Skip to main content
Journal of Cell Communication and Signaling logoLink to Journal of Cell Communication and Signaling
. 2017 Feb 20;11(1):79–87. doi: 10.1007/s12079-017-0381-y

What roles do colon stem cells and gap junctions play in the left and right location of origin of colorectal cancers?

James E Trosko 1,, Heinz-Josef Lenz 2
PMCID: PMC5362582  PMID: 28220297

Abstract

This “Commentary” examines an important clinical observation that right-sided colorectal cancers appear less treatable than the left-sided cancers. The concepts of (a) the “initiation/promotion/progression” process, (b) the stem cell hypothesis, (c) the role gap junctional intercellular communication, (d) cancer cells lacking GJIC either because of the non-expression of connexin genes or of non-functional gap junction proteins, and (e) the role of the microbiome in promoting initiated colon stem cells to divide symmetrically or asymmetrically are examined to find an explanation. It has been speculated that “embryonic-like” lesions in the ascending colon are initiated stem cells, promoted via symmetrical cell division, while the polyp-type lesions in the descending colon are initiated stem cells stimulated to divide asymmetrically. To test this hypothesis, experiments could be designed to examine if right-sided lesions might express Oct4A and ABCG2 genes but not any connexin genes, whereas the left-sided lesions might express a connexin gene, but not Oct4A or the ABCG2 genes. Treatment of the right sided lesions might include transcriptional regulators, whereas the left-sided lesions would need to restore the posttranslational status of the connexin proteins.

Keywords: Colorectal cancers, Adult stem cells, Gap junction intercellular communication, Colon stem cells, Cancer stem cells, Connexin genes, Microbiome

Introduction

With the important observation that the location of the origin of colorectal cancers seems to determine the lethality of the tumors (Loupakis et al. 2015a, b), the significance of this observation is telling us to look for some fundamental biological underpinning that might serve to provide guides to either or both prevention and treatment of colon cancers. Metastatic colorectal cancers (CRC) can be defined based on the location of the primary tumour. Historically, publications have defined CRCs within three compartments of the gut: distal colon, proximal colon and rectum ( Bufill 1990;. Gervaz et al. 2004; Iacopetta 2002) Right-sided colon carcinomas (RCC) are located within the midgut, which encompasses the proximal two-thirds of the transverse colon, ascending colon and caecum. Left-sided colon carcinomas (LCC) lie within the hindgut, which includes the known as the descending colon, sigmoid colon and rectum. Patients with RCC and LCC differ in their microbiome, clinical characteristics, molecular profiling, clinical outcome and response to treatment. However, embryology cannot explain these differences since the midgut and hindgut are derive from the same origin, the endoderm, and the rectum derives from the cloacal membrane (Hill 2016; Sadler 2014) Microbiome differs significantly between the right- and left-sided colon which may explain the gradual changes in MSI and CIMP types of CRC along the length of the gut (Yamauchi et al. 2012; Gagniere et al. 2016). Regional differences in the physiological functioning of the right & left regions of the colon might expose these two types of initiated stem cells could affect either the anti-mitogenic factors ( niche extra-cellular matrices and soluble factors, low oxygen microenvironment) for the non-expressing connexin stem cells or the contact- inhibiting factors (pollutants; dietary components; drugs, hormones, growth factors, cytokines, microbial toxins, etc.) for the connexin-expressing, but non GJIC-functioning initiated stem cells.”

LCC are more chromosomally instable and RCC have a higher frequency of MSI-high phenotypes, and KRAS and BRAF mutations (Venook et al. 2016; Heinemann et al. 2014), which may account for the poorer prognosis of these patients. Location of the primary tumor is a known prognostic factor for patients with CRC (Loupakis et al. 2015; Nitsche et al. 2016; Price et al. 2015; Sinicrope et al. 2015; Venook et al. 2016; Wong et al. 2016). RCC are associated with a poorer prognosis than LCC but may be also predictive for anti-EGFR and anti-angiogenic therapies. (Loupakis et al. 2015a, b; Venook et al.; 2016; Wong et al. 2016; Houts et al. 2016; Lenz et al. 2016)

As with all scientific and medical problems, we have a choice to tackle the problem either with an empirical or a hypothesis-testing experimental approach. Given today’s propensity to use powerful and sophisticated technologies to solve medical problems, much of current cancer research (basic; chemo-preventive; chemo-therapeutic; translational, etc.) hopes to generate massive amounts of data, from which various uses of different algorithms of bioinformatics will lead to identification of critical patterns for “personalized” medicine. Yet, the alternative approach to use biologically-based hypothesis-testing seems to depend on the apparent lack of universally-accepted mechanisms to explain the complex multi-stage, multi-mechanism origin of all cancers. The seemingly complex interactions of genetic, developmental, gender, physical/chemical/microbiological agent interactions, doses/concentrations, chemical mixture interactions affecting the redox state of cells, psychological, behavioral, social and cultural factors make for simple hypothesis-testing very difficult. Any cancer researcher knows that to submit a research proposal with a hypothesis-testing strategy will meet with a great chance of a critical review, challenging the hypothesis used to propose the experimental testing, compared to a proposal with no hypothesis being proposed but with the highlighting of the state of the art technologies to generate tons of data.

It is within this broad background that there exists a rationale for a hypothesis to explain this observation that the location of the polyp-type, partially differentiated colorectal cancers is different from the flat, undifferentiated- type colorectal tumors that, somehow, makes one type more treatable than the other. The rationale for the hypothesis to be presented is independent on the reality that the former type is easily detected during colonoscopy, whereas the flat-type is more easily missed. The hypothesis to be proposed here is founded on an old, but solid, observation that Loewenstein and Kanno made decades ago (1966), namely, that all cancer cells, unlike normal cells, lack gap junctional intercellular communication. Before expanding on his observation, it should be noted that his papers are rarely cited by the most cited cancer researchers. In addition, while the role of gap junctions in the carcinogenic process is clearly complex, and the link between “cancer cells” and the lack of functional gap junctions in cancer cells, itself, is complex (Aasen et al. 2016; Leithe et al. 2006; Saez 2016; Jiang and Penuela 2016; Trosko 2016a, b) such as the observation of “selective” lack of gap junctional intercellular communication (Yamasaki et al. 1987), the assumption being made here is that GJIC is a necessary, if not sufficient, requirement for a cancer cell to be immortal, have uncontrolled growth, cannot terminally differentiate and does not apoptose correctly.

In order to characterize the origin of cancer cells, two additional hypotheses have driven cancer research, off and on, for many years, namely that: (a) cancers originate from adult-organ-specific stem cells (Markert 1968; Pierce 1974; Fialkow 1976; Potter 1978; Till 1982; Trosko and Tai 2006) or (b) from the “de-differentiation” or “re-programming” of somatic differentiated cells (Sell 1993). Only with the discovery of human embryonic stem cells (Bongso et al. 1994; Thomson et al. 1998; Shamblott et al. 1998), induced- pluripotent stem cells (Takahashi and Yamanaka 2006), organ-specific adult human stem cells (Tai et al. 2005), as well as the re-introduction of the idea of “cancer-initiating” or “cancer stem cells” (Trosko 2014) has it been possible to start to characterize these normal stem cells and cancer stem cells. While it can be said, up front, that there is no universal acceptance of which of these two hypotheses might explain what is the “target cell” for cancers, there is some strong evidence that the adult organ-specific stem cell is the “target” cell for the conversion of this adult stem cell to be initiated (Tai et al. 2005; Bonnet and Dick 1997; Barker et al. 2009; Wang et al. 2009; Sanchez et al. 2016). The interpretation of an organ-specific adult stem cell being “initiated” by some agent that can influence carcinogenesis is that this initiated adult stem cell will be unable to terminally differentiate and remain in its normal “undifferentiated” state [ or promoted] (Trosko and Kang 2012), to accrue all those other mutagenic and epigenetic changes to acquire the “hallmarks” of cancer (Hanahan and Weinberg 2000; Hanahan and Weinberg 2011).

