Hostname: page-component-7c8c6479df-27gpq Total loading time: 0 Render date: 2024-03-29T12:11:44.851Z Has data issue: false hasContentIssue false

New insights into adipose tissue atrophy in cancer cachexia

Symposium on ‘Frontiers in adipose tissue biology’

Published online by Cambridge University Press:  01 September 2009

Chen Bing*
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Liverpool L69 3GA, UK
Paul Trayhurn
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Liverpool L69 3GA, UK
*
*Corresponding author: Dr Chen Bing, fax +44 151 7065802, email bing@liverpool.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Profound loss of adipose and other tissues is a hallmark of cancer cachexia, a debilitating condition associated with increased morbidity and mortality. Fat loss cannot be attributable to reduced appetite alone as it precedes the onset of anorexia and is much more severe in experimental models of cachexia than in food restriction. Morphological examination has shown marked remodelling of adipose tissue in cancer cachexia. It is characterised by the tissue containing shrunken adipocytes with a major reduction in cell size and increased fibrosis in the tissue matrix. The ultrastructure of ‘slimmed’ adipocytes has revealed severe delipidation and modifications in cell membrane conformation. Although the molecular mechanisms remain to be established, evidence suggests that altered adipocyte metabolism may lead to adipose atrophy in cancer cachexia. Increased lipolysis appears to be a key factor underlying fat loss, while inhibition of adipocyte development and lipid deposition may also contribute. Both tumour and host-derived factors are implicated in adipose atrophy. Zinc-α2-glycoprotein (ZAG), which is overexpressed by certain malignant tumours, has been identified as a novel adipokine. ZAG transcripts and protein expression in adipose tissue are up regulated in cancer cachexia but reduced with adipose tissue expansion in obesity. Studies in vitro demonstrate that recombinant ZAG stimulates lipolysis. ZAG may therefore act locally, as well as systemically, to promote lipid mobilisation in cancer cachexia. Further elucidation of ZAG function in adipose tissue may lead to novel targets for preventing adipose atrophy in malignancy.

Type
Research Article
Copyright
Copyright © The Authors 2009

Abbreviations:
UCP

uncoupling protein

ZAG

zinc-α2-glycoprotein

Cancer cachexia

Cachexia is derived from the Greek words ‘kakos’ and ‘hexis’, meaning ‘bad condition’. It is a complex metabolic syndrome that comprises weight loss with reductions in skeletal muscle and adipose tissue mass, anorexia and weakness(Reference Morley, Thomas and Wilson1). It usually occurs in chronic diseases such as cancer, chronic obstructive pulmonary disease, chronic heart failure and end-stage renal failure(Reference Morley, Thomas and Wilson1). Cachexia not only markedly impairs the quality of life but is associated with increased morbidity and mortality.

Most patients with cancer develop cachexia at some point during the course of their disease and approximately half all patients with cancer have weight loss at diagnosis(Reference Tisdale2). Clinically, cachexia should be suspected if involuntary weight loss of >5% premorbid weight occurs within a 6-month period(Reference Morley, Thomas and Wilson1). The frequency of weight loss varies with the type of malignancy, being more common and severe in patients with cancers of the gastrointestinal tract, prostate and lung(Reference Tisdale3). Cachexia has a detrimental effect on cancer treatment as a result of, for example, poor responses to chemotherapy(Reference Slaviero, Read and Clarke4). Weight loss is also a prognostic indicator of decreased survival in patients with cancer(Reference Fearon, Voss and Hustead5, Reference Deans, Wigmore and de Beaux6). It is considered that ⩽20% of all cancer deaths are directly attributable to cachexia(Reference Tisdale2, Reference Skipworth, Stewart and Dejong7).

Evidence is accumulating that cancer cachexia arises from multiple metabolic alterations, such as reduced appetite, increased energy expenditure and tissue breakdown. The main tissues that are affected during the development of cachexia are skeletal muscle and white adipose tissue. The mechanisms of muscle wasting have been the focus of intensive research, with the demonstration of reduced protein synthesis and enhanced proteolysis in experimental models of cachexia and in patients with cancer cachexia(Reference Tisdale3, Reference Argiles, Busquets and Lopez-Soriano8). Although profound loss of adipose tissue is a hallmark of cancer cachexia, much less is known of the underlying cellular and molecular mechanisms. A better understanding of fat depletion in malignancy is crucial for the development of effective treatments for the syndrome. The present article reviews studies on the mechanisms and potential mediators of adipose atrophy in cancer cachexia.

Adipose tissue in metabolic health

It is well documented that adipose tissue plays an important metabolic role by storing TAG in periods when energy input exceeds expenditure and releasing NEFA during energy deprivation(Reference Spiegelman and Flier9). As the largest energy reserve in the body, adipose tissue has a major impact on energy flux, plasma lipid levels and glucose uptake. There is compelling evidence that alterations in adipose tissue mass and metabolism have a major impact on whole-body energy homeostasis(Reference Bluher10). It has been shown that too little fat in lipodystrophy, as with too much fat in obesity, is a major risk for insulin resistance, dyslipidaemia and vascular diseases(Reference Rosen and Spiegelman11).

In addition to its primary role as a fuel reservoir, white adipose tissue has been affirmed as a major endocrine organ, since the tissue synthesises and secretes an array of hormones and proteins, termed adipokines(Reference Trayhurn and Wood12). Adipose tissue has extensive cross talk with other organs including the brain, liver and skeletal muscle through these adipokines. Over the last decade a growing number of adipokines have been identified, such as leptin, adiponectin, TNFα, visfatin and chemerin, which act locally in an autocrine and/or paracrine manner and/or as endocrine signals to modulate appetite, nutrient metabolism, insulin sensitivity, inflammation and adipose tissue development(Reference Mohamed-Ali, Pinkney and Coppack13Reference Trayhurn and Bing17).

