Skip to main content

Mammalian tumor-like organs. 2. Mammalian adipose has many tumor features and obesity is a tumor-like process

Abstract

Background

In previous publications, the author developed the theory of carcino-evo-devo, which predicts that evolutionarily novel organs should recapitulate some features of tumors in their development.

Main text

Mammalian adipose is currently recognized as a multi-depot metabolic and endocrine organ consisting of several adipose tissues. Although lipid-storing cells and proteins are ancient, the adipose organ as a whole is evolutionarily novel to mammals. The adipose expansion has remarkable similarities with the growth of solid tumors. These similarities are the following: (1) The capability to unlimited expansion; (2) Reversible plasticity; (3) Induction of angiogenesis; (4) Chronic inflammation; (5) Remodeling and disfunction; (6) Systemic influence on the organism; (7) Hormone production; (8) Production of miRNAs that influence other tissues; (9) Immunosuppression; (10) DNA damage and resistance to apoptosis; (11) Destructive infiltration in other organs and tissues. These similarities include the majority of “hallmarks of cancer”. In addition, lipomas are the most frequent soft tissue tumors, and similar drugs may be used for the treatment of obesity and cancer by preventing infiltration. This raises the possibility that obesity, at least in part, may represent an oncological problem. The existing similarities between adipose and tumors suggest the possible evolutionary origin of mammalian adipose from some ancestral benign mesenchymal hereditary tumors. Indeed, using a transgenic inducible zebrafish tumor model, we described many genes, which originated in fish and were expressed in fish tumors. Their human orthologs LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA acquired functions connected with the adipose organ. They are also involved in tumor development in humans.

Conclusion

If the hypothesis of the evolutionary origin of the adipose organ from the ancestral hereditary tumor is correct, it may open new opportunities to resolve the oncological problem and the problem of the obesity epidemic. New interventions targeting LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA gene network, in addition to what already is going on, can be designed for treatment and prevention of both obesity and tumors.

Background

In previous publications, the author developed the theory of carcino-evo-devo, which describes the coevolution of normal and neoplastic development [1, 2]. I formulated the hypothesis of evolution by tumor neofunctionalization (below I will call it "the main hypothesis"), which suggested that the evolutionary role of hereditary tumors might consist in supplying evolving multicellular organisms with extra cells masses for expression of evolutionarily novel genes and the origin of new cell types, tissues, and organs [1, 2].

Several non-trivial predictions of the main hypothesis have been confirmed in my laboratory ([1,2,3,4], reviewed in [5]). One non-trivial prediction of the main hypothesis is that evolutionarily novel organs if they indeed originated from hereditary tumors or tumor-like structures, should recapitulate some features of tumors in their development. That is why in previous articles [2, 3, 5] I was looking for the data that might confirm this prediction in the literature, and also performed some experiments in my lab. The first paper in this series [5] reviewed the evidence that evolutionarily novel organs such as the placenta, mammary gland, prostate, and infantile brain indeed have many features of tumors including the regulated invasiveness at certain stages of their development and higher incidence of cancer. In that paper, I suggested calling evolutionarily new normal organs, which have many tumor features, the tumor-like organs for brevity [5].

In the present paper, the author reviews the evidence that mammalian adipose, the other evolutionarily novel organ of mammals, has many tumor features as well, and obesity is a tumor-like process. A hypothesis of the possible evolutionary origin of mammalian adipose from hereditary mesenchymal tumors is formulated and supporting data obtained in the author’s lab are discussed.

Main text

Mammalian adipose is a metabolic and endocrine organ evolutionarily novel to mammals

Adipose is a metabolic and endocrine organ operating “as a structured whole” [6, 7]. The concept of adipose as a large multi-depot organ with discrete anatomy was developed by S. Cinti [6, 8,9,10]. The adipose organ consists of white adipose tissue and brown adipose tissue distributed in a series of subcutaneous and visceral depots. Each depot of the organ has its own vascular and nerve supply. White adipose tissue (WAT) is involved in triglyceride/energy storage, and brown adipose tissue (BAT)—in energy expenditure.WAT and BAT differ in morphology and location [10]. The third type of fat, beige adipose tissue, resembles brown adipose morphologically and functionally, but its development is closer to the development of white adipose tissue [10, 11]. Some authors also consider bone marrow adipose as a separate type of adipose tissue [12].

Adipose has a mesodermal origin, but white and brown adipocytes develop from separate precursor cells, through separate differentiating lineages, and by using different differentiation factors. Beige adipocytes develop from precursors of white adipocytes [13, 14]. Adipose cells can also originate from tumor cells—trans-differentiation of breast cancer cells into functional adipocytes was reported [15].

Although the storage of energy in lipids is evolutionarily conserved, and lipid-storing cells and proteins (FIT) are ancient [16, 17], the adipose organ is evolutionarily novel to mammals [18]. BAT has not been described in fishes, amphibians, reptiles, or birds, and is present only in higher mammals [19, 20]. The overview of the evolution of adipose tissue depots shows the gradual accumulation of features such as the way of fat storage, leptin, BAT, uncoupling protein-1 (UCP-1) in BAT, and thermoregulation in mammalian evolutionary lineage [19]. Adipose organ acquired several fundamental metabolic functions since the early evolution of mammals as an adaptation to new diets and thermoregulation [18]. Adipose plays a central role in the energy metabolism and maintenance of glucose homeostasis [21]. Adipose associated with other organs has a diversity of additional functions and adaptations, and participate in morphogenetic processes [17].

Similarities of mammalian adipose to tumors

The capability of unlimited expansion

The main similarity of adipose to tumors is its capability to almost unlimited expansion. Variations in nutrition or environmental temperature cause dramatic anatomical changes in the adipose organ. In obesity, it can increase its mass tremendously. Adipose tissue expands due to hypertrophy and hyperplasia of adipocytes. The authors stress that there are remarkable similarities between adipose expansion and growth of solid tumors” [22]. The adipose expansion may lead to a pathological condition, i.e. obesity and related metabolic disorders. Obesity is a risk factor for developing lipomas and other types of tumors, not only adipose tumors.

Relatively high prevalence of mammalian adipose tumors

Adipose tumors comprise a large group of human tumors. Lipomas are the most frequent soft tissue tumors (50% of all soft-tissue masses) and are found in 2% of the population [23,24,25]. The mammary gland and prostate, the other evolutionarily novel mammalian organs, are also characterized by the highest incidence of tumors [26]. Lipomas are benign tumors, while liposarcomas are malignant adipose tumors with different degrees of malignancy. Liposarcomas are the most prevalent soft tissue malignancy [24, 2729].

The remarkable plasticity of mammalian adipose

The reversible plasticity of cancer cells is well known (reviewed in [2]). Cell plasticity is defined as “the ability of cells to change their phenotypes without genetic mutations in response to environmental cues” [30]. Neoplasms have been associated with increased plasticity, although cell plasticity was first observed during normal development.

The plasticity of the adipose is remarkable and reminds that of tumors. The adipose organ can increase in size or regress, depending on the energy balance. Earlier studies suggested adipocytes transdifferentiation during cold exposure, physical exercise, lactation, and obesity [9, 10]. At present, the appearance of beige adipocytes in WAT depots after cold exposure or stimulation (beiging or browning of WAT) is viewed as direct transdifferentiation of white adipocytes, or as differentiation from progenitor cells. The process of WAT beiging is reversible [31,32,33].

