Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions
Abstract
:1. Introduction
2. Clinical Relevance and Features of CRA
3. Iron Homeostasis
4. Pathogenesis of CRA
Potential Role of Microbiota in Iron Metabolism
5. First-Line Therapy
6. Evidence of CRA Modulation by Lifestyle Interventions in Inflammatory Conditions
6.1. Mediterranean Diet: Microbiota-Mediated Anti-Inflammatory Effects
6.2. Supplementation to Reduce Chronic Inflammation and Potentially Counteract Anemia in Cancer Patients
6.2.1. Polyphenols
6.2.2. Curcumin
6.2.3. Resveratrol
6.2.4. Quercetin
6.2.5. Epigallocatechin Gallate
6.2.6. Vitamin D Supplementation
6.2.7. Lipoic Acid
6.2.8. Lactoferrin
6.2.9. Omega 3 Fatty Acids
6.2.10. Probiotics
Dietary Supplementation | Study Design (R/MA) | Effects | Major Targets | ||
---|---|---|---|---|---|
Anti-Inflammatory | Antioxidant | Others | |||
Iron | R [274] | Replenishment of iron stores | 1α-hydroxylase, IL-6, IL-1β, MCP-1 | ||
Vit. D | R/MA [275] | Yes | Stimulation of erythroid progenitor cell proliferation, reduction of hepcidin production | ||
Polyphenols | R [152] | Yes | Yes | Modulation of gut microbiota and mucosal integrity, anti-infective | NFkB, COX, NOX, SOD |
Curcumin | MA [276] | Yes | Antineoplastic | TNF-α, IL-1β, COX, ICAM-1, VCAM-1, STAT3, NfKb, TLR, PPAR | |
Resveratrol | MA [277] | Yes | Yes | Cardioprotective | COX, PPAR, eNOS, NFkB, VCAM-1 |
Quercetin | MA [278] | Yes | Metabolic | NFkB, NOS, COX-2, TNF-α, adiponectin | |
Epigallocatechin gallate | R [193] | Yes | Yes | Immunomodulation | NFkB, iNOS, NO |
Lipoic acid | MA [279] | Yes | Yes | Antineoplastic | VCAM-1, MMP-9 NFkB, eNOS, Nrf2 |
Lactoferrin | R [280] | Yes | Yes | Regulation of iron absorption, immunomodulation, anti-microbial, anti-viral, antineoplastic | COX-1, IL-10, INF-γ, TNF-α |
Omega 3 | MA [281] | Yes | Metabolic | NFkB, PPAR-γ, IL-6 | |
Probiotics | MA [268] | Yes | restoration of microbial balance, immunomodulation, metabolic, promotion of enterocyte iron uptake | Hepcidin, p-hydroxyphenyllactic acid |
7. Potential Role of Exercise in CRA
7.1. The Endocrine Function of the Skeletal Muscle and the Immunomodulatory Function of Exercise
7.2. Exercise Prescription: The Possible Role of Acute and Chronic Exercise
7.3. Overview of Evidence of the Effects of Acute Exercise on the Immune Response in Patients with Chronic Inflammatory Diseases
7.4. Overview of Evidence of the Effects of Chronic Exercise on the Immune Response in Patients with a Chronic Inflammatory Disease
7.5. Overview of Evidence of the Effects of Acute and Chronic Exercise on Serum Hepcidin Levels
7.6. Overview of Evidence of the Effects of Acute and Chronic Exercise on Microbiota Diversity
7.7. Overview of the Role and Efficacy of Exercise Interventions in Patients with Cancer
8. Future Direction: A Multitarget Approach and the Age of COVID-19
9. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Madeddu, C.; Gramignano, G.; Astara, G.; Demontis, R.; Sanna, E.; Atzeni, V.; Macciò, A. Pathogenesis and Treatment Options of Cancer Related Anemia: Perspective for a Targeted Mechanism-Based Approach. Front. Physiol. 2018, 9, 1294. [Google Scholar] [CrossRef]
- Ludwig, H.; Van Belle, S.; Barrett-Lee, P.; Birgegård, G.; Bokemeyer, C.; Gascón, P.; Kosmidis, P.; Krzakowski, M.; Nortier, J.; Olmi, P.; et al. The European Cancer Anaemia Survey (ECAS): A large, multinational, prospective survey defining the prevalence, incidence, and treatment of anaemia in cancer patients. Eur. J. Cancer 2004, 40, 2293–2306. [Google Scholar] [CrossRef]
- Maccio, A.; Madeddu, C.; Gramignano, G.; Mulas, C.; Tanca, L.; Cherchi, M.C.; Floris, C.; Omoto, I.; Barracca, A.; Ganz, T. The role of inflammation, iron, and nutritional status in cancer-related anemia: Results of a large, prospective, observational study. Haematologica 2015, 100, 124–132. [Google Scholar] [CrossRef] [Green Version]
- Kundu, J.K.; Surh, Y.J. Emerging avenues linking inflammation and cancer. Free Radic. Biol. Med. 2012, 52, 2013–2037. [Google Scholar] [CrossRef]
- Neumcke, I.; Schneider, B.; Fandrey, J.; Pagel, H. Effects of pro-and antioxidative compounds on renal production of erythropoietin. Endocrinology 1999, 140, 641–645. [Google Scholar] [CrossRef] [PubMed]
- Macciò, A.; Madeddu, C.; Massa, D.; Mudu, M.C.; Lusso, M.R.; Gramignano, G.; Serpe, R.; Melis, G.B.; Mantovani, G. Hemoglobin levels correlate with interleukin-6 levels in patients with advanced untreated epithelial ovarian cancer: Role of inflammation in cancer-related anemia. Blood 2005, 106, 362–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Falkensammer, C.E.; Thurnher, M.; Leonhartsberger, N.; Ramoner, R. C-reactive protein is a strong predictor for anaemia in renal cell carcinoma: Role of IL-6 in overall survival. BJU Int. 2011, 107, 1893–1898. [Google Scholar] [CrossRef] [PubMed]
- Birgegård, G.; Aapro, M.S.; Bokemeyer, C.; Dicato, M.; Drings, P.; Hornedo, J.; Krzakowski, M.; Ludwig, H.; Pecorelli, S.; Schmoll, H.; et al. Cancer-related anemia: Pathogenesis, prevalence and treatment. Oncology 2005, 68 (Suppl. 1), 3–11. [Google Scholar] [CrossRef]
- Schwartz, R.N. Anemia in patients with cancer: Incidence, causes, impact, management, and use of treatment guidelines and protocols. Am. J. Health Syst. Pharm. 2007, 64, S5–S13. quiz S28–30. [Google Scholar] [CrossRef] [PubMed]
- Knight, K.; Wade, S.; Balducci, L. Prevalence and outcomes of anemia in cancer: A systematic review of the literature. Am. J. Med. 2004, 116 (Suppl. 7A), S11–S26. [Google Scholar] [CrossRef]
- Caro, J.J.; Salas, M.; Ward, A.; Goss, G. Anemia as an independent prognostic factor for survival in patients with cancer: A systemic, quantitative review. Cancer 2001, 91, 2214–2221. [Google Scholar] [CrossRef]
- Dunn, A.; Carter, J.; Carter, H. Anemia at the end of life: Prevalence, significance, and causes in patients receiving palliative care. J. Pain Symptom Manag. 2003, 26, 1132–1139. [Google Scholar] [CrossRef] [PubMed]
- Bower, J.E. Cancer-related fatigue--mechanisms, risk factors, and treatments. Nat. Rev. Clin. Oncol. 2014, 11, 597–609. [Google Scholar] [CrossRef] [PubMed]
- Delano, M.J.; Moldawer, L.L. The origins of cachexia in acute and chronic inflammatory diseases. Nutr. Clin. Pract. 2006, 21, 68–81. [Google Scholar] [CrossRef] [PubMed]
- Macdougall, I.C. Quality of life and anemia: The nephrology experience. Semin. Oncol. 1998, 25, 39–42. [Google Scholar] [PubMed]
- Ludwig, H.; Fritz, E. Anemia in cancer patients. Semin. Oncol. 1998, 25, 2–6. [Google Scholar]
- Ludwig, H.; Strasser, K. Symptomatology of anemia. Semin. Oncol. 2001, 28, 7–14. [Google Scholar] [CrossRef]
- Ganz, T. Hepcidin and iron regulation, 10 years later. Blood 2011, 117, 4425–4433. [Google Scholar] [CrossRef] [Green Version]
- Ganz, T.; Nemeth, E. Hepcidin and iron homeostasis. Biochim. Et Biophys. Acta 2012, 1823, 1434–1443. [Google Scholar] [CrossRef] [Green Version]
- Michels, K.; Nemeth, E.; Ganz, T.; Mehrad, B. Hepcidin and Host Defense against Infectious Diseases. PLoS Pathog. 2015, 11, e1004998. [Google Scholar] [CrossRef] [Green Version]
- Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicolas, G.; Chauvet, C.; Viatte, L.; Danan, J.L.; Bigard, X.; Devaux, I.; Beaumont, C.; Kahn, A.; Vaulont, S. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J. Clin. Investig. 2002, 110, 1037–1044. [Google Scholar] [CrossRef] [PubMed]
- Qiao, B.; Sugianto, P.; Fung, E.; Del-Castillo-Rueda, A.; Moran-Jimenez, M.J.; Ganz, T.; Nemeth, E. Hepcidin-induced endocytosis of ferroportin is dependent on ferroportin ubiquitination. Cell Metab. 2012, 15, 918–924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganz, T. The Discovery of the Iron-Regulatory Hormone Hepcidin. Clin. Chem. 2019, 65, 1330–1331. [Google Scholar] [CrossRef] [PubMed]
- Kautz, L.; Jung, G.; Valore, E.V.; Rivella, S.; Nemeth, E.; Ganz, T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat. Genet. 2014, 46, 678–684. [Google Scholar] [CrossRef] [Green Version]
- Vallet, N. The role of erythroferrone in iron metabolism: From experimental results to pathogenesis. La Rev. De Med. Interne 2018, 39, 178–184. [Google Scholar] [CrossRef]
- Weiss, G.; Goodnough, L.T. Anemia of chronic disease. N. Engl. J. Med. 2005, 352, 1011–1023. [Google Scholar] [CrossRef] [Green Version]
- Adamson, J.W. The anemia of inflammation/malignancy: Mechanisms and management. Hematology. American Society of Hematology. Educ. Program. 2008, 2008, 159–165. [Google Scholar] [CrossRef] [Green Version]
- Jelkmann, W. Proinflammatory cytokines lowering erythropoietin production. J. Interferon Cytokine Res. 1998, 18, 555–559. [Google Scholar] [CrossRef]
- Spivak, J.L. The anaemia of cancer: Death by a thousand cuts. Nature reviews. Cancer 2005, 5, 543–555. [Google Scholar] [CrossRef]
- Libregts, S.F.; Gutierrez, L.; de Bruin, A.M.; Wensveen, F.M.; Papadopoulos, P.; van Ijcken, W.; Ozgur, Z.; Philipsen, S.; Nolte, M.A. Chronic IFN-gamma production in mice induces anemia by reducing erythrocyte life span and inhibiting erythropoiesis through an IRF-1/PU.1 axis. Blood 2011, 118, 2578–2588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faquin, W.C.; Schneider, T.J.; Goldberg, M.A. Effect of inflammatory cytokines on hypoxia-induced erythropoietin production. Blood 1992, 79, 1987–1994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jelkmann, W.; Pagel, H.; Wolff, M.; Fandrey, J. Monokines inhibiting erythropoietin production in human hepatoma cultures and in isolated perfused rat kidneys. Life Sci. 1992, 50, 301–308. [Google Scholar] [CrossRef]
- Patra, S.K.; Arora, S. Integrative role of neuropeptides and cytokines in cancer anorexia-cachexia syndrome. Clin. Chim. Acta Int. J. Clin. Chem. 2012, 413, 1025–1034. [Google Scholar] [CrossRef] [PubMed]
- Gautron, L.; Layé, S. Neurobiology of inflammation-associated anorexia. Front. Neurosci. 2009, 3, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saini, A.; Al-Shanti, N.; Stewart, C.E. Waste management-cytokines, growth factors and cachexia. Cytokine Growth Factor Rev. 2006, 17, 475–486. [Google Scholar] [CrossRef]
- Burke, S.J.; Stadler, K.; Lu, D.; Gleason, E.; Han, A.; Donohoe, D.R.; Rogers, R.C.; Hermann, G.E.; Karlstad, M.D.; Collier, J.J. IL-1β reciprocally regulates chemokine and insulin secretion in pancreatic β-cells via NF-κB. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E715–E726. [Google Scholar] [CrossRef] [Green Version]
- Oburoglu, L.; Romano, M.; Taylor, N.; Kinet, S. Metabolic regulation of hematopoietic stem cell commitment and erythroid differentiation. Curr. Opin. Hematol. 2016, 23, 198–205. [Google Scholar] [CrossRef]
