51
|
Juárez-Cruz JC, Okoniewski M, Ramírez M, Ortuño-Pineda C, Navarro-Tito N, Castañeda-Saucedo E. Chronic Leptin Treatment Induces Epithelial-Mesenchymal Transition in MCF10A Mammary Epithelial Cells. J Mammary Gland Biol Neoplasia 2022; 27:19-36. [PMID: 35195812 DOI: 10.1007/s10911-022-09515-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 02/08/2022] [Indexed: 01/06/2023] Open
Abstract
Leptin is a cytokine-like hormone that functions as a link between obesity and breast cancer (BC). Leptin treatment induces Epithelial to Mesenchymal Transition (EMT) in BC cell lines. In non-tumoral breast epithelial MCF10A cells, acute leptin treatment induces partial EMT. However, the effect of chronic leptin treatment on EMT in non-tumorigenic breast cells has not been fully explored. This study aimed to evaluate the effect of chronic leptin treatment on the induction of EMT in MCF10A cells. We found that chronic leptin treatment induces a switch from an epithelial to a mesenchymal morphology, partial loss of E-cadherin and gain of vimentin expression. Immunolocalization experiments showed a partial loss of E-cadherin at cell junctions and increased cytoplasmic localization of vimentin in leptin-treated cells. Moreover, chronic leptin treatment increased collective cell migration and invasion. Furthermore, when cultured in non-adherent conditions leptin treated cells exhibited reduced cell aggregation, increased survival, and decreased apoptosis, which correlates with increased FAK and AKT phosphorylation. Finally, bioinformatic analysis in two publicly available RNAseq datasets from normal breast tissue shows that high levels of leptin mRNA correlate positively with the expression of mesenchymal markers, and negatively with epithelial markers. Thus, our results demonstrate that chronic leptin treatment induces EMT in non-tumorigenic MCF10A cells and suggest that high leptin expression in normal breast tissue may induce EMT and contribute to increased risk of breast cancer.
Collapse
Affiliation(s)
- Juan Carlos Juárez-Cruz
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México
| | | | - Mónica Ramírez
- CONACYT, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Guerrero, México
| | - Carlos Ortuño-Pineda
- Laboratorio de Ácidos Nucleicos y Proteínas. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo, Guerrero, México
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer. Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero. Av. Lázaro Cárdenas S/N Ciudad Universitaria. C.P, 39087, Chilpancingo de los Bravo, Guerrero, México.
| |
Collapse
|
52
|
Petrescu AD, Grant S, Williams E, An SY, Seth N, Shell M, Amundsen T, Tan C, Nadeem Y, Tjahja M, Weld L, Chu CS, Venter J, Frampton G, McMillin M, DeMorrow S. Leptin Enhances Hepatic Fibrosis and Inflammation in a Mouse Model of Cholestasis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:484-502. [PMID: 34896073 PMCID: PMC8895426 DOI: 10.1016/j.ajpath.2021.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 10/27/2021] [Accepted: 11/17/2021] [Indexed: 01/09/2023]
Abstract
Leptin is an adipokine with roles in food intake and energy metabolism through its actions on neurons in the hypothalamus. The role of leptin in obesity and cardiovascular disorders is well documented. However, its influence on liver conditions such as cholestasis is poorly understood. The effects of exogenous leptin and leptin-neutralizing antibody on biliary hyperplasia, hepatic fibrosis, and inflammation in the multidrug resistance protein 2 knockout (Mdr2KO) mouse model of cholestasis were assessed by quantifying markers specific for cholangiocytes, activated hepatic stellate cells (HSCs), and cytokines. Serum and hepatic leptin were increased in Mdr2KO mice compared with FVB/NJ (FVBN) controls, and exogenous leptin enhanced biliary hyperplasia and liver fibrosis in Mdr2KO and FVBN mice. Leptin administration increased hepatic expression of C-C motif chemokine ligand 2 and IL-6 in Mdr2KO mice. In contrast, leptin-neutralizing antibody reduced intrahepatic bile duct mass and decreased HSC activation in Mdr2KO mice compared with FVBN controls. Sex-related differences were noted, with female Mdr2KO mice having more leptin than males. In cholangiocytes and LX2 cells in vitro, leptin increased phosphorylated Akt and stimulated cell proliferation. Leptin receptor siRNA and inhibitors of Akt phosphorylation impaired leptin-induced cell proliferation and proinflammatory cytokines. The current data suggest that leptin is abnormally increased in cholestatic mice, and excess leptin increases ductular reaction, hepatic fibrosis, and inflammation via leptin receptor-mediated phosphorylation of Akt in cholangiocytes and HSCs.
Collapse
Affiliation(s)
- Anca D Petrescu
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Stephanie Grant
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Elaina Williams
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Su Yeon An
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Nikhil Seth
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Mark Shell
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Tyson Amundsen
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Christopher Tan
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Yusra Nadeem
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Matthew Tjahja
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Lancaster Weld
- Department of Internal Medicine, Baylor Scott & White Health, Temple, Texas
| | - Christopher S Chu
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Julie Venter
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Gabriel Frampton
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Matthew McMillin
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Central Texas Veterans Health Care System, Temple, Texas
| | - Sharon DeMorrow
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Central Texas Veterans Health Care System, Temple, Texas.
| |
Collapse
|
53
|
Rodríguez-Valentín R, Torres-Mejía G, Martínez-Matsushita L, Angeles-Llerenas A, Gómez-Flores-Ramos L, Wolff RK, Baumgartner KB, Hines LM, Ziv E, Flores-Luna L, Sánchez-Zamorano LM, Ortiz-Panozo E, Slattery ML. Energy homeostasis genes modify the association between serum concentrations of IGF-1 and IGFBP-3 and breast cancer risk. Sci Rep 2022; 12:1837. [PMID: 35115550 PMCID: PMC8813998 DOI: 10.1038/s41598-022-05496-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is a multifactorial disease in which the interplay among multiple risk factors remains unclear. Energy homeostasis genes play an important role in carcinogenesis and their interactions with the serum concentrations of IGF-1 and IGFBP-3 on the risk of breast cancer have not yet been investigated. The aim of this study was to assess the modifying effect of the genetic variation in some energy homeostasis genes on the association of serum concentrations of IGF-1 and IGFBP-3 with breast cancer risk. We analyzed 78 SNPs from 10 energy homeostasis genes in premenopausal women from the 4-Corner’s Breast Cancer Study (61 cases and 155 controls) and the Mexico Breast Cancer Study (204 cases and 282 controls). After data harmonization, 71 SNPs in HWE were included for interaction analysis. Two SNPs in two genes (MBOAT rs13272159 and NPY rs16131) showed an effect modification on the association between IGF-1 serum concentration and breast cancer risk (Pinteraction < 0.05, adjusted Pinteraction < 0.20). In addition, five SNPs in three genes (ADIPOQ rs182052, rs822391 and rs7649121, CARTPT rs3846659, and LEPR rs12059300) had an effect modification on the association between IGFBP-3 serum concentration and breast cancer risk (Pinteraction < 0.05, adjusted Pinteraction < 0.20). Our findings showed that variants of energy homeostasis genes modified the association between the IGF-1 or IGFBP-3 serum concentration and breast cancer risk in premenopausal women. These findings contribute to a better understanding of this multifactorial pathology.
Collapse
Affiliation(s)
- Rocío Rodríguez-Valentín
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Gabriela Torres-Mejía
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico.
| | | | - Angélica Angeles-Llerenas
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Liliana Gómez-Flores-Ramos
- Cátedras CONACYT-Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Roger K Wolff
- Department of Medicine, University of Utah, Salt Lake City, UT, 84108, USA
| | - Kathy B Baumgartner
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Lisa M Hines
- Department of Biology, University of Colorado Colorado Springs, Colorado Springs, CO, 80918, USA
| | - Elad Ziv
- Department of Medicine, Institute of Human Genetics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Lourdes Flores-Luna
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Luisa Ma Sánchez-Zamorano
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Eduardo Ortiz-Panozo
- Center for Population Health Research, National Institute of Public Health, 62100, Cuernavaca, Morelos, Mexico
| | - Martha L Slattery
- Department of Medicine, University of Utah, Salt Lake City, UT, 84108, USA
| |
Collapse
|
54
|
Barone I, Caruso A, Gelsomino L, Giordano C, Bonofiglio D, Catalano S, Andò S. Obesity and endocrine therapy resistance in breast cancer: Mechanistic insights and perspectives. Obes Rev 2022; 23:e13358. [PMID: 34559450 PMCID: PMC9285685 DOI: 10.1111/obr.13358] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
The incidence of obesity, a recognized risk factor for various metabolic and chronic diseases, including numerous types of cancers, has risen dramatically over the recent decades worldwide. To date, convincing research in this area has painted a complex picture about the adverse impact of high body adiposity on breast cancer onset and progression. However, an emerging but overlooked issue of clinical significance is the limited efficacy of the conventional endocrine therapies with selective estrogen receptor modulators (SERMs) or degraders (SERDs) and aromatase inhibitors (AIs) in patients affected by breast cancer and obesity. The mechanisms behind the interplay between obesity and endocrine therapy resistance are likely to be multifactorial. Therefore, what have we actually learned during these years and which are the main challenges in the field? In this review, we will critically discuss the epidemiological evidence linking obesity to endocrine therapeutic responses and we will outline the molecular players involved in this harmful connection. Given the escalating global epidemic of obesity, advances in understanding this critical node will offer new precision medicine-based therapeutic interventions and more appropriate dosing schedule for treating patients affected by obesity and with breast tumors resistant to endocrine therapies.
Collapse
Affiliation(s)
- Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| |
Collapse
|
55
|
Asal DM, Mesbah NM, Abo-Elmatty DM, Fathy H, Abdel-Hamed AR. Association of vaspin rs2236242 and Val109Asp omentin genes polymorphism and their serum levels with increased risk of breast cancer. Meta Gene 2022. [DOI: 10.1016/j.mgene.2022.101016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
56
|
Anisman H, Kusnecov AW. Cancer biology and pathology. Cancer 2022. [DOI: 10.1016/b978-0-323-91904-3.00004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
57
|
Li S, Lin J, Wei J, Zhou L, Wang P, Qu S. Sinigrin impedes the breast cancer cell growth through inhibition of PI3K/AKT/mTOR phosphorylation-mediated cell cycle arrest. J Environ Pathol Toxicol Oncol 2022; 41:33-43. [DOI: 10.1615/jenvironpatholtoxicoloncol.2022041136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
58
|
Chen P, Wang B, Li M, Cui C, Liu F, Gao Y. Celastrol inhibits the proliferation and migration of MCF-7 cells through the leptin-triggered PI3K/AKT pathway. Comput Struct Biotechnol J 2022; 20:3173-3181. [PMID: 35782744 PMCID: PMC9234344 DOI: 10.1016/j.csbj.2022.06.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Leptin is the pivotal modulator in the onset and progression of breast cancer and obesity. Celastrol, which is extracted from the roots of Tripterygium wilfordi plants, exerts various anticancer bioactivities and has recently emerged as a candidate to treat obesity by improving leptin sensitivity. However, the relationship between leptin and celastrol in the treatment of breast cancer is unknown. Here, the growth and migration of MCF-7 cells induced by leptin were tested to demonstrate the antineoplastic activity of celastrol. Transcriptomic analysis and western blotting were conducted to explore the biological roles of leptin in treating breast cancer with celastrol. The present findings showed that celastrol remarkably reversed leptin-triggered cell proliferation and migration in MCF-7 cells. Fifty-two mRNAs with fivefold higher counts and 149 mRNAs with fivefold lower counts were identified in the celastrol-treated MCF-7 cells. According to the GO and KEGG analyses, the effects of celastrol on MCF-7 cells forced lipid metabolism and the endocrine system. Moreover, leptin treatment induced phosphorylation of leptin receptor and PI3K/AKT in MCF-7 cells, whereas pretreatment with celastrol partly abrogated leptin activation. The binding of celastrol to the leptin receptor was also confirmed by molecular docking. The antitumor effect of celastrol is proposed to be mediated by its binding to the leptin receptor and controlled downregulation of the PI3K/AKT pathway.
