; Abu-Amero, S.N.; Moore, G.E.; Kaneda, M.; Perry, G.H.; Stone, A.C.; et al. Identification of the imprinted KLF14 transcription factor undergoing human-specific accelerated evolution. PLoS Genet. 2007, 3, e65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Small, K.S.; Hedman, A.K.; Grundberg, E.; Nica, A.C.; Thorleifsson, G.; Kong, A.; Thorsteindottir, U.; Shin, S.Y.; Richards, H.B.; Consortium, G.; et al. Identification of an imprinted master trans regulator at the KLF14 locus related to multiple metabolic phenotypes. Nat. Genet. 2011, 43, 561–564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Weng, J.; Wu, J.; Chen, W.; Fan, H.; Liu, H. KLF14 inhibits osteogenic differentiation of human bone marrow mesenchymal stem cells by downregulating WNT3A. Am. J. Transl. Res. 2020, 12, 4445–4455. [Google Scholar] [PubMed]
  123. Compston, J. Management of glucocorticoid-induced osteoporosis: What is new? Int. J. Rheum. Dis. 2019, 22, 1595–1597. [Google Scholar] [CrossRef] [PubMed]
  124. Yang, Y.; Su, Y.; Wang, D.; Chen, Y.; Liu, Y.; Luo, S.; Wu, T.; Cui, L. Tanshinol Rescues the Impaired Bone Formation Elicited by Glucocorticoid Involved in KLF15 Pathway. Oxidative Med. Cell Longev. 2016, 2016, 1092746. [Google Scholar] [CrossRef] [Green Version]
  125. Mori, T.; Sakaue, H.; Iguchi, H.; Gomi, H.; Okada, Y.; Takashima, Y.; Nakamura, K.; Nakamura, T.; Yamauchi, T.; Kubota, N.; et al. Role of Kruppel-like factor 15 (KLF15) in transcriptional regulation of adipogenesis. J. Biol. Chem. 2005, 280, 12867–12875. [Google Scholar] [CrossRef] [Green Version]
  126. Yang, Y.J.; Zhu, Z.; Wang, D.T.; Zhang, X.L.; Liu, Y.Y.; Lai, W.X.; Mo, Y.L.; Li, J.; Liang, Y.L.; Hu, Z.Q.; et al. Tanshinol alleviates impaired bone formation by inhibiting adipogenesis via KLF15/PPARgamma2 signaling in GIO rats. Acta Pharmacol. Sin. 2018, 39, 633–641. [Google Scholar] [CrossRef]
  127. Song, Z.; Lian, X.; Wang, Y.; Xiang, Y.; Li, G. KLF15 regulates in vitro chondrogenic differentiation of human mesenchymal stem cells by targeting SOX9. Biochem. Biophys. Res. Commun. 2017, 493, 1082–1088. [Google Scholar] [CrossRef]
  128. Hwang, C.K.; D’Souza, U.M.; Eisch, A.J.; Yajima, S.; Lammers, C.H.; Yang, Y.; Lee, S.H.; Kim, Y.M.; Nestler, E.J.; Mouradian, M.M. Dopamine receptor regulating factor, DRRF: A zinc finger transcription factor. Proc. Natl. Acad. Sci. USA 2001, 98, 7558–7563. [Google Scholar] [CrossRef] [Green Version]
  129. Jang, M.K.; Lee, S.; Jung, M.H. RNA-Seq Analysis Reveals a Negative Role of KLF16 in Adipogenesis. PLoS ONE 2016, 11, e0162238. [Google Scholar] [CrossRef] [Green Version]
  130. van Vliet, J.; Crofts, L.A.; Quinlan, K.G.; Czolij, R.; Perkins, A.C.; Crossley, M. Human KLF17 is a new member of the Sp/KLF family of transcription factors. Genomics 2006, 87, 474–482. [Google Scholar] [CrossRef] [Green Version]
  131. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef] [PubMed]
  132. Fellows, C.R.; Matta, C.; Mobasheri, A. Applying Proteomics to Study Crosstalk at the Cartilage-Subchondral Bone Interface in Osteoarthritis: Current Status and Future Directions. EBioMedicine 2016, 11, 2–4. [Google Scholar] [CrossRef] [PubMed]
