Skip to main content
Cell Cycle logoLink to Cell Cycle
. 2014 May 5;13(11):1677–1693. doi: 10.4161/cc.29082

The other side of the coin

The tumor-suppressive aspect of oncogenes and the oncogenic aspect of tumor-suppressive genes, such as those along the CCND–CDK4/6–RB axis

Xiaomin Lou 1,*, Ju Zhang 1, Siqi Liu 1, Ningzhi Xu 2,*, D Joshua Liao 3,*
PMCID: PMC4111714  PMID: 24799665

Abstract

Although cancer-regulatory genes are dichotomized to oncogenes and tumor-suppressor gene s, in reality they can be oncogenic in one situation but tumor-suppressive in another. This dual-function nature, which sometimes hampers our understanding of tumor biology, has several manifestations: (1) Most canonically defined genes have multiple mRNAs, regulatory RNAs, protein isoforms, and posttranslational modifications; (2) Genes may interact at different levels, such as by forming chimeric RNAs or by forming different protein complexes; (3) Increased levels of tumor-suppressive genes in normal cells drive proliferation of cancer progenitor cells in the same organ or tissue by imposing compensatory proliferation pressure, which presents the dual-function nature as a cell–cell interaction. All these manifestations of dual functions can find examples in the genes along the CCND–CDK4/6–RB axis. The dual-function nature also underlies the heterogeneity of cancer cells. Gene-targeting chemotherapies, including that targets CDK4, are effective to some cancer cells but in the meantime may promote growth or progression of some others in the same patient. Redefining “gene” by considering each mRNA, regulatory RNA, protein isoform, and posttranslational modification from the same genomic locus as a “gene” may help in better understanding tumor biology and better selecting targets for different sub-populations of cancer cells in individual patients for personalized therapy.

Keywords: CDK4, cyclin D1, oncogenes, tumor-suppressor gene s, targeted chemotherapy, radiation therapy


Cancer regulatory genes are often dichotomized to oncogenes and tumor-suppressor gene s. Those that promote cell proliferation or migration and/or inhibit differentiation or cell death are considered oncogenes, which may be amplified in tumor cells with gain of function. Conversely, those that inhibit cell proliferation or migration and/or promote differentiation or cell death are regarded as tumor-suppressor gene s, which may be deleted from tumor cells with loss of function. Despite being oversimplified, this dichotomy has greatly helped in delineating molecular mechanisms behind tumor biology. However, in reality, virtually all cancer regulatory genes manifest oncogenic features in one situation while showing tumor-suppressive properties in another.1 To provide examples, in this primer we review the dual functions of cyclin-dependent kinase 4 (CDK4) as well as its regulatory CCNDs (cyclins) and other proteins and analyze the meaning of the dual functions from the evolutionary point of view, as assigning multiple functions to one single gene can make the genome more efficient and thus smaller. Since CDK4 is a key target for cancer chemotherapies and some of its inhibitors such as PD0332991 are currently on clinical trials, the clinical relevance of this dual function nature of genes to the CDK4 inhibitors in particular and other targeted chemotherapies in general are reviewed and discussed as well.

There Is No Such Thing Called Tumor-suppressor gene s or Oncogenes

By the current estimation, the human genome encompasses about 3.0–3.2 billion bases but only slightly over 20 000 protein coding genes,2,3 when those open reading frames smaller than 100 amino acids are regarded as non-coding and are thus excluded.4 Human exome, the part of the genome formed by all (roughly 180 000) exons that encode amino acids, constitutes about 1% of the whole genome, or about 30–32 megabases of DNA.2,3,5-7 These figures together suggest that the majority of the human genome may not function via canonically defined genes, or, to keep the notion that the human genome functions via the “gene” that is the basic physical and functional unit of heredity, we need to redefine the “gene”, as pointed out by Gerstein et al.8

Most genes can be expressed to multiple RNA transcripts by using different transcription initiation sites or termination sites, and some of the transcripts are pre-mRNAs, while others may be precursors of regulatory RNAs. By alternative splicing, one pre-mRNA may produce not only different mature mRNAs but also different regulatory RNAs, such as long non-coding RNAs, microRNAs, and small interfering RNAs. One mature mRNA may yield different protein isoforms by using different translation start codons or stop codons. Each of the protein isoforms may undergo many posttranslational modifications, such as phosphorylation, methylation, and even splicing.9,10

For many genes, each of their pre-mRNAs, mRNAs, regulatory RNAs, protein isoforms, and protein modifications may function differently or even oppositely. For instance, the long mRNA form of the Bcl-x gene, i.e., Bcl-xL, encodes an oncoprotein, whereas its short mRNA form, i.e., Bcl-xS, encodes a tumor suppressor.11 In the FANCL gene, on the other hand, it is the long mRNA form that is tumor-suppressive, while the short form is oncogenic.12 The titin gene expressed in muscle encodes the largest (4.2 MDa) and the third most abundant protein in the human, with over one million mRNA variants derived from different alternatives of splicing of its 363 exons.13,14 At the DNA level, many genes can change their functions by mutating their sequence, as exemplified by p53, for which over 30 000 mutation types have been identified.15 While the wild-type (wt) p53 often manifests tumor-suppressive features, some of its normal mRNA variants16 or DNA mutants17,18 are oncogenic. Actually, p53 was originally taken to be an oncogene, because its protein level is high in many cancers. Gene mutations can also change any of the above-described mechanisms for gene product multiplicity; for instance, mutations at splice sites can change the splice pattern.19 More complexly, nucleotide change can also occur at the mRNA level by RNA editing that, unlike DNA mutation, provides cells with a temporary and reversible mechanism for altering protein sequence.

In addition to the gene product multiplicity, interactions among genes also diversify genes’ functions at yet another level of complexity. The best-known manner of interaction is the formation of complexes among different proteins, as to be detailed later in this primer for the complexes containing CDK4. Chromosomal translocation, which results in a fusion gene and ensuing chimeric RNA that may or may not encode a fusion protein, can be regarded as another manifestation of gene–gene interaction. One less-discussed manner is that DNA mutation or RNA editing of a regulatory gene may affect products of its target gene via any of the above-described mechanisms. For example, mutation of some splisosome genes can change titin protein isoforms.20 Formation of chimeric RNA at the RNA level,21 such as via trans-splicing, can also be regarded as a manner of gene–gene interaction. Actually, a new definition of “gene” is needed8 to cover such observations, such as that the 7 Msh4 mRNA variants in mouse involve sequences from 4 different chromosomes,22 and that there may be many chimeric RNAs in human formed between nuclear RNA and mitochondrial RNA.23

Summarizing the above information with 2 examples of the extremes, i.e., that the titin gene is expressed to over a million mRNA variants, and that p53 has over 30 000 mutation types leads us to a conclusion that the whole genetic repertoire consists of not just 20 000 genes but 20 000 groups of genes, if each of the pre-mRNAs, mature mRNAs, regulatory RNAs, protein isoforms, and protein modifications is considered one individual “gene” with somewhat different functions. The real number of gene groups is actually much larger, because both strands of the DNA double helix in most loci, including unannotated ones, are transcribed.24-26 Redefining “gene” in this way explains better the so-diversified human biology and related sociology relative to the canonical gene definition.

Each gene is just a tool used by the cell to elicit some specific function, and whether it should be oncogenic or tumor-suppressive is more up to the cell in a particular situation than to the gene per se, similar to a weapon that can be used by a person to defend or to attack. To use an analogy, an actor may play the hero in some movies but the villain in some others, depending on the script. Some actors almost always play the hero, making us forget that they can also do well as a negative role if required. Similarly, p53 usually acts as a tumor-suppressor gene , but, if the cell needs it, it can act as an oncogene as well, as described above. In conclusion, each gene is just an actor or actress whose role is assigned by the host cell to deal with a particular situation.

There is probably evolutionarily determined programming for which (canonically defined) genes use permanent DNA mutations or flexible RNA editing and for which other genes use the aforementioned means such as alternative splicing or protein modification to respond to environmental changes. Obviously, p53 is allowed to more often change its DNA sequence, whereas titin has more mRNA variants as alternatives. Actually, the United States adapts this elegant way to prevent any chance of administrative chaos by listing 16 persons on the presidential line of succession in the Constitution, with the vice president, the speaker of the House, the president pro tempore of the Senate, and the Secretary of State at the top of the list. Similarly, human cells reserve over 30 000 mutation types of p53 as backups to deal with different situations. Since about 12% single-nucleotide polymorphisms in the human genome are actually harmful,27 some mutations may actually be more beneficial than the wt to the host.27-29 An intriguing question is whether cells can convert some mutations back to wt if the situation returns to normal, since such “back mutation” or “reverse mutation” of inherited mutations has been well documented.30-33 It deserves attention whether during sporadic carcinogenesis there is a short period of window for such reverse mutation or secondary mutations, both of which have been observed for the BRCA1 and BRCA2 genes after chemotherapy of some cancers,34-36 leading to spontaneous abortion of the carcinogenesis. Some mutations of a gene occur more frequently than the others and are thus called “hot spots”, which are reminiscent of those at the top of the presidential line of succession as the backups for the “wild-type” (current) president of the USA. p53 knockout animals or cell lines are so often used in cancer research with a rationale that “p53 functions are often lost in cancer cells”. Unfortunately, this rationale is much closer to wrong than to right and has little human relevance, because cancer cells rarely lose the entire 19148-bp human p53 gene. Instead, the vast majority of cancer cells manifest one or some of the 30 000 alterations (including partial deletions) that in many cases lose some tumor-suppressive functions but gain some oncogenecity.

In a Tissue, Tumor Suppressors in Normal Cells Drive Proliferation of Pre-Malignant Cells

In an in vivo situation, cancer progenitor cells, e.g., initiated cells that are collectively dubbed hereinafter as pre-malignant cells regardless of how they have emerged, need to proliferate continuously in order to evolve toward a malignant state. Many, probably most, environmental tumor-promoting agents such as chemical carcinogens and physical (e.g., radiation) or biological (e.g., virus) factors that drive their proliferation are actually inhibitors of cell proliferation, as detailed before.37 When insulted by these tumor-promoting agents, called tumor promoters hereafter, normal cells stop replicating, which is referred to as “mitoinhibition”, to avoid DNA damage and avoid passing damaged DNA to their progenies. This mitoinhibition is likely achieved by elevated expression of tumor-suppressor genes such as p53, probably in association with decreased expression of oncogenes (Fig. 1). However, the organ or tissue still has to regenerate a certain number of cells to balance the routine cell loss. This need for regeneration is actually enhanced by tumor promoters, not only because decreased expression of oncogenes often weakens cell survival, but more also because elevated expression of tumor-suppressor gene s often induces cell death, which is commonly considered by peers as apoptosis but by us as “stress-induced cell death (SICD)”, a programmed cellular death mechanism different from physiological apoptosis.38,39 “Fortunately”, those pre-malignant cells that already exist in the organ or tissue are resistant to the effects of tumor promoters, i.e., being less mitoinhibited and having less SICD potential, probably due to inactivation of some critical tumor-suppressor gene s and/or activation of some critical oncogenes (Fig. 1). These cells with the “resistant phenotype”40 are the only ones capable of proliferating, thus replicate robustly to compensate for the cell loss of the organ or tissue while evolving to a malignant state. Therefore, activation of tumor-suppressor genes in the normal cells provides an impetus for pre-malignant cells to proliferate. Because of this interaction between pre-malignant cells and their surrounding normal cells, tumor-suppressor genes are as important as oncogenes in driving carcinogenesis in vivo, which is another manifestation of genes’ dual-function nature at the tissue level. Knockout of a tumor-suppressor gene from the whole tissue or organ will weaken the mitoinhibition and decrease the SICD potential of the normal cells, thus decreasing the compensatory proliferation pressure onto the pre-malignant cells. This major but less often discussed mechanism of carcinogenesis is one of the major reasons why animals with a tumor-suppressor gene deleted, such as p53−/− mice, do not develop cancer as efficiently as some peers expected and do not much resemble the human situation, since no tumor-suppressor gene is deleted from all cells of a human organ or tissue as a genetic basis for a sporadic cancer (germline mutation triggered carcinogenesis starts embryonically and thus is different). If one day it becomes applicable to knockout one gene from only one single cell of an organ or tissue with its surrounding cells intact, this “mitoinhibition-resistant phenotype” principle can be further tested, although, as reviewed before,37 it has been tested intensively by Farber’s team41-47 and many other researchers48-50 since it was proposed by Haddow in 1938.40

graphic file with name cc-13-1677-g1.jpg

Figure 1. Depiction of how tumor-suppressor genes (TSG) promote carcinogenesis via the “mitoinhibition-resistant phenotype” principle, with liver carcinogenesis as an example. There already is a cell in the liver that has been initiated to undergo carcinogenic process, due to such as spontaneous or chemical-induced mutation of some critical gene(s). This so-called “initiated cell” may show decreased expression of TSG and/or increased expression of oncogenes (ONG). If the liver encounters a chronic insult from some tumor-promoting agent (promoter) such as a long-term exposure to a chemical carcinogen, normal hepatocytes will react by increasing their TSG expression and likely also decreasing their ONG expression, so as to arrest cell growth to prevent DNA damage or to repair damaged DNA. If the DNA damage is irreparable, the hepatocyte will die of SICD, which will further enhance the demand for regeneration. Partly because of its low TSG levels and/or high ONG levels, the initiated cell is resistant to the promoter, i.e., being much less growth-arrested and having a less potential to SICD, and thus proliferates robustly to compensate for the cell loss of the liver, eventually evolving to a cancer (Ca). Therefore, sporadic cancer only occurs in those organs or tissues that have a routine cell turnover, but not in those that can no longer regenerate such as neurons or heart muscle.

Many genetically engineered animal models of carcinogenesis, usually with 2 or more genes manipulated, such as in ras-and-myc double transgenic mice, can produce solid tumors at 100% incidence, i.e., in all animals.51,52 However, although in these models all cells in the target organ or tissue bear the same genotype, usually only one to several cancer masses appear until the animal capitulates.51,52 Since there are millions of target cells in the same organ or tissue, the carcinogenesis efficiency is less than one of a million. In other words, there are few, if any, genetically engineered animal models that can convert even a tiny portion of individual target cells into individual cancer masses, although at the animal level the penetrance is 100%. This situation, which to our knowledge has not been mentioned and addressed in the literature, has been puzzling us for a long time and may not be explained until we are able to manipulate genes in single cells of the target organ.

For Example, the CCND–CDK4/6–RB Axis Also Plays Tumor-Suppressive Roles

CDK4 or its close sibling CDK6 binds to 1 of the 3 CCND proteins, mainly CCND1, to form a holoenzyme, and p27 or p21 protein facilitates the CDK4/6-CCND assembly.53 The holoenzymes phosphorylate RB1 or its sibling p107 or p130. Once phosphorylated, these RB proteins are inactivated and release E2F1 or other E2F proteins to regulate transcription of many proliferation regulatory genes, usually leading to acceleration of G1 progression of the cell cycle. On the other hand, p16 and related proteins can bind to CDK4 and inhibit its kinase activity, thus collectively coined as inhibitors of CDK4 (INK4), although they also bind to CDK6. Many types of cancer manifest increased expression of CCND1 or CDK4 with high frequencies of gene amplification, which makes this CCND–CDK4/6–RB–E2F axis a key promoter of cancer formation and progression. Since the oncogenic property of this axis has been reviewed extensively, herein we just focus on its tumor-suppressive aspect, which, to our knowledge, has so far not yet been reviewed comprehensively.

It is often forgotten, and is thus worth mentioning, that CCND-CDK4/6 can drive cell proliferation but cannot render cells competent for proliferation, meaning that once a cell has fully differentiated and lost its replication capacity, CCND-CDK4/6 cannot make it proliferating. CDK4 is often used in combination with other genes to immortalize cells, as reviewed before,54 but likely the cells need to be relatively young. Aged cells that can only undergo a few rounds of replication are probably hard, if not impossible, to immortalize by a CDK4-containing regimen.

