Category: Home

Caloric restriction and disease prevention

caloric restriction and disease prevention

Over 2 years, the team assessed just restrlction people, restrictjon 21—50 years. Viskaitis, P. Home News Media Calorie Restriction Rewires Metabolism, Immunity for Longer Health Span. Chong, C. Madeo F, Carmona-Gutierrez D, Hofer SJ, Kroemer G. caloric restriction and disease prevention

Caloric restriction and disease prevention -

Intermittent fasting refers to periods with intervals during which no food but only clear fluids are ingested — such as a period of daily time-restricted eating with a window of 8 to 12 hours for any caloric intake — and could be combined with overall calorie restriction and variants of the Mediterranean diet which may contribute to long-term cardiovascular health and longevity.

The study was designed to mimic dietary conditions during World War II. Participants could only eat kcal per day, but were required to walk 5 km per day and expend calories. Despite the extreme calorie restriction, the experiment was not representative of true calorie-restrictive diets, which adhere to intake guidelines for macronutrients and micronutrients.

A systematic review investigated whether people in intensive care units have different outcomes with normocaloric feeding or hypocaloric feeding, and found no difference. A calorie restriction study started in by the National Institute on Aging showed that calorie restriction did not extend years of life or reduce age-related deaths in non-obese rhesus macaques.

In a report on rhesus monkeys, caloric restriction in the presence of adequate nutrition was effective in delaying the effects of aging. Calorie restriction preserves muscle tissue in nonhuman primates [31] [32] and rodents.

However, studies show that overall activity levels are no higher in calorie restriction than ad libitum animals in youth. Preliminary research indicates that sirtuins are activated by fasting and serve as "energy sensors" during metabolism.

Contents move to sidebar hide. Article Talk. Read Edit View history. Tools Tools. What links here Related changes Upload file Special pages Permanent link Page information Cite this page Get shortened URL Download QR code Wikidata item.

Download as PDF Printable version. In other projects. Wikimedia Commons. Dietary regime. For caloric restriction for the purpose of weight loss, see dieting. Main article: Caloric restriction mimetic. doi : PMC PMID Annual Review of Nutrition.

Skyhorse Publishing Inc. Retrieved 30 September September June July Endocrine Practice. Lifestyle Management: Standards of Medical Care in Diabetes ". Diabetes Care. May Diabetes Care Professional society guidelines. BMC Pregnancy and Childbirth. The Biology of Human Starvation, The American Journal of Clinical Nutrition.

Journal of the American Dietetic Association. Clinics in Geriatric Medicine. A Systematic Review and Meta-Analysis of Randomized Controlled Trials". Journal of the American College of Cardiology. S2CID Ageing Research Reviews.

Nutritional interventions modulating aging and age-associated diseases. The biology of human starvation 2 vols.

of Minnesota Press. Intensive Care Medicine. Retrieved May 17, The New York Times. January Nature Communications. Bibcode : NatCo Taken together, it is clear that either hyperinsulinemia or increased IGF-1 or both can augment tumor growth by signaling through these receptors.

This pathway integrates intracellular and environmental cues, such as growth factor concentrations and nutrient availability, to regulate cellular survival, proliferation, protein translation, and metabolism. Akt regulates the mammalian target of rapamycin mTOR [ 22 ], which regulates cell growth, cell proliferation, and survival through downstream mediators.

Increased activation of mTOR is common in tumors and many normal tissues from obese or diabetic mice, while CR decreases mTOR signaling in these same tumors and normal tissues[ 23 ].

Moreover, mTOR activation is inhibited by increased AMP-activated kinase AMPK under low nutrient conditions[ 24 ]. Specific mTOR inhibitors block the tumor-enhancing effects of obesity in mouse models[ 25 , 26 ].

Adiponectin is a peptide hormone primarily secreted from visceral white adipose tissue. In contrast to leptin and other adipokines, circulating levels of adiponectin negatively correlate with adiposity, and are thus increased by CR and decreased by obesity[ 27 ].

Adiponectin functions to counter obesity-related metabolic perturbations, such as insulin resistance and leptin resistance, that impact glucose and fatty acid metabolism, alter insulin responses, and increase production of inflammatory cytokines[ 27 ].

Adiponectin also reduces proinflammatory cytokine expression via inhibition of the nuclear factor κ-light-chain-enhancer of activated B-cells NF-κB [ 28 , 29 ]. Circulating leptin levels positively correlate with adipose stores and nutritional status, and function as an energy sensor to signal the brain to reduce appetite.

Leptin has direct effects on peripheral tissues, indirect effects on neuroendocrine regulators of appetite and energy expenditure in the hypothalamus, and impacts carcinogenesis, angiogenesis, immune responses, cytokine production, and other biological processes[ 31 ].

