1
|
Mohammad Mirzaei N, Kevrekidis PG, Shahriyari L. Oxygen, angiogenesis, cancer and immune interplay in breast tumour microenvironment: a computational investigation. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240718. [PMID: 39665095 PMCID: PMC11631512 DOI: 10.1098/rsos.240718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 12/13/2024]
Abstract
Breast cancer is a challenging global health problem among women. This study investigates the intricate breast tumour microenvironment (TME) dynamics utilizing data from mammary-specific polyomavirus middle T antigen overexpression mouse models (MMTV-PyMT). It incorporates endothelial cells (ECs), oxygen and vascular endothelial growth factors (VEGF) to examine the interplay of angiogenesis, hypoxia, VEGF and immune cells in cancer progression. We introduce an approach to impute immune cell fractions within the TME using single-cell RNA-sequencing (scRNA-seq) data from MMTV-PyMT mice. We quantify our analysis by estimating cell counts using cell size data and laboratory findings from existing literature. We perform parameter estimation via a Hybrid Genetic Algorithm (HGA). Our simulations reveal various TME behaviours, emphasizing the critical role of adipocytes, angiogenesis, hypoxia and oxygen transport in driving immune responses and cancer progression. Global sensitivity analyses highlight potential therapeutic intervention points, such as VEGFs' role in EC growth and oxygen transportation and severe hypoxia's effect on cancer and the total number of cells. The VEGF-mediated production rate of ECs shows an essential time-dependent impact, highlighting the importance of early intervention in slowing cancer progression. These findings align with clinical observations demonstrating the VEGF inhibitors' efficacy and suggest a timely intervention for better outcomes.
Collapse
Affiliation(s)
- Navid Mohammad Mirzaei
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York10032, USA
| | - Panayotis G. Kevrekidis
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA01003-4515, USA
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA01003-4515, USA
| |
Collapse
|
2
|
Yang Z, Chen F, Zhang Y, Ou M, Tan P, Xu X, Li Q, Zhou S. Therapeutic targeting of white adipose tissue metabolic dysfunction in obesity: mechanisms and opportunities. MedComm (Beijing) 2024; 5:e560. [PMID: 38812572 PMCID: PMC11134193 DOI: 10.1002/mco2.560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/09/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
White adipose tissue is not only a highly heterogeneous organ containing various cells, such as adipocytes, adipose stem and progenitor cells, and immune cells, but also an endocrine organ that is highly important for regulating metabolic and immune homeostasis. In individuals with obesity, dynamic cellular changes in adipose tissue result in phenotypic switching and adipose tissue dysfunction, including pathological expansion, WAT fibrosis, immune cell infiltration, endoplasmic reticulum stress, and ectopic lipid accumulation, ultimately leading to chronic low-grade inflammation and insulin resistance. Recently, many distinct subpopulations of adipose tissue have been identified, providing new insights into the potential mechanisms of adipose dysfunction in individuals with obesity. Therefore, targeting white adipose tissue as a therapeutic agent for treating obesity and obesity-related metabolic diseases is of great scientific interest. Here, we provide an overview of white adipose tissue remodeling in individuals with obesity including cellular changes and discuss the underlying regulatory mechanisms of white adipose tissue metabolic dysfunction. Currently, various studies have uncovered promising targets and strategies for obesity treatment. We also outline the potential therapeutic signaling pathways of targeting adipose tissue and summarize existing therapeutic strategies for antiobesity treatment including pharmacological approaches, lifestyle interventions, and novel therapies.
Collapse
Affiliation(s)
- Zi‐Han Yang
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang‐Zhou Chen
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Xiang Zhang
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Min‐Yi Ou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Poh‐Ching Tan
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue‐Wen Xu
- Department of Plastic and Burn SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Qing‐Feng Li
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shuang‐Bai Zhou
- Department of Plastic & Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
3
|
Guo Y, Hu Z, Chen J, Zhang J, Fan Z, Qu Q, Miao Y. Feasibility of adipose-derived therapies for hair regeneration: Insights based on signaling interplay and clinical overview. J Am Acad Dermatol 2023; 89:784-794. [PMID: 34883154 DOI: 10.1016/j.jaad.2021.11.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/13/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Dermal white adipose tissue (dWAT) is a dynamic component of the skin and closely interacts with the hair follicle. Interestingly, dWAT envelops the hair follicle during anagen and undergoes fluctuations in volume throughout the hair cycle. dWAT-derived extracellular vesicles can significantly regulate the hair cycle, and this provides a theoretical basis for utilizing adipose tissue as a feasible clinical strategy to treat hair loss. However, the amount and depth of the available literature are far from enough to fully elucidate the prominent role of dWAT in modulating the hair growth cycle. This review starts by investigating the hair cycle-coupled dWAT remodeling and the reciprocal signaling interplay underneath. Then, it summarizes the current literature and assesses the advantages and limitations of clinical research utilizing adipose-derived therapies for hair regeneration.
Collapse
Affiliation(s)
- Yilong Guo
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jian Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhexiang Fan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qian Qu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
4
|
Chiang YF, Huang KC, Chen HY, Huang TC, Ali M, Chang HY, Shieh TM, Shih YH, Wang KL, Huang YJ, Chung CP, Hsia SM. The Adipokine Visfatin Modulates Cancer Stem Cell Properties in Triple-Negative Breast Cancer. Biomedicines 2023; 11:biomedicines11020297. [PMID: 36830834 PMCID: PMC9953233 DOI: 10.3390/biomedicines11020297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/30/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
Obesity is a cancer progression risk factor; excessive adipocytes increase adipokine secretion. Visfatin, a novel adipokine highly expressed in cancer patients, is related to breast cancer risk. The modulation of nicotinamide adenine dinucleotide (NAD+) metabolism and the induction of a tumorigenic environment plays a vital role in cancer progression. Among cancer cell types, cancer stem-like cells (CSCs) with self-renewal and chemotherapy-resistance abilities could modulate tumor progression and cancer recurrence ability. In this study, we focused on visfatin's modulation effect on stemness-related properties using the high-malignancy breast cancer cell line MDA-MB-231 in in vitro and in vivo studies. Visfatin treatment significantly increased both the sphere number and sphere diameter and increased the protein expression of NANOG homeobox (NANOG), sex-determining region Y-box 2 (SOX2), and octamer-binding transcription factor 4 (OCT4), as well as SIRT1 protein levels. The serum angiogenesis marker VEGF and extracellular nicotinamide phosphoribosyl transferase (NAMPT, visfatin) were induced after visfatin treatment, increasing the stemness and angiogenesis environment, which were significantly reduced by the visfatin inhibitor FK866. Our results demonstrate that the visfatin-activated SIRT-SOX2 axis promotes triple-negative breast cancer stemness and enriches the tumorigenic microenvironment.
Collapse
Affiliation(s)
- Yi-Fen Chiang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Ko-Chieh Huang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Hsin-Yuan Chen
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
| | - Tsui-Chin Huang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110301, Taiwan
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Hsin-Yi Chang
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei 114201, Taiwan
| | - Tzong-Ming Shieh
- School of Dentistry, College of Dentistry, China Medical University, Taichung 40402, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| | - Kai-Lee Wang
- Department of Nursing, Ching Kuo Institute of Management and Health, Keelung 20301, Taiwan
| | - Yun-Ju Huang
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan City 710301, Taiwan
| | - Cheng-Pei Chung
- Department of Nutrition and Health Sciences, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
- School of Food and Safety, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Correspondence: ; Tel.: +886-2-27361661 (ext. 6558)
| |
Collapse
|
5
|
García-Chico C, López-Ortiz S, Peñín-Grandes S, Pinto-Fraga J, Valenzuela PL, Emanuele E, Ceci C, Graziani G, Fiuza-Luces C, Lista S, Lucia A, Santos-Lozano A. Physical Exercise and the Hallmarks of Breast Cancer: A Narrative Review. Cancers (Basel) 2023; 15:324. [PMID: 36612320 PMCID: PMC9818971 DOI: 10.3390/cancers15010324] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Growing evidence suggests that, among the different molecular/cellular pathophysiological mechanisms associated with cancer, there are 14 hallmarks that play a major role, including: (i) sustaining proliferative signaling, (ii) evading growth suppressors, (iii) activating invasion and metastasis, (iv) enabling replicative immortality, (v) inducing angiogenesis, (vi) resisting cell death, (vii) reprogramming energy metabolism, (viii) evading immune destruction, (ix) genome instability and mutations, (x) tumor-promoting inflammation, (xi) unlocking phenotypic plasticity, (xii) nonmutational epigenetic reprogramming, (xiii) polymorphic microbiomes, and (xiv) senescent cells. These hallmarks are also associated with the development of breast cancer, which represents the most prevalent tumor type in the world. The present narrative review aims to describe, for the first time, the effects of physical activity/exercise on these hallmarks. In summary, an active lifestyle, and particularly regular physical exercise, provides beneficial effects on all major hallmarks associated with breast cancer, and might therefore help to counteract the progression of the disease or its associated burden.