Adipose is involved in the development of the mammary gland, the other novel mammalian organ with many tumor features. Subcutaneous adipose depots participate in mammary gland formation during lactation and pregnancy [9]. Earlier data suggested that mammary adipocytes transdifferentiate into mammary epithelial cells during mammary gland development [8, 9]. The latest cell lineage tracing studies showed that white adipocytes in the mammary gland and skin can reversibly dedifferentiate into preadipocytes [33], and adipocyte progenitor cells can differentiate into epithelial cells of the mammary gland [34]. White adipocytes can reversibly transdifferentiate into myofibroblasts and cancer-associated fibroblasts during fibrosis and cancer, and dedifferentiate in liposarcomas [33]. These new data support the earlier evidence on the possibility of mammary adipocytes transdifferentiation into mammary epithelial cells [8, 9]. On the other hand, as already mentioned above, EMT-derived breast cancer cells can trans-differentiate into post-mitotic functional adipocytes [15]. Mammary adipose controls breast cancer progression: mammary preadipocytes act locally by releasing cytokines, growth factors, and extracellular matrix components [35]. Interestingly, liposarcomas are very rare in the mammary gland that is characterized by the high incidence of cancer [27, 28].

Adipose expansion induces angiogenesis

Tumor growth induces angiogenesis. Similarly, adipose expansion also induces angiogenesis. The angiogenic activity of adipocytes is connected with the secretion of pro-angiogenic molecules. The newly formed vasculature is important for adipogenesis. Angiogenesis is a rate-limiting step for adipose expansion [22, 36].

Chronic inflammation

Like solid tumors, adipose expansion is connected with hypoxia. Hypoxia is one of the factors that cause macrophage infiltration of obese adipose tissue. The other factors are adipocyte death, chemotactic regulation, and fatty acid flux [22]. Infiltrated macrophages participate in adipose tissue inflammation. Saturated fatty acids released from adipocytes are ligands for Toll-like receptor 4 complex located on macrophages. Their interaction induces inflammatory changes in macrophages, which include TNFα production. A paracrine loop involving saturated fatty acids and TNFα causes chronic inflammatory responses in adipose tissue [37]. Low-grade chronic inflammation connected with obesity is a risk factor for many cancers [38].

Similar interactions between endogenous ligands and pathogen sensors occur in tumors which are also chronic inflammatory diseases. Tumor-associated macrophages are a major type of inflammatory cells infiltrating most tumors. The recruitment of immune cells and increased expression of inflammatory mediators in tumors constitute the phenomenon of tumor-elicited inflammation. Inflammation is connected with the initiation of tumors and with different stages of tumor progression [38].

Remodeling and disfunction

Considerable changes in obese adipose tissue, including changes of extracellular matrix (ECM), adipogenesis, and metabolism, constitute adipose tissue remodeling [22, 37]. Tumors also undergo considerable dynamic changes and remodeling of chromatin [39], ECM [40, 41], vasculature [42], and metabolism [43].

Inflammation, fibrosis, and impaired angiogenesis cause disfunction of adipose organ, which leads to obesity and related metabolic complications [44, 45]. The loss of function and differentiation features is also connected with tumor development (reviewed in [2]).

DNA damage and resistance to apoptosis

Obesity-related inflammation and oxidative stress cause DNA damage in adipocytes and other tissues [46] that can lead to obesity-related carcinogenesis [47]. This suggests the similarity of DNA damage mechanisms in obesity and carcinogenesis. There are even more similarities. DNA damage in obese adipocytes activates the p53 pathway [48], as it does in tumors [49]. p53 negatively regulates both tumorigenesis and adipogenesis [50].

DNA damage is an initial stage of molecular processes that leads to genomic instability. Genomic instability is a feature of most tumors [51].

In hereditary cancers, mutations in DNA repair genes cause genomic instability [51]. CIDE proteins involved in regulating lipid metabolism belong to the family of Cell death-Inducing DNA fragmentation Factor Alpha (DFFA)-like Effector proteins. They may participate in the DNA fragmentation step in apoptosis [52, 53].

Apoptosis of adipocytes is anticipated in the stressful obese environment. However, anti-apoptotic factors such as YAP, TAZ, and Bcl2 are activated in obese adipocytes protecting them from cell death [54], a situation similar to that in tumor cells. Survivin, another potent apoptosis inhibitor, is upregulated in obesity by inflammation and oxidative stress. Survivin is also the regulator of lipid storage and metabolism. On the other hand, survivin is an oncogene expressed in most tumors. Thus, survivin is the direct molecular link between obese adipose and tumors [55].

Systemic influence in the organism

Adipose, as the central energy metabolism regulator, influences other tissues' metabolism. Adipose regulates the other tissues' metabolism according to the nutritional balance of the organism. Obesity causes systemic metabolic disorders such as insulin resistance and diabetes [7, 56, 57]. The systemic influence of tumors on the organism has also been known since long ago [58, 59].

Tumor cells have a higher rate of glucose consumption than normal cells [60,61,62]. That is why tumors are called "the glucose trap" [61]. Adipose has enhanced glucose utilization during accelerated body-fat recovery (catch-up fat), which is connected with muscle-adipose glucose redistribution [63, 64]. The authors use the terminology “the glucose sink” to describe the role of enhanced de novo lipogenesis in regulating glycemia during catch-up growth [65].

Cancer can cause cachexia, a wasting syndrome. Cachexia is associated with systemic inflammation connected with tumors and tumor-induced changes in the metabolism [66, 67]. Brown adipose tissue and energy expenditure are increased in cachexic patients. The uncoupling protein UCP3, found in brown adipose tissue, is increased in cancer and is connected with high energy expenditure [68]. Tumor-derived parathyroid hormone-related protein (PTHrP) is involved in adipose beiging, energy-wasting, and cancer cachexia [69].

Hormone production

Both adipose and tumors produce hormones. The concept of adipose as an endocrine organ is widely accepted [7, 56, 70, 71]. Adipose organ secretes several hormones (adipokines and batokines [72]) and classical cytokines, especially TNFα. The energy metabolism is regulated by adipokines leptin, adiponectin, resistin, and others [56]. Leptin has an important physiological role in the central control of energy and lipid metabolism and the regulation of metabolism-immune system interplay (immunometabolism) [73]. Mammalian leptin is defined as a lipostatic signal, which regulates energy balance by controlling food intake. It also regulates glucose homeostasis maintenance and participates in the regulation of immunometabolism [56, 73].

The phenomenon of hormone secretion by non-endocrine tumors is known as “ectopic” hormone production [74,75,76]. It causes unique clinical syndromes or endocrine paraneoplastic syndromes. These syndromes represent an important cause of morbidity and mortality [77]. Ectopic hormones are similar to normal hormones, but in tumors, they usually are present in lower amounts per unit mass than in normal endocrine organs.

miRNA production

Both adipose and tumors produce miRNAs that influence other tissues. For example, adipose-derived circulating miRNAs can regulate gene expression in other tissues [78]. Tumor-derived immune-modulatory miRNAs influence cancer immune surveillance and immune escape [79].

Immunosuppression

Obesity and related metabolic syndrome cause negative effects on immunity [56, 73, 80, 81]. Cancer immunosuppression is also a well-known phenomenon [82,83,84,85]. Obese metabolism suppresses antitumor immunity [86].