In the obese state, adipose tissue overproduces leptin, and the brain no longer responds to the signal, resulting in leptin resistance. Insulin, glucocorticoids, tumor necrosis factor-α TNF-α , and estrogens all stimulate leptin release[ 31 ]. Calorie restriction consistently and robustly decreases systemic leptin levels in a manner dependent on the extent of the adiposity loss[ 1 ].

In vitro , animal, and epidemiologic evidence linking adiponectin[ 32 — 36 ] or leptin[ 37 — 39 ] individually to cancer risk is mixed. Intermittent CR suppresses murine mammary tumor incidence in association with decreased leptin-to-adiponectin ratio[ 32 ].

Associations between the leptin-to-adiponectin ratio and the metabolic syndrome[ 40 — 42 ] and some cancers[ 43 — 45 ] have been reported, but further characterization of these links is needed.

Chronic inflammation is characterized by increased circulating free fatty acids, cytokines, and chemokines that attract immune cells such as macrophages that also produce inflammatory mediators into the local microenvironment[ 46 — 48 ]. The inflammatory cascade is further amplified by the release of inflammatory cytokines, such as interleukin IL -1β, IL-6, TNF-α, and monocyte chemoattractant protein-1, primarily from macrophages, into the local and systemic circulation.

Adipocytes can enlarge past the point of effective oxygen diffusion, which results in hypoxia and eventually necrosis. Free fatty acids escape the engorged or necrotic adipocytes and deposit in other tissues, and this in turn promotes insulin resistance, diabetes through downregulation of insulin receptors and glucose transporters , hepatic steatosis, and pancreatic steatosis, and also activates signaling molecules involved in epithelial carcinogenesis, such as NF-κB and cyclooxygenase COX -2[ 49 ].

The transcription factor NF-κB is activated in response to bacterial and viral stimuli, growth factors, and inflammatory molecules for example, TNF-α, IL-6, and IL-1β , and is responsible for inducing gene expression associated with cell proliferation, apoptosis, inflammation, metastasis, and angiogenesis.

Activation of NF-κB is a common characteristic of many tumors and is associated with insulin resistance and elevated circulating levels of leptin, insulin, or IGF-1[ 46 , 50 , 51 ].

A connection between chronic inflammation and cancer development was observed years ago when Rudolph Virchow noted an abundance of leukocytes in neoplastic tissue[ 52 ]. Indeed, several tissue-specific inflammatory lesions are established neoplastic precursors for invasive cancer, including inflammatory bowel disease for colon cancer, pancreatitis for pancreatic cancer, dermatitis for certain forms of skin cancer, and gastritis for gastric cancer[ 56 , 57 ].

Tumor and preneoplastic microenvironments are composed of mixtures of cell types including epithelial cells, fibroblasts, mast cells, and cells of the innate and adaptive immune system[ 58 ].

As discussed previously, macrophages, which are activated in the obese state, infiltrate tumors and amplify the inflammatory tumor microenvironment, often through NF-κB-dependent production of cytokines and angiogenic factors[ 58 ].

COX-2 is another important cancer-related inflammatory mediator that is upregulated in most tumors and catalyzes the synthesis of the potent inflammatory lipid metabolite, prostaglandin E 2. COX-2 expression, an indicator of poor prognosis in many cancer types, is increased in response to obesity[ 59 ].

Calorie restriction can prevent much of the inflammation associated with preneoplasia or neoplasia[ 46 , 60 — 62 ]. Specifically, CR decreases the number of tumor-infiltrating macrophages, levels of circulating and tissue cytokines, and NF-κB signaling and COX-2 expression in many tissues and tumor types[ 46 , 61 , 62 ].

Thus evidence is accumulating that the anti-inflammatory effects of CR contribute significantly to its cancer preventive effects[ 1 , 46 ]. Imbalances in the production or interactions of several factors influence key functions of the endothelium, including its roles in regulating angiogenesis, hemostasis, vascular density, inflammation, and vascular wall integrity.

One such factor is PAI-1, a serine protease inhibitor produced by endothelial cells, stromal cells, and adipocytes in visceral white adipose tissue[ 63 ].

PAI-1, through its inhibition of urokinase-type and tissue-type plasminogen activators, regulates fibrinolysis and integrity of the extracellular matrix[ 64 ]. Increased circulating PAI-1 levels, frequently found in obese subjects, are associated with increased risk of atherogenesis and cardiovascular disease, diabetes, and several cancers[ 63 — 66 ].

PAI-1 is also involved in angiogenesis and thus may contribute to obesity-driven tumor cell growth, invasion, and metastasis[ 66 ]. Circulating levels of PAI-1 are consistently decreased in response to CR[ 1 ], although the mechanistic link between PAI-1 and cancer requires further study.