Yet, this alone will not explain the original differences in the left and right origin of two different kinds of cancer. Another series of observations, usually dismissed in current cancer research papers and research proposals, was built on Loewenstein’s original characterization of the lack of gap junctional intercellular communication (GJIC ) in cancer cells. That observation came from historic observations on the ability of some chemicals and physical conditions that could cause a single “initiated” cell to be selectively expanded by hyperplasic growth and a loss of apoptosis (Berenblum 1954; Yamagiwa and Ichikawa 1977; Trosko 2001). This process, seen in skin, liver, breast, and bladder tissues in vivo, was termed, tumor promotion (Weinstein et al. 1984; Pitot and Dragon 1991). What is interesting about these tumor-promoting chemicals is that they were not mutagenic, but acted by “epigenetic” mechanisms (Trosko et al. 1990; Trosko and Ruch 1998; Trosko et al. 1998; Upham and Trosko 2009). In brief, the powerful mouse skin tumor promoter (phorbol ester or TPA), as shown to inhibit, reversibly, GJIC (Yotti et al. 1979). Later, more non-mutagenic chemicals, e.g., DDT, phthalates, phenobarbital, etc., could inhibit GJIC, reversibly, by triggering redox changes in cells and trigger various intracellular signals to alter GJIC (Upham and Trosko 2009; Trosko and Chang 1988). Species specificity and threshold levels for these tumor promoting chemicals were discovered (Pitot and Dragon 1991; Klaunig and Ruch 1987).

Later, using the logic that there exist chemicals that could reversibly inhibit GJIC to be tumor promoters, there might be chemicals that could do exactly the opposite to GJIC. This led to predict, successfully, that both natural and synthetic chemicals do exist, which could either prevent GJIC-inhibiting tumor promoters or that could enhance, transcriptionally, enhanced expression of connexin genes. (Trosko and Ruch 2002; Leone et al. 2012).

In addition, various oncogenes, such as src, ras, and neu, could stably inhibit gap junctions (Trosko and Ruch 1998). This observation starts to provide some insight as to why there might be two types of cancer cells that do not have functional GJIC intercellular communication (Trosko 2003). To understand the basis of this statement, another observation, rarely mentioned, is that stem cells (embryonic, induced pluripotent stem cells, adult organ-specific stem cells) do not express their gap junction or connexin genes [of which there are twenty in this highly evolutionarily-conserved family (Cruciani and Mikalsen 2006). While there have been contradictory studies suggesting the expression of connexin genes or the functionality of GJIC in various types of stem cells, due to different methodologies used, the fact that embryonic-, induced pluri-potent- and organ-specific- stem cells are propagated on feeder layer cells, suggests that there was no GJIC between the stem cells and the feeder layer cells. One example is seen in Figs. 5 and 6 (Chang et al. 1987) and Fig. 3 (Kao et al. 1995). This observation that stem cells do not express their connexin genes or have functional GJIC is because of the evolutionary role of gap junctions to help maintain homeostatic control of cell proliferation, cell differentiation and apoptosis (Trosko 2016a, b). While the evolutionary appearance of this family of connexin or gap junction genes, as well as the germinal and somatic stem cells (Crosnier et al. 2006; Mentink and Tsiantis 2015; Weissman 2015; Horn et al. 2015), has yet to be explained, the appearance of multi-cellularity and gap junctions and stem cells seemed to depend on each other (Trosko 2016a, b).

With the accepted definition of a stem cell as a cell having the ability to divide either symmetrically to maintain self-renewal or by asymmetrical division to produce one daughter that maintains self -renewal and another daughter that can terminally differentiate, it seems that, while there are obvious genes needed to control which division process to initiate, critical endogenous and exogenous factors must trigger the symmetrical or asymmetrical division process during development and subsequent state of health during adolescent, mature and geriatric stages of life (Lane et al. 2014). With the stem cell seemingly having metabolic characteristics of anaerobic single cell organisms, namely, metabolism of glucose via glycolysis, having few mitochondria (Neste et al. 2007; Armstrong et al. 2010; Chen et al. 2008; Prigione et al. 2010), and aversion to high levels of oxygen with their protective niches (Csete 2005; Pervaiz et al. 2009; Mohyeldin et al. 2010; Brigelius-Flohé and Flohé 2011; Atena et al. 2014), the stem cell’s phenotype mimics that of the single cell organism (Trosko 2014). In addition, given the old observation that cancer cells exhibit the Warburg metabolism (Warburg 1956) and the more recent identification of the “cancer initiating” or “cancer stem cells”(Al Hajj et al. 2003; Cozzio et al. 2003), as well as evidence that the organ-specific adult stem cell as being the origin of these “cancer stem” cells (Tai et al. 2005; Hemmati et al. 2003; Hope et al. 2003; Singh et al. 2003; Galli et al. 2004; Kondo et al. 2004; Yuan et al. 2004; De Jong et al. 2005; Locke et al. 2005; Ponti et al. 2005; Krivtsov et al. 2006; Li and Yurchenco 2006; Gu et al. 2007; O’Brien et al. 2007; Ricci-Vitiani et al. 2007; Wu et al. 2007; Schatton et al. 2008), several observations bring the stem cell hypothesis and “de-differentiation’ or “reprogramming” hypothesis of cancer for further examination.

One of the prevailing paradigms in the cancer field is that the first step of the “initiation”, “promotion” and “progression” process of carcinogenesis is the “immortalization” of a normal “mortal cell” (Land et al. 1983). In effect, it is generally assumed that the normal, mortal cell, which was “initiated” or “immortalized”, was a somatic differentiated cell. Even more recent evidence of the Nobel Prize work by S. Yamanaka, that one could “re-program” a normal differentiated cell by the genetic insertion of a few “embryonic genes” (Takahashi and Yamanaka 2006), seemed to support the “de-differentiation” or “reprogramming” hypothesis of the cancer process. However, an alternative hypothesis has been proposed (Trosko 2006, 2008a, b, 2009, 2014).

In brief, if one assumes that a stem cell is fundamentally “immortal” until it is induced to terminally differentiate or to become “mortal”, then there are important facts, for which there must be some accounting. First, all organs of the human body are known to have organ-specific adult stem cells (e.g., skin, liver, intestine, eye, brain, blood, pancreas, kidney, ovary, testes, bone, lung, etc.). Second, in all early primary in vitro cultures of tissues, there are a few adult organ-specific adult stem cells, which, when grown under typical in vitro conditions (e.g., on plastic; high oxygen levels, etc.), are lost after a few passages. That is why primary cultures usually die out after a finite number of passages [Hayflick phenomenon] (Hayflick 1965). Third, when one places the Yamanaka-type “induced pluripotent stem cells” back into an adult host animal, they form teratomas. If, in fact, cancers are induced in vivo in human beings by the “re-programming” process of “immortalizing” a differentiated somatic cell, then teratomas, not sarcomas or carcinomas, would be formed. Last, there is strong evidence that adult stem cells are the target cells for initiating the cancer (Tai et al. 2005; Bonnet and Dick 1997; Barker et al. 2009;Wang et al. 2009; Sanchez et al. 2016).