  133. Yuan, Y.; Tan, H.; Dai, P. Kruppel-Like Factor 2 Regulates Degradation of Type II Collagen by Suppressing the Expression of Matrix Metalloproteinase (MMP)-13. Cell Physiol. Biochem. 2017, 42, 2159–2168. [Google Scholar] [CrossRef] [PubMed]
  134. Fisch, K.M.; Gamini, R.; Alvarez-Garcia, O.; Akagi, R.; Saito, M.; Muramatsu, Y.; Sasho, T.; Koziol, J.A.; Su, A.I.; Lotz, M.K. Identification of transcription factors responsible for dysregulated networks in human osteoarthritis cartilage by global gene expression analysis. Osteoarthr. Cartil. 2018, 26, 1531–1538. [Google Scholar] [CrossRef] [Green Version]
  135. Li, Y.; Zhao, M.; Xiao, W. KLF15 Regulates the Expression of MMP-3 in Human Chondrocytes. J. Interferon Cytokine Res. 2018, 38, 356–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Sun, X.; Huang, H.; Pan, X.; Li, S.; Xie, Z.; Ma, Y.; Hu, B.; Wang, J.; Chen, Z.; Shi, P. EGR1 promotes the cartilage degeneration and hypertrophy by activating the Kruppel-like factor 5 and beta-catenin signaling. Biochim. Et Biophys. Acta Mol. Basis Dis. 2019, 1865, 2490–2503. [Google Scholar] [CrossRef]
  137. Huang, Y.; Chen, D.; Yan, Z.; Zhan, J.; Xue, X.; Pan, X.; Yu, H. LncRNA MEG3 Protects Chondrocytes From IL-1beta-Induced Inflammation via Regulating miR-9-5p/KLF4 Axis. Front. Physiol. 2021, 12, 617654. [Google Scholar] [CrossRef]
  138. Li, J.; Jiang, M.; Xiong, C.; Pan, J.; Jia, S.; Zhang, Y.; Zhang, J.; Xu, N.; Zhou, X.; Huang, Y. KLF4, negatively regulated by miR-7, suppresses osteoarthritis development via activating TGF-beta1 signaling. Int. Immunopharmacol. 2022, 102, 108416. [Google Scholar] [CrossRef]
  139. Gao, X.; Jiang, S.; Du, Z.; Ke, A.; Liang, Q.; Li, X. KLF2 Protects against Osteoarthritis by Repressing Oxidative Response through Activation of Nrf2/ARE Signaling In Vitro and In Vivo. Oxidative Med. Cell Longev. 2019, 2019, 8564681. [Google Scholar] [CrossRef] [Green Version]
  140. Yu, S.M.; Kim, S.J. Kruppel-like factor 4 (KLF-4) plays a crucial role in simvastatin (SVT)-induced differentiation of rabbit articular chondrocytes. Biochem. Biophys. Res. Commun. 2018, 501, 814–819. [Google Scholar] [CrossRef]
  141. Kawata, M.; Teramura, T.; Ordoukhanian, P.; Head, S.R.; Natarajan, P.; Sundaresan, A.; Olmer, M.; Asahara, H.; Lotz, M.K. Kruppel-like factor-4 and Kruppel-like factor-2 are important regulators of joint tissue cells and protect against tissue destruction and inflammation in osteoarthritis. Ann. Rheum. Dis. 2022. [Google Scholar] [CrossRef] [PubMed]
  142. Liu, W.; Chen, Y.; Zeng, G.; Yang, T.; Song, W. INSR mediated by transcription factor KLF4 and DNA methylation ameliorates osteoarthritis progression via inactivation of JAK2/STAT3 signaling pathway. Am. J. Transl. Res. 2020, 12, 7953–7967. [Google Scholar]
  143. Lai, X.; Song, Y.; Tian, J. CircCDK14 ameliorates interleukin-1beta-induced chondrocyte damage by the miR-1183/KLF5 pathway in osteoarthritis. Autoimmunity 2022, 55, 408–417. [Google Scholar] [CrossRef] [PubMed]
