Review

Pdf file on Synergy OPEN

omments

Acta Biochim Biophys Sin 2008, 40: 554-564

doi:10.1111/j.1745-7270.2008.00439.x

Roles of Krüppel-like factor 4 in normal homeostasis, cancer and stem cells

 

Paul M Evans and Chunming Liu*

 

Department of Biochemistry and Molecular Biology, Sealy Center for Cancer Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555-1448, USA

 

Received: April 20, 2008       

Accepted: May 15, 2008

This work was supported by the grants from the Sealy Center for Cancer Cell Biology, the National Institutes of Health (No. T32CA117834), and the Charlotte Geyer Foundation

*Corresponding author: Tel, 1-409-747-1909; E-mail, [email protected]

 

Krüppel-like factor 4 (KLF4) is a zinc finger-type transcription factor expressed in a variety of tissues, including the epithelium of the intestine and the skin, and it plays an important role in differentiation and cell cycle arrest. Depending on the gene targeted, KLF4 can both activate and repress transcription. Moreover, in certain cellular contexts, KLF4 can function as a tumor suppressor or an oncogene. Finally, KLF4 is important in reprogramming differentiated fibroblasts into inducible pluripotent stem cells, which highly resemble embryonic stem cells. This review summarizes what is known about the diverse functions of KLF4 as well as their molecular mechanisms.

 

Keywords        Krüppel-like factor 4; colorectal cancer; stem cell

 

Krüppel-like factor 4 (KLF4) is a transcription factor expressed in a wide variety of tissues in humans, including the intestine and the skin, which is important for many different physiologic processes, including development, differentiation, and maintenance of normal tissue homeostasis. KLF4 is a bi-functional transcription factor that can either activate or repress transcription using different mechanisms, depending on the target gene. In addition, KLF4 can function as an oncogene or a tumor suppressor depending on the type of cancer involved. In concert with three other transcription factors, KLF4 can reprogram differentiated fibroblasts into a state resembling embryonic stem cells in every possible manner tested so far. This review will provide a detailed summary of what is currently known about KLF4 and its role in the homeostasis of tissues, in cancer and in stem cell reprogramming.

 

The Krüppel-like Factor Family

 

Krüppel-like factors are a family of transcription factors that play important roles in many fundamental biologic processes, including development, proliferation, differentiation and apoptosis (Fig. 1). Krüppel-like factor family members contain three C-terminal C2H2-type zinc fingers that bind DNA. They were named “Krüppel-like” due to strong homology in this region with the Drosophila gene product Krüppel, which is important in segmentation of the developing embryo. Genetic deletion of Krüppel results in complete absence of the thoracic and anterior abdominal segments [1]. KLF4 was cloned independently by two groups, and given two different names: gut-enriched Krüppel-like factor due to the fact that it was found to be highly expressed in the intestine [2], and epithelial zinc finger due to its high expression in the skin epithelium [3]. It was later renamed KLF4 to avoid confusion, as expression of KLF4 is also detectable in the lung, skin, testis [2-5], thymus [6], cornea [7], cardiac myocytes [8], and lymphocytes [9]. In addition, KLF4 is important in development, as it is detectable in the mouse embryo, with the highest expression occurring in the later stages [3,4].

 

Roles of KLF4 in Homeostasis of the Colonic Epithelium

 

The colonic epithelium consists of three major types of differentiated cells: enterocytes, goblet cells and enteroendocrine cells. Actively proliferating cells reside at the base of the crypts and migrate towards the luminal surface as they differentiate, eventually to be sloughed off. KLF4 inhibits proliferation and promotes differentiation; consistent with this role, expression of KLF4 is greatest near the luminal surface and gradually decreases toward the base of the crypts [2,10]. Klf4-/- mice lack goblet cells, which does not affect the total number of enterocytes, suggesting that KLF4 may be specifically required for goblet cell differentiation [11]. In addition, KLF4 can interact with b-catenin and antagonize Wnt signaling [10], a key pathway in driving proliferation of the intestinal epithelium [12-14]. Thus, KLF4 may also be important in mediating the switch from transit-amplifying cells to the various differentiated cell types in the colonic crypts.

Butyrate is constantly produced in the colon by bacterial fermentation of dietary fiber in the intestine [15], and it can induce expression of KLF4 [5,16]. In cell culture, butyrate stimulates expression of the enterocyte-specific marker intestinal alkaline phosphatase [17], and induces colon cancer cells to acquire a more differentiated, enterocyte-like phenotype [18]. KLF4 positively regulates expression of intestinal alkaline phosphatase [19,20], and overexpression of KLF4 in cell culture inhibits proliferation [2,5]. 

KLF4 appears to have inhibitory effect on a wide variety of cellular processes, including protein and cholesterol synthesis, transcription, cell growth and DNA repair [21,22]. Consistent with its anti-proliferative role, KLF4 simultaneously induces the expression of cyclin-dependent kinase inhibitor proteins p21Cip1/WAF1 and p57Kip2 [21,23–25], and represses the expression of Cyclin D1 [5,26,27], Cyclin D2 [28], Cyclin E [29], and Cyclin B1 [30] (Fig. 2). In addition, KLF4 represses expression of ornithine decarboxylase [7,31], an enzyme involved in the production of a class of molecules known as polyamines, which are also important in proliferation. KLF4 is required for both the G1/S-phase and G2/M-phase checkpoints [30,32,33]. Finally, KLF4 represses expression of p53 and may be important in determining whether cells decide to undergo apoptosis or cell cycle arrest [34].

 

Roles in other Homeostasis of Other Tissues

 

Although the importance of KLF4 in the intestine is well characterized, increasing evidence demonstrates its importance in other organs and tissues as well. For example, KLF4-/- mice die of dehydration soon after birth due to defects in the epidermal barrier of the skin [35], yet targeted overexpression of KLF4 results in early formation of the epithelial permeability barrier [36]. These data clearly implicate KLF4 as an important molecule in differentiation of the skin epithelium. 

Furthermore, overexpressed KLF4 can synergize with maternally injected corticosteroids in accelerating the formation of the skin barrier. This is likely due to overlap between the genes targeted by KLF4 and the glucocorticoid receptor [37]. The utility of glucocorticoids in lung maturation of premature infants is well-established [38], thus it might be interesting to determine whether KLF4 or possibly other Krüppel-like factors could synergize with glucocorticoids in fetal lung maturation as well. Also, in the developing fetus, KLF4 synergizes with Sp1 in up-regulating expression of PSG-5, a protein secreted into the maternal circulation by the placenta [39]. PSG-5 is thought be required for maintenance of a term pregnancy and may protect the fetus from attack by the maternal immune system. In addition, KLF4 and PSG-5 have closely overlapping patterns of expression in the placenta, suggesting an in vivo role for KLF4 in the regulation of PSG-5 expression [40].