Collapse
Affiliation(s)
- Celia García-Chico
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Susana López-Ortiz
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Saúl Peñín-Grandes
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - José Pinto-Fraga
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Pedro L. Valenzuela
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
- Department of Systems Biology, University of Alcalá, 28871 Madrid, Spain
| | | | - Claudia Ceci
- Departmental Faculty of Medicine, Saint Camillus International University of Health and Medical Sciences, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carmen Fiuza-Luces
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
| | - Simone Lista
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
- Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH, Miguel de Cervantes European University, 27038 Valladolid, Spain
- Research Institute of the Hospital 12 de Octubre (‘Imas12’ [PaHerg Group]), 28041 Madrid, Spain
| |
Collapse
|
6
|
Guo D, Lin C, Lu Y, Guan H, Qi W, Zhang H, Shao Y, Zeng C, Zhang R, Zhang H, Bai X, Cai D. FABP4 secreted by M1-polarized macrophages promotes synovitis and angiogenesis to exacerbate rheumatoid arthritis. Bone Res 2022; 10:45. [PMID: 35729106 PMCID: PMC9213409 DOI: 10.1038/s41413-022-00211-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 03/10/2022] [Accepted: 03/20/2022] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence shows that adipokines play a vital role in the development of rheumatoid arthritis (RA). Fatty acid-binding protein 4 (FABP4), a novel adipokine that regulates inflammation and angiogenesis, has been extensively studied in a variety of organs and diseases. However, the effect of FABP4 on RA remains unclear. Here, we found that FABP4 expression was upregulated in synovial M1-polarized macrophages in RA. The increase in FABP4 promoted synovitis, angiogenesis, and cartilage degradation to exacerbate RA progression in vivo and in vitro, whereas BMS309403 (a FABP4 inhibitor) and anagliptin (dipeptidyl peptidase 4 inhibitor) inhibited FABP4 expression in serum and synovial M1-polarized macrophages in mice to alleviate RA progression. Further studies showed that constitutive activation of mammalian target of rapamycin complex 1 (mTORC1) by TSC1 deletion specifically in the myeloid lineage regulated FABP4 expression in macrophages to exacerbate RA progression in mice. In contrast, inhibition of mTORC1 by ras homolog enriched in brain (Rheb1) disruption specifically in the myeloid lineage reduced FABP4 expression in macrophages to attenuate RA development in mice. Our findings established an essential role of FABP4 that is secreted by M1-polarized macrophages in synovitis, angiogenesis, and cartilage degradation in RA. BMS309403 and anagliptin inhibited FABP4 expression in synovial M1-polarized macrophages to alleviate RA development. Hence, FABP4 may represent a potential target for RA therapy.
Collapse
Affiliation(s)
- Dong Guo
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Chuangxin Lin
- Department of Orthopedic Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Yuheng Lu
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hong Guan
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Weizhong Qi
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Hongbo Zhang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yan Shao
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Chun Zeng
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Rongkai Zhang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Haiyan Zhang
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China. .,State Key Laboratory of Organ Failure Research, Department of Cell Biology, Southern Medical University School of Basic Medical Sciences, Guangzhou, China.
| | - Daozhang Cai
- Department of Joint Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. .,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China.
| |
Collapse
|
7
|
Mocellin D, Bratti LDOS, Silva AH, Assunção LS, Kretzer IF, Filippin-Monteiro FB. Serum from morbidly obese patients affects melanoma cell behavior in vitro. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-979020201000x42e19375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/03/2023] Open
|
8
|
Rae JM, Lippman ME. The role of estrogen receptor signaling in suppressing the immune response to cancer. J Clin Invest 2021; 131:155476. [PMID: 34907918 DOI: 10.1172/jci155476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapies are standard of care for the treatment of many solid tumors. While some patients with cancer experience exceptional and long-term responses, intrinsic and acquired mechanisms of resistance limit the clinical efficacy of ICBs. In addition, ICBs can elicit life-threatening side effects. Alternative options that can increase ICB responses without added toxicities are needed. In this issue of the JCI, Chakraborty et al. explored the role of estrogen receptor α (ERα) in modulating ICB activity. Using transcriptomics and preclinical melanoma models, the authors show that ERα signaling in tumor-associated macrophages contributed to an immune-suppressive state within the tumor microenvironment (TME) by promoting CD8+ T cell dysfunction and exhaustion. Further, in murine melanoma models, the addition of fulvestrant, a selective estrogen receptor downregulator (SERD) approved for the treatment of breast cancer, enhanced the antitumor effects of ICB. These results provide a rationale for human trials to test the combination of antiestrogens with ICBs.
Collapse
Affiliation(s)
- James M Rae
- Division of Hematology and Oncology, Department of Internal Medicine, and.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Marc E Lippman
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA
| |
Collapse
|
9
|
Lim HY, Seung BJ, Cho SH, Kim SH, Bae MK, Sur JH. Canine mammary cancer in overweight or obese female dogs is associated with intratumoral microvessel density and macrophage counts. Vet Pathol 2021; 59:39-45. [PMID: 34547936 DOI: 10.1177/03009858211040481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Obesity is a major health condition owing to its effects on chronic diseases and cancers in humans, but little information is available regarding the role of obesity in canine mammary cancer (CMC). In the present study, we performed immunohistochemistry to investigate the effect of obesity on CMC by analyzing the number of tumor-associated macrophages, intratumoral microvessel density (iMVD), and the expression of prognostic factors including epidermal growth factor receptor (EGFR), cyclooxygenase 2 (COX-2), and Ki67 in CMC specimens. These data were compared in CMC specimens from lean or ideal body weight (Group 1) versus overweight or obese (Group 2) female dogs (n = 60 for each group). Associations between obesity status and histologic characteristics, such as histologic subtype, grading, and lymphatic invasion, were also investigated. Compared with lean or ideal body weight dogs, TAM (tumor-associated macrophage) counts (P < .005) and iMVD (P < .001) were significantly higher in overweight or obese dogs. CMC specimens of dogs in the overweight or obese group also showed higher histologic grade (P < .001). In addition, although no association was found between obesity status and either COX-2 or EGFR expression, Ki67 expression was greater in CMC specimens of overweight or obese dogs (P < .005). The results of this study suggest that obesity may influence CMC development and progression, being associated with higher histologic grade, greater infiltration of TAMs, and increased tumor angiogenesis.
Collapse
Affiliation(s)
- Ha-Young Lim
- Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | | | - Seung-Hee Cho
- Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Soo-Hyeon Kim
- Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Min-Kyung Bae
- Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| | - Jung-Hyang Sur
- Konkuk University, Gwangjin-gu, Seoul, Republic of Korea
| |
Collapse
|
10
|
Li WM, Sun QW, Fan XF, Zhang JC, Xu T, Shen QQ, Jia L. Mammography breast density: an effective supplemental modality for the precise grading of ultrasound BI-RADS 4 categories. Gland Surg 2021; 10:2010-2018. [PMID: 34268085 DOI: 10.21037/gs-21-313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/17/2021] [Indexed: 11/06/2022]
Abstract
Background High breast density is significantly associated with an increased risk of breast diseases. Presently, suspected breast masses assessed as Breast Imaging-Reporting and Data System (BI-RADS) grade 4 provide a wide range of positive predictive values. Moreover, subcategories (4a, 4b, and 4c) are still under consideration as the diagnostic criteria are neither comprehensive nor objective. However, whether mammography breast density (MBD) has any impact on the accurate grading of BI-RADS 4 assessed by ultrasound (US) remains unknown. Methods A total of 1,086 women with 1,293 breast masses were included and assessed as BI-RADS 3-5 by US. The subcategories of MBD (from the ACR-a to the ACR-d group) were assessed by mammography according to the criteria of the American College of Radiology (ACR). The clinicopathological characteristics of these patients were reviewed retrospectively. The malignancy rates of breast masses among different subgroups assessed by BI-RADS were re-estimated with MBD. Results Almost all BI-RADS 3 masses were classified as benign and nearly all BI-RADS 5 masses were identified as malignant. Significant inverse associations between MBD and malignancy rates were detected between the BI-RADS 4a and BI-RADS 4b groups. Moreover, malignancy rates decreased significantly from ACR-a to ACR-d for BI-RADS 4a and 4b breast lesions (P<0.001). However, this trend was not observed in BI-RADS 4c breast lesions. Conclusions MBD could serve as a crucial factor for the accurate grading of BI-RADS 4 lesions assessed by US. We strongly recommend the adoption of the MBD as a possible supplemental screening modality for US. Furthermore, it is equally beneficial for accurate risk assessment and screening recommendations based on MBD.
Collapse
Affiliation(s)
- Wei-Min Li
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Qiu-Wei Sun
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiao-Fang Fan
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jun-Chao Zhang
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ting Xu
- Department of Clinical and Research, Shenzhen Mindray Biomedical Electronics Co., Ltd, Shenzhen, China
| | - Qi-Qi Shen
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Lei Jia
- Department of Ultrasonography, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Relationships between Exercise Modality and Activity Restriction, Quality of Life, and Hematopoietic Profile in Korean Breast Cancer Survivors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17186899. [PMID: 32967252 PMCID: PMC7559845 DOI: 10.3390/ijerph17186899] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022]
Abstract
This study aimed to examine the relationships between activity restriction, quality of life (QoL), and hematopoietic profile in breast cancer survivors according to exercise modality. The subjects in this study were 187 female breast cancer survivors among a total of 32,631 participants in the Korea National Health and Nutrition Examination Survey, which was conducted from 2016 to 2018. The selected subjects participated in a questionnaire survey and blood analysis. A cross-analysis was conducted to determine the relationship between participation in various modality of exercise (e.g., aerobic exercise, resistance exercise, walking exercise). The phi coefficients or Cramer’s V value for activity restriction and QoL were calculated; an independent t-test was conducted to evaluate the differences between hematopoietic profiles based on the modality of exercise. Statistically significant correlations were seen between obesity and aerobic exercise and walking frequency, as well as between diabetes and aerobic exercise and activity restriction. With respect to QoL, there was a statistically significant correlation between participation in aerobic exercise and exercise ability, participation in aerobic exercise and anxiety/depression, participation in resistance exercise and subjective health status, participation in resistance exercise and exercise ability, and participation in weekly walking exercise and self-care ability. Regarding hemodynamic changes, red blood cells increased significantly in breast cancer survivors who participated in weekly resistance exercise compared to in those who did not. In conclusion, exercise participation had a positive effect on activity restriction, QoL, and hematopoietic profile in breast cancer survivors; in particular, some modalities of aerobic exercise were more effective.