As reviewed extensively before,55 nearly 2 decades ago ectopic expression of CCND1 had already been shown to inhibit cell proliferation and induce apoptosis or senescence of several cell types56-61 in addition to driving cell proliferation, although such ectopic expression usually results in only modest accumulation of the CCND1 protein.62 Transformation of human fibroblasts by SV40 coincides with a downregulation of CCND1,63-65 whereas many cancer cell lines manifest a decreased CCND1 protein level compared with normal diploid fibroblasts,64-69 although fibroblasts are not vigorous controls for epithelial-originated cancers. In transgenic mice, c-myc induces mammary tumors while it inhibits CCND1.70 In the pioneer study by Land et al. in 1983, colonies of ras-transformed rat primary embryonic fibroblasts appear in the dish at very early time point, but the cells have not yet been immortalized and eventually die; however, co-transfection with c-myc can immortalize the ras-transformed cells.71 Since ras72,73 induces whereas c-myc37,54,55 inhibits CCND1 expression, decrease in the CCND1 may be a mechanism that prevents the death of ras-transformed cells and thus be a mechanism for the ras-myc collaboration in transformation.54,55

In a cohort of breast cancer cases, low proliferation was observed in tumors with low p16, with high CCND1 but normal or high p16 expression, or without cell cycle defects.74 A study of another cohort concludes that CCND1 upregulation is not associated with increased proliferative activity in the tumors, although loss of Rb1 expression is significantly associated with an increased proliferation of the tumor cells regardless of CCND1 abnormalities.75 Oyama et al. also show that the vast majority of invasive lobular breast carcinomas manifest overexpression of CCND1 protein, but most CCND1-positive cells do not stain for Ki67 (a marker for proliferating cells), suggesting that CCND1 may contribute to the tumor progression via a mechanism irrelevant to its promotion of cell proliferation.76 Indeed, CCND1 expression in human breast cancer does not seem to be clustered into the group of proliferation-related genes.77 However, while the above-described studies, using cultured cells, animals, and human samples, seem to suggest a lower CCND1 level in transformed or malignant cells, it may sometimes be a misperception, because this “feature” may simply reflect a changed cell cycle distribution with decreased G1 and increased S fractions. This is because in cycling cells CCND1 protein level starts to elevate in the G2 phase of the cell cycle and remains high in the M phase and the G1 phase of the next cycle, but it levels off in S phase to allow DNA synthesis.72,73 Therefore, a smaller G1 fraction and a larger S fraction seen in transformed or malignant cells that grow faster, relative to the normal counterparts, will collectively show a decreased CCND1 level (Fig. 2). This decrease may be discerned on immunoblots, but immunohistochemical staining intensity may still be stronger in individual malignant cells than in normal cells, according to our experience. Some proliferation markers such as Ki67 and proliferation cell nuclear antigen (PCNA) stain cells at the S and G2–M phases and thus may or may not statistically differ from CCND1 staining, depending on how large the G2–M portion is, which is usually the smallest wherein both CCND1 and proliferation markers are highly expressed (Fig. 2).

graphic file with name cc-13-1677-g2.jpg

Figure 2. Illustration of the cell cycle and CCND1 relationship. In slowly growing tissues or cultures, G1-phase cells constitute the largest portion, while the S-phase portion is much smaller, whereas the situation is the opposite in fast-growing tissues or cultures. Cells in the G2/M phases usually constitute the smallest portion, although they may still be larger in the fast-growing than in the slow-growing tissues or cultures. CCND1 level is higher in G2/M and G1 but lower in S, and thus cannot sensitively reflect the proliferative rate.

CDK4, CDK6, CCND2, and CCND3 may manifest tumor-suppressive features as well in some situations. Transfection with a CDK4R24C or CDK6R31C mutant that cannot be inhibited by p16 may lead to cell death.78 Even transfection with the wt CDK6 can cause cell death.79 Overexpression of CDK6 and CCND1 causes apoptosis of chondrocytes without enhancing proliferation.80 Ectopic expression of CDK6 inhibits cell proliferation, which is probably elicited via the p53 and RB pathways as p53 and p130 are increased.79,81,82 Actually, overexpression of CDK6 or CCND3 in transgenic mice inhibits skin tumor development and, in the case of CCND3, the CCND3–CDK4 kinase activity may be involved.83,84 CCND3 is required for differentiation of several tissues.85-87 CCND2, including its splice variant CycD2SV,88 is actually regarded by many peers as a tumor suppressor, because its expression level is decreased in many malignancies,89-93 whereas restoration of its expression prevents prostate carcinogenesis.94

p16 is commonly regarded as a tumor suppressor, but it sometimes manifests oncogenic properties. Its mRNA and protein levels are unexpectedly higher in basal cell carcinomas than in normal human skin.95 A subset of breast, ovarian, and lung cancers manifests high levels of p16, which is usually associated with Rb1 mutation.96-100 p16 is shown to be required for the survival of some cervical carcinoma cell lines in a CDK4/6-dependent manner.78 Free p16 protein in the cytoplasm has been shown to promote migration of liver cancer cells in culture, which is somewhat opposite to the functions of the cytoplasmic CCND1 and CDK4.101

The canonical tumor suppressor RB1 sometimes helps cell proliferation or survival102,103 and is overexpressed or amplified in some cancers,104 whereas its loss may induce cell death.103,105,106 More surprisingly, transgenic mice expressing a constitutively active Rb1 gene actually develop mammary gland cancer, which is thought to occur via the aforementioned “mitoinhibition” mechanism, i.e., the growth suppression of active RB1 protein imposes a selection pressure in favor of transforming mutations that accelerate cell proliferation.107 Since RB1 regulates a plethora of genes that inhibit cell proliferation or promote apoptosis, besides those cell cycle-promoting genes such as CCNE1, CCNA2, and PCNA,108 it remains possible that RB1 functions differently in normal cells and in cancer progenitor cells. RB1 also plays roles in replication of DNA, repair of DNA damage, and G2/M progression of the cell cycle.108 The Rb1 gene produces not only the wt, but also several N-terminally truncated proteins by alternative splicing,109,110 by using downstream ATG start codons, or by caspase-cleavage.111-117 There are at least 932 mutations identified for the Rb1,118 some of which may result in not only oncogenic proteins but also smaller protein isoforms.119 These smaller protein isoforms have different abilities in growth inhibition, with some N-terminal truncated ones being more potent, partly because the C-terminus contains a docking sequence that cannot be inactivated by CCND-CDK4/6.120 However, chimerism of the wt with some smaller isoforms manifests decreased tumor suppression.121 Whether different mechanisms for generation of different RB1 protein isoforms are differentially used in cancer and non-cancer cells remains elusive.

Being the major target of RB1, E2F1 not only activates transcription of genes that drive cell proliferation but also induces cell apoptosis in some situations,122-124 such as when RB1 is lacking.125 E2F1 knockout mice develop spontaneous tumors in a number of tissues, demonstrating its tumor-suppressive property in vivo in some situations.126

CCND1 Not Only Drives G1 Progression, but Also Arrests It, Allowing DNA Repair

The functional duality of CCND1 may be attributed more to the cellular context than to the CCND1, per se, but until now it still baffles us as to what cellular properties direct the high-CCND1 cells to replicate, arrest proliferation, undergo apoptosis, or enter into senescence. Serving as a cellular sensor for extracellular growth stimuli, CCND1 is activated by Ras, ERK, and many growth stimuli-induced molecules at the G2 phase and remains high until the next G1 phase.72,73 At the G1–S boundary, CCND1 protein is quickly degraded to allow DNA synthesis (Fig. 2). Phosphorylation of CCND1 protein at T286 by glycogen synthase kinase 3β (GSK-3β) is required for CCND1 ubiquitination by 26S proteasome, and this degradation is enhanced by its binding to CDK4,127,128 but T286 phosphorylation does not promote its disassociation from CDK4.129 CCND1 that is unbound to CDK4 is more rapidly degraded in some,130 but probably not all,129 cells via ubiquitination without phosphorylation at T286.129

If a cell encounters a genomic insult such as radiation that causes DNA damage, the cell arrests G1 progression in part by rapidly degrading the CCND1 protein,131,132 or, if it is too late for the cell to hold itself at G1, it arrests S progression by sustaining a high CCND1 level.133 Both G1 and S arrests allow the cell to repair its DNA and to prevent passing the genetic change to the daughter cells. If the DNA damage is irreparable, the sustained CCND1 protein at the S phase will instead trigger more DNA damages and ensuing apoptosis73,134 that is considered by us as SICD.38,39 Although CCND1 is bound by CDK4 and p21 at the G1, genomic stress only degrades CCND1, but not CDK4 and p21. In consequence, CDK4 is released to bind another CCND1 molecule and brings it for degradation while p21 is released to bind CDK2, thus sequestering CDK2 activity and further arresting G1 progression.131,132 It has been shown that CCND1 and CCNA2 undergo cleavage by caspases during irradiation-induced apoptosis of Xenopus embryo, and the cleaved CCND1, alone or in complex with CDK4/6, is much more potent in binding to p27, leading to reduced phosphorylation of RB1.135

Irradiation-induced apoptosis has been shown to be more evident in CCND1 knockout (−/−) mouse fibroblasts (MEFs) than in the wt counterparts, suggesting that CCND1 confers protection against irradiation.136 However, ectopic expression of CCND1 in several premalignant and malignant cell lines of breast origin also enhances irradiation-induced apoptosis.137,138 This incongruity may in part be related to the method of irradiation, as Shimura et al. show that single irradiation downregulates CCND1 protein level, but fractionated irradiation causes CCND1 accumulation via DNA-PK/AKT-mediated inhibition of its proteolysis.133 Chronic irradiation is thought to result in cytoplasmic accumulation of CCND1 protein, wherein it binds and thus sequesters Bax, leading to inhibition of mitochondrial-mediated cell death.139 Consonantly, CCND1 overexpression is shown to be associated with poor prognosis in oral and head and neck cancers after radiotherapy or concurrent chemoradiotherapy.140,141 The persistently high level of CCND1 during the S phase inhibits DNA replication by preventing replication fork progression, which will, in turn, trigger double-strand breaks.133 The cell will then remove the aberrant replication fork and reconstruct the fork to resume DNA replication.133 It is only CCND1 protein, but not the CCND1–CDK4 complex, that binds to the fork,133 meaning that this function is irrelevant to CDK4.142-144 Hence, the role of CCND1 varies among different cell types,145 varies between acute and chronic irradiations,133 and has CDK4-dependent146,147 or -independent133,148 mechanisms. The paradoxical roles of CCND1 in driving G1 progression on the one hand, and in promoting DNA repair on the other, again shows its functional duality.

Association with Different Proteins Diversifies CCND, CDK4/6, or CCND-CDK4/6 Functions

At least 132 proteins can bind to CCND1 in breast cancer cells,143 some of which bind to CCND1 in a way independent of CDK4, such as the DNA repair proteins RAD51, BRCA1, BRCA2, PCNA, and replication factor C.143,144 BRCA2 brings CCND1 to damaged chromosomal sites, where CCND1 recruits RAD51 to perform homologous recombination (but not other types of DNA repair).142-144 Another group of CCND1-binding proteins, which may be mechanistically related to its growth promotion,149 belongs to transcription factors,150 such as Sp1,151,152 DMP1,149 as well as steroid hormone and thyroid hormone receptors, as reviewed previously.55 Interestingly, CCND1 binds to and activates estrogen receptor α,153 but it binds to and inhibits androgen receptor.154,155 Moreover, many CCND1-regulated genes encode molecular chaperones.156,157

p16 and probably also other INK4 members form mainly binary INK4–CDK4/6 complexes. INK4–CDK4/6–CCND ternary complexes may also be formed at a lesser abundance158,159 and probably mainly in senescent cells,160 but inhibition of CDK4 kinase activity by p16 is not affected by whether CDK4 is alone or is bound to a CCND.161 Many proteins that contain ankyrin-repeat domain, such as IkBalpha, can bind to CDK4 as well.162 Gankyrin that contains 7 ankyrin repeats is known to compete with p16 in binding to CDK4, but it does not inhibit CDK4 activity; actually, this difference makes gankyrin an oncoprotein but makes p16 a tumor suppressor.163,164 Survivin competes with p16 or p21 for binding to the CDK4165 and brings CDK4 into the nucleus, which is a mechanism for its promotion of cell cycle entry and cell survival.166,167 Cdc37 is a molecular chaperone important for the stability and activity of several protein kinases; like Hsp90, Cdc37 binds to the N terminus of CDK4 by competing with p16.168 The portion of cdc37-bound CDK4 is largely devoid of CCND1, suggesting that this complex functions to prepare CDK4 for CCND1 interaction.169,170 In addition, some viral proteins, such as the HTLV-1 Tax protein,171 can bind to CDK4 as well. Unlike CDK4 and other CDKs, CDK6 can bind to androgen receptor and activates its transcriptional activity, which does not require its kinase activity and is independent of CCND1.172 Actually, binding of CCND1 to CDK6 inhibits the transactivation of the androgen receptor.172

Although p27 and p21 are similar in their CDK binding regions, only p27, but not p21, can bind to CDK4 alone, but in mid-G1 p21 assembles CCND1–CDK4 complex by replacing p27 in binding to CDK4.160 Binding of p27 to CCND1-CDK4 may or may not inhibit CDK4 kinase activity, depending on whether the Y88 and Y89 of p27 are phosphorylated, in part because p27 can also bind to CCND1, but not just CDK4.173,174

It goes without saying that CCND–CDK4/6 holoenzymes bind to their substrates as well. In addition to RB1, p107, and p130, 68 proteins have been identified as CCND1–CDK4/6 substrates,175 including FOXM1, Smad3, Cdt1, MARCKS, and PRMT5.53,176 CCND1-CDK4 has also been reported to phosphorylate microtubule-associated protein Tau in neurons to promote neurite extension.177 Filamin A can bind to CDK4 and may be a substrate of the CCND1–CDK4 complex as well, as regulation of its phosphorylation by CCND1 promotes cell migration and invasion.178 Moreover, p220NPAT may be a CCND2–CDK4 substrate.179 Thus, a gene has dual functions not only because it can produce multiple products, but also because one protein can have different partners and, if it is an enzyme like CDK4, different substrates.

Some Tumor-Suppressive Roles of CDK4 May Be Independent of CCND1 and Occur in the S and G2/M phases

A question that has been baffling us for a long time is why CDK4 has mainly been shown to function at the G1, while its main partner, CCND1, has already, as aforementioned, been elevated in the G2-M of the previous cycle. Our cloning of a CDK4 mRNA variant that lacks exon 2, coined as ΔE2, provides some of the answers.180 Initiation of protein translation of this variant is shifted from the canonical ATG start codon in exon 2 to an in-frame ATG in exon 3, resulting in an isoform lacking an N-terminal region of 74 amino acids that harbors not only the ATP binding sequence, but also the PISTVRE domain required for binding to the p16 and CCND. Co-immunoprecipitation assays confirm that this truncated protein has indeed lost the CCND1- and RB1-binding ability. Surprisingly, however, this ΔE2 protein can still drive the S–G2/M progression as does the wt CDK4. More surprisingly, it is as potent as its wt in enhancing serum starvation-related apoptosis of RB1-inactivated 293T and Hela cells181 but inhibiting the death of RB1-proficient HEK293 cells. It is also as potent as its wt in arresting the Hela, but not the HEK293, cells at the G1 when treated with a CDK4-inhibitory compound NPCD. These results suggest that CDK4 may use some CCND- and RB-irrelevant mechanisms not only in the well-established promotion of cell proliferation, but also in the unexpected inhibition of cell cycle progression and induction of cell death, somewhat similar to CDK6, which can activate androgen receptor transcriptional activity without involving its CCND1 binding and its kinase activity.172 A study on B-cell lymphoma has also suggested, though indirectly, the possible existence of cyclin-independent functions of CDK4.182,183 Since Warenius et al. have reported that the 251–256th hexapeptide (PRGPRP) of human CDK4 has killing effects on cancer cells,184 the growth arrest and apoptotic effects of the ΔE2 and the wt may be attributed to this C-terminal region. The dual-function nature, i.e., promotion of cell growth or death, has also been discerned for CDK8185 and thus is not unique to CDK4.