Human KLF4 was isolated from an umbilical vein complementary DNA library and is expressed in the vascular endothelium [41]. Expression of KLF4 is induced by shear stress in endothelial cells [42], whereas KLF4 appears to block differentiation and is expressed at low levels in differentiated arterial smooth muscle cells [43]. However, expression of KLF4 is rapidly up-regulated in smooth muscle cells in response to vascular injury [44].

Overexpression of KLF4 in a pro-myelocytic cell line increases the expression of monocyte markers, whereas knockdown of KLF4 decreases TPA-induced over­expression of these same markers. In addition, KLF4-/- hematopoietic stem cells less frequently differentiate into monocytes [45]. When fetal liver cells from KLF4-/- mice were transplanted into lethally irradiated wild-type mice, they had undetectable levels of circulating inflammatory monocytes [46]. Thus, KLF4 appears to be important for both resident and inflammatory monocyte differentiation.

KLF4 is highly expressed in the corneal epithelium, where it is important in differentiation. Targeted deletion of KLF4 in the eye results in corneal fragility, edema and a lack of goblet cells in the conjunctiva [47]. In a cell culture model of adipocyte differentiation using 3T3-L1 cells, short interfering RNA-mediated knockdown of KLF4 completely blocked expression of several phenotypic markers of differentiated adipocytes [48]. Collectively, these data strongly implicate KLF4 as a factor involved in the differentiation of many tissues. 

 

Roles of KLF4 in Cancers

 

As an anti-proliferative factor expressed in differentiated epithelia, it seems logical that KLF4 might act as a tumor suppressor, and indeed this appears to be the case in the gastrointestinal tract [49,50]. However, recent evidence suggests that KLF4 might also act as an oncogene in certain contexts [51]. This section will investigate these two contrasting roles.

 

KLF4 as a tumor suppressor

Increasing evidence implicates KLF4 as a tumor suppressor in the intestinal epithelium. In human colorectal carcinoma, expression of KLF4 is down-regulated, with evidence of both hypermethylation and loss of heterozygosity [52-54]. However, no association has been found between down-regulation of KLF4 and tumor staging or 5-year survival in patients with metastatic carcinoma, suggesting that loss of KLF4 in colorectal cancer may be an early event [53,54].

Examination of KLF4 expression in mouse models of colorectal cancer has yielded similar results.  The APCmin/+ mouse develops hundred of intestinal adenomas early in life and is a widely used model of intestinal tumorigenesis [55,56]. In adenomas from these mice, KLF4 is down-regulated, with expression inversely related to the size of the tumor [4,57]. As APC is a critical component of the Wnt/b-catenin pathway and APCmin/+ mice express a truncated form of the APC protein, these mice have deregulated Wnt signaling in their intestine [58,59]. Interestingly, KLF4 can interact with b-catenin in the nucleus and repress Wnt signaling in vivo, as well as inhibit tumor growth in tumor xenografts [10]. In addition, crossing APCmin/+ mice with KLF4+/- heterozygotes resulted in significantly more adenomas than in APCmin/+ mice alone [60]. Notably, this phenotype was similar to that found with another double mutant, APCMin/+/TCF-1-/-. The most abundant isoform of TCF-1 expressed in the intestine is also an antagonist of Wnt/b-catenin signaling, suggesting that an important effect of decreased KLF4 expression during colorectal tumorigenesis may be de-repression of Wnt signaling.

In human colon cancer cell lines, several point mutations have been found in the KLF4 gene. One mutation had a significant effect on the ability to activate a p21Cip1/WAF1 reporter construct in NIH3T3 cells [52]. However, an investigation to identify mutations in tissue samples of human colorectal cancers has not yet been performed. In the HCT116 colorectal cancer cell line, KLF4 is required to prevent centrosome amplification after gamma-irradiation, and loss of KLF4 may promote chromosomal instability [29]. In addition, KLF4 represses expression of the enzyme ornithine decarboxylase [31], a proto-oncogene that alone is sufficient to transform NIH3T3 cells [61].  Collectively, these data strongly implicate KLF4 as a tumor suppressor in the colon.

Strong evidence also implicates KLF4 as a tumor suppressor in the gastric epithelium. Similar to colorectal cancer, KLF4 is down-regulated in gastric cancer, with evidence of hypermethylation and loss of heterozygosity [62-64]. Moreover, targeted loss of the KLF4 gene in the gastric mucosa of mice results in pre-cancerous changes in the stomach [65]. In examining both normal and cancerous gastric mucosal tissue from humans, one study found an inverse relationship between the expression of KLF4 and Sp1, a distantly related Krüppel-like factor family member (Fig. 1) [62]. In addition, the same study found that in gastric cancer cell lines, KLF4 can directly repress the expression of Sp1. Given that strong expression of Sp1 is correlated with poor survival in gastric cancer [66], loss of KLF4 may contribute to gastric cancer progression.

In addition to gastric and colorectal cancer, KLF4 is down-regulated in esophageal cancer [67,68], bladder cancer [69], non-small-cell lung carcinoma [70], and leukemia [71,72].

 

KLF4 as an oncogene

Although these data clearly demonstrate that KLF4 can act as tumor suppressor in multiple tissues, the possibility that KLF4 might be an oncogene as well was first demonstrated in the late 1990s. Using E1A-immortalized rat kidney epithelial cells to screen for factors that could induce transformation, KLF4 was identified. Moreover, KLF4-transformed rat kidney epithelial cells could produce tumors in xenografted mice [73]. KLF4 is overexpressed in laryngeal squamous cell carcinoma as an early event in its progression [73]. Expression of KLF4 is increased in ductal carcinoma of the breast [74], and increased nuclear staining is associated with a more aggressive phenotype and poorer prognosis [75]. In the skin, overexpression of KLF4 results in hyperplasia and dysplasia [76], eventually leading to squamous cell carcinoma [77].