Collapse
|
12
|
Qureshi R, Picon-Ruiz M, Aurrekoetxea-Rodriguez I, Nunes de Paiva V, D'Amico M, Yoon H, Radhakrishnan R, Morata-Tarifa C, Ince T, Lippman ME, Thaller SR, Rodgers SE, Kesmodel S, Vivanco MDM, Slingerland JM. The Major Pre- and Postmenopausal Estrogens Play Opposing Roles in Obesity-Driven Mammary Inflammation and Breast Cancer Development. Cell Metab 2020; 31:1154-1172.e9. [PMID: 32492394 DOI: 10.1016/j.cmet.2020.05.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/26/2019] [Accepted: 05/11/2020] [Indexed: 12/30/2022]
Abstract
Many inflammation-associated diseases, including cancers, increase in women after menopause and with obesity. In contrast to anti-inflammatory actions of 17β-estradiol, we find estrone, which dominates after menopause, is pro-inflammatory. In human mammary adipocytes, cytokine expression increases with obesity, menopause, and cancer. Adipocyte:cancer cell interaction stimulates estrone- and NFκB-dependent pro-inflammatory cytokine upregulation. Estrone- and 17β-estradiol-driven transcriptomes differ. Estrone:ERα stimulates NFκB-mediated cytokine gene induction; 17β-estradiol opposes this. In obese mice, estrone increases and 17β-estradiol relieves inflammation. Estrone drives more rapid ER+ breast cancer growth in vivo. HSD17B14, which converts 17β-estradiol to estrone, associates with poor ER+ breast cancer outcome. Estrone and HSD17B14 upregulate inflammation, ALDH1 activity, and tumorspheres, while 17β-estradiol and HSD17B14 knockdown oppose these. Finally, a high intratumor estrone:17β-estradiol ratio increases tumor-initiating stem cells and ER+ cancer growth in vivo. These findings help explain why postmenopausal ER+ breast cancer increases with obesity, and offer new strategies for prevention and therapy.
Collapse
Affiliation(s)
- Rehana Qureshi
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA.
| | - Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Human Anatomy and Embryology, Faculty of Medicine, Biopathology and Medicine Regenerative Institute (IBIMER), and Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), Granada, Spain.
| | - Iskander Aurrekoetxea-Rodriguez
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Vanessa Nunes de Paiva
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Massimo D'Amico
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Hyunho Yoon
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Ramya Radhakrishnan
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Cynthia Morata-Tarifa
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | - Tan Ince
- Department of Pathology, Weill Cornell Medicine and New York Presbyterian Brooklyn Methodist Hospital, New York, NY, USA
| | - Marc E Lippman
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Seth R Thaller
- Department of Plastic Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Steven E Rodgers
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Susan Kesmodel
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Maria Del Mar Vivanco
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA; Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
13
|
Kessous R, Wainstock T, Sheiner E. Pre-pregnancy obesity and childhood malignancies: A population-based cohort study. Pediatr Blood Cancer 2020; 67:e28269. [PMID: 32196946 DOI: 10.1002/pbc.28269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Exploring the effect of maternal obesity during pregnancy on the long-term health of offspring is of great importance. The aim of this study was to evaluate the association between maternal pre-pregnancy obesity and future risk of childhood malignancies. STUDY DESIGN A population-based cohort analysis comparing the risk for long-term childhood malignancies (up to the age of 18 years) in children born (1991-2014) to mothers with and without pre-pregnancy obesity (body mass index > 30) was conducted in July 2017. Childhood malignancies were predefined based on ICD-9 codes, as recorded in the hospital medical files. Children with congenital malformations and multiple gestations were excluded from the analysis. The Kaplan-Meier survival curve was constructed to compare cumulative oncological morbidity in both groups over time. The Cox proportional hazards model was used to control for confounders. RESULTS During the study period, 241 273 infants met the inclusion criteria; 3268 were born to mothers with pre-pregnancy obesity. Children of obese women had significantly increased risk for several childhood malignancies (including brain tumors) as well as increased risk for total hospitalizations with malignancy diagnoses, even after controlling for several confounders (adjusted HR 1.90, 95% CI 1.07-3.37, P = 0.028). Cumulative incidence of oncological morbidity was also significantly increased over time in the studied group (log-rank P = 0.023). CONCLUSION Maternal pre-pregnancy obesity is significantly associated with an increased long-term risk for general childhood malignancies, and specifically brain tumors in the offspring. These results are important when counseling mothers regarding potential future risks and recommended lifestyle modifications.
Collapse
Affiliation(s)
- Roy Kessous
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tamar Wainstock
- The Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eyal Sheiner
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
14
|
Martínez-Rodríguez OP, Thompson-Bonilla MDR, Jaramillo-Flores ME. Association between obesity and breast cancer: Molecular bases and the effect of flavonoids in signaling pathways. Crit Rev Food Sci Nutr 2020; 60:3770-3792. [PMID: 31899947 DOI: 10.1080/10408398.2019.1708262] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity is an abnormal or excessive accumulation of fat that leads to different health problems, such as cancer, where the adipocytes promote the proliferation, migration, and invasion of cancer cells, especially in the breast, where the epithelial cells are immersed in a fatty environment, and the interactions between these two types of cells involve, not only adipokines but also local pro-inflammatory mechanisms and hypoxic processes generating anti-apoptotic signals, which are a common result in leptin signaling. The expression of the Vascular Endothelial Growth Factor (VEGF) and cyclin D1, results in the decrease in phosphorylation of AMPK, increasing the activity of the aromatase enzyme; alternatively, the adiponectin activates AMPK to reduce inflammation. Nevertheless, alterations of the JAK/STAT pathways contribute to mammary carcinogenesis, while the PI3K/AKT/mTOR pathway controls most of the cancer's characteristics such as the cell cycle, survival, differentiation, proliferation, motility, metabolism, and genetic stability. Therefore, the purpose of the present review is, through the accumulated scientific evidence, to find the concordance between the signaling pathways involved among obesity and breast cancer, which can be modulated by using flavonoids.
Collapse
Affiliation(s)
- Oswaldo Pablo Martínez-Rodríguez
- Departamento de Ingeniería Bioquímica, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Ciudad de México, México
| | - María Del Rocío Thompson-Bonilla
- Laboratorio de Medicina Genómica, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado ISSSTE, Ciudad de México, México
| | - María Eugenia Jaramillo-Flores
- Departamento de Ingeniería Bioquímica, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Ciudad de México, México
| |
Collapse
|
15
|
Abstract
The tumor microenvironment is the primary location in which tumor cells and the host immune system interact. There are many physiological, biochemical, cellular mechanisms in the neighbor of tumor which is composed of various cell types. Interactions of chemokines and chemokine receptors can recruit immune cell subsets into the tumor microenvironment. These interactions can modulate tumor progression and metastasis. In this chapter, we will focus on chemokine (C-C motif) ligand 7 (CCL7) that is highly expressed in the tumor microenvironment of various cancers, including colorectal cancer, breast cancer, oral cancer, renal cancer, and gastric cancer. We reviewed how CCL7 can affect cancer immunity and tumorigenesis by describing its regulation and roles in immune cell recruitment and stromal cell biology.
Collapse
|
16
|
Becerril S, Rodríguez A, Catalán V, Ramírez B, Unamuno X, Portincasa P, Gómez-Ambrosi J, Frühbeck G. Functional Relationship between Leptin and Nitric Oxide in Metabolism. Nutrients 2019; 11:nu11092129. [PMID: 31500090 PMCID: PMC6769456 DOI: 10.3390/nu11092129] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/23/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
Leptin, the product of the ob gene, was originally described as a satiety factor, playing a crucial role in the control of body weight. Nevertheless, the wide distribution of leptin receptors in peripheral tissues supports that leptin exerts pleiotropic biological effects, consisting of the modulation of numerous processes including thermogenesis, reproduction, angiogenesis, hematopoiesis, osteogenesis, neuroendocrine, and immune functions as well as arterial pressure control. Nitric oxide (NO) is a free radical synthesized from L-arginine by the action of the NO synthase (NOS) enzyme. Three NOS isoforms have been identified: the neuronal NOS (nNOS) and endothelial NOS (eNOS) constitutive isoforms, and the inducible NOS (iNOS). NO mediates multiple biological effects in a variety of physiological systems such as energy balance, blood pressure, reproduction, immune response, or reproduction. Leptin and NO on their own participate in multiple common physiological processes, with a functional relationship between both factors having been identified. The present review describes the functional relationship between leptin and NO in different physiological processes.
Collapse
Affiliation(s)
- Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Medical Engineering Laboratory, University of Navarra, 31008 Pamplona, Spain.
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Policlinico Hospital, 70124 Bari, Italy.
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 31008 Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain.
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain.
| |
Collapse
|
17
|
Pearce JV, Farrar JS, Lownik JC, Ni B, Chen S, Kan TW, Celi FS. E0771 and 4T1 murine breast cancer cells and interleukin 6 alter gene expression patterns but do not induce browning in cultured white adipocytes. Biochem Biophys Rep 2019; 18:100624. [PMID: 31193642 PMCID: PMC6536889 DOI: 10.1016/j.bbrep.2019.100624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 03/05/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer remains a substantial clinical problem worldwide, and cancer-associated cachexia is a condition associated with poor prognosis in this and other malignancies. Adipose tissue is involved in the development and progression of cancer-associated cachexia, but its various roles and mechanisms of action are not completely defined, especially as it relates to breast cancer. Interleukin 6 has been implicated in several mechanisms contributing to increased breast cancer tumorigenesis, as well as a net-negative energy balance and cancer-associated cachexia via adipose tissue remodeling in other models of cancer; however, its potential role in breast cancer-associated white adipose browning has not been explored. In this study, we demonstrate localized white adipose tissue browning in a spontaneous model of murine mammary cancer. We then used an in vitro murine adipocyte culture system with the E0771 and 4T1 cell lines as models of breast cancer. We demonstrate that while the E0771 and 4T1 secretomes and cross-talk with white adipocytes alter white adipocyte mRNA expression, they do not directly induce white adipocyte browning. Additionally, we show that neither exogenous administration of interleukin 6 alone or with its soluble receptor directly induce white adipocyte browning. Together, these results demonstrate that neither the E0771 or 4T1 murine breast cancer cell lines, nor interleukin 6, directly cause browning of cultured white adipocytes. This suggests that their roles in adipose tissue remodeling are more complex and indirect in nature.
