Mechanisms underlying the effects of stress on tumorigenesis and metastasis (Review)

  • Authors:
    • Zhaozhou Zhang
    • Yan Wang
    • Qi Li
  • View Affiliations

  • Published online on: September 24, 2018     https://doi.org/10.3892/ijo.2018.4570
  • Pages: 2332-2342
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Stress is one of the fundamental survival mechanisms in nature. Although chronic or long-lasting stress can be detrimental to health, acute or short-term stress can have health benefits. The aim of the present review was to address the complexity and significance of stress in tumorigenesis. The review covers an evaluation of previously used and reported experimental animal models of stress, as well as the effects of stress on the neuroendocrine system, immune function, gut microbiota, and inflammation and multidrug resistance, all of which are closely associated with cancer occurrence, progression and treatment. The review concludes that understanding the efficacy of stress management (prevention and rehabilitation) is crucial to the development of comprehensive and individualized strategies for cancer prevention and treatment.

1. Introduction

Stress is a constellation of events that begins with a stressor, leading to stress perception and subsequently resulting in stress response (1). Stress can impair the balance or homeostatic state of an organism physiologically or psychologically when exposed to extrinsic or intrinsic adverse forces (2). It is well known that stress is ubiquitous and unavoidable in life. For certain individuals, stress can serve as a stimulant under certain conditions and as a burden under others. In addition, stress serving as a stimulant in certain individuals may be a burden for others, which indicates that there exists a varying degree of stress susceptibility among individuals (3,4). Stress occurs when an organism perceives a disruption, or a threat of disruption, of homeostasis. Factors that can induce stress include, among others, social circumstances, physical environment, hypoxia, emotional state, pain, glucose deprivation, accidents and chronic anxiety (5). Stress-related symptoms include negative emotions or altered mood and behavior, such as anxiety, irritability, anger, startle response, hostility and depression, which can negatively affect various organs of the human body (6). Studies have preliminarily revealed that differences in the stress experienced by different individuals can be due to genetic and environmental factors (7,8). It is also generally believed that moderate stress, such as routine or regular exercise or physical activity, can enhance the body’s immune response and reduce the risk of cancer occurrence, progression and mortality (9,10), while chronic stress can affect a number of physiological functions and lead to several diseases, including cancer (11-13) (Table I). Stressors can result in mood and anxiety disorders, including depression, anxiety and irritability, which may be associated with malignancies (14,15).

Table I

Differences between acute stress and chronic stress.

Table I

Differences between acute stress and chronic stress.

DifferencesChronic stressAcute stress(Ref.)
DurationProlonged-term (months to years)Short-term (minutes to hours)(16,17)
IntensityStrong/weak intensity stimulationStrong intensity stimulation(1,2,8,9,12)
Animal modelsSocial isolation, chronic mild stress, intimidationElectric shock, immobilization(19,20,21)
Response system(s)Mainly activates the HPA axisActivates the HPA axis and the sympathetic neural system(5,25,26,31,32)
OriginAdrenal medullaAdrenal medulla and sympathetic ganglia(5,27,28,45,46)
Response outcomesElevations in catecholamines, i.e., epinephrine and norepinephrineElevations in catecholamines and glucocorticoids(5,25-28)
Immune responseImmune suppression/immune pathologyImmune protection/immune pathology(56,57,59-62)
Possible significanceDetrimentalBeneficial(9,10,11,13)
TumorTumor development and progressionInhibits tumor development and progression(11-13,24-26)

[i] HPA, hypothalamic-pituitary-adrenal.

In order to elucidate the underlying mechanisms linking stress with tumorigenesis and metastasis, it is necessary to establish effective animal models. At present, the most common animal stress models are those developed for chronic restraint stress, maternal separation, and dietary and environmental stress. Tumor metastasis is a complex process that consists of proliferation/angiogenesis, detachment/invasion, embolism/circulation and evasion of immune system surveillance. The present review attempted to classify and summarize the mechanisms underlying the role of stress in tumorigenesis and cancer progression.

2. Stress and animal models of stress

Animal models mimicking the pattern of human diseases serve a key role in understanding the effects of stress on cancer. To study this effect, multiple physiological stress models have been utilized to promote fear or anxiety in rodents, including the following: i) Social isolation model, where laboratory rodents, which are highly social creatures, are housed individually in cages for extended periods to elicit loneliness; ii) restraint stress model, in which the animals are immobilized or confined to small spaces; and iii) intimidation-induced stress model, which involves placing rodents into the cage of another animal. When categorized by type, stressors are classified as physical (trauma/injury and exercise/exhaustion), cognitive (anxiety and depression), a combination of physical and cognitive (fire-fighting on a 24-h shift) or chemical (environmental toxins and diet). When categorized by duration, stressors are classified as acute (minutes to hours), and chronic (months to years) (16,17).

At present, the chronic mild stress (CMS) model is one of the most widely used stress models, allowing for a combination of a large variety of stressors with different numbers/lengths of intervals and the measurement of different behaviors as a response to rewards (18). Specifically, the CMS model consists of immobilization, forced swimming, noise, hypothermia, social isolation, resident/intruder aggression, maternal deprivation, stroboscopic illumination, cage titling, and food, water or sleep deprivation (19). Meanwhile, the chronic restraint stress animal model is one of the most common immobilizations, in which the duration of restraint has ranged from 1 to 12 h daily (20,21). Generally speaking, these stress models are evaluated based on three major criteria: i) Construct validity, where the experimental conditions are replicating causes of human diseases; ii) face validity, where the symptoms observed in diseased animals are consistent with clinical observations; and iii) predictive validity, where the animal responds to the treatment currently used in the clinic (22). However, no studies have systematically evaluated the validity or the efficiency of the CMS model, as researchers have often chosen a particular combination of stressors with timing based on previous practical experience and/or the particular requirements of their experiments (18). Stress can be harmful when it is chronic or long lasting, but a fact that is often overlooked is that a stress response can have salubrious adaptive effects in the short run, and that short-term stress may enhance cellular immunity and increase early resistance to cancer (23,24). The timing or duration of the stress models is an important factor that requires consideration. Therefore, it is essential to distinguish between acute and chronic stress in the following discussion (Fig. 1).

Figure 1

Types of stress and their effects on immune function. Stress can be categorized as either chronic or acute, based on its effects on the body and its duration. Certain individuals are more prone to stress, while others more resistant; this is due to constitution-associated differences among individuals. The fight-or-flight stress response is one of the fundamental defense mechanisms in nature that enables the body to survive. Acute stress can serve an important role in the ‘fight’ response by exerting immune protection functions, which can prove beneficial to the body and strengthen immune surveillance and resistance to cancer. Chronic stress, on the other hand, can serve an important role in the ‘fight’ response, which is detrimental to the body and may lead to immune suppression or immune escape, as well as promote cancer. It is worth noting that acute stress may also lead to immunopathology when it is experienced during self/innocuous antigen/allergen exposure, which can result in the increase of pro-inflammatory cytokines and is harmful to the body. According to the aforementioned model, there exists a resting zone between chronic (harmful) and acute (beneficial) stress, and the extent and efficiency with which an organism returns to its resting zone following stress depends on its resilience. Certain factors, such as lifestyle, psychosocial buffers and activities, can maximize the resting zone by optimizing beneficial stress and minimizing harmful stress, which enables the body to stay on the positive side of the stress spectrum.