Whether KLF4 acts as a tumor suppressor or an oncogene is likely due to differences in cell context, expression patterns of other genes and the chromatin environment of individual cells. However, the mechanism to explain these differences fully is unknown. A recent study that found that KLF4 could override RasV12-induced senescence in primary fibroblasts and induce transformation provided some insight [34]. Additionally, this study demonstrated that the status of p21Cip1/WAF1, a transcriptional target of KLF4, determined whether overexpression of KLF4 induced transformation or resulted in cell cycle arrest. Overexpression of KLF4 alone increases expression of p21Cip1/WAF1 and results in cell cycle arrest. However, the addition of RasV12 resulted in inhibition of p21Cip1/WAF1 expression, allowing KLF4’s ability to repress p53 to predominate. Repression of p53 effectively blocked apoptosis and, in concert with the decreased expression of p21Cip1/WAF1, eventually led to transformation. Thus, KLF4 can be added to a growing list of genes that have multiple, context-dependent roles in cancer, including CDKN1A (p21), transforming growth factor-b, Ras and NOTCH1 genes [51].

 

Roles of KLF4 in Stem Cell Renewal and Reprogramming

 

Recently, it was found that overexpression of KLF4, in combination with three other transcription factors, could transform mouse fibroblasts into a state resembling embryonic stem cells (ES cells). These cells have been termed “inducible pluripotent stem cells” (iPS cells) [78]. By replacing the open reading frame of Fbx15, a non-essential marker of ES cells, with a neomycin resistance gene, it was hypothesized that neomycin-resistant colonies might have somehow reprogrammed themselves into ES cells. After screening a short list of potential factors, it was found that the simultaneous infection of retroviruses expressing Oct3/4, Sox2, c-Myc and KLF4 were able to produce resistant clones. These cells could form teratomas that contained differentiated tissues from all three germ layers, confirming their pluripotency. This approach was further refined by screening for neomycin resistance based on Nanog or Oct4 expression instead of Fbx15 expression. Unlike Fbx15-iPS cells, Nanog and Oct4-iPS could produce chimeric mice, and generate live late-term embryos when injected into tetraploid blastocysts [79-81]. Thus, Nanog- and Oct4-iPS are even more stringent tests of pluripotency than Fbx15-iPS cells.

Researchers are currently trying to gain a better understanding of the molecular events that occur during stem cell reprogramming as well as the precise role of the four individual factors required. The importance of Oct3/4 and Sox2 in ES cell renewal is well established [82]. What is less clear is the function of the other two factors that make up the "magic brew": c-Myc and KLF4. One possibility is that c-Myc and KLF4 confer increased proliferative capacity on potential iPS cells, since both can function as oncogenes [83]. Since c-Myc regulates a significant number of genes, its function may be to affect global changes in the chromatin environment by recruiting histone acetyl-transferase complexes. According this model, KLF4 may then function to inhibit apoptosis induced by overexpression of c-Myc. KLF4 represses c-Myc expression in colon cancer cells by inhibiting Wnt signaling [10]. While the role of Wnt signaling in iPS cells remains unresolved, c-Myc may provide a balance for KLF4.

Overexpression of KLF4 in ES cells inhibited differentiation in erythroid progenitors and increased their capacity to generate secondary embryoid bodies, suggesting a role for KLF4 in self-renewal [84]. In concert with Oct3/4 and Sox2, KLF4 activates expression of Lefty1, a gene expressed in ES cells but lost during differentiation [85]. In addition, KLF4-null mice survive to term and have no detectable defects during embryogenesis in their pluripotent stem cell population [11,35], suggesting that KLF4 may be dispensable in normal ES cells. More recently, human iPS have been produced using a slightly different mix of factors, substituting Nanog and LIN28 for c-Myc and KLF4 [86], further calling into question the overall importance of c-Myc and KLF4. It has even been suggested that c-Myc and KLF4 are merely molecular catalysts, in that they might accelerate or increase the efficiency of the reprogramming process, but are otherwise not absolutely required [87].

However, a recent study has found that KLF4’s function in ES cell self-renewal is partially redundant; knockdown of KLF4, KLF2 and KLF5, but not any one individually, resulted in spontaneous ES cell differentiation [88]. In addition, significant overlap was found between genes regulated by Nanog and the three Krüppel-like factors. Clearly, a complete understanding of the role of KLF4 in ES cell self-renewal and iPS cell reprogramming awaits further study

 

Molecular Mechanisms of KLF4

Human and mouse KLF4 are 470 and 483 amino acids in length, respectively, and produce a 55 kDa protein. KLF4 can be roughly divided into three separate domains: a N-terminal activation domain [3,41,89], a central repressive domain [41], and a C-terminal DNA-binding domain (Fig. 3). The DNA-binding domain consists of three successive zinc fingers, each containing an anti-parallel b-sheet, followed by a short loop and an a-helix. Two cysteines within the b-sheet and two histidines within the a-helix work together to coordinate a single zinc ion, which stabilizes the fold. Each zinc finger interacts with three consecutive nucleotides on a target DNA sequence, and the sequence specificity of a zinc finger protein can be increased simply by adding zinc fingers [90].

In general, KLF4 interacts with GT-rich or CACCC elements on target genes [41,91]. Although one report suggests that KLF4 prefers to bind a RRGGYGY sequence (where R=A/G and Y=C/T) [92], it is still not clear whether this is a true consensus in vivo. KLF4 is exclusively nuclear, like many other transcription factors, and appears to contain two discrete nuclear localization sequences: a basic hexapeptide sequence of a N-terminal to the three C-terminal zinc fingers and a sequence contained within the first two zinc fingers themselves [93].

Given the large number of genes regulated by KLF4, it is not surprising that the expression of KLF4 is highly regulated (Table 1). In the colon cancer cell line HCT116, KLF4 has a half-life of only 2 h and is quickly degraded by the proteasome [94]. However, a variety of stimuli can induce KLF4 expression, including serum starvation, contact inhibition [3], interferon-g [31,95], sodium butyrate [5,16], cAMP [48], gastrin [96], DNA damage [24,33], and oxidative stress [8,25]. The precise mechanism of how the majority these stimuli increase the expression of KLF4 is unclear, although possibilities include increased transcription of the KLF4 gene, increased mRNA stability and/or increased protein stability. 