Adipose atrophy in cachexia

Extensive loss of adipose tissue is a prominent feature of cancer cachexia. Although it is not clear whether there are site differences in fat loss, a study using computed tomography scanning has revealed that patients with cancer cachexia who have gastrointestinal carcinoma have a smaller visceral adipose tissue area than control subjects(Reference Ogiwara, Takahashi and Kato18). In a follow-up study of patients with cancer the analysis of body composition (by dual-energy X-ray absorptiometry) has shown that in progressive cancer cachexia the loss of body fat is more rapid than that of lean mass and occurs preferentially from the trunk followed by leg and arm adipose tissue(Reference Fouladiun, Korner and Bosaeus19). A recent study using retrospective computed tomography scan images of patients with advanced colo-rectal cancer has shown that the most rapid loss of adipose tissues (⩽41%) occurs within 3 months of death(Reference Lieffers, Mourtzakis and Hall20). Marked falls of ⩽85% body fat have been observed in patients with lung cancer, which may lead to hyperlipidaemia and insulin resistance as well as complicating anti-tumour therapies(Reference Fearon and Preston21, Reference Fearon22). With the current escalation in obesity, the paradox of higher BMI as a long-term risk factor but having better survival has been observed in several wasting diseases, including chronic obstructive pulmonary disease, chronic heart failure, end-stage renal failure and cancer(Reference Kalantar-Zadeh, Horwich and Oreopoulos23). A recent study has reported that obesity estimated by an elevated BMI appears to have a protective effect against prostate cancer-specific mortality(Reference Halabi, Ou and Vogelzang24).

Fat loss cannot be explained by reduced appetite alone, as it often precedes the onset of anorexia and is much more severe in animal models of cachexia than in food restriction(Reference Ishiko, Nishimura and Yasui25). This pattern is also observed at the tissue level, as morphological examination has shown marked changes in adipose tissue plasticity in mice with cancer cachexia compared with ad libitum-fed and pair-fed controls(Reference Bing, Russell and Becket26). Adipose tissue from tumour-bearing mice contains shrunken adipocytes of various sizes with a dilated interstitial space. Further morphometric analysis has revealed that the adipocyte size is dramatically reduced. In the tissue matrix increased fibrosis is evident with strong collagen-fibril staining (Fig. 1). Moreover, the ultrastructural features of ‘slimmed’ adipocytes are characterised by severe delipidation and alterations in cell membrane conformation, with irregular cytoplamic projections and increased mitochondria that are electron dense. Taken together, these changes illustrate adipose remodelling in cancer cachexia. A recent study of adipose tissue from patients with cancer has also shown tissue atrophy in subjects with cachexia but not in those without cachexia (C Bing and NA Stephens, unpublished results).

Fig. 1. Light microscopy of Sirius Red-stained sections of epididymal adipose tissue from control (A), pair-fed (B) and MAC16 tumour-bearing (C) mice at day 18 after tumour inoculation. (Adapted from Bing et al. 2006(Reference Bing, Russell and Becket26).)

Mechanisms of adipose atrophy

Although the molecular basis of adipose atrophy is poorly understood, evidence suggests that fat loss may arise from an enhanced catabolic response and disrupted anabolic processes. Increased lipolysis appears to be a key factor(Reference Zuijdgeest-van Leeuwen, van den Berg and Wattimena27, Reference Agustsson, Ryden and Hoffstedt28). In patients with cancer cachexia there is an increase in glycerol and fatty acid turnover compared with patients with cancer without cachexia(Reference Shaw and Wolfe29). It has also been shown that whole-body lipolysis, measured by the rate of appearance of glycerol, is higher in patients with cancer who are losing weight than in healthy subjects(Reference Zuijdgeest-van Leeuwen, van den Berg and Wattimena27). Studies have demonstrated that lipolytic activity (fasting plasma glycerol or fatty acids) is increased in patients with cancer cachexia(Reference Agustsson, Ryden and Hoffstedt28, Reference Drott, Persson and Lundholm30). Increased expression and activity of hormone-sensitive lipase, a rate-limiting enzyme of the lipolytic pathway, is thought to promote lipolysis. Hormone-sensitive lipase mRNA and protein levels have been shown to be increased in adipose tissue of patients with cancer cachexia(Reference Agustsson, Ryden and Hoffstedt28, Reference Thompson, Cooper and Parry31). It is therefore proposed that inhibition of hormone-sensitive lipase may prevent or reverse cachexia-associated fat loss. Furthermore, in mature adipocytes isolated from subcutaneous fat of patients with gastrointestinal adenocarcinoma the lipolytic effects of catecholamines and natriuretic peptide are increased by >2-fold in patients with cachexia, although the basal lipolysis is unchanged(Reference Agustsson, Ryden and Hoffstedt28). However, gene expression of adipose TAG lipase is not affected in patients with cachexia(Reference Agustsson, Ryden and Hoffstedt28). It is also postulated that the fatty acids liberated by lipolysis may serve as substrates for oxidation(Reference Laurencikiene, Stenson and Arvidsson Nordstrom32), which might be mediated by the adipocyte-specific gene cell death-inducing DNA fragmentation factor-α-like effector A. Cell death-inducing DNA fragmentation factor-α-like effector A mRNA levels are increased in patients with cancer cachexia and its overexpression in vitro stimulates adipocyte fatty acid oxidation while decreasing glucose oxidation through inactivation of the pyruvate dehydrogenase complex(Reference Laurencikiene, Stenson and Arvidsson Nordstrom32).

In addition to increased lipolysis, fat loss may be attributable to a decrease in lipid deposition. Circulating insulin, the hormone that promotes fat deposition and glucose transport in adipose tissue, is reduced in the tumour-bearing state(Reference Yoshikawa, Noguchi and Satoh33Reference Bing, Taylor and Tisdale35). A fall in lipoprotein lipase activity in white fat has been reported in tumour-bearing mice(Reference Thompson, Koons and Tan36). This outcome may lead to reduced cleavage of TAG from plasma lipoproteins into glycerol and NEFA for storage, resulting in an increased net flux of lipid into the circulation. There is also evidence that fat diminution in cachexia could be the result of impairment in the formation and development of adipose tissue. A recent study has shown that the expression of the genes encoding several key adipogenic transcription factors, including CCAAT/enhancer-binding protein-α and -β, PPARγ and sterol regulatory element-binding protein-1c, is markedly reduced in white fat of mice with cancer cachexia(Reference Bing, Russell and Becket26). mRNA levels of sterol regulatory element-binding protein-1c targets, genes encoding lipogenic enzymes fatty acid synthase, acetyl-CoA carboxylase, stearoyl-CoA desaturase 1 and glycerol-3-phosphate acyl transferase, also fall markedly(Reference Bing, Russell and Becket26, Reference Hale, Price and Sanchez37). Finally, glucose, which serves as a substrate for lipid synthesis, is transported into the adipocyte via the insulin-responsive facilitative glucose transporter GLUT-4. However, there is a decrease in GLUT-4 mRNA in white fat of mice with cancer cachexia(Reference Bing, Russell and Becket26), which could be a downstream effect of inhibited CCAAT/enhancer binding protein α since its deficiency is associated with abnormal subcellular localisation of GLUT-4(Reference Rosen38).