3. Mechanisms underlying the effects of stress on tumorigenesis and metastasis

Effects of stress on the neuroendocrine system. In the fast-paced society of today, the pressure on individuals is high, and it often manifests itself in the form of anxiety, tension, insomnia and depression, all of which can lead to chronic stress. The hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS) are the two branches of the neuroendocrine system that govern the response to stress (25,26). Corticotrophin-releasing hormone (CRH) and arginine vasopressin (AVP), released from the hypothalamus, can activate the pituitary gland to release adrenocorticotropic hormone (ACTH), encephalin and endorphin (END), and the adrenal cortex to release ACTH-induced glucocorticoid (GC) (3,27). The release of CRH is under excitatory input from the amygdala and inhibitory input from the hippocampus (28). Meanwhile, the secretion of CRH and AVP is characterized by a precise circadian rhythm that can be disrupted by imposed stressors, and a circadian rhythm disorder can have the same detrimental effects as chronic stress (29,30). Stress can also result in increased secretion of catecholamines, including norepinephrine (NE) and epinephrine (E), which can enhance alertness and physiological functions, and elicit the fight-or-flight response (31,32). GC serves an important role in the treatment and chemo-resistance of tumors, and catecholamine can promote tumor growth and metastasis (33,34). Adolescent chronic stress can cause HPA hypo-responsiveness and depression-like behavior (35). Psychosocial stressors in cancer can result in the dysregulation of the HPA axis, and vice versa (36). Restraint stress facilitates cancer angiogenesis and metastasis by releasing β-END, prolactin, increasing concentrations of circulating catecholamine and GC, and increasing tumor-associated macrophage infiltration into the primary tumor (37,38). Psychological stress may attenuate antiangiogenic therapy, primarily through activating β-adrenergic signaling to promote tumor angiogenesis (39), and β-blockers or behavioral therapies can limit skeletal metastasis of breast cancer cells (40). Dopamine (DA) is also a catecholamine hormone, which can stabilize tumor blood vessels to block the effects of chronic stress on tumor vasculature, as the depletion of DA under chronic stress conditions creates a permissive microenvironment for tumor growth (41,42). Notably, studies have revealed that DA acts through five types of DA receptors; DA type-1 receptor overexpression is associated with advanced breast cancer and a poor prognosis (43), but DA type-2 receptor has been found to inhibit tumor growth (44).

Furthermore, since the lymphatic system and the pancreas are innervated by fibers of the SNS and have receptors for SNS neurotransmitters, chronic stress-induced SNS activity can increase pancreatic cancer growth, lymphatic vessel contraction and lymphocyte output into the lymphatic circulation. These processes may affect tumor lymphatic dissemination and cancer progression (45,46). Chronic restraint stress can attenuate the levels and function of p53 proteins, and promote the growth of human xenograft tumors, which is mediated by GC elevation during chronic restraint stress (47). A study has shown that exposure to chronic psychological stress may lead to significant changes in the proteomic profile of tumors (48). It is noteworthy that cytokines are potent activators of the central stress response and have regulatory effects on the HPA axis by forming a feedback loop through which the immune/inflammatory system communicates with the brain (49). The duration and magnitude of stress-induced increases in NE, Epi and GC have significant effects on immune cell redistribution and function (50,51) (Fig. 2). However, several studies have indicated that β-adrenergic receptor blockers can reverse the effects of chronic stress on cancer progression and abrogate drug resistance, which merits further investigation as a novel strategy for cancer treatment (52-54).

Figure 2

Systemic effects of stress on the neuroendocrine system. The HPA axis and the SNS are the two key branches of the neuroendocrine system that govern the response of an organism to stress. In response to stress, the paraventricular nucleus of the hypothalamus secretes CRH and AVP, which cause the pituitary gland to secrete ACTH. Meanwhile, the CRH/AVP neurons and central catecholaminergic neurons of the locus coeruleus/NE system are reciprocally innervated and activated. The secretion of CRH and AVP is characterized by a precise circadian rhythm. Subsequently, ACTH stimulates the adrenal cortex to release GC and cortisol. In addition, the HPA axis is controlled by feedback loops that tend to normalize the time-integrated secretion of cortisol. The SNS originates in the brain stem, with the sympathetic nerves located between the intermediolateral column of the T1 and L3 vertebrae. From there, cells synapse with post-synaptic ganglia located in the sympathetic trunk or in splanchnic ganglia throughout the body, and ultimately release NE. Under the condition of stress, particularly chronic stress, NE can promote tumor growth and metastasis. In the image, solid lines indicate stimulation and dashed lines indicate inhibition. HPA, hypothalamic-pituitary-adrenal; SNS, sympathetic nervous system; CRH, corticotrophin-releasing hormone; CRF, corticotrophin releasing factor; AVP, arginine vasopressin; ACTH, adrenocorticotropic hormone; IL-6, interleukin-6; NE, norepinephrine; Epi, epinephrine; GC, glucocorticoid; DA, dopamine.

Effects of stress on immune function. Stress exerts pleiotropic effects on the immune system, affecting the innate and adaptive immune responses (55). Stress-induced immune responses can be categorized as immune-protective, immune-pathological and immune-inhibitory. Notably, stress can be categorised as good or bad, based on the duration of the biological stress response (11). It is well known that acute stress can enhance immune function, whereas chronic stress can suppress it, as well as increasing the susceptibility to cancer (1). Stress can have beneficial and harmful effects, depending on the type of immune response; factors that determine the effects of stress on immune function include duration (acute or chronic), endogenous versus synthetic GCs and time of stressor (at early stages or late stages of the immune response) (1,56) (Fig. 1). It has been reported that chronic stress can affect individual components of the cellular immune system and downregulate the cellular immune response (57,58), which manifests in the form of a significant decrease in body weight and lymphatic organs (spleen, thymus and axillary lymph nodes) and a significant increase in the apoptotic cell count in all lymphatic organs (59). Chronic stress may influence the immune function and promote tumor growth by depressing T-cell-mediated immunity and reducing the lymphocyte count, which may depend on toll-like receptor 9 and β-arrestin 2 (60,61). Cytotoxic T lymphocytes (CTLs) are capable of secreting cytokines, such as interferon-γ (IFN-γ), and other effector molecules that serve a role in immune surveillance against tumor cells and the eradication of cancer stem cells (62), but NE and GC can decrease the number of CTLs or impair their function to attenuate their antitumor effect (63). However, a previous study found that psychological stress could augment immune response (64). With regards to the effect on non-specific cellular immunity, previous studies have demonstrated that chronic stress can weaken natural killer (NK) cell function and inhibit their activity, promoting tumor progression (65). Stress can also stimulate macrophages or monocytes to secrete interleukin (IL)-1, IL-6 and tumor necrosis factor (TNF)-α, but reduce the secretion of IL-2, IFN-γ and major histocompatibility complex (MHC), which may aid tumor cells to evade immune surveillance (66,67). Tumor associated-macrophages (TAMs) are one of the components of the tumor microenvironment in which stress can increase tumor infiltration by macrophages, and macrophage infiltration can mediate stress-enhanced metastasis in primary breast cancer (68). GC can induce macrophage phenotype changes from M1 to M2 to promote tumor progression (69). Daily restraint stress can also lead to increased monocyte chemoattractant protein-1 (MCP-1) expression and infiltration of cluster of differentiation (CD)14+ and CD68+ cells. In addition, the elevation of peripheral blood monocytes and TAMs has been associated with a worse progression-free survival time in patients with ovarian cancer (70). The matricellular protein thrombospondin-1 response to stress via the cluster of differentiation 47 (CD47) and thrombospondin-1/CD47 signaling pathways serves an important role in tumor angiogenesis (71). The upregulated CD47 protein on the surface of cancer cells can combine with signal regulatory protein-α, located on the surface of macrophages, in order to avoid phagocytosis. The expression of β2-microglobulin (a component of MHC class I molecules) in cancer cells directly protects them from phagocytosis, which is mediated by the inhibitory receptor leukocyte immunoglobulin-like receptor B1, whose expression is upregulated on the surface of macrophages (72). Dendritic cells (DCs) are specialized antigen-presenting cells that have a direct cytotoxic effect on tumor cells (73), but restraint stress compromises the suppressor function of regulatory T cells (Tregs) and alters DCs to contribute to intestinal inflammation (74). A short-term, stress-induced increase in IFN-γ, macrophage inflammatory protein-3α, TNF-α, MCP-1, IL-1α, IL-1β and IL-6 can enhance the immunization phase of cell-mediated immunity (75,76). Nevertheless, chronic stress can lead to a reduction in the number of cytokines, including IFN-γ, IL-2 and IL-12, due to the inhibitory effect of GC, NE and E, which have an important antitumor function (77,78). Chronic stress-induced neuroendocrine changes have been found to suppress the immune response, including NK cell cytotoxicity, phagocytosis, inflammatory cytokine production and cytotoxic T-cell activity, compromising the most important effectors of the immune response against tumors (66). It is worth highlighting that, as the largest organ and the body’s first line of defence, cutaneous cell-mediated immunity serves an important role in the elimination of immune-responsive tumors such as squamous cell carcinoma (24). Short-term stress not only enhances primary cutaneous immune responses, but also augments secondary or recall responses in the skin (79). In addition, the lymphatic system serves an important role in immune function, while also contributing to tumor cell invasion and metastasis. Studies have found that chronic stress can promote tumor cell dissemination by remodeling the lymph vasculature (80,81).