- Ganz, T.; Nemeth, E. Hepcidin and disorders of iron metabolism. Annu. Rev. Med. 2011, 62, 347–360. [Google Scholar] [CrossRef]
- McCranor, B.J.; Kim, M.J.; Cruz, N.M.; Xue, Q.L.; Berger, A.E.; Walston, J.D.; Civin, C.I.; Roy, C.N. Interleukin-6 directly impairs the erythroid development of human TF-1 erythroleukemic cells. Blood CellsMol. Dis. 2014, 52, 126–133. [Google Scholar] [CrossRef]
- Strassmann, G.; Fong, M.; Kenney, J.S.; Jacob, C.O. Evidence for the involvement of interleukin 6 in experimental cancer cachexia. J. Clin. Investig. 1992, 89, 1681–1684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Babitt, J.L.; Huang, F.W.; Xia, Y.; Sidis, Y.; Andrews, N.C.; Lin, H.Y. Modulation of bone morphogenetic protein signaling in vivo regulates systemic iron balance. J. Clin. Investig. 2007, 117, 1933–1939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Besson-Fournier, C.; Latour, C.; Kautz, L.; Bertrand, J.; Ganz, T.; Roth, M.P.; Coppin, H. Induction of activin B by inflammatory stimuli up-regulates expression of the iron-regulatory peptide hepcidin through Smad1/5/8 signaling. Blood 2012, 120, 431–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madeddu, C.; Mantovani, G.; Gramignano, G.; Astara, G.; Macciò, A. Muscle wasting as main evidence of energy impairment in cancer cachexia: Future therapeutic approaches. Future Oncol. (Lond. Engl.) 2015, 11, 2697–2710. [Google Scholar] [CrossRef]
- Chung, J.; Bauer, D.E.; Ghamari, A.; Nizzi, C.P.; Deck, K.M.; Kingsley, P.D.; Yien, Y.Y.; Huston, N.C.; Chen, C.; Schultz, I.J.; et al. The mTORC1/4E-BP pathway coordinates hemoglobin production with L-leucine availability. Sci. Signal. 2015, 8, ra34. [Google Scholar] [CrossRef] [Green Version]
- Nathan, D.G. Amino acid uptake in erythropoiesis. Sci. Signal. 2015, 8, fs9. [Google Scholar] [CrossRef]
- Buck, I.; Morceau, F.; Grigorakaki, C.; Dicato, M.; Diederich, M. Linking anemia to inflammation and cancer: The crucial role of TNFalpha. Biochem. Pharmacol. 2009, 77, 1572–1579. [Google Scholar] [CrossRef] [Green Version]
- Patel, H.J.; Patel, B.M. TNF-α and cancer cachexia: Molecular insights and clinical implications. Life Sci. 2017, 170, 56–63. [Google Scholar] [CrossRef]
- Carbó, N.; Costelli, P.; Tessitore, L.; Bagby, G.J.; López-Soriano, F.J.; Baccino, F.M.; Argilés, J.M. Anti-tumour necrosis factor-alpha treatment interferes with changes in lipid metabolism in a tumour cachexia model. Clin. Sci. 1994, 87, 349–355. [Google Scholar] [CrossRef]
- Cawthorn, W.P.; Sethi, J.K. TNF-α and adipocyte biology. FEBS Lett. 2008, 582, 117–131. [Google Scholar] [CrossRef] [Green Version]
- Bing, C.; Russell, S.; Becket, E.; Pope, M.; Tisdale, M.J.; Trayhurn, P.; Jenkins, J.R. Adipose atrophy in cancer cachexia: Morphologic and molecular analysis of adipose tissue in tumour-bearing mice. Br. J. Cancer 2006, 95, 1028–1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagasawa, E.; Abe, Y.; Nishimura, J.; Yanase, T.; Nawata, H.; Muta, K. Pivotal role of peroxisome proliferator-activated receptor gamma (PPARgamma) in regulation of erythroid progenitor cell proliferation and differentiation. Exp. Hematol. 2005, 33, 857–864. [Google Scholar] [CrossRef] [PubMed]
- Means, R.T., Jr. Recent developments in the anemia of chronic disease. Curr. Hematol. Rep. 2003, 2, 116–121. [Google Scholar] [PubMed]
- Sailaja, Y.R.; Baskar, R.; Saralakumari, D. The antioxidant status during maturation of reticulocytes to erythrocytes in type 2 diabetics. Free Radic. Biol. Med. 2003, 35, 133–139. [Google Scholar] [CrossRef]
- Olszewska, M.; Wiatrow, J.; Bober, J.; Stachowska, E.; Gołembiewska, E.; Jakubowska, K.; Stańczyk-Dunaj, M.; Pietrzak-Nowacka, M. Oxidative stress modulates the organization of erythrocyte membrane cytoskeleton. Postepy Hig. I Med. Dosw. (Online) 2012, 66, 534–542. [Google Scholar] [CrossRef] [PubMed]
- Lang, F.; Abed, M.; Lang, E.; Föller, M. Oxidative stress and suicidal erythrocyte death. Antioxid. Redox Signal. 2014, 21, 138–153. [Google Scholar] [CrossRef]
- Bukowska, B.; Sicińska, P.; Pająk, A.; Koceva-Chyla, A.; Pietras, T.; Pszczółkowska, A.; Górski, P.; Koter-Michalak, M. Oxidative stress and damage to erythrocytes in patients with chronic obstructive pulmonary disease--changes in ATPase and acetylcholinesterase activity. Biochem. Cell Biol. Biochim. Et Biol. Cell. 2015, 93, 574–580. [Google Scholar] [CrossRef]
- Prince, O.D.; Langdon, J.M.; Layman, A.J.; Prince, I.C.; Sabogal, M.; Mak, H.H.; Berger, A.E.; Cheadle, C.; Chrest, F.J.; Yu, Q.; et al. Late stage erythroid precursor production is impaired in mice with chronic inflammation. Haematologica 2012, 97, 1648–1656. [Google Scholar] [CrossRef]
- Millonig, G.; Ganzleben, I.; Peccerella, T.; Casanovas, G.; Brodziak-Jarosz, L.; Breitkopf-Heinlein, K.; Dick, T.P.; Seitz, H.K.; Muckenthaler, M.U.; Mueller, S. Sustained submicromolar H2O2 levels induce hepcidin via signal transducer and activator of transcription 3 (STAT3). J. Biol. Chem. 2012, 287, 37472–37482. [Google Scholar] [CrossRef] [Green Version]
- Rodgers, G.M.; Gilreath, J.A. The Role of Intravenous Iron in the Treatment of Anemia Associated with Cancer and Chemo-therapy. Acta Haematol. 2019, 142, 13–20. [Google Scholar] [CrossRef]
- Bennett, C.L.; Becker, P.S.; Kraut, E.H.; Samaras, A.T.; West, D.P. Intersecting guidelines: Administering erythropoiesis-stimulating agents to chronic kidney disease patients with cancer. Semin. Dial. 2009, 22, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yilmaz, B.; Li, H. Gut Microbiota and Iron: The Crucial Actors in Health and Disease. Pharmaceuticals (Basel) 2018, 11, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.; Clavel, T.; Smirnov, K.; Schmidt, A.; Lagkouvardos, I.; Walker, A.; Lucio, M.; Michalke, B.; Schmitt-Kopplin, P.; Fedorak, R.; et al. Oral versus intravenous iron replacement therapy distinctly alters the gut microbiota and metabolome in patients with IBD. Gut 2017, 66, 863–871. [Google Scholar] [CrossRef] [PubMed]
- Raines, D.J.; Moroz, O.V.; Blagova, E.V.; Turkenburg, J.P.; Wilson, K.S.; Duhme-Klair, A.-K. Bacteria in an intense competition for iron: Key component of the Campylobacter jejuni iron uptake system scavenges enterobactin hydrolysis product. Proc. Natl. Acad. Sci. USA 2016, 113, 5850–5855. [Google Scholar] [CrossRef] [Green Version]
- Andrews, S.C.; Robinson, A.K.; Rodriguez-Quinones, F. Bacterial iron homeostasis. FEMS Microbiol. Rev. 2003, 27, 215–237. [Google Scholar] [CrossRef]
- Drakesmith, H.; Prentice, A.M. Hepcidin and the iron-infection axis. Science 2012, 338, 768–772. [Google Scholar] [CrossRef] [Green Version]
- Soares, M.P.; Hamza, I. Macrophages and Iron Metabolism. Immunity 2016, 44, 492–504. [Google Scholar] [CrossRef] [Green Version]
- Cronin, S.J.F.; Woolf, C.J.; Weiss, G.; Penninger, J.M. The Role of Iron Regulation in Immunometabolism and Immune-Related Disease. Front. Mol. Biosci. 2019, 6, 116. [Google Scholar] [CrossRef] [Green Version]
- Ratledge, C. Iron metabolism and infection. Food Nutr. Bull. 2007, 28, S515–S523. [Google Scholar] [CrossRef]
- Ganz, T.; Nemeth, E. Iron Balance and the Role of Hepcidin in Chronic Kidney Disease. Semin. Nephrol. 2016, 36, 87–93. [Google Scholar] [CrossRef] [Green Version]
- Finch, C.A. Erythropoiesis, erythropoietin, and iron. Blood 1982, 60, 1241–1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crawford, J.; Cella, D.; Cleeland, C.S.; Cremieux, P.Y.; Demetri, G.D.; Sarokhan, B.J.; Slavin, M.B.; Glaspy, J.A. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002, 95, 888–895. [Google Scholar] [CrossRef] [PubMed]
- Bohlius, J.; Weingart, O.; Trelle, S.; Engert, A. Cancer-related anemia and recombinant human erythropoietin--an updated overview. Nat. Clin. Pract. Oncol. 2006, 3, 152–164. [Google Scholar] [CrossRef] [PubMed]
- Grogan, M.; Thomas, G.M.; Melamed, I.; Wong, F.L.; Pearcey, R.G.; Joseph, P.K.; Portelance, L.; Crook, J.; Jones, K.D. The importance of hemoglobin levels during radiotherapy for carcinoma of the cervix. Cancer 1999, 86, 1528–1536. [Google Scholar] [CrossRef]
- Kader, A.S.; Lim, J.T.W.; Berthelet, E.; Petersen, R.; Ludgate, D.; Truong, P.T. Prognostic significance of blood transfusions in patients with esophageal cancer treated with combined chemoradiotherapy. Am. J. Clin. Oncol 2007, 30, 492–497. [Google Scholar] [CrossRef]
- Gleeson, C.; Spencer, D. Blood transfusion and its benefits in palliative care. Palliat. Med. 1995, 9, 307–313. [Google Scholar] [CrossRef]
- Mercadante, S.; Ferrera, P.; Villari, P.; David, F.; Giarratano, A.; Riina, S. Effects of red blood cell transfusion on anemia-related symptoms in patients with cancer. J. Palliat. Med. 2009, 12, 60–63. [Google Scholar] [CrossRef]
- Tanneberger, S.; Melilli, G.; Strocchi, E.; Frenquelli, C.; Pannuti, Q.F. Use of red blood cell transfusion in palliative care services: Is it still up to date or is cancer-related anaemia controlled better with erythropoietic agents? Ann. Oncol. 2004, 15, 839–840. [Google Scholar] [CrossRef]
- Brown, E.; Hurlow, A.; Rahman, A.; Closs, S.J.; Bennett, M.I. Assessment of fatigue after blood transfusion in palliative care patients: A feasibility study. J. Palliat. Med. 2010, 13, 1327–1330. [Google Scholar] [CrossRef] [Green Version]
- Goodnough, L.T. Risks of blood transfusion. Anesthesiol. Clin. North. Am. 2005, 23, 241–252. [Google Scholar] [CrossRef]
- Toy, P.; Popovsky, M.A.; Abraham, E.; Ambruso, D.R.; Holness, L.G.; Kopko, P.M.; McFarland, J.G.; Nathens, A.B.; Silliman, C.C.; Stroncek, D. Transfusion-related acute lung injury: Definition and review. Crit. Care Med. 2005, 33, 721–726. [Google Scholar] [CrossRef] [PubMed]
- Khorana, A.A.; Francis, C.W.; Blumberg, N.; Culakova, E.; Refaai, M.A.; Lyman, G.H. Blood transfusions, thrombosis, and mortality in hospitalized patients with cancer. Arch. Intern. Med. 2008, 168, 2377–2381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Storring, P.L.; Tiplady, R.J.; Gaines Das, R.E.; Stenning, B.E.; Lamikanra, A.; Rafferty, B.; Lee, J. Epoetin alfa and beta differ in their erythropoietin isoform compositions and biological properties. Br. J. Haematol. 1998, 100, 79–89. [Google Scholar] [CrossRef] [PubMed]
- Tonia, T.; Mettler, A.; Robert, N.; Schwarzer, G.; Seidenfeld, J.; Weingart, O.; Hyde, C.; Engert, A.; Bohlius, J. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst. Rev. 2012, 12, Cd003407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quirt, I.; Robeson, C.; Lau, C.Y.; Kovacs, M.; Burdette-Radoux, S.; Dolan, S.; Tang, S.C.; McKenzie, M.; Couture, F. Epoetin alfa therapy increases hemoglobin levels and improves quality of life in patients with cancer-related anemia who are not receiving chemotherapy and patients with anemia who are receiving chemotherapy. J. Clin. Oncol. 2001, 19, 4126–4134. [Google Scholar] [CrossRef] [PubMed]