If the organ-specific adult stem cell, which, by definition, has an unlimited life span and which, under normal conditions, mutations could be formed by “error –prone DNA repair” (Cleaver and Trosko 1970; Maher and McCormick 1976; Cleaver 1978; Glover et al. 1979; Brash et al. 1991) or “error-prone- DNA Replication ( Warren et al. 1981). If those mutations occurred in a stem cell’s genes to regulate asymmetrical cell division, but does not affect symmetrical cell division, then that stem cell would be able to expand but not terminally differentiate (Kang and Trosko 2011). In effect, the initiation process does not induce “immortalization” of a “mortal” somatic differentiated cell, but it blocks “mortalization” or terminal differentiation of an “immortal” adult stem cell.

At this point, the observation of “immortalizing viruses”, such as SV40 and human papilloma viruses, should be brought into this analysis (Aasen et al. 2016). Historically, in the many attempts to obtain transformed cells in vitro by exposing normal primary cultured cells to various radiations or carcinogenic chemicals, nothing but failures were reported (DiPaolo 1983; Kuroki and Huh 1993; Rhim 1993). Only with the infection or transfection of the immortalizing viruses or some of their genes in primary fibroblastic or epithelial cells, could one isolate a few “immortalized” cells. Hence, the original logical designation of the terms, “immortalizing viruses” occurred. However, another interpretation of these observations can, again, be made. In those primary cultures, there exist a few adult organ-specific stem cells. If, these viruses or their “immortalizing” genes, infected or transfected, all the cells (differentiated and the few adult stem cells), only the stem cells survived the subsequent passages. The infected differentiated cells died during “crisis period” via the Hayflick phenomenon. However, if the SV40 and HPV E-6, E-7 genes rendered the p53 and RB gene products unable to bring about differentiation of the stem cells (Ahuja et al. 2005), these cells remain “immortal”, undifferentiated, since they were not “re-programmed” to convert their differentiated state or state of “mortality” to that of the “immortalized” phenotype. This is a radical change of the paradigm to explain the first step of carcinogenesis.

If this explanation is correct, how might this be of help to either prevent or treat colorectal cancers, based on the observation that right-sided tumors have a worse prognosis than those of the left sided colorectal tumors? An explanation can be offered. It is based on the assumption that, if all tumors contain cells that cannot perform GJIC or performs “selective” GJIC ( Yamasaki et al. 1987) , which is needed for “contact inhibition” or growth control (Eagle 1965), needed for differentiation (Yamasaki and Naus 1996) and for apoptosis (Wilson et al. 2000), then one must note that there exist two kinds of cancer cells that do not have functional GJIC (Trosko 2007). There are those cancer cells, which started from a stem cell that was “initiated” before the connexin genes were transcriptionally expressed. These cells are “frozen” in their “immortalized” and undifferentiated state. While not being neoplastically- transformed at this “initiated” stage, they can still be mitogenically- repressed by insoluble or soluble anti-mitogenic growth factors, such as niche- extracellular matrix molecules or negative soluble growth- regulators that bind to their receptors and signal transducers that control mitogenesis.

Given that prevention of initiation of carcinogenesis can be reduced, but not to zero levels, due to the normal production of mutations by “errors of replication” caused by normal stem cell replication, the promotion phase of carcinogenesis is the “rate-limiting” step of carcinogenesis, as it takes place of decades in adults (Trosko 2006). Therefore, in both the right and left regions of the colon, promotion is the process that would cause the initiated stem cell expansion. The promotion process requires an endogenous (growth factor, hormone, cytokine) or exogenous (pollutant, medication, microbial toxin, dietary chemical, etc.) factors to release the initiated stem cell from anti-mitogenic agents, as well to block apoptotic factors, in order to expand this pre-malignant population. In addition, promoters must exceed “threshold” levels in order to block gap junctional intercellular communication, while at the same time, work in the absence of “anti-promoters” (Leone et al. 2012).Only after stimulation by “promoters” that release these “initiated”, undifferentiated stem cells from anti-mitotic restraint, could these initiated stem cells expand to form flat, undifferentiated or “embryonic-like” lesions.

On the other hand, if an early stem cell, which has its connexin genes transcriptionally expressed, but has either (a) an expressed mutated oncogene, such as neu, src, or ras, these oncogene products can, posttranslationally, modify the connexin proteins to render them non-functional (Trosko and Ruch 2002) or (b) exposed chronically to promoters that can inhibit gap junctions, then these cells can be clonally amplified or promoted to form polyp –type lesions. These cells are also phenotypically like the initiated stem cell, in that they both are unable to communicate intercellular signals for growth control.

So, what might be the promoters of these two types of “initiated” stem cells? There are many agents that can inhibit mitogenesis of (a) an initiated stem cell without expressed connexins or gap junctions or (b) a contact- inhibited, gap junctional intercellular communication –expressing, partially differentiated stem cells. This is a major barrier in screening for a “specific” promoter for colon cancers. So many factors can cause “promotion” of either type of “initiated” colon cancer stem cell. This is seen in a major observation that, depending on the stage of development when initiation occurred (pre- and post- weaning), the tumor promoter, phenobarbital, affected two types of lesions in the rat (Lee 2000)

To recapitulate the logical chain of thought, human intestinal tissue contains adult stem cells (Barker et al. 2007; Potten et al. 2009; Kemper et al. 2012; Sato and Clevers 2013). If the adult stem cell is capable of either symmetrical or asymmetrical cell division, depending on endogenous or exogenous signals it receives (Barker 2014), then either expansion of the stem cell population or of its progenitor/differentiated lineages will follow. Also, if one assumes that these intestinal stem cells are the “target” cells for colon cancers (Huels and Sansom 2015), the first step of the carcinogenic process is to block its ability to divide asymmetrically under normal conditions to be “initiated” (Trosko 2016a, b). However, if that stem cell is “initiated” in the stem cell state, and “promoted” by agents (endogenous or exogenous) that favors symmetrical cell division, these initiated colon stem cell cells will form, undifferentiated-type or “flat” lesions. On the other hand, if the initiated colon stem cell is promoted by endogenous or exogenous factors that can force some “partial differentiation (“oncogeny as partially block ontogeny” (Potter 1978), then lesions will form polyps.

One has to note that normal growth modulators of cell proliferation, such as growth factors (Ruch et al. 2001), cytokines and chemokines (Martin and Prince 1982; Brosnan et al. 2001), hormones (Puri and Garfield 1982), as well as dietary factors and drugs and pollutants, and products of food preparation (Dougal et al. 2012), can alter GJIC . Therefore, the question arises, “Why are the lesions arising from the right-sided adult colon initiated stem cells giving rise to “flat lesions, while those adult initiated stem cells promoted to form the polyp –type partially differentiated lesion”? Could it be related to the fact that the ascending colon has a very different physiological function than the descending colon? Or could it be related to the possibility that the internal milieu of these two regions of the colon are home to two different populations of microbiome organisms that secrete different factors that might affect the initiated adult stem cells of the two regions of the colon differently?

With the explosion of new physiological and pathological roles of the intestinal microbiome, one might be able to hypothesize that there exists in the right and left regions of the colon, products of normal colon physiology, as well as products of regional microbiome populations, that might explain differential “promoting” effects on the “initiated” stem cell that triggers their mitogenesis and prevents their differentiation, leading to the resistant, flat type of tumors on the right side. On the other hand, differential normal physiological by-products of the left region or of the microbiome population of the left side could cause the clonal expansion of the partially-differentiated stem cell with expressed connexin genes. With expanding literature of the differential distribution of micro-organisms in these two regions (Dougal et al. 2012; Nava et al. 2012; Flemer et al. 2016), it might be reasonable to predict the microbial toxins of these two regions might have differential effects on these two types of initiated but non-communicating stem cells.