Effects of stress on the gut microbiota. The gut microbiota serves an important role in maintaining gut homeostasis (82). The most common bacterial phyla in the gut include the Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria, and the composition of the gut microbiota is highly sensitive to chronic stress (83,84). Furthermore, stress during the perinatal period can markedly influence the microbiota, leading to long-lasting immunological aberrations (85). It has been demonstrated that microbiota dysbiosis is closely associated with the occurrence, development and treatment of cancer (86,87). Certain bioactive substances of the microbiota metabolite production have also been found to be involved in carcinogenesis (88). The influence of the gut microbiota on tumorigenesis and development is mainly through several methods that include direct contact with the tumor (89), affecting the tumor cells by regulating body metabolism indirectly (90,91) and promoting tumor progression by regulating the immune system (92,93). Studies performed on a restraint stress mouse model suggested that commensal microbiota can affect the postnatal development of the HPA stress response (94), and that gut dysbiosis is associated with brain dysfunction and stress-related behavior, including anxiety and depression (95,96). It was shown that the absence of the gut microbiota enhanced anxiety-like behavior and neuroendocrine response to acute stress in rats (97). Chronic stress can not only cause an imbalance in and disorders of the gut microbiota, but also behavioral, cognitive and biochemical aberrations, which may be involved in the microbiotagut-brain axis. A study revealed that Lactobacillus helveticus NS8 can improve chronic restraint stress-induced behavioral and cognitive dysfunction (98). Rifaximin can alter the bacterial population in the ileum of rats and lead to a relative abundance of Lactobacillus, which can prevent intestinal abnormalities and visceral hyperalgesia in response to chronic psychological stress (99). A study showed that probiotic treatment attenuated the HPA response to acute stress (100). Notably, a recent study demonstrated that dietary bioactive compounds and probiotics could reduce the risk of colon cancer by shaping functional gut microbiota (101). In addition, resident gut bacteria can affect patient response to cancer immunotherapy, and maintaining a healthy gut flora could assist patients in their fight against cancer (102). Based on the aforementioned literature, we tentatively conclude that there may be an association of mutual causality between tumorigenesis and gut microbiota imbalance.

Effects of stress on inflammation. Approximately 25% of all cancer types are associated with chronic inflammations of broad origin (103). Just as stress can be divided into acute and chronic according to its beneficial or detrimental effects on the body, so can inflammation. It has been shown that chronic inflammatory processes affect all stages of tumor development, as well as the efficacy of therapy, particularly in gastric, hepatic and colorectal cancer (104,105). There are two major signaling pathways underlying cancer-related inflammation: The transcription factor nuclear factor-κB (NF-κB) and the signal transducer and activator of transcription 3 signaling pathways, which can be activated by the majority of cancer risk factors, including stress, diet, infectious agents and environmental pollutants (106). Studies have also revealed that inflammatory cells can be recruited by stress to tumor sites, increasing the formation of blood vessels (107). Meanwhile, stress-inducible inflammatory factors and genes, including IL-6, IL-8 and vascular endothelial growth factor, are increased in the circulation following stressor exposure. Stress can lead to metastatic invasion and metabolic syndrome by the activation of the HPA axis and SNS, respectively, which are characterized by an increased production of IL-6, TNF-α, llasminogen activator inhibitor-1 and metalloproteinase-2 and -9 (108). Reciprocally, adipose-derived IL-6 may further stimulate the HPA axis, forming a deleterious vicious cycle (109). Chronic psychological stress can induce vascular inflammation via the TNF-α and p38/c-Jun N-terminal kinase pathways and increase the expression of inflammatory molecules, including mRNA and proteins such as TNF-α, C-reactive protein, MCP-1, macrophage migration inhibitory factor and intercellular adhesion molecule-1 (110). Psychological stress-derived prolactin has been shown not only to induce IL-6 and IL-23 production by DCs, the former of which serve a critical role in altering the phenotypes of Tregs, but also to alter Treg properties, leading to intestinal inflammation (75). Chronic restraint stress has been shown to result in a marked decrease in CD4 T cell numbers and intracellular IFN-γ expression, while increasing IL-4 production. It was also found that, in chronically stressed mice, treatment with 4-methylhistamine (4-MeH) agonist was able to restore the immune response, particularly via the production of Th1 cytokines. Stimulation of the histamine 4 receptor with 4-MeH modulates the effects of chronic stress on the Th1/Th2 cytokine balance (111). The sympathetic and neuroendocrine responses to psychosocial stress have been shown to have a significant impact on cancer, partly through the regulation of inflammatory mediators (112). Psychological stress increases extracellular adenosine triphosphate (ATP), IL-1β and TNF-α in the hippocampus, and activates the inflammasomes via the release of ATP and the stimulation of the purinergic type 2X7 receptor (16). Inflammasomes are multiprotein complexes that operate as platforms for the activation of caspase-1 and can be categorized based on their main constituent as either NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain containing 4, NLRP6 or absent in melanoma 2. The activation of inflammasomes can lead to the conversion of inactive inflammation mediators to active ones (IL-1β and IL-18), and, subsequently, the active inflammation mediators are secreted to the cell exterior to modulate cell function in an autocrine or paracrine manner. This process may mechanistically explain the link between inflammasome activation and tumorigenesis, angiogenesis or metastasis (113,114). Furthermore, the external IL-1β can initiate self-reinforcing feedback loops to further perpetuate its existence through the IL-1R-MyD88-NF-κB pathway by inflammasome activators (115). Based on the aforementioned findings, the inhibition of inflammasomes or neutralization of their products can have profound effects on carcinogenesis and tumor progression. The stress hormones NE and Epi can enhance IL-8 expression and thereby mediate the effects of stress on the growth and metastasis of ovarian cancer (116). IL-8 gene silencing with liposomal small interfering RNA incorporated in 1,2-dioleoyl-sn-glycero-3-phosphocholine has been shown to decrease tumor growth and angiogenesis in ovarian cancer. In addition, the increase of pro-inflammatory cytokines has been associated with irritability, insomnia and fatigue, which, in turn, are associated with cancer (117) (Fig. 3).

Figure 3

Effects of stress-induced inflammation on tumorigenesis, invasion, angiogenesis and metastasis. Stress, particularly chronic stress, can not only lead to the activation of the HPA axis and SNS, but also activate inflammatory mediators in extrinsic and intrinsic pathways. The transcription factors NF-κB and STAT3 are the two major pathways linking cancer to inflammation, which can be activated by the majority of cancer risk factors, including stress, diet, infectious agents and environmental pollutants. Stress-induced cytokines and chemokines, including IL-8, IL-8, VEGF, MMP-2 and inflammasomes, have a damaging effect on DNA and can result in gene instability and mutations closely associated with cell transformation, invasion, angiogenesis and metastasis. Furthermore, there exists a vicious circle between the activation of the SNS and the HPA axis, and the secretion of IL-6. Specifically, the activation of SNS can increase the release of IL-6, while IL-6 can trigger the activation of the HPA axis and promote the release of CRH, ACTH and cortisol. HPA, hypothalamic-pituitary-adrenal; SNS, sympathetic nervous system; NF-κB, nuclear factor-κB; STAT3, signal transducer and activator of transcription 3; IL, interleukin; VEGF, vascular endothelial growth factor; MMP, metalloproteinase; CRH, corticotrophin-releasing hormone; ACTH, adrenocorticotropic hormone; NE, norepinephrine; Epi, epinephrine; GC, glucocorticoid; IL, interleukin; TNF-α, tumor necrosis factor-α; MMP, matrix metalloproteinase; MCP-1, monocyte chemoattractant protein-1.