Potential mediators of adipose atrophy

Several factors produced by tumours and host tissues in the presence of a tumour burden are suggested to be able to mediate fat loss in cachexia. These factors include pro-inflammatory cytokines such as TNFα, IL-1β and IL-6 and the lipid-mobilising factor zinc-α2-glycoprotein (ZAG; also known as AZGP1), each of which can be derived from the tumour and also from the host tissues. Their potential involvement in cachexia-associated fat depletion will be discussed.

Cytokines

TNFα, also termed cachectin, was first identified as the cachexia-inducing factor in chronic diseases such as cancer and persistent infection(Reference Cerami, Ikeda and Le Trang39). Recent data have shown that TNFα infusion can induce systemic lipolysis in human subjects(Reference Plomgaard, Fischer and Ibfelt40). Treatment with TNFα in vitro increases glycerol release from rodent and human adipocytes, probably by inhibiting lipoprotein lipase activity(Reference Yang, Koo and Yoon41) and down regulating the expression of perilipin, which then enables hormone-sensitive lipase to access the surface of lipid droplets(Reference Ryden and Arner42). TNFα-induced adipocyte lipolysis is the outcome of activation of the TNFα receptor 1-dependent pathway(Reference Lopez-Soriano, Llovera and Carbo43, Reference Sethi, Xu and Uysal44), which involves the stimulation of extracellular signal-regulated kinase 1 and 2, mitogen-activated protein kinase, c-Jun N-terminal kinase and protein kinase A(Reference Ryden, Dicker and van Harmelen45, Reference Zhang, Halbleib and Ahmad46). TNFα also has an inhibitory effect on adipocyte differentiation via the Wnt-signalling pathway(Reference Hammarstedt, Isakson and Gustafson47, Reference Cawthorn, Heyd and Hegyi48). In addition, both TNFα and IL-1β are able to inhibit glucose transport in murine and human adipocytes(Reference Hauner, Petruschke and Russ49) and consequently decrease the availability of substrates for lipogenesis. Some studies suggest that TNFα also increases lipid deployment, probably via up-regulation of uncoupling protein (UCP) 2 and UCP3 expression in skeletal muscle(Reference Masaki, Yoshimatsu and Chiba50, Reference Busquets, Carbo and Almendro51), offering a mechanism to remove NEFA resulting from lipolysis. Although these studies indicate a role for TNFα in reducing fat mass, its importance in cancer-related adipose atrophy is still debatable. It is largely a result of the observations that circulating TNFα levels are unchanged or undetectable(Reference Kayacan, Karnak and Beder52, Reference Iwase, Murakami and Saito53), as well as elevated(Reference Karayiannakis, Syrigos and Polychronidis54, Reference Fortunati, Manti and Birocco55), in patients with cancer cachexia.

IL-6 has been shown to moderately increase lipolysis in human adipose tissue in vitro (Reference Trujillo, Sullivan and Harten56). Treatment with CNTO-328, a monoclonal antibody to IL-6, is able to reverse tumour-induced cachexia in nude mice(Reference Zaki, Nemeth and Trikha57). Recent work has shown that IL-6 is necessary for the onset of adipose and skeletal muscle wasting in the Apc(Min/+) mouse(Reference Baltgalvis, Berger and Pena58). Despite serum IL-6 levels being elevated in patients with cancer(Reference Richey, George and Couch59, Reference Krzystek-Korpacka, Matusiewicz and Diakowska60), it is still unclear whether circulating IL-6 correlates with the extent of cachexia(Reference Iwase, Murakami and Saito53, Reference Richey, George and Couch59Reference Kuroda, Nakashima and Kanao61). Since TNFα and IL-6 are also produced by adipose tissue, although probably mostly from non-fat cells(Reference Fain62), their autocrine and/or paracrine effects may be important in cachexia. However, studies in mice with cancer cachexia have shown that TNFα and IL-6 mRNA levels in white fat are unaffected by the tumour burden(Reference Bing, Russell and Becket26, Reference Bing, Brown and King63). In a recent study of patients with gastrointestinal cancer no alterations were found in gene expression of TNFα and IL-6 and their protein release by adipose tissue under cachectic states(Reference Ryden, Agustsson and Laurencikiene64). In addition, there is no apparent infiltration of macrophages and lymphocytes in adipose tissue of mice with cachexia(Reference Bing, Russell and Becket26) and patients with cancer cachexia(Reference Ryden, Agustsson and Laurencikiene64).

ZAG, a lipid-mobilising factor

ZAG is a 41 kDa soluble protein first isolated from human plasma(Reference Burgi and Schmid65) and subsequently identified in secretory epithelial cells, including those of liver, breast, prostate and the gastrointestinal tract(Reference Tada, Ohkubo and Niwa66). The crystal structure of ZAG reveals that it belongs to the class I MHC family. There is a non-peptidic ligand in the ZAG counterpart of the MHC peptide-binding groove, which may relate to its signalling function(Reference Sanchez, Chirino and Bjorkman67). ZAG is overexpressed by several types of malignant tumour, such as breast, prostate and bladder cancers(Reference Hale, Price and Sanchez37, Reference Diez-Itza, Sanchez and Allende68, Reference Irmak, Tilki and Heukeshoven69), and ZAG levels are elevated in serum and seminal fluid of patients with prostate cancer(Reference Hale, Price and Sanchez37, Reference Hassan, Kumar and Kashav70). The biological functions of ZAG were largely unknown until a lipid-mobilising factor, purified from the urine of patients with cancer cachexia, was shown to be identical to ZAG in electrophoretic mobility, immunoreactivity and amino acid sequence(Reference Todorov, McDevitt and Meyer71). ZAG has also been purified from a murine adenocarcinoma (MAC16) that induces profound cachexia(Reference Hirai, Hussey and Barber72). Amino acid sequence analysis has revealed that murine and human ZAG display an overall homology of 59%(Reference Ueyama, Naitoh and Ohkubo73), but share up to 100% identity in specific regions thought to be important in lipid metabolism(Reference Sanchez, Chirino and Bjorkman67).