The above-described novel properties of CDK4, i.e.: (1) its CCND- and RB-independent functions; (2) its promotion of S and G2–M progression; and (3) its driving of G1 arrest and apoptosis, occur in a cell line-specific manner and have so far been observed only in some situations, wherein cells were treated with CDK4 inhibitors or were serum starved and then replenished.180 Since the ΔE2 isoform still differs from the wt CDK4 in some functions, it remains possible that some functions of the wt CDK4 at the G2-M may still involve CCND1 that has already been high at the G2 phase.72,73 Regardless of whether a CCND is involved, there actually is some inkling of CDK4 activity at the S to G2/M phases.130,186-188 Ectopic expression of the wt p16, but not its mutant, which cannot inhibit CDK4/6, lengthens the S-phase progression of MCF7 and other cancer cell lines;189,190 although the underlying mechanism is still unclear, the involvement of CDK4/6 remains highly possible in part because CDK4 is active throughout the cell cycle.188 Transient transfection with either a wt p21 or a dominant-negative CDK4 construct can suppress doxorubicin-induced apoptosis at the G2–M phase, indicating that the CDK4 mutant can act as a tumor suppressor, just like p21, during G2-M.191 CDK2 is shown to be required for p53-independent checkpoint control of the G2–M phase.192 Inactivation of RB1 by expressing polyomavirus large tumor antigen (PyLT) or SV40 increases CCNA2 expression and activates CDK2.193,194 Since CDK4 indirectly regulates CDK2 activity via such means as sequestration of p27 binding to CDK2, CDK4 may indirectly regulate G2/M checkpoint as well.

The CDK4 effects in the G2-M may be more evident in fully differentiated cells. In postmitotic neurons, cell cycle activation is essential for DNA repair, as suppression of CDK4/6 activity attenuates DNA repair.195 Likely, the CCND1–CDK4 complex is mainly localized in the cytoplasm of postmitotic neurons, because its internationalization into the nucleus causes apoptosis.195 Disruption of RB1 accelerates the G2–M progression in the presence of DNA damage by elevating the expression of a set of mitotic regulatory genes.196 Conditional deletion of the Rb1 gene reactivates cell cycle-regulating genes in skeletal muscle, indicating that an RB1-independent mechanism preserves the postmitotic state of differentiated cells.197 Adenovirus-mediated RB1 deletion in the liver of adult mice leads to DNA replication in the absence of productive mitotic condensation; the replication induced by RB1 loss is E2F-mediated and is associated with the induction of DNA damage and a non-transcriptional G2/M checkpoint that targets the accumulation of CCNB1.198 Inactivation of Rb1 by siRNA or other means can also achieve similar results.108,199

Inhibition of CDK4 for Cancer Therapy May Also Have Weaknesses

Inhibition of CDK4 activity is considered an attractive strategy for cancer chemotherapy.53,200-202 Many laboratory studies have indeed shown that chemical inhibitors of CDK4 kinase activity can cause cytostasis, senescence, and even death of cancer cells.175,200,203-212 Moreover, knockdown of CDK4 using shRNA also significantly increases radiation-induced apoptosis of malignant and nonmalignant breast cell lines without significantly altering cell cycle progression or post-irradiation DNA repair.213 Several chemical inhibitors specific for CDK4/6 have been on clinical trials, including PD0332991 (Palbociclib), LY2835219, and LEE011,200,214,215 of which the trials on PD0332991 have been promising for lymphomas and liposarcomas.216,217 Studies with cancer cell lines and ex vivo breast205,218-220 or prostate221 cancer tissues all suggest that PD0332991 is efficient in RB-competent cases, but the RB-deficient counterparts are relatively refractory. Treatment of glioblastoma cells with PD0332991 in an xenograft mouse model also suggests that RB-proficient cells are more sensitive,222 which makes sense, as RB-deficient cancer cells do not seem to require CCND1 for proliferation.130,223 A single dose of 10 Gy irradiation therapy followed by 7 d of treatment with PD0332991 increased, by 19% in comparison with irradiation alone, the survival of the transgenic mice that bear brainstem glioma.224 PD0332991 has also been shown to have an additive effect on irradiation of prostate cancer cells in culture.221

PD0332991 has been shown to reduce the overall irradiation toxicity without evidently compromising the therapeutic effect of the irradiation on autochthonous cancer in the mice,225 although it cannot be ruled out that some tumor cells are also protected. Co-administration of PD0332991 with carboplatin in mice bearing RB-competent breast cancer decreases the antitumor effect of the carboplatin, although the combination significantly attenuates the myelosuppression caused by carboplatin and thus is still beneficial.226 In contrast, RB-deficient breast tumors in mice are resistant to PD0332991, and concomitant administration of carboplatin does not have an additive effect.226 Treatment of RB-competent breast cancer cells in culture or in xenograft animals with PD0332991 in combination with genotoxic doxorubicin may have additive effects on cytostasis but actually antagonizes the doxorubicin’s cytotoxic effects.219 An indolocarbazole-derived CDK4 inhibitor has been shown to sensitize MDA-MB231 but antagonize MCF7, both being breast cancer cell lines, to irradiation, whereas more evident growth inhibition and apoptosis by the inhibitor alone are discerned in MCF7 than in MDA–MB231 cells.227 These results suggest that the chemotherapeutic effect of the CDK4 inhibitor is disassociated from its addition or antagonism to irradiation, both being cell type-specific. A more disturbing observation is that when taxanes or doxorubicin induces DNA damage in triple-negative breast cancer cells, a subtype of breast cancer with poor prognosis, PD0332991 shifts the DNA repair from homologous recombination (HR) to non-homologous end joining (NHEJ).220 Since HR is error-proof whereas NHEJ is error-prone, this shift may, presumably, cause more mutations and thus may promote cancer progression toward more aggressiveness.

CCND1 can inhibit migration of several cell types including breast cancer cells.145,210,228,229 This tumor-suppressor function is in part attributed to its inhibition of epithelial–mesenchymal transition (EMT), which is a morphological feature for cancer progression toward metastasis.229,230 A recent study shows that Panc-1 cells established from a primary pancreatic cancer are sensitive to PD0332991, whereas AsPC-1 cells established from ascites of a pancreatic cancer patient are resistant to it.231 In AsPC-1 and another relatively resistant pancreatic cancer cell line (COLO-357), PD0332991 induces expression of those genes that promote EMT,231 which not only suggests indirectly that the aforementioned inhibition of cell migration and EMT by CCND1 may involve CDK4 activity, but also implies that treatment with CDK4 inhibitors may facilitate cancer metastasis in at least breast and pancreatic cancers in the long run.

CCND1 has been shown to inhibit mitochondrial metabolism and biogenesis,145,232 and both addition and loss of CCND1–CDK4 functions have been shown to increase mitochondrial superoxide production and decrease the cell’s lifespan, indicating that an imbalance in mitobiogenesis may lead to oxidative stress and aging.233 These effects on mitochondria and ensuing aging may facilitate not only the progression of the existing cancer cells, but also the development of a second malignancy.234-236 Moreover, CDK4 inhibition may affect pancreatic β cells’ survival and insulin secretion,234-236 and the long-term consequence of this effect to patients needs to be evaluated.

More Information Is Needed For Fully Assessing CDK4-Targeted Cancer Therapy

PD0332991 and other CDK4 inhibitory compounds do not function by lowering the CDK4 or CCND1 level, but by competing with ATP for binding to CDK4/6.237-239 Actually, PD0332991 treatment increases the protein levels of CCND1211,212,219,231,240 as well as CDK4 and CDK6.231 However, it remains esoteric whether the elevated CCND1 and/or CDK4/6 contribute to PD0332991-induced cytostasis, senescence, or death of cancer cells, and whether the elevation is because decreased RB phosphorylation triggers a feedback regulation, because PD0332991 stabilizes these proteins, or because of other reasons. Whether these increased proteins are in a free status or in a binary or ternary protein complex is also unknown, although the abundance of the CCND1–CDK4 complex is shown to be modestly increased in a study.221 So far it is still elusive whether PD0332991-bound CDK4/6 has the same binding affinity to CCND or other partner proteins, despite the fact that the crystal structure of PD0332991–CDK6 complex has been studied.239 Persistently high level of CCND1 probably is more abnormal than that of CDK4 to the cell, since normally the CDK4 level and activity are relatively stable,188 whereas the CCND1 level should be “oscillating” around the cell cycle. We need to know whether accumulated CCND1 and CDK4/6 in the presence of PD0332991 have any effect on the cells, so as to make a decision whether PD0332991 should be used with an approach that decreases the levels of these proteins. Since currently there has not yet been an approach available for clinical use that can specifically decrease CCND1 or CDK4/6 protein, some peers are considering designing peptides or compounds that block other functional, such as the substrate-binding, domains of CDK4/6 proteins.241,242 Whether these alternatives, alone and in combination with PD0332991 or other ATP site blockers, are more effective is an intriguing question.

Treatment of the HepG2 liver cancer cells with a CDK4 inhibitor, flavopiridol, increases cellular ATP concentration up to 3-fold,161 whereas inhibition of CDK2-CCNA2 activity by p27 is known to be dependent on cellular ATP concentration.243 Therefore, it would be interesting to know whether CDK4 inhibitors attenuate the inhibitory effects of p27 and other endogenous CDK-inhibitory proteins by raising the ATP level. Why and how PD0332991 shifts drug-induced DNA repair from an error-proof to an error-prone mechanism220 should also be studied in more detail, as these details are prerequisite for making a correct decision on whether and how CDK4 inhibitors should be used in combination with irradiation and with genotoxic chemotherapeutic agents in cancer treatment.

Spontaneous tumors or cancers hardly regress spontaneously, but in experimental conditions, these lesions may be dependent on the inducer.244,245 For instance, in animals, coal tar-induced tumors in the skin,246 estrogen-induced cancers in several organs,247-251 as well as c-myc- and k-ras-induced lung cancer and lymphomas252 may regress after withdrawal of the inducer(s). Similarly, although spontaneous senescence should be irreversible, induced senescence has frequently been shown to be reversible.245,253-255 Blagosklonny’s group dichotomizes growth arrest to the earlier, reversible phase and the later, irreversible phase and points out that only the irreversible arrest is senescence.211,212,253,256,257 For those cells that are already at the end of their proliferative life span, i.e., are already at a permanent growth-arrested or senescent state, such as fully differentiated neurons and striated (both skeletal and cardiac) muscle, growth stimuli such as those acting via the MEK/MAPK pathways may still induce CCND1. For those cells that still retain a replication capacity but are growth-arrested, such as by p21 or p16 overexpression or by PD0332991, growth stimuli will induce CCND1 but eventually drive the cells to senescence if MTOR (mechanistic target of Rapamycin) is active to stimulate cellular mass growth and cellular super function.211,212,253,256,257 Likely in these situations, senescent cells display a super elevation of the CCND1 protein level as a biomarker for senescence.211,212,253,256,257 Entry into either a reversible growth arrest or a senescent state decreases the cell turnover, which is more efficient and affordable, relative to death from apoptosis, for some tissues to maintain the physiological cell number and thus physiological function.258 For those cells whose replication capacity is closer to the Hayflick limit, which is about 50 generations in vitro,259,260 entry into a growth arrest status saves them replication capacity, while the high CCND1 level prevents DNA synthesis and thus prevents passing damaged DNA to the progeny. Hence, even if the cells already bear irreparable DNA damage, they are allowed to survive at an arrested or senescent state to continue their service. Therefore, growth arrest or senescence is an equally effective tumor-suppressive mechanism as is apoptosis for the organ or tissue to protect itself.261 Nevertheless, it deserves further study in many more cell lines whether growth arrest induced by CDK4 inhibitors eventually turns to true, irreversible senescence, since senescent cells are, in general, relatively resistant to apoptosis258,262 and thus may be refractory to other apoptosis inducers such as radiation.

The Functional Duality of Genes Arouses New Considerations on Cancer Therapies

Dichotomy of cancer-regulatory genes to “oncogenes” and “tumor-suppressor genes” has, on the one hand, greatly helped us in learning molecular details of tumor biology, but has, on the other hand, created a lot of confusion for some cancer researchers as well. This is largely because virtually all these genes are dual-functional in reality, with those along the CCND–CDK4/6–RB axis as tangible examples. Much of the confusion may be eliminated if we redefine each of the mRNAs, regulatory RNAs, protein isoforms, and protein modifications produced from the same genomic locus as a “gene”. For instance, by this definition, the oncogenic bcl-xL and suppressive bcl-xS are 2 different genes.11, Unfortunately, this new gene definition still cannot eliminate all the perplexities, because dual-function phenomena occur at gene–gene and cell–cell interactions as well.

Cancer cells are characterized by their uncontrolled replication, which is often misunderstood as “faster growth”.263,264 A cancer mass usually has a much larger percentage of cells that are in a proliferating status, compared with the normal part of the host tissue or organ and to most other normal tissues or organs of the patient.263,264 However, this does not mean that individual cancer cells replicate faster than individual normal cells.264 Actually, the opposite is true in many occasions (Fig. 3). For example, many hematopoietic progenitor cells complete one round of the cell cycle in 12–24 h, while cells in many cancer types require 2–3 d for a cycle,264 with those dormant cancer cells and those cancer stem cells that proliferate only occasionally as the extremes. In other words, that some cancers grow very fast is often because they have a very large fraction of cells in the proliferating status, but not because individual cancer cells replicate faster. Cancer tissues usually have a larger fraction of dead cells than their host tissues or organs as well (Fig. 3),265 although the much larger fraction of proliferating cancer cells still makes the tumor mass larger and larger until, if without treatment, about 40 tumor doublings, when the patient capitulates.264 Despite that it is a misconception in many occasions, “cancer cells grow faster” has been a general rationale for anticancer drug development. Indeed, most classical chemotherapies target cell proliferation and have some side-effects in common, including low blood cell counts, skin itch, hair loss, and gastrointestinal (GI) tract symptoms. This is because the drugs also inhibit proliferation of those normal cells that are highly proliferating, including hematopoietic progenitor cells, skin epithelial cells, hair follicles, and GI epithelial cells. When most cells in a tumor lump do not actually replicate faster than the normal cells in these tissues, this type of side-effect is more severe.

graphic file with name cc-13-1677-g3.jpg

Figure 3. Illustration of cell type distributions in normal and cancerous tissues and the locations of the 2 different types of adverse effects of chemotherapies. The data are rationalized from an average of most situations and thus may not be applied to a given case. A much larger percentage of cells in a cancer tissue is in cycling, i.e., in a proliferating status, relative to those normal tissues or organs that retain regeneration capacity. On the other hand, proliferating cells in many cancer types usually take a longer time to complete a cell cycle than many proliferating cells in normal tissues, such as those hematopoietic progenitor cells and those in the basal layer of skin epidermis and gastrointestinal mucosa. Common side-effects of traditional chemotherapeutic agents occur systemically, i.e., are the off-target effects on the proliferating cells in the normal tissues or organs. In contrast, the potential adversities of targeted therapies that are derived from the functional duality of targeted genes are attributed to the heterogeneity of the cancer cells, which, in turn, is related to the heterogeneous expression and function of the canonically defined target gene within the same cancer tissue.

In addition to the above-described adversities that occur systemically, i.e., outside the cancer, many targeted therapies, which target a particular gene such as CDK4 and are considered “magic bullets”,266 may also have another type of weakness, including the induction of resistance as well as invasion and metastasis, which occur within the cancer per se (Fig. 3). Cancer cells in any individual patient are heterogeneous and, even worse, continue to be more heterogeneous as therapy goes on, especially when the treatments are genotoxic. The dual-function nature of genes is one of the main reasons behind the heterogeneity and, thus, a main mechanism underlying these in-tumor adversities. Targeting one gene or one form of the gene products may be effective for some cancer cells, but in the meantime may promote survival or progression of other cells, eventually manifested as tumor recurrence or metastasis, besides the possible development of a second primary tumor. For example, the canonically oncogenic ERK/MAPK signaling also has a strong tumor-suppressive activity in culture cells, in animals, and probably in a subset of more aggressive cases of different human cancers as well, as discussed by Deschenes-Simard et al.267 Blocking this pathway may promote the growth and progression of this subset of cases. Hence, knowing the dual-function nature of genes and redefining “genes” may help us not only to clear up much of confusion, but also to steer our train of thought to a new track in finding targets for cancer therapy. For instance, targeting the bcl-xL or the splicing mechanism that converts bcl-xL to bcl-xS may be better than targeting the expression of the bcl-x gene.11 A good combination of targeted therapies to simultaneously block 2 or more pathways may be synthetically lethal to some cancer cells,268,269 but in the meantime they probably are synthetically promoting to some other cancer cells as well. Currently, most animal studies for testing drug efficacy use xenograft or orthotopic graft model that transplants culture cells or tumor tissue developed from a single cell line to an immunodeficient mouse. Our assumption rationalizes that using primary tumor tissue from an animal or a human patient as the tumor source, which contains heterogeneous cancer cell populations, will be a better animal model to test the efficacy of anticancer drugs.