- Bogdanos, J.; Karamanolakis, D.; Milathianakis, K.; Repousis, P.; Chloraki-Bobota, A.; Majed, H.; Pagalou-Thoua, E.; Tsintavis, A.; Koutsilieris, M. Epoetin beta (NeoRecormon) corrects anaemia in patients with hormone-refractory prostate cancer and bone metastases. Anticancer Res. 2004, 24, 1957–1961. [Google Scholar]
- Mystakidou, K.; Kalaidopoulou, O.; Katsouda, E.; Parpa, E.; Kouskouni, E.; Chondros, C.; Tsiatas, M.L.; Vlahos, L. Evaluation of epoetin supplemented with oral iron in patients with solid malignancies and chronic anemia not receiving anticancer treatment. Anticancer Res. 2005, 25, 3495–3500. [Google Scholar]
- Wright, J.R.; Ung, Y.C.; Julian, J.A.; Pritchard, K.I.; Whelan, T.J.; Smith, C.; Szechtman, B.; Roa, W.; Mulroy, L.; Rudinskas, L.; et al. Randomized, double-blind, placebo-controlled trial of erythropoietin in non-small-cell lung cancer with disease-related anemia. J. Clin. Oncol. 2007, 25, 1027–1032. [Google Scholar] [CrossRef]
- Smith, R.E., Jr.; Aapro, M.S.; Ludwig, H.; Pintér, T.; Smakal, M.; Ciuleanu, T.E.; Chen, L.; Lillie, T.; Glaspy, J.A. Darbepoetin alpha for the treatment of anemia in patients with active cancer not receiving chemotherapy or radiotherapy: Results of a phase III, multicenter, randomized, double-blind, placebo-controlled study. J. Clin. Oncol. 2008, 26, 1040–1050. [Google Scholar] [CrossRef]
- Nekoui, A.; Blaise, G. Erythropoietin and Nonhematopoietic Effects. Am. J. Med. Sci. 2017, 353, 76–81. [Google Scholar] [CrossRef]
- Toblli, J.E.; Angerosa, M. Optimizing iron delivery in the management of anemia: Patient considerations and the role of ferric carboxymaltose. Drug Des. Dev. Ther. 2014, 8, 2475–2491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Keating, G.M. Ferric carboxymaltose: A review of its use in iron deficiency. Drugs 2015, 75, 101–127. [Google Scholar] [CrossRef] [PubMed]
- Steensma, D.P.; Sloan, J.A.; Dakhil, S.R.; Dalton, R.; Kahanic, S.P.; Prager, D.J.; Stella, P.J.; Rowland, K.M., Jr.; Novotny, P.J.; Loprinzi, C.L. Phase III, randomized study of the effects of parenteral iron, oral iron, or no iron supplementation on the erythropoietic response to darbepoetin alfa for patients with chemotherapy-associated anemia. J. Clin. Oncol. 2011, 29, 97–105. [Google Scholar] [CrossRef] [PubMed]
- Million, M.; Diallo, A.; Raoult, D. Gut microbiota and malnutrition. Microb. Pathog. 2017, 106, 127–138. [Google Scholar] [CrossRef] [PubMed]
- Jensen, G.L.; Mirtallo, J.; Compher, C.; Dhaliwal, R.; Forbes, A.; Grijalba, R.F.; Hardy, G.; Kondrup, J.; Labadarios, D.; Nyulasi, I.; et al. Adult starvation and disease-related malnutrition: A proposal for etiology-based diagnosis in the clinical practice setting from the International Consensus Guideline Committee. JPEN J. Parenter Enter. Nutr. 2010, 34, 156–159. [Google Scholar] [CrossRef] [PubMed]
- Chrysohoou, C.; Panagiotakos, D.B.; Pitsavos, C.; Das, U.N.; Stefanadis, C. Adherence to the Mediterranean diet attenuates inflammation and coagulation process in healthy adults: The ATTICA Study. J. Am. Coll. Cardiol. 2004, 44, 152–158. [Google Scholar] [CrossRef] [Green Version]
- Knoops, K.T.; de Groot, L.C.; Kromhout, D.; Perrin, A.E.; Moreiras-Varela, O.; Menotti, A.; van Staveren, W.A. Mediterranean diet, lifestyle factors, and 10-year mortality in elderly European men and women: The HALE project. JAMA 2004, 292, 1433–1439. [Google Scholar] [CrossRef]
- Brunner, E.J.; Mosdøl, A.; Witte, D.R.; Martikainen, P.; Stafford, M.; Shipley, M.J.; Marmot, M.G. Dietary patterns and 15-y risks of major coronary events, diabetes, and mortality. Am. J. Clin. Nutr. 2008, 87, 1414–1421. [Google Scholar] [CrossRef] [Green Version]
- Bonaccio, M.; Pounis, G.; Cerletti, C.; Donati, M.B.; Iacoviello, L.; de Gaetano, G. Mediterranean diet, dietary polyphenols and low grade inflammation: Results from the MOLI-SANI study. Br. J. Clin. Pharmacol. 2017, 83, 107–113. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
- Raetz, C.R.; Whitfield, C. Lipopolysaccharide endotoxins. Annu. Rev. Biochem. 2002, 71, 635–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
- Derrien, M.; Veiga, P. Rethinking Diet to Aid Human–Microbe Symbiosis. Trends Microbiol. 2017, 25, 100–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gaucher, F.; Bonnassie, S.; Rabah, H.; Marchand, P.; Blanc, P.; Jeantet, R.; Jan, G. Review: Adaptation of Beneficial Propionibacteria, Lactobacilli, and Bifidobacteria Improves Tolerance Toward Technological and Digestive Stresses. Front. Microbiol. 2019, 10, 841. [Google Scholar] [CrossRef] [PubMed]
- Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alipour, B.; Homayouni-Rad, A.; Vaghef-Mehrabany, E.; Sharif, S.K.; Vaghef-Mehrabany, L.; Asghari-Jafarabadi, M.; Nakhjavani, M.R.; Mohtadi-Nia, J. Effects of Lactobacillus casei supplementation on disease activity and inflammatory cytokines in rheumatoid arthritis patients: A randomized double-blind clinical trial. Int. J. Rheum. Dis. 2014, 17, 519–527. [Google Scholar] [CrossRef]
- Pei, R.; DiMarco, D.M.; Putt, K.K.; Martin, D.A.; Gu, Q.; Chitchumroonchokchai, C.; White, H.M.; Scarlett, C.O.; Bruno, R.S.; Bolling, B.W. Low-fat yogurt consumption reduces biomarkers of chronic inflammation and inhibits markers of endotoxin exposure in healthy premenopausal women: A randomised controlled trial. Br. J. Nutr. 2017, 118, 1043–1051. [Google Scholar] [CrossRef] [PubMed]
- St-Onge, M.P.; Farnworth, E.R.; Savard, T.; Chabot, D.; Mafu, A.; Jones, P.J. Kefir consumption does not alter plasma lipid levels or cholesterol fractional synthesis rates relative to milk in hyperlipidemic men: A randomized controlled trial [ISRCTN10820810]. BMC Complementary Altern. Med. 2002, 2, 1. [Google Scholar] [CrossRef]
- O’Brien, K.V.; Stewart, L.K.; Forney, L.A.; Aryana, K.J.; Prinyawiwatkul, W.; Boeneke, C.A. The effects of postexercise consumption of a kefir beverage on performance and recovery during intensive endurance training. J. Dairy Sci. 2015, 98, 7446–7449. [Google Scholar] [CrossRef]
- Huang, C.C.; Hsu, M.C.; Huang, W.C.; Yang, H.R.; Hou, C.C. Triterpenoid-Rich Extract from Antrodia camphorata Improves Physical Fatigue and Exercise Performance in Mice. Evid. Based Complementary Altern. Med. 2012, 2012, 364741. [Google Scholar] [CrossRef] [Green Version]
- Lollo, P.C.B.; Morato, P.N.; de Moura, C.S.; de Oliveira, M.M.; Cruz, A.G.; Faria, J.d.A.F.; Amaya-Farfan, J.; Cristianini, M. Ultra-high temperature plus dynamic high pressure processing: An effective combination for potential probiotic fermented milk processing which attenuate exercise-induced immune suppression in Wistar rats. J. Funct. Foods 2015, 14, 541–548. [Google Scholar] [CrossRef]
- Slavin, J. Fiber and Prebiotics: Mechanisms and Health Benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salehi-Abargouei, A.; Saraf-Bank, S.; Bellissimo, N.; Azadbakht, L. Effects of non-soy legume consumption on C-reactive protein: A systematic review and meta-analysis. Nutrition 2015, 31, 631–639. [Google Scholar] [CrossRef] [PubMed]
- Zhu, F.; Du, B.; Xu, B. Anti-inflammatory effects of phytochemicals from fruits, vegetables, and food legumes: A review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1260–1270. [Google Scholar] [CrossRef]
- Bach Knudsen, K.E. Microbial Degradation of Whole-Grain Complex Carbohydrates and Impact on Short-Chain Fatty Acids and Health. Adv. Nutr. 2015, 6, 206–213. [Google Scholar] [CrossRef] [Green Version]
- De Filippis, F.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef]
- Russell, W.R.; Hoyles, L.; Flint, H.J.; Dumas, M.E. Colonic bacterial metabolites and human health. Curr. Opin. Microbiol. 2013, 16, 246–254. [Google Scholar] [CrossRef] [Green Version]
- Holscher, H.D.; Bauer, L.L.; Gourineni, V.; Pelkman, C.L.; Fahey, G.C., Jr.; Swanson, K.S. Agave Inulin Supplementation Affects the Fecal Microbiota of Healthy Adults Participating in a Randomized, Double-Blind, Placebo-Controlled, Crossover Trial. J. Nutr. 2015, 145, 2025–2032. [Google Scholar] [CrossRef] [Green Version]
- Rivière, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front. Microbiol. 2016, 7, 979. [Google Scholar] [CrossRef] [Green Version]
- Paul, B.; Barnes, S.; Demark-Wahnefried, W.; Morrow, C.; Salvador, C.; Skibola, C.; Tollefsbol, T.O. Influences of diet and the gut microbiome on epigenetic modulation in cancer and other diseases. Clin. Epigenetics 2015, 7, 112. [Google Scholar] [CrossRef] [Green Version]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef] [PubMed]
- Anhê, F.F.; Roy, D.; Pilon, G.; Dudonné, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Donati Zeppa, S.; Agostini, D.; Gervasi, M.; Annibalini, G.; Amatori, S.; Ferrini, F.; Sisti, D.; Piccoli, G.; Barbieri, E.; Sestili, P.; et al. Mutual Interactions among Exercise, Sport Supplements and Microbiota. Nutrients 2019, 12, 17. [Google Scholar] [CrossRef] [Green Version]
- González-Sarrías, A.; Espín, J.C.; Tomás-Barberán, F.A. Non-extractable polyphenols produce gut microbiota metabolites that persist in circulation and show anti-inflammatory and free radical-scavenging effects. Trends Food Sci. Technol. 2017, 69, 281–288. [Google Scholar] [CrossRef]
- Tian, S.; Liu, X.; Lei, P.; Zhang, X.; Shan, Y. Microbiota: A mediator to transform glucosinolate precursors in cruciferous vegetables to the active isothiocyanates. J. Sci. Food Agric. 2018, 98, 1255–1260. [Google Scholar] [CrossRef]
- De Wit, N.; Derrien, M.; Bosch-Vermeulen, H.; Oosterink, E.; Keshtkar, S.; Duval, C.; de Vogel-van den Bosch, J.; Kleerebezem, M.; Müller, M.; van der Meer, R. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G589–G599. [Google Scholar] [CrossRef] [Green Version]
- Kaysen, G.A.; Johansen, K.L.; Chertow, G.M.; Dalrymple, L.S.; Kornak, J.; Grimes, B.; Dwyer, T.; Chassy, A.W.; Fiehn, O. Associations of Trimethylamine N-Oxide with Nutritional and Inflammatory Biomarkers and Cardiovascular Outcomes in Patients New to Dialysis. J. Ren. Nutr. 2015, 25, 351–356. [Google Scholar] [CrossRef] [Green Version]
- Tang, W.H.W.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ. Res. 2015, 116, 448–455. [Google Scholar] [CrossRef] [Green Version]
- Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [Green Version]