Another important mediator of vascular integrity is the heparin-binding glycoprotein vascular endothelial growth factor VEGF produced by adipocytes and tumor cells.

VEGF has mitogenic, angiogenic, and vascular permeability-enhancing activities specific for endothelial cells[ 67 ]. The need for nutrients and oxygen triggers tumor cells to produce VEGF, which leads to the formation of new blood vessels angiogenesis to nourish the rapidly growing tumor.

VEGF may also facilitate the metastatic spread of tumors cells[ 68 ]. Adipocytes communicate with endothelial cells by producing a variety of pro-angiogenic and vascular permeability-enhancing factors, including VEGF and PAI-1[ 69 ].

In the obese, nontumor setting, these factors stimulate neovascularization in support of the expanding fat mass[ 69 ]. Circulating levels of VEGF are increased in obese, relative to lean, human beings and animals, and increased tumoral expression of VEGF is associated with poor prognosis in several obesity-related cancers[ 70 — 73 ].

Data to date for several experimental tumor models[ 71 — 73 ] suggest that CR decreases systemic and tissue VEGF and has anti-angiogenic effects. The sirtuin family of proteins has been implicated in the regulation of endocrine signaling, stress-induced apoptosis, and the metabolic changes associated with energy balance modulation and aging[ 74 — 76 ].

Sirtuins were originally studied in yeast and nematodes, where CR increases lifespan in association with the levels and activity of the Sir2 protein[ 77 — 79 ]. The levels of Sir2, or its mammalian homolog SIRT1, rise in response to CR[ 75 — 79 ]. SIRT1 is an NAD-dependent deacetylase that inhibits stress-induced apoptotic cell death, and may modulate IGF-1, adiponectin, and insulin production, and insulin sensitivity, in different tissues[ 79 — 81 ].

The specific roles of sirtuins in cancer development or progression are not yet clear. SIRT1 is upregulated in several tumor types and can inhibit apoptosis and downregulate the expression of tumor suppressor genes to enhance survival of epithelial cancer cells[ 82 — 85 ].

In addition, the SIRT1 activator SRT promotes tumor cell migration and lung metastases in a murine breast cancer model[ 86 ]. In contrast, there is also evidence that SIRT1 can act to suppress polyp formation in the APC Min intestinal tumor model[ 87 ].

Additionally, in preclinical studies the phytochemical resveratrol activates SIRT1 and reduces cancer development in several models[ 88 ]. SIRT1-overexpression did not influence the anticancer effects of an every-other-day fasting regimen a variation of CR in a pdeficient mouse model of cancer, suggesting that SIRT1 may have a limited role in the effects of CR on cancer[ 89 ].

Given the conflicting data to date regarding the tumor-enhancing, versus inhibitory, effects of SIRT1 activation, and the apparently limited role of SIRT1 in the response to CR, it remains unclear whether SIRT1 or other sirtuins represent mechanistic targets for cancer prevention.

Autophagy is a cellular degradation pathway involved in the clearance of damaged or unnecessary proteins and organelles. It also provides an alternative source of energy and substrates during periods of restricted dietary intake such as CR or metabolic stress to enhance survival.

One of the proposed mechanisms of CR is that under conditions of nutrient limitation, there is a shift in metabolic investment from cell replication and growth to maintenance, to ensure extended survival[ 92 ].

This tightly regulated process is driven by a group of autophagy-related proteins, and is suppressed by the conserved nutrient sensor TOR[ 93 ]. CR regulates TOR complex 1 and, to a lesser extent TOR complex 2, in many species, including flies, worms, yeast, and mammals.

TOR complex 1 signaling regulates protein translation and many cellular processes, including metabolism and autophagy[ 93 ]. In addition, suppression of nutrient-activated TOR signaling is sufficient to trigger an energy stress response that is coordinated by AMPK, and this metabolic program blunts the growth responses to nutrient availability and promotes autophagy[ 94 ].

Several longevity-promoting regimens, including inhibition of TOR with rapamycin, resveratrol, or the natural polyamine spermidine, may require autophagy for their effects[ 95 ].

Autophagy activation is essential for clearing cellular damage and disease prevention in normal cells, and tumor cells also utilize autophagy to maintain a favorable metabolic state for daughter cell production, especially under limiting nutrient conditions[ 96 ]. However, little is known about what role autophagy plays in CR-mediated effects on tumor development or progression.