Treatment with purified ZAG can cause weight loss in genetically-obese ob/ob mice(Reference Hirai, Hussey and Barber72) and normal mice(Reference Bing, Russell and Beckett74, Reference Russell, Zimmerman and Domin75), and body composition analysis indicates that ZAG-induced weight loss is a result of selective reduction in body fat but not lean mass. ZAG has been shown in vitro to stimulate glycerol release from isolated murine adipocytes in a dose-dependent manner(Reference Hirai, Hussey and Barber72, Reference Russell, Zimmerman and Domin75). The lipolytic effect of ZAG has been postulated to be mediated by β3-adrenoceptors and the activation of the intracellular cAMP pathway. ZAG has been shown to be able to produce a comparable increase in cAMP levels to that obtained with isoprenaline and ZAG-induced lipolysis can be attenuated by the specific β3-adrenoceptor antagonist SR59230 in adipocytes(Reference Hirai, Hussey and Barber72, Reference Russell, Zimmerman and Domin75, Reference Russell, Hirai and Tisdale76).

In addition to lipid mobilisation there is also evidence that ZAG promotes lipid utilisation in brown adipose tissue and skeletal muscle. ZAG administration in vivo in mice leads to an up-regulation of UCP1 mRNA and protein expression in brown adipose tissue and of skeletal muscle UCP2 and UCP3 mRNA(Reference Bing, Russell and Beckett74). ZAG induces expression of UCP1 protein and O2 uptake in vitro in primary cultures of brown adipose tissue(Reference Hirai, Hussey and Barber72, Reference Sanders and Tisdale77). Hence, by up regulating UCP in brown adipose tissue and muscle, ZAG may provide a mechanism for the disposal of excess fatty acids liberated from enhanced lipolysis, which could lead to increased energy expenditure during cachexia.

Adipose-derived ZAG in cancer cachexia

The secretory function of adipose tissue and the potent fat-mobilising effect of ZAG have led to the postulation that this protein could also be produced by adipose tissue, thereby modulating adipocyte metabolism(Reference Bing, Bao and Jenkins78). Work from the authors' group has demonstrated that the ZAG gene and protein are expressed by the major white fat depots (epididymal, perirenal, subcutaneous, mammary gland) and the interscapular brown fat of mice(Reference Bing, Bao and Jenkins78). Further detection using immunocytochemistry has shown the presence of ZAG protein in the cytoplasm of adipocytes in adipose tissue(Reference Bing, Bao and Jenkins78). In human subjects ZAG mRNA and protein have been shown to be expressed in both visceral and subcutaneous fat depots(Reference Bing, Bao and Jenkins78). Futhermore, ZAG mRNA and protein are detected in differentiated human Simpson-Golabi-Behmel syndrome adipocytes. Most importantly, ZAG, which contains a secretory signal sequence(Reference Hale, Price and Sanchez37), has been shown to be secreted into the culture medium by differentiated Simpson-Golabi-Behmel syndrome adipocytes(Reference Bao, Bing and Hunter79). Subsequent quantification of ZAG secretion levels in the culture medium has revealed its concentration to be in the range of ng/ml per 24 h, close to that of adiponectin(Reference Mracek, Ding and Tzanavari80). Taken together, these findings indicate that ZAG is indeed a novel adipokine produced abundantly by adipocytes and the protein may have a major action locally in the regulation of fat mass.

Adipose-derived ZAG appears to be inversely associated with body fat mass. ZAG mRNA and protein levels are markedly increased in adipose tissue of mice with cancer cachexia and, furthermore, the increase in ZAG protein content is related to the extent of weight loss in these animals(Reference Bing, Bao and Jenkins78). In contrast, as a reference adipokine, leptin mRNA and circulating leptin levels are strikingly repressed in tumour-bearing mice(Reference Bing, Russell and Becket26, Reference Bing, Taylor and Tisdale35, Reference Bing, Bao and Jenkins78) (Fig. 2). Very recently, it has been shown that ZAG mRNA and protein expression are also up regulated in adipose tissue in patients with cancer cachexia (T Mracek, NA Stephens, X Xiao and C Bing, unpublished results). Contrarily, studies of obese subjects have shown that ZAG gene expression is down regulated in subcutaneous adipose tissue of obese women(Reference Dahlman, Kaaman and Olsson81) and men(Reference Marrades, Martinez and Moreno-Aliaga82). Furthermore, recent work has demonstrated that ZAG mRNA levels are negatively correlated with total fat mass in human subjects with a wide range of BMI(Reference Mracek, Ding and Tzanavari80).

Fig. 2. mRNA levels of zinc-α2-glycoprotein (A), leptin (B), CCAAT/enhancer-binding protein α (C) and sterol regulatory element-binding protein-1c (D) in epididymal adipose tissue of control (□), pair-fed (PF; ) and MAC16 tumour-bearing (▪) mice, quantified by real-time PCR and normalised to β-actin. Values presented as fold changes relative to controls are means with their standard errors represented by vertical bars for eight mice per group. Mean values were significantly different from those for the control group: *P<0·05, **P<0·01. Mean values were significantly different from those for the PF group: †P<0·05, ††P<0·01. (Adapted from Bing et al. 2006(Reference Bing, Russell and Becket26).)

Although the role of ZAG in adipose tissue remains to be established, its effect on fat loss has been further supported by a recent study that shows that ZAG-knock-out mice are vulnerable to weight gain when fed a high-fat diet and this outcome appears to be the result of decreased lipolytic response to several stimuli, such as isoprenaline, CL316243, foskolin and isobutylmethylxanthine, in adipocytes(Reference Rolli, Radosavljevic and Astier83). Recent work has demonstrated that recombinant ZAG stimulates lipolysis in human adipocytes (T Mracek, P Trayhurn and C Bing, unpublished results). Further studies are required to unravel the nature of the action of ZAG in human cancer cachexia. Overall, current data point to ZAG, as well as TNFα, as a potential candidate for mediating lipid catabolism in cancer cachexia (Fig. 3). Other factors may also be involved in fat loss in malignancy, e.g. macrophage inhibitory cytokine-1, which causes cachexia and a reduction in fat mass via its central effects on appetite(Reference Johnen, Lin and Kuffner84). Interestingly, recent work has found that macrophage inhibitory cytokine-1 is also produced by adipocytes and this factor may have autocrine and/or paracrine effects in adipose tissue(Reference Ding, Mracek and Gonzalez-Muniesa85).