Collapse
|
59
|
Shastri AA, Lombardo J, Okere SC, Higgins S, Smith BC, DeAngelis T, Palagani A, Hines K, Monti DA, Volpe S, Mitchell EP, Simone NL. Personalized Nutrition as a Key Contributor to Improving Radiation Response in Breast Cancer. Int J Mol Sci 2021; 23:175. [PMID: 35008602 PMCID: PMC8745527 DOI: 10.3390/ijms23010175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding metabolic and immune regulation inherent to patient populations is key to improving the radiation response for our patients. To date, radiation therapy regimens are prescribed based on tumor type and stage. Patient populations who are noted to have a poor response to radiation such as those of African American descent, those who have obesity or metabolic syndrome, or senior adult oncology patients, should be considered for concurrent therapies with radiation that will improve response. Here, we explore these populations of breast cancer patients, who frequently display radiation resistance and increased mortality rates, and identify the molecular underpinnings that are, in part, responsible for the radiation response and that result in an immune-suppressive tumor microenvironment. The resulting immune phenotype is discussed to understand how antitumor immunity could be improved. Correcting nutrient deficiencies observed in these populations should be considered as a means to improve the therapeutic index of radiation therapy.
Collapse
Affiliation(s)
- Anuradha A. Shastri
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Joseph Lombardo
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Samantha C. Okere
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Stephanie Higgins
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Brittany C. Smith
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Tiziana DeAngelis
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Ajay Palagani
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Kamryn Hines
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| | - Daniel A. Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Stella Volpe
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA;
| | - Edith P. Mitchell
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Nicole L. Simone
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; (A.A.S.); (J.L.); (S.C.O.); (S.H.); (B.C.S.); (T.D.); (A.P.); (K.H.)
| |
Collapse
|
60
|
Holm JB, Rosendahl AH, Borgquist S. Local Biomarkers Involved in the Interplay between Obesity and Breast Cancer. Cancers (Basel) 2021; 13:cancers13246286. [PMID: 34944905 PMCID: PMC8699696 DOI: 10.3390/cancers13246286] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Breast cancer is the second most common cancer in women worldwide. The risk of developing breast cancer depends on various mechanisms, such as age, heredity, reproductive factors, physical inactivity, and obesity. Obesity increases the risk of breast cancer and worsens outcomes for breast cancer patients. The rate of obesity is increasing worldwide, stressing the need for awareness of the association between obesity and breast cancer. In this review, we outline the biomarkers—including cellular and soluble factors—in the breast, associated with obesity, that affect the risk of breast cancer and breast cancer prognosis. Through these biomarkers, we aim to better identify patients with obesity with a higher risk of breast cancer and an inferior prognosis. Abstract Obesity is associated with an increased risk of breast cancer, which is the most common cancer in women worldwide (excluding non-melanoma skin cancer). Furthermore, breast cancer patients with obesity have an impaired prognosis. Adipose tissue is abundant in the breast. Therefore, breast cancer develops in an adipose-rich environment. During obesity, changes in the local environment in the breast occur which are associated with breast cancer. A shift towards a pro-inflammatory state is seen, resulting in altered levels of cytokines and immune cells. Levels of adipokines, such as leptin, adiponectin, and resistin, are changed. Aromatase activity rises, resulting in higher levels of potent estrogen in the breast. Lastly, remodeling of the extracellular matrix takes place. In this review, we address the current knowledge on the changes in the breast adipose tissue in obesity associated with breast cancer initiation and progression. We aim to identify obesity-associated biomarkers in the breast involved in the interplay between obesity and breast cancer. Hereby, we can improve identification of women with obesity with an increased risk of breast cancer and an impaired prognosis. Studies investigating mammary adipocytes and breast adipose tissue in women with obesity versus women without obesity are, however, sparse and further research is needed.
Collapse
Affiliation(s)
- Jonas Busk Holm
- Department of Oncology, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
- Correspondence: (J.B.H.); (S.B.)
| | - Ann H. Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Barngatan 4, 221 85 Lund, Sweden;
| | - Signe Borgquist
- Department of Oncology, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
- Department of Clinical Sciences Lund, Oncology, Lund University, Skåne University Hospital, Barngatan 4, 221 85 Lund, Sweden;
- Correspondence: (J.B.H.); (S.B.)
| |
Collapse
|
61
|
Obi N, Jung AY, Maurer T, Huebner M, Johnson T, Behrens S, Jaskulski S, Becher H, Chang-Claude J. Association of circulating leptin, adiponectin, and resistin concentrations with long-term breast cancer prognosis in a German patient cohort. Sci Rep 2021; 11:23526. [PMID: 34876619 PMCID: PMC8651788 DOI: 10.1038/s41598-021-02958-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/19/2021] [Indexed: 12/23/2022] Open
Abstract
Adipokines including leptin, adiponectin and resistin have been linked to risk of obesity-related cancers potentially through low-grade chronic inflammation pathways. We aimed to assess the role of post-diagnosis circulating adipokines on long-term prognosis in a prospective breast cancer cohort. Adipokines were measured in blood collected at baseline shortly after diagnosis (2002-2005) and at follow-up (2009) from 3112 breast cancer patients enrolled in the population-based MARIE study. Half of the patients had measurements at both time-points. All-cause mortality, breast cancer specific mortality and recurrences were ascertained up to June 2015 (11 years median follow-up). Associations with time-varying adipokine concentrations overall and stratified by estrogen and progesterone receptor (ERPR) were evaluated using adjusted proportional hazard regression. At baseline (n = 2700) and follow-up (n = 2027), median concentrations for leptin, adiponectin and resistin were 4.6 and 2.7 ng/ml, 24.4 and 30.0 mg/l, 15.4 and 26.2 ng/ml, respectively. After adjustment, there was no evidence for associations between adipokines and any outcome overall. In ERPR negative tumors, highest vs. lowest quintile of adiponectin was significantly associated with increased breast cancer specific mortality (HR 2.51, 95%CI 1.07-5.92). Overall, post-diagnosis adipokines were not associated with long-term outcomes after breast cancer. In patients with ERPR negative tumors, higher concentrations of adiponectin may be associated with increased breast cancer specific mortality and warrant further investigation.
Collapse
Affiliation(s)
- Nadia Obi
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Audrey Y Jung
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Tabea Maurer
- Cancer Epidemiology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marianne Huebner
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Statistics and Probability, Michigan State University, East Lansing, MI, 48824, USA
| | - Theron Johnson
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Sabine Behrens
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
| | - Stefanie Jaskulski
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Heiko Becher
- Institute for Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology/Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- Cancer Epidemiology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
62
|
Leptin and Beyond: Actors in Cancer. Biomolecules 2021; 11:biom11121836. [PMID: 34944480 PMCID: PMC8699167 DOI: 10.3390/biom11121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
|
63
|
Renna ME. A review and novel theoretical model of how negative emotions influence inflammation: The critical role of emotion regulation. Brain Behav Immun Health 2021; 18:100397. [PMID: 34927103 PMCID: PMC8649080 DOI: 10.1016/j.bbih.2021.100397] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/02/2022] Open
Abstract
Psychological distress is an inevitable part of life. Research drawing on theories from clinical psychology, health psychology, and psychoneuroimmunology (PNI) has identified relationships between negative emotions such as anxiety and sadness with inflammation. When not regulated properly, negative emotions can create biological wear and tear on the body that can increase risk for morbidity and mortality. This review discusses previously available research on relationships between negative emotions and emotion regulation with inflammation among both physically healthy adults and those with chronic illnesses. I then present a novel comprehensive biobehavioral model of negative emotionality. This model emphasizes the influence of negative emotions and their contribution to heightened inflammation. Further, I also discuss how emotion regulation (including perseverative processes such as worry and rumination) mediates this association. The relationships between negative emotionality and emotion regulation may be bidirectional, and empirical investigation of this model should specifically seek to disentangle these relationships. The proposed model offers the opportunity to advance PNI research through understanding how emotional factors alter inflammation and contribute to accelerated biological aging and disease risk.
Collapse
Affiliation(s)
- Megan E. Renna
- School of Psychology, University of Southern Mississippi, USA
| |
Collapse
|
64
|
Renna ME, Shrout MR, Madison AA, Jaremka LM, Alfano CM, Povoski SP, Agnese DM, Carson WE, Kiecolt-Glaser JK. Fluctuations in depression and anxiety predict dysregulated leptin among obese breast cancer survivors. J Cancer Surviv 2021; 15:847-854. [PMID: 33409856 DOI: 10.1007/s11764-020-00977-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/27/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Leptin influences inflammation and tumor growth and leptin signaling is often dysregulated among obese breast cancer survivors. This leads to a lack of satiety and, ultimately, risk for further weight gain. Breast cancer survivors also experience high rates of depression and anxiety, which are linked to leptin production. This study examined how a woman's anxiety and depressive symptoms, in combination with their obesity status, were associated with leptin. METHODS Breast cancer survivors (n = 200, stages 0-IIIa) completed a baseline visit before treatment and two follow-up visits, 6 and 18 months after treatment ended. Women completed anxiety and depression measures, and blood samples provided leptin data at each visit. This study related fluctuations in a survivor's own depression and anxiety (i.e., within-person effects), as well as average effects of depression and anxiety (i.e., between-person effects) to changes in leptin depending on BMI. RESULTS Obese survivors' leptin was significantly higher at visits when they had higher anxiety and depression symptoms than their own average level of symptoms. In contrast, within-person fluctuations in depression and anxiety were not related to leptin levels among non-obese survivors. No significant between-person effects of depression or anxiety on leptin emerged. CONCLUSIONS Leptin is a critical risk factor for recurrence and further health consequences. Our findings highlight how psychological health influences leptin production among breast cancer survivors. IMPLICATIONS FOR CANCER SURVIVORS These results highlight a biological pathway that may facilitate further weight gain and health risks among distressed, obese breast cancer survivors.
Collapse
Affiliation(s)
- Megan E Renna
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
- Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH, USA.
| | - M Rosie Shrout
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Annelise A Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Lisa M Jaremka
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Catherine M Alfano
- Northwell Health Cancer Institute & Center for Personalized Health, New York, NY, USA
| | - Stephen P Povoski
- Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Doreen M Agnese
- Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH, USA
- Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Janice K Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
65
|
Brena D, Huang MB, Bond V. Extracellular vesicle-mediated transport: Reprogramming a tumor microenvironment conducive with breast cancer progression and metastasis. Transl Oncol 2021; 15:101286. [PMID: 34839106 PMCID: PMC8636863 DOI: 10.1016/j.tranon.2021.101286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles’ (EVs) role in breast tumor microenvironment and pre-metastatic niche development. Breast cancer EV-mediated transmission of pro-metastatic and drug-resistant phenotypes. Precision medicine with EVs as biomarkers and delivery vehicles for drug and anticancer genetic material.
Breast cancer metastatic progression to critical secondary sites is the second leading cause of cancer-related mortality in women. While existing therapies are highly effective in combating primary tumors, metastatic disease is generally deemed incurable with a median survival of only 2, 3 years. Extensive efforts have focused on identifying metastatic contributory targets for therapeutic antagonism and prevention to improve patient survivability. Excessive breast cancer release of extracellular vesicles (EVs), whose contents stimulate a metastatic phenotype, represents a promising target. Complex breast cancer intercellular communication networks are based on EV transport and transference of molecular information is in bulk resulting in complete reprogramming events within recipient cells. Other breast cancer cells can acquire aggressive phenotypes, endothelial cells can be induced to undergo tubule formation, and immune cells can be neutralized. Recent advancements continue to implicate the critical role EVs play in cultivating a tumor microenvironment tailored to cancer proliferation, metastasis, immune evasion, and conference of drug resistance. This literature review serves to frame the role of EV transport in breast cancer progression and metastasis. The following five sections will be addressed: (1) Intercellular communication in developing a tumor microenvironment & pre-metastatic niche. (2) Induction of the epithelial-to-mesenchymal transition (EMT). (3). Immune suppression & evasion. (4) Transmission of drug resistance mechanisms. (5) Precision medicine: clinical applications of EVs.