- Craciun, S.; Balskus, E.P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl. Acad. Sci. USA 2012, 109, 21307–21312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Jameson, E.; Crosatti, M.; Schafer, H.; Rajakumar, K.; Bugg, T.D.; Chen, Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc. Natl. Acad. Sci. USA 2014, 111, 4268–4273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ley, S.H.; Sun, Q.; Willett, W.C.; Eliassen, A.H.; Wu, K.; Pan, A.; Grodstein, F.; Hu, F.B. Associations between red meat intake and biomarkers of inflammation and glucose metabolism in women. Am. J. Clin. Nutr. 2013, 99, 352–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samraj, A.N.; Pearce, O.M.T.; Läubli, H.; Crittenden, A.N.; Bergfeld, A.K.; Banda, K.; Gregg, C.J.; Bingman, A.E.; Secrest, P.; Diaz, S.L.; et al. A red meat-derived glycan promotes inflammation and cancer progression. Proc. Natl. Acad. Sci. USA 2015, 112, 542. [Google Scholar] [CrossRef] [Green Version]
- Demmer, E.; Van Loan, M.D.; Rivera, N.; Rogers, T.S.; Gertz, E.R.; German, J.B.; Zivkovic, A.M.; Smilowitz, J.T. Consumption of a high-fat meal containing cheese compared with a vegan alternative lowers postprandial C-reactive protein in overweight and obese individuals with metabolic abnormalities: A randomised controlled cross-over study. J. Nutr. Sci. 2016, 5, e9. [Google Scholar] [CrossRef] [Green Version]
- Boudry, G.; Hamilton, M.K.; Chichlowski, M.; Wickramasinghe, S.; Barile, D.; Kalanetra, K.M.; Mills, D.A.; Raybould, H.E. Bovine milk oligosaccharides decrease gut permeability and improve inflammation and microbial dysbiosis in diet-induced obese mice. J. Dairy Sci. 2017, 100, 2471–2481. [Google Scholar] [CrossRef] [Green Version]
- Müller, S.; März, R.; Schmolz, M.; Drewelow, B.; Eschmann, K.; Meiser, P. Placebo-controlled Randomized Clinical Trial on the Immunomodulating Activities of Low- and High-Dose Bromelain after Oral Administration–New Evidence on the Antiinflammatory Mode of Action of Bromelain. Phytother. Res. 2013, 27, 199–204. [Google Scholar] [CrossRef]
- Pandey, S.; Cabot, P.J.; Shaw, P.N.; Hewavitharana, A.K. Anti-inflammatory and immunomodulatory properties of Carica papaya. J. Immunotoxicol. 2016, 13, 590–602. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.; Salem, A.; Nanavati, J.; Mullin, G.E. The Role of Diet in the Treatment of Irritable Bowel Syndrome: A Systematic Review. Gastroenterol. Clin. N. Am. 2018, 47, 107–137. [Google Scholar] [CrossRef]
- Sinha, K.; Khare, V. Review on: Antinutritional factors in vegetable crops. Pharma Innov. J. 2017, 6, 353–358. [Google Scholar]
- Lerner, A.; Matthias, T. Rheumatoid arthritis–celiac disease relationship: Joints get that gut feeling. Autoimmun. Rev. 2015, 14, 1038–1047. [Google Scholar] [CrossRef] [PubMed]
- Bendsen, N.T.; Stender, S.; Szecsi, P.B.; Pedersen, S.B.; Basu, S.; Hellgren, L.I.; Newman, J.W.; Larsen, T.M.; Haugaard, S.B.; Astrup, A. Effect of industrially produced trans fat on markers of systemic inflammation: Evidence from a randomized trial in women. J. Lipid Res. 2011, 52, 1821–1828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, B.; Leung, J.C.K.; Chan, L.Y.Y.; Yiu, W.H.; Tang, S.C.W. A global perspective on the crosstalk between saturated fatty acids and Toll-like receptor 4 in the etiology of inflammation and insulin resistance. Prog. Lipid Res. 2020, 77, 101020. [Google Scholar] [CrossRef] [PubMed]
- Lorente-Cebrián, S.; Costa, A.G.V.; Navas-Carretero, S.; Zabala, M.; Laiglesia, L.M.; Martínez, J.A.; Moreno-Aliaga, M.J. An update on the role of omega-3 fatty acids on inflammatory and degenerative diseases. J. Physiol. Biochem. 2015, 71, 341–349. [Google Scholar] [CrossRef] [PubMed]
- Virgili, E.; Emili, R.; Ferri, H.I.; Palermo, F.; Calza, L.; Gardarelli, M.; Spina, M. Lipidomics and Nutrylipidomics in Oncology: Review of the Literature. J. Bioinform Syst. Biol. 2018, 1, 11–14. [Google Scholar]
- Zárate, R.; El Jaber-Vazdekis, N.; Tejera, N.; Pérez, J.A.; Rodríguez, C. Significance of long chain polyunsaturated fatty acids in human health. Clin. Transl. Med. 2017, 6, 25. [Google Scholar] [CrossRef] [Green Version]
- Zamani, S.A.; McClain, K.M.; Graubard, B.I.; Liao, L.M.; Abnet, C.C.; Cook, M.B.; Petrick, J.L. Dietary polyunsaturated fat intake in relation to head and neck, esophageal, and gastric cancer incidence in the NIH-AARP Diet and Health Study. Am. J. Epidemiol. 2020. [Google Scholar] [CrossRef] [Green Version]
- Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef] [Green Version]
- De la Puerta, R.; Ruiz Gutierrez, V.; Hoult, J.R. Inhibition of leukocyte 5-lipoxygenase by phenolics from virgin olive oil. Biochem. Pharmacol. 1999, 57, 445–449. [Google Scholar] [CrossRef]
- Jiang, Q. Natural forms of vitamin E: Metabolism, antioxidant, and anti-inflammatory activities and their role in disease prevention and therapy. Free Radic. Biol. Med. 2014, 72, 76–90. [Google Scholar] [CrossRef] [Green Version]
- Sassi, F.; Tamone, C.; D’Amelio, P. Vitamin D: Nutrient, Hormone, and Immunomodulator. Nutrients 2018, 10, 1656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef] [PubMed]
- Basnet, P.; Skalko-Basnet, N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011, 16, 4567–4598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sultan, M.T.; Butt, M.S.; Qayyum, M.M.; Suleria, H.A. Immunity: Plants as effective mediators. Crit. Rev. Food Sci. Nutr. 2014, 54, 1298–1308. [Google Scholar] [CrossRef] [PubMed]
- Toden, S.; Theiss, A.L.; Wang, X.; Goel, A. Essential turmeric oils enhance anti-inflammatory efficacy of curcumin in dextran sulfate sodium-induced colitis. Sci. Rep. 2017, 7, 814. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.T.; Cui, W.Q.; Pan, D.; Jiang, M.; Chang, B.; Sang, L.X. Tea polyphenols and their chemopreventive and therapeutic effects on colorectal cancer. World J. Gastroenterol. 2020, 26, 562–597. [Google Scholar] [CrossRef]
- Uribarri, J.; del Castillo, M.D.; de la Maza, M.P.; Filip, R.; Gugliucci, A.; Luevano-Contreras, C.; Macías-Cervantes, M.H.; Markowicz Bastos, D.H.; Medrano, A.; Menini, T.; et al. Dietary advanced glycation end products and their role in health and disease. Adv. Nutr. (BethesdaMd.) 2015, 6, 461–473. [Google Scholar] [CrossRef]
- Poulsen, M.W.; Hedegaard, R.V.; Andersen, J.M.; de Courten, B.; Bügel, S.; Nielsen, J.; Skibsted, L.H.; Dragsted, L.O. Advanced glycation endproducts in food and their effects on health. Food Chem. Toxicol. 2013, 60, 10–37. [Google Scholar] [CrossRef]
- Uribarri, J.; Woodruff, S.; Goodman, S.; Cai, W.; Chen, X.; Pyzik, R.; Yong, A.; Striker, G.E.; Vlassara, H. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J. Am. Diet. Assoc. 2010, 110, 911–916.e912. [Google Scholar] [CrossRef] [Green Version]
- Goldberg, T.; Cai, W.; Peppa, M.; Dardaine, V.; Baliga, B.S.; Uribarri, J.; Vlassara, H. Advanced glycoxidation end products in commonly consumed foods. J. Am. Diet. Assoc. 2004, 104, 1287–1291. [Google Scholar] [CrossRef]
- Qu, W.; Yuan, X.; Zhao, J.; Zhang, Y.; Hu, J.; Wang, J.; Li, J. Dietary advanced glycation end products modify gut microbial composition and partially increase colon permeability in rats. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
- Macciò, A.; Madeddu, C.; Gramignano, G.; Mulas, C.; Sanna, E.; Mantovani, G. Efficacy and safety of oral lactoferrin supplementation in combination with rHuEPO-Beta for the treatment of anemia in advanced cancer patients undergoing chemotherapy: Open-Label, randomized controlled study. Oncologist 2010, 15, 894–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korobkova, E.A. Effect of Natural Polyphenols on CYP Metabolism: Implications for Diseases. Chem. Res. Toxicol. 2015, 28, 1359–1390. [Google Scholar] [CrossRef] [PubMed]
- Sprouse, A.A.; van Breemen, R.B. Pharmacokinetic Interactions between Drugs and Botanical Dietary Supplements. Drug Metab. Dispos. 2016, 44, 162–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaufman, D.W.; Kelly, J.P.; Rosenberg, L.; Anderson, T.E.; Mitchell, A.A. Recent patterns of medication use in the ambulatory adult population of the United States: The Slone survey. JAMA 2002, 287, 337–344. [Google Scholar] [CrossRef] [Green Version]
- Santino, A.; Scarano, A.; De Santis, S.; De Benedictis, M.; Giovinazzo, G.; Chieppa, M. Gut Microbiota Modulation and Anti-Inflammatory Properties of Dietary Polyphenols in IBD: New and Consolidated Perspectives. Curr. Pharm. Des. 2017, 23, 2344–2351. [Google Scholar] [CrossRef]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [Green Version]
- Rahman, I.; Marwick, J.; Kirkham, P. Redox modulation of chromatin remodeling: Impact on histone acetylation and deacetylation, NF-kappaB and pro-inflammatory gene expression. Biochem. Pharmacol. 2004, 68, 1255–1267. [Google Scholar] [CrossRef]
- Bae, J.S. Role of high mobility group box 1 in inflammatory disease: Focus on sepsis. Arch. Pharmacal Res. 2012, 35, 1511–1523. [Google Scholar] [CrossRef]
- Wang, H.K.; Yeh, C.H.; Iwamoto, T.; Satsu, H.; Shimizu, M.; Totsuka, M. Dietary flavonoid naringenin induces regulatory T cells via an aryl hydrocarbon receptor mediated pathway. J. Agric. Food Chem. 2012, 60, 2171–2178. [Google Scholar] [CrossRef]
- Rahman, I.; Biswas, S.K.; Kirkham, P.A. Regulation of inflammation and redox signaling by dietary polyphenols. Biochem. Pharmacol. 2006, 72, 1439–1452. [Google Scholar] [CrossRef] [PubMed]
- Sakaguchi, S.; Miyara, M.; Costantino, C.M.; Hafler, D.A. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 2010, 10, 490–500. [Google Scholar] [CrossRef] [PubMed]
- Boissier, M.C.; Assier, E.; Biton, J.; Denys, A.; Falgarone, G.; Bessis, N. Regulatory T cells (Treg) in rheumatoid arthritis. Jt. Bone Spine 2009, 76, 10–14. [Google Scholar] [CrossRef] [PubMed]
- Robinson, D.S.; Larché, M.; Durham, S.R. Tregs and allergic disease. J. Clin. Investig. 2004, 114, 1389–1397. [Google Scholar] [CrossRef] [Green Version]
- Noorafshan, A.; Ashkani-Esfahani, S. A review of therapeutic effects of curcumin. Curr. Pharm. Des. 2013, 19, 2032–2046. [Google Scholar]
- Marchiani, A.; Rozzo, C.; Fadda, A.; Delogu, G.; Ruzza, P. Curcumin and curcumin-like molecules: From spice to drugs. Curr. Med. Chem. 2014, 21, 204–222. [Google Scholar] [CrossRef]
- Gupta, S.C.; Prasad, S.; Kim, J.H.; Patchva, S.; Webb, L.J.; Priyadarsini, I.K.; Aggarwal, B.B. Multitargeting by curcumin as revealed by molecular interaction studies. Nat. Prod. Rep. 2011, 28, 1937–1955. [Google Scholar] [CrossRef] [Green Version]
- Leiherer, A.; Mündlein, A.; Drexel, H. Phytochemicals and their impact on adipose tissue inflammation and diabetes. Vasc. Pharmacol. 2013, 58, 3–20. [Google Scholar] [CrossRef]
