Review
Mechanism of colorectal carcinogenesis triggered by heme iron from red meat

https://doi.org/10.1016/j.bbcan.2019.188334Get rights and content

Abstract

Colorectal cancer (CRC) is one of the major tumor entities worldwide, with an increasing incidence in younger people. CRC formation is causally linked to various genetic, life-style and dietary risk factors. Among the ladder, the consumption of red meat has emerged as important risk factor contributing to CRC. A large body of evidence shows that heme iron is the critical component of red meat, which promotes colorectal carcinogenesis.

In this review, we describe the uptake and cellular fate of both heme and inorganic iron in intestinal epithelial cells. Next, an overview on the DNA damaging properties of heme iron is provided, highlighting the DNA adducts relevant for CRC etiology. Moreover, heme triggered mechanisms leading to colonic hyperproliferation are presented, which are intimately linked to changes in the intestinal microbiota induced by heme. A special focus was set on the impact of heme iron on innate and adaptive immune cells, which could be relevant in the context of CRC. Finally, we recapitulate in vivo studies providing evidence for the tumor-promoting potential of dietary heme iron. Altogether, heme iron affects numerous key pathways involved in the pathogenesis of CRC.

Introduction

Colorectal cancer (CRC) ranks among the most frequent tumor entities worldwide with a global burden of around 1.8 Mio. people newly diagnosed for CRC every year [1]. Furthermore, an increasing incidence of CRC has recently been observed in adults under the age of fifty in both the US and Europe [2,3], highlighting the need to better understand the mechanism driving CRC formation in order to improve primary prevention. CRC is a multifactorial disease, which develops in the majority of cases sporadically due to acquired somatic mutations and aberrant epigenetic alterations in critical genes [1]. CRC is causally linked to life-style factors (e.g. alcohol, tobacco, physical inactivity) and dietary habits (e.g. red and processed meat intake, low dietary fiber) [1,4]. Epidemiological studies showed a clear association between meat consumption and CRC development [5,6]. In 2015, the International Agency for Research on Cancer classified the consumption of processed meat as carcinogenic and red meat as probably carcinogenic in humans [7]. A large prospective cohort study revealed that the intake of red meat also correlates with an increased risk for overall cancers, particularly breast cancer [8]. Red meat, e.g. beef and lamb, is characterized by its high myoglobin content and contains higher levels of heme iron as compared to white meat, such as chicken [9]. Interestingly, meta-analysis of epidemiological studies showed that dietary heme significantly increases the risk for CRC [10]. A recent prospective cohort study further supported this correlation and displayed a dose-effect relationship between heme iron uptake and the risk to develop colorectal adenoma [11]. A body of evidence suggests that heme iron is the critical component of red meat, which promotes colorectal carcinogenesis. In this review, we will provide a comprehensive overview on the underlying mechanisms known so far, involving, amongst others, genotoxic effects, hyperproliferation of the gut epithelium and changes in the intestinal microbiota. We will further highlight the impact of heme iron on immune cells and its possible link to CRC formation. It should be mentioned that also other red meat-derived agents might contribute to the elevated CRC risk. This comprises N-glycolylneuraminic acid (Neu5Gc), which is a sialic acid not occurring in humans. Neu5Gc was reported to cause systemic inflammation and to promote the progression of hepatocellular carcinoma in mice [12]. Furthermore, latent infections with the so-called bovine meat and milk factors (BMMF) and associated inflammatory processes might play a role in CRC etiology [13], particularly in the presence of food-borne carcinogens like N-nitroso compounds or heterocyclic aromatic amines [14,15].

Section snippets

Structure of heme

Heme is the prosthetic group of various proteins including the oxygen transporter proteins hemoglobin and myoglobin [16]. Hemoglobin consists of four subunits, two α- and two β-chains, which bind up to four oxygen molecules in the lung and transport them through the bloodstream to the tissue (Fig. 1). Due to the Bohr effect, the oxygen is transferred to the monomeric myoglobin in tissue consisting of only one β-chain [17]. The myoglobin then distributes the oxygen in the muscle, where it is

Gastrointestinal resorption of heme iron and inorganic iron

Iron is an essential micronutrient found in two dietary forms, heme iron and non-heme iron or inorganic iron. The latter occurs in vegetables and cereals, while the main nutritional source of heme iron represents animal tissue. Interestingly, heme iron has a greater bioavailability than non-heme iron [18]. Following its dietary uptake, heme is released from myoglobin and hemoglobin in the acidic environment of the stomach as well as by the proteolytic activity of different enzymes in the

Heme iron-mediated formation of genotoxic compounds and DNA damage induction

Heme iron gives rise to different DNA damaging agents, including reactive oxygen species (ROS), lipid peroxidation end-products and N-nitroso compounds (NOC).