Collapse
Affiliation(s)
- Dara Brena
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | - Vincent Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| |
Collapse
|
66
|
Leptin Protein Expression and Promoter Methylation in Ovarian Cancer: A Strong Prognostic Value with Theranostic Promises. Int J Mol Sci 2021; 22:ijms222312872. [PMID: 34884678 PMCID: PMC8657586 DOI: 10.3390/ijms222312872] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer (OC) is the deadliest among all gynecological cancers. Epidemiological studies showed that obesity might influence many cancers including OC. One of the key factors that may link obesity and OC is leptin (LEP), known as an adipokine with pleiotropic effects on body homeostasis. This study aims to investigate the expression pattern of LEP, assess the methylation profiles of LEP and their associations with clinicopathological features including survival outcomes of OC patients. The protein expression of LEP was evaluated in 208 samples using both tissue microarray and immunohistochemistry techniques. The methylation profiles of LEP were measured in 63 formalin-fixed, paraffin-embedded tumor tissues by quantitative polymerase chain reaction using a MethyLight assay. Our results showed a significant association of LEP protein overexpression with several clinicopathological variables, mainly tumor subtype, LVI, age of menarche, tumor size and stage (p < 0.04). Kaplan-Meier analysis (using low expression versus high expression as a discriminator) indicated that LEP protein overexpression is a powerful positive prognosticator of both OC recurrence (DFS) and disease-specific survival (DSS) in our OC cohort (log-rank p = 0.01 and p = 0.002, respectively). This implies that patients with high LEP expression profiles live longer with less recurrence rates. Methylation analysis results demonstrated a clear association between no/low LEP protein expression pattern (38%) and LEP promoter CpG island hypermethylation (43%). Results of this study suggest that LEP is a powerful prognosticator of OC recurrence and DSS. LEP expression in OC seems to be regulated by its promoter hypermethylation through gene partial/total silencing. Further multi-institutional studies using larger cohorts are required to demystify the intricate molecular functions of this leptin-driven effects in OC pathophysiology and to accurately assess its theranostic potential and validate its prognostic/predictive power in OC onset, progression towards more effective and personalized management of OC patients.
Collapse
|
67
|
Choromańska B, Myśliwiec P, Dadan J, Maleckas A, Zalewska A, Maciejczyk M. Effects of age and gender on the redox homeostasis of morbidly obese people. Free Radic Biol Med 2021; 175:108-120. [PMID: 34390781 DOI: 10.1016/j.freeradbiomed.2021.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/04/2021] [Accepted: 08/09/2021] [Indexed: 01/28/2023]
Abstract
Obesity is a chronic disease of complex etiology. Recent evidence suggests that obesity is caused by inflammation of adipose tissue leading to metabolic disorders, cardiovascular disease and cancer. This is the first study to evaluated the effects of age and gender on redox homeostasis, glutathione metabolism, and oxidative damage to plasma/serum lipids and proteins in morbidly obese patients. The study included 120 (60 men and 60 women) morbidly obese patients with class 3 obesity (BMI > 40 kg/m2), classified into three groups depending on age: 20-39 years (n = 20), 40-59 years (n = 20) and 60 years or older (n = 20). The number of patients was calculated a priori based on our previous experiment. We observed a reduction in serum activity of antioxidant enzymes (↓SOD) and plasma concentration of non-enzymatic antioxidants (↓GSH) in obese patients compared to the lean controls, which further decreased with age. Redox status (↑TAC, ↑TOS and ↓OSI) in morbidly obese men and women was shifted towards oxidation. Moreover, lipid (↑MDA and ↑LOOH) and protein (↑AOPP, ↑AGE and ↑Amadori products) damage products of oxidation and nitrosylation/nitration (↑total NO, ↑S-nitrosothiols, ↑peroxynitrite and ↑nitrotyrosine) were elevated in both genders of morbidly obese patients and were higher in the elderly. Interestingly, the concentrations of oxidative and nitrosative stress markers were generally higher in obese men compared to obese women at the same age. Summarizing, we showed that the total antioxidant/oxidant potential of obese patients is significantly increased and shifted towards oxidation. Obese patients have increased lipid and protein oxidation, glycation and nitration as compared to the lean controls. Disturbances in redox homeostasis increase with age in obese patients. Oxidative and nitrosative stress are more intense in men than in women at the same age.
Collapse
Affiliation(s)
- Barbara Choromańska
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276, Bialystok, Poland.
| | - Piotr Myśliwiec
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276, Bialystok, Poland.
| | - Jacek Dadan
- 1st Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276, Bialystok, Poland.
| | - Almantas Maleckas
- Department of Surgery, Lithuanian University of Health Sciences, Kaunas, Lithuania; Department of Gastrosurgical Research and Education, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Anna Zalewska
- Experimental Dentistry Laboratory, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274, Bialystok, Poland.
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 2c Mickiewicza Street, 15-233, Bialystok, Poland.
| |
Collapse
|
68
|
Takada K, Kashiwagi S, Asano Y, Goto W, Ishihara S, Morisaki T, Shibutani M, Tanaka H, Hirakawa K, Ohira M. Clinical verification of body mass index and tumor immune response in patients with breast cancer receiving preoperative chemotherapy. BMC Cancer 2021; 21:1129. [PMID: 34670511 PMCID: PMC8529767 DOI: 10.1186/s12885-021-08857-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/08/2021] [Indexed: 01/02/2023] Open
Abstract
Purpose The body mass index (BMI) is commonly used as a simple indicator of obesity; patients with early-stage breast cancer who are obese (OB) per BMI measurements have been shown to have high postoperative recurrence and low survival rates. On the other hand, it has been shown that lymphocytes present in the vicinity of malignant growths that are involved in the tumors’ immune responses influence the efficacy chemotherapy. Therefore, we hypothesized that OB patients with breast cancer have a lower density of tumor-infiltrating lymphocytes (TILs), which may influence the therapeutic effect of preoperative chemotherapy (POC). In this study, we measured pretreatment BMI and TILs in patients with breast cancer who underwent POC, examined the correlations between these two factors, and retrospectively analyzed their therapeutic outcomes and prognoses. Methods The participants in this study were 421 patients with breast cancer who underwent surgical treatment after POC between February 2007 and January 2019. The patient’s height and weight were measured before POC to calculate the BMI (weight [kg] divided by the square of the height [m2]). According to the World Health Organization categorization, patients who weighed under 18.5 kg/m2 were classified as underweight (UW), those ≥18.5 kg/m2 and > 25 kg/m2 were considered normal weight (NW), those ≥25 kg/m2 and < 30 kg/m2 were overweight (OW), and those ≥30 kg/m2 were OB. The TILs were those lymphocytes that infiltrated the tumor stroma according to the definition of the International TILs Working Group 2014. Results The median BMI was 21.9 kg/m2 (range, 14.3–38.5 kg/m2); most patients (244; 64.5%) were NW. Among all 378 patients with breast cancer, the TIL density was significantly lower in OB than in NW and OW patients (vs. NW: p = 0.001; vs. OW: p = 0.003). Furthermore, when examining patients with each breast cancer type individually, the OS of those with TNBC who had low BMIs was significantly poorer than that of their high-BMI counterparts (log rank p = 0.031). Conclusions Our data did not support the hypothesis that obesity affects the tumor immune microenvironment; however, we showed that being UW does affect the tumor immune microenvironment. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08857-7.
Collapse
Affiliation(s)
- Koji Takada
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Shinichiro Kashiwagi
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| | - Yuka Asano
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Wataru Goto
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Sae Ishihara
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Tamami Morisaki
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masatsune Shibutani
- Department of Gastrointestinal Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Hiroaki Tanaka
- Department of Gastrointestinal Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Kosei Hirakawa
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.,Department of Gastrointestinal Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masaichi Ohira
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.,Department of Gastrointestinal Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| |
Collapse
|
69
|
Hariharan H, Kesavan Y, Raja NS. Impact of native and external factors on exosome release: understanding reactive exosome secretion and its biogenesis. Mol Biol Rep 2021; 48:7559-7573. [PMID: 34626311 DOI: 10.1007/s11033-021-06733-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/29/2021] [Indexed: 02/04/2023]
Abstract
Exosomes are minuscule vesicles secreted in the endolytic region of most mammalian cells. The release of exosomes from the cell engenders cell-to-cell signaling between cellular-compartments. The trading of exosomes between tumor and yonder cells plays a hypercritical role in tumor growth and progression. The exosome released from each tumor cell sequestrates a unique biogenetic pathway reflecting its cellular origin depending on the tumor type. However, treatment of tumor cells with certain physiological factors like drugs, chemotherapy, radiation, etc., enhance the release of exosomes and alters its biogenetic pathway compared with untreated tumor cells. In this review, we will discuss how the non-native physiological factors influence the release of exosomes and how these reactive exosomes orchestrate a unique patterning of a cargo sorting mechanism. We will also discuss the role of reactively secreted exosomes in mediating tumor metastasis, angiogenesis, and tumor progression.
Collapse
Affiliation(s)
- Harini Hariharan
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India
| | - Yasodha Kesavan
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India
| | - Natesan Sella Raja
- MPI Lab, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu, India.
| |
Collapse
|
70
|
Sharifalhoseini M, Es‐haghi A, Vaezi G, Shajiee H. Biosynthesis and characterisation of solid lipid nanoparticles and investigation of toxicity against breast cancer cell line. IET Nanobiotechnol 2021; 15:654-663. [PMID: 34694719 PMCID: PMC8675850 DOI: 10.1049/nbt2.12062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Solid lipid nanoparticles (SLNs) comprise non-toxic surface-active lipidic agents combined with appropriate ratios of drugs or essential oils. The goal of this research was to investigate the effects of the SLN synthesised using essential oils of Foeniculum vulgare on the MCF-7 breast cancer cell line. SLNs were prepared by homogenisation and ultrasound techniques and characterised by dynamic light scattering (DLS), zeta potential assessment, and transmission electron microscopy (TEM). 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay (MTT assay), flow-cytometry, and Acridine-Orange assay were employed for assessing the biological activities of the SLNs. The average particle size was 55.43 nm and the net surface charge was -29.54 ± 11.67 mV. TEM showed that the mean particle size was 33.55 nm and the synthesised SLNs had a uniform round morphology. The MTT assay showed that the prepared SLNs had high toxicity against MCF-7 cells and low toxicity against normal HUVECs cells. Flow-cytometry revealed a noteworthy rise in the subG1 peak of the cell cycle in the cancer cells treated with SLNs compared to the controls, indicating apoptosis in cancer cells. The results also showed discolouration in SLNs-treated cells, which further confirmed the induction of apoptosis and the toxicity of the SLNs against MCF-7 cells.