Collapse
Affiliation(s)
- Janina V. Pearce
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jared S. Farrar
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Joseph C. Lownik
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Bin Ni
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Shanshan Chen
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Tiffany W. Kan
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Francesco S. Celi
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
18
|
Identification of CTRP1 as a Prognostic Biomarker and Oncogene in Human Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2582416. [PMID: 31183364 PMCID: PMC6515110 DOI: 10.1155/2019/2582416] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/21/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022]
Abstract
Introduction Glioblastoma (GBM) is the most frequent and malignant type of primary brain tumors in adults. The valuable prognostic biomarkers and therapeutic targets for GBM remain to be elucidated. The association of adipokines with cancer has been well documented. The C1q/TNF-related protein 1 (CTRP1), a novel adipokine, belongs to the CTRP family. Methods In the present study, the expression and potential roles of CTRP1 in GBM were explored based on in silico evaluation, including GEPIA, the Pathology Atlas of the Human Protein Atlas, cBioPortal, TIMER, and SurvExpress. The CCK8, transwell, and wound healing assays were used to detect cell proliferation and migration. Results It was found that mRNA expression levels of CTRP1 were significantly upregulated in GBM tissues compared with those in nontumor tissues according to the analysis on public dataset and immunohistochemical results of GBM tissues (P<0.05). CTRP1 was mainly localized in the cytoplasm and cell membrane of GBM cells. The genetic alterations of CTRP1 occurred at a low rate in GBM (2 of 591 sequenced cases/patients, 0.33%). The mRNA expression levels of CTRP1 were positively associated with the tumor-infiltrating macrophages and CCL2 in GBM (P<0.05, respectively). The higher mRNA expression levels of CTRP1 were significantly correlated with higher risk and shorter overall survival time in GBM (P<0.05). CTRP1 knockdown significantly inhibited the proliferation and migration in human GBM cells, suggesting the inhibition of CTRP1 on human GMB progression. Moreover, CTRP1 knockdown inhibited CCL2 expression, and CCL2 overexpression reversed the inhibition of cell proliferation and migration induced by CTRP1 knockdown, suggesting that CTRP1 promoted tumor progression by regulating CCL2 expression. Conclusions These findings suggest that CTRP1 potentially indicates poor prognosis in GBM and promotes the progression of human GBM.
Collapse
|
19
|
Amin MN, Hussain MS, Sarwar MS, Rahman Moghal MM, Das A, Hossain MZ, Chowdhury JA, Millat MS, Islam MS. How the association between obesity and inflammation may lead to insulin resistance and cancer. Diabetes Metab Syndr 2019; 13:1213-1224. [PMID: 31336467 DOI: 10.1016/j.dsx.2019.01.041] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/22/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND OBJECTIVES Obesity is associated with metabolic dysfunction and over nutrition. Increased body mass index and obesity are strongly amalgamated with changes in the physiological function of adipose tissue, leading to altered secretion of adipocytokines, inflammatory mediators release as well as chronic inflammation and insulin resistance. The purposes of this study were to review the evidence of how obesity and inflammation may lead to insulin resistance and cancer. SUMMARY Recent findings suggested that increased level of inflammatory mediators in obesity, plays an introductory and cabalistic role in the development of different types of inflammatory disorders including type 2 diabetes mellitus. Link between elevated body mass index and type 2 diabetes mellitus (T2DM). Several of the factors-such as increased levels of leptin, plasminogen activator inhibitor-1, decreased levels of adiponectin, insulin resistance, chronic inflammation etc. consequently result in carcinogenesis and carcinogenic progression too. CONCLUSION This review summarizes how cytokine production in adipose tissue of obese subject creates a chronic inflammatory environment that favors tumor cell motility and invasion to enhance the metastatic potential of tumor cells. High levels of cytokine in the circulation of affected individuals have been associated with a significantly worse outcome. This article also reconnoiters the mechanisms that link obesity to numerous disorders such as inflammation, diabetes, cancers and most specifically combine these processes in a single image. Understanding these mechanisms may assist to understand the consequences of obesity.
Collapse
Affiliation(s)
- Mohammad Nurul Amin
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh; Department of Pharmacy, Atish Dipankar University of Science and Technology, Sonapur, Uttara, Dhaka, Bangladesh
| | - Md Saddam Hussain
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh
| | - Md Shahid Sarwar
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh
| | - Md Mizanur Rahman Moghal
- Department of Pharmacy, Mawlana Bhashani Science and Technology University, Santosh, 1902, Tangail, Bangladesh
| | - Abhijit Das
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh
| | - Mohammad Zahid Hossain
- Department of Pharmacy, State University of Bangladesh, Dhanmondi, Dhaka, 1206, Bangladesh
| | - Jakir Ahmed Chowdhury
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Md Shalahuddin Millat
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh
| | - Mohammad Safiqul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Sonapur, Noakhali, 3814, Bangladesh.
| |
Collapse
|
20
|
Wang L, Wang R, Ye Z, Wang Y, Li X, Chen W, Zhang M, Cai C. PVT1 affects EMT and cell proliferation and migration via regulating p21 in triple-negative breast cancer cells cultured with mature adipogenic medium. Acta Biochim Biophys Sin (Shanghai) 2018; 50:1211-1218. [PMID: 30371726 DOI: 10.1093/abbs/gmy129] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Indexed: 11/14/2022] Open
Abstract
Excessive adiposity has long been proved to be associated with greater incidence and mortality of breast cancer in post-menopausal women. However, the effects and underlying mechanisms of human adipocytes on breast cancer cells remain largely unknown. In recent years, several reports have revealed the oncogenic role of long non-coding RNA PVT1 in breast cancer. Here, we aimed to investigate the role and underlying mechanisms of PVT1 in triple-negative breast cancer (TNBC) cells cultured with mature adipogenic medium. At first, we successfully induced adipogenic differentiation from human adipose-derived mesenchymal stem cells and collected the mature adipogenic medium to mimic excessive adiposity. Our results demonstrated that the mature adipogenic medium promoted the epithelial-mesenchymal transition, enhanced the cell viability and migration potential of TNBC cells. In addition, we proved that mature adipogenic medium affected the PVT1 expression and inhibition of the PVT1 disturbed the role of mature adipogenic medium in TNBC cells. Finally, we illustrated that repression of p21 restored the phenotype caused by PVT1 knockdown in TNBC cells treated with mature adipogenic medium. Taken together, our results demonstrated that PVT1 affected the role of mature adipogenic medium in TNBC cells via modulating p21 expression.
Collapse
Affiliation(s)
- Lingli Wang
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruirui Wang
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Zi Ye
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Yanyan Wang
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Xiao Li
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Weizhen Chen
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Mengna Zhang
- Medical Research Institute, Wuhan University, Wuhan, China
| | - Cheguo Cai
- Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Pancreatic Cancer and Obesity: Molecular Mechanisms of Cell Transformation and Chemoresistance. Int J Mol Sci 2018; 19:ijms19113331. [PMID: 30366466 PMCID: PMC6274743 DOI: 10.3390/ijms19113331] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer and obesity are the two major epidemics of the 21st century. Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of death, with a five-year overall survival rate of only 8%. Its incidence and mortality have increased in recent years, and this cancer type is expected to be among the top five leading causes of cancer-related death by 2030 in the United States (US). In the last three decades, the prevalence of overweight people has boosted with a consequent increase in obesity-related diseases. Considerable epidemiologic evidence correlates overweight and obese conditions to an increased risk of several types of cancer, including PDAC. Besides being a risk factor for multiple metabolic disorders, the tumor-promoting effects of obesity occur at the local level via inflammatory mediators that are associated with adipose inflammation and metabolic or hormones mediators and microbiota dysbiosis. Although an excess of body mass index (BMI) represents the second most modifiable risk factor for PDAC with an increased cancer related-death of more than 20–40%, still little is known about the molecular mechanisms that underlie this strong association. In this review, we focused on the role of obesity as a preventable risk factor of PDAC, discussing the molecular mechanisms linking obesity to cancer initiation and progression. Moreover, we highlighted the role of obesity in defining chemoresistance, showing how a high BMI can actually reduce response to chemotherapy.
Collapse
|
22
|
Impact of X-ray Exposure on the Proliferation and Differentiation of Human Pre-Adipocytes. Int J Mol Sci 2018; 19:ijms19092717. [PMID: 30208657 PMCID: PMC6163807 DOI: 10.3390/ijms19092717] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/20/2023] Open
Abstract
Radiotherapy is a widely used treatment option for cancer patients as well as for patients with musculoskeletal disorders. Adipocytes, the dominant cell type of adipose tissue, are known to constitute an active part of the tumor microenvironment. Moreover, adipocytes support inflammatory processes and cartilage degradation in chronic inflammatory diseases, i.e., rheumatoid and osteoarthritis. Since the production of inflammatory factors is linked to their differentiation stages, we set out to explore the radiation response of pre-adipocytes that may influence their inflammatory potential and differentiation capacity. This is the first study investigating the effects of X-ray irradiation on the proliferation and differentiation capacity of human primary pre-adipocytes, in comparison to Simpson–Golabi–Behmel Syndrome (SGBS) pre-adipocytes, an often-used in vitro model of human primary pre-adipocytes. Our results demonstrate a dose-dependent reduction of the proliferation capacity for both cell strains, whereas the potential for differentiation was mostly unaffected by irradiation. The expression of markers of adipogenic development, such as transcription factors (PPARγ, C/EBPα and C/EBPβ), as well as the release of adipokines (visfatin, adiponectin and leptin) were not significantly changed upon irradiation. However, after irradiation with high X-ray doses, an increased lipid accumulation was observed, which suggests a radiation-induced response of adipocytes related to inflammation. Our results indicate that pre-adipocytes are radio-resistant, and it remains to be elucidated whether this holds true for the overall inflammatory response of adipocytes upon irradiation.