The identification and development of natural or synthetic agents that mimic some of the protective effects of CR may facilitate new strategies for cancer prevention. Given how difficult it is for many people to adopt a low-calorie diet for an extended period, the identification of drugs or other agents that could either complement or even reproduce the anticancer effects of CR without drastic changes in diet and lifestyle is a goal for many pharmaceutical companies.

Numerous studies have used microarray analyses to profile the molecular targets responding to CR and other dietary energy balance modulations[ 97 — ]. Most of these studies were focused on understanding CR effects related to aging, and they revealed that the extent to which CR modulates the transcriptome is species-specific, tissue-specific, and dependent on the duration and intensity of CR.

Nonetheless, some emerging patterns from these studies suggest that transcripts involved in inflammation, growth factor signaling particularly related to the insulin and IGF-1 pathways , oxidative stress, and nutrient metabolism are commonly altered by CR.

Application of the emerging field of metabolomics to this question should accelerate the identification of additional targets. Sirtuin modulators, including resveratrol and its analogs, and pharmacologic modulators of SIRT1[ 82 ], exert some anticancer activity, although much of this work has been limited to in vitro systems and awaits verification in vivo.

Agents or interventions that safely reduce IGF-1, or inhibit one or more components of the signaling pathways downstream of IGF-1 and other growth factors including Akt and mTOR without requiring drastic dietary changes, may provide an effective physiological or pharmacological mimetic of those effects.

The hope is that these agents or interventions could be readily adopted by a large proportion of the population, particularly those unable to lose weight and at high risk of cancer or other chronic diseases associated with obesity.

As recently reviewed[ 16 , ], antireceptor antibodies, small-molecule receptor kinase inhibitors, and to a lesser extent anti-IGF ligand antibodies are being developed to target the IR or IGF-1 receptor, and several promising agents from each of these classes have advanced to clinical trials.

The antireceptor antibodies have been the subject of the most intense translational research activity, extending to phase 3 trials, while the other classes are currently in phase 1 or phase 2 trials.

The various antireceptor antibodies that have been developed were designed to avoid IR inhibition blocking IR would be likely to have significant adverse effects , and this is generally being accomplished.

Each targets ligand binding to the IGF-IR, and preliminary evidence suggests that the effects extend to hybrid receptors. Despite the lack of interference with insulin binding, the use of these antibodies causes hyperglycemia and hyperinsulinemia, and can also lead to increased levels of serum IGF-1 in compensation for the reduced IGF-IR signaling.

This can contribute to insulin resistance in patients receiving these antibodies, and these untoward effects, along with generally disappointing trial results to date, is limiting the pace of development of these agents[ ].

Although the initial development of small-molecule tyrosine kinase inhibitors involved attempts to achieve IGF-IR specificity, the newer agents tend to partially inhibit several members of the insulin and IGF-1 receptor family, which may limit side effects and provide a therapeutic advantage of more specific inhibitors.

Early clinical experience suggests that these agents are safer than was originally anticipated, possibly because the drug concentrations that are achieved are fairly low in muscle, which is a major metabolic regulator, perhaps accounting for a modest rather than a severe effect of these kinase inhibitors on metabolic perturbations.

Nevertheless, insulin levels are generally increased in patients treated with these kinase inhibitors, possibly limiting their efficacy and the pace of their development[ 16 ].

Pharmacological mTOR inhibitors have emerged as lead candidates for CR mimetics. Rapamycin treatment extends lifespan and delays cancer in mice, providing additional support for mTOR as a target for mimicking the effects of CR[ ].

We have shown that rapamycin or its analog, RAD everolimus , can offset the obesity-associated increased growth of mammary or pancreatic tumors[ 25 , 62 ].

Rapamycin is a potent inhibitor of the mTOR complex 1, but chronic rapamycin exposure has been linked in some studies to disruption of mTOR complex 2 signaling, resulting in impaired glucose tolerance and insulin action[ ].

Thus while inhibiting mTOR complex 1 appears to be a good strategy for mimicking many of the anticancer effects of CR, the search for agents that can do so without disrupting mTOR complex 2 signaling is ongoing.

An mTOR-inhibiting drug with great promise as a CR mimetic that overcomes the concerns about glucose intolerance associated with rapamycin is metformin, a biguanide commonly used to treat type 2 diabetes[ ]. Metformin inhibits gluconeogenesis through indirect activation of AMPK in the liver and possibly cancer cells, and may also exert direct effects on AMPK in cancer cells[ ].

Administration of metformin suppresses tumor development or growth in multiple experimental models, including colon, mammary, and hematopoietic cancer models[ ].

Epidemiological studies have suggested that type 2 diabetic patients treated with metformin have a lower risk of developing from or dying from cancer, relative to diabetic patients receiving sulfonylurea, insulin, or other therapies[ — ]. A randomized trial is now underway to evaluate the effect of metformin on breast cancer recurrence[ ].