Although much remains to be known about how KLF4 expression is regulated, several transcription factors have been found to regulate its promoter. For example, p53 transactivates the KLF4 gene, and p53 is required for the induction of KLF4 after DNA damage [24,33]. CDX2, another protein important in differentiation of the intestinal epithelium, can activate a KLF4 reporter construct [97]. This suggests that KLF4 may act downstream of CDX2, although more work is necessary to demonstrate this in vivo. KLF4 up-regulates its own expression by binding to its promoter, whereas KLF5 inhibits KLF4 expression and blocks the binding of KLF4 to its promoter [98]. Although KLF4 and KLF5 are closely related transcription factors, expression of KLF5 is in a completely opposite pattern in the colonic intestine, with the strongest expression found in the actively proliferating cells at the base of the crypts; expression is absent in differentiated cells at the luminal surface [99,100]. In fact, KLF4 and KLF5 have several antagonizing roles in the intestinal epithelium [49].

 

Mechanisms of activation

A major function of KLF4 is to activate transcription of target genes (Table 2). Consistent with this function, the N-terminus of KLF4 contains a strong transactivation domain [3,41,89]. This domain alone, when directly fused to its three C-terminal zinc fingers, is sufficient to activate a synthetic reporter construct [89]. In addition, the N-terminal domain interacts with the transcriptional co-activators p300/CBP, which is required for its function, as point mutations that block interactions with CBP also completely abrogate its ability to activate transcription [20,89]. p300/CBP are histone acetyltransferase proteins, and recruitment of p300/CBP results in an increase in localized histone acetylation at the promoter. Acetylation of histones facilitates the recruitment of other transcription factors as well as the basal transcriptional machinery. In addition, KLF4 itself is acetylated by p300/CBP at lysine residues 225 and 229. Mutation of these two lysines to arginine significantly decreases the ability of KLF4 to transactivate target genes and to inhibit proliferation [20], suggesting that acetylation of KLF4 is important for its function.

One report found that KLF4 can interact with Tip60, a bi-functional cofactor that contains intrinsic histone acetyltransferase activity, but it can also recruit HDAC7 [96]. Tip60 is a co-activator for several nuclear hormone receptors and APP [101,102], but appears to function as a co-repressor for STAT3 by recruiting HDAC7 [103].  Krox20, another zinc finger protein, can directly interact with KLF4 and synergistically activate the C/EBPb gene in 3T3-L1 cells [48]. KLF4 interacts with the NF-kB subunit p65/RelA and synergistically activates expression of inducible nitric oxide synthase [104]. Thus, the mechanisms of transactivation mediated by KLF4 may be gene dependent.

 

Mechanism of repression

One mechanism for repression by a transcription factor is to simple competition with an activator for binding to a target DNA sequence. This mechanism is known as a form of passive repression. On the CYP1A1, HDC, and Sp1 genes, KLF4 binds to a sequence overlapping that recognized by the activator Sp1, displacing Sp1 from the promoter and resulting in repression of the target gene [62,105,106]. Since Sp1 is ubiquitously expressed and positively regulates many genes [107], it is likely this mechanism is used by KLF4 to repress many of its target genes.

GAL4 fusion assays demonstrate that KLF4 contains central repressive domain in addition to its more fully characterized transactivation domain [41]. This suggests that KLF might actively repress expression of some genes, in addition to or instead of passive repression via competition with a transcriptional activator. In KLF4-mediated repression of the CD11d gene, KLF4 interacts with and recruits HDAC1 and HDAC2 [108], whereas KLF4 represses Cyclin B1 by specifically recruiting HDAC3 [20]. On the TP53 gene, MUC1-C recruits KLF4, as well as HDAC1 and HDAC3, to mediate repression [109]. KLF4 inhibits Smad3-mediated activation of PAI-1 by directly competing with Smad3 for p300 binding [104]. Finally, KLF4 represses transcriptional targets of Wnt signaling by directly interacting with b-catenin/TCF-4 [10]. These data strongly suggest that KLF4-mediated activation and repression is complex and gene-dependent.

 

Final Thoughts

 

KLF4 is complex transcription factor that, depending on the context, can act as a transcriptional activator, a transcriptional repressor, an oncogene, and a tumor suppressor. In considering such a transcription factor, questions arise as to how it can switch between these modes and what molecular mechanisms govern its function in normal cells, in cancer and in stem cell reprogramming. Although this review discusses much of what is already known in regard to these issues, more work is needed to fully understand them. Attaining a greater understanding of the molecular function of KLF4 will ultimately provide a deeper insight into these many different fundamental processes.

 

 

References

 

  1    Reiss A, Rosenberg UB, Kienlin A, Seifert E, Jackle H. Molecular genetics of Krüppel, a gene required for segmentation of the Drosophila embryo. Nature 1985, 313: 27-32

  2    Shields JM, Christy RJ, Yang VW. Identification and characterization of a gene encoding a gut-enriched Krüppel-like factor expressed during growth arrest. J Biol Chem 1996, 271: 20009-20017

  3   Garrett-Sinha LA, Eberspaecher H, Seldin MF, de Crombrugghe B. A gene for a novel zinc-finger protein expressed in differentiated epithelial cells and transiently in certain mesenchymal cells. J Biol Chem 1996, 271: 31384-31390

  4    Ton-That H, Kaestner KH, Shields JM, Mahatanankoon CS, Yang VW. Expression of the gut-enriched Krüppel-like factor gene during development and intestinal tumorigenesis. FEBS Lett 1997, 419: 239-243

  5    Shie JL, Chen ZY, Orien MJ, Pestell RG, Lee ME, Tseng CC. Role of gut-enriched Krüppel-like factor in colonic cell growth and differentiation. Am J Physiol Gastrointest Liver Physiol 2000, 279: G806-G814

  6   Panigada M, Porcellini S, Sutti F, Doneda L, Pozzoli O, Consalez GG, Guttinger M et al. GKLF in thymus epithelium as a developmentally regulated element of thymocyte-stroma cross-talk. Mech Dev 1999, 81: 103-113

  7    Chiambaretta F, De Graeve F, Turet G, Marceau G, Gain P, Dastugue B, Rigal D et al. Cell and tissue specific expression of human Krüppel-like transcription factors in human ocular surface. Mol Vis 2004, 10: 901-909

  8    Cullingford TE, Butler MJ, Marshall AK, Tham EL, Sugden PH, Clerk A. Differential regulation of Krüppel-like factor family transcription factor expression in neonatal rat cardiac myocytes: effects of endothelin-1, oxidative stress and cytokines. Biochim Biophys Acta 2008, 1783: 1229-1236

  9    Fruman DA, Ferl GZ, An SS, Donahue AC, Satterthwaite AB, Witte ON. Phosphoinositide 3-kinase and Bruton tyrosine kinase regulate overlapping sets of genes in B lymphocytes. Proc Natl Acad Sci USA 2002, 99: 359-364