Destructive infiltration in other organs and tissues

Ectopic lipid deposition (ELD) in skeletal muscles, heart, liver, pancreas, placenta, and kidney during obesity is a major cause of metabolism distortion [87,88,89]. ELD is caused by the formation of lipid droplets in the organ's parenchymal cells; in adipocytes originated by differentiation of resident adipogenic progenitors; or in adipocytes differentiated after infiltration of organs with adipocyte progenitors from subcutaneous adipose tissue [90,91,92,93]. Infiltration of adipocyte progenitors in other organs with a negative influence on these organs' functions is the most important similarity with tumor metastasis.

CXCL12/CXCR4 chemokine axis participates in tumor progression and metastasis [94,95,96]. Adipocyte progenitors trafficking is also regulated by the CXCL12/CXCR4 axis [93].

Similar drugs may be used for the treatment of obesity and cancer

Thiazolidinediones (or glitazones) are used for the treatment of type 2 diabetes. By binding peroxisome proliferator-activated receptor gamma (PPARγ) they promote the maturation of adipocytes. They also suppress tumor cell invasion, migration, and invasiveness through CXCL12/CXCR4 pathway. In addition, it was found that treatment of mice with pioglitazone (a member of the glitazone group) prevents infiltration of adipocyte progenitors in skeletal muscles [93]. The other class of antidiabetic drugs—biguanides—also act as anti-carcinogens and inhibitors of tumor growth [97,98,99].

The connection between obesity and cancer

Obesity-related inflammation and oxidative stress cause DNA damage that can lead to obesity-related carcinogenesis [47].

Obesity and type 2 diabetes are associated with the risk of cancer and cancer-related mortality, as shown in epidemiological studies [100]. The link can be related to the insulin/insulin-like growth factor (IGF) system [57, 100]. Throughout evolution, this system “has integrated the control of tissue growth with metabolic status" [57]. Tumors are connected with the insulin/IGF system and systemic metabolism. The development and progression of several types of cancer are determined by the insulin/IGF system [98]. The factors that play a role in this connection include insulin resistance, hyperinsulinemia, increased levels of insulin growth factors (IGFs), hormones, and inflammatory markers [57, 100, 101].

Using Paget’s terminology of “seed” and “soil” as related to tumor metastasis [102], Holly and co-authors describe the internal milieu of obese individuals, or "soil," as containing high levels of glucose, insulin, insulin-like growth factors, inflammatory cytokines, and adipokines. These authors believe that such an environment stimulates the latent neoplastic lesions, the "seeds," to progress to clinical cancer [57]. Indeed, obese metabolism suppresses antitumor immunity [86].

The paradox of some benefits of obesity in cancer is also known: moderate overweight and early obese states can improve the survival and response to therapy [103, 104].

Mammalian adipose is a tumor-like organ, and obesity is a tumor-like process

Thus, adipose, an evolutionarily young organ of mammals, shares many features with tumors. Adipose unlimited expansion is similar to tumor growth, and lipomas are the most frequent soft tissue tumors. Chronic inflammation is characteristic of obese adipose and tumors. Both adipose and tumors exert systemic metabolic and immunological influence on the organism; both participate in paracrine and endocrine interactions with other tissues; both produce miRNAs that influence other tissues; both are characterized by plasticity, induce angiogenesis and participate in morphogenetic processes. Tumors act as “glucose trap”, and adipose during catch-up fat—as “glucose sink”. Obese adipose and tumors can cause immunosuppression; obese adipose and tumors are connected with remodeling and disfunction, with DNA damage and cell death resistance. Most important, adipose cells can metastasize into normal organs and impair their functions, similarly to malignant tumors. Finally, the same drugs and interventions are used against obesity, diabetes, ectopic lipid deposition, and tumors. Many of the common features of tumors and adipose organ are in the list of so-called "hallmarks of cancer" [105], and many of them are connected with the obese state of the adipose organ.

As discussed earlier, other evolutionarily novel organs of mammals, such as the placenta, mammary gland, and prostate, also have many tumor features [2, 5]. However, evolutionarily older organs are characterized by lower cancer rates [26] and do not have (or have fewer) tumor features. The author suggested calling normal organs, which have many tumor features, the "tumor-like organs" [3, 5]. We may conclude that mammalian adipose is a tumor-like organ and obesity is a tumor-like process.

The possible origin of mammalian adipose from ancestral mesenchymal hereditary tumors

The mammalian adipose organ's tumor features suggest its recent evolutionary origin from ancestral hereditary tumors. Following the main hypothesis, the adipose organ's origin may be represented as follows. Some diffuse mesenchymal hereditary tumors in eutherian ancestors, which produced several biologically active compounds (future adipokine hormones), acquired the capability to synthesize and accumulate fat using pre-existing and evolutionarily novel genes. Accumulation of lipids inhibited the potential of progression to malignancy ("gain fat—loose metastasis," [15]) and, together with future adipokine substances, was selected in ancestral Mammalia for control of energy metabolism in connection to the nutritional status of the organism, as an adaptation to new diets and thermoregulation. As a result of this evolutionary process, the evolutionarily novel mammalian organ involved in the storage and expenditure of energy with many ancestral tumor features—the adipose organ—originated.

The lab of the author has already obtained the evidence in support of a hypothesis specific to the origin of mammalian adipose organ.

In our previous article [4], we studied fish genes expressed in transgenic zebrafish inducible tumors, tumors after regression, and spontaneous zebrafish tumors. Among these genes, using the Orthologous Matrix (OMA) approach, we selected genes evolutionarily novel to fishes (as compared to lamprey, myxine, and other organisms in fish evolutionary lineage), and studied their human orthologs. We described many human orthologs that acquired progressive functions (such as involvement in the development of the placenta, mammary gland, lungs, neocortex, according to Gene Ontology studies), which are not encountered in fish [4]. Several of those human genes with progressive functions—LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA genes—also acquired functions connected with the adipose organ.

Thus, human LEP, which encodes leptin, became the central regulator of energy metabolism in mammals. It is involved in beige/brown fat cell differentiation regulation [4, 106] and lipostatic function (fish leptin is not an adipostat [107, 108]). Mammalian leptin is also involved in thermoregulation [109, 110].

NOTCH1 regulates adipose browning, energy metabolism, and thermogenesis [111, 112].

SPRY1 is mandatory for the initiation and regulation of adipogenesis, for maintaining proliferation and differentiation of human adipose stem/progenitor cells (ASCs). It is induced in ASCs after weight loss [113, 114]. SPRY1 can suppress PPARG [115].

PPARG, the ortholog of the fish pparg gene, was selected in [4] because of its involvement in placenta development. Peroxisome proliferator-activated receptor gamma (PPARγ), encoded by PPARG, is the target of thiazolidinediones antidiabetic treatment, as discussed above. PPARγ participates in the differentiation of adipocytes and activation of thermogenic gene expression in brown adipocytes [116]. PPARγ is a major regulator of adipocyte differentiation and function [117]. It plays a role in lipodystrophy, obesity, and diabetes [118] and can downregulate LEP gene expression [104, 119].

ID2 stimulates PPARγ expression, adipocyte differentiation, and adipogenesis. Its expression is elevated in adipose tissues during obesity [120].

The CIDEA gene was also found among human orthologs of novel fish genes expressed in fish tumors [4]. CIDE proteins are associated with lipid droplets and regulate lipid metabolism. CIDE protein family includes CIDEA, CIDEB, and CIDEC proteins [52, 53]. In mice, CIDEA is a marker of brown and brite adipocytes [121]. In humans, the CIDEA gene regulates adipocyte beiging [122]. It means that in mammals CIDEA gene also acquired progressive functions not encountered in fish. Transcription of CIDEA gene is activated by PPARγ [52].