- Siddiqui, A.M.; Cui, X.; Wu, R.; Dong, W.; Zhou, M.; Hu, M.; Simms, H.H.; Wang, P. The anti-inflammatory effect of curcumin in an experimental model of sepsis is mediated by up-regulation of peroxisome proliferator-activated receptor-gamma. Crit. Care Med. 2006, 34, 1874–1882. [Google Scholar] [CrossRef]
- Lainé, F.; Laviolle, B.; Bardou-Jacquet, E.; Faith, N.; Jezequel, C.; Collet, N.; Ropert, M.; Morcet, J.; Hamon, C.; Reymann, J.M.; et al. Curcuma decreases serum hepcidin levels in healthy volunteers: A placebo-controlled, randomized, double-blind, cross-over study. Fundam Clin. Pharm. 2017, 31, 567–573. [Google Scholar] [CrossRef] [Green Version]
- Yang, C.; Ma, X.; Wang, Z.; Zeng, X.; Hu, Z.; Ye, Z.; Shen, G. Curcumin induces apoptosis and protective autophagy in castration-resistant prostate cancer cells through iron chelation. Drug Des. Dev. Ther. 2017, 11, 431–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonucci, M.; Ferri, I.H.; Mantovani, M.; Di Fede, G. New Formulation of Curcumin: Study of Oral Absorption and Bioavailability. Int. J. Pharm. Pharm. Chem. 2018, 13, 122–130. [Google Scholar]
- Mohar, D.S.; Malik, S. The Sirtuin System: The Holy Grail of Resveratrol? J. Clin. Exp. Cardiol. 2012, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Biasutto, L.; Mattarei, A.; Zoratti, M. Resveratrol and health: The starting point. Chembiochem 2012, 13, 1256–1259. [Google Scholar] [CrossRef]
- Carluccio, M.A.; Siculella, L.; Ancora, M.A.; Massaro, M.; Scoditti, E.; Storelli, C.; Visioli, F.; Distante, A.; De Caterina, R. Olive oil and red wine antioxidant polyphenols inhibit endothelial activation: Antiatherogenic properties of Mediterranean diet phytochemicals. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 622–629. [Google Scholar] [CrossRef] [Green Version]
- Capiralla, H.; Vingtdeux, V.; Venkatesh, J.; Dreses-Werringloer, U.; Zhao, H.; Davies, P.; Marambaud, P. Identification of potent small-molecule inhibitors of STAT3 with anti-inflammatory properties in RAW 264.7 macrophages. FEBS J. 2012, 279, 3791–3799. [Google Scholar] [CrossRef] [Green Version]
- Domitrovic, R. The molecular basis for the pharmacological activity of anthocyans. Curr. Med. Chem. 2011, 18, 4454–4469. [Google Scholar] [CrossRef]
- Sato, M.; Miyazaki, T.; Kambe, F.; Maeda, K.; Seo, H. Quercetin, a bioflavonoid, inhibits the induction of interleukin 8 and monocyte chemoattractant protein-1 expression by tumor necrosis factor-alpha in cultured human synovial cells. J. Rheumatol. 1997, 24, 1680–1684. [Google Scholar]
- Min, Y.D.; Choi, C.H.; Bark, H.; Son, H.Y.; Park, H.H.; Lee, S.; Park, J.W.; Park, E.K.; Shin, H.I.; Kim, S.H. Quercetin inhibits expression of inflammatory cytokines through attenuation of NF-kappaB and p38 MAPK in HMC-1 human mast cell line. Inflamm. Res. 2007, 56, 210–215. [Google Scholar] [CrossRef]
- Chen, J.C.; Ho, F.M.; Pei-Dawn Lee, C.; Chen, C.P.; Jeng, K.C.; Hsu, H.B.; Lee, S.T.; Wen Tung, W.; Lin, W.W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of IkappaB kinase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 2005, 521, 9–20. [Google Scholar] [CrossRef]
- Comalada, M.; Camuesco, D.; Sierra, S.; Ballester, I.; Xaus, J.; Gálvez, J.; Zarzuelo, A. In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-kappaB pathway. Eur. J. Immunol. 2005, 35, 584–592. [Google Scholar] [CrossRef] [PubMed]
- Ruiz, P.A.; Braune, A.; Hölzlwimmer, G.; Quintanilla-Fend, L.; Haller, D. Quercetin inhibits TNF-induced NF-kappaB transcription factor recruitment to proinflammatory gene promoters in murine intestinal epithelial cells. J. Nutr. 2007, 137, 1208–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wadsworth, T.L.; McDonald, T.L.; Koop, D.R. Effects of Ginkgo biloba extract (EGb 761) and quercetin on lipopolysaccharide-induced signaling pathways involved in the release of tumor necrosis factor-alpha. Biochem. Pharmacol. 2001, 62, 963–974. [Google Scholar] [CrossRef]
- Kanwar, J.; Taskeen, M.; Mohammad, I.; Huo, C.; Chan, T.H.; Dou, Q.P. Recent advances on tea polyphenols. Front. Biosci. (Elite Ed.) 2012, 4, 111–131. [Google Scholar] [CrossRef]
- Singh, B.N.; Shankar, S.; Srivastava, R.K. Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem. Pharmacol. 2011, 82, 1807–1821. [Google Scholar] [CrossRef] [Green Version]
- Landis-Piwowar, K.; Chen, D.; Foldes, R.; Chan, T.H.; Dou, Q.P. Novel epigallocatechin gallate analogs as potential anticancer agents: A patent review (2009-present). Expert Opin. Ther. Pat. 2013, 23, 189–202. [Google Scholar] [CrossRef] [Green Version]
- Wong, C.P.; Nguyen, L.P.; Noh, S.K.; Bray, T.M.; Bruno, R.S.; Ho, E. Induction of regulatory T cells by green tea polyphenol EGCG. Immunol. Lett. 2011, 139, 7–13. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Pae, M.; Meydani, S.N.; Wu, D. Green tea epigallocatechin-3-gallate modulates differentiation of naïve CD4+ T cells into specific lineage effector cells. J. Mol. Med. (Berlin, Germany) 2013, 91, 485–495. [Google Scholar] [CrossRef]
- Ichikawa, D.; Matsui, A.; Imai, M.; Sonoda, Y.; Kasahara, T. Effect of various catechins on the IL-12p40 production by murine peritoneal macrophages and a macrophage cell line, J774.1. Biol. Pharm. Bull. 2004, 27, 1353–1358. [Google Scholar] [CrossRef]
- Cho, S.Y.; Park, S.J.; Kwon, M.J.; Jeong, T.S.; Bok, S.H.; Choi, W.Y.; Jeong, W.I.; Ryu, S.Y.; Do, S.H.; Lee, C.S.; et al. Quercetin suppresses proinflammatory cytokines production through MAP kinases andNF-kappaB pathway in lipopolysaccharide-stimulated macrophage. Mol. Cell. Biochem. 2003, 243, 153–160. [Google Scholar] [CrossRef]
- Hou, D.X.; Luo, D.; Tanigawa, S.; Hashimoto, F.; Uto, T.; Masuzaki, S.; Fujii, M.; Sakata, Y. Prodelphinidin B-4 3′-O-gallate, a tea polyphenol, is involved in the inhibition of COX-2 and iNOS via the downregulation of TAK1-NF-kappaB pathway. Biochem. Pharmacol. 2007, 74, 742–751. [Google Scholar] [CrossRef] [PubMed]
- Hou, D.X.; Masuzaki, S.; Hashimoto, F.; Uto, T.; Tanigawa, S.; Fujii, M.; Sakata, Y. Green tea proanthocyanidins inhibit cyclooxygenase-2 expression in LPS-activated mouse macrophages: Molecular mechanisms and structure-activity relationship. Arch. Biochem. Biophys. 2007, 460, 67–74. [Google Scholar] [CrossRef] [PubMed]
- Carvalho, C.; Isakova, T.; Collerone, G.; Olbina, G.; Wolf, M.; Westerman, M.; Gutierrez, O.M. Hepcidin and disordered mineral metabolism in chronic kidney disease. Clin. Nephrol. 2011, 76, 90–98. [Google Scholar] [CrossRef] [PubMed]
- Perlstein, T.S.; Pande, R.; Berliner, N.; Vanasse, G.J. Prevalence of 25-hydroxyvitamin D deficiency in subgroups of elderly persons with anemia: Association with anemia of inflammation. Blood 2011, 117, 2800–2806. [Google Scholar] [CrossRef] [PubMed]
- Icardi, A.; Paoletti, E.; De Nicola, L.; Mazzaferro, S.; Russo, R.; Cozzolino, M. Renal anaemia and EPO hyporesponsiveness associated with vitamin D deficiency: The potential role of inflammation. Nephrol. Dial. Transplant. 2013, 28, 1672–1679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, E.M.; Alvarez, J.A.; Kearns, M.D.; Hao, L.; Sloan, J.H.; Konrad, R.J.; Ziegler, T.R.; Zughaier, S.M.; Tangpricha, V. High-dose vitamin D3 reduces circulating hepcidin concentrations: A pilot, randomized, double-blind, placebo-controlled trial in healthy adults. Clin. Nutr. 2017, 36, 980–985. [Google Scholar] [CrossRef] [PubMed]
- Smith, E.M.; Jones, J.L.; Han, J.E.; Alvarez, J.A.; Sloan, J.H.; Konrad, R.J.; Zughaier, S.M.; Martin, G.S.; Ziegler, T.R.; Tangpricha, V. High-Dose Vitamin D3 Administration Is Associated with Increases in Hemoglobin Concentrations in Mechanically Ventilated Critically Ill Adults: A Pilot Double-Blind, Randomized, Placebo-Controlled Trial. JPEN J. Parenter Enter. Nutr. 2018, 42, 87–94. [Google Scholar] [CrossRef]
- Moran-Lev, H.; Galai, T.; Yerushalmy-Feler, A.; Weisman, Y.; Anafy, A.; Deutsch, V.; Cipok, M.; Lubetzky, R.; Cohen, S. Vitamin D Decreases Hepcidin and Inflammatory Markers in Newly Diagnosed Inflammatory Bowel Disease Paediatric Patients: A Prospective Study. J. Crohn’s Colitis 2019, 13, 1287–1291. [Google Scholar] [CrossRef]
- Roig, E.A.; Richer, E.; Canonne-Hergaux, F.; Gros, P.; Cellier, M.F.M. Regulation of NRAMP1 gene expression by 1α,25-dihydroxy-vitamin D3 in HL-60 phagocytes. J. Leukoc. Biol. 2002, 71, 890–904. [Google Scholar] [CrossRef]
- Zughaier, S.M.; Alvarez, J.A.; Sloan, J.H.; Konrad, R.J.; Tangpricha, V. The role of vitamin D in regulating the iron-hepcidin-ferroportin axis in monocytes. J. Clin. Transl. Endocrinol. 2014, 1, 19–25. [Google Scholar] [CrossRef]
- McCarley, P. The KDOQI clinical practice guidelines and clinical practice recommendations for treating anemia in patients with chronic kidney disease: Implications for nurses. Nephrol. Nurs. J. 2006, 33, 423–428, 445. [Google Scholar]
- Alvarez, J.A.; Zughaier, S.M.; Law, J.; Hao, L.; Wasse, H.; Ziegler, T.R.; Tangpricha, V. Effects of high-dose cholecalciferol on serum markers of inflammation and immunity in patients with early chronic kidney disease. Eur. J. Clin. Nutr. 2013, 67, 264–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.D.; Zhou, D.B.; Li, S.L.; Wang, X.; Zhang, J.P.; Duan, M.H.; Shen, T.; Wu, Y.J. Effect of recombinant human erythropoietin on hepcidin mRNA expression in patients with multiple myeloma. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2011, 19, 390–394. [Google Scholar] [PubMed]
- Bacchetta, J.; Zaritsky, J.J.; Sea, J.L.; Chun, R.F.; Lisse, T.S.; Zavala, K.; Nayak, A.; Wesseling-Perry, K.; Westerman, M.; Hollis, B.W.; et al. Suppression of iron-regulatory hepcidin by vitamin D. J. Am. Soc. Nephrol. 2014, 25, 564–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, J.S.; Hewison, M. Unexpected actions of vitamin D: New perspectives on the regulation of innate and adaptive immunity. Nat. Clin. Pr. Endocrinol. Metab. 2008, 4, 80–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hewison, M. Antibacterial effects of vitamin D. Nat. Rev. Endocrinol. 2011, 7, 337–345. [Google Scholar] [CrossRef]
- Yuk, J.M.; Shin, D.M.; Lee, H.M.; Yang, C.S.; Jin, H.S.; Kim, K.K.; Lee, Z.W.; Lee, S.H.; Kim, J.M.; Jo, E.K. Vitamin D3 induces autophagy in human monocytes/macrophages via cathelicidin. Cell Host Microbe 2009, 6, 231–243. [Google Scholar] [CrossRef] [Green Version]
- Shin, D.-M.; Yuk, J.-M.; Lee, H.-M.; Lee, S.-H.; Son, J.W.; Harding, C.V.; Kim, J.-M.; Modlin, R.L.; Jo, E.-K. Mycobacterial lipoprotein activates autophagy via TLR2/1/CD14 and a functional vitamin D receptor signalling. Cell Microbiol 2010, 12, 1648–1665. [Google Scholar] [CrossRef] [Green Version]
- Sun, C.C.; Vaja, V.; Babitt, J.L.; Lin, H.Y. Targeting the hepcidin-ferroportin axis to develop new treatment strategies for anemia of chronic disease and anemia of inflammation. Am. J. Hematol. 2012, 87, 392–400. [Google Scholar] [CrossRef] [Green Version]