Collapse
Affiliation(s)
| | - Ali Es‐haghi
- Department of BiologyMashhad BranchIslamic Azad UniversityMashhadIran
| | | | - Hooman Shajiee
- Department of BiologyDamghan BranchIslamic Azad UniversityDamghanIran
| |
Collapse
|
71
|
Danforth DN. The Role of Chronic Inflammation in the Development of Breast Cancer. Cancers (Basel) 2021; 13:3918. [PMID: 34359821 PMCID: PMC8345713 DOI: 10.3390/cancers13153918] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammation contributes to the malignant transformation of several malignancies and is an important component of breast cancer. The role of chronic inflammation in the initiation and development of breast cancer from normal breast tissue, however, is unclear and needs to be clarified. A review of the literature was conducted to define the chronic inflammatory processes in normal breast tissue at risk for breast cancer and in breast cancer, including the role of lymphocyte and macrophage infiltrates, chronic active adipocytes and fibroblasts, and processes that may promote chronic inflammation including the microbiome and factors related to genomic abnormalities and cellular injury. The findings indicate that in healthy normal breast tissue there is systemic evidence to suggest inflammatory changes are present and associated with breast cancer risk, and adipocytes and crown-like structures in normal breast tissue may be associated with chronic inflammatory changes. The microbiome, genomic abnormalities, and cellular changes are present in healthy normal breast tissue, with the potential to elicit inflammatory changes, while infiltrating lymphocytes are uncommon in these tissues. Chronic inflammatory changes occur prominently in breast cancer tissues, with important contributions from tumor-infiltrating lymphocytes and tumor-associated macrophages, cancer-associated adipocytes and crown-like structures, and cancer-associated fibroblasts, while the microbiome and DNA damage may serve to promote inflammatory events. Together, these findings suggest that chronic inflammation may play a role in influencing the initiation, development and conduct of breast cancer, although several chronic inflammatory processes in breast tissue may occur later in breast carcinogenesis.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
72
|
Iterative principal component analysis method for improvised classification of breast cancer disease using blood sample analysis. Med Biol Eng Comput 2021; 59:1973-1989. [PMID: 34331636 DOI: 10.1007/s11517-021-02405-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/01/2021] [Indexed: 10/20/2022]
Abstract
Breast cancer is the most common cancer in women occurring worldwide. Some of the procedures used to diagnose breast cancer are mammogram, breast ultrasound, biopsy, breast magnetic resonance imaging, and blood tests such as complete blood count. Detecting breast cancer at an early stage plays an important role in diagnostic and curative procedures. This paper aims to develop a predictive model for detecting the breast cancer using blood samples data containing age, body mass index (BMI), glucose, insulin, homeostasis model assessment (HOMA), leptin, adiponectin, resistin, and chemokine monocyte chemoattractant protein 1 (MCP-1).The two main challenges encountered in this process are identification of biomarkers and the precision of disease prediction accuracy. The proposed methodology employs principal component analysis in a peculiar approach followed by random forest tree prediction model to discriminate between healthy and breast cancer patients. This approach extracts high communalities, a linear combination of input attributes in a systematic procedure as principal axis elements. The iteratively extracted principal axis elements combined with minimum number of input attributes are able to predict the disease with higher accuracy of classification with increased sensitivity and specificity score. The results proved that the proposed approach generates a higher predictor performance than the previous reported results by opting relevant extracted principal axis elements and attributes that commend the classifier with increased performance measures.
Collapse
|
73
|
Leptin-Activity Modulators and Their Potential Pharmaceutical Applications. Biomolecules 2021; 11:biom11071045. [PMID: 34356668 PMCID: PMC8301849 DOI: 10.3390/biom11071045] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Leptin, a multifunctional hormone primarily, but not exclusively, secreted in adipose tissue, is implicated in a wide range of biological functions that control different processes, such as the regulation of body weight and energy expenditure, reproductive function, immune response, and bone metabolism. In addition, leptin can exert angiogenic and mitogenic actions in peripheral organs. Leptin biological activities are greatly related to its interaction with the leptin receptor. Both leptin excess and leptin deficiency, as well as leptin resistance, are correlated with different human pathologies, such as autoimmune diseases and cancers, making leptin and leptin receptor important drug targets. The development of leptin signaling modulators represents a promising strategy for the treatment of cancers and other leptin-related diseases. In the present manuscript, we provide an update review about leptin-activity modulators, comprising leptin mutants, peptide-based leptin modulators, as well as leptin and leptin receptor specific monoclonal antibodies and nanobodies.
Collapse
|
74
|
Houghton SC, Eliassen H, Tamimi RM, Willett WC, Rosner BA, Hankinson SE. Central Adiposity and Subsequent Risk of Breast Cancer by Menopause Status. J Natl Cancer Inst 2021; 113:900-908. [PMID: 33367714 PMCID: PMC8491796 DOI: 10.1093/jnci/djaa197] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/25/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Increased body mass index (BMI) is associated with higher postmenopausal breast cancer risk and lower premenopausal breast cancer risk. Less is known about the central adiposity-breast cancer risk association, particularly for tumor subtypes. METHODS We used prospective waist (WC) and hip circumference (HC) measures in the Nurses' Health Studies. We examined associations of WC, HC, and waist-to-hip ratio (WHR) with breast cancer independent of BMI, by menopausal status. Cox proportional hazards models estimated the hazard ratios (HRs) and 95% confidence intervals (CIs) adjusting for breast cancer risk factors, with and without BMI. RESULTS Adjusting for BMI, WC and HC were not associated, and WHR was positively associated with premenopausal breast cancer risk (WHR, quintile 5 vs 1: HRQ5vQ1, BMI-adjusted = 1.27, 95% CI = 1.04 to 1.54; Ptrend = .01), particularly for estrogen receptor-negative (ER-) and progesterone receptor-negative (PR-) and basal-like breast cancers. Premenopausal WC, HC, and WHR were not associated with postmenopausal breast cancer risk, with or without BMI adjustment. Postmenopausal WC, HC, and WHR were each positively associated with postmenopausal breast cancer (eg, WC HRQ5vsQ1 = 1.59, 95% CI = 1.36 to 1.86); after adjustment for BMI, only WC remained statistically significant (HRQ5vsQ1, BMI-adjusted = 1.38, 95% CI = 1.15 to 1.64; Ptrend = .002). In postmenopausal women, associations were stronger among never-users of hormone therapy and for ER+/PR+ breast cancers. CONCLUSIONS Central adiposity was positively associated with pre- and postmenopausal breast cancers independent of BMI. This suggests that mechanisms other than estrogen may also play a role in the relationship between central adiposity and breast cancer. Maintaining a healthy waist circumference may decrease pre- and postmenopausal breast cancer risk.
Collapse
Affiliation(s)
- Serena C Houghton
- Department of Biostatistics and Epidemiology,
University of Massachusetts Amherst, Amherst, MA, USA
| | - Heather Eliassen
- Channing Division of Network Medicine, Brigham and
Women’s Hospital and Harvard Medical School, Boston, MA,
USA
- Department of Epidemiology, Harvard T.H. Chan School
of Public Health, Boston, MA, USA
| | - Rulla M Tamimi
- Department of Epidemiology, Harvard T.H. Chan School
of Public Health, Boston, MA, USA
- Department of Population Health Sciences, Weill
Cornell Medicine, New York, NY, USA
| | - Walter C Willett
- Channing Division of Network Medicine, Brigham and
Women’s Hospital and Harvard Medical School, Boston, MA,
USA
- Department of Epidemiology, Harvard T.H. Chan School
of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of
Public Health, Boston, MA, USA
| | - Bernard A Rosner
- Channing Division of Network Medicine, Brigham and
Women’s Hospital and Harvard Medical School, Boston, MA,
USA
- Department of Biostatistics, Harvard T.H. Chan
School of Public Health, Boston, MA, USA
| | - Susan E Hankinson
- Department of Biostatistics and Epidemiology,
University of Massachusetts Amherst, Amherst, MA, USA
| |
Collapse
|
75
|
Machado VMQ, Justa RMDE, Lopes da Costa S, Barbosa MC, Damasceno NRT, Verde SMML. Bioelectrical impedance vector applied to body composition evaluation of women survivors of breast cancer: A longitudinal study. Clin Nutr ESPEN 2021; 44:247-253. [PMID: 34330474 DOI: 10.1016/j.clnesp.2021.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/05/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS Thus, the aim of this study was to assess the changes in the body composition of the women survivors of breast cancer and its association with clinical staging, using the bioelectrical impedance vector analysis (BIVA), over five years of follow-up. METHODS At baseline time 114 patients (T0) were selected and after 5 years (T1) of follow-up (n = 35) data of weight, body mass index, waist circumference, phase angle, resistance/height, reactance/height, %fat free mass and %fat mass were monitored. Bioelectrical impedance vector analysis (BIVA) and the vector displacement assessment took place according to the initial and advanced clinical staging (CS 1 and 2, respectively) and time of follow-up and have been showed using mean graph and RXc score graph. RESULTS Patients on CS 1 showed a reduction in reactance/height, phase angle, %fat free mass and an increase in %fat mass. The same group presented during the follow-up significant vector displacement by mean graph and a change to the 95% ellipse by the RXc score graph. CONCLUSIONS Women with breast cancer in the initial CS showed a more significant displacement of bioelectric vectors, indicating worsening in body composition.
Collapse
Affiliation(s)
| | | | - Sâmia Lopes da Costa
- Postgraduate Program in Nutrition and Health, State University of Ceara, Fortaleza, Ceara, Brazil.
| | - Manuela Cunha Barbosa
- Postgraduate Program in Nutrition and Health, State University of Ceara, Fortaleza, Ceara, Brazil.
| | | | | |
Collapse
|
76
|
Huang J, Yang M, Liu Z, Li X, Wang J, Fu N, Cao T, Yang X. PPFIA4 Promotes Colon Cancer Cell Proliferation and Migration by Enhancing Tumor Glycolysis. Front Oncol 2021; 11:653200. [PMID: 34094943 PMCID: PMC8173052 DOI: 10.3389/fonc.2021.653200] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/27/2021] [Indexed: 12/22/2022] Open
Abstract
Dysregulated glycolysis is one of the mechanisms employed by cancer cells to facilitate growth and metastasis. Here we aimed to characterize the PPFIA4 gene, as a glycolysis-related oncogene in promoting the proliferation and migration of colon cancer cells. Using bioinformatical tools including The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA), we found that PPFIA4 expression and methylation levels were higher in colon cancer tissues of different stages than in normal tissues. Higher PPFIA4 level was also positively correlated with poorer survival of patients. PPFIA4 upregulation also correlated with poor prognosis and higher clinical stages of colon cancer patients. Colon cancer cell viability, migration and migration were enhanced after PPFIA4 overexpression. EMT markers and glycolysis were upregulated after PPFIA4 overexpression. PPFIA4 expression was found to be positively correlated with PFKFB3 and ENO2 levels, while knockdown of PFKFB3 and ENO2 reduced cell proliferation, migration, invasion and glycolysis. PPFIA4 upregulation is a potential biomarker in colon cancer which promotes proliferation, migration, invasion and glycolysis. The upregulation of PFKFB3/ENO2 signaling by PPFIA4 is a potential mechanism underlying the oncogenic effects of PPFIA4.
Collapse
Affiliation(s)
- Jia Huang
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Meiling Yang
- Department of Oncology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Zhaoxia Liu
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaoqian Li
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Junfeng Wang
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Nian Fu
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Ting Cao
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuefeng Yang
- Department of Gastroenterology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
77
|
Lv W, Ding B, Qian L, Wu W, Wen Y. Safety of Breast Cancer Mastoscopic Surgery from the Perspective of Immunity and Adipokines. J INVEST SURG 2021; 35:632-638. [PMID: 33998356 DOI: 10.1080/08941939.2021.1919945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: This study was performed to explore the safety of breast cancer (BC) mastoscopic surgery from the perspective of immunity and adipokines. Method: A single-center, prospective, randomized controlled trial was carried out among 42 patients who had undergone surgery from December 2018 to July 2019. All patients were randomly divided into an open surgery group (n = 21) and a mastoscopic surgery group (n = 21). Flow cytometry was used to detect natural killer (NK), CD4+ T cells, CD8+ T cells, and regulatory T (Treg) cells in each group 1 d before surgery, 1 h after operation, and 1, 5, and 7 d after operation. The levels of serum leptin and adiponectin were detected by enzyme-linked immunosorbent assay before and after operation. Results: There were no significant differences in the percentages of NK (p = 0.984), CD4+ T (p = 0.591), Treg (p = 0.676), and CD8 + T (p = 0.341) lymphocytes between the two groups during the perioperative period. There were no significant differences in the levels of serum leptin and adiponectin before and after operation between the two groups (all p > 0.05). There were no significant differences between patients undergoing open surgery and mastoscopic surgery from the perspective of immunity and adipokines. Conclusion: Mastoscopic surgery is a suitable surgical choice for patients with BC.