In summary, it is speculated that the differences of the right- and left-sided origin of colorectal cancers’ sensitivity to anti-cancer treatments might be related to differential factors that could affect initiated stem cells’ ability to divide either symmetrically to form flat-type or embryonic-like lesions, which express Oct4A and ABCG2 genes, but not the connexin genes. On the other hand, if those factors in the left side of the colon affect the initiated stem cells to divide asymmetrically, then partially-differentiated or polyp-type lesions would be formed. These lesions probably do not express Oct4A or ABCG2 drug transporter genes, but do express some connexin gene. In addition, it has been shown that genetic and epigenetic changes in right and left colon cancer are significant different including microsatellite instability, methylation status and mutations, such as ras and raf, suggesting differential etiology of carcinogenesis and possible different initiated stem cells (Missiaglia et al. 2014).

To find a rationale for treatment strategies for lesions starting from the two different regions of the colon, it is proposed that, starting from Loewenstein’s original observation that cancer cells: (a) do not “contact inhibit” or have growth control; (b) do not terminally differentiate; and (c) do not apoptose properly under normal conditions, and (d) do not have functional gap junctional intercellular communication [Loewenstein and Kanno 1966], one should try to get these cancer cells to restore these properties. Also, since it has been shown that there are two kinds of cancer cells that lack deficient gap junctional intercellular communication (Trosko 2003), first, those that never expressed their connexin genes and are very embryonic-like and second, those that do have expressed connexin genes but these proteins are rendered non-functional by some posttranscriptional/posttranslational modification, be treated differently.

This, then, leads to the phenotypic differences in responses of the colon cancers of right and left regions to anti-cancer treatments. The cancer cells are very different, epigenetically, in a manner that makes them very different, phenotypically, to be treated as “cancer cells”. The same anti-cancer agent given to an “initiated” stem cell, which has its connexin genes, epigenetically repressed at the transcriptional level, will respond to this agent very differently that a cancer cell that is partially differentiated because it has its connexin gene expressed, but is unable to communicate with a dysfunctional gap junction protein. From a theoretical perspective, to convert a cancer cell, which has no expressed connexin and no functional GJIC, to one that might be contact inhibited, differentiate or apoptose as a normal cell, one needs to transcriptionally activate the connexin gene. The use of agents, such as suberoylanilide hydroxamic acid ,SAHA, a histone deacetylase (HDAC) inhibitor, (Ogawa et al. 2005), might be a strategy for the colorectal right located, embryonic-like undifferentiated tumors. On the other hand, treatment of tumor cells with expressed connexin genes but no functional GJIC because of a post-translationally-modified connexin protein might be treated with various inhibitors of oncogenes or agents that ameliorate the action of GJIC-inhibiting chemical promoters that could be rending the gap junction protein from being transported to membrane or from forming gap junctions.

To test this hypothesis to explain this important observation of the location of the tumor in the colon that determines its sensitivity or resistance to treatment, there seems to be a straight forward testable series of experiments. Since stem cells and “cancer stem cells” express OCT4A, express drug transporter genes, such ABCG2, but do not express connexin genes, the right-sided colorectal cancer tissues should be positive for these and other stem cell markers. The differentiated left- sided colorectal tumors, on the other hand, should express some differentiated colon genes, express connexin genes and gap junction proteins, but little or no OCT4A gene and no expressed drug transporter genes. Of course, the additional real problem of all tumors is the fact that tumors are mixtures of “cancer stem cells” and “cancer non-stem cells” (particularly, the polyp-type), as well as stromal and invasive immune cells. This complex micro-environment of many interacting soluble growth regulators, and cytokines, as well as insoluble extracellular matrix factors, must also be considered in both the design and interpretation of future experiments.

In conclusion, one critical hallmark of all normal cells (organ-specific stem cells; progenitor cells) is their ability to respond to growth suppression signals (secreted factors; extra-cellular matrix; gap junctions). On the other hand, cancer cells are characterized by their loss of growth control, inability to terminally-differentiate and dysfunctional apoptosis, which is correlated with their loss of gap junction function, either by the cancer cells having no transcription of any connexin gene or by having their expressed connexin protein made dysfunctional by posttranslational modification. Gap junction function can be modulated by many induced intra-cellular signaling pathways, e.g. phorbol ester- induced PK-C; src- induced signaling; DDT- altered Ca++ signaling; etc.). Stem cells can be stimulated to divide either symmetrically or asymmetrically by many factors. Initiated stem cells appear to be able only to divide symmetrically by promoting, epigenetic agents that can trigger various intra-cellular signals. In vivo, depending on the tissue/organ, various endogenous hormones, growth factors or cytokines could act as these agents to stimulate initiated stem cells. In addition, exogenous agents, such as pollutants, drugs, dietary factors, or toxins from the microbiome, could act to promote the symmetrical cell division of initiated organ-specific stem cells. In the case of the differences in response to cancer therapeutic agents, the resistance or sensitivity of the right- and left derived colorectal cancers might be related to the micro-environmental milieu of the adult stem cells’ response to different promoting factors in the ascending and descending colon.

Compliance with ethical standards

Funding

This Commentary was done independent of any outside support.