A drawback closely related to selection of therapy targets is that we still know too little about cancer invasion and early metastasis and thus lack specific targets for these stages of cancer, while metastasis is the major reason why solid cancers are lethal. There is evidence suggesting that invasive and early metastatic cells, which locate in the invadopodia,270 are quiescent (G0 phase) and thus are small in number.271 While those cycling cells can be killed by chemo- or irradiation therapies, the quiescent cells are the ones that may recur later. Hence, identification of target “genes” (defined by our new concept) specific for those invasive and early metastatic cells in the invadopodia is imperative. Moreover, so far tumor shrinkage is still the major, if not the only, efficacy criterion for evaluation of whether a drug can enter into the pharmaceutical pipeline, which may be a manmade barrier that inhibits the development of metastatic-specific drugs, as such drugs may not cause tumor shrinkage in xenograft animal models, as pointed out by Rosel et al.272 Therefore, there is also a need for evaluating invasion or metastasis as criteria for drugs to enter into the pharmaceutical pipeline.272

Summary

Genes defined canonically have dual functions, which are manifested at multiple levels: (1) Different mRNAs, regulatory RNAs, protein isoforms and protein modifications derived from the same genomic locus may function differently and even oppositely; (2) Genes may interact at different levels, such as by forming chimeric RNAs and by forming different protein complexes to exert different and even opposite functions in different situations; (3) High levels of tumor-suppressor genes in normal cells drive proliferation of cancer progenitor cells in the same organ or tissue by imposing compensatory proliferation pressure, which presents the functional duality of genes as a cell–cell interaction at the tissue level. This “mitoinhibition-resistant phenotype” principle can be further tested if one day technology allows the manipulation of genes specifically in cancer progenitor cells without affecting their normal surrounding cells in the same organ or tissue. All these manifestations can find tangible examples along the CCND–CDK4/6–RB axis. The dual-function nature of genes, which often creates confusion to hamper our understanding of tumor biology, may be a main reason behind the heterogeneity of cancer cells in individual patients. Redefining “gene” by considering each of the RNAs, proteins or protein modifications from the same genomic locus as an individual “gene” should help us in clearing up much of the confusion on tumor biology and in selecting target molecules for treatment of different subpopulations of cancer cells in individual patients.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We also want to thank Fred Bogott, MD, PhD, at Austin Medical Center, Austin, Minnesota, for his excellent English editing of this manuscript. This work was supported by a grant from the Department of Defense of United States (DOD Award W81XWH-11-1-0119) to D.J. Liao and a grant from the National High Technology Research and Development program of China (2012AA020206) to Xiaomin Lou. The funding agencies had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Orr B, Compton DA. A double-edged sword: how oncogenes and tumor-suppressor gene s can contribute to chromosomal instability. Front Oncol. 2013;3:164. doi: 10.3389/fonc.2013.00164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Belizário JE. The humankind genome: from genetic diversity to the origin of human diseases. Genome. 2013;56:705–16. doi: 10.1139/gen-2013-0125. [DOI] [PubMed] [Google Scholar]
  • 3.Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ, Nickerson DA, Shendure J. Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet. 2011;12:745–55. doi: 10.1038/nrg3031. [DOI] [PubMed] [Google Scholar]
  • 4.Kageyama Y, Kondo T, Hashimoto Y. Coding vs non-coding: Translatability of short ORFs found in putative non-coding transcripts. Biochimie. 2011;93:1981–6. doi: 10.1016/j.biochi.2011.06.024. [DOI] [PubMed] [Google Scholar]
  • 5.Ng SB, Turner EH, Robertson PD, Flygare SD, Bigham AW, Lee C, Shaffer T, Wong M, Bhattacharjee A, Eichler EE, et al. Targeted capture and massively parallel sequencing of 12 human exomes. Nature. 2009;461:272–6. doi: 10.1038/nature08250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pennisi E. Genomics. ENCODE project writes eulogy for junk DNA. Science. 2012;337:1159–61, 1161. doi: 10.1126/science.337.6099.1159. [DOI] [PubMed] [Google Scholar]
  • 7.Skipper M, Dhand R, Campbell P. Presenting ENCODE. Nature. 2012;489:45. doi: 10.1038/489045a. [DOI] [PubMed] [Google Scholar]
  • 8.Gerstein MB, Bruce C, Rozowsky JS, Zheng D, Du J, Korbel JO, Emanuelsson O, Zhang ZD, Weissman S, Snyder M. What is a gene, post-ENCODE? History and updated definition. Genome Res. 2007;17:669–81. doi: 10.1101/gr.6339607. [DOI] [PubMed] [Google Scholar]
  • 9.Volkmann G, Mootz HD. Recent progress in intein research: from mechanism to directed evolution and applications. Cell Mol Life Sci. 2013;70:1185–206. doi: 10.1007/s00018-012-1120-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Aranko AS, Oeemig JS, Kajander T, Iwaï H. Intermolecular domain swapping induces intein-mediated protein alternative splicing. Nat Chem Biol. 2013;9:616–22. doi: 10.1038/nchembio.1320. [DOI] [PubMed] [Google Scholar]
  • 11.Akgul C, Moulding DA, Edwards SW. Alternative splicing of Bcl-2-related genes: functional consequences and potential therapeutic applications. Cell Mol Life Sci. 2004;61:2189–99. doi: 10.1007/s00018-004-4001-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Yuan C, Xu N, Liao J. Switch of FANCL, a key FA-BRCA component, between tumor suppressor and promoter by alternative splicing. Cell Cycle. 2012;11:3356. doi: 10.4161/cc.21852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Krüger M, Linke WA. The giant protein titin: a regulatory node that integrates myocyte signaling pathways. J Biol Chem. 2011;286:9905–12. doi: 10.1074/jbc.R110.173260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Guo W, Bharmal SJ, Esbona K, Greaser ML. Titin diversity--alternative splicing gone wild. J Biomed Biotechnol. 2010;2010:753675. doi: 10.1155/2010/753675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Soussi T, Hamroun D, Hjortsberg L, Rubio-Nevado JM, Fournier JL, Béroud C. MUT-TP53 2.0: a novel versatile matrix for statistical analysis of TP53 mutations in human cancer. Hum Mutat. 2010;31:1020–5. doi: 10.1002/humu.21313. [DOI] [PubMed] [Google Scholar]
  • 16.Marcel V, Dichtel-Danjoy ML, Sagne C, Hafsi H, Ma D, Ortiz-Cuaran S, Olivier M, Hall J, Mollereau B, Hainaut P, et al. Biological functions of p53 isoforms through evolution: lessons from animal and cellular models. Cell Death Differ. 2011;18:1815–24. doi: 10.1038/cdd.2011.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dent P. Non-canonical p53 signaling to promote invasion. Cancer Biol Ther. 2013;14:879–80. doi: 10.4161/cbt.26174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol. 2013;15:2–8. doi: 10.1038/ncb2641. [DOI] [PubMed] [Google Scholar]
  • 19.Ward AJ, Cooper TA. The pathobiology of splicing. J Pathol. 2010;220:152–63. doi: 10.1002/path.2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.LeWinter MM, Granzier HL. Titin is a major human disease gene. Circulation. 2013;127:938–44. doi: 10.1161/CIRCULATIONAHA.112.139717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Frenkel-Morgenstern M, Gorohovski A, Lacroix V, Rogers M, Ibanez K, Boullosa C, Andres Leon E, Ben-Hur A, Valencia A. ChiTaRS: a database of human, mouse and fruit fly chimeric transcripts and RNA-sequencing data. Nucleic Acids Res. 2013;41:D142–51. doi: 10.1093/nar/gks1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hirano M, Noda T. Genomic organization of the mouse Msh4 gene producing bicistronic, chimeric and antisense mRNA. Gene. 2004;342:165–77. doi: 10.1016/j.gene.2004.08.016. [DOI] [PubMed] [Google Scholar]
  • 23.Yang W, Wu JM, Bi AD, Ou-Yang YC, Shen HH, Chirn GW, Zhou JH, Weiss E, Holman EP, Liao DJ. Possible formation of mitochondrial-RNA containing chimeric or trimeric RNA implies a post-transcriptional and post-splicing mechanism for RNA fusion. PLoS One. 2013;8:e77016. doi: 10.1371/journal.pone.0077016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, Nishida H, Yap CC, Suzuki M, Kawai J, et al. RIKEN Genome Exploration Research Group. Genome Science Group (Genome Network Project Core Group) FANTOM Consortium Antisense transcription in the mammalian transcriptome. Science. 2005;309:1564–6. doi: 10.1126/science.1112009. [DOI] [PubMed] [Google Scholar]
  • 25.Li K, Ramchandran R. Natural antisense transcript: a concomitant engagement with protein-coding transcript. Oncotarget. 2010;1:447–52. doi: 10.18632/oncotarget.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Werner A, Sayer JA. Naturally occurring antisense RNA: function and mechanisms of action. Curr Opin Nephrol Hypertens. 2009;18:343–9. doi: 10.1097/MNH.0b013e32832cb982. [DOI] [PubMed] [Google Scholar]
  • 27.Subramanian S. Quantifying harmful mutations in human populations. Eur J Hum Genet. 2012;20:1320–2. doi: 10.1038/ejhg.2012.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Subramanian S. Significance of population size on the fixation of nonsynonymous mutations in genes under varying levels of selection pressure. Genetics. 2013;193:995–1002. doi: 10.1534/genetics.112.147900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Subramanian S. Fixation of deleterious mutations at critical positions in human proteins. Mol Biol Evol. 2011;28:2687–93. doi: 10.1093/molbev/msr097. [DOI] [PubMed] [Google Scholar]
  • 30.Hirschhorn R. In vivo reversion to normal of inherited mutations in humans. J Med Genet. 2003;40:721–8. doi: 10.1136/jmg.40.10.721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lai-Cheong JE, McGrath JA, Uitto J. Revertant mosaicism in skin: natural gene therapy. Trends Mol Med. 2011;17:140–8. doi: 10.1016/j.molmed.2010.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pasmooij AM, Jonkman MF, Uitto J. Revertant mosaicism in heritable skin diseases: mechanisms of natural gene therapy. Discov Med. 2012;14:167–79. [PubMed] [Google Scholar]
  • 33.van Dyk E, Pretorius PJ, van DE Point mutation instability (PIN) mutator phenotype as model for true back mutations seen in hereditary tyrosinemia type 1 - a hypothesis. J Inherit Metab Dis. 2012;35:407–11. doi: 10.1007/s10545-011-9401-x. [DOI] [PubMed] [Google Scholar]
  • 34.Bouwman P, Jonkers J. Molecular pathways: how can BRCA-mutated tumors become resistant to PARP inhibitors? Clin Cancer Res. 2014;20:540–7. doi: 10.1158/1078-0432.CCR-13-0225. [DOI] [PubMed] [Google Scholar]
  • 35.Dhillon KK, Swisher EM, Taniguchi T. Secondary mutations of BRCA1/2 and drug resistance. Cancer Sci. 2011;102:663–9. doi: 10.1111/j.1349-7006.2010.01840.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ashworth A. Drug resistance caused by reversion mutation. Cancer Res. 2008;68:10021–3. doi: 10.1158/0008-5472.CAN-08-2287. [DOI] [PubMed] [Google Scholar]
  • 37.Wang C, Tai Y, Lisanti MP, Liao DJ. c-Myc induction of programmed cell death may contribute to carcinogenesis: a perspective inspired by several concepts of chemical carcinogenesis. Cancer Biol Ther. 2011;11:615–26. doi: 10.4161/cbt.11.7.14688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liao DJ. The scavenger cell hypothesis of apoptosis: apoptosis redefined as a process by which a cell in living tissue is destroyed by phagocytosis. Med Hypotheses. 2005;65:23–8. doi: 10.1016/j.mehy.2005.01.028. [DOI] [PubMed] [Google Scholar]
  • 39.Liu B, Xu N, Man Y, Shen H, Avital I, Stojadinovic A, Liao DJ. Apoptosis in Living Animals Is Assisted by Scavenger Cells and Thus May Not Mainly Go through the Cytochrome C-Caspase Pathway. J Cancer. 2013;4:716–23. doi: 10.7150/jca.7577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Haddow A. Cellular inhibition and the origin of cancer. Acta Unio Int Contra Cancrum. 1938;3:342–53. [Google Scholar]
  • 41.Laconi E, Pani P, Farber E. The resistance phenotype in the development and treatment of cancer. Lancet Oncol. 2000;1:235–41. doi: 10.1016/S1470-2045(00)00154-6. [DOI] [PubMed] [Google Scholar]
  • 42.Farber E. Hepatocyte proliferation in stepwise development of experimental liver cell cancer. Dig Dis Sci. 1991;36:973–8. doi: 10.1007/BF01297150. [DOI] [PubMed] [Google Scholar]
  • 43.Farber E, Rubin H. Cellular adaptation in the origin and development of cancer. Cancer Res. 1991;51:2751–61. [PubMed] [Google Scholar]
  • 44.Farber E. Cell proliferation as a major risk factor for cancer: a concept of doubtful validity. Cancer Res. 1995;55:3759–62. [PubMed] [Google Scholar]
  • 45.Farber E. Cell proliferation is not a major risk factor for cancer. Mod Pathol. 1996;9:606. [PubMed] [Google Scholar]
  • 46.Farber E. The step-by-step development of epithelial cancer: from phenotype to genotype. Adv Cancer Res. 1996;70:21–48. doi: 10.1016/S0065-230X(08)60870-2. [DOI] [PubMed] [Google Scholar]
  • 47.Farber E. Risk assessment for possible carcinogens: a critical look. Drug Metab Rev. 2000;32:143–51. doi: 10.1081/DMR-100100568. [DOI] [PubMed] [Google Scholar]
  • 48.Eriksson LC, Andersson GN. Membrane biochemistry and chemical hepatocarcinogenesis. Crit Rev Biochem Mol Biol. 1992;27:1–55. doi: 10.3109/10409239209082558. [DOI] [PubMed] [Google Scholar]
  • 49.Eriksson LC, Blanck A, Bock KW, Mannervik B. Metabolism of xenobiotics in hepatocyte nodules. Toxicol Pathol. 1987;15:27–42. doi: 10.1177/019262338701500104. [DOI] [PubMed] [Google Scholar]
  • 50.Björkhem-Bergman L, Ekström L, Eriksson LC. Review: Exploring anticarcinogenic agents in a rat hepatocarcinogenesis model--focus on selenium and statins. In Vivo. 2012;26:527–35. [PubMed] [Google Scholar]