Collapse
Affiliation(s)
- Wenzhi Lv
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Boni Ding
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Liyuan Qian
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Wei Wu
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanguang Wen
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
78
|
Maliniak ML, Miller-Kleinhenz J, Cronin-Fenton DP, Lash TL, Gogineni K, Janssen EAM, McCullough LE. Crown-Like Structures in Breast Adipose Tissue: Early Evidence and Current Issues in Breast Cancer. Cancers (Basel) 2021; 13:2222. [PMID: 34066392 PMCID: PMC8124644 DOI: 10.3390/cancers13092222] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/29/2022] Open
Abstract
Obesity is an established risk factor for postmenopausal breast cancer and has been linked to worse breast cancer prognosis, most clearly for hormone receptor-positive breast cancers. The underlying mechanisms of the obesity-breast cancer association are not fully understood, but growing evidence points to the breast adipose tissue microenvironment playing an important role. Obesity-induced adipose tissue dysfunction can result in a chronic state of low-grade inflammation. Crown-like structures of the breast (CLS-B) were recently identified as a histologic marker of local inflammation. In this review, we evaluate the early evidence of CLS-B in breast cancer. Data from preclinical and clinical studies show that these inflammatory lesions within the breast are associated with local NF-κB activation, increased aromatase activity, and elevation of pro-inflammatory mediators (TNFα, IL-1β, IL-6, and COX-2-derived PGE2)-factors involved in multiple pathways of breast cancer development and progression. There is also substantial evidence from epidemiologic studies that CLS-B are associated with greater adiposity among breast cancer patients. However, there is insufficient evidence that CLS-B impact breast cancer risk or prognosis. Comparisons across studies of prognosis were complicated by differences in CLS-B evaluation and deficiencies in study design, which future studies should take into consideration. Breast adipose tissue inflammation provides a plausible explanation for the obesity-breast cancer association, but further study is needed to establish its role and whether markers such as CLS-B are clinically useful.
Collapse
Affiliation(s)
- Maret L. Maliniak
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (J.M.-K.); (T.L.L.); (L.E.M.)
| | - Jasmine Miller-Kleinhenz
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (J.M.-K.); (T.L.L.); (L.E.M.)
| | | | - Timothy L. Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (J.M.-K.); (T.L.L.); (L.E.M.)
- Department of Clinical Epidemiology, Aarhus University Hospital, 8200 Aarhus, Denmark;
- Glenn Family Breast Center, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA;
| | - Keerthi Gogineni
- Glenn Family Breast Center, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA;
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway;
| | - Lauren E. McCullough
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (J.M.-K.); (T.L.L.); (L.E.M.)
- Glenn Family Breast Center, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA;
| |
Collapse
|
79
|
LEP (-2548G>A LEP) and LEPR (223Gln>Arg, 109Lys>Arg) polymorphisms as breast cancer risk factors in the Polish female population. Mol Biol Rep 2021; 48:3237-3244. [PMID: 33864589 PMCID: PMC8172510 DOI: 10.1007/s11033-021-06328-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/31/2021] [Indexed: 12/02/2022]
Abstract
On a global scale, breast cancer is the most common type of cancer in women, and it is still a growing problem. Therefore, new prognostic or diagnostic markers are required that would facilitate the assessment of patients or provide more efficient therapy, respectively. In these studies, we analyzed the contribution of LEP (2548G>A) and LEPR (109 Lys>Arg and 223Gln>Arg) genes polymorphisms to the risk of breast cancer development. The study involved 209 women aged 59.6 ± 11 years diagnosed with breast cancer and 202 healthy women aged 57.8 ± 8.2 years, who were blood donors. Polymorphism were evaluated by PCR–RFLP reaction followed by the verification of part of the samples by sequencing. The results of the study confirmed obesity as a significant breast cancer development risk factor in Polish women. However, no significant association between the studied polymorphisms and breast cancer risk or severity of the neoplastic disease was found. Interestingly, it was shown that wild type 223Gln>Gln leptin receptor (LEPR) was statistically more common in women with human epidermal growth factor receptor 2 negative (HER2−) than human epidermal groth factor receptor 2 positive (HER2+) breast cancer and wild type form of 2548G>A LEP was more common in women with progesterone receptor positive (PR+) than progesterone receptor negative (PR−) breast cancer. Studied polymorphisms of the LEP and LEPR genes do not increase breast cancer risk in the population of Polish women. However, they can affect PR an HER receptors expression and thus the severity of the disease. Noteworthy, this interesting correlation is being reported for the first time and might constitute an essential contribution to the identification of molecular mechanisms of carcinogenesis.
Collapse
|
80
|
Performance of functionality measures and phase angle in women exposed to chemotherapy for early breast cancer. Clin Nutr ESPEN 2021; 42:105-116. [DOI: 10.1016/j.clnesp.2021.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/07/2021] [Accepted: 02/14/2021] [Indexed: 12/21/2022]
|
81
|
Sørensen M, Poulsen AH, Kroman N, Hvidtfeldt UA, Thacher JD, Roswall N, Brandt J, Frohn LM, Jensen SS, Levin G, Raaschou-Nielsen O. Road and railway noise and risk for breast cancer: A nationwide study covering Denmark. ENVIRONMENTAL RESEARCH 2021; 195:110739. [PMID: 33460635 DOI: 10.1016/j.envres.2021.110739] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/07/2021] [Accepted: 01/07/2021] [Indexed: 05/25/2023]
Abstract
OBJECTIVE Previous studies have suggested that transportation noise may increase risk for breast cancer, but existing literature is scarce and inconclusive. We aimed to investigate associations between road traffic and railway noise and risk for breast cancer across the entire Danish female population. METHODS For all 2.8 million residential addresses across Denmark, we modelled road and railway noise at the most and least exposed façades for the period 1990-2017. We calculated 10-year time-weighted mean noise exposure for 1.8 million women aged >35 years, of whom 66,006 developed breast cancer during follow-up from 2000 to 2017. We analysed data using Cox proportional hazards models with noise exposure included as 10-year running means and adjusted for a number of individual and area-level socioeconomic co-variates and air pollution with fine particles estimated for all addresses. RESULTS For exposures at the least exposed façade, we found that a 10 dB increase in 10-year time-weighted noise was associated with incidence rate ratios (IRRs) and 95% confidence intervals (CI) for breast cancer of 1.032 (1.019-1.046) for road noise and 1.023 (0.993-1.053) for railway noise. For exposures at the most exposed façade, the IRRs (95% CIs) were 1.012 (1.002-1.022) for road noise and 1.020 (1.001-1.039) for railway noise. Associations were strongest among women with human epidermal growth factor receptor 2 negative breast cancer. CONCLUSIONS Road traffic and railway noise were associated with higher risk for breast cancer, especially noise at the least exposed façade, which is a proxy for noise exposure during sleep.
Collapse
Affiliation(s)
- Mette Sørensen
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Roskilde, Denmark.
| | - Aslak Harbo Poulsen
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Niels Kroman
- Danish Cancer Society, Copenhagen, Denmark; Department of Breast Surgery, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Jesse Daniel Thacher
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Nina Roswall
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Jørgen Brandt
- Department of Environmental Science, Aarhus University, Roskilde, Denmark; IClimate - Aarhus University Interdisciplinary Centre for Climate Change, Roskilde, Denmark
| | - Lise Marie Frohn
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | | | - Gregor Levin
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Ole Raaschou-Nielsen
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark; Department of Environmental Science, Aarhus University, Roskilde, Denmark
| |
Collapse
|
82
|
Silva C, Andrade N, Guimarães JT, Patrício E, Martel F. The in vitro effect of the diabetes-associated markers insulin, leptin and oxidative stress on cellular characteristics promoting breast cancer progression is GLUT1-dependent. Eur J Pharmacol 2021; 898:173980. [PMID: 33647254 DOI: 10.1016/j.ejphar.2021.173980] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 01/09/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) associate with increased incidence and mortality from many cancers, including breast cancer. The mechanisms involved in this relation remain poorly understood. Our study aimed to investigate the in vitro effect of high levels of glucose, insulin, leptin, TNF-α, INF-γ and oxidative stress (induced with tert-butylhydroperoxide (TBH)), which are associated with T2DM, upon glucose uptake by breast cancer (MCF-7 and MDA-MB-231) and non-cancer (MCF-12A) cells and to correlate this effect with their effects upon cellular characteristics associated with cancer progression (cell proliferation, viability, migration, angiogenesis and apoptosis). 3H-DG uptake was markedly inhibited by a selective GLUT1 inhibitor (BAY-876) in all cell lines, proving that 3H-DG uptake is mainly GLUT1-mediated. TBH (2.5 μM), insulin (50 nM), leptin (500 ng/ml) and INF-y (100 ng/ml) stimulate GLUT1-mediated 3H-DG (1 mM) uptake by both ER-positive and triple-negative breast cancer cell lines. TBH and leptin, but not insulin and INF-γ, increase GLUT1 mRNA levels. Insulin and leptin (in both ER-positive and triple-negative breast cancer cell lines) and TBH (in the triple-negative cell line) have a proproliferative effect and leptin possesses a cytoprotective effect in both breast cancer cell lines that can contribute to cancer progression. The effects of TBH, insulin, leptin and INF-γ upon breast cancer cell proliferation and viability are GLUT1-dependent. In conclusion, T2DM-associated characteristics induce changes in GLUT1-mediated glucose uptake that can contribute to cancer progression. Moreover, we conclude that BAY-876 can be a strong candidate for development of a new effective anticancer agent against breast cancer.
Collapse
Affiliation(s)
- Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal; REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Portugal
| | - João Tiago Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal; Institute of Public Health, University of Porto, Porto, Portugal
| | - Emília Patrício
- Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal; Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal.
| |
Collapse
|
83
|
Soni S, Torvund M, Mandal CC. Molecular insights into the interplay between adiposity, breast cancer and bone metastasis. Clin Exp Metastasis 2021; 38:119-138. [PMID: 33591548 DOI: 10.1007/s10585-021-10076-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023]
Abstract
Cancer is a complex disease, with various pre-existing health ailments enhancing its pathology. In cancer, the extracellular environment contains various intrinsic physiological factors whose levels are altered with aging and pre-existing conditions. In obesity, the tumor microenvironment and metastases are enriched with factors that are both derived locally, and from other physiological compartments. Similarly, in obesity, the cancer cell environment both at the site of origin and at the secondary site i.e., metastatic niche, contains significantly more phenotypically-altered adipocytes than that of un-obese cancer patients. Indeed, obesity has been linked with cancer progression, metastasis, and therapy resistance. Adipocytes not only interact with tumor cells, but also with adjacent stromal cells at primary and metastatic sites. This review emphasizes the importance of bidirectional interactions between adipocytes and breast tumor cells in breast cancer progression and its bone metastases. This paper not only chronicles the role of various adipocyte-derived factors in tumor growth, but also describes the significance of adipocyte-derived bone metastatic factors in the development of bone metastasis of breast cancer. It provides a molecular view of the interplay between the adipocytes and tumor cells involved in breast cancer bone metastasis. However, more research is needed to determine if targeting cancer-associated adipocytes holds promise as a potential therapeutic approach for breast cancer bone metastasis treatment. Interplay between adipocytes and breast cancer cells at primary cancer site and metastatic bone microenvironment. AMSC Adipose-derived mesenchymal stem cell, CAA Cancer associated adipocytes, CAF Cancer associated fibroblast, BMSC Bone marrow derived mesenchymal stem cell, BMA Bone marrow adipocyte.