Fig. 3. A schematic diagram of lipid catabolism in cancer cachexia. Certain tumour and host tissue-produced factors, such as TNFα and zinc-α2-glycoprotein (ZAG), may act systemically and/or as autocrine and/or paracrine signals to stimulate adipocyte lipid metabolism. TNFα-induced lipolysis acts through a TNFα receptor 1 (TNFR-1) dependent pathway, which inhibits perilipin allowing hormone-sensitive lipase (HSL) to access the surface of lipid droplets. ZAG-stimulated lipolysis may be mediated by β3-adrenoceptors (β3AR) and the activation of the intracellular cAMP pathway. NEFA generated by enhanced lipolysis in cachexia may serve as substrates for lipid utilisation through uncoupling proteins (UCP) in brown adipose tissue (BAT) and skeletal muscle.↑, Increased; ↓, decreased.

Conclusion

Cancer cachexia, manifested by progressive weight loss, is a metabolic disorder associated with increased morbidity and mortality. Extensive loss of adipose tissue is a prominent feature of cachexia. The marked alterations in adipose morphology indicate tissue atrophy and this outcome cannot be attributable to reduced appetite alone. Evidence suggests that altered adipocyte metabolism may have an important role. Increased lipolysis appears to be a key factor, while impairment in lipid deposition and adipocyte development may also contribute. Adipose atrophy could be mediated by the tumour and/or host-derived factors. TNFα, as a potential mediator, has been linked with increased lipolysis. ZAG, a potent lipid-mobilising factor, has been identified as a novel adipokine and its expression in adipose tissue is up regulated in cancer cachexia. ZAG may therefore act locally, as well as systemically, to promote lipid breakdown. Further elucidation of the function of ZAG in adipose tissue may lead to novel targets for preventing adipose atrophy in malignancy.

Acknowledgement

The authors declare no conflict of interest. This work is being carried out with financial support from the University of Liverpool R&D Fund, the Biotechnology and Biological Sciences Research Council (BBE015379) and the Medical Research Council (87972). C. B. wrote the draft of the article, with subsequent revision from P. T., and C. B. prepared the final version.