Effects of stress on chemo-resistance (multidrug resistance). At present, chemotherapy is one of the main treatment strategies for malignant tumors. However, a number of chemotherapeutic approaches fail due to intrinsic or acquired drug resistance, particularly multidrug resistance. Multiple studies have focused on the mechanisms underlying multidrug resistance in cancer, including DNA damage repair (118) and abnormalities in the expression and function of transporters of the ATP-binding cassette superfamily. P-glycoprotein is the member of this mechanism that has been most frequently reported in association with tumor drug resistance, cancer stem cell, epithelial-mesenchymal transitions and hypoxia (119,120). A study on mice showed that psychological stress reduced the antitumor effects of chemotherapeutic drugs and induced chemo-resistance in breast cancer by upregulating multidrug resistance protein 1 via adrenergic stimulation (121). Stress hormones induced by restraint stress, including cortisol and epinephrine, can decrease the efficacy of paclitaxel in triple-negative breast cancer through the induction of DNA damage and ATR serine/threonine kinase and p21 expression (122). Adrenaline can induce chemo-resistance in HT-29 colon adenocarcinoma cells by upregulating the ATP binding cassette subfamily B member 1 gene expression via α2-adrenergic receptors (123), as well as cisplatin resistance through the activation of the NF-κB pathway and subsequent induction of miR-155 (124). One study showed that miR-155 is responsible for the drug resistance in breast cancer cells, by targeting fork-head box O3 (125). Meanwhile, injections of epinephrine or immobilization stress can counteract the antitumor effects of phosphatidylinositol-4,5-bisphosphate 3-kinase inhibitors on prostate cancer xenografts in mice (126). A dynamic network model of apoptosis regulation in prostate cancer indicated that psychological stress could trigger a synergism pattern switch in drug combination therapy (127). However, it is important to note that chemotherapy itself may generate or deteriorate psychological stress, which is known to be accompanied by chronic elevation of plasma catecholamine, conversely leading to chemo-resistance (121). Psychological stress has been found to attenuate the anti-angiogenic efficacy of sunitinib through the activation of β-adrenergic signaling and the promotion of tumor angiogenesis, which, however, can be improved by the use of β-blockers (39). Stress hormones have also been shown to promote resistance to epidermal growth factor receptor (EGFR) inhibitor in non-small cell lung cancer, which can be abrogated by combinations of β-blockers and EGFR tyrosine kinase inhibitors (53). Furthermore, certain gut microbiota, such as Escherichia coli strains, can decrease the efficacy of chemotherapeutic agent gemcitabine by metabolizing and deactivating the active form of the drug (128).

4. Conclusion

Stress is one of the fundamental survival mechanisms in nature; it begins with a stressor, leading to stress perception and subsequently resulting in a stress response. Stress can be divided into acute and chronic, mainly according to its respective salubrious or detrimental effects on the health of an individual. Several studies have demonstrated that stress, particularly chronic stress, serves an important role in tumorigenesis and metastasis. Relevant and effective animal models are essential for the study of the effects of stress on cancer and the underlying mechanisms. The induction of CMS or chronic restraint stress has been widely used for the development of experimental models. The stress-induced persistent activation of the HPA axis and SNS has been shown to result in a cascade reaction leading to cross-talk among the neuroendocrine system, the immune system, gut microbiota and inflammation. Further elucidation of the influence of the interactions among these factors in mediating stress-associated effects on tumorigenesis and metastasis is imperative. Novel approaches to the prevention and blockage of the harmful effects of stress on tumorigenesis and metastasis are required.

5. Practical perspectives

Stress and stress-associated disorders have become prevalent in modern societies due to the fast-paced nature of contemporary lifestyles. Due to their prevalence, such disorders are often concealed, which has detrimental effects to health. Studies have demonstrated that exposure to stress during critical periods in human development is sufficient for it to have severe, long-term consequences (35,129). In addition, chronic stress may lead to epigenetic heritable modifications, suggesting a possible propagation across generations (55). It is noteworthy that stress may exist universally during the diagnosis and subsequent treatment process in patients with cancer (130). On a positive note, it has been shown that effective intervention in cancer patients presenting with stress can improve their immune function and physical activity, and social support can modulate cancer-related pathways and improve the levels of certain biomarkers associated with a better prognosis and longer survival (131). Studies have also revealed that disturbances in mood, anxiety and irritability may precede the appearance of a medical disorder, and that stress-prone personalities or unfavorable coping mechanisms and negative emotional responses are associated with a higher incidence, poorer survival and higher mortality rates in patients with cancer (132). Dhabhar (11) proposed the stress spectrum model and concept, so as to reconcile the potentially beneficial effect of stress with the harmful; according to the stress spectrum model, one can keep fit by minimizing chronic stress, maximizing the resting zone of low/no stress and optimizing the acute stress response.

In the research findings discussed within the present review, it was demonstrated that effective stress prevention and management are plausible and imperative, and that the primary, secondary and tertiary prevention strategies should receive adequate recognition. In particular, moderate exercise, a healthy diet, high-quality sleep and emotional management are key aspects that can be improved in the lives of patients with stress-induced cancer, so that they benefit from them. Regular exercise can assist in keeping the short-term stress response well-oiled, finely-tuned and ready for fight-or-flight (11), as well as improving the physical and emotional well-being of an individual. Furthermore, regular exercise has also been shown to reduce the risk for breast cancer recurrence (133). The type, intensity, duration and frequency of exercise should be tailored to the constitution an individual. A number of traditional methods of exercise, including five-animal boxing, eight-section brocade, changing tendon exercise, meditation and yoga, may contribute to successful stress management.

The present review found that diet and gut microbiota play an important role in tumorigenesis and metastasis. Therefore, closer attention should be paid to dietary strategies, and particularly the maintenance of healthy-eating habits. Furthermore, developing effective microbial agents is necessary and promising with regard to the reduction of cancer risks. An association has been identified between lack of sleep or sleep disturbances such as insomnia, fatigue, obstructive sleep apnea and restless legs syndrome, and depression or anxiety (134). The latter are also common among cancer patients, and often comprise the symptoms that lead to the diagnosis of certain types of cancer (135). In addition, circadian cortisol rhythm disruptions serve as an important indicator and/or mediator of the deleterious effects of chronic stress (1). In view of this, future studies are warranted to better understand the mechanisms underlying the effects of stress on tumorigenesis and metastasis.

Funding

The present review was supported by the National Natural Science Foundation of China (grant nos. 81573764 and 81774095), the Program for Outstanding Medical Academic Leader, Shanghai Academic/Technology Research Leader (grant no. 16XD1403600), the Shanghai Rising-Star Program (grant no. 16QA1403700) and the Municipal Human Resources Development Program for Outstanding Leaders in Medical Disciplines in Shanghai (grant no. 2017BR031).

Availability of data and materials

Not applicable.

Authors’ contributions

QL and YW put forward the conception and design of the manuscript, and ZZZ and YW were major contributors in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Professor Junfeng Zhang (Duke University, Durham, NC, USA) for the revision of the original manuscript.