Each of the LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA genes is also involved in tumor development in humans.

Leptin is overexpressed in breast cancer [104] and many other types of cancer [123], has a role at different levels and participates in cancer progression. Its activation results in the activation of multiple oncogenic pathways. Leptin oncogenic functions are reinforced through crosstalk with oncogenes, e.g. NOTCH [104].

NOTCH1 has both oncogenic and tumor suppressor abilities [124, 125].

SPRY1 is downregulated in some tumors and overexpressed in other tumors. Depending on the cellular context, it may serve either as a tumor suppressor or tumor promoter. SPRY1 expression is essential for induction, maintenance, and progression of tumors [126,127,128,129,130,131].

PPARγ plays oncogenic and tumor suppressor roles. PPARγ functions as a tumor suppressor in colon, lung, pancreatic, and breast cancers. A tumor-promoting role for PPARγ has been suggested in a variety of cancers as well [117].

ID family of proteins participates in the regulation of pathways essential to the progression of cancer. ID gene transcription is sensitive to signals from the cellular environment including oncoproteins. Depending on the context, ID proteins can play tumor-promoting or tumor-suppressing roles [132,133,134]. The tumor-suppressive role of ID2 has been described in [135, 136], and its oncogenic role—in [137].

CIDE proteins control lipid droplets' size and metabolism [53]. Lipid droplets actively participate in tumor processes and accumulate in a variety of cancer cells [138, 139]. CIDEA controls the beiging of adipocytes [122], and beige adipocytes contribute to breast cancer progression [140]. CIDEA plays an important role in human cachexia [141]. CIDE proteins were originally discovered as apoptotic proteins. They induce caspase-independent cell death in various cell types [53] that can be connected with cancer processes.

Once more, orthologs of LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA genes originated in fishes and were expressed in fish tumors. In humans, these genes acquired progressive functions not encountered in fishes, including functions connected with mammalian adipose organ, form a gene network with mutual influences, and participate in tumor processes. These genomic and transcriptomic data support the possibility of mammalian adipose origin from ancestral hereditary tumors and the tumor-like nature of mammalian adipose.

The hypothesis of adipose origin by ancestral tumor neofunctionalization is also strongly supported by the experimental cytology approach: it was demonstrated that breast cancer cells can trans-differentiate, in experimental conditions of adipogenesis, into post-mitotic functional adipocytes with the loss of malignancy [15]. Similarly, by trans-differentiation after the expression of evolutionarily novel genes and gene combinations involved in adipogenesis, mammalian adipocytes could originate in evolution from ancestral hereditary tumor cells.

The possible origin of mammalian adipose from ancestral tumors is in correspondence with other examples of hereditary tumors, which played roles in the origin of new cell types, tissues, and organs discussed in our previous publications [1,2,3, 5].

Conclusion

The ongoing synthesis of evolutionary biology and health sciences attempts to find evolutionary roots of disease [1, 142, 143]. If the hypothesis of the evolutionary origin of the adipose organ from the ancestral hereditary tumor is correct, it may help find new clues to obesity and cancer. Approaches developed to prevent and treat obesity may be examined to prevent and treat tumors and vice-versa. Empirically, some of such approaches are already underway [144]. Cancer metabolism is currently being studied for therapeutic opportunities, along with calorie restriction interventions for the prevention and treatment of cancer. Obese adipose supports tumor growth in various ways, and interventions aimed at metabolic disorders caused by adipose expansion may also be effective against tumors.

Our hypothesis may add a theoretical ground to such studies and may open new opportunities to resolve the oncological problem and the problem of the obesity epidemic. New interventions targeting LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA gene network, in addition to what already is going on, can be designed for treatment and prevention of both obesity and tumors.

Availability of data and materials

Not applicable.

Abbreviations

ASCs:

Adipose stem/progenitor cells

BAT:

Brown adipose tissue

DFFA:

DNA fragmentation factor alpha

ECM:

Extracellular matrix

ELD:

Ectopic lipid deposition

EMT:

Epithelial-mesenchymal transition

FIT:

Fat storage-inducing transmembrane

IGF:

Insulin-like growth factor

OMA:

Orthologous matrix

PPARγ:

Peroxisome proliferator-activated receptor gamma

PTHrP:

Tumor-derived parathyroid hormone-related protein

UCP:

Uncoupling protein

WAT:

White adipose tissue

References

  1. Kozlov AP. Evolution by tumor neofunctionalization: the role of tumors in the origin of new cell types, tissues, and organs. Amsterdam: Elsevier; 2014.

    Google Scholar 

  2. Kozlov AP. The role of heritable tumors in evolution of development: a new theory of carcino-evo-devo. Acta Nat. 2019;11:65–72.

    Article  CAS  Google Scholar 

  3. Kozlov AP, Zabezhinski MA, Popovich IG, Polev DE, Shilov ES, Murashev BV. Hyperplastic skin growth on the head of goldfish—comparative oncology aspects. Probl Oncol (Voprosi Oncologii). 2012;58:387–93.

    CAS  Google Scholar 

  4. Matyunina EA, Emelyanov AV, Kurbatova TV, Makashov AA, Mizgirev IV, Kozlov AP. Evolutionarily novel genes are expressed in transgenic fish tumors and their orthologs are involved in the development of progressive traits in humans. Infect Agents Cancer. 2019;14:46.

    Article  CAS  Google Scholar 

  5. Kozlov AP. Mammalian tumor-like organs. 1. The role of tumor-like normal organs and atypical tumor organs in the evolution of development (carcino-evo-devo). Infect Agents Cancer. 2022;17:2.

    Article  CAS  Google Scholar 

  6. Cinti S. The adipose organ. Milan: Editrice Kurtis; 2001.

    Google Scholar 

  7. Frayn KN, Karpe F, Fielding BA, Macdonald IA, Coppack SW. Integrative physiology of human adipose tissue. Int J Obes Relat Metab Disord. 2003;27:875–88.

    Article  CAS  PubMed  Google Scholar 

  8. Cinti S. The adipose organ. Prostaglandins Leukot Essent Fatty Acids. 2005;73:9–15.

    Article  CAS  PubMed  Google Scholar 

  9. Cinti S. The adipose organ. In: Fantuzzi G, Mazzone T, editors. Adipose tissue and adipokines in health and disease. Nutrition and health. Totowa: Humana Press Inc; 2007. p. 3–19.

    Chapter  Google Scholar 

  10. Cinti S. The adipose organ at a glance. Dis Model Mech. 2012;5:588–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kaisanlahti A, Glumoff T. Browning of white fat: agents and implications for beige adipose tissue to type 2 diabetes. J Physiol Biochem. 2019;75:1–10.

    Article  CAS  PubMed  Google Scholar 

  12. Horowitz MA, Berry R, Holtrup B, Sebo Z, Nelson T, Fretz JA, et al. Bone marrow adipocytes. Adipocyte. 2017;6:193–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Seal P, Bjork B, Yang W, Kajimura S, Chin S, Kuang S, et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature. 2008;454:961–7.

    Article  CAS  Google Scholar 

  14. Tseng Y-H, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature. 2008;454:1000–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ishay-Ronen D, Diepenbruck M, Kalathur RKR, Sugiyama N, Tiede S, Ivanek R, et al. Gain fat-lose metastasis: converting invasive breast cancer cells into adipocytes inhibits cancer metastasis. Cancer Cell. 2019;35:17–32.