References

  1. Aasen T, Mesnil M, Naus CC, Lampe PD, Laird DW. Gap junctions and cancer: communicating for 50 years. Nat Rev Cancer. 2016;16:775–788. doi: 10.1038/nrc.2016.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Ahuja D, Sáenz-Robles MT, Pipas JM. SV40 large T antigen targets multiple cellular pathways to elicit cellular transformation. Oncogene. 2005;24:7729–7745. doi: 10.1038/sj.onc.1209046. [DOI] [PubMed] [Google Scholar]
  3. Al Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Armstrong L, Tilgner K, Saretzki G, Atkinson SP, Stojkovic M, Moreno R, Przyborski S, Lako M. Human induced pluripotent stem cell lines show stress defense mechanisms and mitochondrial regulation similar to those of human embryonic stem cells. Stem Cells. 2010;28:661–673. doi: 10.1002/stem.307. [DOI] [PubMed] [Google Scholar]
  5. Atena M, Reza AM, Meehran G. A review on the biology of cancer stem cells. Stem Cell Discov. 2014;4:83–89. doi: 10.4236/scd.2014.44009. [DOI] [Google Scholar]
  6. Barker N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat Rev/Mol Cell Biol. 2014;15:19–33.02. doi: 10.1038/nrm3721. [DOI] [PubMed] [Google Scholar]
  7. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, Clevers H. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003–1007. doi: 10.1038/nature06196. [DOI] [PubMed] [Google Scholar]
  8. Barker N, Ridgway RA, van Es JH, van de Wetering M, Begthel H, van den Born M, Danenberg E, Clarke AR, Sansom OJ, Clevers H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009;457:608–611. doi: 10.1038/nature07602. [DOI] [PubMed] [Google Scholar]
  9. Berenblum PM. A speculative review: the probable nature of promoting action and its significance in the understanding of the mechanisms of carcinogenesis. Cancer Res. 1954;14:471–477. [PubMed] [Google Scholar]
  10. Bongso A, Fong CY, Ng SC, Ratnam S. Isolation and culture of inner cell mass cells from human blastocyts. Hum Reprod. 1994;9:2110–2117. doi: 10.1093/oxfordjournals.humrep.a138401. [DOI] [PubMed] [Google Scholar]
  11. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730–737. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
  12. Brash DE, Rudolph JA, Simon JA, Lin A, McKenna GJ, Baden HP, Halperin AJ, Pontén J. A role for sunlight in skin cancer: UV-induced p53 mutations in squamous cell carcinomas. Proc Natl Acad Sci U S A. 1991;88(22):10124–11018. doi: 10.1073/pnas.88.22.10124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brigelius-Flohé R, Flohé L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid Redox Signal. 2011;15:2335–2381. doi: 10.1089/ars.2010.3534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brosnan CF, Scemes E, Spray DC. Cytokine regulation of gap junction connectivity: an open-and-shut case or changing partners at the Nnxus? Am J Pathol. 2001;158(5):1565–1569. doi: 10.1016/S0002-9440(10)64110-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bufill JA. Colorectal cancer: evidence for distinct genetic categories based on proximal or distal tumor location. Ann Intern Med. 1990;113(10):779–788. doi: 10.7326/0003-4819-113-10-779. [DOI] [PubMed] [Google Scholar]
  16. Chang CC, Trosko JE, El-Fouly MH, Gibson-D’Ambrosio R, D’Ambrosio SM. Contact insensitivity of a subpopulation of normal human fetal kidney epithelial cells and of human carcinoma cell lines. Cancer Res. 1987;47:1634–1645. [PubMed] [Google Scholar]
  17. Chen CT, Shih YR, Kuo TK, Lee OK, Wei YH. Coordinated changes of mitochondrial biogenesis and antioxidant enzymes during osteogenic differentiation of human mesenchymal stem cells. Stem Cells. 2008;26:960–968. doi: 10.1634/stemcells.2007-0509. [DOI] [PubMed] [Google Scholar]
  18. Cleaver JE. Xeroderma pigmentosum: genetic and environmental influences in –skin carcinogenesis. J Dermatol. 1978;17(6):435–444. doi: 10.1111/j.1365-4362.1978.tb06178.x. [DOI] [PubMed] [Google Scholar]
  19. Cleaver JE, Trosko JE. Absence of excision of ultraviolet-induced cyclobutane dimers in xeroderma pigmentosum. Photochem Photobiol. 1970;11(6):547–550. doi: 10.1111/j.1751-1097.1970.tb06025.x. [DOI] [PubMed] [Google Scholar]
  20. Cozzio A, Passegue E, Ayton PM, Karsunky H, Cleary ML, Weissman IL. Similar MLL associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003;17:3029–3035. doi: 10.1101/gad.1143403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Crosnier C, Stamataki D, Lewis J. Organizing cell renewal in the intestine: stem cell, signal and combinatorial control. Nat Rev/Genet. 2006;7:349–359. doi: 10.1038/nrg1840. [DOI] [PubMed] [Google Scholar]
  22. Cruciani V, Mikalsen SO. The vertebrate connexin family. Cell Mol Life Sci. 2006;63:1125–1140. doi: 10.1007/s00018-005-5571-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Csete M. Oxygen in the cultivation of stem cells. Ann N Y Acad Sci. 2005;1049:1–8. doi: 10.1196/annals.1334.001. [DOI] [PubMed] [Google Scholar]
  24. de Jong J, Stoop H, Dohle GR, Bangma CH, Kliffen M, van Esser JW, van den Bent M, Kros JM, Oosterhuis JW, Looijenga LH. Diagnostic value of OCT3/4 for pre-invasive an invasive testicular germ cell tumours. J Pathol. 2005;206:242–249. doi: 10.1002/path.1766. [DOI] [PubMed] [Google Scholar]
  25. DiPaolo JA. Relative difficulties in transforming human and animal cells in vitro. J Natl Cancer Inst. 1983;70(1):3–8. [PubMed] [Google Scholar]
  26. Dougal K, Harris PA, Edwards A, Pachebat JA, Blackmore TM, Worgan HJ, Newbold CJ. A comparison of the microbiome and the metabolome4 of different regions of the equine hindgut. FEMS Microbiol Ecol. 2012;82:642–652. doi: 10.1111/j.1574-6941.2012.01441.x. [DOI] [PubMed] [Google Scholar]
  27. Eagle H. Growth inhibitor effects of cellular interactions. Isr J Med Sci. 1965;1:1220–1228. [Google Scholar]
  28. Fialkow PJ. Clonal origin of human tumors. Annu Rev Med. 1976;30:135–176. doi: 10.1146/annurev.me.30.020179.001031. [DOI] [PubMed] [Google Scholar]
  29. Flemer B, Lynch DB, Brown JM, Jeffery IB, Ryan FJ, Claesson MJ, O'Riordain M, Shanahan F, O'Toole PW (2016). Tumour-associated and non-tumour associated microbiota in colorectal cancer. Gut. pii: gutjnl-2015-309595. doi: 10.1136/gutjnl-2015-309595. [DOI] [PMC free article] [PubMed]
  30. Gagniere J, Raisch J, Veziant J, Barnich N, Bonnet R, Buc E, et al. Gut microbiota imbalance and colorectal cancer. World J Gastroenterol. 2016;22(2):501–518. doi: 10.3748/wjg.v22.i2.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Galli R, Binda E, Orfanelli U, Cipelletti B, Gritti A, De Vitis S, Fiocco R, Foroni C, Dimeco F, Vescovi A. Isolation and characterization of tumorigenic, stem-cell neural precursors from human glioblastoma. Cancer Res. 2004;64:7011–7021. doi: 10.1158/0008-5472.CAN-04-1364. [DOI] [PubMed] [Google Scholar]
  32. Gervaz P, Bucher P, Morel P. Two colons-two cancers: paradigm shift and clinical implications. J Surg Oncol. 2004;88(4):261–266. doi: 10.1002/jso.20156. [DOI] [PubMed] [Google Scholar]
  33. Glover TW, Chang CC, Trosko JE, Li SSL. Ultraviolet light induction of diphtheria toxin resistant mutations in normal and xeroderma pigmentosum human fibroblasts. Proc Natl Acad Sci. 1979;76(3982):3986. doi: 10.1073/pnas.76.8.3982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Gu G, Yuan J, Wills M, Kasper S. Prostate cancer cells with stem cell characteristics reconstitute the original human tumor in vivo. Cancer Res. 2007;67:4807–4815. doi: 10.1158/0008-5472.CAN-06-4608. [DOI] [PubMed] [Google Scholar]