References

1.Morley, JE, Thomas, DR & Wilson, MM (2006) Cachexia: pathophysiology and clinical relevance. Am J Clin Nutr 83, 735743.CrossRefGoogle ScholarPubMed
2.Tisdale, MJ (2002) Cachexia in cancer patients. Nat Rev Cancer 2, 862871.CrossRefGoogle ScholarPubMed
3.Tisdale, MJ (2005) Molecular pathways leading to cancer cachexia. Physiology (Bethesda) 20, 340348.Google ScholarPubMed
4.Slaviero, KA, Read, JA, Clarke, SJ et al. (2003) Baseline nutritional assessment in advanced cancer patients receiving palliative chemotherapy. Nutr Cancer 46, 148157.CrossRefGoogle ScholarPubMed
5.Fearon, KC, Voss, AC & Hustead, DS (2006) Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr 83, 13451350.CrossRefGoogle ScholarPubMed
6.Deans, DA, Wigmore, SJ, de Beaux, AC et al. (2007) Clinical prognostic scoring system to aid decision-making in gastro-oesophageal cancer. Br J Surg 94, 15011508.CrossRefGoogle ScholarPubMed
7.Skipworth, RJ, Stewart, GD, Dejong, CH et al. (2007) Pathophysiology of cancer cachexia: Much more than host-tumour interaction? Clin Nutr 26, 667676.CrossRefGoogle ScholarPubMed
8.Argiles, JM, Busquets, S & Lopez-Soriano, FJ (2006) Cytokines as mediators and targets for cancer cachexia. Cancer Treat Res 130, 199217.CrossRefGoogle ScholarPubMed
9.Spiegelman, BM & Flier, JS (2001) Obesity and the regulation of energy balance. Cell 104, 531543.CrossRefGoogle ScholarPubMed
10.Bluher, M (2005) Transgenic animal models for the study of adipose tissue biology. Best Pract Res Clin Endocrinol Metab 19, 605623.CrossRefGoogle Scholar
11.Rosen, ED & Spiegelman, BM (2006) Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444, 847853.CrossRefGoogle ScholarPubMed
12.Trayhurn, P & Wood, IS (2004) Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br J Nutr 92, 347355.CrossRefGoogle ScholarPubMed
13.Mohamed-Ali, V, Pinkney, JH & Coppack, SW (1998) Adipose tissue as an endocrine and paracrine organ. Int J Obes Relat Metab Disord 22, 11451158.CrossRefGoogle ScholarPubMed
14.Ahima, RS & Flier, JS (2000) Adipose tissue as an endocrine organ. Trends Endocrinol Metab 11, 327332.CrossRefGoogle ScholarPubMed
15.Fruhbeck, G, Gomez-Ambrosi, J, Muruzabal, FJ et al. (2001) The adipocyte: a model for integration of endocrine and metabolic signaling in energy metabolism regulation. Am J Physiol Endocrinol Metab 280, E827E847.CrossRefGoogle Scholar
16.Rajala, MW & Scherer, PE (2003) Minireview: The adipocyte – at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 144, 37653773.CrossRefGoogle ScholarPubMed
17.Trayhurn, P & Bing, C (2006) Appetite and energy balance signals from adipocytes. Philos Trans R Soc Lond B Biol Sci 361, 12371249.CrossRefGoogle ScholarPubMed
18.Ogiwara, H, Takahashi, S, Kato, Y et al. (1994) Diminished visceral adipose tissue in cancer cachexia. J Surg Oncol 57, 129133.CrossRefGoogle ScholarPubMed
19.Fouladiun, M, Korner, U, Bosaeus, I et al. (2005) Body composition and time course changes in regional distribution of fat and lean tissue in unselected cancer patients on palliative care – correlations with food intake, metabolism, exercise capacity, and hormones. Cancer 103, 21892198.CrossRefGoogle ScholarPubMed
20.Lieffers, JR, Mourtzakis, M, Hall, KD et al. (2009) A viscerally driven cachexia syndrome in patients with advanced colorectal cancer: contributions of organ and tumor mass to whole-body energy demands. Am J Clin Nutr 89, 11731179.CrossRefGoogle ScholarPubMed
21.Fearon, KC & Preston, T (1990) Body composition in cancer cachexia. Infusionstherapie 17, Suppl. 3, 6366.Google ScholarPubMed
22.Fearon, KC (1992) The mechanisms and treatment of weight loss in cancer. Proc Nutr Soc 51, 251265.CrossRefGoogle ScholarPubMed
23.Kalantar-Zadeh, K, Horwich, TB, Oreopoulos, A et al. (2007) Risk factor paradox in wasting diseases. Curr Opin Clin Nutr Metab Care 10, 433442.CrossRefGoogle ScholarPubMed
24.Halabi, S, Ou, SS, Vogelzang, NJ et al. (2007) Inverse correlation between body mass index and clinical outcomes in men with advanced castration-recurrent prostate cancer. Cancer 110, 14781484.CrossRefGoogle ScholarPubMed
25.Ishiko, O, Nishimura, S, Yasui, T et al. (1999) Metabolic and morphologic characteristics of adipose tissue associated with the growth of malignant tumors. Jpn J Cancer Res 90, 655659.CrossRefGoogle ScholarPubMed
26.Bing, C, Russell, S, Becket, E et al. (2006) Adipose atrophy in cancer cachexia: morphologic and molecular analysis of adipose tissue in tumour-bearing mice. Br J Cancer 95, 10281037.CrossRefGoogle ScholarPubMed
27.Zuijdgeest-van Leeuwen, SD, van den Berg, JW, Wattimena, JL et al. (2000) Lipolysis and lipid oxidation in weight-losing cancer patients and healthy subjects. Metabolism 49, 931936.CrossRefGoogle ScholarPubMed
28.Agustsson, T, Ryden, M, Hoffstedt, J et al. (2007) Mechanism of increased lipolysis in cancer cachexia. Cancer Res 67, 55315537.CrossRefGoogle ScholarPubMed
29.Shaw, JH & Wolfe, RR (1987) Fatty acid and glycerol kinetics in septic patients and in patients with gastrointestinal cancer. The response to glucose infusion and parenteral feeding. Ann Surg 205, 368376.CrossRefGoogle ScholarPubMed
30.Drott, C, Persson, H & Lundholm, K (1989) Cardiovascular and metabolic response to adrenaline infusion in weight-losing patients with and without cancer. Clin Physiol 9, 427439.CrossRefGoogle ScholarPubMed
31.Thompson, MP, Cooper, ST, Parry, BR et al. (1993) Increased expression of the mRNA for hormone-sensitive lipase in adipose tissue of cancer patients. Biochim Biophys Acta 1180, 236242.CrossRefGoogle ScholarPubMed
32.Laurencikiene, J, Stenson, BM, Arvidsson Nordstrom, E et al. (2008) Evidence for an important role of CIDEA in human cancer cachexia. Cancer Res 68, 92479254.CrossRefGoogle ScholarPubMed
33.Yoshikawa, T, Noguchi, Y, Satoh, S et al. (1997) Insulin resistance and the alterations of glucose transporter-4 in adipose cells from cachectic tumor-bearing rats. JPEN J Parenter Enteral Nutr 21, 347349.CrossRefGoogle ScholarPubMed
34.Inadera, H, Nagai, S, Dong, HY et al. (2002) Molecular analysis of lipid-depleting factor in a colon-26-inoculated cancer cachexia model. Int J Cancer 101, 3745.CrossRefGoogle Scholar
35.Bing, C, Taylor, S, Tisdale, MJ et al. (2001) Cachexia in MAC16 adenocarcinoma: suppression of hunger despite normal regulation of leptin, insulin and hypothalamic neuropeptide Y. J Neurochem 79, 10041012.CrossRefGoogle ScholarPubMed
36.Thompson, MP, Koons, JE, Tan, ET et al. (1981) Modified lipoprotein lipase activities, rates of lipogenesis, and lipolysis as factors leading to lipid depletion in C57BL mice bearing the preputial gland tumor, ESR-586. Cancer Res 41, 32283232.Google ScholarPubMed
37.Hale, LP, Price, DT, Sanchez, LM et al. (2001) Zinc alpha-2-glycoprotein is expressed by malignant prostatic epithelium and may serve as a potential serum marker for prostate cancer. Clin Cancer Res 7, 846853.Google ScholarPubMed
38.Rosen, ED (2005) The transcriptional basis of adipocyte development. Prostaglandins Leukot Essent Fatty Acids 73, 3134.CrossRefGoogle ScholarPubMed
39.Cerami, A, Ikeda, Y, Le Trang, N et al. (1985) Weight loss associated with an endotoxin-induced mediator from peritoneal macrophages: the role of cachectin (tumor necrosis factor). Immunol Lett 11, 173177.CrossRefGoogle ScholarPubMed
40.Plomgaard, P, Fischer, CP, Ibfelt, T et al. (2008) Tumor necrosis factor-alpha modulates human in vivo lipolysis. J Clin Endocrinol Metab 93, 543549; Epublication 20 November 2007.CrossRefGoogle ScholarPubMed
41.Yang, JY, Koo, JH, Yoon, HY et al. (2007) Effect of scopoletin on lipoprotein lipase activity in 3T3-L1 adipocytes. Int J Mol Med 20, 527531.Google ScholarPubMed
42.Ryden, M & Arner, P (2007) Tumour necrosis factor-alpha in human adipose tissue – from signalling mechanisms to clinical implications. J Intern Med 262, 431438.CrossRefGoogle ScholarPubMed
43.Lopez-Soriano, J, Llovera, M, Carbo, N et al. (1997) Lipid metabolism in tumour-bearing mice: studies with knockout mice for tumour necrosis factor receptor 1 protein. Mol Cell Endocrinol 132, 9399.Google ScholarPubMed
44.Sethi, JK, Xu, H, Uysal, KT et al. (2000) Characterisation of receptor-specific TNFalpha functions in adipocyte cell lines lacking type 1 and 2 TNF receptors. FEBS Lett 469, 7782.CrossRefGoogle ScholarPubMed
45.Ryden, M, Dicker, A, van Harmelen, V et al. (2002) Mapping of early signaling events in tumor necrosis factor-alpha-mediated lipolysis in human fat cells. J Biol Chem 277, 10851091.CrossRefGoogle ScholarPubMed