    Article  CAS  PubMed  Google Scholar 

  16. Kadereit B, Kumar P, Wang W-J, Miranda D, Snapp EL, Severina N, et al. Evolutionarily conserved gene family important for fat storage. Proc Natl Acad Sci USA. 2008;105:94–9.

    Article  CAS  PubMed  Google Scholar 

  17. Zwick RK, Guerrero-Juarez CF, Horsley V, Plikus MV. Anatomical, physiological and functional diversity of adipose tissue. Cell Metab. 2018;27:63–83.

    Article  CAS  Google Scholar 

  18. Pond CM. The evolution of mammalian adipose tissue. In: Symonds M, editor. Adipose tissue biology. New York: Springer; 2011. p. 227–69.

    Google Scholar 

  19. Gesta S, Tseng Y-H, Kahn CR. Developmental origin of fat: tracking obesity to its source. Cell. 2007;131:242–56.

    Article  CAS  PubMed  Google Scholar 

  20. Mezentseva NV, Kumaratilake JS, Newman SA. The brown adipocyte differentiation pathway in birds: an evolutionary toad not taken. BMC Biol. 2008;6:17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature. 2006;444:847–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling, and obesity. J Clin Investig. 2011;121:2094–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Murphey MD, Carrol JF, Flemming DJ, Pope TL, Gannon FH, Kransdorf MJ, et al. From the archives of the AFIP: benign musculoskeletal lipomatous lesions. Radiographics. 2004;24:1433–66.

    Article  PubMed  Google Scholar 

  24. Awh MA. Lipoma and liposarcoma. In: Radsource. MRI Web Clinic. 2014. https://radsource.us/lipoma-and-liposarcoma/. Accessed Jan 2014.

  25. Johnson CN, Ha AS, Chen E, Davidson D. Lipomatous soft-tissue tumors. J Am Acad Orthop Surg. 2018;26:779–88.

    Article  PubMed  Google Scholar 

  26. Davies JA. Inverse correlation between an organ’s cancer rate and its evolutionary antiquity. Organogenesis. 2004;1:60–3.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Pedeutour F, Italiano A. Adipose tumors. In: Schwab M, editor. Encyclopedia of cancer. Berlin: Springer; 2011. https://doi.org/10.1007/978-3-642-16483-5_106.

    Chapter  Google Scholar 

  28. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.

    Article  PubMed  Google Scholar 

  29. Conyers R, Young S, Thomas DM. Liposarcoma: molecular genetics and therapeutics. Sarcoma. 2011;2011:483154.

    Article  PubMed  CAS  Google Scholar 

  30. Shen S, Clairambault J. Cell plasticity in cancer cell populations. F1000 Res. 2020;9:635.

    Article  CAS  Google Scholar 

  31. Pisani DF, Djedani M, Beranger GE, Elabd C, Scheideler M, Ailhaud G, et al. Differentiation of human adipose-derived stem cells into “brite” (brown-in-white) adipocytes. Front Endocrinol. 2011;2:87.

    Article  CAS  Google Scholar 

  32. Yadav H, Rane SG. TGF-β/Smad3 signaling regulates brown adipocyte induction in white adipose tissue. Front Endocrinol. 2012;3:35.

    Article  CAS  Google Scholar 

  33. Bielczyk-Maczynska E. White adipocyte plasticity in physiology and disease. Cells. 2019;8:1507.

    Article  CAS  PubMed Central  Google Scholar 

  34. Joshi PA, Waterhouse PD, Kasaian K, Fang H, Gulyaeva O, Sul HS, et al. PDGFRα+ stromal adipocyte progenitors transition into epithelial cells during lobulo-alveologenesis in the murine mammary gland. Nat Commun. 2019;10:1760.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. D’Esposito V, Ambrosio MR, Giuliano M, Cabaro S, Miele C, Beguinot F, et al. Mammary adipose tissue control of breast cancer progression: Impact of obesity and diabetes. Front Oncol. 2020;10:1554.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Herold J, Kalucka J. Angiogenesis in adipose tissue: the interplay between adipose and endothelial cells. Front Physiol. 2021;11:624903.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Itoh M, Suganami T, Hachiya R, Ogawa Y. Adipose tissue remodeling as homeostatic inflammation. Int J Inflam. 2011;2011:720926.

    PubMed  PubMed Central  Google Scholar 

  38. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity. 2019;51:27–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nair SS, Kumar R. Chromatin remodeling in cancer: a gateway to regulate gene transcription. Mol Oncol. 2012;6:611–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Models Mech. 2011;4:165–78.

    Article  CAS  Google Scholar 

  41. Mohan V, Das A, Sagi I. Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol. 2019;62:192–200.

    Article  PubMed  CAS  Google Scholar 

  42. Farnsworth RH, Lackmann M, Achen MG, Stacker SA. Vascular remodeling in cancer. Oncogene. 2014;33:3496–505.

    Article  CAS  PubMed  Google Scholar 

  43. Chang L, Fang S, Gu W. The molecular mechanism of metabolic remodeling in lung cancer. J Cancer. 2020;11:1403–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Crewe C, An YA, Scherer PE. The ominous triad of adipose tissue dysfunction: inflammation, fibrosis, and impaired angiogenesis. J Clin Investig. 2017;127:74–82.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose tissue disfunction as the determinant of obesity-associated metabolic complications. Int J Mol Sci. 2019;20:2358.

    Article  CAS  PubMed Central  Google Scholar 

  46. Wlodarczyk M, Nowika G. Obesity, DNA damage, and development of obesity-related diseases. Int J Mol Sci. 2019;20:1146.

    Article  CAS  PubMed Central  Google Scholar 

  47. Cerda C, Sanchez C, Climent B, Vázquez A, Iradi A, El Amrani F, et al. Oxidative stress and DNA damage in obesity-related tumorigenesis. Adv Exp Med Biol. 2014;824:5–17.

    Article  CAS  PubMed  Google Scholar 

  48. Vergoni B, Cornejo PJ, Gilleron J, Djedaini M, Ceppo F, Jacquel A, et al. DNA damage and the activation of the p53 pathway mediate alterations in metabolic and secretory functions of adipocytes. Diabetes. 2016;65:3062–74.

    Article  CAS  PubMed  Google Scholar 

  49. Moon JJ, Lu A, Moon C. Role of genomic instability in human carcinogenesis. Exp Biol Med (Maywood). 2019;244:227–40.

    Article  CAS  Google Scholar 

  50. Lee YK, Chung YS, Lee JH, Chun JM, Park JH. The intricate role of p53 in adipocyte differentiation and function. Cells. 2020;9:2621.

    Article  CAS  PubMed Central  Google Scholar 

  51. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instability—an evolving hallmark of cancer. Nature Rev Mol Cell Biol. 2010;11:220–8.

    Article  CAS  Google Scholar 

  52. Slayton M, Gupta A, Balakrishnan B, Puri V. CIDE proteins in human health and disease. Cells. 2019;8:238.

    Article  CAS  PubMed Central  Google Scholar 

  53. Chen F-J, Yin Y, Chua BT, Li P. CIDE family proteins control lipid homeostasis and the development of metabolic diseases. Traffic. 2020;21:94–105.