Lipid peroxidation and cytotoxicity

As mentioned in the last chapter, dietary heme catalyzes lipid peroxidation and gives rise to reactive aldehydes, which are detected as TBARS. High levels of heme-induced TBARS in fecal water were associated with luminal injury and a high cytotoxicity if fecal water is applied to cultured colonocytes [21,36,63,103]. Further studies provided evidence that dietary heme decreases caspase-3 activity and inhibits apoptosis in the colon mucosa, which goes along with an increased proliferation and

Impact of heme on hyperproliferation

First evidence for an increased epithelial proliferation rate upon dietary heme intake was provided in a rodent feeding study by autoradiographic analysis of methyl-[3H]thymidine incorporation [21]. Interestingly, this effect could be blocked by dietary supplementation with high amounts of calcium [111]. Further short-term studies with moderate doses of heme (0.2 and 0.5 μmol/g diet) confirmed this finding, showing an increased number of Ki67-positive cells in colon crypts and an increased

Influence of heme iron on the intestinal microbiome

A growing body of evidence links the heme-induced hyperproliferation of colon epithelium to heme-dependent alterations of the gut flora. The intestinal microbiota plays a crucial role in host nutrient and drug metabolism, but also in maintaining the integrity of the gut mucosal barrier and in immunomodulation [116,117]. More than 90% of the gut microbiota is composed of four major phyla. The most prevailing phylum are gram-positive Firmicutes followed by gram-negative Bacteroidetes.

Modulation of immune cell function by heme

Dietary heme causes a microbial dysbiosis and impairs the intestinal barrier, thereby exposing the epithelium directly to enterobacteria and, thus, also bacterial lipopolysaccharides (LPS). Although one study reported no evidence for a Toll-like receptor 4 (TLR4)-triggered innate immune reaction and inflammation pathways in heme-fed mice [122], it is conceivable that bacteria reaching the epithelium will elicit an immune response and may promote intestinal inflammation. It is well-known that

Heme iron/red meat and intestinal tumorigenesis

Epidemiological studies provided evidence that the consumption of red meat and heme iron increases the risk to develop CRC (see Introduction). These data were supported by a series of experiments conducted in rodents, which were fed with a diet containing hemoglobin, hemin or other heme-rich meat sources. The first study indicating a contribution of heme iron to colon carcinogenesis was reported two decades ago [155]. The alkylating agent methylnitrosurea was administered intrarectally to rats

Concluding remarks

Several lines of evidence showed convincingly that heme iron, either free or bound as prosthetic group to hemoglobin or myoglobin, promotes CRC formation. Dietary heme exerts its pro-tumorigenic activity in the colorectum on multiple layers (Fig. 7) and thereby favors sporadic CRC formation, which is known to occur primarily via the adenoma-carcinoma sequence [1,163]. On the one hand, heme iron catalyzes the formation of genotoxic species and concomitant DNA damage induction. On the other hand,

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was supported by the German Research Foundation (DFG FA1034/3-1 and FA1034/3-3).

References (168)

  • R.R. Crichton et al.

    Molecular and cellular mechanisms of iron homeostasis and toxicity in mammalian cells

    J. Inorg. Biochem.

    (2002)
  • G.O. Latunde-Dada et al.

    Recent advances in mammalian haem transport

    Trends Biochem. Sci.

    (2006)
  • D. Campa et al.

    A gene-wide investigation on polymorphisms in the ABCG2/BRCP transporter and susceptibility to colorectal cancer

    Mutat. Res.

    (2008)
  • T.A. Rouault

    The intestinal heme transporter revealed

    Cell

    (2005)
  • K. Gkouvatsos et al.

    Regulation of iron transport and the role of transferrin

    Biochim. Biophys. Acta

    (2012)
  • S.J. Stohs et al.

    Oxidative mechanisms in the toxicity of metal ions

    Free Radic. Biol. Med.

    (1995)
  • J.P. Angeli et al.

    Lipid hydroperoxide-induced and hemoglobin-enhanced oxidative damage to colon cancer cells

    Free Radic. Biol. Med.

    (2011)
  • M.M. Gaschler et al.

    Lipid peroxidation in cell death

    Biochem. Biophys. Res. Commun.

    (2017)
  • K.A. Johnson et al.

    Repair of propanodeoxyguanosine by nucleotide excision repair in vivo and in vitro

    J. Biol. Chem.

    (1997)
  • D. Tsikas

    Assessment of lipid peroxidation by measuring malondialdehyde (MDA) and relatives in biological samples: analytical and biological challenges

    Anal. Biochem.

    (2017)
  • S.A. Bingham et al.

    Effect of white versus red meat on endogenous N-nitrosation in the human colon and further evidence of a dose response

    J. Nutr.

    (2002)
  • B. Kaina et al.

    MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents

    DNA Repair (Amst)

    (2007)
  • M. Glei et al.

    Hemoglobin and hemin induce DNA damage in human colon tumor cells HT29 clone 19A and in primary human colonocytes

    Mutat. Res.

    (2006)
  • N. Keum et al.

    Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies

    Nat. Rev. Gastroenterol. Hepatol.

    (2019)
  • F.E. Vuik et al.

    Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years

    Gut

    (2019)
  • R.L. Siegel et al.

    Colorectal cancer incidence patterns in the United States, 1974-2013

    J. Natl. Cancer Inst.

    (2017)
  • A. Chao et al.