References

1 

Dhabhar FS and McEwen BS: Acute stress enhances while chronic stress suppresses cell-mediated immunity in vivo: A potential role for leukocyte trafficking. Brain Behav Immun. 11:286–306. 1997. View Article : Google Scholar

2 

Amin SN, El-Aidi AA, Ali MM, Attia YM and Rashed LA: Modification of hippocampal markers of synaptic plasticity by memantine in animal models of acute and repeated restraint stress: Implications for memory and behavior. Neuromolecular Med. 17:121–136. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Charmandari E, Tsigos C and Chrousos G: Endocrinology of the stress response. Annu Rev Physiol. 67:259–284. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Gunnar M and Quevedo K: The neurobiology of stress and development. Annu Rev Psychol. 58:145–173. 2007. View Article : Google Scholar

5 

Krizanova O, Babula P and Pacak K: Stress, catecholaminergic system and cancer. Stress. 19:419–428. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Hering D, Lachowska K and Schlaich M: Role of the sympathetic nervous system in stress-mediated cardiovascular disease. Curr Hypertens Rep. 17:802015. View Article : Google Scholar : PubMed/NCBI

7 

Dhabhar FS, McEwen BS and Spencer RL: Stress response, adrenal steroid receptor levels and corticosteroid-binding globulin levels - a comparison between Sprague-Dawley, Fischer 344 and Lewis rats. Brain Res. 616:89–98. 1993. View Article : Google Scholar : PubMed/NCBI

8 

Dhabhar FS, McEwen BS and Spencer RL: Adaptation to prolonged or repeated stress - comparison between rat strains showing intrinsic differences in reactivity to acute stress. Neuroendocrinology. 65:360–368. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Ambarish V, Chandrashekara S and Suresh KP: Moderate regular exercises reduce inflammatory response for physical stress. Indian J Physiol Pharmacol. 56:7–14. 2012.PubMed/NCBI

10 

Clague J and Bernstein L: Physical activity and cancer. Curr Oncol Rep. 14:550–558. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Dhabhar FS: Effects of stress on immune function: The good, the bad, and the beautiful. Immunol Res. 58:193–210. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Lagraauw HM, Kuiper J and Bot I: Acute and chronic psychological stress as risk factors for cardiovascular disease: Insights gained from epidemiological, clinical and experimental studies. Brain Behav Immun. 50:18–30. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Muffly LS, Hlubocky FJ, Khan N, Wroblewski K, Breitenbach K, Gomez J, McNeer JL, Stock W and Daugherty CK: Psychological morbidities in adolescent and young adult blood cancer patients during curative-intent therapy and early survivorship. Cancer. 122:954–961. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Cohen L, Cole SW, Sood AK, Prinsloo S, Kirschbaum C, Arevalo JM, Jennings NB, Scott S, Vence L, Wei Q, et al: Depressive symptoms and cortisol rhythmicity predict survival in patients with renal cell carcinoma: Role of inflammatory signaling. PLoS One. 7:e423242012. View Article : Google Scholar : PubMed/NCBI

15 

Shan T, Ma J, Ma Q, Guo K, Guo J, Li X, Li W, Liu J, Huang C, Wang F, et al: β2-AR-HIF-1α: A novel regulatory axis for stress-induced pancreatic tumor growth and angiogenesis. Curr Mol Med. 13:1023–1034. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Iwata M, Ota KT, Li XY, Sakaue F, Li N, Dutheil S, Banasr M, Duric V, Yamanashi T, Kaneko K, et al: Psychological stress activates the inflammasome via release of adenosine triphosphate and stimulation of the purinergic type 2X7 receptor. Biol Psychiatry. 80:12–22. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Agarwal SK and Marshall GD Jr: Stress effects on immunity and its application to clinical immunology. Clin Exp Allergy. 31:25–31. 2001. View Article : Google Scholar : PubMed/NCBI

18 

Yin X, Guven N and Dietis N: Stress-based animal models of depression: Do we actually know what we are doing? Brain Res. 1652:30–42. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Heinrichs SC and Koob GF: Application of experimental stressors in laboratory rodents. Curr Protoc Neurosci. Chapter 8: Unit8.4. 2006. View Article : Google Scholar

20 

Zorzet S, Perissin L, Rapozzi V and Giraldi T: Restraint stress reduces the antitumor efficacy of cyclophosphamide in tumor-bearing mice. Brain Behav Immun. 12:23–33. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Nukina H, Sudo N, Aiba Y, Oyama N, Koga Y and Kubo C: Restraint stress elevates the plasma interleukin-6 levels in germ-free mice. J Neuroimmunol. 115:46–52. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Willner P: The validity of animal models of predisposition to depression. Behav Pharmacol. 13:169–188. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Glaser R and Kiecolt-Glaser JK: Stress-induced immune dysfunction: Implications for health. Nat Rev Immunol. 5:243–251. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Dhabhar FS, Saul AN, Daugherty C, Holmes TH, Bouley DM and Oberyszyn TM: Short-term stress enhances cellular immunity and increases early resistance to squamous cell carcinoma. Brain Behav Immun. 24:127–137. 2010. View Article : Google Scholar

25 

Eng JW, Kokolus KM, Reed CB, Hylander BL, Ma WW and Repasky EA: A nervous tumor microenvironment: The impact of adrenergic stress on cancer cells, immunosuppression, and immunotherapeutic response. Cancer Immunol Immunother. 63:1115–1128. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Frick LR, Rapanelli M, Bussmann UA, Klecha AJ, Arcos ML, Genaro AM and Cremaschi GA: Involvement of thyroid hormones in the alterations of T-cell immunity and tumor progression induced by chronic stress. Biol Psychiatry. 65:935–942. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Volpi S, Rabadan-Diehl C and Aguilera G: Vasopressinergic regulation of the hypothalamic pituitary adrenal axis and stress adaptation. Stress. 7:75–83. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Meltzer HY, Lowy MT and Koenig JI: The hypothalamic-pituitary-adrenal axis in depression. Adv Biochem Psychopharmacol. 43:165–182. 1987.PubMed/NCBI

29 

Chrousos GP: Ultradian, circadian, and stress-related hypothalamic-pituitary-adrenal axis activity - a dynamic digital-to-analog modulation. Endocrinology. 139:437–440. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Sephton S and Spiegel D: Circadian disruption in cancer: A neuroendocrine-immune pathway from stress to disease? Brain Behav Immun. 17:321–328. 2003. View Article : Google Scholar : PubMed/NCBI

31 

McEwen BS: Physiology and neurobiology of stress and adaptation: Central role of the brain. Physiol Rev. 87:873–904. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Moreno-Smith M, Lutgendorf SK and Sood AK: Impact of stress on cancer metastasis. Future Oncol. 6:1863–1881. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Gündisch S, Boeckeler E, Behrends U, Amtmann E, Ehrhardt H and Jeremias I: Glucocorticoids augment survival and proliferation of tumor cells. Anticancer Res. 32:4251–4261. 2012.PubMed/NCBI

34 

Wang HM, Liao ZX, Komaki R, Welsh JW, O’Reilly MS, Chang JY, Zhuang Y, Levy LB, Lu C and Gomez DR: Improved survival outcomes with the incidental use of beta-blockers among patients with non-small-cell lung cancer treated with definitive radiation therapy. Ann Oncol. 24:1312–1319. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Wulsin AC, Wick-Carlson D, Packard BA, Morano R and Herman JP: Adolescent chronic stress causes hypothalamo-pituitary-adrenocortical hypo-responsiveness and depression-like behavior in adult female rats. Psychoneuroendocrinology. 65:109–117. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Bortolato B, Hyphantis TN, Valpione S, Perini G, Maes M, Morris G, Kubera M, Köhler CA, Fernandes BS, Stubbs B, et al: Depression in cancer: The many biobehavioral pathways driving tumor progression. Cancer Treat Rev. 52:58–70. 2017. View Article : Google Scholar

37 

Zhao L, Xu J, Liang F, Li A, Zhang Y and Sun J: Effect of chronic psychological stress on liver metastasis of colon cancer in mice. PLoS One. 10:e01399782015. View Article : Google Scholar : PubMed/NCBI

38 

Xie H, Li C, He Y, Griffin R, Ye Q and Li L: Chronic stress promotes oral cancer growth and angiogenesis with increased circulating catecholamine and glucocorticoid levels in a mouse model. Oral Oncol. 51:991–997. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Liu J, Deng GH, Zhang J, Wang Y, Xia XY, Luo XM, Deng YT, He SS, Mao YY, Peng XC, et al: The effect of chronic stress on anti-angiogenesis of sunitinib in colorectal cancer models. Psychoneuroendocrinology. 52:130–142. 2015. View Article : Google Scholar

40 

Elefteriou F: Chronic stress, sympathetic activation and skeletal metastasis of breast cancer cells. Bonekey Rep. 4:6932015. View Article : Google Scholar : PubMed/NCBI