 10   Zhang W, Chen X, Kato Y, Evans PM, Yuan S, Yang J, Rychahou PG et al. Novel cross talk of Krüppel-like factor 4 and b-catenin regulates normal intestinal homeostasis and tumor repression. Mol Cell Biol 2006, 26: 2055-2064

 11   Katz JP, Perreault N, Goldstein BG, Lee CS, Labosky PA, Yang VW, Kaestner KH. The zinc-finger transcription factor KLF4 is required for terminal differentiation of goblet cells in the colon. Development 2002, 129: 2619-2628

 12   Korinek V, Barker N, Moerer P, van Donselaar E, Huls G, Peters PJ, Clevers H. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking TCF-4. Nat Genet 1998, 19: 379-383

 13   van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I, Hurlstone A, van der Horn K et al. The b-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells. Cell 2002, 111: 241-250

 14   Batlle E, Henderson JT, Beghtel H, van den Born MM, Sancho E, Huls G, Meeldijk J et al. b-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell 2002, 111: 251-263

 15   Roediger WE. Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 1980, 21: 793-798

 16   Chen ZY, Rex S, Tseng CC. Krüppel-like factor 4 is transactivated by butyrate in colon cancer cells. J Nutr 2004, 134: 792-798

 17   Wang Q, Wang X, Hernandez A, Kim S, Evers BM. Inhibition of the phosphatidylinositol 3-kinase pathway contributes to HT29 and Caco-2 intestinal cell differentiation. Gastroenterology 2001, 120: 1381-1392

 18   Heerdt BG, Houston MA, Augenlicht LH. Potentiation by specific short-chain fatty acids of differentiation and apoptosis in human colonic carcinoma cell lines. Cancer Res 1994, 54: 3288-3293

 19   Hinnebusch BF, Siddique A, Henderson JW, Malo MS, Zhang W, Athaide CP, Abedrapo MA et al. Enterocyte differentiation marker intestinal alkaline phosphatase is a target gene of the gut-enriched Krüppel-like factor. Am J Physiol Gastrointest Liver Physiol 2004, 286: G23-G30

 20   Evans PM, Zhang W, Chen X, Yang J, Bhakat KK, Liu C. Krüppel-like factor 4 is acetylated by p300 and regulates gene transcription via modulation of histone acetylation. J Biol Chem 2007, 282: 33994-4002

 21   Chen X, Whitney EM, Gao SY, Yang VW. Transcriptional profiling of Krüppel-like factor 4 reveals a function in cell cycle regulation and epithelial differentiation. J Mol Biol 2003, 326: 665-677

 22   Whitney EM, Ghaleb AM, Chen X, Yang VW. Transcriptional profiling of the cell cycle checkpoint gene Krüppel-like factor 4 reveals a global inhibitory function in macromolecular biosynthesis. Gene Expr 2006, 13: 85-96

 23   Mahatan CS, Kaestner KH, Geiman DE, Yang VW. Characterization of the structure and regulation of the murine gene encoding gut-enriched Krüppel-like factor (Krüppel-like factor 4). Nucleic Acids Res 1999, 27: 4562-4569

 24   Zhang W, Geiman DE, Shields JM, Dang DT, Mahatan CS, Kaestner KH, Biggs JR et al. The gut-enriched Krüppel-like factor (Krüppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J Biol Chem 2000, 275: 18391-18398

 25   Nickenig G, Baudler S, Muller C, Werner C, Werner N, Welzel H, Strehlow K et al. Redox-sensitive vascular smooth muscle cell proliferation is mediated by GKLF and Id3 in vitro and in vivo. FABSEB J 2002, 16: 1077-1086

 26   Shie JL, Pestell RG, TC C. Repression of the cyclin D1 promoter by gut-enriched Krüppel-like factor. Gastroenterology 1999, 11: A520

 27   Shie JL, Chen ZY, Fu M, Pestell RG, Tseng CC. Gut-enriched Krüppel-like factor represses cyclin D1 promoter activity through Sp1 motif. Nucleic Acids Res 2000, 28: 2969-2976

 28   Klaewsongkram J, Yang Y, Golech S, Katz J, Kaestner KH, Weng NP. Krüppel-like factor 4 regulates B cell number and activation-induced B cell proliferation. J Immunol 2007, 179: 4679-4684

 29   Yoon HS, Ghaleb AM, Nandan MO, Hisamuddin IM, Dalton WB, Yang VW. Krüppel-like factor 4 prevents centrosome amplification following g-irradiation-induced DNA damage. Oncogene 2005, 24: 4017-4025

 30   Yoon HS Yang VW. Requirement of Krüppel-like factor 4 in preventing entry into mitosis following DNA damage. J Biol Chem 2004, 279: 5035-5041

 31   Chen ZY, Shie JL, Tseng CC. Gut-enriched Krüppel-like factor represses ornithine decarboxylase gene expression and functions as checkpoint regulator in colonic cancer cells. J Biol Chem 2002, 277: 46831-46839

 32   Chen X, Johns DC, Geiman DE, Marban E, Dang DT, Hamlin G, Sun R et al. Krüppel-like factor 4 (gut-enriched Krüppel-like factor) inhibits cell proliferation by blocking G1/S progression of the cell cycle. J Biol Chem 2001, 276: 30423-30428

 33   Yoon HS, Chen X, Yang VW. Krüppel-like factor 4 mediates p53-dependent G1/S cell cycle arrest in response to DNA damage. J Biol Chem 2003, 278: 2101-2105

 34   Rowland BD, Bernards R, Peeper DS. The KLF4 tumor suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol 2005, 7: 1074-1082

 35   Segre JA, Bauer C, Fuchs E. KLF4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet 1999, 22: 356-260

 36   Jaubert J, Cheng J, Segre JA. Ectopic expression of Krüppel like factor 4 (KLF4) accelerates formation of the epidermal permeability barrier. Development 2003, 130: 2767-2777

 37   Patel S, Xi ZF, Seo EY, McGaughey D, Segre JA. KLF4 and corticosteroids activate an overlapping set of transcriptional targets to accelerate in utero epidermal barrier acquisition. Proc Natl Acad Sci USA 2006, 103: 18668-18673

 38   Effect of corticosteroids for fetal maturation on perinatal outcomes. NIH consensus development panel on the effect of corticosteroids for fetal maturation on perinatal outcomes. JAMA 1995, 273: 413-418