- Kenar, G.; Köksoy, E.B.; Ürün, Y.; Utkan, G. Prevalence, etiology and risk factors of anemia in patients with newly diagnosed cancer. Supportive Care Cancer 2020, 28, 5235–5242. [Google Scholar] [CrossRef]
- Raza, U.; Sheikh, A.; Jamali, S.N.; Turab, M.; Zaidi, S.A.; Jawaid, H. Post-treatment Hematological Variations and the Role of Hemoglobin as a Predictor of Disease-free Survival in Stage 2 Breast Cancer Patients. Cureus 2020, 12, e7259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moura, F.A.; de Andrade, K.Q.; dos Santos, J.C.; Goulart, M.O. Lipoic Acid: Its antioxidant and anti-inflammatory role and clinical applications. Curr. Top. Med. Chem. 2015, 15, 458–483. [Google Scholar] [CrossRef] [PubMed]
- Shay, K.P.; Moreau, R.F.; Smith, E.J.; Smith, A.R.; Hagen, T.M. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim. Et Biophys. Acta 2009, 1790, 1149–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El-Nakib, G.A.; Mostafa, T.M.; Abbas, T.M.; El-Shishtawy, M.M.; Mabrouk, M.M.; Sobh, M.A. Role of alpha-lipoic acid in the management of anemia in patients with chronic renal failure undergoing hemodialysis. Int. J. Nephrol. Renov. Dis. 2013, 6, 161–168. [Google Scholar] [CrossRef] [Green Version]
- González-Chávez, S.A.; Arévalo-Gallegos, S.; Rascón-Cruz, Q. Lactoferrin: Structure, function and applications. Int. J. Antimicrob. Agents 2009, 33, 301.e1–301.e8. [Google Scholar] [CrossRef]
- Moastafa, T.M.; El-Sissy Ael, D.; El-Saeed, G.K.; Koura, M.S. Study on the Therapeutic Benefit on Lactoferrin in Patients with Colorectal Cancer Receiving Chemotherapy. Int. Sch. Res. Not. 2014, 2014, 184278. [Google Scholar] [CrossRef]
- Varadhachary, A.; Wolf, J.S.; Petrak, K.; O’Malley, B.W., Jr.; Spadaro, M.; Curcio, C.; Forni, G.; Pericle, F. Oral lactoferrin inhibits growth of established tumors and potentiates conventional chemotherapy. Int. J. Cancer 2004, 111, 398–403. [Google Scholar] [CrossRef]
- Giansanti, F.; Panella, G.; Leboffe, L.; Antonini, G. Lactoferrin from Milk: Nutraceutical and Pharmacological Properties. Pharmaceuticals (Basel) 2016, 9, 61. [Google Scholar] [CrossRef] [Green Version]
- Kruzel, M.L.; Zimecki, M.; Actor, J.K. Lactoferrin in a Context of Inflammation-Induced Pathology. Front. Immunol. 2017, 8, 1438. [Google Scholar] [CrossRef]
- Legrand, D. Lactoferrin, a key molecule in immune and inflammatory processes. Biochem. Cell Biol. Biochim. Et Biol. Cell. 2012, 90, 252–268. [Google Scholar] [CrossRef]
- Sill, C.; Biehl, R.; Hoffmann, B.; Radulescu, A.; Appavou, M.S.; Farago, B.; Merkel, R.; Richter, D. Structure and domain dynamics of human lactoferrin in solution and the influence of Fe(III)-ion ligand binding. BMC Biophys. 2016, 9, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu. Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef] [PubMed]
- Hamam, F.; Shahidi, F. Synthesis of structured lipids containing medium-chain and omega-3 fatty acids. J. Agric. Food Chem. 2006, 54, 4390–4396. [Google Scholar] [CrossRef]
- Senanayake, S.P.; Shahidi, F. Structured lipids via lipase-catalyzed incorporation of eicosapentaenoic acid into borage (Borago officinalis L.) and evening primrose (Oenothera biennis L.) oils. J. Agric. Food Chem. 2002, 50, 477–483. [Google Scholar] [CrossRef] [PubMed]
- Hixson, S.M.; Shukla, K.; Campbell, L.G.; Hallett, R.H.; Smith, S.M.; Packer, L.; Arts, M.T. Long-Chain Omega-3 Polyunsaturated Fatty Acids Have Developmental Effects on the Crop Pest, the Cabbage White Butterfly Pieris rapae. PLoS ONE 2016, 11, e0152264. [Google Scholar] [CrossRef] [Green Version]
- Beckermann, B.; Beneke, M.; Seitz, I. Comparative bioavailability of eicosapentaenoic acid and docasahexaenoic acid from triglycerides, free fatty acids and ethyl esters in volunteers. Arzneim. Forsch. 1990, 40, 700–704. [Google Scholar]
- Mozaffarian, D.; Lemaitre, R.N.; King, I.B.; Song, X.; Huang, H.; Sacks, F.M.; Rimm, E.B.; Wang, M.; Siscovick, D.S. Plasma phospholipid long-chain ω-3 fatty acids and total and cause-specific mortality in older adults: A cohort study. Ann. Intern. Med. 2013, 158, 515–525. [Google Scholar] [CrossRef]
- Laidlaw, M.; Cockerline, C.A.; Rowe, W.J. A randomized clinical trial to determine the efficacy of manufacturers’ recommended doses of omega-3 fatty acids from different sources in facilitating cardiovascular disease risk reduction. Lipids Health Dis. 2014, 13, 99. [Google Scholar] [CrossRef] [Green Version]
- National Institute of Health. Omega-3 Fatty Acids. Available online: https://ods.od.nih.gov/factsheets/Omega3FattyAcids-HealthProfessional/#en5 (accessed on 5 October 2020).
- Finley, J.W.; Shahidi, F. The Chemistry, Processing, and Health Benefits of Highly Unsaturated Fatty Acids: An Overview. In Omega-3 Fatty Acids; American Chemical Society: Washington, DC, USA, 2001; Volume 788, pp. 2–11. [Google Scholar]
- Calder, P.C. Omega-3 fatty acids and inflammatory processes: From molecules to man. Biochem. Soc. Trans. 2017, 45, 1105–1115. [Google Scholar] [CrossRef] [Green Version]
- Shahidi, F. Omega-3 fatty acids and marine oils in cardiovascular and general health: A critical overview of controversies and realities. J. Funct. Foods 2015, 19. [Google Scholar] [CrossRef]
- Shahidi, F.; Ambigaipalan, P. Novel functional food ingredients from marine sources. Curr. Opin. Food Sci. 2015, 2, 123–129. [Google Scholar] [CrossRef]
- Shahidi, F.; Ambigaipalan, P. Beverages Fortified with Omega-3 Fatty Acids, Dietary Fiber, Minerals, and Vitamins. In Handbook of Functional Beverages and Human Health; Shahidi, F., Alasalvar, C., Eds.; Routledge: London, UK, 2016; pp. 801–813. [Google Scholar] [CrossRef]
- Costantini, L.M.R.; Farinon, B.; Merendino, N. Impact of Omega-3 Fatty Acids on the Gut Microbiota. Int. J. Mol. Sci. 2017, 18, 2645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ministero Della Salute. Probiotici e Prebiotici. 2019. Available online: http://www.salute.gov.it/portale/temi/p2_6.jsp?lingua=italiano&id=1426&area=Alimenti%20particolari%20e%20integratori&menu=integratori (accessed on 5 October 2020).
- Ministero Della Salute. Linee Guida su Probiotici e Prebiotici-Revisione Marzo 2018. Available online: http://www.salute.gov.it/portale/news/p3_2_1_1_1.jsp?id=3353&lingua=italiano&menu=notizie&p=dalministero (accessed on 5 October 2020).
- Cruchet, S.; Furnes, R.; Maruy, A.; Hebel, E.; Palacios, J.; Medina, F.; Ramirez, N.; Orsi, M.; Rondon, L.; Sdepanian, V.; et al. The use of probiotics in pediatric gastroenterology: A review of the literature and recommendations by Latin-American experts. Paediatr. Drugs 2015, 17, 199–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R.B.; et al. Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018, 174, 1388–1405.e1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Metchnikoff, E. Essais optimistes. In The Prolongation of Life: Optimistic Studies; Mitchell, P.C., Ed.; Mauro Liistro Editore: London, UK, 1907. [Google Scholar]
- Williams, N.T. Probiotics. Am. J. Health Syst. Pharm. 2010, 67, 449–458. [Google Scholar] [CrossRef]
- Lawrence, K.; Hyde, J. Microbiome restoration diet improves digestion, cognition and physical and emotional wellbeing. PLoS ONE 2017, 12, e0179017. [Google Scholar] [CrossRef] [Green Version]
- Shi, L.H.; Balakrishnan, K.; Thiagarajah, K.; Mohd Ismail, N.I.; Yin, O.S. Beneficial Properties of Probiotics. Trop. Life Sci. Res. 2016, 27, 73–90. [Google Scholar] [CrossRef]
- Wilkins, T.; Sequoia, J. Probiotics for Gastrointestinal Conditions: A Summary of the Evidence. Am. Fam. Physician 2017, 96, 170–178. [Google Scholar]
- Mack, D.R.; Ahrne, S.; Hyde, L.; Wei, S.; Hollingsworth, M.A. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut 2003, 52, 827–833. [Google Scholar] [CrossRef] [Green Version]
- Duncan, S.H.; Louis, P.; Flint, H.J. Lactate-utilizing bacteria, isolated from human feces, that produce butyrate as a major fermentation product. Appl. Environ. Microbiol. 2004, 70, 5810–5817. [Google Scholar] [CrossRef] [Green Version]
- Lamprecht, M.; Bogner, S.; Schippinger, G.; Steinbauer, K.; Fankhauser, F.; Hallstroem, S.; Schuetz, B.; Greilberger, J.F. Probiotic supplementation affects markers of intestinal barrier, oxidation, and inflammation in trained men; a randomized, double-blinded, placebo-controlled trial. J. Int. Soc. Sports Nutr. 2012, 9, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bravo, J.A.; Julio-Pieper, M.; Forsythe, P.; Kunze, W.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Communication between gastrointestinal bacteria and the nervous system. Curr. Opin. Pharmacol. 2012, 12, 667–672. [Google Scholar] [CrossRef] [PubMed]
- Benton, D.; Williams, C.; Brown, A. Impact of consuming a milk drink containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 2007, 61, 355–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gruenwald, J.; Graubaum, H.J.; Harde, A. Effect of a probiotic multivitamin compound on stress and exhaustion. Adv. Ther. 2002, 19, 141–150. [Google Scholar] [CrossRef]
- Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef] [Green Version]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
- Roy, S.; Trinchieri, G. Microbiota: A key orchestrator of cancer therapy. Nat. Rev. Cancer 2017, 17, 271–285. [Google Scholar] [CrossRef]
- Gori, S.; Inno, A.; Belluomini, L.; Bocus, P.; Bisoffi, Z.; Russo, A.; Arcaro, G. Gut microbiota and cancer: How gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit. Rev. Oncol. Hematol. 2019, 143, 139–147. [Google Scholar] [CrossRef]
- Tsilimigras, M.C.; Fodor, A.; Jobin, C. Carcinogenesis and therapeutics: The microbiota perspective. Nat. Microbiol. 2017, 2, 17008. [Google Scholar] [CrossRef]
- Ciernikova, S.; Mego, M.; Semanova, M.; Wachsmannova, L.; Adamcikova, Z.; Stevurkova, V.; Drgona, L.; Zajac, V. Probiotic Survey in Cancer Patients Treated in the Outpatient Department in a Comprehensive Cancer Center. Integr. Cancer Ther. 2017, 16, 188–195. [Google Scholar] [CrossRef]
- Dasari, S.; Kathera, C.; Janardhan, A.; Praveen Kumar, A.; Viswanath, B. Surfacing role of probiotics in cancer prophylaxis and therapy: A systematic review. Clin. Nutr. (Edinbburgh Scotl.) 2017, 36, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