Collapse
Affiliation(s)
- Sneha Soni
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | - Meaghan Torvund
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Chandi C Mandal
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
84
|
Nigro E, Orlandella FM, Polito R, Mariniello RM, Monaco ML, Mallardo M, De Stefano AE, Iervolino PLC, Salvatore G, Daniele A. Adiponectin and leptin exert antagonizing effects on proliferation and motility of papillary thyroid cancer cell lines. J Physiol Biochem 2021; 77:237-248. [PMID: 33587254 PMCID: PMC8121733 DOI: 10.1007/s13105-021-00789-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/13/2021] [Indexed: 12/20/2022]
Abstract
Adiponectin (Acrp30) and leptin, adipokines produced and secreted mainly by the adipose tissue, are involved in human carcinogenesis. Thyroid carcinomas are frequent endocrine cancers, and several evidences suggest that they are correlated with obesity. In this study, we first analyzed the expression levels and prognostic values of Acrp30, leptin, and their receptors in thyroid cancer cells. Then, we investigated the role of Acrp30 and leptin in proliferation, migration, and invasion. We found that Acrp30 treatment alone inhibits cell proliferation and cell viability in a time and dose-dependent manner; leptin alone does not influence thyroid cancer cells (BCPAP and K1) proliferation, but the combined treatment reverts Acrp30-induced effects on cell proliferation. Additionally, through wound healing and Matrigel Matrix invasion assays, we unveiled that Acrp30 inhibits thyroid cancer cell motility, while leptin induces the opposite effect. Importantly, in the combined treatment, Acrp30 and leptin exert antagonizing effects on papillary thyroid cancer cells’ migration and invasion in both BCPAP and K1 cell lines. Highlights of these studies suggest that Acrp30 and leptin could represent therapeutic targets and biomarkers for the management of thyroid cancer.
Collapse
Affiliation(s)
- Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli,", Via G. Vivaldi 42, 81100, Caserta, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy
| | | | - Rita Polito
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli,", Via G. Vivaldi 42, 81100, Caserta, Italy.,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy
| | - Raffaela Mariarosaria Mariniello
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Dipartimento di Scienze Motorie e del Benessere, Università degli Studi di Napoli "Parthenope,", Via Medina 40, 80133, Napoli, Italy
| | - Maria Ludovica Monaco
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy
| | - Marta Mallardo
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli,", Via G. Vivaldi 42, 81100, Caserta, Italy
| | - Anna Elisa De Stefano
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Dipartimento di Scienze Motorie e del Benessere, Università degli Studi di Napoli "Parthenope,", Via Medina 40, 80133, Napoli, Italy
| | - Paola Lucia Chiara Iervolino
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy.,Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli "Federico II,", Via Pansini 5, 80131, Napoli, Italy
| | - Giuliana Salvatore
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy. .,IRCCS SDN, Via Emanuele Gianturco 113, 80143, Napoli, Italy. .,Dipartimento di Scienze Motorie e del Benessere, Università degli Studi di Napoli "Parthenope,", Via Medina 40, 80133, Napoli, Italy.
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università degli Studi della Campania, "Luigi Vanvitelli,", Via G. Vivaldi 42, 81100, Caserta, Italy. .,CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145, Napoli, Italy.
| |
Collapse
|
85
|
Gameiro A, Nascimento C, Urbano AC, Correia J, Ferreira F. Serum and Tissue Expression Levels of Leptin and Leptin Receptor Are Putative Markers of Specific Feline Mammary Carcinoma Subtypes. Front Vet Sci 2021; 8:625147. [PMID: 33644151 PMCID: PMC7902695 DOI: 10.3389/fvets.2021.625147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 01/02/2023] Open
Abstract
Obesity is an established risk factor for breast cancer in post-menopausal women, being associated with elevated serum levels of leptin. Although overweight is a common condition in cat, the role of leptin and its receptor in feline mammary carcinoma remains unsettled. In this study, serum leptin and leptin receptor (ObR) levels were investigated in 58 cats with mammary carcinoma and compared with those of healthy animals, as were the expression levels of leptin and ObR in tumor tissues. The results showed that the Free Leptin Index is significantly decreased in cats with mammary carcinoma (p = 0.0006), particularly in those with luminal B and HER2-positive tumors, and that these animals also present significantly lower serum leptin levels (p < 0.0001 and p < 0.005, respectively). Interestingly, ulcerating tumors (p = 0.0005) and shorter disease-free survival (p = 0.0217) were associated to serum leptin levels above 4.17 pg/mL. In contrast, elevated serum ObR levels were found in all cats with mammary carcinoma (p < 0.0001), with levels above 16.89 ng/mL being associated with smaller tumors (p = 0.0118), estrogen receptor negative status (p = 0.0291) and increased serum levels of CTLA-4 (p = 0.0056), TNF-α (p = 0.0025), PD-1 (p = 0.0023), and PD-L1 (p = 0.0002). In tumor samples, leptin is overexpressed in luminal B and triple-negative carcinomas (p = 0.0046), whereas ObR is found to be overexpressed in luminal B tumors (p = 0.0425). Altogether, our results support the hypothesis that serum levels of leptin and ObR can be used as biomarkers of specific feline mammary carcinoma subtypes, and suggests the use of leptin antagonists as a therapeutic tool, reinforcing the utility of the cat as a cancer model.
Collapse
Affiliation(s)
- Andreia Gameiro
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária da Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Nascimento
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária da Universidade de Lisboa, Lisbon, Portugal
| | - Ana Catarina Urbano
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária da Universidade de Lisboa, Lisbon, Portugal
| | - Jorge Correia
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária da Universidade de Lisboa, Lisbon, Portugal
| | - Fernando Ferreira
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
86
|
Muoio MG, Talia M, Lappano R, Sims AH, Vella V, Cirillo F, Manzella L, Giuliano M, Maggiolini M, Belfiore A, De Francesco EM. Activation of the S100A7/RAGE Pathway by IGF-1 Contributes to Angiogenesis in Breast Cancer. Cancers (Basel) 2021; 13:cancers13040621. [PMID: 33557316 PMCID: PMC7915817 DOI: 10.3390/cancers13040621] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Breast cancer mortality is increased in patients affected by metabolic disorders associated with dysregulation of the Insulin-like growth factor-1 (IGF-1) axis, like obesity and type-2 diabetes. Despite the oncogenic role of this complex signaling system is widely known, the clinical targeting of IGF-1 and its receptor (IGF-1R) has provided valuable benefit only on small sub-populations of cancer patients, thus suggesting that a further characterization of the biological effects of the IGF-1/IGF-1R pathway could pave the way for a better manipulation of this crucial signaling system at the clinical level. In this study, we have identified the protein S100A7 as novel molecular target of IGF-1 action in the breast tumor microenvironment, toward increased cancer-associated angiogenesis. Targeting the IGF-1/IGF-1R/S100A7 pathway may therefore represent a further useful approach for blocking disease progression in breast cancer patients with dysregulated IGF-1 signaling. Abstract Background: Breast cancer (BC) mortality is increased among obese and diabetic patients. Both obesity and diabetes are associated with dysregulation of both the IGF-1R and the RAGE (Receptor for Advanced Glycation End Products) pathways, which contribute to complications of these disorders. The alarmin S100A7, signaling through the receptor RAGE, prompts angiogenesis, inflammation, and BC progression. Methods: We performed bioinformatic analysis of BC gene expression datasets from published studies. We then used Estrogen Receptor (ER)-positive BC cells, CRISPR-mediated IGF-1R KO BC cells, and isogenic S100A7-transduced BC cells to investigate the role of IGF-1/IGF-1R in the regulation of S100A7 expression and tumor angiogenesis. To this aim, we also used gene silencing and pharmacological inhibitors, and we performed gene expression and promoter studies, western blotting analysis, ChIP and ELISA assays, endothelial cell proliferation and tube formation assay. Results: S100A7 expression correlates with worse prognostic outcomes in human BCs. In BC cells, the IGF-1/IGF-1R signaling engages STAT3 activation and its recruitment to the S100A7 promoter toward S100A7 increase. In human vascular endothelial cells, S100A7 activates RAGE signaling and prompts angiogenic effects. Conclusions: In ER-positive BCs the IGF-1 dependent activation of the S100A7/RAGE signaling in adjacent endothelial cells may serve as a previously unidentified angiocrine effector. Targeting S100A7 may pave the way for a better control of BC, particularly in conditions of unopposed activation of the IGF-1/IGF-1R axis.
Collapse
Affiliation(s)
- Maria Grazia Muoio
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Andrew H. Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh EH4 2XU, UK;
| | - Veronica Vella
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Livia Manzella
- Center of Experimental Oncology and Hematology, A.O.U. Policlinico Vittorio Emanuele, 95122 Catania, Italy;
- Department of Clinical and Experimental Medicine, University of Catania, 95122 Catania, Italy
| | - Marika Giuliano
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.T.); (R.L.); (F.C.); (M.M.)
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Correspondence: (A.B.); (E.M.D.F.); Tel.: 39-095-7598-700 (A.B.); +39-095-7598-831 (E.M.D.F.)
| | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122 Catania,
Italy; (M.G.M.); (V.V.); (M.G.)
- Correspondence: (A.B.); (E.M.D.F.); Tel.: 39-095-7598-700 (A.B.); +39-095-7598-831 (E.M.D.F.)
| |
Collapse
|
87
|
Scully T, Ettela A, LeRoith D, Gallagher EJ. Obesity, Type 2 Diabetes, and Cancer Risk. Front Oncol 2021; 10:615375. [PMID: 33604295 PMCID: PMC7884814 DOI: 10.3389/fonc.2020.615375] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and type 2 diabetes have both been associated with increased cancer risk and are becoming increasingly prevalent. Metabolic abnormalities such as insulin resistance and dyslipidemia are associated with both obesity and type 2 diabetes and have been implicated in the obesity-cancer relationship. Multiple mechanisms have been proposed to link obesity and diabetes with cancer progression, including an increase in insulin/IGF-1 signaling, lipid and glucose uptake and metabolism, alterations in the profile of cytokines, chemokines, and adipokines, as well as changes in the adipose tissue directly adjacent to the cancer sites. This review aims to summarize and provide an update on the epidemiological and mechanistic evidence linking obesity and type 2 diabetes with cancer, focusing on the roles of insulin, lipids, and adipose tissue.
Collapse
Affiliation(s)
- Tiffany Scully
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Emily Jane Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
88
|
Delort L, Cholet J, Decombat C, Vermerie M, Dumontet C, Castelli FA, Fenaille F, Auxenfans C, Rossary A, Caldefie-Chezet F. The Adipose Microenvironment Dysregulates the Mammary Myoepithelial Cells and Could Participate to the Progression of Breast Cancer. Front Cell Dev Biol 2021; 8:571948. [PMID: 33505957 PMCID: PMC7829501 DOI: 10.3389/fcell.2020.571948] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common cancer among women worldwide. Overweight and obesity are now recognized as established risk factors for this pathology in postmenopausal women. These conditions are also believed to be responsible for higher recurrence and mortality rates. Reciprocal interactions have been described between adipose and cancer cells. An adipose microenvironment favors a greater proliferation of cancer cells, their invasion and even resistance to anti-cancer treatments. In addition, the chronic low-grade inflammation observed in obese individuals is believed to amplify these processes. Among the cell types present in the breast, myoepithelial cells (MECs), located at the interface of the epithelial cells and the stroma, are considered "tumor suppressor" cells. During the transition from ductal carcinoma in situ to invasive cancer, disorganization or even the disappearance of MECs is observed, thereby enhancing the ability of the cancer cells to migrate. As the adipose microenvironment is now considered as a central actor in the progression of breast cancer, our objective was to evaluate if it could be involved in MEC functional modifications, leading to the transition of in situ to invasive carcinoma, particularly in obese patients. Through a co-culture model, we investigated the impact of human adipose stem cells from women of normal weight and obese women, differentiated or not into mature adipocytes, on the functionality of the MECs by measuring changes in viability, apoptosis, gene, and miRNA expressions. We found that adipose cells (precursors and differentiated adipocytes) could decrease the viability of the MECs, regardless of the original BMI. The adipose cells could also disrupt the expression of the genes involved in the maintenance of the extracellular matrix and to amplify the expression of leptin and inflammatory markers. miR-122-5p and miR-132-3p could also be considered as targets for adipose cells. The metabolite analyses revealed specific profiles that may be involved in the growth of neoplastic cells. All of these perturbations could thus be responsible for the loss of tumor suppressor status of MECs and promote the transition from in situ to invasive carcinoma.