Collapse
|
23
|
LeBlanc ES, Rizzo JH, Pedula KL, Yaffe K, Ensrud KE, Cauley JA, Cawthon PM, Cummings SR, Hillier TA. Long-Term Weight Trajectory and Risk of Hip Fracture, Falls, Impaired Physical Function, and Death. J Am Geriatr Soc 2018; 66:1972-1979. [PMID: 30151825 DOI: 10.1111/jgs.15532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To determine the association between weight trajectory, health status, and mortality in older women. DESIGN Cohort study. SETTING Study of Osteoporotic Fractures. PARTICIPANTS Older community-dwelling women (age: baseline (1986-88), mean 68, range 65-81; Year 20 (2006-08), mean 88, range 83-102 (N = 1,323)). MEASUREMENTS Body weight measured repeatedly over 20 years (mean 8 times). Logistic and Cox proportional hazard models were used to evaluate whether 20-year weight trajectory measures were associated with hip fracture, falls, physical performance, and mortality. RESULTS In models adjusted for age, clinic, calcium use, Year 20 weight, walking speed, comorbidity score, smoking, self-reported health, and walking for exercise, women with moderate weight loss (>9.0 kg) over 20 years had a 74% greater risk of death (hazard ratio (HR) = 1.74, 95% confidence interval (CI) = 1.37-2.20) in the 5 years after the Year 20 visit than those with no weight loss and more than twice the risk of hip fracture (HR = 2.56, 95% CI = 1.39-4.70). They were 3.6 times (odds ratio (OR) = 3.60, 95% CI = 1.86-6.95) as likely to have poor physical function at the Year 20 visit as women with no weight loss but no greater risk of 2 or more falls in the 1.5 years after the Year 20 visit. Weight variability and abrupt weight decline were not associated with adverse health oucomes (falls, fractures, mortality), but those in the highest quartile of variability were 2.3 times (OR = 2.26, 95% CI = 1.34-3.80) as likely to have poor physical function scores. CONCLUSION In women surviving past 80 years of age, moderate weight loss over 20 years was associated with greater risk of hip fracture, poor physical function, and mortality but not of falls. Future work should separate voluntary from involuntary weight loss.
Collapse
Affiliation(s)
- Erin S LeBlanc
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon
| | - Joanne H Rizzo
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon
| | - Kathryn L Pedula
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon
| | - Kristine Yaffe
- Departments of Psychiatry.,Neurology.,Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, California.,San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Kristine E Ensrud
- Department of Medicine, School of Medicine.,Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota.,Center for Chronic Disease Outcomes Research, Veterans Affairs Health Care System, Minneapolis, Minnesota
| | - Jane A Cauley
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Peggy M Cawthon
- Research Institute, California Pacific Medical Center, San Francisco, California
| | - Steven R Cummings
- Research Institute, California Pacific Medical Center, San Francisco, California
| | - Teresa A Hillier
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon.,Center for Health Research, Kaiser Permanente Hawaii, Honolulu, Hawaii
| | -
- Center for Health Research, Kaiser Permanente Northwest, Portland, Oregon
| |
Collapse
|
24
|
Challenges and perspectives in the treatment of diabetes associated breast cancer. Cancer Treat Rev 2018; 70:98-111. [PMID: 30130687 DOI: 10.1016/j.ctrv.2018.08.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is one of the most common chronic disease worldwide and affects all cross-sections of the society including children, women, youth and adults. Scientific evidence has linked diabetes to higher incidence, accelerated progression and increased aggressiveness of different cancers. Among the different forms of cancer, research has reinforced a link between diabetes and the risk of breast cancer. Some studies have specifically linked diabetes to the highly aggressive, triple negative breast cancers (TNBCs) which do not respond to conventional hormonal/HER2 targeted interventions, have chances of early recurrence, metastasize, tend to be more invasive in nature and develop drug resistance. Commonly used anti-diabetic drugs, such as metformin, have recently gained importance in the treatment of breast cancer due to their proposed anti-cancer properties. Here we discuss the link between diabetes and breast cancer, the metabolic disturbances in diabetes that support the development of breast cancer, the challenges involved and future perspective and directions. We link the three main metabolic disturbances (dyslipidemia, hyperinsulinemia and hyperglycemia) that occur in diabetes to potential aberrant molecular pathways that may lead to the development of an oncogenic phenotype of the breast tissue, thereby leading to acceleration of cell growth, proliferation, migration, inflammation, angiogenesis, EMT and metastasis and inhibition of apoptosis in breast cancer cells. Furthermore, managing diabetes and treating cancer using a combination of anti-diabetic and classical anti-cancer drugs should prove to be more efficient in the treatment diabetes associated cancers.
Collapse
|
25
|
Danthala M, Rajesh GR, Gundeti S, Raju GS, Chandran P, Srinivas ML. Obesity and Breast Cancer: Association of Serum Adiponectin, Leptin, and Adiponectin–Leptin Ratio as Risk Biomarkers. Indian J Med Paediatr Oncol 2018. [DOI: 10.4103/ijmpo.ijmpo_203_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Abstract
Introduction: Obesity has been associated with the development of breast cancer. The objectives were to study the association of serum adiponectin, serum leptin, and adiponectin–leptin ratio (ALR) in patients with breast cancer and matched controls, and to study their relationship with the various clinicopathological characteristics of breast cancer. Materials and Methods: A prospective, hospital-based case–control study was conducted on 40 patients with a first-confirmed histopathology diagnosis of breast cancer and 40 controls comprising individuals without a history of cancer simultaneously recruited from the health examination clinics during the same study period. Serum adiponectin and leptin concentrations were measured in a single run using commercially available kits (Human ADP/Acrp30 [adiponectin] enzyme-linked immune-sorbent assay [ELISA] kit and Human Leptin ELISA kit, Elabscience Biotechnology Co., Ltd) according to the manufacturer's instructions. Results:: Serum adiponectin levels were reduced significantly in breast cancer patients, in comparison to controls (P = 0.04), while serum leptin levels were increased significantly in breast cancer patients, in comparison to controls (P = 0.03). ALR was significantly lower in breast cancer cases, in comparison to controls (P = 0.05). There was no correlation between receptor status (estrogen receptor, progesterone receptor, Her2/neu), aggressiveness of disease in terms of tumor size, nodal metastases, stage, tumor grade, and serum adiponectin levels, leptin levels, or ALR. Body mass index was negatively correlated with serum adiponectin levels and ALR (r = 0.33, P = 0.03; r = 0.39, P = 0.01, respectively) and positively correlated with serum leptin levels (r = 0.34, P = 0.02). Conclusion: In summary, our results suggest that low serum adiponectin levels, ALR, and high serum leptin levels are associated with breast cancer.
Collapse
Affiliation(s)
- Madhav Danthala
- Department of Medical Oncology, Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| | - Gogulamudi Ratna Rajesh
- Department of Biochemistry, Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| | - Sadashivudu Gundeti
- Department of Medical Oncology, Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| | | | - Priscilla Chandran
- Department of Biochemistry, Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| | - Maddali Lakshmi Srinivas
- Department of Medical Oncology, Nizam's Institute of Medical Sciences, Panjagutta, Hyderabad, Telangana, India
| |
Collapse
|
26
|
Gyamfi J, Lee YH, Eom M, Choi J. Interleukin-6/STAT3 signalling regulates adipocyte induced epithelial-mesenchymal transition in breast cancer cells. Sci Rep 2018; 8:8859. [PMID: 29891854 PMCID: PMC5995871 DOI: 10.1038/s41598-018-27184-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/22/2018] [Indexed: 01/08/2023] Open
Abstract
The tumour microenvironment is a key regulators of tumour progression through the secretion of growth factors that activate epithelial-mesenchymal transition (EMT). Induction of EMT is a key step for transition from a benign state to a metastatic tumour. Adipose tissue forms a bulk portion of the breast cancer microenvironment, emerging evidence indicates the potential for adipocytes to influence tumour progression through the secretion of adipokines that can induce EMT. The molecular mechanisms underlying how adipocytes enhance breast cancer progression is largely unknown. We hypothesized that paracrine signalling by adipocytes can activate EMT and results in increased migration and invasion characteristics of breast cancer cells. We found that IL-6 secreted by adipocytes induce EMT in breast cancer cells. The effect of IL-6 expression on EMT is mediated through activation of the signal transducer and activated of transcription 3 (STAT3). Blocking of IL-6 signalling in breast cancer cells and adipocytes, decreased proliferation, migration and invasion capabilities and altered the expression of genes regulating EMT. Together, our results suggest that matured human adipocytes can enhance the aggressive behaviour of breast cancer cells and induce an EMT-phenotype through paracrine IL-6/STAT3 signalling.