  • 51.Liao DJ, Dickson RB. c-Myc in breast cancer. Endocr Relat Cancer. 2000;7:143–64. doi: 10.1677/erc.0.0070143. [DOI] [PubMed] [Google Scholar]
  • 52.Liao JD, Adsay NV, Khannani F, Grignon D, Thakur A, Sarkar FH. Histological complexities of pancreatic lesions from transgenic mouse models are consistent with biological and morphological heterogeneity of human pancreatic cancer. Histol Histopathol. 2007;22:661–76. doi: 10.14670/hh-22.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Choi YJ, Anders L. Signaling through cyclin D-dependent kinases. Oncogene. 2014;33:1890–903. doi: 10.1038/onc.2013.137. [DOI] [PubMed] [Google Scholar]
  • 54.Wang C, Lisanti MP, Liao DJ. Reviewing once more the c-myc and Ras collaboration: converging at the cyclin D1-CDK4 complex and challenging basic concepts of cancer biology. Cell Cycle. 2011;10:57–67. doi: 10.4161/cc.10.1.14449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Liao DJ, Thakur A, Wu J, Biliran H, Sarkar FH. Perspectives on c-Myc, Cyclin D1, and their interaction in cancer formation, progression, and response to chemotherapy. Crit Rev Oncog. 2007;13:93–158. doi: 10.1615/CritRevOncog.v13.i2.10. [DOI] [PubMed] [Google Scholar]
  • 56.Atadja P, Wong H, Veillete C, Riabowol K. Overexpression of cyclin D1 blocks proliferation of normal diploid fibroblasts. Exp Cell Res. 1995;217:205–16. doi: 10.1006/excr.1995.1080. [DOI] [PubMed] [Google Scholar]
  • 57.Han EK, Sgambato A, Jiang W, Zhang YJ, Santella RM, Doki Y, Cacace AM, Schieren I, Weinstein IB. Stable overexpression of cyclin D1 in a human mammary epithelial cell line prolongs the S-phase and inhibits growth. Oncogene. 1995;10:953–61. [PubMed] [Google Scholar]
  • 58.Han EK, Begemann M, Sgambato A, Soh JW, Doki Y, Xing WQ, Liu W, Weinstein IB. Increased expression of cyclin D1 in a murine mammary epithelial cell line induces p27kip1, inhibits growth, and enhances apoptosis. Cell Growth Differ. 1996;7:699–710. [PubMed] [Google Scholar]
  • 59.Han EK, Ng SC, Arber N, Begemann M, Weinstein IB. Roles of cyclin D1 and related genes in growth inhibition, senescence and apoptosis. Apoptosis. 1999;4:213–9. doi: 10.1023/A:1009618824145. [DOI] [PubMed] [Google Scholar]
  • 60.Freeman RS, Estus S, Johnson EM., Jr. Analysis of cell cycle-related gene expression in postmitotic neurons: selective induction of Cyclin D1 during programmed cell death. Neuron. 1994;12:343–55. doi: 10.1016/0896-6273(94)90276-3. [DOI] [PubMed] [Google Scholar]
  • 61.Pagano M, Theodoras AM, Tam SW, Draetta GF. Cyclin D1-mediated inhibition of repair and replicative DNA synthesis in human fibroblasts. Genes Dev. 1994;8:1627–39. doi: 10.1101/gad.8.14.1627. [DOI] [PubMed] [Google Scholar]
  • 62.Russell A, Thompson MA, Hendley J, Trute L, Armes J, Germain D. Cyclin D1 and D3 associate with the SCF complex and are coordinately elevated in breast cancer. Oncogene. 1999;18:1983–91. doi: 10.1038/sj.onc.1202511. [DOI] [PubMed] [Google Scholar]
  • 63.Peterson SR, Gadbois DM, Bradbury EM, Kraemer PM. Immortalization of human fibroblasts by SV40 large T antigen results in the reduction of cyclin D1 expression and subunit association with proliferating cell nuclear antigen and Waf1. Cancer Res. 1995;55:4651–7. [PubMed] [Google Scholar]
  • 64.Lukas J, Pagano M, Staskova Z, Draetta G, Bartek J. Cyclin D1 protein oscillates and is essential for cell cycle progression in human tumour cell lines. Oncogene. 1994;9:707–18. [PubMed] [Google Scholar]
  • 65.Buchou T, Kranenburg O, van Dam H, Roelen D, Zantema A, Hall FL, van der Eb A. Increased cyclin A and decreased cyclin D levels in adenovirus 5 E1A-transformed rodent cell lines. Oncogene. 1993;8:1765–73. [PubMed] [Google Scholar]
  • 66.Bartek J, Lukas J. DNA repair: Cyclin D1 multitasks. Nature. 2011;474:171–2. doi: 10.1038/474171a. [DOI] [PubMed] [Google Scholar]
  • 67.Lukas J, Müller H, Bartkova J, Spitkovsky D, Kjerulff AA, Jansen-Dürr P, Strauss M, Bartek J. DNA tumor virus oncoproteins and retinoblastoma gene mutations share the ability to relieve the cell’s requirement for cyclin D1 function in G1. J Cell Biol. 1994;125:625–38. doi: 10.1083/jcb.125.3.625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jiang W, Zhang YJ, Kahn SM, Hollstein MC, Santella RM, Lu SH, Harris CC, Montesano R, Weinstein IB. Altered expression of the cyclin D1 and retinoblastoma genes in human esophageal cancer. Proc Natl Acad Sci U S A. 1993;90:9026–30. doi: 10.1073/pnas.90.19.9026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Spitkovsky D, Steiner P, Lukas J, Lees E, Pagano M, Schulze A, Joswig S, Picard D, Tommasino M, Eilers M, et al. Modulation of cyclin gene expression by adenovirus E1A in a cell line with E1A-dependent conditional proliferation. J Virol. 1994;68:2206–14. doi: 10.1128/jvi.68.4.2206-2214.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Liao DJ, Natarajan G, Deming SL, Jamerson MH, Johnson M, Chepko G, Dickson RB. Cell cycle basis for the onset and progression of c-Myc-induced, TGFalpha-enhanced mouse mammary gland carcinogenesis. Oncogene. 2000;19:1307–17. doi: 10.1038/sj.onc.1203430. [DOI] [PubMed] [Google Scholar]
  • 71.Land H, Parada LF, Weinberg RA. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature. 1983;304:596–602. doi: 10.1038/304596a0. [DOI] [PubMed] [Google Scholar]
  • 72.Stacey DW. Cyclin D1 serves as a cell cycle regulatory switch in actively proliferating cells. Curr Opin Cell Biol. 2003;15:158–63. doi: 10.1016/S0955-0674(03)00008-5. [DOI] [PubMed] [Google Scholar]
  • 73.Stacey DW. Three Observations That Have Changed Our Understanding of Cyclin D1 and p27 in Cell Cycle Control. Genes Cancer. 2010;1:1189–99. doi: 10.1177/1947601911403475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Nielsen NH, Lodén M, Cajander J, Emdin SO, Landberg G. G1-S transition defects occur in most breast cancers and predict outcome. Breast Cancer Res Treat. 1999;56:105–12. doi: 10.1023/A:1006208419350. [DOI] [PubMed] [Google Scholar]
  • 75.Jares P, Rey MJ, Fernández PL, Campo E, Nadal A, Muñoz M, Mallofré C, Muntané J, Nayach I, Estapé J, et al. Cyclin D1 and retinoblastoma gene expression in human breast carcinoma: correlation with tumour proliferation and oestrogen receptor status. J Pathol. 1997;182:160–6. doi: 10.1002/(SICI)1096-9896(199706)182:2<160::AID-PATH814>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 76.Oyama T, Kashiwabara K, Yoshimoto K, Arnold A, Koerner F. Frequent overexpression of the cyclin D1 oncogene in invasive lobular carcinoma of the breast. Cancer Res. 1998;58:2876–80. [PubMed] [Google Scholar]
  • 77.Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–52. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  • 78.McLaughlin-Drubin ME, Park D, Munger K. Tumor suppressor p16INK4A is necessary for survival of cervical carcinoma cell lines. Proc Natl Acad Sci U S A. 2013;110:16175–80. doi: 10.1073/pnas.1310432110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Tay SP, Yeo CW, Chai C, Chua PJ, Tan HM, Ang AX, Yip DL, Sung JX, Tan PH, Bay BH, et al. Parkin enhances the expression of cyclin-dependent kinase 6 and negatively regulates the proliferation of breast cancer cells. J Biol Chem. 2010;285:29231–8. doi: 10.1074/jbc.M110.108241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ito K, Maruyama Z, Sakai A, Izumi S, Moriishi T, Yoshida CA, Miyazaki T, Komori H, Takada K, Kawaguchi H, et al. Overexpression of Cdk6 and Ccnd1 in chondrocytes inhibited chondrocyte maturation and caused p53-dependent apoptosis without enhancing proliferation. Oncogene. 2014;33:1862–71. doi: 10.1038/onc.2013.130. [DOI] [PubMed] [Google Scholar]
  • 81.Nagasawa M, Gelfand EW, Lucas JJ. Accumulation of high levels of the p53 and p130 growth-suppressing proteins in cell lines stably over-expressing cyclin-dependent kinase 6 (cdk6) Oncogene. 2001;20:2889–99. doi: 10.1038/sj.onc.1204396. [DOI] [PubMed] [Google Scholar]
  • 82.Lucas JJ, Domenico J, Gelfand EW. Cyclin-dependent kinase 6 inhibits proliferation of human mammary epithelial cells. Mol Cancer Res. 2004;2:105–14. [PubMed] [Google Scholar]
  • 83.Wang X, Sistrunk C, Rodriguez-Puebla ML. Unexpected reduction of skin tumorigenesis on expression of cyclin-dependent kinase 6 in mouse epidermis. Am J Pathol. 2011;178:345–54. doi: 10.1016/j.ajpath.2010.11.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Rojas P, Cadenas MB, Lin PC, Benavides F, Conti CJ, Rodriguez-Puebla ML. Cyclin D2 and cyclin D3 play opposite roles in mouse skin carcinogenesis. Oncogene. 2007;26:1723–30. doi: 10.1038/sj.onc.1209970. [DOI] [PubMed] [Google Scholar]
  • 85.Gurung R, Parnaik VK. Cyclin D3 promotes myogenic differentiation and Pax7 transcription. J Cell Biochem. 2012;113:209–19. doi: 10.1002/jcb.23346. [DOI] [PubMed] [Google Scholar]
  • 86.Sankaran VG, Ludwig LS, Sicinska E, Xu J, Bauer DE, Eng JC, Patterson HC, Metcalf RA, Natkunam Y, Orkin SH, et al. Cyclin D3 coordinates the cell cycle during differentiation to regulate erythrocyte size and number. Genes Dev. 2012;26:2075–87. doi: 10.1101/gad.197020.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bartkova J, Lukas J, Strauss M, Bartek J. Cyclin D3: requirement for G1/S transition and high abundance in quiescent tissues suggest a dual role in proliferation and differentiation. Oncogene. 1998;17:1027–37. doi: 10.1038/sj.onc.1202016. [DOI] [PubMed] [Google Scholar]
  • 88.Wafa K, MacLean J, Zhang F, Pasumarthi KB. Characterization of growth suppressive functions of a splice variant of cyclin D2. PLoS One. 2013;8:e53503. doi: 10.1371/journal.pone.0053503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ko E, Kim Y, Park SE, Cho EY, Han J, Shim YM, Park J, Kim DH. Reduced expression of cyclin D2 is associated with poor recurrence-free survival independent of cyclin D1 in stage III non-small cell lung cancer. Lung Cancer. 2012;77:401–6. doi: 10.1016/j.lungcan.2012.03.027. [DOI] [PubMed] [Google Scholar]
  • 90.Kobayashi T, Nakamura E, Shimizu Y, Terada N, Maeno A, Kobori G, Kamba T, Kamoto T, Ogawa O, Inoue T. Restoration of cyclin D2 has an inhibitory potential on the proliferation of LNCaP cells. Biochem Biophys Res Commun. 2009;387:196–201. doi: 10.1016/j.bbrc.2009.06.146. [DOI] [PubMed] [Google Scholar]
  • 91.Padar A, Sathyanarayana UG, Suzuki M, Maruyama R, Hsieh JT, Frenkel EP, Minna JD, Gazdar AF. Inactivation of cyclin D2 gene in prostate cancers by aberrant promoter methylation. Clin Cancer Res. 2003;9:4730–4. [PubMed] [Google Scholar]
  • 92.Tsutsui M, Iizuka N, Moribe T, Miura T, Kimura N, Tamatsukuri S, Ishitsuka H, Fujita Y, Hamamoto Y, Tsunedomi R, et al. Methylated cyclin D2 gene circulating in the blood as a prognosis predictor of hepatocellular carcinoma. Clin Chim Acta. 2010;411:516–20. doi: 10.1016/j.cca.2010.01.004. [DOI] [PubMed] [Google Scholar]
  • 93.Witt D, Burfeind P, von Hardenberg S, Opitz L, Salinas-Riester G, Bremmer F, Schweyer S, Thelen P, Neesen J, Kaulfuss S. Valproic acid inhibits the proliferation of cancer cells by re-expressing cyclin D2. Carcinogenesis. 2013;34:1115–24. doi: 10.1093/carcin/bgt019. [DOI] [PubMed] [Google Scholar]
  • 94.Kobayashi T, Nakamura E, Shimizu Y, Terada N, Maeno A, Kobori G, Kamba T, Kamoto T, Ogawa O, Inoue T. Restoration of cyclin D2 has an inhibitory potential on the proliferation of LNCaP cells. Biochem Biophys Res Commun. 2009;387:196–201. doi: 10.1016/j.bbrc.2009.06.146. [DOI] [PubMed] [Google Scholar]
  • 95.Eshkoor SA, Ismail P, Rahman SA, Oshkour SA. p16 gene expression in basal cell carcinoma. Arch Med Res. 2008;39:668–73. doi: 10.1016/j.arcmed.2008.06.003. [DOI] [PubMed] [Google Scholar]
  • 96.Milde-Langosch K, Riethdorf S. Role of cell-cycle regulatory proteins in gynecological cancer. J Cell Physiol. 2003;196:224–44. doi: 10.1002/jcp.10286. [DOI] [PubMed] [Google Scholar]
  • 97.Milde-Langosch K, Hagen M, Bamberger AM, Löning T. Expression and prognostic value of the cell-cycle regulatory proteins, Rb, p16MTS1, p21WAF1, p27KIP1, cyclin E, and cyclin D2, in ovarian cancer. Int J Gynecol Pathol. 2003;22:168–74. doi: 10.1097/00004347-200304000-00009. [DOI] [PubMed] [Google Scholar]
  • 98.Herschkowitz JI, He X, Fan C, Perou CM. The functional loss of the retinoblastoma tumour suppressor is a common event in basal-like and luminal B breast carcinomas. Breast Cancer Res. 2008;10:R75. doi: 10.1186/bcr2142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kommoss S, du Bois A, Ridder R, Trunk MJ, Schmidt D, Pfisterer J, Kommoss F, AGO-OVAR Independent prognostic significance of cell cycle regulator proteins p16(INK4a) and pRb in advanced-stage ovarian carcinoma including optimally debulked patients: a translational research subprotocol of a randomised study of the Arbeitsgemeinschaft Gynaekologische Onkologie Ovarian Cancer Study Group. Br J Cancer. 2007;96:306–13. doi: 10.1038/sj.bjc.6603531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Andujar P, Wang J, Descatha A, Galateau-Sallé F, Abd-Alsamad I, Billon-Galland MA, Blons H, Clin B, Danel C, Housset B, et al. p16INK4A inactivation mechanisms in non-small-cell lung cancer patients occupationally exposed to asbestos. Lung Cancer. 2010;67:23–30. doi: 10.1016/j.lungcan.2009.03.018. [DOI] [PubMed] [Google Scholar]
  • 101.Chen YW, Chu HC, Ze-Shiang Lin, Shiah WJ, Chou CP, Klimstra DS, Lewis BC. p16 Stimulates CDC42-dependent migration of hepatocellular carcinoma cells. PLoS One. 2013;8:e69389. doi: 10.1371/journal.pone.0069389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Manning AL, Dyson NJRB. RB: mitotic implications of a tumour suppressor. Nat Rev Cancer. 2012;12:220–6. doi: 10.1038/nrc3216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ciavarra G, Zacksenhaus E. Multiple pathways counteract cell death induced by RB1 loss: implications for cancer. Cell Cycle. 2011;10:1533–9. doi: 10.4161/cc.10.10.15520. [DOI] [PubMed] [Google Scholar]