46.Zhang, HH, Halbleib, M, Ahmad, F et al. (2002) Tumor necrosis factor-alpha stimulates lipolysis in differentiated human adipocytes through activation of extracellular signal-related kinase and elevation of intracellular cAMP. Diabetes 51, 29292935.CrossRefGoogle ScholarPubMed
47.Hammarstedt, A, Isakson, P, Gustafson, B et al. (2007) Wnt-signaling is maintained and adipogenesis inhibited by TNFalpha but not MCP-1 and resistin. Biochem Biophys Res Commun 357, 700706.CrossRefGoogle Scholar
48.Cawthorn, WP, Heyd, F, Hegyi, K et al. (2007) Tumour necrosis factor-alpha inhibits adipogenesis via a beta-catenin/TCF4(TCF7L2)-dependent pathway. Cell Death Differ 14, 13611373.CrossRefGoogle Scholar
49.Hauner, H, Petruschke, T, Russ, M et al. (1995) Effects of tumour necrosis factor alpha (TNF alpha) on glucose transport and lipid metabolism of newly-differentiated human fat cells in cell culture. Diabetologia 38, 764771.CrossRefGoogle ScholarPubMed
50.Masaki, T, Yoshimatsu, H, Chiba, S et al. (1999) Tumor necrosis factor-alpha regulates in vivo expression of the rat UCP family differentially. Biochim Biophys Acta 1436, 585592.CrossRefGoogle ScholarPubMed
51.Busquets, S, Carbo, N, Almendro, V et al. (2001) Hyperlipemia: a role in regulating UCP3 gene expression in skeletal muscle during cancer cachexia? FEBS Lett 505, 255258.CrossRefGoogle ScholarPubMed
52.Kayacan, O, Karnak, D, Beder, S et al. (2006) Impact of TNF-alpha and IL-6 levels on development of cachexia in newly diagnosed NSCLC patients. Am J Clin Oncol 29, 328335.CrossRefGoogle ScholarPubMed
53.Iwase, S, Murakami, T, Saito, Y et al. (2004) Steep elevation of blood interleukin-6 (IL-6) associated only with late stages of cachexia in cancer patients. Eur Cytokine Netw 15, 312316.Google ScholarPubMed
54.Karayiannakis, AJ, Syrigos, KN, Polychronidis, A et al. (2001) Serum levels of tumor necrosis factor-alpha and nutritional status in pancreatic cancer patients. Anticancer Res 21, 13551358.Google ScholarPubMed
55.Fortunati, N, Manti, R, Birocco, N et al. (2007) Pro-inflammatory cytokines and oxidative stress/antioxidant parameters characterize the bio-humoral profile of early cachexia in lung cancer patients. Oncol Rep 18, 15211527.Google ScholarPubMed
56.Trujillo, ME, Sullivan, S, Harten, I et al. (2004) Interleukin-6 regulates human adipose tissue lipid metabolism and leptin production in vitro. J Clin Endocrinol Metab 89, 55775582.CrossRefGoogle ScholarPubMed
57.Zaki, MH, Nemeth, JA & Trikha, M (2004) CNTO 328, a monoclonal antibody to IL-6, inhibits human tumor-induced cachexia in nude mice. Int J Cancer 111, 592595.CrossRefGoogle ScholarPubMed
58.Baltgalvis, KA, Berger, FG, Pena, MM et al. (2008) Interleukin-6 and cachexia in ApcMin/+ mice. Am J Physiol Regul Integr Comp Physiol 294, R393R401.CrossRefGoogle ScholarPubMed
59.Richey, LM, George, JR, Couch, ME et al. (2007) Defining cancer cachexia in head and neck squamous cell carcinoma. Clin Cancer Res 13, 65616567.CrossRefGoogle ScholarPubMed
60.Krzystek-Korpacka, M, Matusiewicz, M, Diakowska, D et al. (2007) Impact of weight loss on circulating IL-1, IL-6, IL-8, TNF-alpha, VEGF-A, VEGF-C and midkine in gastroesophageal cancer patients. Clin Biochem 40, 13531360.CrossRefGoogle ScholarPubMed
61.Kuroda, K, Nakashima, J, Kanao, K et al. (2007) Interleukin 6 is associated with cachexia in patients with prostate cancer. Urology 69, 113117.CrossRefGoogle ScholarPubMed
62.Fain, JN (2006) Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm 74, 443477.CrossRefGoogle Scholar
63.Bing, C, Brown, M, King, P et al. (2000) Increased gene expression of brown fat uncoupling protein (UCP)1 and skeletal muscle UCP2 and UCP3 in MAC16-induced cancer cachexia. Cancer Res 60, 24052410.Google ScholarPubMed
64.Ryden, M, Agustsson, T, Laurencikiene, J et al. (2008) Lipolysis – not inflammation, cell death, or lipogenesis – is involved in adipose tissue loss in cancer cachexia. Cancer 113, 16951704.CrossRefGoogle ScholarPubMed
65.Burgi, W & Schmid, K (1961) Preparation and properties of Zn-alpha 2-glycoprotein of normal human plasma. J Biol Chem 236, 10661074.CrossRefGoogle ScholarPubMed
66.Tada, T, Ohkubo, I, Niwa, M et al. (1991) Immunohistochemical localization of Zn-alpha 2-glycoprotein in normal human tissues. J Histochem Cytochem 39, 12211226.CrossRefGoogle ScholarPubMed
67.Sanchez, LM, Chirino, AJ & Bjorkman, P (1999) Crystal structure of human ZAG, a fat-depleting factor related to MHC molecules. Science 283, 19141919.CrossRefGoogle Scholar
68.Diez-Itza, I, Sanchez, LM, Allende, MT et al. (1993) Zn-alpha 2-glycoprotein levels in breast cancer cytosols and correlation with clinical, histological and biochemical parameters. Eur J Cancer 29A, 12561260.CrossRefGoogle ScholarPubMed
69.Irmak, S, Tilki, D, Heukeshoven, J et al. (2005) Stage-dependent increase of orosomucoid and zinc-alpha2-glycoprotein in urinary bladder cancer. Proteomics 5, 42964304.CrossRefGoogle ScholarPubMed
70.Hassan, MI, Kumar, V, Kashav, T et al. (2007) Proteomic approach for purification of seminal plasma proteins involved in tumor proliferation. J Sep Sci 30, 19791988.CrossRefGoogle ScholarPubMed
71.Todorov, PT, McDevitt, TM, Meyer, DJ et al. (1998) Purification and characterization of a tumor lipid-mobilizing factor. Cancer Res 58, 23532358.Google ScholarPubMed
72.Hirai, K, Hussey, HJ, Barber, MD et al. (1998) Biological evaluation of a lipid-mobilizing factor isolated from the urine of cancer patients. Cancer Res 58, 23592365.Google ScholarPubMed
73.Ueyama, H, Naitoh, H & Ohkubo, I (1994) Structure and expression of rat and mouse mRNAs for Zn-alpha 2-glycoprotein. J Biochem (Tokyo) 116, 677681.CrossRefGoogle ScholarPubMed
74.Bing, C, Russell, ST, Beckett, EE et al. (2002) Expression of uncoupling proteins-1, -2 and -3 mRNA is induced by an adenocarcinoma-derived lipid-mobilizing factor. Br J Cancer 86, 612618.CrossRefGoogle ScholarPubMed
75.Russell, ST, Zimmerman, TP, Domin, BA et al. (2004) Induction of lipolysis in vitro and loss of body fat in vivo by zinc-alpha2-glycoprotein. Biochim Biophys Acta 1636, 5968.CrossRefGoogle ScholarPubMed
76.Russell, ST, Hirai, K & Tisdale, MJ (2002) Role of beta3-adrenergic receptors in the action of a tumour lipid mobilizing factor. Br J Cancer 86, 424428.CrossRefGoogle ScholarPubMed
77.Sanders, PM & Tisdale, MJ (2004) Role of lipid-mobilising factor (LMF) in protecting tumour cells from oxidative damage. Br J Cancer 90, 12741278.CrossRefGoogle ScholarPubMed
78.Bing, C, Bao, Y, Jenkins, J et al. (2004) Zinc-alpha2-glycoprotein, a lipid mobilizing factor, is expressed in adipocytes and is up-regulated in mice with cancer cachexia. Proc Natl Acad Sci USA 101, 25002505.CrossRefGoogle ScholarPubMed
79.Bao, Y, Bing, C, Hunter, L et al. (2005) Zinc-alpha2-glycoprotein, a lipid mobilizing factor, is expressed and secreted by human (SGBS) adipocytes. FEBS Lett 579, 4147.CrossRefGoogle ScholarPubMed
80.Mracek, T, Ding, Q, Tzanavari, T et al. (2009) The adipokine zinc-alpha2-glycoprotein is downregulated with fat mass expansion in obesity. Clin Endocrinol (Oxf) (Epublication ahead of print version; doi: 10.1111/j.1365-2265.2009.03658.x).Google ScholarPubMed
81.Dahlman, I, Kaaman, M, Olsson, T et al. (2005) A unique role of monocyte chemoattractant protein 1 among chemokines in adipose tissue of obese subjects. J Clin Endocrinol Metab 90, 58345840.CrossRefGoogle ScholarPubMed
82.Marrades, MP, Martinez, JA & Moreno-Aliaga, MJ (2008) ZAG, a lipid mobilizing adipokine, is downregulated in human obesity. J Physiol Biochem 64, 6166.CrossRefGoogle Scholar
83.Rolli, V, Radosavljevic, M, Astier, V et al. (2007) Lipolysis is altered in MHC class I zinc-alpha(2)-glycoprotein deficient mice. FEBS Lett 581, 394400.CrossRefGoogle ScholarPubMed
84.Johnen, H, Lin, S, Kuffner, T et al. (2007) Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-1. Nat Med 13, 13331340.CrossRefGoogle ScholarPubMed
85.Ding, Q, Mracek, T, Gonzalez-Muniesa, P et al. (2009) Identification of macrophage inhibitory cytokine-1 in adipose tissue and its secretion as an adipokine by human adipocytes. Endocrinology 150, 16881696.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Light microscopy of Sirius Red-stained sections of epididymal adipose tissue from control (A), pair-fed (B) and MAC16 tumour-bearing (C) mice at day 18 after tumour inoculation. (Adapted from Bing et al. 2006(26).)