- Vonderheid, S.C.; Tussing-Humphreys, L.; Park, C.; Pauls, H.; OjiNjideka Hemphill, N.; LaBomascus, B.; McLeod, A.; Koenig, M.D. A Systematic Review and Meta-Analysis on the Effects of Probiotic Species on Iron Absorption and Iron Status. Nutrients 2019, 11, 2938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korčok, D.J.; Tršić-Milanović, N.A.; Ivanović, N.D.; Đorđević, B.I. Development of Probiotic Formulation for the Treatment of Iron Deficiency Anemia. Chem. Pharm. Bull. 2018, 66, 347–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hoppe, M.; Önning, G.; Berggren, A.; Hulthén, L. Probiotic strain Lactobacillus plantarum 299v increases iron absorption from an iron-supplemented fruit drink: A double-isotope cross-over single-blind study in women of reproductive age. Br. J. Nutr. 2015, 114, 1195–1202. [Google Scholar] [CrossRef] [Green Version]
- Wells, J.M. Immunomodulatory mechanisms of lactobacilli. Microb. Cell Fact. 2011, 10 (Suppl. 1), S17. [Google Scholar] [CrossRef] [Green Version]
- Hoppe, M.; Önning, G.; Hulthén, L. Freeze-dried Lactobacillus plantarum 299v increases iron absorption in young females-Double isotope sequential single-blind studies in menstruating women. PLoS ONE 2017, 12, e0189141. [Google Scholar] [CrossRef]
- González, A.; Gálvez, N.; Martín, J.; Reyes, F.; Pérez-Victoria, I.; Dominguez-Vera, J.M. Identification of the key excreted molecule by Lactobacillus fermentum related to host iron absorption. Food Chem. 2017, 228, 374–380. [Google Scholar] [CrossRef] [Green Version]
- Madu, A.J.; Ughasoro, M.D. Anaemia of Chronic Disease: An In-Depth Review. Med. Princ. Pract. 2017, 26, 1–9. [Google Scholar] [CrossRef]
- Akbari, M.; Ostadmohammadi, V.; Lankarani, K.B.; Tabrizi, R.; Kolahdooz, F.; Heydari, S.T.; Kavari, S.H.; Mirhosseini, N.; Mafi, A.; Dastorani, M.; et al. The Effects of Vitamin D Supplementation on Biomarkers of Inflammation and Oxidative Stress Among Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Horm. Metab. Res. 2018, 50, 271–279. [Google Scholar] [CrossRef]
- Tabrizi, R.; Vakili, S.; Akbari, M.; Mirhosseini, N.; Lankarani, K.B.; Rahimi, M.; Mobini, M.; Jafarnejad, S.; Vahedpoor, Z.; Asemi, Z. The effects of curcumin-containing supplements on biomarkers of inflammation and oxidative stress: A systematic review and meta-analysis of randomized controlled trials. Phytother Res. 2019, 33, 253–262. [Google Scholar] [CrossRef]
- Koushki, M.; Dashatan, N.A.; Meshkani, R. Effect of Resveratrol Supplementation on Inflammatory Markers: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Clin. Ther. 2018, 40, 1180–1192. [Google Scholar] [CrossRef] [PubMed]
- Tabrizi, R.; Tamtaji, O.R.; Mirhosseini, N.; Lankarani, K.B.; Akbari, M.; Heydari, S.T.; Dadgostar, E.; Asemi, Z. The effects of quercetin supplementation on lipid profiles and inflammatory markers among patients with metabolic syndrome and related disorders: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2020, 60, 1855–1868. [Google Scholar] [CrossRef] [PubMed]
- Saboori, S.; Falahi, E.; Eslampour, E.; Zeinali Khosroshahi, M.; Yousefi Rad, E. Effects of alpha-lipoic acid supplementation on C-reactive protein level: A systematic review and meta-analysis of randomized controlled clinical trials. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 779–786. [Google Scholar] [CrossRef] [PubMed]
- Hao, L.; Shan, Q.; Wei, J.; Ma, F.; Sun, P. Lactoferrin: Major Physiological Functions and Applications. Curr. Protein. Pept. Sci. 2019, 20, 139–144. [Google Scholar] [CrossRef] [PubMed]
- Custodero, C.; Mankowski, R.T.; Lee, S.A.; Chen, Z.; Wu, S.; Manini, T.M.; Hincapie Echeverri, J.; Sabbà, C.; Beavers, D.P.; Cauley, J.A.; et al. Evidence-based nutritional and pharmacological interventions targeting chronic low-grade inflammation in middle-age and older adults: A systematic review and meta-analysis. Ageing Res. Rev. 2018, 46, 42–59. [Google Scholar] [CrossRef]
- Mathur, N.; Pedersen, B.K. Exercise as a mean to control low-grade systemic inflammation. Mediat. Inflamm 2008, 2008, 109502. [Google Scholar] [CrossRef] [Green Version]
- Hoffmann, C.; Weigert, C. Skeletal Muscle as an Endocrine Organ: The Role of Myokines in Exercise Adaptations. Cold Spring Harb Perspect Med. 2017, 7, a029793. [Google Scholar] [CrossRef]
- Petersen, A.M.; Pedersen, B.K. The anti-inflammatory effect of exercise. J. Appl. Physiol. (Bethesda Md. 1985) 2005, 98, 1154–1162. [Google Scholar] [CrossRef] [Green Version]
- Pedersen, B.K.; Febbraio, M.A. Muscles, exercise and obesity: Skeletal muscle as a secretory organ. Nat. Rev. Endocrinol. 2012, 8, 457–465. [Google Scholar] [CrossRef]
- Fischer, C.P.; Hiscock, N.J.; Penkowa, M.; Basu, S.; Vessby, B.; Kallner, A.; Sjöberg, L.B.; Pedersen, B.K. Supplementation with vitamins C and E inhibits the release of interleukin-6 from contracting human skeletal muscle. J. Physiol. 2004, 558, 633–645. [Google Scholar] [CrossRef]
- Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nat. Rev. Immunol. 2011, 11, 85–97. [Google Scholar] [CrossRef] [PubMed]
- Fuster, J.J.; Ouchi, N.; Gokce, N.; Walsh, K. Obesity-Induced Changes in Adipose Tissue Microenvironment and Their Impact on Cardiovascular Disease. Circ. Res. 2016, 118, 1786–1807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pedersen, B.K.; Hoffman-Goetz, L. Exercise and the immune system: Regulation, integration, and adaptation. Physiol. Rev. 2000, 80, 1055–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nemeth, E.; Rivera, S.; Gabayan, V.; Keller, C.; Taudorf, S.; Pedersen, B.K.; Ganz, T. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Investig. 2004, 113, 1271–1276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kong, W.N.; Gao, G.; Chang, Y.Z. Hepcidin and sports anemia. Cell Biosci. 2014, 4, 19. [Google Scholar] [CrossRef] [Green Version]
- Peeling, P.; Dawson, B.; Goodman, C.; Landers, G.; Wiegerinck, E.T.; Swinkels, D.W.; Trinder, D. Effects of exercise on hepcidin response and iron metabolism during recovery. Int. J. Sport Nutr. Exerc. Metab. 2009, 19, 583–597. [Google Scholar] [CrossRef] [Green Version]
- Domínguez, R.; Sánchez-Oliver, A.J. Effects of an Acute Exercise Bout on Serum Hepcidin Levels. Nutrients 2018, 10, 209. [Google Scholar] [CrossRef] [Green Version]
- Sim, M.; Dawson, B.; Landers, G.; Trinder, D.; Peeling, P. Iron regulation in athletes: Exploring the menstrual cycle and effects of different exercise modalities on hepcidin production. Int. J. Sport Nutr. Exerc. Metab. 2014, 24, 177–187. [Google Scholar] [CrossRef]
- Ishibashi, A.; Maeda, N.; Sumi, D.; Goto, K. Elevated Serum Hepcidin Levels during an Intensified Training Period in Well-Trained Female Long-Distance Runners. Nutrients 2017, 9, 277. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.Q.; Duan, X.L.; Chang, Y.Z.; Wang, H.T.; Qian, Z.M. Molecular analysis of increased iron status in moderately exercised rats. Mol. Cell. Biochem. 2006, 282, 117–123. [Google Scholar] [CrossRef]
- Troadec, M.B.; Lainé, F.; Daniel, V.; Rochcongar, P.; Ropert, M.; Cabillic, F.; Perrin, M.; Morcet, J.; Loréal, O.; Olbina, G.; et al. Daily regulation of serum and urinary hepcidin is not influenced by submaximal cycling exercise in humans with normal iron metabolism. Eur. J. Appl. Physiol. 2009, 106, 435–443. [Google Scholar] [CrossRef] [PubMed]
- Riebe, D.; Ehrman, J.K.; Ligouri, G.; Magal, M. ACSM’s Guidelines for Excercise Testing and Prescription (American College and Sports Medicine: Indianapolis), 10th ed.; Lippincott Williams Wilkins: Philadelphia, PA, USA, 2017. [Google Scholar]
- Pedersen, B.K. Exercise and cytokines. Immunol. Cell Biol. 2000, 78, 532–535. [Google Scholar] [CrossRef] [PubMed]
- Luo, H.; Galvão, D.; Newton, R.; Fairman, C.M.; Taaffe, D.R. Sport Medicine in the Prevention and Management of Cancer. Integr. Cancer. 2019, 18, 1534735419894063. [Google Scholar] [CrossRef] [PubMed]
- Agostini, D.; Natalucci, V.; Baldelli, G.; De Santi, M.; Donati Zeppa, S.; Vallorani, L.; Annibalini, G.; Lucertini, F.; Federici, A.; Izzo, R.; et al. New Insights into the Role of Exercise in Inhibiting mTOR Signaling in Triple-Negative Breast Cancer. Oxid. Med. Cell Longev. 2018, 2018, 5896786. [Google Scholar] [CrossRef]
- Baldelli, G.; De Santi, M.; Gervasi, M.; Annibalini, G.; Sisti, D.; Højman, P.; Sestili, P.; Stocchi, V.; Barbieri, E.; Brandi, G. The effects of human sera conditioned by high-intensity exercise sessions and training on the tumorigenic potential of cancer cells. Clin. Transl. Oncol. 2020. [Google Scholar] [CrossRef]
- De Santi, M.; Baldelli, G.; Lucertini, F.; Natalucci, V.; Brandi, G.; Barbieri, E. A dataset on the effect of exercise-conditioned human sera in three-dimensional breast cancer cell culture. Data Brief. 2019, 27, 104704. [Google Scholar] [CrossRef]
- Pedersen, B.K.; Saltin, B. Evidence for prescribing exercise as therapy in chronic disease. Scand. J. Med. Sci. Sports 2006, 16 (Suppl. 1), 3–63. [Google Scholar] [CrossRef]
- Codella, R.; Luzi, L.; Inverardi, L.; Ricordi, C. The anti-inflammatory effects of exercise in the syndromic thread of diabetes and autoimmunity. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 3709–3722. [Google Scholar]
- Cronin, O.; Keohane, D.M.; Molloy, M.G.; Shanahan, F. The effect of exercise interventions on inflammatory biomarkers in healthy, physically inactive subjects: A systematic review. QJM Mon. J. Assoc. Physicians 2017, 110, 629–637. [Google Scholar] [CrossRef] [Green Version]
- Peake, J.M.; Neubauer, O.; Walsh, N.P.; Simpson, R.J. Recovery of the immune system after exercise. J. Appl. Physiol. (Bethesda Md. 1985) 2017, 122, 1077–1087. [Google Scholar] [CrossRef]
- Moldoveanu, A.I.; Shephard, R.J.; Shek, P.N. The cytokine response to physical activity and training. Sports Med. (Auckland N.Z.) 2001, 31, 115–144. [Google Scholar] [CrossRef] [PubMed]
- Allen, J.; Sun, Y.; Woods, J.A. Exercise and the Regulation of Inflammatory Responses. Prog. Mol. Biol. Transl. Sci. 2015, 135, 337–354. [Google Scholar] [CrossRef] [PubMed]
- Bigley, A.B.; Simpson, R.J. NK cells and exercise: Implications for cancer immunotherapy and survivorship. Discov. Med. 2015, 19, 433–445. [Google Scholar] [PubMed]
- Silveira, L.S.; Antunes Bde, M.; Minari, A.L.; Dos Santos, R.V.; Neto, J.C.; Lira, F.S. Macrophage Polarization: Implications on Metabolic Diseases and the Role of Exercise. Crit. Rev. Eukaryot. Gene Expr. 2016, 26, 115–132. [Google Scholar] [CrossRef] [Green Version]
- Hennigar, S.R.; McClung, J.P.; Pasiakos, S.M. Nutritional interventions and the IL-6 response to exercise. FASEB J. 2017, 31, 3719–3728. [Google Scholar] [CrossRef] [Green Version]
- Fischer, C.P. Interleukin-6 in acute exercise and training: What is the biological relevance? Exerc. Immunol. Rev. 2006, 12, 6–33. [Google Scholar] [PubMed]
- Steensberg, A.; Fischer, C.P.; Keller, C.; Møller, K.; Pedersen, B.K. IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. American journal of physiology. Endocrinol. Metab. 2003, 285, E433–E437. [Google Scholar] [CrossRef]