Collapse
Affiliation(s)
- Jones Gyamfi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Yun-Hee Lee
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea.
| |
Collapse
|
27
|
Dittus KL, Harvey JR, Bunn JY, Kokinda ND, Wilson KM, Priest J, Pratley RE. Impact of a behaviorally-based weight loss intervention on parameters of insulin resistance in breast cancer survivors. BMC Cancer 2018; 18:351. [PMID: 29587682 PMCID: PMC5872579 DOI: 10.1186/s12885-018-4272-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 03/20/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer survivors with excess weight are more likely to have negative breast cancer outcomes. Biomarkers related to insulin resistance may help explain this negative association. Weight loss is associated with improvements in insulin sensitivity. Our goal was to identify the impact of a behaviorally based weight loss intervention on indices of insulin resistance. METHODS Overweight, early stage breast cancer survivors who completed initial cancer therapy were enrolled in a 6 month behaviorally based weight loss intervention that included calorie reduction, exercise and behavior modification. Biomarkers related to insulin resistance were obtained at baseline and after the intervention. Results from participants who achieved ≥5% weight loss were compared to those who lost less weight. RESULTS Despite not having diabetes as a preexisting diagnosis prior to the study, 69% of all participants were considered to have pre-diabetes or diabetes at baseline based on American Diabetes Association definitions. Participants who achieved ≥5% weight loss had significantly lower fasting insulin, AUC insulin, and insulin resistance as measured by HOMA-IR. Beta cell function decreased as anticipated when insulin resistance improved. Additionally, leptin levels declined. CONCLUSIONS Breast cancer survivors who achieved ≥5% weight loss demonstrated significant improvements in indices of insulin resistance. Despite an exclusion criteria of diabetes at the time of enrolment, a high proportion met criteria for pre-diabetes or diabetes at baseline. Pre-diabetes appears to be under recognized in overweight breast cancer survivors. Behaviorally based weight loss interventions can result in weight loss and improvements in biomarkers related to breast cancer outcomes and additionally may decrease the chance of developing diabetes. TRIAL REGISTRATION NCT01482702 4/12/2010 (retrospectively registered). https://clinicaltrials.gov/ct2/show/NCT01482702?term=Dittus&rank=4.
Collapse
Affiliation(s)
- Kim L. Dittus
- Department of Internal Medicine, Vermont Center on Behavior and Health, University of Vermont, Burlington, VT 05405 USA
| | - Jean R. Harvey
- Department of Nutrition and Food Sciences, Vermont Center on Behavior and Health, University of Vermont, Burlington, VT 05405 USA
| | - Janice Y. Bunn
- Department of Mathematics and Statistics, University of Vermont, Burlington, VT 05405 USA
| | - Nathan D. Kokinda
- Department of Rehabilitation and Movement Science, University of Vermont, Burlington, VT 05405 USA
| | | | - Jeff Priest
- Department of Mathematics and Statistics, University of Vermont, Burlington, VT 05405 USA
| | - Richard E. Pratley
- Florida Hospital Translational Research Institute for Metabolism and Diabetes, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32804 USA
| |
Collapse
|
28
|
Seiler A, Chen MA, Brown RL, Fagundes CP. Obesity, Dietary Factors, Nutrition, and Breast Cancer Risk. CURRENT BREAST CANCER REPORTS 2018; 10:14-27. [PMID: 30662586 PMCID: PMC6335046 DOI: 10.1007/s12609-018-0264-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW To synthesize the critical role of obesity-associated inflammation, dietary factors, and nutrition in determining breast cancer risk. RECENT FINDINGS Obesity-associated inflammation is strongly linked to breast cancer risk and progression, largely via two processes: inflammatory pathways and dysregulated metabolism. Cytokine production in excess adipose tissues creates a chronic inflammatory microenvironment, which favors tumor development. Lifestyle factors, including diet, have long been recognized as important determinants of breast cancer risk and mortality. SUMMARY Obesity increases the risk of developing breast cancer in both pre- and postmenopausal women and also negatively affects breast cancer recurrence and survival. Poor dietary habits characterized by the high intake of refined starches, sugar, and both saturated and trans-saturated fats, as well as the low intake of omega-3 fatty acids, natural antioxidants, and fiber, modulate inflammation and, thereby, appear to be linked to increased risk of breast cancer and mortality.
Collapse
Affiliation(s)
- Annina Seiler
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, Haldenbachstrasse 18, 8091 Zurich, Switzerland
| | | | - Ryan L Brown
- Department of Psychology, Rice University, Houston, TX, USA
| | - Christopher P Fagundes
- Department of Psychology, Rice University, Houston, TX, USA
- Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Prepregnancy obesity: a risk factor for future development of ovarian and breast cancer. Eur J Cancer Prev 2018; 26:151-155. [PMID: 26908154 DOI: 10.1097/cej.0000000000000228] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The aim of this study was to investigate the association between a history of prepregnancy obesity and a woman's future long-term risk for the development of female malignancies. A population-based study compared the incidence of long-term female malignancies in a cohort of consecutive women with and without a diagnosis of prepregnancy obesity. Deliveries occurred between the years 1988 and 2013, with a mean follow-up duration of 11.6 years. Women with known malignancies before the index pregnancy and known genetic predisposition for malignancy were excluded from the study. Female malignancies were divided according to specific type (ovary, uterine, breast, and uterine cervix). A Kaplan-Meier survival curve was used to estimate the cumulative incidence of malignancies. A Cox proportional hazards model was used to estimate the adjusted hazard ratios for female malignancy. During the study period, 106 251 deliveries fulfilled the inclusion criteria; 2.2% (n=2360) occurred in patients with a history of prepregnancy obesity. During the follow-up period, patients with prepregnancy obesity had a significantly increased risk for hospitalization because of female malignancies as a group and specifically ovarian and breast cancer. Using a Kaplan-Meier survival curve, patients with a previous diagnosis of prepregnancy obesity had a significantly higher cumulative incidence of female malignancies. Using a Cox proportional hazards model, adjusted for confounders such as gestational diabetes mellitus and maternal age, prepregnancy obesity remained independently associated with long-term risk for female malignancies (adjusted hazard ratio: 1.4; 95% confidence interval: 1.1-1.9; P=0.045). Prepregnancy obesity is an independent risk factor for long-term female malignancies such as ovarian and breast cancer.
Collapse
|
30
|
Gyamfi J, Eom M, Koo JS, Choi J. Multifaceted Roles of Interleukin-6 in Adipocyte-Breast Cancer Cell Interaction. Transl Oncol 2018; 11:275-285. [PMID: 29413760 PMCID: PMC5884177 DOI: 10.1016/j.tranon.2017.12.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide, with a developmental process spanning decades. The malignant cells recruit a variety of cells including fibroblasts, endothelial cells, immune cells, and adipocytes, creating the tumor microenvironment. The tumor microenvironment has emerged as active participants in breast cancer progression and response to treatment through autocrine and paracrine interaction with the malignant cells. Adipose tissue is abundant in the breast cancer microenvironment; interactions with cancer cells create cancer-associated adipocytes which produce a variety of adipokines that influence breast cancer initiation, metastasis, angiogenesis, and cachexia. Interleukin (IL)-6 has emerged as key compound significantly produced by breast cancer cells and adipocytes, with the potential of inducing proliferation, epithelial-mesenchymal phenotype, stem cell phenotype, angiogenesis, cachexia, and therapeutic resistance in breast cancer cells. Our aim is to present a brief knowledge of IL-6’s role in breast cancer. This review summarizes our current understanding of the breast microenvironment, with emphasis on adipocytes as key players in breast cancer tumorigenesis. The effects of key adipocytes such as leptin, adipokines, TGF-b, and IL-6 are discussed. Finally, we discuss the role of IL-6 in various aspects of cancer progression.
Collapse
Affiliation(s)
- Jones Gyamfi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ja-Seung Koo
- Department of Pathology, Yonsei University College of Medicine.
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea.
| |
Collapse
|
31
|
Šnajder D, Perić Kačarević Ž, Grgić A, Bijelić N, Fenrich M, Belovari T, Radić R. Effect of different combination of maternal and postnatal diet on adipose tissue morphology in male rat offspring. J Matern Fetal Neonatal Med 2018; 32:1838-1846. [PMID: 29295664 DOI: 10.1080/14767058.2017.1419181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE Adipose tissue expansion can occur through several different ways and, under certain conditions, can be connected with chronic inflammation. TNF-α is one of the important cytokines involved in this process. Prolonged inflammation in obesity can lead to obesity-related insulin resistance and tissue dysfunction. The aim of our study was to investigate how different combination of maternal and postnatal diet affects offspring adipose tissue morphology and adipose tissue TNF-α expression. METHODS Ten female Sprague Dawley rats, 9 weeks old, were randomly divided into two groups and fed either standard laboratory chow or food rich in saturated fatty acids during 6 weeks and then mated with the same male rat. After birth and lactation male rat offspring from both groups were divided into four subgroups depending on the diet they were fed until 22 weeks old. Samples of white adipose tissue were taken from the subcutaneous, epididymal, and perirenal fat pad. On tissue sections, histomorphometric analysis was conducted using CellProfiler program v 2.1.1, and immunohistochemical staining for TNF-α was performed. RESULTS Greater mean surface area of subcutaneous and epididymal adipocytes was found in groups of male rat offspring with altered diet. In perirenal adipose tissue, the highest number of adipocytes was measured in the group where both mother and offspring were fed a high-fat diet. Adipocyte staining intensity for TNF-α did not differ significantly between the groups. CONCLUSIONS Together with our previously published data, our results lead to the conclusion that alteration of postnatal diet can lead to TNF-α and adipocyte morphology changes.