    Meat consumption and risk of colorectal cancer

    JAMA

    (2005)
  • T. Norat et al.

    Meat, fish, and colorectal cancer risk: the European Prospective Investigation into cancer and nutrition

    J. Natl. Cancer Inst.

    (2005)
  • A. Diallo et al.

    Red and processed meat intake and cancer risk: results from the prospective NutriNet-Sante cohort study

    Int. J. Cancer

    (2018)
  • G. Lombardi-Boccia et al.

    Total heme and non-heme iron in raw and cooked meats

    J. Food Sci.

    (2002)
  • L. Qiao et al.

    Intakes of heme iron and zinc and colorectal cancer incidence: a meta-analysis of prospective studies

    Cancer Causes Control

    (2013)
  • N. Bastide et al.

    Heme iron intake, dietary antioxidant capacity, and risk of colorectal adenomas in a large cohort study of French women

    Cancer Epidemiol. Biomark. Prev.

    (2016)
  • A.N. Samraj et al.

    A red meat-derived glycan promotes inflammation and cancer progression

    Proc. Natl. Acad. Sci. U. S. A.

    (2015)
  • H. Zur Hausen et al.

    Specific nutritional infections early in life as risk factors for human colon and breast cancers several decades later

    Int. J. Cancer

    (2019)
  • M. Mimmler et al.

    DNA damage response curtails detrimental replication stress and chromosomal instability induced by the dietary carcinogen PhIP

    Nucleic Acids Res.

    (2016)
  • C.J. Reedy et al.

    Heme protein assemblies

    Chem. Rev.

    (2004)
  • C. Valenzuela et al.

    Total iron and heme iron content and their distribution in beef meat and viscera

    Biol. Trace Elem. Res.

    (2009)
  • J. Hooda et al.

    Heme, an essential nutrient from dietary proteins, critically impacts diverse physiological and pathological processes

    Nutrients

    (2014)
  • G.P. Young et al.

    Haem in the gut. I. Fate of haemoproteins and the absorption of haem

    J. Gastroenterol. Hepatol.

    (1989)
  • A.L. Sesink et al.

    Red meat and colon cancer: the cytotoxic and hyperproliferative effects of dietary heme

    Cancer Res.

    (1999)
  • R. Grasbeck et al.

    An intestinal receptor for heme

    Scand. J. Haematol.

    (1979)
  • Y. Gottlieb et al.

    Endoplasmic reticulum anchored heme-oxygenase 1 faces the cytosol

    Haematol. Hematol. J.

    (2012)
  • S.B. Raffin et al.

    Intestinal absorption of hemoglobin iron-heme cleavage by mucosal heme oxygenase

    J. Clin. Invest.

    (1974)
  • P.R. Montellano

    The mechanism of heme oxygenase

    Curr. Opin. Chem. Biol.

    (2000)
  • K.R. Kranc et al.

    Oxidative degradation of bilirubin produces vasoactive compounds

    Eur. J. Biochem.

    (2000)
  • E. Kuntz et al.

    Hepatology Textbook and Atlas

    (2008)
  • A.R. West et al.

    Mechanisms of heme iron absorption: current questions and controversies

    World J. Gastroenterol.

    (2008)
  • N. IJssennagger et al.

    Dietary haem stimulates epithelial cell turnover by downregulating feedback inhibitors of proliferation in murine colon

    Gut

    (2012)
  • P.S. Oates

    Heme in intestinal epithelial cell turnover, differentiation, detoxification, inflammation, carcinogenesis, absorption and motility

    World J. Gastroenterol.

    (2006)
  • I. Hlavata et al.

    The role of ABC transporters in progression and clinical outcome of colorectal cancer

    Mutagenesis

    (2012)
  • Cited by (57)

    • Association of buccal micronucleus cytome assay (BMNCyt) biomarkers with inorganic element concentration and genetic polymorphisms in welders

      2023, Environmental Toxicology and Pharmacology
      Citation Excerpt :

      Furthermore, the high Fe content of red meat can increase oxidative damage and the formation of N-nitroso compounds (Joosen et al., 2009). This effect is enhanced when red meat products have a high fat content, which has been associated with the development of breast cancer and colon cancer (Inoue-Choi et al., 2016; Seiwert et al., 2020). Age was significantly correlated with the MN and KRL biomarkers.

    • The role of diet in genotoxicity of fecal water derived from IBD patients and healthy controls

      2022, Food and Chemical Toxicology
      Citation Excerpt :

      Excessive intake of heme iron give rise to different DNA damaging agents, including reactive oxygen species, lipid peroxidation end-products and NOC. Moreover, heme iron support intestinal carcinogenesis via inducing colonic hyperproliferation and outgrowth of (pre)neoplastic cells, modulation of immune cells, and promoting gut dysbiosis (Gamage et al., 2021; Seiwert et al., 2020). The association of cancer risk with high intakes of dietary sugar has been investigated in several epidemiological studies (Debras et al., 2020; Makarem et al., 2016, 2018).

    View all citing articles on Scopus
    1

    Present address: Department of Pharmacology, University Medical Center Mainz, 55131 Mainz, Germany.

    View full text