  144. Zheng, L.; Lu, H.; Li, H.; Xu, X.; Wang, D. KLF10 is upregulated in osteoarthritis and inhibits chondrocyte proliferation and migration by upregulating Acvr1 and suppressing inhbb expression. Acta Histochem. 2020, 122, 151528. [Google Scholar] [CrossRef] [PubMed]
  145. Han, F.; Jiang, H.; Qu, W.; Rui, Y.J. KLF11 protects chondrocytes via inhibiting p38 MAPK signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 6505–6516. [Google Scholar] [CrossRef] [PubMed]
  146. Compston, J.E.; McClung, M.R.; Leslie, W.D. Osteoporosis. Lancet 2019, 393, 364–376. [Google Scholar] [CrossRef]
  147. Miller, P.D.; Barlas, S.; Brenneman, S.K.; Abbott, T.A.; Chen, Y.T.; Barrett-Connor, E.; Siris, E.S. An approach to identifying osteopenic women at increased short-term risk of fracture. Arch. Intern. Med. 2004, 164, 1113–1120. [Google Scholar] [CrossRef] [Green Version]
  148. Siris, E.S.; Chen, Y.T.; Abbott, T.A.; Barrett-Connor, E.; Miller, P.D.; Wehren, L.E.; Berger, M.L. Bone mineral density thresholds for pharmacological intervention to prevent fractures. Arch. Intern. Med. 2004, 164, 1108–1112. [Google Scholar] [CrossRef] [Green Version]
  149. Rachner, T.D.; Khosla, S.; Hofbauer, L.C. Osteoporosis: Now and the future. Lancet 2011, 377, 1276–1287. [Google Scholar] [CrossRef] [Green Version]
  150. Viprakasit, V.; Ekwattanakit, S.; Riolueang, S.; Chalaow, N.; Fisher, C.; Lower, K.; Kanno, H.; Tachavanich, K.; Bejrachandra, S.; Saipin, J.; et al. Mutations in Kruppel-like factor 1 cause transfusion-dependent hemolytic anemia and persistence of embryonic globin gene expression. Blood 2014, 123, 1586–1595. [Google Scholar] [CrossRef] [Green Version]
  151. Kulczynska-Figurny, K.; Bieker, J.J.; Siatecka, M. Severe anemia caused by dominant mutations in Kruppel-like factor 1 (KLF1). Mutat. Res. Rev. Mutat. Res. 2020, 786, 108336. [Google Scholar] [CrossRef] [PubMed]
  152. Shi, L.; Lin, C.L.; Su, C.H.; Lin, K.C.; Leong, K.H.; Wang, Y.T.; Kuo, C.F.; Tsai, S.Y. The Risk of Developing Osteoporosis in Hemolytic Anemia-What Aggravates the Bone Loss? J. Clin. Med. 2021, 10, 3364. [Google Scholar] [CrossRef] [PubMed]
  153. Kristjansdottir, H.L.; Mellstrom, D.; Johansson, P.; Karlsson, M.; Vandenput, L.; Lorentzon, M.; Herlitz, H.; Ohlsson, C.; Lerner, U.H.; Lewerin, C. Anemia is associated with increased risk of non-vertebral osteoporotic fractures in elderly men: The MrOS Sweden cohort. Arch. Osteoporos. 2022, 17, 85. [Google Scholar] [CrossRef] [PubMed]
  154. Ding, W.G.; Wei, Z.X.; Liu, J.B. Reduced local blood supply to the tibial metaphysis is associated with ovariectomy-induced osteoporosis in mice. Connect. Tissue Res. 2011, 52, 25–29. [Google Scholar] [CrossRef] [PubMed]
  155. Portal-Nunez, S.; Lozano, D.; Esbrit, P. Role of angiogenesis on bone formation. Histol. Histopathol. 2012, 27, 559–566. [Google Scholar] [CrossRef]