Collapse
Affiliation(s)
- Darija Šnajder
- a Department of Anatomy and Neuroscience , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia.,b Clinical Institute of Nuclear Medicine and Radiation Protection, University Hospital Osijek , Osijek , Croatia
| | - Željka Perić Kačarević
- a Department of Anatomy and Neuroscience , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia
| | - Anđela Grgić
- a Department of Anatomy and Neuroscience , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia.,c Department of Physical Medicine and Rehabilitation , University Hospital Osijek , Bizovac , Croatia
| | - Nikola Bijelić
- d Department of Histology and Embryology , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia
| | - Matija Fenrich
- a Department of Anatomy and Neuroscience , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia
| | - Tatjana Belovari
- d Department of Histology and Embryology , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia
| | - Radivoje Radić
- a Department of Anatomy and Neuroscience , Josip Juraj Strossmayer University of Osijek , Osijek , Croatia
| |
Collapse
|
32
|
Dalasanur Nagaprashantha L, Adhikari R, Singhal J, Chikara S, Awasthi S, Horne D, Singhal SS. Translational opportunities for broad-spectrum natural phytochemicals and targeted agent combinations in breast cancer. Int J Cancer 2017; 142:658-670. [PMID: 28975625 DOI: 10.1002/ijc.31085] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/18/2017] [Accepted: 09/12/2017] [Indexed: 12/17/2022]
Abstract
Breast cancer (BC) prevention and therapy in the context of life-style risk factors and biological drivers is a major focus of developmental therapeutics in oncology. Obesity, alcohol, chronic estrogen signaling and smoking have distinct BC precipitating and facilitating effects that may act alone or in combination. A spectrum of signaling events including enhanced oxidative stress and changes in estrogen-receptor (ER)-dependent and -independent signaling drive the progression of BC. Breast tumors modulate ERα/ERβ ratio, upregulate proliferative pathways driven by ERα and HER2 with a parallel loss and/or downregulation of tumor suppressors such as TP53 and PTEN which together impact the efficacy of therapeutic strategies and frequently lead to emergence of drug resistance. Natural phytochemicals modulate oxidative stress, leptin, integrin, HER2, MAPK, ERK, Wnt/β-catenin and NFκB signaling along with regulating ERα and ERβ, thereby presenting unique opportunities for both primary and combinatorial interventions in BC. In this regard, this article focuses on critical analyses of the evidence from multiple studies on the efficacy of natural phytochemicals in BC. In addition, areas in which the combinations of such effective natural phytochemicals with approved and/or developing anticancer agents can be translationally beneficial are discussed to derive evidence-based inference for addressing challenges in BC control and therapy.
Collapse
Affiliation(s)
| | | | - Jyotsana Singhal
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Shireen Chikara
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Sanjay Awasthi
- Texas Tech University Health Sciences Center, Lubbock, TX
| | - David Horne
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| | - Sharad S Singhal
- Department of Molecular Medicine, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
33
|
Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 2017; 48:52-108. [PMID: 28816105 DOI: 10.1080/10408444.2017.1351420] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thiazolidinedione (TZD) drugs used in the treatment of type 2 diabetes mellitus (T2DM) have proven effective in improving insulin sensitivity, hyperglycemia, and lipid metabolism. Though well tolerated by some patients, their mechanism of action as ligands of peroxisome proliferator-activated receptors (PPARs) results in the activation of several pathways in addition to those responsible for glycemic control and lipid homeostasis. These pathways, which include those related to inflammation, bone formation, and cell proliferation, may lead to adverse health outcomes. As treatment with TZDs has been associated with adverse hepatic, cardiovascular, osteological, and carcinogenic events in some studies, the role of TZDs in the treatment of T2DM continues to be debated. At the same time, new therapeutic roles for TZDs are being investigated, with new forms and isoforms currently in the pre-clinical phase for use in the prevention and treatment of some cancers, inflammatory diseases, and other conditions. The aims of this review are to provide an overview of the mechanism(s) of action of TZDs, a review of their safety for use in the treatment of T2DM, and a perspective on their current and future therapeutic roles.
Collapse
Affiliation(s)
- Melissa A Davidson
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada
| | - Donald R Mattison
- b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada
| | - Laurent Azoulay
- d Center for Clinical Epidemiology , Lady Davis Research Institute, Jewish General Hospital , Montreal , Canada.,e Department of Oncology , McGill University , Montreal , Canada
| | - Daniel Krewski
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada.,f Faculty of Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
34
|
Han G, Li Y, Cao Y, Yue Z, Zhang Y, Wang L, Liu J. Overexpression of leptin receptor in human glioblastoma: Correlation with vasculogenic mimicry and poor prognosis. Oncotarget 2017; 8:58163-58171. [PMID: 28938545 PMCID: PMC5601641 DOI: 10.18632/oncotarget.17344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 04/11/2017] [Indexed: 01/28/2023] Open
Abstract
Vasculogenic mimicry (VM) was an important tumor blood supply to complement the endothelial cell-dependent angiogenesis, while leptin and receptor (ObR) involved in angiogenesis in glioblastoma has been reported on previous study, but the relationship between ObR expression and VM formation in human glioblastoma tissues, as well as their prognostic significance still remains unclear. In our study, we found that VM recognized by CD31-/PAS+ immunohistochemical staining in glioblastoma tissues showed a positive correlation with leptin expression (r = 0.58, P < 0.01), as well as ObR expression in glioblastoma tissues (r = 0.61, P < 0.01). Association of glial to mesenchymal transition (GMT)-related molecular with ObR expression and VM formation in glioblastoma tissues indicated that ObR-positive glioblastoma cells with GMT phenotype might be more likely to constitute VM, and co-expression of ObR and CD133 or Nestin to constitute the channel impliated that ObR-positive glioblastoma cells displayed glioblastoma stem cells (GSC) properties. Moreover, Kaplan-Meier statistical analysis showed that patients with more VM or ObR expression displayed poorer prognosis for overall survival times than patients with less expression (VMhigh vs. VMlow: P = 0.033; ObRhigh vs. ObRlow: P = 0.009). And ObR+ glioblastoma cells with GSC characteristic were mostly involved in VM formation, whereas a little part of cells were also related to microvascular density (MVD), which suggested that ObR was an important target for anticancer therapy, so further related studies were needed to improve glioblastoma treatment.
Collapse
Affiliation(s)
- Guosheng Han
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yanan Li
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yiqun Cao
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijian Yue
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuhui Zhang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Laixing Wang
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianmin Liu
- Department of Neurosurgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
35
|
Zeinalian R, Farhangi MA, Shariat A, Saghafi-Asl M. The effects of Spirulina Platensis on anthropometric indices, appetite, lipid profile and serum vascular endothelial growth factor (VEGF) in obese individuals: a randomized double blinded placebo controlled trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:225. [PMID: 28431534 PMCID: PMC5399840 DOI: 10.1186/s12906-017-1670-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 03/08/2017] [Indexed: 12/27/2022]
Abstract
Background In recent years, a great attention has been focused on Spirulina platensis as a source of potential valuable nutrients for prevention and treatment of chronic diseases. The objectives of the current study were to determine the effects of Spirulina platensis on anthropometric parameters, serum lipids, appetite and serum Vascular Endothelial Growth Factor (VEGF) in obese individuals. Methods In the current study sixty four obese individuals aged 20–50 years were enrolled and randomly allocated into two groups of intervention and placebo. Intervention group (n = 29) received each 500 mg of the Spirulina platensis a twice-daily dosage while the control group (n = 27) received two pills daily starch for 12 weeks. Anthropometric parameters and serum VEGF and lipid profile were measured in fasting blood samples at the beginning and end of the study period. Dietary intakes were assessed by a 24-h recall method and appetite was measured using standard visual analogue scale (VAS). Results Body weight and body mass index (BMI) were decreased in intervention and placebo treated groups although the mean reduction in Spirulina platensis-treated group was significantly higher (P < 0.05). Serum total cholesterol (TC) significantly reduced in intervention group (P < 0.05). Also, treatment with Spirulina platensis significantly reduced appetite (P = 0.008). Mean serum VEGF, low density lipoprotein-cholesterol, and triglycerides did not change significantly after intervention. Serum high density lipoprotein-cholesterol concentrations (HDL-c) significantly increased in both groups while no difference in mean difference of this change has been observed. Conclusion Spirulina supplementation at a dose of 1 g/d for 12 weeks is effective in modulating body weight and appetite and partly modifies serum lipids. This can further confirm the efficacy of this herbal supplement in control and prevention of obesity and obesity- related disorders. Trial registration Iranian registry of clinical trials (IRCT registration number: IRCT2015071219082N7; Date registered: September 12, 2015).
Collapse
|
36
|
Bariatric Surgery Significantly Reduces Serum Concentration of Vascular Endothelial Growth Factor A and Increases Apelin-12 in Patients with Morbid Obesity. Bariatr Surg Pract Patient Care 2017. [DOI: 10.1089/bari.2016.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
37
|
Matafome P, Rodrigues T, Sena C, Seiça R. Methylglyoxal in Metabolic Disorders: Facts, Myths, and Promises. Med Res Rev 2017; 37:368-403. [PMID: 27636890 DOI: 10.1002/med.21410] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/07/2016] [Accepted: 08/12/2016] [Indexed: 08/26/2024]
Abstract
Glucose and fructose metabolism originates the highly reactive byproduct methylglyoxal (MG), which is a strong precursor of advanced glycation end products (AGE). The MG has been implicated in classical diabetic complications such as retinopathy, nephropathy, and neuropathy, but has also been recently associated with cardiovascular diseases and central nervous system disorders such as cerebrovascular diseases and dementia. Recent studies even suggested its involvement in insulin resistance and beta-cell dysfunction, contributing to the early development of type 2 diabetes and creating a vicious circle between glycation and hyperglycemia. Despite several drugs and natural compounds have been identified in the last years in order to scavenge MG and inhibit AGE formation, we are still far from having an effective strategy to prevent MG-induced mechanisms. This review summarizes the endogenous and exogenous sources of MG, also addressing the current controversy about the importance of exogenous MG sources. The mechanisms by which MG changes cell behavior and its involvement in type 2 diabetes development and complications and the pathophysiological implication are also summarized. Particular emphasis will be given to pathophysiological relevance of studies using higher MG doses, which may have produced biased results. Finally, we also overview the current knowledge about detoxification strategies, including modulation of endogenous enzymatic systems and exogenous compounds able to inhibit MG effects on biological systems.