Collapse
Affiliation(s)
- Laetitia Delort
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Juliette Cholet
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Caroline Decombat
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Marion Vermerie
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | - Charles Dumontet
- Université Lyon 1, INSERM U1052, CNRS 5286, Cancer Research Center of Lyon, Lyon, France
| | - Florence A Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif-sur-Yvette, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Gif-sur-Yvette, France
| | - Céline Auxenfans
- Banque de Tissus et de Cellules, Hôpital Edouard-Herriot, Lyon, France
| | - Adrien Rossary
- Université Clermont Auvergne, INRAE, UNH, ECREIN, Clermont-Ferrand, France
| | | |
Collapse
|
89
|
COX-2 promotes mammary adipose tissue inflammation, local estrogen biosynthesis, and carcinogenesis in high-sugar/fat diet treated mice. Cancer Lett 2021; 502:44-57. [PMID: 33429006 DOI: 10.1016/j.canlet.2021.01.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/22/2020] [Accepted: 01/04/2021] [Indexed: 12/26/2022]
Abstract
Obesity is a major risk factor for breast cancer, especially in post-menopausal women. In the breast tissue of obese women, cyclooxygenase-2 (COX-2)-dependent prostaglandin E2 (PGE2) production has been correlated with inflammation and local estrogen biosynthesis via aromatase. Using a mouse model of 7,12-dimethylbenz[a]anthracene/medroxyprogesterone-acetate (DMBA/MPA)-induced carcinogenesis, we demonstrated that an obesogenic diet promotes mammary tissue inflammation and local estrogen production, and accelerates mammary tumor formation in a COX-2-dependent manner. High-sugar/fat (HSF) diet augmented the levels of the pro-inflammatory mediators MCP-1, IL-6, COX-2, and PGE2 in mammary tissue, and this was accompanied by crown-like structures of breast (CLS-B) formation and aromatase/estrogen upregulation. Treatment with a COX-2 selective inhibitor, etoricoxib, decreased PGE2, IL-6, MCP-1, and CLS-B formation as well as reduced aromatase protein and estrogen levels in the mammary tissue of mice fed a HSF diet. Etoricoxib-treated mice showed increased latency and decreased incidence of mammary tumors, which resulted in prolonged animal survival when compared to HSF diet alone. Inhibition of tumor angiogenesis also seemed to account for the prolonged survival of COX-2 inhibitor-treated animals. In conclusion, obesogenic diet-induced COX-2 is sufficient to trigger inflammation, local estrogen biosynthesis, and mammary tumorigenesis.
Collapse
|
90
|
Acheva A, Kärki T, Schaible N, Krishnan R, Tojkander S. Adipokine Leptin Co-operates With Mechanosensitive Ca 2 +-Channels and Triggers Actomyosin-Mediated Motility of Breast Epithelial Cells. Front Cell Dev Biol 2021; 8:607038. [PMID: 33490070 PMCID: PMC7815691 DOI: 10.3389/fcell.2020.607038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
In postmenopausal women, a major risk factor for the development of breast cancer is obesity. In particular, the adipose tissue-derived adipokine leptin has been strongly linked to tumor cell proliferation, migration, and metastasis, but the underlying mechanisms remain unclear. Here we show that treatment of normal mammary epithelial cells with leptin induces EMT-like features characterized by higher cellular migration speeds, loss of structural ordering of 3D-mammo spheres, and enhancement of epithelial traction forces. Mechanistically, leptin triggers the phosphorylation of myosin light chain kinase-2 (MLC-2) through the interdependent activity of leptin receptor and Ca2+ channels. These data provide evidence that leptin-activated leptin receptors, in co-operation with mechanosensitive Ca2+ channels, play a role in the development of breast carcinomas through the regulation of actomyosin dynamics.
Collapse
Affiliation(s)
- Anna Acheva
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Niccole Schaible
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Beth Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Sari Tojkander
- Section of Pathology, Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
91
|
Luo Y, Li HB, Zhang Y, Wu YX, Shen D, Che YQ. Combination of Endogenous Estradiol and Adipokine Leptin in Breast Cancer Risk and Prognosis Assessment in Postmenopausal Chinese Women. Front Endocrinol (Lausanne) 2021; 12:766463. [PMID: 34970222 PMCID: PMC8712642 DOI: 10.3389/fendo.2021.766463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Our study aims to clarify the role of estradiol and leptin in breast cancer risk and prognostic assessment in postmenopausal Chinese women. DESIGN The serum circulating estradiol and leptin level was detected by ELISA. Then the correlation between estradiol, leptin level, and clinical characteristics was analyzed using Fisher's exact test. Next, the Kaplan-Meier model was used to analyze the association between estradiol, leptin, and prognosis of postmenopausal breast cancer patients in our cohort and the TCGA dataset. SETTING The study was conducted at the National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College. PATIENTS A total of 182 postmenopausal breast cancer patients and 111 healthy subjects from January 2010 to August 2010 were included in the analysis. Another 702 cases with breast cancer were retrieved from The Cancer Genome Atlas (TCGA) database for subsequent analysis. MAIN OUTCOME MEASURE Serum circulating estradiol and leptin level. RESULTS The level of estradiol was significantly higher (P<0.001) but the level of leptin had no significant difference (P = 0.764) in postmenopausal breast cancer patients compared with healthy subjects. The level of estradiol and leptin was not significantly different between estrogen receptor (ER) positive and ER-negative groups (P>0.05). Estradiol was significantly correlated with tumor T stage (P = 0.002) and leptin was significantly associated with perineural invasion (P = 0.014). In addition, the disease-free survival of patients with a high level of estradiol was significantly shorter (P = 0.025) but leptin tended to be a protective factor for overall survival in TCGA analysis (P = 0.038). CONCLUSION Circulating estradiol and leptin played important roles in the risk of postmenopausal breast cancer even in low-estrogen nations with an independent expression of ER status. High circulating estradiol was a poor prognostic factor and leptin may be a protection signal in Chinese postmenopausal patients with breast cancer.
Collapse
Affiliation(s)
- Yang Luo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han-Bing Li
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhang
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Xin Wu
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Di Shen
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Qun Che
- Center for Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yi-Qun Che,
| |
Collapse
|
92
|
Kim JH, Lim JS. Early menarche and its consequence in Korean female: reducing fructose intake could be one solution. Clin Exp Pediatr 2021; 64:12-20. [PMID: 32403898 PMCID: PMC7806406 DOI: 10.3345/cep.2019.00353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 12/27/2019] [Accepted: 02/28/2020] [Indexed: 12/31/2022] Open
Abstract
The mean age at menarche (AAM) of Korean females has been rapidly decreasing over the last 50 years; currently, the prevalence of early menarche (<12 years) is 22.3%. Female adolescents who experience early menarche are known to be at greater risk of psychosocial and behavioral problems along with several physical health problems such as menstrual problems. They also tend to achieve a shorter final height and develop obesity. Population-based Korean studies have shown a strong association between early menarche and the risk of obesity, insulin resistance, metabolic syndrome, nonalcoholic fatty liver disease, diabetes, breast cancer, and cardiovascular disease in adulthood. Although the exact mechanism of how early menarche causes cardiometabolic derangement in later adulthood is unknown, childhood obesity and insulin resistance might be major contributors. Recent studies demonstrated that an excessive consumption of fructose might underlie the development of obesity and insulin resistance along with an earlier AAM. A positive association was observed between sugar-sweetened beverages (a major source of fructose) intake and obesity, metabolic syndrome, insulin resistance, and cardiometabolic risk in Korean females. In pediatrics, establishing risk factors is important in preventing disease in later life. In this regard, early menarche is a simple and good marker for the management of cardiometabolic diseases in adulthood. Decreasing one's fructose intake might prevent early menarche as well as the development of obesity, insulin resistance, and cardiometabolic diseases.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Jung Sub Lim
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Korea
| |
Collapse
|
93
|
Ortega MA, Fraile-Martínez O, García-Montero C, Pekarek L, Guijarro LG, Castellanos AJ, Sanchez-Trujillo L, García-Honduvilla N, Álvarez-Mon M, Buján J, Zapico Á, Lahera G, Álvarez-Mon MA. Physical Activity as an Imperative Support in Breast Cancer Management. Cancers (Basel) 2020; 13:E55. [PMID: 33379177 PMCID: PMC7796347 DOI: 10.3390/cancers13010055] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy and the second cause of cancer-related death among women. It is estimated that 9 in 10 cases of BC are due to non-genetic factors, and approximately 25% to 30% of total breast cancer cases should be preventable only by lifestyle interventions. In this context, physical activity represents an excellent and accessible approach not only for the prevention, but also for being a potential support in the management of breast cancer. The present review will collect the current knowledge of physical activity in the background of breast cancer, exploring its systemic and molecular effects, considering important variables in the training of these women and the evidence regarding the benefits of exercise on breast cancer survival and prognosis. We will also summarize the various effects of physical activity as a co-adjuvant therapy in women receiving different treatments to deal with its adverse effects. Finally, we will reveal the impact of physical activity in the enhancement of quality of life of these patients, to conclude the central role that exercise must occupy in breast cancer management, in an adequate context of a healthy lifestyle.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Oscar Fraile-Martínez
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
| | - Cielo García-Montero
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
| | - Leonel Pekarek
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
| | - Luis G. Guijarro
- Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Alejandro J. Castellanos
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
| | - Lara Sanchez-Trujillo
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
| | - Natalio García-Honduvilla
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Melchor Álvarez-Mon
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Julia Buján
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain
| | - Álvaro Zapico
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
- Obstetrics and Gynecology Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, 28806 Alcalá de Henares, Spain
| | - Guillermo Lahera
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, 28806 Alcalá de Henares, Spain
| | - Miguel A. Álvarez-Mon
- Unit of Histology and Pathology, Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.G.-M.); (L.P.); (A.J.C.); (L.S.-T.); (N.G.-H.); (M.Á.-M.); (J.B.); (G.L.); (M.A.Á.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Psychiatry and Medical Psychology, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| |
Collapse
|
94
|
Augimeri G, Gelsomino L, Plastina P, Giordano C, Barone I, Catalano S, Andò S, Bonofiglio D. Natural and Synthetic PPARγ Ligands in Tumor Microenvironment: A New Potential Strategy against Breast Cancer. Int J Mol Sci 2020; 21:E9721. [PMID: 33352766 PMCID: PMC7767156 DOI: 10.3390/ijms21249721] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple lines of evidence indicate that activation of the peroxisome proliferator-activated receptor γ (PPARγ) by natural or synthetic ligands exerts tumor suppressive effects in different types of cancer, including breast carcinoma. Over the past decades a new picture of breast cancer as a complex disease consisting of neoplastic epithelial cells and surrounding stroma named the tumor microenvironment (TME) has emerged. Indeed, TME is now recognized as a pivotal element for breast cancer development and progression. Novel strategies targeting both epithelial and stromal components are under development or undergoing clinical trials. In this context, the aim of the present review is to summarize PPARγ activity in breast TME focusing on the role of this receptor on both epithelial/stromal cells and extracellular matrix components of the breast cancer microenvironment. The information provided from the in vitro and in vivo research indicates PPARγ ligands as potential agents with regards to the battle against breast cancer.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
| | - Pierluigi Plastina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (L.G.); (P.P.); (C.G.); (I.B.); (S.C.); (S.A.)