  156. Tong, X.; Chen, X.; Zhang, S.; Huang, M.; Shen, X.; Xu, J.; Zou, J. The Effect of Exercise on the Prevention of Osteoporosis and Bone Angiogenesis. BioMed Res. Int. 2019, 2019, 8171897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Rodda, S.J.; McMahon, A.P. Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors. Development 2006, 133, 3231–3244. [Google Scholar] [CrossRef] [Green Version]
  158. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
  159. Rowland, B.D.; Peeper, D.S. KLF4, p21 and context-dependent opposing forces in cancer. Nat. Rev. Cancer 2006, 6, 11–23. [Google Scholar] [CrossRef]
  160. Zhang, M.; Xuan, S.; Bouxsein, M.L.; von Stechow, D.; Akeno, N.; Faugere, M.C.; Malluche, H.; Zhao, G.; Rosen, C.J.; Efstratiadis, A.; et al. Osteoblast-specific knockout of the insulin-like growth factor (IGF) receptor gene reveals an essential role of IGF signaling in bone matrix mineralization. J. Biol. Chem. 2002, 277, 44005–44012. [Google Scholar] [CrossRef] [Green Version]
  161. Lu, Y.; Xie, Y.; Zhang, S.; Dusevich, V.; Bonewald, L.F.; Feng, J.Q. DMP1-targeted Cre expression in odontoblasts and osteocytes. J. Dent. Res. 2007, 86, 320–325. [Google Scholar] [CrossRef] [PubMed]
  162. Logan, M.; Martin, J.F.; Nagy, A.; Lobe, C.; Olson, E.N.; Tabin, C.J. Expression of Cre Recombinase in the developing mouse limb bud driven by a Prxl enhancer. Genesis 2002, 33, 77–80. [Google Scholar] [CrossRef] [PubMed]
  163. Sosic, D.; Richardson, J.A.; Yu, K.; Ornitz, D.M.; Olson, E.N. Twist regulates cytokine gene expression through a negative feedback loop that represses NF-kappaB activity. Cell 2003, 112, 169–180. [Google Scholar] [CrossRef] [PubMed]
  164. Yu, K.; Xu, J.; Liu, Z.; Sosic, D.; Shao, J.; Olson, E.N.; Towler, D.A.; Ornitz, D.M. Conditional inactivation of FGF receptor 2 reveals an essential role for FGF signaling in the regulation of osteoblast function and bone growth. Development 2003, 130, 3063–3074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. DeFalco, J.; Tomishima, M.; Liu, H.; Zhao, C.; Cai, X.; Marth, J.D.; Enquist, L.; Friedman, J.M. Virus-assisted mapping of neural inputs to a feeding center in the hypothalamus. Science 2001, 291, 2608–2613. [Google Scholar] [CrossRef]
  166. Hopwood, B.; Tsykin, A.; Findlay, D.M.; Fazzalari, N.L. Gene expression profile of the bone microenvironment in human fragility fracture bone. Bone 2009, 44, 87–101. [Google Scholar] [CrossRef]
  167. You, M.; Zhang, L.; Zhang, X.; Fu, Y.; Dong, X. MicroRNA-197-3p Inhibits the Osteogenic Differentiation in Osteoporosis by Down-Regulating KLF 10. Clin. Interv. Aging 2021, 16, 107–117. [Google Scholar] [CrossRef]
  168. Tau, K.R.; Hefferan, T.E.; Waters, K.M.; Robinson, J.A.; Subramaniam, M.; Riggs, B.L.; Spelsberg, T.C. Estrogen regulation of a transforming growth factor-beta inducible early gene that inhibits deoxyribonucleic acid synthesis in human osteoblasts. Endocrinology 1998, 139, 1346–1353. [Google Scholar] [CrossRef]