  • 104.Viatour P, Sage J. Newly identified aspects of tumor suppression by RB. Dis Model Mech. 2011;4:581–5. doi: 10.1242/dmm.008060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Morgenbesser SD, Williams BO, Jacks T, DePinho RA. p53-dependent apoptosis produced by Rb-deficiency in the developing mouse lens. Nature. 1994;371:72–4. doi: 10.1038/371072a0. [DOI] [PubMed] [Google Scholar]
  • 106.Macleod KF, Hu Y, Jacks T. Loss of Rb activates both p53-dependent and independent cell death pathways in the developing mouse nervous system. EMBO J. 1996;15:6178–88. [PMC free article] [PubMed] [Google Scholar]
  • 107.Jiang Z, Zacksenhaus E. Activation of retinoblastoma protein in mammary gland leads to ductal growth suppression, precocious differentiation, and adenocarcinoma. J Cell Biol. 2002;156:185–98. doi: 10.1083/jcb.200106084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bosco EE, Knudsen ES. RB in breast cancer: at the crossroads of tumorigenesis and treatment. Cell Cycle. 2007;6:667–71. doi: 10.4161/cc.6.6.3988. [DOI] [PubMed] [Google Scholar]
  • 109.Murakami Y, Katahira M, Makino R, Hayashi K, Hirohashi S, Sekiya T. Inactivation of the retinoblastoma gene in a human lung carcinoma cell line detected by single-strand conformation polymorphism analysis of the polymerase chain reaction product of cDNA. Oncogene. 1991;6:37–42. [PubMed] [Google Scholar]
  • 110.Latil A, Pesche S, Valeri A, Cussenot O, Fournier G, Lidereau R. Common delta2 and delta8 RB1 variants in human breast and prostate tissues. Int J Cancer. 1999;80:632–3. doi: 10.1002/(SICI)1097-0215(19990209)80:4<632::AID-IJC25>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 111.Liu HP, Thompson AM, Macleod KF. A novel form of pRb expressed during normal myelopoiesis and in tumour-associated macrophages. Cell Prolif. 2005;38:13–24. doi: 10.1111/j.1365-2184.2005.00326.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lemaire C, Godefroy N, Costina-Parvu I, Rincheval V, Renaud F, Trotot P, Bouleau S, Mignotte B, Vayssière JL. Caspase-9 can antagonize p53-induced apoptosis by generating a p76(Rb) truncated form of Rb. Oncogene. 2005;24:3297–308. doi: 10.1038/sj.onc.1208493. [DOI] [PubMed] [Google Scholar]
  • 113.Le Floch N, Rincheval V, Ferecatu I, Ali-Boina R, Renaud F, Mignotte B, Vayssière JL. The p76(Rb) and p100(Rb) truncated forms of the Rb protein exert antagonistic roles on cell death regulation in human cell lines. Biochem Biophys Res Commun. 2010;399:173–8. doi: 10.1016/j.bbrc.2010.07.041. [DOI] [PubMed] [Google Scholar]
  • 114.Roig JM, Molina MA, Cascante A, Calbó J, Carbó N, Wirtz U, Sreedharan S, Fillat C, Mazo A. Adenovirus-mediated retinoblastoma 94 gene transfer induces human pancreatic tumor regression in a mouse xenograft model. Clin Cancer Res. 2004;10:1454–62. doi: 10.1158/1078-0432.CCR-0442-03. [DOI] [PubMed] [Google Scholar]
  • 115.Xu HJ, Hu SX, Hashimoto T, Takahashi R, Benedict WF. The retinoblastoma susceptibility gene product: a characteristic pattern in normal cells and abnormal expression in malignant cells. Oncogene. 1989;4:807–12. [PubMed] [Google Scholar]
  • 116.Xu HJ, Xu K, Zhou Y, Li J, Benedict WF, Hu SX. Enhanced tumor cell growth suppression by an N-terminal truncated retinoblastoma protein. Proc Natl Acad Sci U S A. 1994;91:9837–41. doi: 10.1073/pnas.91.21.9837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sánchez-Sánchez F, Ramírez-Castillejo C, Weekes DB, Beneyto M, Prieto F, Nájera C, Mittnacht S. Attenuation of disease phenotype through alternative translation initiation in low-penetrance retinoblastoma. Hum Mutat. 2007;28:159–67. doi: 10.1002/humu.20394. [DOI] [PubMed] [Google Scholar]
  • 118.Valverde JR, Alonso J, Palacios I, Pestaña A. RB1 gene mutation up-date, a meta-analysis based on 932 reported mutations available in a searchable database. BMC Genet. 2005;6:53–61. doi: 10.1186/1471-2156-6-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Onadim Z, Hogg A, Baird PN, Cowell JK. Oncogenic point mutations in exon 20 of the RB1 gene in families showing incomplete penetrance and mild expression of the retinoblastoma phenotype. Proc Natl Acad Sci U S A. 1992;89:6177–81. doi: 10.1073/pnas.89.13.6177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wallace M, Ball KL. Docking-dependent regulation of the Rb tumor suppressor protein by Cdk4. Mol Cell Biol. 2004;24:5606–19. doi: 10.1128/MCB.24.12.5606-5619.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yang H, Williams BO, Hinds PW, Shih TS, Jacks T, Bronson RT, Livingston DM. Tumor suppression by a severely truncated species of retinoblastoma protein. Mol Cell Biol. 2002;22:3103–10. doi: 10.1128/MCB.22.9.3103-3110.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Worku D, Jouhra F, Jiang GW, Patani N, Newbold RF, Mokbel K. Evidence of a tumour suppressive function of E2F1 gene in human breast cancer. Anticancer Res. 2008;28(4B):2135–9. [PubMed] [Google Scholar]
  • 123.Johnson DG, Degregori J. Putting the Oncogenic and Tumor Suppressive Activities of E2F into Context. Curr Mol Med. 2006;6:731–8. doi: 10.2174/1566524010606070731. [DOI] [PubMed] [Google Scholar]
  • 124.Bell LA, Ryan KM. Life and death decisions by E2F-1. Cell Death Differ. 2004;11:137–42. doi: 10.1038/sj.cdd.4401324. [DOI] [PubMed] [Google Scholar]
  • 125.Sun B, Wingate H, Swisher SG, Keyomarsi K, Hunt KK. Absence of pRb facilitates E2F1-induced apoptosis in breast cancer cells. Cell Cycle. 2010;9:1122–30. doi: 10.4161/cc.9.6.10990. [DOI] [PubMed] [Google Scholar]
  • 126.Yamasaki L, Jacks T, Bronson R, Goillot E, Harlow E, Dyson NJ. Tumor induction and tissue atrophy in mice lacking E2F-1. Cell. 1996;85:537–48. doi: 10.1016/S0092-8674(00)81254-4. [DOI] [PubMed] [Google Scholar]
  • 127.Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 1998;12:3499–511. doi: 10.1101/gad.12.22.3499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Diehl JA, Zindy F, Sherr CJ. Inhibition of cyclin D1 phosphorylation on threonine-286 prevents its rapid degradation via the ubiquitin-proteasome pathway. Genes Dev. 1997;11:957–72. doi: 10.1101/gad.11.8.957. [DOI] [PubMed] [Google Scholar]
  • 129.Germain D, Russell A, Thompson A, Hendley J. Ubiquitination of free cyclin D1 is independent of phosphorylation on threonine 286. J Biol Chem. 2000;275:12074–9. doi: 10.1074/jbc.275.16.12074. [DOI] [PubMed] [Google Scholar]
  • 130.Bates S, Parry D, Bonetta L, Vousden K, Dickson C, Peters G. Absence of cyclin D/cdk complexes in cells lacking functional retinoblastoma protein. Oncogene. 1994;9:1633–40. [PubMed] [Google Scholar]
  • 131.Agami R, Bernards R. Convergence of mitogenic and DNA damage signaling in the G1 phase of the cell cycle. Cancer Lett. 2002;177:111–8. doi: 10.1016/S0304-3835(01)00785-6. [DOI] [PubMed] [Google Scholar]
  • 132.Agami R, Bernards R. Distinct initiation and maintenance mechanisms cooperate to induce G1 cell cycle arrest in response to DNA damage. Cell. 2000;102:55–66. doi: 10.1016/S0092-8674(00)00010-6. [DOI] [PubMed] [Google Scholar]
  • 133.Shimura T, Fukumoto M, Kunugita N. The role of cyclin D1 in response to long-term exposure to ionizing radiation. Cell Cycle. 2013;12:2738–43. doi: 10.4161/cc.25746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Shimura T. Acquired radioresistance of cancer and the AKT/GSK3β/cyclin D1 overexpression cycle. J Radiat Res. 2011;52:539–44. doi: 10.1269/jrr.11098. [DOI] [PubMed] [Google Scholar]
  • 135.Finkielstein CV, Chen LG, Maller JL. A role for G1/S cyclin-dependent protein kinases in the apoptotic response to ionizing radiation. J Biol Chem. 2002;277:38476–85. doi: 10.1074/jbc.M206184200. [DOI] [PubMed] [Google Scholar]
  • 136.Albanese C, D’Amico M, Reutens AT, Fu M, Watanabe G, Lee RJ, Kitsis RN, Henglein B, Avantaggiati M, Somasundaram K, et al. Activation of the cyclin D1 gene by the E1A-associated protein p300 through AP-1 inhibits cellular apoptosis. J Biol Chem. 1999;274:34186–95. doi: 10.1074/jbc.274.48.34186. [DOI] [PubMed] [Google Scholar]
  • 137.Coco Martin JM, Balkenende A, Verschoor T, Lallemand F, Michalides R. Cyclin D1 overexpression enhances radiation-induced apoptosis and radiosensitivity in a breast tumor cell line. Cancer Res. 1999;59:1134–40. [PubMed] [Google Scholar]
  • 138.Zhou Q, Fukushima P, DeGraff W, Mitchell JB, Stetler Stevenson M, Ashkenazi A, Steeg PS. Radiation and the Apo2L/TRAIL apoptotic pathway preferentially inhibit the colonization of premalignant human breast cells overexpressing cyclin D1. Cancer Res. 2000;60:2611–5. [PubMed] [Google Scholar]
  • 139.Alexandrou AT, Li JJ. Cell cycle regulators guide mitochondrial activity in radiation-induced adaptive response. Antioxid Redox Signal. 2014;20:1463–80. doi: 10.1089/ars.2013.5684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Jayasurya R, Francis G, Kannan S, Lekshminarayanan K, Nalinakumari KR, Abraham T, Abraham EK, Nair MK. p53, p16 and cyclin D1: molecular determinants of radiotherapy treatment response in oral carcinoma. Int J Cancer. 2004;109:710–6. doi: 10.1002/ijc.20042. [DOI] [PubMed] [Google Scholar]
  • 141.Higuchi E, Oridate N, Homma A, Suzuki F, Atago Y, Nagahashi T, Furuta Y, Fukuda S. Prognostic significance of cyclin D1 and p16 in patients with intermediate-risk head and neck squamous cell carcinoma treated with docetaxel and concurrent radiotherapy. Head Neck. 2007;29:940–7. doi: 10.1002/hed.20632. [DOI] [PubMed] [Google Scholar]
  • 142.Bartek J, Lukas J. DNA repair: Cyclin D1 multitasks. Nature. 2011;474:171–2. doi: 10.1038/474171a. [DOI] [PubMed] [Google Scholar]
  • 143.Jirawatnotai S, Hu Y, Michowski W, Elias JE, Becks L, Bienvenu F, Zagozdzon A, Goswami T, Wang YE, Clark AB, et al. A function for cyclin D1 in DNA repair uncovered by protein interactome analyses in human cancers. Nature. 2011;474:230–4. doi: 10.1038/nature10155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Jirawatnotai S, Hu Y, Livingston DM, Sicinski P. Proteomic identification of a direct role for cyclin d1 in DNA damage repair. Cancer Res. 2012;72:4289–93. doi: 10.1158/0008-5472.CAN-11-3549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pestell RG. New roles of cyclin D1. Am J Pathol. 2013;183:3–9. doi: 10.1016/j.ajpath.2013.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Shimura T, Kakuda S, Ochiai Y, Nakagawa H, Kuwahara Y, Takai Y, Kobayashi J, Komatsu K, Fukumoto M. Acquired radioresistance of human tumor cells by DNA-PK/AKT/GSK3beta-mediated cyclin D1 overexpression. Oncogene. 2010;29:4826–37. doi: 10.1038/onc.2010.238. [DOI] [PubMed] [Google Scholar]
  • 147.Shimura T, Ochiai Y, Noma N, Oikawa T, Sano Y, Fukumoto M. Cyclin D1 overexpression perturbs DNA replication and induces replication-associated DNA double-strand breaks in acquired radioresistant cells. Cell Cycle. 2013;12:773–82. doi: 10.4161/cc.23719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Aggarwal P, Lessie MD, Lin DI, Pontano L, Gladden AB, Nuskey B, Goradia A, Wasik MA, Klein-Szanto AJ, Rustgi AK, et al. Nuclear accumulation of cyclin D1 during S phase inhibits Cul4-dependent Cdt1 proteolysis and triggers p53-dependent DNA rereplication. Genes Dev. 2007;21:2908–22. doi: 10.1101/gad.1586007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Inoue K, Sherr CJ. Gene expression and cell cycle arrest mediated by transcription factor DMP1 is antagonized by D-type cyclins through a cyclin-dependent-kinase-independent mechanism. Mol Cell Biol. 1998;18:1590–600. doi: 10.1128/mcb.18.3.1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Bienvenu F, Jirawatnotai S, Elias JE, Meyer CA, Mizeracka K, Marson A, Frampton GM, Cole MF, Odom DT, Odajima J, et al. Transcriptional role of cyclin D1 in development revealed by a genetic-proteomic screen. Nature. 2010;463:374–8. doi: 10.1038/nature08684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Opitz OG, Rustgi AK. Interaction between Sp1 and cell cycle regulatory proteins is important in transactivation of a differentiation-related gene. Cancer Res. 2000;60:2825–30. [PubMed] [Google Scholar]
  • 152.Tapias A, Ciudad CJ, Roninson IB, Noé V. Regulation of Sp1 by cell cycle related proteins. Cell Cycle. 2008;7:2856–67. doi: 10.4161/cc.7.18.6671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zwijsen RM, Wientjens E, Klompmaker R, van der Sman J, Bernards R, Michalides RJ. CDK-independent activation of estrogen receptor by cyclin D1. Cell. 1997;88:405–15. doi: 10.1016/S0092-8674(00)81879-6. [DOI] [PubMed] [Google Scholar]
  • 154.Knudsen KE, Cavenee WK, Arden KC. D-type cyclins complex with the androgen receptor and inhibit its transcriptional transactivation ability. Cancer Res. 1999;59:2297–301. [PubMed] [Google Scholar]
  • 155.Reutens AT, Fu M, Wang C, Albanese C, McPhaul MJ, Sun Z, Balk SP, Jänne OA, Palvimo JJ, Pestell RG. Cyclin D1 binds the androgen receptor and regulates hormone-dependent signaling in a p300/CBP-associated factor (P/CAF)-dependent manner. Mol Endocrinol. 2001;15:797–811. doi: 10.1210/mend.15.5.0641. [DOI] [PubMed] [Google Scholar]
  • 156.Lamb J, Ramaswamy S, Ford HL, Contreras B, Martinez RV, Kittrell FS, Zahnow CA, Patterson N, Golub TR, Ewen ME. A mechanism of cyclin D1 action encoded in the patterns of gene expression in human cancer. Cell. 2003;114:323–34. doi: 10.1016/S0092-8674(03)00570-1. [DOI] [PubMed] [Google Scholar]
  • 157.Lamb J, Ewen ME. Cyclin D1 and molecular chaperones: implications for tumorigenesis. Cell Cycle. 2003;2:525–7. doi: 10.4161/cc.2.6.584. [DOI] [PubMed] [Google Scholar]
  • 158.Jeffrey PD, Tong L, Pavletich NP. Structural basis of inhibition of CDK-cyclin complexes by INK4 inhibitors. Genes Dev. 2000;14:3115–25. doi: 10.1101/gad.851100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Russo AA, Tong L, Lee JO, Jeffrey PD, Pavletich NP. Structural basis for inhibition of the cyclin-dependent kinase Cdk6 by the tumour suppressor p16INK4a. Nature. 1998;395:237–43. doi: 10.1038/26155. [DOI] [PubMed] [Google Scholar]
  • 160.Morisaki H, Ando A, Nagata Y, Pereira-Smith O, Smith JR, Ikeda K, Nakanishi M. Complex mechanisms underlying impaired activation of Cdk4 and Cdk2 in replicative senescence: roles of p16, p21, and cyclin D1. Exp Cell Res. 1999;253:503–10. doi: 10.1006/excr.1999.4698. [DOI] [PubMed] [Google Scholar]