Phenformin, another biguanide that has been abandoned for diabetes therapy due to its toxicity from lactic acidosis is a more potent AMPK inhibitor than metformin and may also have some potential as a CR mimetic at lower, nontoxic doses[ ].

An emerging issue in the area of mTOR inhibitors as CR mimetics is that of the relative effects of nature versus nurture, that is, the contribution of systemic factors which has been the focus of this review in the context of cell autonomous effects.

The observations of Kalaany and Sabatini[ ] that cancer cells with constitutively activated PI3K mutations are proliferative in vitro in the absence of insulin or IGF-1 and form CR-resistant tumors in vivo illustrate this issue.

We also found that constitutive activation of mTOR in MMTV-Wnt-1 mammary tumor cells blocked the anticancer effects of CR[ 26 ]. These findings suggest that cell autonomous alterations, such as activating mutations of PI3K or downstream mTOR pathway components, may influence the response of cells to CR or CR mimetics.

Another emerging issue is that, in addition to impacting the growth and survival of aberrant cells, CR and mTOR inhibition may also affect the stem cell compartment and enhance maintenance or repair of tissues. Yilmaz and colleagues[ ] showed that CR, through its inhibitory effects of mTOR signaling in Paneth cells immune-related support cells in the stem cell niche adjacent to intestinal stem cells, preserves and even enriches intestinal stem cells.

The augmenting effects of CR via Paneth cells on intestinal stem cell self-renewal can be mimicked by rapamycin. Cerletti et al. In addition, we showed that mammary tumors highly enriched in breast cancer stem cells have heightened sensitivity to the anticancer effects of CR[ ].

Calorie reduction almost completely ablates M-Wnt tumor growth relative to tumors induced by E-Wnt cells, also cloned from a MMTV-Wnt-1 tumor but with basal-like epithelial morphology and low expression of stem cell markers.

Furthermore, CR promotes a mesenchymal-to-epithelial transition in the mammary gland by increasing the expression of the epithelial markers, such as E-cadherin, and decreasing the expression of mesenchymal markers, such as N-cadherin and fibronectin[ ].

Taken together, these studies suggest an important role for the microenvironment in the response of stem cells including cancer stem cells to CR or CR mimetics targeting the mTOR pathway, and this will no doubt be an important and exciting research area in the coming years. As summarized in Figure 1 , this review considers lessons learned from CR and cancer research to discuss promising molecular targets for cancer prevention, particularly for breaking the link between obesity and cancer.

Potential targets include components of energy-responsive growth factor and adipokine signaling pathways, inflammatory pathways, vascular regulators, autophagy regulators, and the sirtuin pathway.

Clearly, no single pathway accounts for all of the anticancer effects of CR. As with most chronic disease intervention strategies, combination approaches involving lifestyle including diet and physical activity and pharmacological interventions that target multiple pathways and that maximize efficacy and minimize adverse effects are likely to be most successful for preventing cancer.

Future studies aimed at further elucidating the mechanisms underlying the anticancer effects of CR, and that exploit this mechanistic information to target CR-responsive pathways will facilitate the translation of CR research into effective cancer prevention strategies in human beings. In this review we discussed possible mechanisms underlying the anticancer effects of CR, with emphasis on CR-associated changes in growth factor signaling, inflammation, and angiogenesis, as well as emerging evidence suggesting that autophagy and the sirtuin pathway may also play roles in the effects of CR on tumor development and progression.

Several natural or synthetic agents have been shown to mimic some of the protective effects of CR and may thus represent new strategies for cancer prevention. Hursting SD, Smith SM, Lashinger LM, Harvey AE, Perkins SN: Calories and carcinogenesis: lessons learned from 30 years of calorie restriction research.

CAS PubMed Google Scholar. Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, Simmons HA, Kemnitz JW, Weindruch R: Caloric restriction delays disease onset and mortality in rhesus monkeys. PubMed Central CAS PubMed Google Scholar.

Mattison JA, Roth GS, Beasley TM, Tilmon EM, Handy AM, Herbert RL, Longo DL, Allison DB, Young JE, Bryant M, Barnard D, Ward WF, Qi W, Ingram DK, de Cabo R: Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study.

Harvie M, Howell A: Energy restriction and the prevention of breast cancer. Proc Nutr Soc. Cancer Res. Kagawa Y: Impact of Westernization on the nutrition of Japanese: changes in physique, cancer, longevity and centenarians. Prev Med. Michels KB, Ekbom A: Caloric restriction and incidence of breast cancer.