41 

Moreno-Smith M, Lu C, Shahzad MM, Pena GN, Allen JK, Stone RL, Mangala LS, Han HD, Kim HS, Farley D, et al: Dopamine blocks stress-mediated ovarian carcinoma growth. Clin Cancer Res. 17:3649–3659. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Moreno-Smith M, Lee SJ, Lu C, Nagaraja AS, He G, Rupaimoole R, Han HD, Jennings NB, Roh JW, Nishimura M, et al: Biologic effects of dopamine on tumor vasculature in ovarian carcinoma. Neoplasia. 15:502–510. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Borcherding DC, Tong W, Hugo ER, Barnard DF, Fox S, LaSance K, Shaughnessy E and Ben-Jonathan N: Expression and therapeutic targeting of dopamine receptor-1 (D1R) in breast cancer. Oncogene. 35:3103–3113. 2016. View Article : Google Scholar

44 

Peters MA, Walenkamp AM, Kema IP, Meijer C, de Vries EG and Oosting SF: Dopamine and serotonin regulate tumor behavior by affecting angiogenesis. Drug Resist Updat. 17:96–104. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Barbieri A, Palma G, Rosati A, Giudice A, Falco A, Petrillo A, Petrillo M, Bimonte S, Di Benedetto M, Esposito G, et al: Role of endothelial nitric oxide synthase (eNOS) in chronic stress-promoted tumour growth. J Cell Mol Med. 16:920–926. 2012. View Article : Google Scholar

46 

Partecke LI, Speerforck S, Käding A, Seubert F, Kühn S, Lorenz E, Schwandke S, Sendler M, Kessler W, Trung DN, et al: Chronic stress increases experimental pancreatic cancer growth, reduces survival and can be antagonised by beta-adrenergic receptor blockade. Pancreatology. 16:423–433. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Feng Z, Liu L, Zhang C, Zheng T, Wang J, Lin M, Zhao Y, Wang X, Levine AJ and Hu W: Chronic restraint stress attenuates p53 function and promotes tumorigenesis. Proc Natl Acad Sci USA. 109:7013–7018. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Gao J, Gao G, Zhang Y and Wang F: Proteomic analysis of human epithelial ovarian cancer xenografts in immunodeficient mice exposed to chronic psychological stress. Sci China Life Sci. 54:112–120. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Turnbull AV and Rivier CL: Regulation of the hypothalamic-pituitary-adrenal axis by cytokines: Actions and mechanisms of action. Physiol Rev. 79:1–71. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Schwab CL, Fan R, Zheng Q, Myers LP, Hebert P and Pruett SB: Modeling and predicting stress-induced immunosuppression in mice using blood parameters. Toxicol Sci. 83:101–113. 2005. View Article : Google Scholar

51 

Dhabhar FS, Malarkey WB, Neri E and McEwen BS: Stress-induced redistribution of immune cells - from barracks to boulevards to battlefields: A tale of three hormones - Curt Richter Award winner. Psychoneuroendocrinology. 37:1345–1368. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Barbieri A, Bimonte S, Palma G, Luciano A, Rea D, Giudice A, Scognamiglio G, La Mantia E, Franco R, Perdonà S, et al: The stress hormone norepinephrine increases migration of prostate cancer cells in vitro and in vivo. Int J Oncol. 47:527–534. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Nilsson MB, Sun H, Diao L, Tong P, Liu D, Li L, Fan Y, Poteete A, Lim SO, Howells K, et al: Stress hormones promote EGFR inhibitor resistance in NSCLC: Implications for combinations with β-blockers. Sci Transl Med. 9:92017. View Article : Google Scholar

54 

Kim-Fuchs C, Le CP, Pimentel MA, Shackleford D, Ferrari D, Angst E, Hollande F and Sloan EK: Chronic stress accelerates pancreatic cancer growth and invasion: A critical role for beta-adrenergic signaling in the pancreatic microenvironment. Brain Behav Immun. 40:40–47. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Nagaraja AS, Sadaoui NC, Dorniak PL, Lutgendorf SK and Sood AK: SnapShot: Stress and Disease. Cell Metab. 23:388–388.e1. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Dhabhar FS: Enhancing versus suppressive effects of stress on immune function: Implications for immunoprotection and immunopathology. Neuroimmunomodulation. 16:300–317. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Glaser R, MacCallum RC, Laskowski BF, Malarkey WB, Sheridan JF and Kiecolt-Glaser JK: Evidence for a shift in the Th-1 to Th-2 cytokine response associated with chronic stress and aging. J Gerontol A Biol Sci Med Sci. 56:M477–M482. 2001. View Article : Google Scholar : PubMed/NCBI

58 

Ochoa CE, Mirabolfathinejad SG, Ruiz VA, Evans SE, Gagea M, Evans CM, Dickey BF and Moghaddam SJ: Interleukin 6, but not T helper 2 cytokines, promotes lung carcinogenesis. Cancer Prev Res (Phila). 4:51–64. 2011. View Article : Google Scholar

59 

Divyashree S, Sarjan HN and Yajurvedi HN: Effects of long-term chronic stress on the lymphoid organs and blood l. Can J Zool. 94:137–143. 2015. View Article : Google Scholar

60 

Frick LR, Arcos ML, Rapanelli M, Zappia MP, Brocco M, Mongini C, Genaro AM and Cremaschi GA: Chronic restraint stress impairs T-cell immunity and promotes tumor progression in mice. Stress. 12:134–143. 2009. View Article : Google Scholar

61 

Li H, Zhao J, Chen M, Tan Y, Yang X, Caudle Y and Yin D: Toll-like receptor 9 is required for chronic stress-induced immune suppression. Neuroimmunomodulation. 21:1–7. 2014. View Article : Google Scholar :

62 

Lakshmi Narendra B, Eshvendar Reddy K, Shantikumar S and Ramakrishna S: Immune system: a double-edged sword in cancer. Inflamm Res. 62:823–834. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Hunzeker JT, Elftman MD, Mellinger JC, Princiotta MF, Bonneau RH, Truckenmiller ME and Norbury CC: A marked reduction in priming of cytotoxic CD8+ T cells mediated by stress-induced glucocorticoids involves multiple deficiencies in cross-presentation by dendritic cells. J Immunol. 186:183–194. 2011. View Article : Google Scholar

64 

Kour K and Bani S: Augmentation of immune response by chicoric acid through the modulation of CD28/CTLA-4 and Th1 pathway in chronically stressed mice. Neuropharmacology. 60:852–860. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Lutgendorf SK, Sood AK, Anderson B, McGinn S, Maiseri H, Dao M, Sorosky JI, De Geest K, Ritchie J and Lubaroff DM: Social support, psychological distress, and natural killer cell activity in ovarian cancer. J Clin Oncol. 23:7105–7113. 2005. View Article : Google Scholar : PubMed/NCBI

66 

Reiche EM, Nunes SO and Morimoto HK: Stress, depression, the immune system, and cancer. Lancet Oncol. 5:617–625. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Nakatani Y, Amano T and Takeda H: Corticosterone suppresses the proliferation of RAW264.7 macrophage cells via glucocor-ticoid, but not mineralocorticoid, receptor. Biol Pharm Bull. 36:592–601. 2013. View Article : Google Scholar

68 

Sloan EK, Priceman SJ, Cox BF, Yu S, Pimentel MA, Tangkanangnukul V, Arevalo JM, Morizono K, Karanikolas BD, Wu L, et al: The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 70:7042–7052. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Schmieder A, Michel J, Schönhaar K, Goerdt S and Schledzewski K: Differentiation and gene expression profile of tumor-associated macrophages. Semin Cancer Biol. 22:289–297. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Armaiz-Pena GN, Gonzalez-Villasana V, Nagaraja AS, Rodriguez-Aguayo C, Sadaoui NC, Stone RL, Matsuo K, Dalton HJ, Previs RA, Jennings NB, et al: Adrenergic regulation of monocyte chemotactic protein 1 leads to enhanced macrophage recruitment and ovarian carcinoma growth. Oncotarget. 6:4266–4273. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Roberts DD, Miller TW, Rogers NM, Yao M and Isenberg JS: The matricellular protein thrombospondin-1 globally regulates cardiovascular function and responses to stress via CD47. Matrix Biol. 31:162–169. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, George BM, Markovic M, Ring NG, Tsai JM, et al: Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 19:76–84. 2018. View Article : Google Scholar :