  • 161.Kim DM, Yang K, Yang BS. Biochemical characterizations reveal different properties between CDK4/cyclin D1 and CDK2/cyclin A. Exp Mol Med. 2003;35:421–30. doi: 10.1038/emm.2003.55. [DOI] [PubMed] [Google Scholar]
  • 162.Guo Y, Mahajan A, Yuan C, Joo SH, Weghorst CM, Tsai MD, Li J. Comparisons of the conformational stability of cyclin-dependent kinase (CDK) 4-interacting ankyrin repeat (AR) proteins. Biochemistry. 2009;48:4050–62. doi: 10.1021/bi802247p. [DOI] [PubMed] [Google Scholar]
  • 163.Mahajan A, Guo Y, Yuan C, Weghorst CM, Tsai MD, Li J. Dissection of protein-protein interaction and CDK4 inhibition in the oncogenic versus tumor suppressing functions of gankyrin and P16. J Mol Biol. 2007;373:990–1005. doi: 10.1016/j.jmb.2007.08.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Li J, Tsai MD. Novel insights into the INK4-CDK4/6-Rb pathway: counter action of gankyrin against INK4 proteins regulates the CDK4-mediated phosphorylation of Rb. Biochemistry. 2002;41:3977–83. doi: 10.1021/bi011550s. [DOI] [PubMed] [Google Scholar]
  • 165.Selent J, Kaczor AA, Guixà-González R, Carrió P, Pastor M, Obiol-Pardo C. Rational design of the survivin/CDK4 complex by combining protein-protein docking and molecular dynamics simulations. J Mol Model. 2013;19:1507–14. doi: 10.1007/s00894-012-1705-8. [DOI] [PubMed] [Google Scholar]
  • 166.Suzuki A, Ito T, Kawano H, Hayashida M, Hayasaki Y, Tsutomi Y, Akahane K, Nakano T, Miura M, Shiraki K. Survivin initiates procaspase 3/p21 complex formation as a result of interaction with Cdk4 to resist Fas-mediated cell death. Oncogene. 2000;19:1346–53. doi: 10.1038/sj.onc.1203429. [DOI] [PubMed] [Google Scholar]
  • 167.Suzuki A, Hayashida M, Ito T, Kawano H, Nakano T, Miura M, Akahane K, Shiraki K. Survivin initiates cell cycle entry by the competitive interaction with Cdk4/p16(INK4a) and Cdk2/cyclin E complex activation. Oncogene. 2000;19:3225–34. doi: 10.1038/sj.onc.1203665. [DOI] [PubMed] [Google Scholar]
  • 168.Zhao Q, Boschelli F, Caplan AJ, Arndt KT. Identification of a conserved sequence motif that promotes Cdc37 and cyclin D1 binding to Cdk4. J Biol Chem. 2004;279:12560–4. doi: 10.1074/jbc.M308242200. [DOI] [PubMed] [Google Scholar]
  • 169.Stepanova L, Leng X, Parker SB, Harper JW. Mammalian p50Cdc37 is a protein kinase-targeting subunit of Hsp90 that binds and stabilizes Cdk4. Genes Dev. 1996;10:1491–502. doi: 10.1101/gad.10.12.1491. [DOI] [PubMed] [Google Scholar]
  • 170.Wang X, Grammatikakis N, Hu J. Role of p50/CDC37 in hepadnavirus assembly and replication. J Biol Chem. 2002;277:24361–7. doi: 10.1074/jbc.M202198200. [DOI] [PubMed] [Google Scholar]
  • 171.Fraedrich K, Müller B, Grassmann R. The HTLV-1 Tax protein binding domain of cyclin-dependent kinase 4 (CDK4) includes the regulatory PSTAIRE helix. Retrovirology. 2005;2:54. doi: 10.1186/1742-4690-2-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Lim JT, Mansukhani M, Weinstein IB. Cyclin-dependent kinase 6 associates with the androgen receptor and enhances its transcriptional activity in prostate cancer cells. Proc Natl Acad Sci U S A. 2005;102:5156–61. doi: 10.1073/pnas.0501203102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Ray A, James MK, Larochelle S, Fisher RP, Blain SW. p27Kip1 inhibits cyclin D-cyclin-dependent kinase 4 by two independent modes. Mol Cell Biol. 2009;29:986–99. doi: 10.1128/MCB.00898-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Paternot S, Bockstaele L, Bisteau X, Kooken H, Coulonval K, Roger PP. Rb inactivation in cell cycle and cancer: the puzzle of highly regulated activating phosphorylation of CDK4 versus constitutively active CDK-activating kinase. Cell Cycle. 2010;9:689–99. doi: 10.4161/cc.9.4.10611. [DOI] [PubMed] [Google Scholar]
  • 175.Anders L, Ke N, Hydbring P, Choi YJ, Widlund HR, Chick JM, Zhai H, Vidal M, Gygi SP, Braun P, et al. A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells. Cancer Cell. 2011;20:620–34. doi: 10.1016/j.ccr.2011.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Baker SJ, Reddy EP. CDK4: A Key Player in the Cell Cycle, Development, and Cancer. Genes Cancer. 2012;3:658–69. doi: 10.1177/1947601913478972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Schmetsdorf S, Arnold E, Holzer M, Arendt T, Gärtner U. A putative role for cell cycle-related proteins in microtubule-based neuroplasticity. Eur J Neurosci. 2009;29:1096–107. doi: 10.1111/j.1460-9568.2009.06661.x. [DOI] [PubMed] [Google Scholar]
  • 178.Zhong Z, Yeow WS, Zou C, Wassell R, Wang C, Pestell RG, Quong JN, Quong AA. Cyclin D1/cyclin-dependent kinase 4 interacts with filamin A and affects the migration and invasion potential of breast cancer cells. Cancer Res. 2010;70:2105–14. doi: 10.1158/0008-5472.CAN-08-1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Becker KA, Ghule PN, Lian JB, Stein JL, van Wijnen AJ, Stein GS. Cyclin D2 and the CDK substrate p220(NPAT) are required for self-renewal of human embryonic stem cells. J Cell Physiol. 2010;222:456–64. doi: 10.1002/jcp.21967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Sun Y, Lou X, Yang M, Yuan C, Ma L, Xie BK, Wu JM, Yang W, Shen SX, Xu N, et al. Cyclin-dependent kinase 4 may be expressed as multiple proteins and have functions that are independent of binding to CCND and RB and occur at the S and G 2/M phases of the cell cycle. Cell Cycle. 2013;12:3512–25. doi: 10.4161/cc.26510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Goodwin EC, DiMaio D. Repression of human papillomavirus oncogenes in HeLa cervical carcinoma cells causes the orderly reactivation of dormant tumor suppressor pathways. Proc Natl Acad Sci U S A. 2000;97:12513–8. doi: 10.1073/pnas.97.23.12513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Snow EC. Cyclin independent role for cdk4/6 during B cell lymphoma survival. Cell Cycle. 2010;9:2932. doi: 10.4161/cc.9.15.12708. [DOI] [PubMed] [Google Scholar]
  • 183.Gumina MR, Xu C, Chiles TC. Cyclin D3 is dispensable for human diffuse large B-cell lymphoma survival and growth: evidence for redundancy with cyclin E. Cell Cycle. 2010;9:820–8. doi: 10.4161/cc.9.4.10783. [DOI] [PubMed] [Google Scholar]
  • 184.Warenius HM, Kilburn JD, Essex JW, Maurer RI, Blaydes JP, Agarwala U, Seabra LA. Selective anticancer activity of a hexapeptide with sequence homology to a non-kinase domain of Cyclin Dependent Kinase 4. Mol Cancer. 2011;10:72. doi: 10.1186/1476-4598-10-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Gu W, Wang C, Li W, Hsu FN, Tian L, Zhou J, Yuan C, Xie XJ, Jiang T, Addya S, et al. Tumor-suppressive effects of CDK8 in endometrial cancer cells. Cell Cycle. 2013;12:987–99. doi: 10.4161/cc.24003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Burgess A, Wigan M, Giles N, Depinto W, Gillespie P, Stevens F, Gabrielli B. Inhibition of S/G2 phase CDK4 reduces mitotic fidelity. J Biol Chem. 2006;281:9987–95. doi: 10.1074/jbc.M512714200. [DOI] [PubMed] [Google Scholar]
  • 187.Gabrielli BG, Sarcevic B, Sinnamon J, Walker G, Castellano M, Wang XQ, Ellem KA. A cyclin D-Cdk4 activity required for G2 phase cell cycle progression is inhibited in ultraviolet radiation-induced G2 phase delay. J Biol Chem. 1999;274:13961–9. doi: 10.1074/jbc.274.20.13961. [DOI] [PubMed] [Google Scholar]
  • 188.Matsushime H, Quelle DE, Shurtleff SA, Shibuya M, Sherr CJ, Kato JY. D-type cyclin-dependent kinase activity in mammalian cells. Mol Cell Biol. 1994;14:2066–76. doi: 10.1128/mcb.14.3.2066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Chien WW, Domenech C, Catallo R, Salles G, Ffrench M. S-phase lengthening induced by p16(INK4a) overexpression in malignant cells with wild-type pRb and p53. Cell Cycle. 2010;9:3286–96. doi: 10.4161/cc.9.16.12600. [DOI] [PubMed] [Google Scholar]
  • 190.Dulić V. p16Ink4A, not only a G1 inhibitor? Cell Cycle. 2010;9:3150. doi: 10.4161/cc.9.16.12887. [DOI] [PubMed] [Google Scholar]
  • 191.Lu Y, Tatsuka M, Takebe H, Yagi T. Involvement of cyclin-dependent kinases in doxorubicin-induced apoptosis in human tumor cells. Mol Carcinog. 2000;29:1–7. doi: 10.1002/1098-2744(200009)29:1<1::AID-MC1>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  • 192.Chung JH, Bunz F. Cdk2 is required for p53-independent G2/M checkpoint control. PLoS Genet. 2010;6:e1000863. doi: 10.1371/journal.pgen.1000863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Varma H, Skildum AJ, Conrad SE. Functional ablation of pRb activates Cdk2 and causes antiestrogen resistance in human breast cancer cells. PLoS One. 2007;2:e1256. doi: 10.1371/journal.pone.0001256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Varma H, Conrad SE. Reversal of an antiestrogen-mediated cell cycle arrest of MCF-7 cells by viral tumor antigens requires the retinoblastoma protein-binding domain. Oncogene. 2000;19:4746–53. doi: 10.1038/sj.onc.1203827. [DOI] [PubMed] [Google Scholar]
  • 195.Schwartz EI, Smilenov LB, Price MA, Osredkar T, Baker RA, Ghosh S, Shi FD, Vollmer TL, Lencinas A, Stearns DM, et al. Cell cycle activation in postmitotic neurons is essential for DNA repair. Cell Cycle. 2007;6:318–29. doi: 10.4161/cc.6.3.3752. [DOI] [PubMed] [Google Scholar]
  • 196.Eguchi T, Takaki T, Itadani H, Kotani H. RB silencing compromises the DNA damage-induced G2/M checkpoint and causes deregulated expression of the ECT2 oncogene. Oncogene. 2007;26:509–20. doi: 10.1038/sj.onc.1209810. [DOI] [PubMed] [Google Scholar]
  • 197.Camarda G, Siepi F, Pajalunga D, Bernardini C, Rossi R, Montecucco A, Meccia E, Crescenzi M. A pRb-independent mechanism preserves the postmitotic state in terminally differentiated skeletal muscle cells. J Cell Biol. 2004;167:417–23. doi: 10.1083/jcb.200408164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Bourgo RJ, Ehmer U, Sage J, Knudsen ES. RB deletion disrupts coordination between DNA replication licensing and mitotic entry in vivo. Mol Biol Cell. 2011;22:931–9. doi: 10.1091/mbc.E10-11-0895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Bosco EE, Wang Y, Xu H, Zilfou JT, Knudsen KE, Aronow BJ, Lowe SW, Knudsen ES. The retinoblastoma tumor suppressor modifies the therapeutic response of breast cancer. J Clin Invest. 2007;117:218–28. doi: 10.1172/JCI28803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Sheppard KE, McArthur GA. The cell-cycle regulator CDK4: an emerging therapeutic target in melanoma. Clin Cancer Res. 2013;19:5320–8. doi: 10.1158/1078-0432.CCR-13-0259. [DOI] [PubMed] [Google Scholar]
  • 201.Musgrove EA, Caldon CE, Barraclough J, Stone A, Sutherland RL. Cyclin D as a therapeutic target in cancer. Nat Rev Cancer. 2011;11:558–72. doi: 10.1038/nrc3090. [DOI] [PubMed] [Google Scholar]
  • 202.Graf F, Mosch B, Koehler L, Bergmann R, Wuest F, Pietzsch J. Cyclin-dependent kinase 4/6 (cdk4/6) inhibitors: perspectives in cancer therapy and imaging. Mini Rev Med Chem. 2010;10:527–39. doi: 10.2174/138955710791384072. [DOI] [PubMed] [Google Scholar]
  • 203.Sun Y, Li YX, Wu HJ, Wu SH, Wang YA, Luo DZ, Liao DJ. Effects of an Indolocarbazole-Derived CDK4 Inhibitor on Breast Cancer Cells. J Cancer. 2011;2:36–51. doi: 10.7150/jca.2.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Sun Y, Luo D, Liao DJ. CyclinD1 protein plays different roles in modulating chemoresponses in MCF7 and MDA-MB231 cells. J Carcinog. 2012;11:12. doi: 10.4103/1477-3163.100401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Finn RS, Dering J, Conklin D, Kalous O, Cohen DJ, Desai AJ, Ginther C, Atefi M, Chen I, Fowst C, et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 2009;11:R77. doi: 10.1186/bcr2419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Choi YJ, Li X, Hydbring P, Sanda T, Stefano J, Christie AL, Signoretti S, Look AT, Kung AL, von Boehmer H, et al. The requirement for cyclin D function in tumor maintenance. Cancer Cell. 2012;22:438–51. doi: 10.1016/j.ccr.2012.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Choi YJ, Sicinski P. Unexpected outcomes of CDK4/6 inhibition. Oncotarget. 2013;4:176–7. doi: 10.18632/oncotarget.912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Lange CA, Yee D. Killing the second messenger: targeting loss of cell cycle control in endocrine-resistant breast cancer. Endocr Relat Cancer. 2011;18:C19–24. doi: 10.1530/ERC-11-0112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Thangavel C, Dean JL, Ertel A, Knudsen KE, Aldaz CM, Witkiewicz AK, Clarke R, Knudsen ES. Therapeutically activating RB: reestablishing cell cycle control in endocrine therapy-resistant breast cancer. Endocr Relat Cancer. 2011;18:333–45. doi: 10.1530/ERC-10-0262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Lamb R, Lehn S, Rogerson L, Clarke RB, Landberg G. Cell cycle regulators cyclin D1 and CDK4/6 have estrogen receptor-dependent divergent functions in breast cancer migration and stem cell-like activity. Cell Cycle. 2013;12:2384–94. doi: 10.4161/cc.25403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Leontieva OV, Demidenko ZN, Blagosklonny MV. MEK drives cyclin D1 hyperelevation during geroconversion. Cell Death Differ. 2013;20:1241–9. doi: 10.1038/cdd.2013.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Leontieva OV, Blagosklonny MV. CDK4/6-inhibiting drug substitutes for p21 and p16 in senescence: duration of cell cycle arrest and MTOR activity determine geroconversion. Cell Cycle. 2013;12:3063–9. doi: 10.4161/cc.26130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Hagen KR, Zeng X, Lee MY, Tucker Kahn S, Harrison Pitner MK, Zaky SS, Liu Y, O’Regan RM, Deng X, Saavedra HI. Silencing CDK4 radiosensitizes breast cancer cells by promoting apoptosis. Cell Div. 2013;8:10. doi: 10.1186/1747-1028-8-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Guha M. Blockbuster dreams for Pfizer’s CDK inhibitor. Nat Biotechnol. 2013;31:187. doi: 10.1038/nbt0313-187a. [DOI] [PubMed] [Google Scholar]