  35. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/S0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  36. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  37. Hayflick L. The limited in vitro lifespan of human diploid cell strains. Exp Cell Res. 1965;37:614–636.815. doi: 10.1016/0014-4827(65)90211-9. [DOI] [PubMed] [Google Scholar]
  38. Heinemann V, Modest DP, von Weikensthal LF, Decker T, Kiani A, Veiling-Kaiser U, et al (2014). Gender and tumor location as predictors for efficacy: Influence on endpoints in firstline treatment with FOLFIRI in combination with cetuximab or bevacizumab in the AIO KRK 0306 (FIRE3) trial. J Clin Oncol 32(suppl; abstr 3600)
  39. Hemmati HD, Nakano I, Lazareff JA, Masterman-Smith M, Geschwind DH, Bronner-Fraser M, Kornblum HI. Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci U S A. 2003;100:15178–15183. doi: 10.1073/pnas.2036535100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hill MA (2016) Embryology gastrointestinal tract development
  41. Hope KJ, Jin L, Dick JE. Human acute myeloid leukemia stem cells. Arch Med Res. 2003;34:507–514. doi: 10.1016/j.arcmed.2003.08.007. [DOI] [PubMed] [Google Scholar]
  42. Horn T, Hilbrant M, Panfilio KA. Evolution of epithelial morphogenesis: phenotypic integration across multiple levels of biological organization. Front Genet. 2015 doi: 10.3389/fgene.2015.00303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Houts AC OS, Sommer N, Walker MS (2016) Treatment patterns and outcomes in patients with KRAS wild type (WT) metastatic colorectal cancer (mCRC) treated in first line with bevacizumab (B) or cetuximab (C) containing regimens. J Clin Oncol 34 [DOI] [PubMed]
  44. Huels DJ, Sansom OJ. Stem versus non-stem cell origin of colorectal cancers. Brit J Cancer. 2015;113(1):1–5. doi: 10.1038/bjc.2015.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Iacopetta B. Are there two sides to colorectal cancer? Int J Cancer. 2002;101(5):403–408. doi: 10.1002/ijc.10635. [DOI] [PubMed] [Google Scholar]
  46. Jiang JX, Penuela S (2016) Connexin and pannexin channels in cancer. BMC Cell Biol 17(Suppl 1):512. doi:10.1186/s12860-016-0094-8 [DOI] [PMC free article] [PubMed]
  47. Kang S, Trosko JE. Stem cells in toxicology: fundamental biology and practical considerations. Toxicol Sci. 2011;120(Suppl 1):S269–S289. doi: 10.1093/toxsci/kfq370. [DOI] [PubMed] [Google Scholar]
  48. Kao CY, Nomata K, Oakley CS, Welsch CW, Chang CC. Two types of normal human breast epithelial cells derived from reduction mammoplasty: phenotypic characterization and response to SV40 transfection. Carcinogenesis. 1995;16:531–538. doi: 10.1093/carcin/16.3.531. [DOI] [PubMed] [Google Scholar]
  49. Kemper K, Prasetyanti PR, De Lau W, Rodermond H, Clevers H, Medema JP. Monoclonal antibodies against Lgr5 identify human colorectal cancer stem cells. Stem Cells. 2012;30:2378–2386. doi: 10.1002/stem.1233. [DOI] [PubMed] [Google Scholar]
  50. Klaunig J, Ruch RJ. Strain and species effects on the inhibition of hepatocyte intercellular communication by liver tumor promoters. Cancer Lett. 1987;36:161–168. doi: 10.1016/0304-3835(87)90087-5. [DOI] [PubMed] [Google Scholar]
  51. Kondo T, Setoguchi T, Taga T. Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A. 2004;101:781–786. doi: 10.1073/pnas.0307618100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, Levine JE, Wang J, Hahn WC, Gilliland DG, Golub TR, Armstrong SA. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature. 2006;442:818–822. doi: 10.1038/nature04980. [DOI] [PubMed] [Google Scholar]
  53. Kuroki T, Huh N-H. Why are human cells resistant to malignant cell transformation in vitro? Jpn J Cancer Res. 1993;84:1091–1100. doi: 10.1111/j.1349-7006.1993.tb02806.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. 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]
  55. Lane SW, Williams DA, Watt FM. Modulating the stem cell niche for tissue regeneration. Nat Biotechnol. 2014;32:795–803. doi: 10.1038/nbt.2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Lee GH. Paradoxical effects of phenobarbital on mouse hepatocarcinogenesis. Toxicol Pathol. 2000;28:215–225. doi: 10.1177/019262330002800201. [DOI] [PubMed] [Google Scholar]
  57. Leithe E, Sirnes S, Omori Y, Rivedal E. Downregulation of gap junctions in +cancer cells. Crit Rev Oncog. 2006;12(3–4):225–256. doi: 10.1615/CritRevOncog.v12.i3-4.30. [DOI] [PubMed] [Google Scholar]
  58. Lenz H, Lee F, Yau L, Koh H, Knost K, Mitchell E, et al. (2016). MAVERICC, a phase 2 study of mFOLFOX6bevacizumab (BV) vs FOLFIRIBV with biomarker stratification as firstline (1 L) chemotherapy (CT) in patients (pts) with metastatic colorectal cancer (mCRC). J Clin Oncol; 34(suppl 4S; abstr 493)
  59. Leone A, Longo C, Trosko JE. The chemopreventive role of dietary phytochemicals through gap junctional intercellular communication. Phytochem Rev. 2012 [Google Scholar]
  60. Li S, Yurchenco PD. Matrix assembly, cell polarization, and cell survival: analysis of peri-implantation development with cultured embryonic stem cells. Methods Mol Biol. 2006;32:113–125. doi: 10.1385/1-59745-037-5:113. [DOI] [PubMed] [Google Scholar]
  61. Locke M, Heywood M, Fawell S, Mackenzie IC. Retention of intrinsic stem cell hierarchies in carcinoma-derived cell lines. Cancer Res. 2005;65:8944–8950. doi: 10.1158/0008-5472.CAN-05-0931. [DOI] [PubMed] [Google Scholar]
  62. Loewenstein WR, Kanno Y. Intercellular communication and the control of tissue growth: lack of communication between cancer cells. Nature. 1966;209:1248–1249. doi: 10.1038/2091248a0. [DOI] [PubMed] [Google Scholar]
  63. Loupakis F, Yang D, Yau L, Feng S, Cremolini C, Zhang W, Maus MK, Antoniotti C, Langer C, Scherer SJ, Müller T, Hurwitz HI, Saltz L, Falcone A, Lenz HJ (2015a) Primary tumor location as a prognostic factor in metastatic colorectal cancer. J Natl Cancer Inst 107(3). doi:10.1093/jnci/dju427 [DOI] [PMC free article] [PubMed]
  64. Loupakis F, Yang D, Yau L, Feng S, Cremolini C, Zhang W et al (2015b) Primary tumor location as a prognostic factor in metastatic colorectal cancer. J Natl Cancer Inst 107(3) [DOI] [PMC free article] [PubMed]
  65. Maher VM, McCormick JJ. Effect of DNA repair on the cytotoxicity and mutagenicity of UV irradiation and of chemical carcinogens in normal and xeroderma pigmentosum cells. In: Yuhas JM, Tennant RW, Regan JD, editors. Biology of radiation carcinogenesis. New York: Raven Press; 1976. pp. 129–145. [Google Scholar]
  66. Markert CL. Neoplasia: a disease of cell differentiation. Cancer Res. 1968;28:1908–1914. [PubMed] [Google Scholar]
  67. Martin FJ, Prince AS. TLR2 regulates gap junction intercellular communication in airway cells. J Imm Biol Reprod. 1982;27(4):967–975. doi: 10.1095/biolreprod27.4.967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Mentink R, Tsiantis M. From limbs to leaves: common themes in evolutionary diversification of organ form. Front Genet. 2015;6:284. doi: 10.3389/fgene.2015.00284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Missiaglia E, Jacobs B, D'Ario G, Di Narzo AF, Soneson C, Budinska E, Popovici V, Vecchione L, Gerster S, Yan P, Roth AD, Klingbiel D, Bosman FT, Delorenzi M, Tejpar S. Distal and proximal colon cancers differ in terms of molecular, pathological, and clinical features. Ann Oncol. 2014;25:1995–2001. doi: 10.1093/annonc/mdu275. [DOI] [PubMed] [Google Scholar]