Figure 1

Fig. 2. mRNA levels of zinc-α2-glycoprotein (A), leptin (B), CCAAT/enhancer-binding protein α (C) and sterol regulatory element-binding protein-1c (D) in epididymal adipose tissue of control (□), pair-fed (PF; ) and MAC16 tumour-bearing (▪) mice, quantified by real-time PCR and normalised to β-actin. Values presented as fold changes relative to controls are means with their standard errors represented by vertical bars for eight mice per group. Mean values were significantly different from those for the control group: *P<0·05, **P<0·01. Mean values were significantly different from those for the PF group: †P<0·05, ††P<0·01. (Adapted from Bing et al. 2006(26).)

Figure 2

Fig. 3. A schematic diagram of lipid catabolism in cancer cachexia. Certain tumour and host tissue-produced factors, such as TNFα and zinc-α2-glycoprotein (ZAG), may act systemically and/or as autocrine and/or paracrine signals to stimulate adipocyte lipid metabolism. TNFα-induced lipolysis acts through a TNFα receptor 1 (TNFR-1) dependent pathway, which inhibits perilipin allowing hormone-sensitive lipase (HSL) to access the surface of lipid droplets. ZAG-stimulated lipolysis may be mediated by β3-adrenoceptors (β3AR) and the activation of the intracellular cAMP pathway. NEFA generated by enhanced lipolysis in cachexia may serve as substrates for lipid utilisation through uncoupling proteins (UCP) in brown adipose tissue (BAT) and skeletal muscle.↑, Increased; ↓, decreased.