    Article  CAS  PubMed  Google Scholar 

  54. Wang L, Wang SP, Shi Y, Li R, Gunter S, Ong YT, et al. YAP and TAZ protect against white adipocyte cell death during obesity. Nat Commun. 2020;11:5455.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Izquierdo AG, Carreira MC, Rodrigez-Carnero G, Fernandez-Quintela A, Sueiro AM, Martinez-Olmos MA, et al. Weight loss normalizes enhanced expression of the oncogene survivin in visceral adipose tissue and blood leukocytes from individuals with obesity. Int J Obes. 2021;45:206–16.

    Article  CAS  Google Scholar 

  56. Booth A, Magnuson A, Fouts J, Foster MT. Adipose tissue: an endocrine organ playing a role in metabolic regulation. Horm Mol Biol Clin Investig. 2016;26:25–42.

    CAS  PubMed  Google Scholar 

  57. Holly JMP, Biernacka K, Perks CM. Systemic metabolism, its regulators, and cancer: past mistakes and future potential. Front Endocrinol (Lausanne). 2019;10:65.

    Article  Google Scholar 

  58. Ertl E. ‘Systemic effects’ during the growth of malignant experimental tumors. Oncology. 1973;27:415–29.

    Article  CAS  PubMed  Google Scholar 

  59. McAllister SS, Weinberg RA. The tumor-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16:717–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol. 1927;8:519–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Yu L, Chen X, Wang L, Chen S. The sweet trap in tumors: aerobic glycolysis and potential targets for therapy. Oncotarget. 2016;7:38908–26.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Lin X, Xiao Z, Chen T, Liang SH, Guo H. Glucose metabolism on tumor plasticity, diagnosis, and treatment. Front Oncol. 2020;10:317.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Cettour-Rose P, Samec S, Russel AP, Summermatter S, Mainieri D, Carrillo-Theander C, et al. Redistribution of glucose from skeletal muscle to adipose tissue during catch-up fat. Diabetes. 2005;54:751–6.

    Article  CAS  PubMed  Google Scholar 

  64. Summermatter S, Marcelino H, Arsenijevic D, Buchala A, Aprikian O, Assimacopoulos-Jeannet F, et al. Adipose tissue plasticity during catch-up fat driven thrifty metabolism: relevance for muscle-adipose glucose redistribution during catch-up growth. Diabetes. 2009;58:2228–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Marcelino H, Veyrat-Durebex C, Summermater S, Sarafian D, Miles-Chan J, Arsenijevic D, et al. A role for adipose tissue de novo lipogenesis in glucose homeostasis during catch-up growth. Diabetes. 2013;62:362–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. DeWys WD. Weight loss and nutritional abnormalities in cancer patients: incidence, severity, and significance. In: Calman KC, Fearon KCH, editors. Clinics in oncology. London: Saunders; 1986. p. 251–61.

    Google Scholar 

  67. Fearon KC, Voss AC, Hustead DS, Cancer Cachexia Study Group. Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr. 2006;83:1345–50.

    Article  CAS  PubMed  Google Scholar 

  68. Dhanapal R, Saraswathi T, Govind RN. Cancer cachexia. J Oral Maxillofac Pathol. 2011;15:257–60.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Kir S, White JP, Kleiner S, Kazak L, Cohen P, Baracos VE, et al. Tumor-derived PTHrP triggers adipose tissue browning and cancer cachexia. Nature. 2014;513:100–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ahima ES. Adipose tissue as an endocrine organ. Obesity (Silver Spring). 2006;14(Suppl 5):242S-249S.

    Article  CAS  Google Scholar 

  71. Galic S, Oakhill JS, Steinberg GR. Adipose tissue as an endocrine organ. Mol Cell Endocrinol. 2010;316:129–39.

    Article  CAS  PubMed  Google Scholar 

  72. Alvarez-Artime A, Garcia-Soler B, Sainz RM, Mayo JC. Emerging roles for browning of white adipose tissue in prostate cancer malignant behavior. Int J Mol Sci. 2021;22:560.

    Article  CAS  Google Scholar 

  73. Francisco V, Pino J, Campos-Cabaleiro V, Ruiz-Fernandez C, Mera A, Gonzalez-Gay MA, et al. Obesity, fat mass and immune system: role for leptin. Front Physiol. 2018;9:640.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Neville AM. Ectopic production of hormones by tumours. Pathological aspects of the para-endocrine syndrome. Proc R Soc Med. 1972;65:55–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Martin TJ. Secretion of hypothalamic and pituitary hormones by non-endocrine tumours. J Clin Pathol Suppl (Assoc Clin Pathol). 1976;7:26–30.

    Article  CAS  Google Scholar 

  76. Baylin SB, Mendelsohn G. Ectopic (inappropriate) hormone production by tumors: mechanisms involved and the biological and clinical implications. Endocr Rev. 1980;1:45–77.

    Article  CAS  PubMed  Google Scholar 

  77. Yeung SCJ, Gagel RF, et al. Endocrine paraneoplastic syndromes (“ectopic” hormone production). In: Kufe DW, Pollock RE, Weichselbaum RR, Bast RC, Gansler TS, Holland JF, et al., editors. Holland-frei cancer medicine. 6th ed. Hamilton: BC Decker; 2003.

    Google Scholar 

  78. Thomou T, Mori MA, Dreyfuss JM, Konishi M, Sakaguchi M, Wolfrum C, et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature. 2017;542:450–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Yi M, Xu L, Jiao Y, Luo S, Li A, Wu K. The role of cancer-derived microRNAs in cancer immune escape. J Hematol Oncol. 2020;13:25.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Andersen CJ, Murphy KE, Fernandez ML. Impact of obesity and metabolic syndrome on immunity. Adv Nutr. 2016;7:66–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Solt CM, Hill JL, Vonderpool K, Foster MT. Obesity-induced immune dysfunction and immunosuppression: TEM observation of visceral and subcutaneous lymph node microarchitecture and immune cell interactions. Horm Mol Biol Clin Investig. 2019. https://doi.org/10.1515/hmbci-2018-0083.

    Article  PubMed  Google Scholar 

  82. Kim R, Emi M, Tanabe K, Arihiro K. Tumor-driven evolution of immunosuppressive networks during malignant progression. Cancer Res. 2006;66:5527–36.

    Article  CAS  PubMed  Google Scholar 

  83. Kim R, Emi M, Tanabe K. Cancer immunosuppression and autoimmune disease: beyond immunosuppressive networks for tumor immunity. Immunology. 2006;119:254–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lu C, Rong D, Zhang B, Zheng W, Wang X, Chen Z. Current perspective on the immunosuppressive tumor microenvironment in hepatocellular carcinoma: challenges and opportunities. Mol Cancer. 2019;18:130.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Togashi Y, Shitara K, Nishikawa H. Regulatory T cells in cancer immunosuppression—implications for anticancer therapy. Nat Rev Clin Oncol. 2019;16:356–71.

    Article  CAS  PubMed  Google Scholar 

  86. Ringel AE, Drijvers JM, Baker GJ, Catozzi A, Garcia-Canaveras JC, Gassaway BM, et al. Obesity shapes metabolism in the tumor microenvironment to suppress antitumor immunity. Cell. 2020;183:1848–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Szendroedi J, Roden M. Ectopic lipids and organ function. Curr Opin Lipidol. 2009;20:50–6.

    Article  CAS  PubMed  Google Scholar 

  88. de Vries AP, Ruggenenti P, Ruan XZ, Praga M, Cruzado JM, Bajema IM, et al. Fatty kidney: emerging role of ectopic lipid in obesity-related renal disease. Lancet Diabetes Endocrinol. 2014;2:417–26.