- Centro Sanitario, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| |
Collapse
|
95
|
Olea-Flores M, Juárez-Cruz JC, Zuñiga-Eulogio MD, Acosta E, García-Rodríguez E, Zacapala-Gomez AE, Mendoza-Catalán MA, Ortiz-Ortiz J, Ortuño-Pineda C, Navarro-Tito N. New Actors Driving the Epithelial-Mesenchymal Transition in Cancer: The Role of Leptin. Biomolecules 2020; 10:E1676. [PMID: 33334030 PMCID: PMC7765557 DOI: 10.3390/biom10121676] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/12/2020] [Accepted: 12/13/2020] [Indexed: 12/24/2022] Open
Abstract
Leptin is a hormone secreted mainly by adipocytes; physiologically, it participates in the control of appetite and energy expenditure. However, it has also been linked to tumor progression in different epithelial cancers. In this review, we describe the effect of leptin on epithelial-mesenchymal transition (EMT) markers in different study models, including in vitro, in vivo, and patient studies and in various types of cancer, including breast, prostate, lung, and ovarian cancer. The different studies report that leptin promotes the expression of mesenchymal markers and a decrease in epithelial markers, in addition to promoting EMT-related processes such as cell migration and invasion and poor prognosis in patients with cancer. Finally, we report that leptin has the greatest biological relevance in EMT and tumor progression in breast, lung, prostate, esophageal, and ovarian cancer. This relationship could be due to the key role played by the enriched tumor microenvironment in adipose tissue. Together, these findings demonstrate that leptin is a key biomolecule that drives EMT and metastasis in cancer.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Juan C. Juárez-Cruz
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Miriam D. Zuñiga-Eulogio
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Erika Acosta
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Eduardo García-Rodríguez
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| | - Ana E. Zacapala-Gomez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Miguel A. Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (A.E.Z.-G.); (M.A.M.-C.); (J.O.-O.)
| | - Carlos Ortuño-Pineda
- Laboratorio de Ácidos Nucleicos y Proteinas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico;
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39090, Mexico; (M.O.-F.); (J.C.J.-C.); (M.D.Z.-E.); (E.A.); (E.G.-R.)
| |
Collapse
|
96
|
Dalimi-Asl S, Babaahmadi-Rezaei H, Mohammadzadeh G. Combination of Silibinin and Curcumin Reduced Leptin Receptor Expression in MCF-7 Human Breast Cancer Cell Line. IRANIAN JOURNAL OF MEDICAL SCIENCES 2020; 45:477-484. [PMID: 33281265 PMCID: PMC7707627 DOI: 10.30476/ijms.2019.81934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Background: Leptin and leptin receptor (Ob-R) are associated with worse prognosis, distant metastasis, and poor survival of breast cancer.
We investigated the cytotoxic effect of silibinin and curcumin, individually and combined, on Ob-R expression in MCF-7 cells. Methods: This study was performed from October 2017 to April 2018 at the Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur
University of Medical Sciences, Ahvaz, Iran. The cytotoxic effect of silibinin and curcumin, individually and combined, and their corresponding
half-maximal inhibitory concentration (IC50) values were determined using the methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay.
The cells were treated with different concentrations of silibinin (50-400 μM), curcumin (10-35 μM), and their combinations for 24 and 48 hours.
The expression of Ob-R was measured using the Western blot analysis by treating the cells with different concentrations of curcumin (10-25 μM),
silibinin (50-250 μM), and their respective combinations. The difference in mean cell viability between the groups was calculated using one-way ANOVA followed by Tukey’s post hoc test. Results: Silibinin and curcumin exerted time- and dose-dependent cytotoxic effect on MCF-7 cells. After treatment with silibinin, the IC50 values
were about 250 and 50 μM at 24 and 48 hours, respectively. In terms of treatment with curcumin, the IC50 values were about 25 and 15 μM at
24 and 48 hours, respectively. Following treatment with silibinin, the Western blot analysis showed that Ob-R expression significantly
decreased at 150 μM (P=0.031) and 200 μM (P=0.023) concentrations. Curcumin did not significantly decrease the Ob-R expression, however,
the expression significantly decreased (P=0.004) when it was combined with silibinin. Conclusion: The combination of silibinin and curcumin significantly reduced Ob-R expression in MCF-7 cells compared with their individual effects.
Collapse
Affiliation(s)
- Somaye Dalimi-Asl
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ghorban Mohammadzadeh
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
97
|
Orlandella FM, De Stefano AE, Iervolino PLC, Buono P, Soricelli A, Salvatore G. Dissecting the molecular pathways involved in the effects of physical activity on breast cancers cells: A narrative review. Life Sci 2020; 265:118790. [PMID: 33220294 DOI: 10.1016/j.lfs.2020.118790] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/12/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023]
Abstract
Epidemiologic evidence suggests that obesity and sedentary are modifiable factors strongly associated with breast cancer risk worldwide. Since breast cancer represents the most frequent malignant neoplasm and the second cause of cancer-related deaths in women worldwide, an insight into the molecular mechanisms clarifying the effects of physical activity in breast cancer cells could have important implication for changing this cancer burden. In this narrative Review article, we summarize the current knowledge, regarding the effects of adapted physical activity program, focusing on the cellular signaling pathways activated and on the molecular markers involved in breast cancer. Regular exercise training in breast cancer patients has been shown to positively affect tumor-growth and survival rate. Indeed, emerging work demonstrates that regular exercise is able to affect multiple cancer hallmarks influencing the development and progression of cancer. In conclusion, changes in the circulating insulin, adipokines and estrogen levels, inflammation and oxidative stress could represent some of the possible biological mechanisms through which exercise may influence breast cancer development and recurrence.
Collapse
Affiliation(s)
| | - Anna Elisa De Stefano
- Dipartimento di Scienze Motorie e del Benessere, Università "Parthenope", Via Medina 40, 80133 Naples, Italy; CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Paola Lucia Chiara Iervolino
- CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy; Dipartimento di Scienze Biomediche Avanzate, Università "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Pasqualina Buono
- Dipartimento di Scienze Motorie e del Benessere, Università "Parthenope", Via Medina 40, 80133 Naples, Italy; CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Andrea Soricelli
- IRCCS SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; Dipartimento di Scienze Motorie e del Benessere, Università "Parthenope", Via Medina 40, 80133 Naples, Italy
| | - Giuliana Salvatore
- IRCCS SDN, Via Emanuele Gianturco 113, 80143 Naples, Italy; Dipartimento di Scienze Motorie e del Benessere, Università "Parthenope", Via Medina 40, 80133 Naples, Italy; CEINGE - Biotecnologie Avanzate S.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
| |
Collapse
|
98
|
Choromańska B, Myśliwiec P, Łuba M, Wojskowicz P, Myśliwiec H, Choromańska K, Dadan J, Żendzian-Piotrowska M, Zalewska A, Maciejczyk M. Bariatric Surgery Normalizes Protein Glycoxidation and Nitrosative Stress in Morbidly Obese Patients. Antioxidants (Basel) 2020; 9:antiox9111087. [PMID: 33158288 PMCID: PMC7694407 DOI: 10.3390/antiox9111087] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
The results of recent studies indicate the key role of nitrosative stress and protein oxidative damage in the development of morbid obesity. Nevertheless, the effect of bariatric surgery on protein oxidation/glycation and nitrosative/nitrative stress is not yet known. This is the first study evaluating protein glycoxidation and protein nitrosative damage in morbidly obese patients before and after (one, three, six and twelve months) laparoscopic sleeve gastrectomy. The study included 50 women with morbid obesity as well as 50 age- and gender-matched healthy controls. We demonstrated significant increases in serum myeloperoxidase, plasma glycooxidative products (dityrosine, kynurenine, N-formyl-kynurenine, amyloid, Amadori products, glycophore), protein oxidative damage (ischemia modified albumin) and nitrosative/nitrative stress (nitric oxide, peroxy-nitrite, S-nitrosothiols and nitro-tyrosine) in morbidly obese subjects as compared to lean controls, whereas plasma tryptophan and total thiols were statistically decreased. Bariatric surgery generally reduces the abnormalities in the glycoxidation of proteins and nitrosative/nitrative stress. Noteworthily, in the patients with metabolic syndrome (MS+), we showed no differences in most redox biomarkers, as compared to morbidly obese patients without MS (MS−). However, two markers: were able to differentiate MS+ and MS− with high specificity and sensitivity: peroxy-nitrite (>70%) and S-nitrosothiols (>60%). Further studies are required to confirm the diagnostic usefulness of such biomarkers.
Collapse
Affiliation(s)
- Barbara Choromańska
- Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276 Bialystok, Poland; (P.M.); (M.Ł.); (P.W.); (J.D.)
- Correspondence: (B.C.); (M.M.)
| | - Piotr Myśliwiec
- Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276 Bialystok, Poland; (P.M.); (M.Ł.); (P.W.); (J.D.)
| | - Magdalena Łuba
- Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276 Bialystok, Poland; (P.M.); (M.Ł.); (P.W.); (J.D.)
| | - Piotr Wojskowicz
- Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276 Bialystok, Poland; (P.M.); (M.Ł.); (P.W.); (J.D.)
| | - Hanna Myśliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 14 Żurawia Street, 15-540 Bialystok, Poland;
| | - Katarzyna Choromańska
- Department of Oral Surgery, Medical University of Gdansk, 7 Dębinki Street, 80-211 Gdansk, Poland;
| | - Jacek Dadan
- Department of General and Endocrine Surgery, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-276 Bialystok, Poland; (P.M.); (M.Ł.); (P.W.); (J.D.)
| | - Małgorzata Żendzian-Piotrowska
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 2c Mickiewicza Street, 15-233 Bialystok, Poland;
| | - Anna Zalewska
- Experimental Dentistry Laboratory, Medical University of Bialystok, 24a M. Sklodowskiej-Curie Street, 15-274 Bialystok, Poland;
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, 2c Mickiewicza Street, 15-233 Bialystok, Poland;
- Correspondence: (B.C.); (M.M.)
| |
Collapse
|
99
|
Liu C, Zhao Q, Yu X. Bone Marrow Adipocytes, Adipocytokines, and Breast Cancer Cells: Novel Implications in Bone Metastasis of Breast Cancer. Front Oncol 2020; 10:561595. [PMID: 33123472 PMCID: PMC7566900 DOI: 10.3389/fonc.2020.561595] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/27/2020] [Indexed: 02/05/2023] Open
Abstract
Accumulating discoveries highlight the importance of interaction between marrow stromal cells and cancer cells for bone metastasis. Bone is the most common metastatic site of breast cancer and bone marrow adipocytes (BMAs) are the most abundant component of the bone marrow microenvironment. BMAs are unique in their origin and location, and recently they are found to serve as an endocrine organ that secretes adipokines, cytokines, chemokines, and growth factors. It is reasonable to speculate that BMAs contribute to the modification of bone metastatic microenvironment and affecting metastatic breast cancer cells in the bone marrow. Indeed, BMAs may participate in bone metastasis of breast cancer through regulation of recruitment, invasion, survival, colonization, proliferation, angiogenesis, and immune modulation by their production of various adipocytokines. In this review, we provide an overview of research progress, focusing on adipocytokines secreted by BMAs and their potential roles for bone metastasis of breast cancer, and investigating the mechanisms mediating the interaction between BMAs and metastatic breast cancer cells. Based on current findings, BMAs may function as a pivotal modulator of bone metastasis of breast cancer, therefore targeting BMAs combined with conventional treatment programs might present a promising therapeutic option.
Collapse
Affiliation(s)
- Chang Liu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zhao
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Department of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
100
|
Fat Body p53 Regulates Systemic Insulin Signaling and Autophagy under Nutrient Stress via Drosophila Upd2 Repression. Cell Rep 2020; 33:108321. [DOI: 10.1016/j.celrep.2020.108321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/05/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
|