73 

Hanke N, Alizadeh D, Katsanis E and Larmonier N: Dendritic cell tumor killing activity and its potential applications in cancer immunotherapy. Crit Rev Immunol. 33:1–21. 2013. View Article : Google Scholar : PubMed/NCBI

74 

Wu W, Sun M, Zhang HP, Chen T, Wu R, Liu C, Yang G, Geng XR, Feng BS, Liu Z, et al: Prolactin mediates psychological stress-induced dysfunction of regulatory T cells to facilitate intestinal inflammation. Gut. 63:1883–1892. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Dhabhar FS, Satoskar AR, Bluethmann H, David JR and McEwen BS: Stress-induced enhancement of skin immune function: A role for gamma interferon. Proc Natl Acad Sci USA. 97:2846–2851. 2000. View Article : Google Scholar : PubMed/NCBI

76 

Dhabhar FS and Viswanathan K: Short-term stress experienced at time of immunization induces a long-lasting increase in immunologic memory. Am J Physiol Regul Integr Comp Physiol. 289:R738–R744. 2005. View Article : Google Scholar : PubMed/NCBI

77 

Levi B, Benish M, Goldfarb Y, Sorski L, Melamed R, Rosenne E and Ben-Eliyahu S: Continuous stress disrupts immunostimulatory effects of IL-12. Brain Behav Immun. 25:727–735. 2011. View Article : Google Scholar : PubMed/NCBI

78 

Lopes RP, Grassi-Oliveira R, de Almeida LR, Stein LM, Luz C, Teixeira AL and Bauer ME: Neuroimmunoendocrine interactions in patients with recurrent major depression, increased early life stress and long-standing posttraumatic stress disorder symptoms. Neuroimmunomodulation. 19:33–42. 2012. View Article : Google Scholar

79 

Dhabhar FS and McEwen BS: Stress-induced enhancement of antigen-specific cell-mediated immunity. J Immunol. 156:2608–2615. 1996.PubMed/NCBI

80 

Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB and Achen MG: Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat Rev Cancer. 14:159–172. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Le CP, Nowell CJ, Kim-Fuchs C, Botteri E, Hiller JG, Ismail H, Pimentel MA, Chai MG, Karnezis T, Rotmensz N, et al: Chronic stress in mice remodels lymph vasculature to promote tumour cell dissemination. Nat Commun. 7:106342016. View Article : Google Scholar : PubMed/NCBI

82 

Boleij A and Tjalsma H: Gut bacteria in health and disease: A survey on the interface between intestinal microbiology and colorectal cancer. Biol Rev Camb Philos Soc. 87:701–730. 2012. View Article : Google Scholar : PubMed/NCBI

83 

O’Toole PW: Gut microbiota and aging. Science. 350:1214–1215. 2015. View Article : Google Scholar

84 

Rook GAW, Raison CL and Lowry CA: Microbiota, immuno-regulatory old friends and psychiatric disorders. Adv Exp Med Biol. 817:319–356. 2014. View Article : Google Scholar

85 

Penders J, Gerhold K, Stobberingh EE, Thijs C, Zimmermann K, Lau S and Hamelmann E: Establishment of the intestinal microbiota and its role for atopic dermatitis in early childhood. J Allergy Clin Immunol. 132:601–607.e8. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Gagnière J, Raisch J, Veziant J, Barnich N, Bonnet R, Buc E, Bringer MA, Pezet D and Bonnet M: Gut microbiota imbalance and colorectal cancer. World J Gastroenterol. 22:501–518. 2016. View Article : Google Scholar : PubMed/NCBI

87 

Paul B, Barnes S, Demark-Wahnefried W, Morrow C, Salvador C, Skibola C and Tollefsbol TO: Influences of diet and the gut microbiome on epigenetic modulation in cancer and other diseases. Clin Epigenetics. 7:1122015. View Article : Google Scholar : PubMed/NCBI

88 

Berni Canani R, Di Costanzo M and Leone L: The epigenetic effects of butyrate: Potential therapeutic implications for clinical practice. Clin Epigenetics. 4:42012. View Article : Google Scholar : PubMed/NCBI

89 

Yamamoto ML, Maier I, Dang AT, Berry D, Liu J, Ruegger PM, Yang JI, Soto PA, Presley LL, Reliene R, et al: Intestinal bacteria modify lymphoma incidence and latency by affecting systemic inflammatory state, oxidative stress, and leukocyte genotoxicity. Cancer Res. 73:4222–4232. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, et al: Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 461:1282–1286. 2009. View Article : Google Scholar : PubMed/NCBI

91 

Garrett WS: Cancer and the microbiota. Science. 348:80–86. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al: Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 342:967–970. 2013. View Article : Google Scholar : PubMed/NCBI

93 

Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al: Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 350:1079–1084. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, Kubo C and Koga Y: Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 558:263–275. 2004. View Article : Google Scholar : PubMed/NCBI

95 

Luna RA and Foster JA: Gut brain axis: Diet microbiota interactions and implications for modulation of anxiety and depression. Curr Opin Biotechnol. 32:35–41. 2015. View Article : Google Scholar

96 

Naseribafrouei A, Hestad K, Avershina E, Sekelja M, Linløkken A, Wilson R and Rudi K: Correlation between the human fecal microbiota and depression. Neurogastroenterol Motil. 26:1155–1162. 2014. View Article : Google Scholar : PubMed/NCBI

97 

Crumeyrolle-Arias M, Jaglin M, Bruneau A, Vancassel S, Cardona A, Daugé V, Naudon L and Rabot S: Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology. 42:207–217. 2014. View Article : Google Scholar : PubMed/NCBI

98 

Liang S, Wang T, Hu X, Luo J, Li W, Wu X, Duan Y and Jin F: Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience. 310:561–577. 2015. View Article : Google Scholar : PubMed/NCBI

99 

Xu D, Gao J, Gillilland M III, Wu X, Song I, Kao JY and Owyang C: Rifaximin alters intestinal bacteria and prevents stress-induced gut inflammation and visceral hyperalgesia in rats. Gastroenterology. 146:484–96.e4. 2014. View Article : Google Scholar :

100 

Ait-Belgnaoui A, Durand H, Cartier C, Chaumaz G, Eutamene H, Ferrier L, Houdeau E, Fioramonti J, Bueno L and Theodorou V: Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology. 37:1885–1895. 2012. View Article : Google Scholar : PubMed/NCBI

101 

Seidel DV, Azcárate-Peril MA, Chapkin RS and Turner ND: Shaping functional gut microbiota using dietary bioactives to reduce colon cancer risk. Semin Cancer Biol. 46:191–204. 2017. View Article : Google Scholar : PubMed/NCBI

102 

Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al: Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 359:97–103. 2018. View Article : Google Scholar

103 

Balkwill FR and Mantovani A: Cancer-related inflammation: Common themes and therapeutic opportunities. Semin Cancer Biol. 22:33–40. 2012. View Article : Google Scholar : PubMed/NCBI

104 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

105 

Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C and Flavell RA: Inflammation-induced cancer: Crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 13:759–771. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Aggarwal BB, Vijayalekshmi RV and Sung B: Targeting inflammatory pathways for prevention and therapy of cancer: short-term friend, long-term foe. Clin Cancer Res. 15:425–430. 2009. View Article : Google Scholar : PubMed/NCBI

107 

Thaker PH, Han LY, Kamat AA, Arevalo JM, Takahashi R, Lu C, Jennings NB, Armaiz-Pena G, Bankson JA, Ravoori M, et al: Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat Med. 12:939–944. 2006. View Article : Google Scholar : PubMed/NCBI