  70. Mohyeldin A, Garzón-Muvdi T, Quiñones-Hinojosa A. Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell. 2010;7(150):161. doi: 10.1016/j.stem.2010.07.007. [DOI] [PubMed] [Google Scholar]
  71. Nava GM, Carbonero F, Croix JA, Greenberg E, Gaskins HR. Abundance and diversity of muscosa-associated hydrogenotropic microbes in the healthy human colon. The ISME J. 2012;6:57–70. doi: 10.1038/ismej.2011.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Neste C, Pasquali L, Vaglini F, Siciliano G, Murri L. The role of mitochondria in stem cell biology. Biosci Rep. 2007;27:165–171. doi: 10.1007/s10540-007-9044-1. [DOI] [PubMed] [Google Scholar]
  73. Nitsche U, Stogbauer F, Spath C, Haller B, Wilhelm D, Friess H, et al. Right sided colon cancer as a distinct Histopathological subtype with reduced prognosis. Dig Surg. 2016;33(2):157–163. doi: 10.1159/000443644. [DOI] [PubMed] [Google Scholar]
  74. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immune-deficient mice. Nature. 2007;445:106–110. doi: 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
  75. Ogawa T, Hayashi T, Tokunou M, Nakachi K, Trosko JE, Chang CC, Yorioka N. Suberoylanilide hydroxamic acid enhances gap junctional intercellular communication on via acetylation of histone containing connexin43 gene locus. Cancer Res. 2005;65:9771–9778. doi: 10.1158/0008-5472.CAN-05-0227. [DOI] [PubMed] [Google Scholar]
  76. Pervaiz S, Taneja R, Ghaffari S. Oxidative stress regulation of stem and progenitor cells. Antioxid Redox Signal. 2009;11:2777–2789. doi: 10.1089/ars.2009.2804. [DOI] [PubMed] [Google Scholar]
  77. Pierce GB. Neoplasms, differentiation and mutations. Am J Pathol. 1974;77:103–118. [PMC free article] [PubMed] [Google Scholar]
  78. Pitot HC, Dragon Y. Facts and theories concerning the mechanism of carcinogenesis. FASEB J. 1991;5:2280–2286. [PubMed] [Google Scholar]
  79. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pierotti MA, Daidone MG. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer Res. 2005;65:5506–5511. doi: 10.1158/0008-5472.CAN-05-0626. [DOI] [PubMed] [Google Scholar]
  80. Potten CS, Gandara R, Mahida YR, Loeffler M, Wright NA. The stem cells of small intestinal crypts: where are they? Cell Prolif. 2009;42:731–750. doi: 10.1111/j.1365-2184.2009.00642.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Potter VR. Phenotypic diversity in experimental hepatomas: the concept of partially blocked ontogeny. Br J Cancer. 1978;38:1–23. doi: 10.1038/bjc.1978.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Price TJ, Beeke C, Ullah S, Padbury R, Maddern G, Roder D, et al. Does the primary site of colorectal cancer impact outcomes for patients with metastatic disease? Cancer. 2015;9121(6):830–835. doi: 10.1002/cncr.29129. [DOI] [PubMed] [Google Scholar]
  83. Prigione A, Fauler B, Lurz R, Lehrach H, Adjaye J. The senescence related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells. Stem Cells. 2010;28:721–733. doi: 10.1002/stem.404. [DOI] [PubMed] [Google Scholar]
  84. Puri CP, Garfield RE. Changes in hormone levels and gap junctions in the rat uterus during pregnancy and parturition. Biol Reprod. 1982;27(4):967–7180. doi: 10.1095/biolreprod27.4.967. [DOI] [PubMed] [Google Scholar]
  85. Rhim JS. Neoplastic transformation of human cells in vitro. Crit Rev Oncog. 1993;4:312–335. [PubMed] [Google Scholar]
  86. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111–115. doi: 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
  87. Ruch RJ, Trosko JE, Madhukar BV. Inhibition of connexin43 gap junctional intercellular communication by TPA requires ERK activation. J Cell Biochem. 2001;83:163–169. doi: 10.1002/jcb.1227. [DOI] [PubMed] [Google Scholar]
  88. Sadler TW (2014) Langman's medical embryology. Wolters Kluwer, 12th edition, Philadelphia
  89. Saez JC. Proceedings of the international gap. BMC Cell Biol. 2016;17(Suppl 1):18. doi: 10.1186/s12860-016-0097-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Sanchez A, Hannezo E, Larsimont J-C, Liagre M, Youssef KK, Simons BD, Blanpain C. Defining the clonal dynamics leading to mouse skin tumor initiation. Nature. 2016;536:298–303. doi: 10.1038/nature19069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Sato T, Clevers H. Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications. Science. 2013;340:1190–1194. doi: 10.1126/science.1234852. [DOI] [PubMed] [Google Scholar]
  92. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH. Identification of cells initiating human melanomas. Nature. 2008;451:345–349. doi: 10.1038/nature06489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Sell S. Cellular origin of cancer: differentiation of stem cell maturation arrest? Environ Health Perspect. 1993;101:15–26. doi: 10.1289/ehp.93101s515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Shamblott MJ, Axelman J, Wang S, Bugg EM, Littlefield JW, Donovan PJ, Blumenthal PD, Huggins GR, Gearhart JD. Derivation of pluripotent stem cells from cultured human primordial germ cells. PNAS, USA. 1998;95:13726–13731. doi: 10.1073/pnas.95.23.13726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821–5828. [PubMed] [Google Scholar]
  96. Sinicrope FA, Mahoney MR, Yoon HH, Smyrk TC, Thibodeau SN, Goldberg RM, et al. Analysis of molecular markers by anatomic tumor site in stage III colon carcinomas from adjuvant chemotherapy trial NCCTG N0147 (alliance) Clin Cancer Res. 2015;21(23):5294–5304. doi: 10.1158/1078-0432.CCR-15-0527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Tai M-H, Chang CC, Kiupel M, Webster JD, Olson LK, Trosko JE. Oct-4 expression in adult stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis. 2005;26:495–502. doi: 10.1093/carcin/bgh321. [DOI] [PubMed] [Google Scholar]
  98. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  99. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocyts. Science. 1998;282:1145–1147. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  100. Till JE. Stem cells in differentiation and neoplasia. J Cell Physiol Suppl. 1982;1:3–11. doi: 10.1002/jcp.1041130405. [DOI] [PubMed] [Google Scholar]
  101. Trosko JE. Is the concept of ‘tumor promotion’ a useful paradigm? Mol Carcinog. 2001;30:131–137. doi: 10.1002/mc.1021. [DOI] [PubMed] [Google Scholar]
  102. Trosko JE. The role of stem cells and gap junctional intercellular communication in carcinogenesis. J Biochem Mol Biol. 2003;36:43–48. doi: 10.5483/bmbrep.2003.36.1.043. [DOI] [PubMed] [Google Scholar]
  103. Trosko JE. From adult stem cells to cancer stem cells: Oct-4 gene, cell-cell communication, and hormones during tumor promotion. Ann NY Acad Sci. 2006;1089:36–58. doi: 10.1196/annals.1386.018. [DOI] [PubMed] [Google Scholar]
  104. Trosko JE. Gap junction intercellular communication as a ‘biological Rosetta stone’ in understanding, in a systems manner, stem cell behavior, mechanisms of epigenetic toxicology, chemoprevention and chemotherapy. J Membr Biol. 2007;218:93–100. doi: 10.1007/s00232-007-9072-6. [DOI] [PubMed] [Google Scholar]