 39   Blanchon L, Nores R, Gallot D, Marceau G, Borel V, Yang VW, Bocco JL et al. Activation of the human pregnancy-specific glycoprotein PSG-5 promoter by KLF4 and Sp1. Biochem Biophys Res Commun 2006, 343: 745-753

 40   Blanchon L, Bocco JL, Gallot D, Gachon AM, Lemery D, Dechelotte P, Dastugue B et al. Co-localization of KLF6 and KLF4 with pregnancy-specific glycoproteins during human placenta development. Mech Dev 2001, 105: 185-189

 41   Yet SF, McAulty MM, Folta SC, Yen HW, Yoshizumi M, Hsieh CM, Layne MD et al. Human EZF, a Krüppel-like zinc finger protein, is expressed in vascular endothelial cells and contains transcriptional activation and repression domains. J Biol Chem 1998, 273: 1026-1031

 42   McCormick SM, Eskin SG, McIntire LV, Teng CL, Lu CM, Russell CG, Chittur KK. DNA microarray reveals changes in gene expression of shear stressed human umbilical vein endothelial cells. Proc Natl Acad Sci USA 2001, 98: 8955-8960

 43   Adam PJ, Regan CP, Hautmann MB, Owens GK. Positive- and negative-acting Krüppel-like transcription factors bind a transforming growth factor b control element required for expression of the smooth muscle cell differentiation marker SM22a in vivo. J Biol Chem 2000, 275: 37798-37806

 44  Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH, Owens GK. Krüppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J Biol Chem 2005, 280: 9719-9727

 45   Feinberg MW, Wara AK, Cao Z, Lebedeva MA, Rosenbauer F, Iwasaki H, Hirai H et al. The Krüppel-like factor KLF4 is a critical regulator of monocyte differentiation. EMBO J 2007, 26: 4138-4148

 46   Alder JK, Georgantas RW III, Hildreth RL, Kaplan IM, Morisot S, Yu X, McDevitt M et al. Krüppel-like factor 4 is essential for inflammatory monocyte differentiation in vivo. J Immunol 2008, 180: 5645-5652

 47   Swamynathan SK, Katz JP, Kaestner KH, Ashery-Padan R, Crawford MA, Piatigorsky J. Conditional deletion of the mouse KLF4 gene results in corneal epithelial fragility, stromal edema, and loss of conjunctival goblet cells. Mol Cell Biol 2007, 27: 182-194

 48   Birsoy K, Chen Z, Friedman J. Transcriptional regulation of adipogenesis by KLF4. Cell Metab 2008, 7: 339-347

 49   McConnell BB, Ghaleb AM, Nandan MO, Yang VW. The diverse functions of Krüppel-like factors 4 and 5 in epithelial biology and pathobiology. Bioessays 2007, 29: 549-557

 50   Wei D, Kanai M, Huang S, Xie K, Emerging role of KLF4 in human gastrointestinal cancer. Carcinogenesis 2006, 27: 23-31

 51   Rowland BD, Peeper DS. KLF4, p21 and context-dependent opposing forces in cancer. Nat Rev Cancer 2006, 6: 11-23

 52   Zhao W, Hisamuddin IM, Nandan MO, Babbin BA, Lamb NE, Yang VW. Identification of Krüppel-like factor 4 as a potential tumor suppressor gene in colorectal cancer. Oncogene 2004, 23: 395-402

 53   Choi BJ, Cho YG, Song JW, Kim CJ, Kim SY, Nam SW, Yoo NJ et al. Altered expression of the KLF4 in colorectal cancers. Pathol Res Pract 2006, 202: 585-589

 54   Xu J, Lu B, Xu F, Gu H, Fang Y, Huang Q, Lai M. Dynamic down-regulation of Krüppel-like factor 4 in colorectal adenoma-carcinoma sequence. J Cancer Res Clin Oncol 2008 (forthcoming)

 55   Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 1990, 247: 322-324

 56   Su LK, Kinzler KW, Vogelstein B, Preisinger AC, Moser AR, Luongo C, Gould KA et al. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 1992, 256: 668-670

 57   Dang DT, Bachman KE, Mahatan CS, Dang LH, Giardiello FM, Yang VW, Decreased expression of the gut-enriched Krüppel-like factor gene in intestinal adenomas of multiple intestinal neoplasia mice and in colonic adenomas of familial adenomatous polyposis patients. FEBS Lett 2000, 476: 203-207

 58   Morin PJ, Sparks AB, Korinek V, Barker N, Clevers H, Vogelstein B, Kinzler KW. Activation of b-catenin-TCF signaling in colon cancer by mutations in b-catenin or APC. Science 1997, 275: 1787-1790

 59   Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzler KW, Vogelstein B et al. Constitutive transcriptional activation by a b-catenin-TCF complex in APC-/- colon carcinoma. Science 1997, 275: 1784-1787

 60   Ghaleb AM, McConnell BB, Nandan MO, Katz JP, Kaestner KH, Yang VW. Haploinsufficiency of Krüppel-like factor 4 promotes adenomatous polyposis coli dependent intestinal tumorigenesis. Cancer Res 2007, 67: 7147-7154

 61   Auvinen M, Paasinen A, Andersson LC, Holtta E. Ornithine decarboxylase activity is critical for cell transformation. Nature 1992, 360: 355-358

 62   Kanai M, Wei D, Li Q, Jia Z, Ajani J, Le X, Yao J et al. Loss of Krüppel-like factor 4 expression contributes to Sp1 overexpression and human gastric cancer development and progression. Clin Cancer Res 2006, 12: 6395-6402

 63   Wei D, Gong W, Kanai M, Schlunk C, Wang L, Yao JC, Wu TT et al. Drastic down-regulation of Krüppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res 2005, 65: 2746-2754

 64   Cho YG, Song JH, Kim CJ, Nam SW, Yoo NJ, Lee JY, Park WS. Genetic and epigenetic analysis of the KLF4 gene in gastric cancer. APMIS 2007, 115: 802-808

 65   Katz JP, Perreault N, Goldstein BG, Actman L, McNally SR, Silberg DG, Furth EE et al. Loss of KLF4 in mice causes altered proliferation and differentiation and precancerous changes in the adult stomach. Gastroenterology 2005, 128: 935-945

 66   Wang L, Wei D, Huang S, Peng Z, Le X, Wu TT, Yao J et al. Transcription factor Sp1 expression is a significant predictor of survival in human gastric cancer. Clin Cancer Res 2003, 9: 6371-6380