J Am Med Assoc. CAS Google Scholar. Tretli S, Gaard M: Lifestyle changes during adolescence and risk of breast cancer: an ecologic study of the effect of World War II in Norway. Cancer Causes Contr. Elias SG, Peeters PH, Grobbee DE, van Noord PA: The — Dutch famine and subsequent overall cancer incidence.

Cancer Epidemiol Biomarkers Prev. PubMed Google Scholar. Keinan-Boker L, Vin-Raviv N, Lipshitz I, Linn S, Barchana M: Cancer incidence in Israeli Jewish survivors of World War II. J Natl Cancer Inst. Koupil I, Plavinskaja S, Parfenova N, Shestov DB, Danziger PD, Vagero D: Cancer mortality in women and men who survived the siege of Leningrad — Int J Cancer.

Hursting SD, Forman MR: Cancer risk from extreme stressors: lessons from European Jewish survivors of World War II. Heilbronn LK, de Jonge L, Frisard MI, DeLany JP, Larson-Meyer DE, Rood J, Nguyen T, Martin CK, Volaufova J, Most MM, Greenway FL, Smith SR, Deutsch WA, Williamson DA, Ravussin E: Effect of 6-month calorie restriction on biomarkers of longevity, metabolic adaptation, and oxidative stress in overweight individuals: a randomized controlled trial.

Google Scholar. Redman LM, Heilbronn LK, Martin CK, de Jonge L, Williamson DA, Delany JP, Ravussin E: Metabolic and behavioral compensations in response to caloric restriction: implications for the maintenance of weight loss.

PLoS One. PubMed Central PubMed Google Scholar. Colditz GA, Wollin KY, Gehlert S: Applying what we know to accelerate cancer prevention.

Sci Transl Med. Pollak M: The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. Hursting SD, Berger NA: Energy balance, host-related factors, and cancer progression.

J Clin Oncol. Gallagher EJ, Fierz Y, Ferguson RD, LeRoith D: The pathway from diabetes and obesity to cancer, on the route to targeted therapy. Endocr Pract. Price AJ, Allen NE, Appleby PN, Crowe FL, Travis RC, Tipper SJ, Overvad K, Gronbæk H, Tjonneland A, Fons Johnsen N, Rinaldi S, Kaaks R, Lukanova A, Boeing H, Aleksandrova K, Trichopoulou A, Trichopoulos D, Andarakis G, Palli D, Krogh V, Tumino R, Sacerdote C, Bueno-de-Mesquita HB, Arguelles MV, Sanchez MJ, Chirlaque MD, Barricarte A, Larranaga N, Gonzalez CA, Stattin P: Insulin-like growth factor-1 concentration and risk of prostate cancer: results from the European Prospective Investigation into Cancer and Nutrition.

Cancer Epidemiol Biom Prev. Garcia-Cao I, Song MS, Hobbs R, Laurent G, Giorgi C, de Boer VC, Anastasiou D, Ito K, Sasaki AT, Rameh L, Carracedo A, VanderHeiden MG, Cantley LC, Pinton P, Haigis MC, Pandolfi PP: Systemic elevation of PTEN induces a tumor-suppressive metabolic state.

Memmott RM, Dennis PA: Akt-dependent and -independent mechanisms of mTOR regulation in cancer. Cell Signal. Cancer Prev Res. Lindsley JE, Rutter J: Nutrient sensing and metabolic decisions. Comp Biochem Physiol B Biochem Mol Biol.

Mol Carcinog. Nogueira LM, Dunlap SM, Ford NA, Hursting SD: Calorie restriction and rapamycin inhibit MMTV-Wnt-1 mammary tumor growth in a mouse model of postmenopausal obesity.

Endocr Relat Cancer. Vaiopoulos AG, Marinou K, Christodoulides C, Koutsilieris M: The role of adiponectin in human vascular physiology. Int J Cardiol. Barb D, Williams CJ, Neuwirth AK, Mantzoros CS: Adiponectin in relation to malignancies: a review of existing basic research and clinical evidence.

Am J Clin Nutr. Stofkova A: Leptin and adiponectin: from energy and metabolic dysbalance to inflammation and autoimmunity. Endocr Regul. Gautron L, Elmquist JK: Sixteen years and counting: an update on leptin in energy balance. J Clin Invest. Villanueva EC, Myers MG: Leptin receptor signaling and the regulation of mammalian physiology.

Int J Obes. Rogozina OP, Bonorden MJ, Seppanen CN, Grande JP, Cleary MP: Effect of chronic and intermittent calorie restriction on serum adiponectin and leptin and mammary tumorigenesis.

Dalamaga M, Diakopoulos KN, Mantzoros CS: The role of adiponectin in cancer: a review of current evidence. Endocr Rev. Grossmann ME, Nkhata KJ, Mizuno NK, Ray A, Cleary MP: Effects of adiponectin on breast cancer cell growth and signaling.