    Article  PubMed  CAS  Google Scholar 

  89. Shulman GI. Ectopic fat in insulin resistance, dyslipidemia, and cardiometabolic disease. N Engl J Med. 2014;371:1131–41.

    Article  PubMed  CAS  Google Scholar 

  90. Pinnick KE, Collins SC, Londos C, Gauguier D, Clark A, Fielding BA. Pancreatic ectopic fat is characterized by adipocyte infiltration and altered lipid composition. Obesity. 2008;16:522–30.

    Article  CAS  PubMed  Google Scholar 

  91. Gaborit B, Abdesselam I, Dutour A. Epicardial fat: More than just an “Epi” phenomenon? Horm Metab Res. 2013;45:991–1001.

    Article  CAS  PubMed  Google Scholar 

  92. Hamrick MW, McGee-Lawrence ME, Frechette DM. Fatty infiltration of skeletal muscle: mechanisms and comparisons with bone marrow adiposity. Front Endocrinol. 2016;7:69.

    Article  Google Scholar 

  93. Girousse A, Gil-Ortega M, Bourlier V, Bergeaud C, Sastourné-Arrey Q, Moro C, et al. The release of adipose stromal cells from subcutaneous adipose tissue regulates intramuscular adipocyte deposition. Cell Rep. 2019;27:323–33.

    Article  CAS  PubMed  Google Scholar 

  94. Teicher BA, Fricker SP. CXCL12/CXCR4 pathway in cancer. Clin Cancer Res. 2010;16:2927–31.

    Article  CAS  PubMed  Google Scholar 

  95. Zhou Y, Cao H-B, Li W-J, Zhao L. The CXCL12 (SDF-1)/CXCR4 chemokine axis: oncogenic properties, molecular targeting, and synthetic and natural product CXCR4 inhibitors for cancer therapy. Clin J Nat Med. 2018;16:801–10.

    CAS  Google Scholar 

  96. Zhou W, Guo S, Liu M, Barrow ME, Wang G. Targeting CXCL12/CXCR4 axis in tumor immunotherapy. Curr Med Chem. 2019;26:3026–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Dilman VM, Berstein LM, Ostroumova MN, Fedorov SN, Poroshina TE, Tsyrlina EV, et al. Metabolic immunosuppression and metabolic immunotherapy. Oncology. 1982;39:13–9.

    Article  CAS  PubMed  Google Scholar 

  98. Anisimov VN, Bartke A. The key role of growth hormone-insulin-IGF-1 signaling in aging and cancer. Crit Rev Oncol Hematol. 2013;87:201–23.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Anisimov VN. Antidiabetic biguanides as anti-aging drugs. In: Vaiserman AM, editor. RSC drug discovery series no. 57: anti-aging drugs: from basic research to clinical practice. Cambridge: Royal Society of Chemistry; 2017. p. 416–31.

    Chapter  Google Scholar 

  100. Cohen DH, LeRoith D. Obesity, type 2 diabetes, and cancer: the insulin and IGF connection. Endocr Relat Cancer. 2012;19:F27–45.

    Article  CAS  PubMed  Google Scholar 

  101. Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nat Rev Cancer. 2004;4:505–18.

    Article  CAS  PubMed  Google Scholar 

  102. Paget S. The distribution of secondary growths in cancer of the breast. Lancet. 1889;133:571–3.

    Article  Google Scholar 

  103. Lennon H, Sperrin M, Badrick E, Renehan AG. The obesity paradox in cancer: a review. Curr Oncol Rep. 2016;18:56.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Sanchez-Jimenez F, Perez-Perez A, de la Cruz-Merino L, Sanchez-Margalet V. Obesity and breast cancer: role of leptin. Front Oncol. 2019;9:596.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.

    Article  CAS  PubMed  Google Scholar 

  106. Wang J, Ge J, Cao H, Zhang X, Guo Y, Li X, et al. Leptin promotes white adipocyte browning by inhibiting the Hh signaling pathway. Cells. 2019;8:372.

    Article  CAS  PubMed Central  Google Scholar 

  107. Deck CA, Honeycutt JL, Cheung E, Reynolds HM, Borski RJ. Assessing the functional role of leptin in energy homeostasis and the stress response in vertebrates. Front Endocrinol. 2017;8:63.

    Article  Google Scholar 

  108. Londraville RL, Prokop JW, Duff RJ, Liu Q, Tuttle M. On the molecular evolution of leptin, leptin receptor, and endospanin. Front Endocrinol. 2017;8:58.

    Article  Google Scholar 

  109. Deem JD, Muta K, Ogimoto K, Nelson JT, Velasco KR, Kaiyala KJ, et al. Leptin regulation of the core body temperature involves mechanisms independent of the thyroid axis. Am J Physiol Endocrinol Metab. 2018;315:E552–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Fisher AW, Cannon B, Negergaard J. Leptin: Is it thermogenic? Endocrine Rev. 2020;41:232–60.

    Article  Google Scholar 

  111. Bi P, Shan T, Liu W, Yue F, Yang X, Liang X-R, et al. Inhibition of Notch signaling promotes browning of white adipose tissue and ameliorates obesity. Nat Med. 2014;20:911–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Shan T, Liu J, Wu W, Xu Z, Wang Y. Roles of Notch signaling in adipocyte progenitor cells and mature adipocytes. J Cel Physiol. 2017;232:1258–61.

    Article  CAS  Google Scholar 

  113. Mandl M, Wagner SA, Hartzman FM, Mitterberger-Vogt MC, Zwierzina ME, Mattesich M, et al. Sprouty1 is a weight-loss target gene in human adipose stem-progenitor cells that is mandatory for the initiation of adipogenesis. Cell Death Dis. 2019;10:411.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Mandl M, Wagner SA, Hartzmann FM, Ejas A, Ritthammer H, Baumgarten S, et al. Sprouty 1 prevents cellular senescence maintaining proliferation and differentiation capacity of human adipose stem/progenitor cells. J Gerontol A Biol Sci Med Sci. 2020;75:2308–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Urs S, Venkatesh D, Tang Y, Henderson T, Yang X, Friesel RE, et al. Sprouty1 is a critical regulatory switch of mesenchymal stem cell lineage allocation. FASEB J. 2010;24:3264–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Yi D, Nguen HP, Sul HS. Epigenetic dynamics of the thermogenic gene program of adipocytes. Biochem J. 2020;477:1137–48.

    Article  CAS  PubMed  Google Scholar 

  117. Hernandez-Quiles M, Broekema MF, Kalkhoven E. PPARgamma in metabolism, immunity, and cancer: unified and diverse mechanisms of action. Front Endocrinol (Lausanne). 2021;12:624112.

    Article  Google Scholar 

  118. Corrales P, Vidal-Puig A, Medina-Gomez G. PPARs and metabolic disorders associated with challenged adipose tissue plasticity. Int J Mol Sci. 2018;19:2124.

    Article  PubMed Central  CAS  Google Scholar 

  119. Rudko OI, Tretiakov AV, Naumova EA, Klimov EA. Role of PPARs in progression to anxiety: literature analysis and signaling pathways reconstruction. PPAR Res. 2020;2020:8859017.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Park KW, Waki H, Villanueva CJ, Monticelli LA, Hong C, Kang S, et al. Inhibitor of DNA binding 2 is a small molecule-inducible modulator of peroxisome proliferator-activated receptor-expression and adipocyte differentiation. Mol Endocrinol. 2008;22:2038–48.