  169. Asada, M.; Rauch, A.; Shimizu, H.; Maruyama, H.; Miyaki, S.; Shibamori, M.; Kawasome, H.; Ishiyama, H.; Tuckermann, J.; Asahara, H. DNA binding-dependent glucocorticoid receptor activity promotes adipogenesis via Kruppel-like factor 15 gene expression. Lab. Investig. A J. Tech. Methods Pathol. 2011, 91, 203–215. [Google Scholar] [CrossRef] [Green Version]
  170. Lam, F.F.; Yeung, J.H.; Chan, K.M.; Or, P.M. Relaxant effects of danshen aqueous extract and its constituent danshensu on rat coronary artery are mediated by inhibition of calcium channels. Vasc. Pharmacol. 2007, 46, 271–277. [Google Scholar] [CrossRef]
  171. Cui, L.; Liu, Y.Y.; Wu, T.; Ai, C.M.; Chen, H.Q. Osteogenic effects of D+beta-3,4-dihydroxyphenyl lactic acid (salvianic acid A, SAA) on osteoblasts and bone marrow stromal cells of intact and prednisone-treated rats. Acta Pharmacol. Sin. 2009, 30, 321–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Luo, S.; Yang, Y.; Chen, J.; Zhong, Z.; Huang, H.; Zhang, J.; Cui, L. Tanshinol stimulates bone formation and attenuates dexamethasone-induced inhibition of osteogenesis in larval zebrafish. J. Orthop. Transl. 2016, 4, 35–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Tetreault, M.P.; Yang, Y.; Katz, J.P. Kruppel-like factors in cancer. Nat. Rev. Cancer 2013, 13, 701–713. [Google Scholar] [CrossRef] [PubMed]
  174. Fernandez-Zapico, M.E.; Mladek, A.; Ellenrieder, V.; Folch-Puy, E.; Miller, L.; Urrutia, R. An mSin3A interaction domain links the transcriptional activity of KLF11 with its role in growth regulation. EMBO J. 2003, 22, 4748–4758. [Google Scholar] [CrossRef] [Green Version]
  175. Rowland, B.D.; Bernards, R.; Peeper, D.S. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat. Cell Biol. 2005, 7, 1074–1082. [Google Scholar] [CrossRef]
  176. Guo, P.; Dong, X.Y.; Zhang, X.; Zhao, K.W.; Sun, X.; Li, Q.; Dong, J.T. Pro-proliferative factor KLF5 becomes anti-proliferative in epithelial homeostasis upon signaling-mediated modification. J. Biol. Chem. 2009, 284, 6071–6078. [Google Scholar] [CrossRef] [Green Version]
  177. Yang, Y.; Nakagawa, H.; Tetreault, M.P.; Billig, J.; Victor, N.; Goyal, A.; Sepulveda, A.R.; Katz, J.P. Loss of transcription factor KLF5 in the context of p53 ablation drives invasive progression of human squamous cell cancer. Cancer Res. 2011, 71, 6475–6484. [Google Scholar] [CrossRef] [Green Version]
  178. Yang, Y.; Tarapore, R.S.; Jarmel, M.H.; Tetreault, M.P.; Katz, J.P. p53 mutation alters the effect of the esophageal tumor suppressor KLF5 on keratinocyte proliferation. Cell Cycle 2012, 11, 4033–4039. [Google Scholar] [CrossRef] [Green Version]
  179. Messerschmitt, P.J.; Garcia, R.M.; Abdul-Karim, F.W.; Greenfield, E.M.; Getty, P.J. Osteosarcoma. J. Am. Acad. Orthop. Surg. 2009, 17, 515–527. [Google Scholar] [CrossRef]
  180. Raymond, A.K.; Jaffe, N. Osteosarcoma multidisciplinary approach to the management from the pathologist’s perspective. Cancer Treat. Res. 2009, 152, 63–84. [Google Scholar] [CrossRef]
  181. Taniguchi, H.; Jacinto, F.V.; Villanueva, A.; Fernandez, A.F.; Yamamoto, H.; Carmona, F.J.; Puertas, S.; Marquez, V.E.; Shinomura, Y.; Imai, K.; et al. Silencing of Kruppel-like factor 2 by the histone methyltransferase EZH2 in human cancer. Oncogene 2012, 31, 1988–1994. [Google Scholar] [CrossRef] [PubMed]