Br J Cancer. Rzepka-Gorska I, Bedner R, Cymbaluk-Ploska A, Chudecka-Glaz A: Serum adiponectin in relation to endometrial cancer and endometrial hyperplasia with atypia in obese women. Eur J Gynaecol Oncol. Tian YF, Chu CH, Wh MH, Chang CL, Yang T, Chou YC, Hsu GC, Yu CP, Yu JC, Sun CA: Anthropometric measures, plasma adiponectin, and breast cancer risk.

Stattin P, Lukanova A, Biessy C, Soderberg S, Palmqvist R, Kaaks R, Olsson T, Jellum E: Obesity and colon cancer: does leptin provide a link?. Wu MH, Chou YC, Chou WY, Hsu GC, Chu CH, Yu CP, Yu JC, Sun CA: Circulating levels of leptin, adiposity and breast cancer risk.

Korean Diabetes J. Clin Invest Med. Cleary MP, Ray A, Rogozina OP, Dogan S, Grossman ME: Targeting the adiponectin: leptin ratio for postmenopausal breast cancer prevention.

Front Biosci. Ashizawa N, Yahata T, Quan J, Adachi S, Yoshihara K, Tanaka K: Serum leptin-adiponectin ratio and endometrial cancer risk in postmenopausal female subjects.

Gynecol Oncol. Chen DC, Chung YF, Yeh YT, Chaung HC, Kuo FC, Fu OY, Chen HY, Hou ME, Yuan SS: Serum adiponectin and leptin levels in Taiwanese breast cancer patients.

Cancer Lett. Harvey AE, Lashinger LM, Hursting SD: The growing challenge of obesity and cancer: an inflammatory subject. Ann NY Acad Sci. Subbaramaiah K, Howe LR, Bhardway P, Du B, Gravaghi C, Yantiss RK, Zhou XK, Blaho VA, Hia T, Yang P, Kopelovich L, Hudis CA, Dannenberg AJ: Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland.

Olefsky JM, Glass CK: Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. Renehan AG, Roberts DL, Dive C: Obesity and cancer: pathophysiological and biological mechanisms.

Arch Physiol Biochem. Karin M: Nuclear factor-κB in cancer development and progression. Virchow R: Die krankhaften Geschwülste. Aggarwal BB, Gehlot P: Inflammation and cancer: how friendly is the relationship for patients?.

Curr Opin Pharmacol. Ono M: Molecular links between tumor angiogenesis and inflammation: inflammatory stimuli of macrophages and cancer cells as targets for therapeutic strategy.

Cancer Sci. Del Prete A, Allavena P, Santoro G, Fumarulo R, Corsi MM, Mantovani A: Molecular pathways in cancer-related inflammation. Biochem Med. Foltz CJ, Fox JG, Cahill R, Murphy JC, Yan L, Shames B, Schauer DB: Spontaneous inflammatory bowel disease in multiple mutant mouse lines: association with colonization by Helicobacter hepaticus.

Coussens LM, Werb Z: Inflammation and cancer. Allavena P, Sica A, Garlanda C, Mantovani A: The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance.

Immunol Rev. Koki A, Khan NK, Woerner BM, Dannenberg AJ, Olson L, Seibert K, Edwards D, Hardy M, Isakson P, Masterrer JL: Cyclooxygenase-2 in human pathological disease. Adv Exp Med Biol. Perkins SN, Hursting SD, Phang JM, Haines DC: Calorie restriction reduces ulcerative dermatitis and infection-related mortality in pdeficient and wild-type mice.

J Invest Dermatol. Harvey A, Lashinger L, Otto G, Malone L, Hursting SD: Decreased systemic insulin-like growth factor-1 in response to calorie restriction modulates tumor growth, NF-κB activation, and inflammation-related gene expression.

Lashinger LM, Malone LM, Brown GW, Daniels EA, Goldberg JA, Otto G, Fischer SM, Hursting SD: Rapamycin partially mimics the anticancer effects of calorie restriction in a murine model of pancreatic cancer.

Iwaki T, Urano T, Umemura K: PAI-1, progress in understanding the clinical problem and its aetiology. Br J Haematol. Skurk T, Hauner H: Obesity and impaired fibrinolysis: role of adipose production of plasminogen activator inhibitor Int J Obes Relat Metab Disord.

Carter JC, Church FC: Obesity and breast cancer: the roles of peroxisome proliferator-activated receptor-gamma and plasminogen activator inhibitor PPAR Res. McMahon GA, Petitclerc E, Stefansson S, Smith E, Wong MK, Westwick RJ, Ginsburg D, Brooks PC, Lawrence DA: Plasminogen activator inhibior-1 regulates tumor growth and angiogenesis.