- Svensson, M.; Lexell, J.; Deierborg, T. Effects of Physical Exercise on Neuroinflammation, Neuroplasticity, Neurodegeneration, and Behavior: What We Can Learn from Animal Models in Clinical Settings. Neurorehabilit. Neural Repair 2015, 29, 577–589. [Google Scholar] [CrossRef]
- Pedersen, B.K.; Toft, A.D. Effects of exercise on lymphocytes and cytokines. Br. J. Sports Med. 2000, 34, 246–251. [Google Scholar] [CrossRef] [Green Version]
- King, D.E.; Carek, P.; Mainous, A.G., 3rd; Pearson, W.S. Inflammatory markers and exercise: Differences related to exercise type. Med. Sci. Sports Exerc. 2003, 35, 575–581. [Google Scholar] [CrossRef]
- Walsh, N.P.; Gleeson, M.; Shephard, R.J.; Gleeson, M.; Woods, J.A.; Bishop, N.C.; Fleshner, M.; Green, C.; Pedersen, B.K.; Hoffman-Goetz, L.; et al. Position statement. Part one: Immune function and exercise. Exerc. Immunol. Rev. 2011, 17, 6–63. [Google Scholar] [PubMed]
- Gokhale, R.; Chandrashekara, S.; Vasanthakumar, K.C. Cytokine response to strenuous exercise in athletes and non-athletes--an adaptive response. Cytokine 2007, 40, 123–127. [Google Scholar] [CrossRef] [PubMed]
- Woods, J.; Lu, Q.; Ceddia, M.A.; Lowder, T. Special feature for the Olympics: Effects of exercise on the immune system: Exercise-induced modulation of macrophage function. Immunol. Cell Biol. 2000, 78, 545–553. [Google Scholar] [CrossRef] [PubMed]
- Nieman, D.C.; Wentz, L.M. The compelling link between physical activity and the body’s defense system. J. Sport Health Sci. 2019, 8, 201–217. [Google Scholar] [CrossRef] [PubMed]
- Kelly, P.; Kahlmeier, S.; Götschi, T.; Orsini, N.; Richards, J.; Roberts, N.; Scarborough, P.; Foster, C. Systematic review and meta-analysis of reduction in all-cause mortality from walking and cycling and shape of dose response relationship. Int. J. Behav. Nutr. Phys. Act. 2014, 11, 132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicklas, B.J.; Hsu, F.C.; Brinkley, T.J.; Church, T.; Goodpaster, B.H.; Kritchevsky, S.B.; Pahor, M. Exercise training and plasma C-reactive protein and interleukin-6 in elderly people. J. Am. Geriatr. Soc. 2008, 56, 2045–2052. [Google Scholar] [CrossRef]
- Timmerman, K.L.; Flynn, M.G.; Coen, P.M.; Markofski, M.M.; Pence, B.D. Exercise training-induced lowering of inflammatory (CD14+CD16+) monocytes: A role in the anti-inflammatory influence of exercise? J. Leukoc Biol. 2008, 84, 1271–1278. [Google Scholar] [CrossRef]
- Sloan, R.P.; Shapiro, P.A.; Demeersman, R.E.; McKinley, P.S.; Tracey, K.J.; Slavov, I.; Fang, Y.; Flood, P.D. Aerobic exercise attenuates inducible TNF production in humans. J. Appl. Physiol. (Bethesda Md. 1985) 2007, 103, 1007–1011. [Google Scholar] [CrossRef]
- Gleeson, M.; McFarlin, B.; Flynn, M. Exercise and Toll-like receptors. Exerc. Immunol. Rev. 2006, 12, 34–53. [Google Scholar]
- Mackinnon, L.T. Chronic exercise training effects on immune function. Med. Sci. Sports Exerc. 2000, 32, S369–S376. [Google Scholar] [CrossRef]
- Gleeson, M. Immune function in sport and exercise. J. Appl. Physiol. (Bethesda Md. 1985) 2007, 103, 693–699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Timmons, B.W. Paediatric exercise immunology: Health and clinical applications. Exerc. Immunol. Rev. 2005, 11, 108–144. [Google Scholar] [PubMed]
- Gropper, S.S.; Blessing, D.; Dunham, K.; Barksdale, J.M. Iron status of female collegiate athletes involved in different sports. Biol. Trace Elem. Res. 2006, 109, 1–14. [Google Scholar] [CrossRef]
- Zoller, H.; Vogel, W. Iron supplementation in athletes--first do no harm. Nutrition 2004, 20, 615–619. [Google Scholar] [CrossRef] [PubMed]
- Woolf, K.; St Thomas, M.M.; Hahn, N.; Vaughan, L.A.; Carlson, A.G.; Hinton, P. Iron status in highly active and sedentary young women. Int. J. Sport Nutr. Exerc. Metab. 2009, 19, 519–535. [Google Scholar] [CrossRef]
- Sinclair, L.M.; Hinton, P.S. Prevalence of iron deficiency with and without anemia in recreationally active men and women. J. Am. Diet. Assoc. 2005, 105, 975–978. [Google Scholar] [CrossRef] [PubMed]
- Roecker, L.; Meier-Buttermilch, R.; Brechtel, L.; Nemeth, E.; Ganz, T. Iron-regulatory protein hepcidin is increased in female athletes after a marathon. Eur. J. Appl. Physiol. 2005, 95, 569–571. [Google Scholar] [CrossRef]
- Pedersen, B.K.; Steensberg, A.; Schjerling, P. Muscle-derived interleukin-6: Possible biological effects. J. Physiol. 2001, 536, 329–337. [Google Scholar] [CrossRef]
- Deng, F.; Li, Y.; Zhao, J. The gut microbiome of healthy long-living people. Aging 2019, 11, 289–290. [Google Scholar] [CrossRef]
- Codella, R.; Luzi, L.; Terruzzi, I. Exercise has the guts: How physical activity may positively modulate gut microbiota in chronic and immune-based diseases. Dig. Liver Dis. 2018, 50, 331–341. [Google Scholar] [CrossRef] [Green Version]
- Hoffman-Goetz, L.; Spagnuolo, P.A.; Guan, J. Repeated exercise in mice alters expression of IL-10 and TNF-alpha in intestinal lymphocytes. Brain Behav. Immun. 2008, 22, 195–199. [Google Scholar] [CrossRef] [PubMed]
- Luo, B.; Xiang, D.; Nieman, D.C.; Chen, P. The effects of moderate exercise on chronic stress-induced intestinal barrier dysfunction and antimicrobial defense. Brain Behav. Immun. 2014, 39, 99–106. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Miguelez, P.; Fernandez-Gonzalo, R.; Almar, M.; Mejías, Y.; Rivas, A.; de Paz, J.A.; Cuevas, M.J.; González-Gallego, J. Role of Toll-like receptor 2 and 4 signaling pathways on the inflammatory response to resistance training in elderly subjects. Age (Dordr. Neth.) 2014, 36, 9734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rehrer, N.J.; Smets, A.; Reynaert, H.; Goes, E.; De Meirleir, K. Effect of exercise on portal vein blood flow in man. Med. Sci. Sports Exerc. 2001, 33, 1533–1537. [Google Scholar] [CrossRef] [PubMed]
- Van Wijck, K.; Lenaerts, K.; van Loon, L.J.; Peters, W.H.; Buurman, W.A.; Dejong, C.H. Exercise-induced splanchnic hypoperfusion results in gut dysfunction in healthy men. PLoS ONE 2011, 6, e22366. [Google Scholar] [CrossRef] [Green Version]
- Oktedalen, O.; Lunde, O.C.; Opstad, P.K.; Aabakken, L.; Kvernebo, K. Changes in the gastrointestinal mucosa after long-distance running. Scand. J. Gastroenterol. 1992, 27, 270–274. [Google Scholar] [CrossRef]
- Pervaiz, N.; Hoffman-Goetz, L. Immune cell inflammatory cytokine responses differ between central and systemic compartments in response to acute exercise in mice. Exerc. Immunol. Rev. 2012, 18, 142–157. [Google Scholar]
- O’Sullivan, O.; Cronin, O.; Clarke, S.F.; Murphy, E.F.; Molloy, M.G.; Shanahan, F.; Cotter, P.D. Exercise and the microbiota. Gut Microbes 2015, 6, 131–136. [Google Scholar] [CrossRef] [Green Version]
- Kerr, J.; Anderson, C.; Lippman, S.M. Physical activity, sedentary behaviour, diet, and cancer: An update and emerging new evidence. Lancet Oncol. 2017, 18, e457–e471. [Google Scholar] [CrossRef]
- Newton, R.U.; Galvão, D.A. Exercise in prevention and management of cancer. Curr. Treat. Options Oncol. 2008, 9, 135–146. [Google Scholar] [CrossRef]
- Ballard-Barbash, R.; Friedenreich, C.M.; Courneya, K.S.; Siddiqi, S.M.; McTiernan, A.; Alfano, C.M. Physical activity, biomarkers, and disease outcomes in cancer survivors: A systematic review. J. Natl. Cancer Inst. 2012, 104, 815–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sasso, J.P.; Eves, N.D.; Christensen, J.F.; Koelwyn, G.J.; Scott, J.; Jones, L.W. A framework for prescription in exercise-oncology research. J. Cachexia Sarcopenia Muscle 2015, 6, 115–124. [Google Scholar] [CrossRef]
- Hojman, P.; Gehl, J.; Christensen, J.F.; Pedersen, B.K. Molecular Mechanisms Linking Exercise to Cancer Prevention and Treatment. Cell Metab. 2018, 27, 10–21. [Google Scholar] [CrossRef] [Green Version]
- Devin, J.L.; Hill, M.M.; Mourtzakis, M.; Quadrilatero, J.; Jenkins, D.G.; Skinner, T.L. Acute high intensity interval exercise reduces colon cancer cell growth. J. Physiol. 2019, 597, 2177–2184. [Google Scholar] [CrossRef] [PubMed]
- Dethlefsen, C.; Lillelund, C.; Midtgaard, J.; Andersen, C.; Pedersen, B.K.; Christensen, J.F.; Hojman, P. Exercise regulates breast cancer cell viability: Systemic training adaptations versus acute exercise responses. Breast Cancer Res. Treat. 2016, 159, 469–479. [Google Scholar] [CrossRef] [PubMed]
- Campbell, K.L.; Winters-Stone, K.M.; Wiskemann, J.; May, A.M.; Schwartz, A.L.; Courneya, K.S.; Zucker, D.S.; Matthews, C.E.; Ligibel, J.A.; Gerber, L.H.; et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Med. Sci. Sports Exerc. 2019, 51, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
- Weinkove, R.; McQuilten, Z.K. Managing haematology and oncology patients during the COVID-19 pandemic: Interim consensus guidance. Med. J. Aust. 2020, 212, 481–489. [Google Scholar] [CrossRef]
- Segelov, E.; Underhill, C.; Prenen, H.; Karapetis, C.; Jackson, C.; Nott, L.; Clay, T.; Pavlakis, N.; Sabesan, S.; Heywood, E.; et al. Practical Considerations for Treating Patients with Cancer in the COVID-19 Pandemic. JCO Oncol. Pract. 2020, 16, 467–482. [Google Scholar] [CrossRef]
- Liang, W.; Liang, H.; Ou, L.; Chen, B.; Chen, A.; Li, C.; Li, Y.; Guan, W.; Sang, L.; Lu, J.; et al. Development and Validation of a Clinical Risk Score to Predict the Occurrence of Critical Illness in Hospitalized Patients With COVID-19. JAMA Intern. Med. 2020, 180, 1081–1089. [Google Scholar] [CrossRef]
- Yu, J.; Ouyang, W.; Chua, M.L.K.; Xie, C. SARS-CoV-2 Transmission in Patients with Cancer at a Tertiary Care Hospital in Wuhan, China. JAMA Oncol. 2020, 6, 1108–1110. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Natalucci, V.; Virgili, E.; Calcagnoli, F.; Valli, G.; Agostini, D.; Zeppa, S.D.; Barbieri, E.; Emili, R. Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients 2021, 13, 482. https://doi.org/10.3390/nu13020482
Natalucci V, Virgili E, Calcagnoli F, Valli G, Agostini D, Zeppa SD, Barbieri E, Emili R. Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients. 2021; 13(2):482. https://doi.org/10.3390/nu13020482
Chicago/Turabian StyleNatalucci, Valentina, Edy Virgili, Federica Calcagnoli, Giacomo Valli, Deborah Agostini, Sabrina Donati Zeppa, Elena Barbieri, and Rita Emili. 2021. "Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions" Nutrients 13, no. 2: 482. https://doi.org/10.3390/nu13020482
APA StyleNatalucci, V., Virgili, E., Calcagnoli, F., Valli, G., Agostini, D., Zeppa, S. D., Barbieri, E., & Emili, R. (2021). Cancer Related Anemia: An Integrated Multitarget Approach and Lifestyle Interventions. Nutrients, 13(2), 482. https://doi.org/10.3390/nu13020482