  105. Trosko JEA. Commentary: re-programming or selecting adult stem cells. Stem Cell Rev. 2008;4:81–88. doi: 10.1007/s12015-008-9017-1. [DOI] [PubMed] [Google Scholar]
  106. Trosko J. Human adult stem cells as the target cells for the initiation of carcinogenesis and for the generation of “cancer stem cells”. Inter J Stem Cells. 2008;1:8–26. doi: 10.15283/ijsc.2008.1.1.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Trosko JE. Cancer stem cells and cancer non-stem cells: from adult stem cells or from re-programming of differentiated somatic cells. Vet Pathol. 2009;46:176–193. doi: 10.1354/vp.46-2-176. [DOI] [PubMed] [Google Scholar]
  108. Trosko JE. Induction of iPS cells and of cancer stem cells: the stem cell or reprogramming hypothesis of cancer? Anat Rec (Hoboken) 2014;297:161–173. doi: 10.1002/ar.22793. [DOI] [PubMed] [Google Scholar]
  109. Trosko JE (2016a). A Conceptual integration of extra-, intra- and gap junctional-intercellular communication in the evolution of multi-cellularity and stem cells: how disrupted cell-cell communication during development can affect diseases later in life. Int J Stem Cell Res Ther 2016, 3:021, ISSN: 2469-570X
  110. Trosko JE (2016b) Evolution of microbial quorum sensing to human global quorum sensing: an insight to how gap junctional intercellular communication might be linked to the global metabolic disease crisis. Biology (Basel).15;5(2). doi: 10.3390/biology5020029 [DOI] [PMC free article] [PubMed]
  111. Trosko JE, Chang CC (1988) Nongenotoxic mechanisms in carcinogenesis: role of inhibited intercellular communication. In: Hart RW and Hoerger FD (eds.) Banbury Report 31: carcinogen risk assessment: new directions in the qualitative and quantitative aspects. Cold Spring Harbor, N.Y.: Cold Spring Harbor Laboratory. Pp.139–170
  112. Trosko JE, Kang KS. Evolution of energy metabolism, stem cells and cancer stem cells: how the Warburg and barker hypotheses might be linked. Int J Stem Cells. 2012;5:39–56. doi: 10.15283/ijsc.2012.5.1.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Trosko JE, Ruch RJ. Cell-cell communication in carcinogenesis. Front Biosci. 1998;3:208–236. doi: 10.2741/A275. [DOI] [PubMed] [Google Scholar]
  114. Trosko JE, Ruch R. Gap junctions as targets for cancer chemoprevention and chemotherapy. Curr Drug Targets. 2002;203:465–482. doi: 10.2174/1389450023347371. [DOI] [PubMed] [Google Scholar]
  115. Trosko JE, Tai MH (2006) Adult stem cell theory of the multi-stage, multi-mechanism theory of carcinogenesis: role of inflammation on the promotion of initiated cells. In Infections and Inflammation: Impacts on Oncogenesis; Dittmar, T., Zaenker, K.S., Schmidt, A., eds. Karger Publishers, Contributions to Microbiology: Basel, Switzerland, pp.45–65 [DOI] [PubMed]
  116. Trosko JE, Chang CC, Madhukar BV, Oh SY. Modulators of gap junction function: the scientific basis of epigenetic toxicology. In Vitro Toxicology. 1990;3:9–26. [Google Scholar]
  117. Trosko JE, Chang CC, Upham BL, Wilson MR. Epigenetic toxicology as toxicant-induced changes in intracellular signaling leading to altered gap junctional intercellular communication. Toxicol Lett. 1998;102-103:71–78. doi: 10.1016/S0378-4274(98)00288-4. [DOI] [PubMed] [Google Scholar]
  118. Upham BL, Trosko JE. Carcinogenic tumor promotion, induced oxidative stress signaling, modulated gap junction function and altered gene expression. Antioxid Redox. 2009;11:297–308. doi: 10.1089/ars.2008.2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Venook A, Niedzwiecki D, Innocenti F, Fruth B, Greene C, O'Neil B et al. (2016) Impact of primary (1°) tumor location on overall survival (OS) and progressionfree survival (PFS) in patients (pts) with metastatic colorectal cancer (mCRC): Analysis of CALGB/SWOG 80405 (Alliance). J Clin Oncol 36(suppl; abstr 3504)
  120. Wang X, Kruithof-de Julio M, Economides KD, Walker D, Yu H, Halili MV, Hu YP, Price SM, Abate-Shen C, Shen MM. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature. 2009;461:495–500. doi: 10.1038/nature08361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Warburg O. On respiratory impairment in cancer cells. Science. 1956;124:269–270. [PubMed] [Google Scholar]
  122. Warren S, Schultz RA, Chang CC, Wade MH, Trosko JE. Elevated spontaneous mutation rate in bloom syndrome fibroblasts. Proc Natl Acad Sci USA. 1981;78:3133–3137. doi: 10.1073/pnas.78.5.3133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Weinstein IB, Gattoni CS, Kirschmeier P, Lambert M, Hsiao W, Backer J, Jeffrey A. Multistage carcinogenesis involves multiple genes and multiple mechanisms. J Cell Physiol. 1984;3:127–137. doi: 10.1002/jcp.1041210416. [DOI] [PubMed] [Google Scholar]
  124. Weissman IL. Stem cells are units of natural selection for tissue formation, for germline development and in cancer development. Proc Natl Acad Sci USA. 2015;112:8922–8928. doi: 10.1073/pnas.1505464112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Wilson MR, Close TW, Trosko JE. Cell population dynamics (apoptosis, mitosis and cell-cell communication) during disruption of homeostasis. Exp Cell Res. 2000;254:257–268. doi: 10.1006/excr.1999.4771. [DOI] [PubMed] [Google Scholar]
  126. Wong HL, Lee B, Field K, Lomax A, Tacey M, Shapiro J, et al. Impact of primary tumor site on bevacizumab efficacy in metastatic colorectal cancer. Clin Colorectal Cancer. 2016;15(2):e9–e15. doi: 10.1016/j.clcc.2016.02.007. [DOI] [PubMed] [Google Scholar]
  127. Wu C, Wei Q, Utomo V, Nadesan P, Whetstone H, Kandel R, Wunder JS, Alman BA. Side population cells isolated from mesenchymal neoplasms have tumor initiating potential. Cancer Res. 2007;67:8216–8222. doi: 10.1158/0008-5472.CAN-07-0999. [DOI] [PubMed] [Google Scholar]
  128. Yamagiwa K, Ichikawa K. Experimental study of the pathogenesis of carcinoma. CA Cancer J Clin. 1977;27:174–181. doi: 10.3322/canjclin.27.3.174. [DOI] [PubMed] [Google Scholar]
  129. Yamasaki H, Naus CCG. Role of connexin genes in growth control. Carcinogenesis. 1996;17:1199–1213. doi: 10.1093/carcin/17.6.1199. [DOI] [PubMed] [Google Scholar]
  130. Yamasaki H, Hollstein M, Mesnil M, Martel AAM. Selectivel lack of intercellular communication between transformed and nontransformed cells as common property of chemical and oncogene transformation of BALB/c3T3 cells. Cancer Res. 1987;47:5658–5664. [PubMed] [Google Scholar]
  131. Yamauchi M, Morikawa T, Kuchiba A, Imamura Y, Qian ZR, Nishihara R, et al. Assessment of colorectal cancer molecular features along bowel subsites challenges the conception of distinct dichotomy of proximal versus distal colorectum. Gut. 2012;61(6):847–854. doi: 10.1136/gutjnl-2011-300865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Yotti LP, Trosko JE, Chang CC. Elimination of metabolic cooperation in Chinese hamster cells by a tumor promoter. Science. 1979;206(1089):1091. doi: 10.1126/science.493994. [DOI] [PubMed] [Google Scholar]
  133. Yuan X, Curtin J, Xiong Y, Liu G, Waschsmann-Hogiu S, Farkas DL, Black KL, Yu JS. Isolation of cancer stem cells from adult glioblastoma multiforme. Oncogene. 2004;23:9392–9400. doi: 10.1038/sj.onc.1208311. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Cell Communication and Signaling are provided here courtesy of The International CCN Society

RESOURCES