J Biol Chem. Byrne AM, Bouchier-Hayes DJ, Harmey JH: Angiogenic and cell survival functions of vascular endothelial growth factor VEGF. J Cell Mol Med. Saharinen P, Eklund L, Pulkki K, Bono P, Alitalo K: VEGF and angiopoietin signaling in tumor angiogenesis and metastasis.

Trends Mol Med. Cao Y: Angiogenesis modulates adipogenesis and obesity. Breast Cancer Res Treat. Powolny AA, Wang S, Carlton PS, Hoot DR, Clinton SK: Interrelationships between dietary restriction, the IGF-1 axis, and expression of vascular endothelial growth factor by prostate adenocarcinoma in rats.

Lashinger LM, Malone LM, MacArthur MJ, Goldberg JA, Daniels EA, Pavone A, Colby JK, Smith NC, Perkins SN, Fischer SM, Hursting SD: Genetic reduction of insulin-like growth factor-1 mimics the anticancer effects of calorie restriction on cyclooxygenasedriven pancreatic neoplasia.

Blando J, Moore T, Hursting SD, Jiang G, Saha A, Beltran L, Shen J, Repass J, Strom S, DiGiovanni J: Dietary energy balance modulates prostate cancer progression in Hi-Myc mice.

Yang T, Fu M, Pestell R, Sauve AA: SIRT1 and endocrine signaling. Trends Endocrinol Metab. Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, Howitz KT, Gorospe M, de Cabo R, Sinclair DA: Calorie restriction promotes mammalian cell survival by inducing Sirt1 deacetylase.

Metoyer CF, Pruitt K: The role of sirtuin proteins in obesity. Lin SJ, Defossez PA, Guarente L: Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Tissenbaum HA, Guarente L: Increased dosage of a sir - 2 gene extends lifespan in Caenorhabditis elegans.

Bordone L, Cohen D, Robinson A, Motta MC, van Veen E, Czopik A, Steele AD, Crowe H, Marmor S, Luo J, Gu W, Guarente L: SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell. Ramsey KM, Mills KF, Satoh A, Imai S: Age-associated loss of Sirt1-mediated enhancement of glucose-stimulated insulin secretion in beta cell-specific Sirt1-overexpressing BESTO mice.

Nemoto S, Fergusson MM, Finkel T: SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1α. Aljada A, Dong L, Mousa SA: Sirtuin-targeting drugs: mechanisms of action and potential therapeutic applications.

Nutrition resgriction all physiological processes occurring anc our body, preventipn those related to the function of the immune system; indeed, metabolism has been closely associated with the differentiation and activity of Holistic cancer prevention methods innate and adaptive immune Kiwi fruit varieties. While excessive energy caliric and adiposity Natural metabolism-boosting lifestyle been EGCG and digestive health to cause systemic resriction, several festriction and experimental evidence show that calorie restriction Caloric restriction and disease preventionnot restriiction to preventlon, is able restrictioj delay aging and exert diseaee anti-inflammatory effects in different pathological conditions. This review provides an overview of the ability of different CR-related nutritional strategies to control autoimmune, cardiovascular, and infectious diseases, as tested by preclinical studies and human clinical trials, with a specific focus on the immunological aspects of these interventions. In particular, we recapitulate the state of the art on the cellular and molecular mechanisms pertaining to immune cell metabolic rewiring, regulatory T cell expansion, and gut microbiota composition, which possibly underline the beneficial effects of CR. Although studies are still needed to fully evaluate the feasibility and efficacy of the nutritional intervention in clinical practice, the experimental observations discussed here suggest a relevant role of CR in lowering the inflammatory state in a plethora of different pathologies, thus representing a promising therapeutic strategy for the control of human health. This article is part of the Spotlight issue on Obesity, Metabolism, and Diabetes. Over the last few decades, lifestyle in the most industrialized countries has undergone profound changes, in particular in the eating habits. Thank you for visiting nature. You are using a browser version with DEXA scan results support for CSS. To obtain the best rrestriction, we recommend you use Natural metabolism-boosting lifestyle faloric up to date browser or turn off anf mode in Internet EGCG and digestive health. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. Nutriments have been deemed to impact all physiopathologic processes. Recent evidences in molecular medicine and clinical trials have demonstrated that adequate nutrition treatments are the golden criterion for extending healthspan and delaying ageing in various species such as yeast, drosophila, rodent, primate and human. It emerges to develop the precision-nutrition therapeutics to slow age-related biological processes and treat diverse diseases.

Author: Sagul

2 thoughts on “Caloric restriction and disease prevention

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com