  182. Zheng, S.; Qian, Z.; Jiang, F.; Ge, D.; Tang, J.; Chen, H.; Yang, J.; Yao, Y.; Yan, J.; Zhao, L.; et al. CircRNA LRP6 promotes the development of osteosarcoma via negatively regulating KLF2 and APC levels. Am. J. Transl. Res. 2019, 11, 4126–4138. [Google Scholar] [PubMed]
  183. Qi, X.T.; Li, Y.L.; Zhang, Y.Q.; Xu, T.; Lu, B.; Fang, L.; Gao, J.Q.; Yu, L.S.; Zhu, D.F.; Yang, B.; et al. KLF4 functions as an oncogene in promoting cancer stem cell-like characteristics in osteosarcoma cells. Acta Pharmacol. Sin. 2019, 40, 546–555. [Google Scholar] [CrossRef] [PubMed]
  184. Li, Y.; Xian, M.; Yang, B.; Ying, M.; He, Q. Inhibition of KLF4 by Statins Reverses Adriamycin-Induced Metastasis and Cancer Stemness in Osteosarcoma Cells. Stem Cell Rep. 2017, 8, 1617–1629. [Google Scholar] [CrossRef] [Green Version]
  185. Zhang, L.; Yang, P.; Liu, Q.; Wang, J.; Yan, F.; Duan, L.; Lin, F. KLF8 promotes cancer stem cell-like phenotypes in osteosarcoma through miR-429-SOX2 signaling. Neoplasma 2020, 67, 519–527. [Google Scholar] [CrossRef] [Green Version]
  186. Wang, Y.; Wu, J.; Chen, H.; Yang, Y.; Xiao, C.; Yi, X.; Shi, C.; Zhong, K.; He, H.; Li, Y.; et al. Genome-wide CRISPR-Cas9 screen identified KLF11 as a druggable suppressor for sarcoma cancer stem cells. Sci. Adv. 2021, 7, eabe3445. [Google Scholar] [CrossRef]
  187. Lin, F.; Shen, Z.; Tang, L.N.; Zheng, S.E.; Sun, Y.J.; Min, D.L.; Yao, Y. KLF8 knockdown suppresses proliferation and invasion in human osteosarcoma cells. Mol. Med. Rep. 2014, 9, 1613–1617. [Google Scholar] [CrossRef] [Green Version]
  188. Wei, W.; Ji, L.; Duan, W.; Zhu, J. CircSAMD4A contributes to cell doxorubicin resistance in osteosarcoma by regulating the miR-218-5p/KLF8 axis. Open Life Sci. 2020, 15, 848–859. [Google Scholar] [CrossRef]
  189. Wang, B.; Yan, L.; Shi, W.; Xie, H.; Chen, R.; Shao, Y.; Liang, W. CircRNA PVT1 promotes proliferation and chemoresistance of osteosarcoma cells via the miR-24-3p/KLF8 axis. Int. J. Clin. Oncol. 2022, 27, 811–822. [Google Scholar] [CrossRef]
  190. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  191. Desouza, C.V.; Shivaswamy, V. Pioglitazone in the treatment of type 2 diabetes: Safety and efficacy review. Clin. Med. Insights. Endocrinol. Diabetes 2010, 3, 43–51. [Google Scholar] [CrossRef] [PubMed]
  192. Luo, A.; Liu, H.; Huang, C. KLF5-induced miR-487a augments the progression of osteosarcoma cells by targeting NKX3-1 in vitro. Oncol. Lett. 2022, 24, 258. [Google Scholar] [CrossRef] [PubMed]
  193. Huang, H.; Han, Y.; Chen, Z.; Pan, X.; Yuan, P.; Zhao, X.; Zhu, H.; Wang, J.; Sun, X.; Shi, P. ML264 inhibits osteosarcoma growth and metastasis via inhibition of JAK2/STAT3 and WNT/beta-catenin signalling pathways. J. Cell Mol. Med. 2020, 24, 5652–5664. [Google Scholar] [CrossRef]