    Article  CAS  PubMed  Google Scholar 

  121. Fisher AW, Shabalina IG, Mattsson CL, Abreu-Vieira G, Cannon B, Nedergaard J, et al. UCP1 inhibition in Cidea-overexpressing mice is physiologically counteracted by brown adipocyte tissue hyperrecruitment. Am J Physiol Endocrinol Metab. 2017;312:E72–87.

    Article  Google Scholar 

  122. Jash S, Banerjee S, Lee M-J, Farmer SR, Puri V. CIDEA transcriptionally regulates UCP1 for britening and thermogenesis in human fat cells. iScience. 2019;20:73–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Lin T-C, Hsiao M. Leptin and cancer: updates functional roles in carcinogenesis, therapeutic niches, and developments. Int J Mol Sci. 2021;22:2870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Lobry C, Oh P, Mansour MR, Look AT, Aifantis I. Notch signaling: Switching an oncogene to a tumor suppressor. Blood. 2014;123:2451–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Brzozowa-Zasada M, Piecuch A, Michlsky M, Segiet O, Kurek J, Harabin-Slowinska M, et al. Notch and its oncogene activity in human malignancies. Eur Surg. 2017;49:199–209.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Lo TL, Fong CW, Yusoff P, Mckie AB, Chua MS, Leung HY, et al. Sprouty and cancer: the first term report. Cancer Lett. 2006;242:141–50.

    Article  CAS  PubMed  Google Scholar 

  127. Edwin F, Anderson K, Ying C, Patel TB. Intermolecular interactions of Sprouty proteins and their implications in development and disease. Mol Pharmacol. 2009;76:679–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. He Q, Jing H, Liaw L, Gower L, Vary C, Hua S, et al. Suppression of SPRY1 inhibits triple-negative breast cancer malignancy by decreasing EGF/EGFR mediated mesenchymal phenotype. Sci Rep. 2016;6:23216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Rosso V, Panuzzo C, Petiti J, Carturan S, Dragani M, Andreani G, et al. Reduced expression of Sprouty1 contributes to the aberrant proliferation and impaired apoptosis of acute myeloid leukemia cells. J Clin Med. 2019;8:972.

    Article  CAS  PubMed Central  Google Scholar 

  130. Montico B, Collizzi F, Girato G, Rizzo A, Salvati A, Baboci L, et al. Loss of Spry1 reduces the growth of BRAFV600-mutant cutaneous melanoma and improves response to targeted therapy. Cell Death Dis. 2020;11:392.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wang Y-Y, Wang W-C, Su C-W, Hsu CW, Yuan SS, Chen YK. Overexpression of Sprouty1 protein in human oral squamous cell carcinogenesis. J Dent Sci. 2021;16:21–8.

    Article  PubMed  Google Scholar 

  132. Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat Rev Cancer. 2014;14:77–91.

    Article  CAS  PubMed  Google Scholar 

  133. Nair R, Teo WS, Mittal V, Swarbrick A. ID proteins regulate diverse aspects of cancer progression and provide novel therapeutic opportunities. Mol Ther. 2014;22:1407–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Roschger C, Cabrele C. The Id-protein family in developmental and cancer-associated pathways. Cell Commun Signal. 2017;15:7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Ghisi M, Kats L, Masson F, Li J, Kratina T, Vidacs E, et al. Id2 and E proteins orchestrate the initiation and maintenance of MLL-rearranged acute myeloid leukemia. Cancer Cell. 2016;11(30):59–74.

    Article  CAS  Google Scholar 

  136. Li J, Roy S, Kim YM, Li S, Zhang B, Love C, et al. D2 collaborates with ID3 to suppress iNKT and innate-like tumors1. J Immunol. 2017;198:3136–48.

    Article  CAS  PubMed  Google Scholar 

  137. Kamata YU, Sumida T, Kobayashi Y, Ishikawa A, Kumamaru W, Mori Y. Introduction of ID2 enhances invasiveness in ID2-null oral squamous cell carcinoma cells via the SNAIL axis. Cancer Genom Proteom. 2016;11–12(13):493–7.

    Article  CAS  Google Scholar 

  138. Cruz ALS, Barreto EA, Fazolini NPB, Viola JPB, Bozza PT. Lipid droplets: platforms with multiple functions in cancer hallmarks. Cell Death Dis. 2020;11:105.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Petan T. Lipid droplets in cancer. In: Pedersen SHF, editor. Reviews in physiology, biochemistry and pharmacology. Berlin, Heidelberg: Springer; 2020. https://doi.org/10.1007/112_2020_51.

    Chapter  Google Scholar 

  140. Gantov M, Pagnotta P, Lotufo C, Rindone GM, Riera MF, Calvo JC, et al. Beige adipocytes contribute to breast cancer progression. Oncol Rep. 2021;45:317–28.

    Article  PubMed  CAS  Google Scholar 

  141. Laurencikiene J, Stenson BM, Nordstrom EA, Agustsson T, Langin D, Isaksson B, et al. Evidence for an important role of CIDEA in human cachexia. Cancer Res. 2008;68:9247–54.

    Article  CAS  PubMed  Google Scholar 

  142. Williams GC, Nesse RM. The Dawn of Darwinian medicine. Q Rev Biol. 1991;66:1–22.

    Article  CAS  PubMed  Google Scholar 

  143. Ewald PW. Evolution of infectious disease. New York: Oxford University Press; 1994.

    Google Scholar 

  144. Kaiser J. Special diets might boost the power of drugs to vanquish cancer. Science. 2021. https://doi.org/10.1126/science.abi7661.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The author is grateful to anonymous reviewers for their helpful comments.

Authors' information

A.K. currently combines several positions: Chief, Lab of Gene Neofunctionalization, Vavilov Institute of General Genetics of the Russian Academy of Sciences, Moscow; Director, The Biomedical Center; Chief, Kozlov Lab of Theoretical Biology, and Professor, Peter the Great St. Petersburg Polytechnic University, St. Petersburg. In 1979 A.K. formulated a concept of the possible evolutionary role of tumors and since then has been working on the development of this hypothesis and its experimental confirmation. In 2014, his book "Evolution by Tumor Neofunctionalization" was published by Elsevier/Academic Press. In 2016, the translation of the book into the Russian language was published by Peter the Great Polytechnic University. In 2019, the book was published in Chinese by China Science Publishing & Media Ltd (Science Press). Based on the Brachyury gene described by Professor A. K. anti-cancer vaccines are currently participating in about 30 clinical trials (https://clinicaltrials.gov/). Editorial Appointments 2006-Date—Member of the Editorial Board, Infectious Agents, and Cancer Journal. 2021-Date—Member of the Editorial Board of the Russian Journal “Advances in Modern Biology” of the Russian Academy of Sciences. 2021-Date—Member of the Editorial Board, “Pathology” Journal.

Funding

This research work was supported by the Academic Excellence Project 5-100 proposed by Peter the Great St. Petersburg Polytechnic University, and by funding from Vavilov Institute of General Genetics, RAS.

Author information

Authors and Affiliations

Authors

Contributions

APK is an only author. The author read and approved the final manuscript.

Corresponding author

Correspondence to A. P. Kozlov.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kozlov, A.P. Mammalian tumor-like organs. 2. Mammalian adipose has many tumor features and obesity is a tumor-like process. Infect Agents Cancer 17, 15 (2022). https://doi.org/10.1186/s13027-022-00423-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13027-022-00423-5

Keywords