 67   Luo A, Kong J, Hu G, Liew CC, Xiong M, Wang X, Ji J et al. Discovery of Ca2+-relevant and differentiation-associated genes downregulated in esophageal squamous cell carcinoma using cDNA microarray. Oncogene 2004, 23: 1291-1299

 68   Wang N, Liu ZH, Ding F, Wang XQ, Zhou CN, Wu M. Down-regulation of gut-enriched Krüppel-like factor expression in esophageal cancer. World J Gastroenterol 2002, 8: 966-970

 69  Ohnishi S, Ohnami S, Laub F, Aoki K, Suzuki K, Kanai Y, Haga K et al. Downregulation and growth inhibitory effect of epithelial-type Krüppel-like transcription factor KLF4, but not KLF5, in bladder cancer. Biochem Biophys Res Commun 2003, 308: 251-256

 70   Bianchi F, Hu J, Pelosi G, Cirincione R, Ferguson M, Ratcliffe C, Di Fiore PP et al. Lung cancers detected by screening with spiral computed tomography have a malignant phenotype when analyzed by cDNA microarray. Clin Cancer Res 2004, 10: 6023-6028

 71   Yasunaga J, Taniguchi Y, Nosaka K, Yoshida M, Satou Y, Sakai T, Mitsuya H et al. Identification of aberrantly methylated genes in association with adult T-cell leukemia. Cancer Res 2004, 64: 6002-6009

 72   Kharas MG, Yusuf I, Scarfone VM, Yang VW, Segre JA, Huettner CS, Fruman DA. KLF4 suppresses transformation of pre-B cells by ABL oncogenes. Blood 2007, 109: 747-755

 73   Foster KW, Ren S, Louro ID, Lobo-Ruppert SM, McKie-Bell P, Grizzle W, Hayes MR et al. Oncogene expression cloning by retroviral transduction of adenovirus E1A-immortalized rat kidney RK3E cells: transformation of a host with epithelial features by c-Myc and the zinc finger protein GKLF. Cell Growth Differ 1999, 10: 423-434

 74   Foster KW, Frost AR, McKie-Bell P, Lin CY, Engler JA, Grizzle WE, Ruppert JM. Increase of GKLF messenger RNA and protein expression during progression of breast cancer. Cancer Res 2000, 60: 6488-6495

 75   Pandya AY, Talley LI, Frost AR, Fitzgerald TJ, Trivedi V, Chakravarthy M, Chhieng DC et al. Nuclear localization of KLF4 is associated with an aggressive phenotype in early-stage breast cancer. Clin Cancer Res 2004, 10: 2709-2719

 76   Foster KW, Liu Z, Nail CD, Li X, Fitzgerald TJ, Bailey SK, Frost AR et al. Induction of KLF4 in basal keratinocytes blocks the proliferation-differentiation switch and initiates squamous epithelial dysplasia. Oncogene 2005, 24: 1491-1500

 77   Huang CC, Liu Z, Li X, Bailey SK, Nail CD, Foster KW, Frost AR et al. KLF4 and PCNA identify stages of tumor initiation in a conditional model of cutaneous squamous epithelial neoplasia. Cancer Biol Ther 2005, 4: 1401-1408

 78   Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126: 663-676

 79   Maherali N, Sridharan R, Xie W, Utikal J, Eminli S, Arnold K, Stadtfeld M et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell 2007, 1: 55-70

 80   Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007, 448: 313-317

 81   Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, Bernstein BE et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 2007, 448: 318-324

 82   Lewitzky M, Yamanaka S. Reprogramming somatic cells towards pluripotency by defined factors. Curr Opin Biotechnol 2007, 18: 467-473

 83   Yamanaka S, Induction of pluripotent stem cells from mouse fibroblasts by four transcription factors. Cell Prolif 2008, 41: 51-56

 84   Li Y, McClintick J, Zhong L, Edenberg HJ, Yoder MC, Chan RJ. Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor KLF4. Blood 2005, 105: 635-637

 85   Nakatake Y, Fukui N, Iwamatsu Y, Masui S, Takahashi K, Yagi R, Yagi K et al. KLF4 cooperates with Oct3/4 and Sox2 to activate the Lefty1 core promoter in embryonic stem cells. Mol Cell Biol 2006, 26: 7772-7782

 86   Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 2007, 318: 1917-1920

 87   Jaenisch R, Young R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 2008, 132: 567-582

 88   Jiang J, Chan YS, Loh YH, Cai J, Tong GQ, Lim CA, Robson P et al. A core KLF circuitry regulates self-renewal of embryonic stem cells. Nat Cell Biol 2008, 10: 353-360

 89   Geiman DE, Ton-That H, Johnson JM, Yang VW. Transactivation and growth suppression by the gut-enriched Krüppel-like factor (Krüppel-like factor 4) are dependent on acidic amino acid residues and protein-protein interaction. Nucleic Acids Res 2000, 28: 1106-1113

 90   Yang VW. Eukaryotic transcription factors: Identification, characterization and functions. J Nutr 1998, 128: 2045-2051

 91   Philipsen S, Suske G. A tale of three fingers: The family of mammalian Sp/XKLF transcription factors. Nucleic Acids Res 1999, 27: 2991-3000

 92   Shields JM, Yang VW. Identification of the DNA sequence that interacts with the gut-enriched Krüppel-like factor. Nucleic Acids Res 1998, 26: 796-802

 93   Shields JM, Yang VW. Two potent nuclear localization signals in the gut-enriched Krüppel-like factor define a subfamily of closely related Krüppel proteins. J Biol Chem 1997, 272: 18504-18507

 94   Chen ZY, Wang X, Zhou Y, Offner G, Tseng CC. Destabilization of Krüppel-like factor 4 protein in response to serum stimulation involves the ubiquitin-proteasome pathway. Cancer Res 2005, 65: 10394-10400

 95   Chen ZY, Shie J, Tseng C. Up-regulation of gut-enriched Krüppel-like factor by interferon-g in human colon carcinoma cells. FEBS Lett 2000, 477: 67-72

 96   Ai W, Zheng H, Yang X, Liu Y, Wang TC. Tip60 functions as a potential corepressor of KLF4 in regulation of HDC promoter activity. Nucleic Acids Res 2007, 35: 6137-6149

 97   Dang DT, Mahatan CS, Dang LH, Agboola IA, Yang VW. Expression of the gut-enriched Krüppel-like factor (Krüppel-like factor 4) gene in the human colon cancer cell line RKO is dependent on CDX2. Oncogene 2001, 20: 4884-4890