  • 215.Flaherty KT, Lorusso PM, Demichele A, Abramson VG, Courtney R, Randolph SS, Shaik MN, Wilner KD, O’Dwyer PJ, Schwartz GK. Phase I, dose-escalation trial of the oral cyclin-dependent kinase 4/6 inhibitor PD 0332991, administered using a 21-day schedule in patients with advanced cancer. Clin Cancer Res. 2012;18:568–76. doi: 10.1158/1078-0432.CCR-11-0509. [DOI] [PubMed] [Google Scholar]
  • 216.Dickson MA, Tap WD, Keohan ML, D’Angelo SP, Gounder MM, Antonescu CR, Landa J, Qin LX, Rathbone DD, Condy MM, et al. Phase II trial of the CDK4 inhibitor PD0332991 in patients with advanced CDK4-amplified well-differentiated or dedifferentiated liposarcoma. J Clin Oncol. 2013;31:2024–8. doi: 10.1200/JCO.2012.46.5476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Leonard JP, LaCasce AS, Smith MR, Noy A, Chirieac LR, Rodig SJ, Yu JQ, Vallabhajosula S, Schoder H, English P, et al. Selective CDK4/6 inhibition with tumor responses by PD0332991 in patients with mantle cell lymphoma. Blood. 2012;119:4597–607. doi: 10.1182/blood-2011-10-388298. [DOI] [PubMed] [Google Scholar]
  • 218.Dean JL, McClendon AK, Hickey TE, Butler LM, Tilley WD, Witkiewicz AK, Knudsen ES. Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors. Cell Cycle. 2012;11:2756–61. doi: 10.4161/cc.21195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.McClendon AK, Dean JL, Rivadeneira DB, Yu JE, Reed CA, Gao E, Farber JL, Force T, Koch WJ, Knudsen ES. CDK4/6 inhibition antagonizes the cytotoxic response to anthracycline therapy. Cell Cycle. 2012;11:2747–55. doi: 10.4161/cc.21127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Dean JL, McClendon AK, Knudsen ES. Modification of the DNA damage response by therapeutic CDK4/6 inhibition. J Biol Chem. 2012;287:29075–87. doi: 10.1074/jbc.M112.365494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Comstock CE, Augello MA, Goodwin JF, de Leeuw R, Schiewer MJ, Ostrander WF, Jr., Burkhart RA, McClendon AK, McCue PA, Trabulsi EJ, et al. Targeting cell cycle and hormone receptor pathways in cancer. Oncogene. 2013;32:5481–91. doi: 10.1038/onc.2013.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Cen L, Carlson BL, Schroeder MA, Ostrem JL, Kitange GJ, Mladek AC, Fink SR, Decker PA, Wu W, Kim JS, et al. p16-Cdk4-Rb axis controls sensitivity to a cyclin-dependent kinase inhibitor PD0332991 in glioblastoma xenograft cells. Neuro Oncol. 2012;14:870–81. doi: 10.1093/neuonc/nos114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Lukas J, Parry D, Aagaard L, Mann DJ, Bartkova J, Strauss M, Peters G, Bartek J. Retinoblastoma-protein-dependent cell-cycle inhibition by the tumour suppressor p16. Nature. 1995;375:503–6. doi: 10.1038/375503a0. [DOI] [PubMed] [Google Scholar]
  • 224.Barton KL, Misuraca K, Cordero F, Dobrikova E, Min HD, Gromeier M, Kirsch DG, Becher OJ. PD-0332991, a CDK4/6 inhibitor, significantly prolongs survival in a genetically engineered mouse model of brainstem glioma. PLoS One. 2013;8:e77639. doi: 10.1371/journal.pone.0077639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Johnson SM, Torrice CD, Bell JF, Monahan KB, Jiang Q, Wang Y, Ramsey MR, Jin J, Wong KK, Su L, et al. Mitigation of hematologic radiation toxicity in mice through pharmacological quiescence induced by CDK4/6 inhibition. J Clin Invest. 2010;120:2528–36. doi: 10.1172/JCI41402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Roberts PJ, Bisi JE, Strum JC, Combest AJ, Darr DB, Usary JE, Zamboni WC, Wong KK, Perou CM, Sharpless NE. Multiple roles of cyclin-dependent kinase 4/6 inhibitors in cancer therapy. J Natl Cancer Inst. 2012;104:476–87. doi: 10.1093/jnci/djs002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Han YK, Lee JH, Park GY, Chun SH, Han JY, Kim SD, Lee J, Lee CW, Yang K, Lee CG. A possible usage of a CDK4 inhibitor for breast cancer stem cell-targeted therapy. Biochem Biophys Res Commun. 2013;430:1329–33. doi: 10.1016/j.bbrc.2012.10.119. [DOI] [PubMed] [Google Scholar]
  • 228.Lehn S, Tobin NP, Berglund P, Nilsson K, Sims AH, Jirström K, Härkönen P, Lamb R, Landberg G. Down-regulation of the oncogene cyclin D1 increases migratory capacity in breast cancer and is linked to unfavorable prognostic features. Am J Pathol. 2010;177:2886–97. doi: 10.2353/ajpath.2010.100303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Tobin NP, Sims AH, Lundgren KL, Lehn S, Landberg G. Cyclin D1, Id1 and EMT in breast cancer. BMC Cancer. 2011;11:417. doi: 10.1186/1471-2407-11-417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Klemke RL. Trespassing cancer cells: ‘fingerprinting’ invasive protrusions reveals metastatic culprits. Curr Opin Cell Biol. 2012;24:662–9. doi: 10.1016/j.ceb.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Liu F, Korc M. Cdk4/6 inhibition induces epithelial-mesenchymal transition and enhances invasiveness in pancreatic cancer cells. Mol Cancer Ther. 2012;11:2138–48. doi: 10.1158/1535-7163.MCT-12-0562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Wang C, Li Z, Lu Y, Du R, Katiyar S, Yang J, Fu M, Leader JE, Quong A, Novikoff PM, et al. Cyclin D1 repression of nuclear respiratory factor 1 integrates nuclear DNA synthesis and mitochondrial function. Proc Natl Acad Sci U S A. 2006;103:11567–72. doi: 10.1073/pnas.0603363103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Icreverzi A, de la Cruz AF, Van Voorhies WA, Edgar BA. Drosophila cyclin D/Cdk4 regulates mitochondrial biogenesis and aging and sensitizes animals to hypoxic stress. Cell Cycle. 2012;11:554–68. doi: 10.4161/cc.11.3.19062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Fajas L. Re-thinking cell cycle regulators: the cross-talk with metabolism. Front Oncol. 2013;3:4. doi: 10.3389/fonc.2013.00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Fajas L, Blanchet E, Annicotte JS. The CDK4-pRB-E2F1 pathway : A new modulator of insulin secretion. Islets. 2010;2:51–3. doi: 10.4161/isl.2.1.10338. [DOI] [PubMed] [Google Scholar]
  • 236.Fajas L. Metabolic control in cancer cells. Ann Endocrinol (Paris) 2013;74:71–3. doi: 10.1016/j.ando.2013.03.021. [DOI] [PubMed] [Google Scholar]
  • 237.Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, Albassam M, Zheng X, Leopold WR, Pryer NK, et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 2004;3:1427–38. [PubMed] [Google Scholar]
  • 238.Fry DW, Bedford DC, Harvey PH, Fritsch A, Keller PR, Wu Z, Dobrusin E, Leopold WR, Fattaey A, Garrett MD. Cell cycle and biochemical effects of PD 0183812.A potent inhibitor of the cyclin D-dependent kinases CDK4 and CDK6. J Biol Chem. 2001;276:16617–23. doi: 10.1074/jbc.M008867200. [DOI] [PubMed] [Google Scholar]
  • 239.Lu H, Schulze-Gahmen U. Toward understanding the structural basis of cyclin-dependent kinase 6 specific inhibition. J Med Chem. 2006;49:3826–31. doi: 10.1021/jm0600388. [DOI] [PubMed] [Google Scholar]
  • 240.Dean JL, Thangavel C, McClendon AK, Reed CA, Knudsen ES. Therapeutic CDK4/6 inhibition in breast cancer: key mechanisms of response and failure. Oncogene. 2010;29:4018–32. doi: 10.1038/onc.2010.154. [DOI] [PubMed] [Google Scholar]
  • 241.Ferrer JL, Dupuy J, Borel F, Jacquamet L, Noel JP, Dulic V. Structural basis for the modulation of CDK-dependent/independent activity of cyclin D1. Cell Cycle. 2006;5:2760–8. doi: 10.4161/cc.5.23.3506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Liu S, Bolger JK, Kirkland LO, Premnath PN, McInnes C. Structural and functional analysis of cyclin D1 reveals p27 and substrate inhibitor binding requirements. ACS Chem Biol. 2010;5:1169–82. doi: 10.1021/cb1001262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Sheaff RJ, Groudine M, Gordon M, Roberts JM, Clurman BE. Cyclin E-CDK2 is a regulator of p27Kip1. Genes Dev. 1997;11:1464–78. doi: 10.1101/gad.11.11.1464. [DOI] [PubMed] [Google Scholar]
  • 244.Bellovin DI, Das B, Felsher DW. Tumor dormancy, oncogene addiction, cellular senescence, and self-renewal programs. Adv Exp Med Biol. 2013;734:91–107. doi: 10.1007/978-1-4614-1445-2_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Erenpreisa J, Cragg MS. Three steps to the immortality of cancer cells: senescence, polyploidy and self-renewal. Cancer Cell Int. 2013;13:92. doi: 10.1186/1475-2867-13-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Mackenzie I, Rous P. The experimental disclosure of latent neoplastic changes in Tarred skin. J Exp Med. 1941;73:391–416. doi: 10.1084/jem.73.3.391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Noble RL, Hochachka BC, King D. Spontaneous and estrogen-produced tumors in Nb rats and their behavior after transplantation. Cancer Res. 1975;35:766–80. [PubMed] [Google Scholar]
  • 248.Noble RL. Sex steroids as a cause of adenocarcinoma of the dorsal prostate in Nb rats, and their influence on the growth of transplants. Oncology. 1977;34:138–41. doi: 10.1159/000225207. [DOI] [PubMed] [Google Scholar]
  • 249.Noble RL. Production of Nb rat carcinoma of the dorsal prostate and response of estrogen-dependent transplants to sex hormones and tamoxifen. Cancer Res. 1980;40:3547–50. [PubMed] [Google Scholar]
  • 250.Noble RL, Cutts JH. Mammary tumors of the rat: a review. Cancer Res. 1959;19:1125–39. [PubMed] [Google Scholar]
  • 251.Liao DJ, Dickson RB. Roles of androgens in the development, growth, and carcinogenesis of the mammary gland. J Steroid Biochem Mol Biol. 2002;80:175–89. doi: 10.1016/S0960-0760(01)00185-6. [DOI] [PubMed] [Google Scholar]
  • 252.Tran PT, Fan AC, Bendapudi PK, Koh S, Komatsubara K, Chen J, Horng G, Bellovin DI, Giuriato S, Wang CS, et al. Combined Inactivation of MYC and K-Ras oncogenes reverses tumorigenesis in lung adenocarcinomas and lymphomas. PLoS One. 2008;3:e2125. doi: 10.1371/journal.pone.0002125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Leontieva OV, Demidenko ZN, Gudkov AV, Blagosklonny MV. Elimination of proliferating cells unmasks the shift from senescence to quiescence caused by rapamycin. PLoS One. 2011;6:e26126. doi: 10.1371/journal.pone.0026126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence. Genes Dev. 2010;24:2463–79. doi: 10.1101/gad.1971610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Kaplon J, Zheng L, Meissl K, Chaneton B, Selivanov VA, Mackay G, van der Burg SH, Verdegaal EM, Cascante M, Shlomi T, et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature. 2013;498:109–12. doi: 10.1038/nature12154. [DOI] [PubMed] [Google Scholar]
  • 256.Leontieva OV, Lenzo F, Demidenko ZN, Blagosklonny MV. Hyper-mitogenic drive coexists with mitotic incompetence in senescent cells. Cell Cycle. 2012;11:4642–9. doi: 10.4161/cc.22937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Leontieva OV, Demidenko ZN, Blagosklonny MV. S6K in geroconversion. Cell Cycle. 2013;12:3249–52. doi: 10.4161/cc.26248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Chandeck C, Mooi WJ. Oncogene-induced cellular senescence. Adv Anat Pathol. 2010;17:42–8. doi: 10.1097/PAP.0b013e3181c66f4e. [DOI] [PubMed] [Google Scholar]
  • 259.Duesberg P, McCormack A. Immortality of cancers: a consequence of inherent karyotypic variations and selections for autonomy. Cell Cycle. 2013;12:783–802. doi: 10.4161/cc.23720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Hayflick L. THE LIMITED IN VITRO LIFETIME OF HUMAN DIPLOID CELL STRAINS. Exp Cell Res. 1965;37:614–36. doi: 10.1016/0014-4827(65)90211-9. [DOI] [PubMed] [Google Scholar]
  • 261.Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307–15. doi: 10.1038/nature03098. [DOI] [PubMed] [Google Scholar]
  • 262.Salminen A, Ojala J, Kaarniranta K. Apoptosis and aging: increased resistance to apoptosis enhances the aging process. Cell Mol Life Sci. 2011;68:1021–31. doi: 10.1007/s00018-010-0597-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Elion GB. Selectivity--key to chemotherapy: presidential address. Cancer Res. 1985;45:2943–50. [PubMed] [Google Scholar]
  • 264.Vincent MD. Cancer: beyond speciation. Adv Cancer Res. 2011;112:283–350. doi: 10.1016/B978-0-12-387688-1.00010-7. [DOI] [PubMed] [Google Scholar]
  • 265.Gregory CD, Pound JD. Cell death in the neighbourhood: direct microenvironmental effects of apoptosis in normal and neoplastic tissues. J Pathol. 2011;223:177–94. doi: 10.1002/path.2792. [DOI] [PubMed] [Google Scholar]
  • 266.Hanahan D. Rethinking the war on cancer. Lancet. 2014;383:558–63. doi: 10.1016/S0140-6736(13)62226-6. [DOI] [PubMed] [Google Scholar]
  • 267.Deschênes-Simard X, Gaumont-Leclerc MF, Bourdeau V, Lessard F, Moiseeva O, Forest V, Igelmann S, Mallette FA, Saba-El-Leil MK, Meloche S, et al. Tumor suppressor activity of the ERK/MAPK pathway by promoting selective protein degradation. Genes Dev. 2013;27:900–15. doi: 10.1101/gad.203984.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Morandell S, Yaffe MB. Exploiting synthetic lethal interactions between DNA damage signaling, checkpoint control, and p53 for targeted cancer therapy. Prog Mol Biol Transl Sci. 2012;110:289–314. doi: 10.1016/B978-0-12-387665-2.00011-0. [DOI] [PubMed] [Google Scholar]
  • 269.Chan DA, Giaccia AJ. Harnessing synthetic lethal interactions in anticancer drug discovery. Nat Rev Drug Discov. 2011;10:351–64. doi: 10.1038/nrd3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Vaškovičová K, Žárský V, Rösel D, Nikolič M, Buccione R, Cvrčková F, Brábek J. Invasive cells in animals and plants: searching for LECA machineries in later eukaryotic life. Biol Direct. 2013;8:8. doi: 10.1186/1745-6150-8-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Wells A, Griffith L, Wells JZ, Taylor DP. The dormancy dilemma: quiescence versus balanced proliferation. Cancer Res. 2013;73:3811–6. doi: 10.1158/0008-5472.CAN-13-0356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Rösel D, Brábek J, Veselý P, Fernandes M. Drugs for solid cancer: the productivity crisis prompts a rethink. Onco Targets Ther. 2013;6:767–77. doi: 10.2147/OTT.S45177. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cell Cycle are provided here courtesy of Taylor & Francis

RESOURCES