108 

Armaiz-Pena GN, Cole SW, Lutgendorf SK and Sood AK: Neuroendocrine influences on cancer progression. Brain Behav Immun. 30(Suppl): S19–S25. 2013. View Article : Google Scholar

109 

Kyrou I, Tsigos C, Seedorf K and Ferré P: Stress hormones: Physiological stress and regulation of metabolism. Curr Opin Pharmacol. 9:787–793. 2009. View Article : Google Scholar : PubMed/NCBI

110 

Lu XT, Liu YF, Zhao L, Li WJ, Yang RX, Yan FF, Zhao YX and Jiang F: Chronic psychological stress induces vascular inflammation in rabbits. Stress. 16:87–98. 2013. View Article : Google Scholar

111 

Ahmad SF, Zoheir KM, Ansari MA, Korashy HM, Bakheet SA, Ashour AE and Attia SM: Stimulation of the histamine 4 receptor with 4-methylhistamine modulates the effects of chronic stress on the Th1/Th2 cytokine balance. Immunobiology. 220:341–349. 2015. View Article : Google Scholar

112 

Powell ND, Tarr AJ and Sheridan JF: Psychosocial stress and inflammation in cancer. Brain Behav Immun. 30(Suppl): S41–S47. 2013. View Article : Google Scholar

113 

Zitvogel L, Kepp O, Galluzzi L and Kroemer G: Inflammasomes in carcinogenesis and anticancer immune responses. Nat Immunol. 13:343–351. 2012. View Article : Google Scholar : PubMed/NCBI

114 

Müzes G and Sipos F: Inflammasome, inflammation and cancer: An interrelated pathobiological triad. Curr Drug Targets. 16:249–257. 2015. View Article : Google Scholar

115 

Dunn JH, Ellis LZ and Fujita M: Inflammasomes as molecular mediators of inflammation and cancer: Potential role in melanoma. Cancer Lett. 314:24–33. 2012. View Article : Google Scholar

116 

Shahzad MM, Arevalo JM, Armaiz-Pena GN, Lu C, Stone RL, Moreno-Smith M, Nishimura M, Lee JW, Jennings NB, Bottsford-Miller J, et al: Stress effects on FosB- and interleukin-8 (IL8)-driven ovarian cancer growth and metastasis. J Biol Chem. 285:35462–35470. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Cosci F, Fava GA and Sonino N: Mood and anxiety disorders as early manifestations of medical illness: A systematic review. Psychother Psychosom. 84:22–29. 2015. View Article : Google Scholar

118 

Casorelli I, Bossa C and Bignami M: DNA damage and repair in human cancer: Molecular mechanisms and contribution to therapy-related leukemias. Int J Environ Res Public Health. 9:2636–2657. 2012. View Article : Google Scholar : PubMed/NCBI

119 

Higgins CF: Multiple molecular mechanisms for multidrug resistance transporters. Nature. 446:749–757. 2007. View Article : Google Scholar : PubMed/NCBI

120 

Abraham J, Salama NN and Azab AK: The role of P-glycoprotein in drug resistance in multiple myeloma. Leuk Lymphoma. 56:26–33. 2015. View Article : Google Scholar

121 

Su F, Ouyang N, Zhu P, Ouyang N, Jia W, Gong C, Ma X, Xu H and Song E: Psychological stress induces chemoresistance in breast cancer by upregulating mdr1. Biochem Biophys Res Commun. 329:888–897. 2005. View Article : Google Scholar : PubMed/NCBI

122 

Reeder A, Attar M, Nazario L, Bathula C, Zhang A, Hochbaum D, Roy E, Cooper KL, Oesterreich S, Davidson NE, et al: Stress hormones reduce the efficacy of paclitaxel in triple negative breast cancer through induction of DNA damage. Br J Cancer. 112:1461–1470. 2015. View Article : Google Scholar : PubMed/NCBI

123 

Yao H, Duan Z, Wang M, Awonuga AO, Rappolee D and Xie Y: Adrenaline induces chemoresistance in HT-29 colon adenocar-cinoma cells. Cancer Genet Cytogenet. 190:81–87. 2009. View Article : Google Scholar : PubMed/NCBI

124 

Pu J, Bai D, Yang X, Lu X, Xu L and Lu J: Adrenaline promotes cell proliferation and increases chemoresistance in colon cancer HT29 cells through induction of miR-155. Biochem Biophys Res Commun. 428:210–215. 2012. View Article : Google Scholar : PubMed/NCBI

125 

Kong W, He L, Coppola M, Guo J, Esposito NN, Coppola D and Cheng JQ: MicroRNA-155 regulates cell survival, growth, and chemosensitivity by targeting FOXO3a in breast cancer. J Biol Chem. 285:17869–17879. 2010. View Article : Google Scholar : PubMed/NCBI

126 

Hassan S, Karpova Y, Baiz D, Yancey D, Pullikuth A, Flores A, Register T, Cline JM, D’Agostino R Jr, Danial N, et al: Behavioral stress accelerates prostate cancer development in mice. J Clin Invest. 123:874–886. 2013.PubMed/NCBI

127 

Sun X, Bao J, Nelson KC, Li KC, Kulik G and Zhou X: Systems modeling of anti-apoptotic pathways in prostate cancer: Psychological stress triggers a synergism pattern switch in drug combination therapy. PLOS Comput Biol. 9:e10033582013. View Article : Google Scholar : PubMed/NCBI

128 

Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, Gavert N, Zwang Y, Cooper ZA, Shee K, et al: Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 357:1156–1160. 2017. View Article : Google Scholar : PubMed/NCBI

129 

van Bodegom M, Homberg JR and Henckens MJAG: Modulation of the hypothalamic-pituitary-adrenal axis by early life stress exposure. Front Cell Neurosci. 11:872017. View Article : Google Scholar : PubMed/NCBI

130 

Antoni MH: Psychosocial intervention effects on adaptation, disease course and biobehavioral processes in cancer. Brain Behav Immun. 30(Suppl): S88–S98. 2013. View Article : Google Scholar

131 

Saxton JM, Scott EJ, Daley AJ, Woodroofe M, Mutrie N, Crank H, Powers HJ and Coleman RE: Effects of an exercise and hypocaloric healthy eating intervention on indices of psychological health status, hypothalamic-pituitary-adrenal axis regulation and immune function after early-stage breast cancer: A randomised controlled trial. Breast Cancer Res. 16:R392014. View Article : Google Scholar : PubMed/NCBI

132 

Chida Y, Hamer M, Wardle J and Steptoe A: Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat Clin Pract Oncol. 5:466–475. 2008. View Article : Google Scholar : PubMed/NCBI

133 

Dieli-Conwright CM and Orozco BZ: Exercise after breast cancer treatment: Current perspectives. Breast Cancer (Dove Med Press). 7:353–362. 2015.

134 

Nota JA and Coles ME: Shorter sleep duration and longer sleep onset latency are related to difficulty disengaging attention from negative emotional images in individuals with elevated transdiagnostic repetitive negative thinking. J Behav Ther Exp Psychiatry. 58:114–122. 2018. View Article : Google Scholar

135 

Guzman-Marin R and Avidan AY: Sleep disorders in patients with cancer. J Community Support Oncol. 13:148–155. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 53 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Z, Wang Y and Li Q: Mechanisms underlying the effects of stress on tumorigenesis and metastasis (Review). Int J Oncol 53: 2332-2342, 2018
APA
Zhang, Z., Wang, Y., & Li, Q. (2018). Mechanisms underlying the effects of stress on tumorigenesis and metastasis (Review). International Journal of Oncology, 53, 2332-2342. https://doi.org/10.3892/ijo.2018.4570
MLA
Zhang, Z., Wang, Y., Li, Q."Mechanisms underlying the effects of stress on tumorigenesis and metastasis (Review)". International Journal of Oncology 53.6 (2018): 2332-2342.
Chicago
Zhang, Z., Wang, Y., Li, Q."Mechanisms underlying the effects of stress on tumorigenesis and metastasis (Review)". International Journal of Oncology 53, no. 6 (2018): 2332-2342. https://doi.org/10.3892/ijo.2018.4570