 98   Dang DT, Zhao W, Mahatan CS, Geiman DE, Yang VW. Opposing effects of Krüppel-like factor 4 (gut-enriched Krüppel-like factor) and Krüppel-like factor 5 (intestinal-enriched Krüppel-like factor) on the promoter of the Krüppel-like factor 4 gene. Nucleic Acids Res 2002, 30: 2736-2741

 99   Watanabe N, Kurabayashi M, Shimomura Y, Kawai-Kowase K, Hoshino Y, Manabe I, Watanabe M et al. BTEB2, a Krüppel-like transcription factor, regulates expression of the SMemb/Nonmuscle myosin heavy chain B (SMemb/NMHC-B) gene. Circ Res 1999, 85: 182-191

100  Conkright MD, Wani MA, Anderson KP, Lingrel JB. A gene encoding an intestinal-enriched member of the Krüppel-like factor family expressed in intestinal epithelial cells. Nucleic Acids Res 1999, 27: 1263-1270

101  Gaughan L, Brady ME, Cook S, Neal DE, Robson CN. Tip60 is a co-activator specific for class I nuclear hormone receptors. J Biol Chem 2001, 276: 46841-46848

102  Cao X, Sudhof TC. A transcriptionally active complex of APP with Fe65 and histone acetyltransferase Tip60. Science 2001, 293: 115-120

103  Xiao H, Chung J, Kao HY, Yang YC. Tip60 is a co-repressor for STAT3. J Biol Chem 2003, 278: 11197-11204

104  Feinberg MW, Cao Z, Wara AK, Lebedeva MA, Senbanerjee S, Jain MK. Krüppel-like factor 4 is a mediator of proinflammatory signaling in macrophages. J Biol Chem 2005, 280: 38247-38258

105  Zhang W, Shields JM, Sogawa K, Fujii-Kuriyama Y, Yang VW. The gut-enriched Krüppel-like factor suppresses the activity of the CYP1A1 promoter in a Sp1-dependent fashion. J Biol Chem 1998, 273: 17917-17925

106  Ai W, Liu Y, Langlois M, Wang TC. Krüppel-like factor 4 (KLF4) represses histidine decarboxylase gene expression through an upstream Sp1 site and downstream gastrin responsive elements. J Biol Chem 2004, 279: 8684-8693

107  Black AR, Black JD, Azizkhan-Clifford J. Sp1 and Krüppel-like factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol 2001, 188: 143-160

108  Noti JD, Johnson AK, Dillon JD. The leukocyte integrin gene CD11d is repressed by gut-enriched Krüppel-like factor 4 in myeloid cells. J Biol Chem 2005, 280: 3449-3457

109  Wei X, Xu H, and Kufe D. Human mucin 1 oncoprotein represses transcription of the p53 tumor suppressor gene. Cancer Res 2007, 67: 1853-1858

110  Hamik A, Lin Z, Kumar A, Balcells M, Sinha S, Katz J, Feinberg MW et al. Krüppel-like factor 4 regulates endothelial inflammation. J Biol Chem 2007, 282: 13769-13779

111  King KE, Iyemere VP, Weissberg PL, and Shanahan CM, Kruppel-like factor 4 (KLF4/GKLF) is a target of bone morphogenetic proteins and transforming growth factor beta 1 in the regulation of vascular smooth muscle cell phenotype. J Biol Chem 2003. 278: 11661-11669

112  Mao Z, Song S, Zhu Y, Yi X, Zhang H, Shang Y, Tong T. Transcriptional regulation of A33 antigen expression by gut-enriched Krüppel-like factor. Oncogene 2003, 22: 4434-4443

113  Saifudeen Z, Dipp S, Fan H, El-Dahr SS. Combinatorial control of the bradykinin B2 receptor promoter by p53, CREB, KLF4, and CBP: implications for terminal nephron differentiation. Am J Physiol Renal Physiol 2005, 288: F899-F909

114  Jenkins TD, Opitz OG, Okano J, Rustgi AK. Transactivation of the human keratin 4 and Epstein-Barr virus ED-L2 promoters by gut-enriched Krüppel-like factor. J Biol Chem 1998, 273: 10747-10754

115  Reidling JC, Said HM. Regulation of the human biotin transporter hSMVT promoter by KLF4 and AP-2: confirmation of promoter activity in vivo. Am J Physiol Cell Physiol 2007, 292: C1305-C1312

116  Siddique A, Malo MS, Ocuin LM, Hinnebusch BF, Abedrapo MA, Henderson JW, Zhang W et al. Convergence of the thyroid hormone and gut-enriched Krüppel-like factor pathways in the context of enterocyte differentiation. J Gastrointest Surg 2003, 7: 1053-1061

117  Piccinni SA, Bolcato-Bellemin AL, Klein A, Yang VW, Kedinger M, Simon-Assmann P, Lefebvre O. Krüppel-like factors regulate the Lama1 gene encoding the laminin alpha1 chain. J Biol Chem 2004, 279: 9103-9114

118  Brembeck FH, Rustgi AK. The tissue-dependent keratin 19 gene transcription is regulated by GKLF/KLF4 and Sp1. J Biol Chem 2000, 275: 28230-28239

119  Miller KA, Eklund EA, Peddinghaus ML, Cao Z, Fernandes N, Turk PW, Thimmapaya B et al. Krüppel-like factor 4 regulates laminin a 3A expression in mammary epithelial cells. J Biol Chem 2001, 276: 42863-42868

120  Higaki Y, Schullery D, Kawata Y, Shnyreva M, Abrass C, Bomsztyk K. Synergistic activation of the rat laminin gamma1 chain promoter by the gut-enriched Krüppel-like factor (GKLF/KLF4) and Sp1. Nucleic Acids Res 2002, 30: 2270-2279

121  Liu Y, Sinha S, Owens G. A transforming growth factor-b control element required for SM a-actin expression in vivo also partially mediates GKLF-dependent transcriptional repression. J Biol Chem 2003, 278: 48004-48011

122  Zeng Y, Zhuang S, Gloddek J, Tseng CC, Boss GR, Pilz RB. Regulation of cGMP-dependent protein kinase expression by Rho and Krüppel-like transcription factor-4. J Biol Chem 2006, 281: 16951-16961

123   Wang H, Yang L, Jamaluddin MS, Boyd DD. The Krüppel-like KLF4 transcription factor, a novel regulator of urokinase receptor expression, drives synthesis of this binding site in colonic crypt luminal surface epithelial cells. J Biol Chem 2004, 279: 22674-22683