  194. Jianwei, Z.; Enzhong, B.; Fan, L.; Jian, L.; Ning, A. Effects of Kruppel-like factor 6 on osteosarcoma cell biological behavior. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2013, 34, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structural and amino acid sequence properties of KLF proteins. (A) Structure of human SP1, Drosophila melanogaster (Fly) Krüppel, and human KLF proteins. TAD: transcriptional activation domain, RD: repression domain. The green region in SP1 protein defines the Buttonhead box motif, and red region defines the zinc finger motif. Most of the KLF proteins contain three zinc finger motifs close at the carboxyl-terminal edge, while KLF9, KLF13, KLF14, and KLF16 contain zinc finger motifs more positionally shifted to the middle part of the protein indicated by the double arrow. (B) Multiple sequence alignment of three zinc fingers and preceding amino-terminal amino acid residues of Drosophila melanogaster (Fly) Buttonhead, Krüppel, human SP1/KLF family proteins. The first three zinc finger sequences are shown for the Fly Krüppel protein. Alignment was generated by MAFFT, ClustalW, and visual inspection. Buttonhead box motifs (Btd motif) are highlighted by green color. Cystein and Histidine residues of the zinc finger motifs are highlighted by yellow and blue color, respectively.
Figure 1. Structural and amino acid sequence properties of KLF proteins. (A) Structure of human SP1, Drosophila melanogaster (Fly) Krüppel, and human KLF proteins. TAD: transcriptional activation domain, RD: repression domain. The green region in SP1 protein defines the Buttonhead box motif, and red region defines the zinc finger motif. Most of the KLF proteins contain three zinc finger motifs close at the carboxyl-terminal edge, while KLF9, KLF13, KLF14, and KLF16 contain zinc finger motifs more positionally shifted to the middle part of the protein indicated by the double arrow. (B) Multiple sequence alignment of three zinc fingers and preceding amino-terminal amino acid residues of Drosophila melanogaster (Fly) Buttonhead, Krüppel, human SP1/KLF family proteins. The first three zinc finger sequences are shown for the Fly Krüppel protein. Alignment was generated by MAFFT, ClustalW, and visual inspection. Buttonhead box motifs (Btd motif) are highlighted by green color. Cystein and Histidine residues of the zinc finger motifs are highlighted by yellow and blue color, respectively.
Ijms 23 15174 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abe, M.; Saeki, N.; Ikeda, Y.; Ohba, S. Kruppel-like Factors in Skeletal Physiology and Pathologies. Int. J. Mol. Sci. 2022, 23, 15174. https://doi.org/10.3390/ijms232315174

AMA Style

Abe M, Saeki N, Ikeda Y, Ohba S. Kruppel-like Factors in Skeletal Physiology and Pathologies. International Journal of Molecular Sciences. 2022; 23(23):15174. https://doi.org/10.3390/ijms232315174

Chicago/Turabian Style

Abe, Makoto, Naoya Saeki, Yuki Ikeda, and Shinsuke Ohba. 2022. "Kruppel-like Factors in Skeletal Physiology and Pathologies" International Journal of Molecular Sciences 23, no. 23: 15174. https://doi.org/10.3390/ijms232315174

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