Collapse
Affiliation(s)
- Paulo Matafome
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
- Department of Complementary Sciences, Coimbra Health School (ESTeSC), Instituto Politécnico de Coimbra, 3045-601, Coimbra, Portugal
| | - Tiago Rodrigues
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Cristina Sena
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Raquel Seiça
- Laboratory of Physiology, Institute of Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
38
|
Abstract
Adipose tissue is an endocrine organ which is responsible for postprandial uptake of glucose and fatty acids, consequently producing a broad range of adipokines controlling several physiological functions like appetite, insulin sensitivity and secretion, immunity, coagulation, and vascular tone, among others. Many aspects of adipose tissue pathophysiology in metabolic diseases have been described in the last years. Recent data suggest two main factors for adipose tissue dysfunction: accumulation of nonesterified fatty acids and their secondary products and hypoxia. Both of these factors are thought to be on the basis of low-grade inflammatory activation, further increasing metabolic dysregulation in adipose tissue. In turn, inflammation is involved in the inhibition of substrate uptake, alteration of the secretory profile, stimulation of angiogenesis, and recruitment of further inflammatory cells, which creates an inflammatory feedback in the tissue and is responsible for long-term establishment of insulin resistance.
Collapse
Affiliation(s)
- Paulo Matafome
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- Department of Complementary Sciences, Coimbra Health School (ESTeSC), Instituto Politécnico de Coimbra, Coimbra, Portugal.
| | - Raquel Seiça
- Institute of Physiology, Institute for Biomedical Imaging and Life Sciences-IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
39
|
Amjadi F, Mehdipoor R, Zarkesh-Esfahani H, Javanmard SH. Leptin serves as angiogenic/mitogenic factor in melanoma tumor growth. Adv Biomed Res 2016; 5:127. [PMID: 27563637 PMCID: PMC4976532 DOI: 10.4103/2277-9175.187005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022] Open
Abstract
Background: Tumor development is angiogenesis dependent. There is evidence that leptin contributes to tumor growth. However, all the mechanisms by which leptin does this has not been clearly established. The objective of the present study was to test the hypothesis that leptin enhances melanoma tumor growth through inducing angiogenesis and cell proliferation. Materials and Methods: We injected 2 × 106 B16F10 melanoma cells subcutaneously to 32 C57BL6 mice. The mice were randomly divided into four groups of eight animals, on day 8. Two groups received twice daily intraperitoneal (i.p.) injections of either phosphate buffered saline or recombinant murine leptin (1 μg/g initial body weight). Two groups received i.p. injections of either 9F8 an anti leptin receptor antibody or the control mouse IgG at 50 μg/injection every 3 consecutive days. By the end of the 2nd week, the animals were euthanized and blood samples and tumors were analyzed. Angiogenesis and proliferation were assessed by immunohistochemical staining for CD31 and Ki-67 respectively. Results: Tumors size, capillary density, plasma levels of vascular endothelial growth factor, and the number of Ki-67-positive stained cells were significantly more in the leptin than 9F8 and both control groups (P < 0.05). Conclusion: Taken together, our findings reinforce the idea that leptin acts as an angiogenic and mitogenic factor to promote melanoma growth.
Collapse
Affiliation(s)
- Fatemehsadat Amjadi
- Department of Physiology, Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Anatomy, Tehran University of Medical Science, Tehran, Iran
| | - Roshanak Mehdipoor
- Isfahan Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Zarkesh-Esfahani
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran; Department of Immunology, Medical School, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Department of Physiology, School of Medicine, Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
Esophageal adenocarcinoma and obesity: peritumoral adipose tissue plays a role in lymph node invasion. Oncotarget 2016; 6:11203-15. [PMID: 25857300 PMCID: PMC4484450 DOI: 10.18632/oncotarget.3587] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/19/2015] [Indexed: 12/15/2022] Open
Abstract
Obesity is associated with cancer risk in esophageal adenocarcinoma (EAC). Adipose tissue directly stimulates tumor progression independently from body mass index (BMI), but the mechanisms are not fully understood. We studied the morphological, histological and molecular characteristics of peritumoral and distal adipose tissue of 60 patients with EAC, to investigate whether depot-specific differences affect tumor behavior. We observed that increased adipocyte size (a hallmark of obesity) was directly associated with leptin expression, angiogenesis (CD31) and lymphangiogenesis (podoplanin); however, these parameters were associated with nodal metastasis only in peritumoral but not distal adipose tissue of patients. We treated OE33 cells with conditioned media (CM) collected from cultured biopsies of adipose tissue and we observed increased mRNA levels of leptin and adiponectin receptors, as well as two key regulator genes of epithelial-to-mesenchymal transition (EMT): alpha-smooth muscle actin (α-SMA) and E-cadherin. This effect was greater in cells treated with CM from peritumoral adipose tissue of patients with nodal metastasis and was partially blunted by a leptin antagonist. Therefore, peritumoral adipose tissue may exert a direct effect on the progression of EAC by secreting depot-specific paracrine factors, and leptin is a key player in this crosstalk.
Collapse
|
41
|
Dodson MV, Allen RE, Du M, Bergen WG, Velleman SG, Poulos SP, Fernyhough-Culver M, Wheeler MB, Duckett SK, Young MRI, Voy BH, Jiang Z, Hausman GJ. INVITED REVIEW: Evolution of meat animal growth research during the past 50 years: Adipose and muscle stem cells. J Anim Sci 2016; 93:457-81. [PMID: 26020737 DOI: 10.2527/jas.2014-8221] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
If one were to compare today's animal growth research to research from a mere 50 yr ago, one would see programs with few similarities. The evolution of this research from whole-animal through cell-based and finally molecular and genomic studies has been enhanced by the identification, isolation, and in vitro evaluation of adipose- and muscle-derived stem cells. This paper will highlight the struggles and the milestones that make this evolving area of research what it is today. The contribution of adipose and muscle stem cell research to development and growth, tissue regeneration, and final carcass composition are reviewed.
Collapse
|
42
|
Picon-Ruiz M, Pan C, Drews-Elger K, Jang K, Besser AH, Zhao D, Morata-Tarifa C, Kim M, Ince TA, Azzam DJ, Wander SA, Wang B, Ergonul B, Datar RH, Cote RJ, Howard GA, El-Ashry D, Torné-Poyatos P, Marchal JA, Slingerland JM. Interactions between Adipocytes and Breast Cancer Cells Stimulate Cytokine Production and Drive Src/Sox2/miR-302b-Mediated Malignant Progression. Cancer Res 2016; 76:491-504. [PMID: 26744520 DOI: 10.1158/0008-5472.can-15-0927] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 10/20/2015] [Indexed: 11/16/2022]
Abstract
Consequences of the obesity epidemic on cancer morbidity and mortality are not fully appreciated. Obesity is a risk factor for many cancers, but the mechanisms by which it contributes to cancer development and patient outcome have yet to be fully elucidated. Here, we examined the effects of coculturing human-derived adipocytes with established and primary breast cancer cells on tumorigenic potential. We found that the interaction between adipocytes and cancer cells increased the secretion of proinflammatory cytokines. Prolonged culture of cancer cells with adipocytes or cytokines increased the proportion of mammosphere-forming cells and of cells expressing stem-like markers in vitro. Furthermore, contact with immature adipocytes increased the abundance of cancer cells with tumor-forming and metastatic potential in vivo. Mechanistic investigations demonstrated that cancer cells cultured with immature adipocytes or cytokines activated Src, thus promoting Sox2, c-Myc, and Nanog upregulation. Moreover, Sox2-dependent induction of miR-302b further stimulated cMYC and SOX2 expression and potentiated the cytokine-induced cancer stem cell-like properties. Finally, we found that Src inhibitors decreased cytokine production after coculture, indicating that Src is not only activated by adipocyte or cytokine exposures, but is also required to sustain cytokine induction. These data support a model in which cancer cell invasion into local fat would establish feed-forward loops to activate Src, maintain proinflammatory cytokine production, and increase tumor-initiating cell abundance and metastatic progression. Collectively, our findings reveal new insights underlying increased breast cancer mortality in obese individuals and provide a novel preclinical rationale to test the efficacy of Src inhibitors for breast cancer treatment.
Collapse
Affiliation(s)
- Manuel Picon-Ruiz
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain. Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, Granada, Spain
| | - Chendong Pan
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Katherine Drews-Elger
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Kibeom Jang
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Alexandra H Besser
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Donald and Sheila Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Dekuang Zhao
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Donald and Sheila Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Cynthia Morata-Tarifa
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain. Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, Granada, Spain
| | - Minsoon Kim
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Tan A Ince
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida. Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Diana J Azzam
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Seth A Wander
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Donald and Sheila Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Bin Wang
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida. Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Burcu Ergonul
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida. Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Ram H Datar
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida. Biomedical Nanoscience Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Richard J Cote
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida. Biomedical Nanoscience Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Guy A Howard
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida. Geriatric Research, Education and Clinical Center, Bruce W. Carter Veterans Affairs Medical Center, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Dorraya El-Ashry
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Pablo Torné-Poyatos
- Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, Granada, Spain. Department of Surgery, San Cecilio University Hospital, University of Granada, Granada, Spain. Department of Mammary Pathology, San Cecilio University Hospital, University of Granada, Granada, Spain
| | - Juan A Marchal
- Biopathology and Medicine Regenerative Institute (IBIMER), University of Granada, Granada, Spain. Biosanitary Institute of Granada (ibs. GRANADA), University of Granada, Granada, Spain. Department of Human Anatomy and Embryology, University of Granada, Granada, Spain
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, UM Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida. Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida. Donald and Sheila Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida. Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
43
|
Shouman S, Wagih M, Kamel M. Leptin influences estrogen metabolism and increases DNA adduct formation in breast cancer cells. Cancer Biol Med 2016; 13:505-513. [PMID: 28154783 PMCID: PMC5250609 DOI: 10.20892/j.issn.2095-3941.2016.0079] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective: The elevated incidence of obesity has been paralleled with higher risks of breast cancer. High adiposity increases leptin secretion from adipose tissue, which in turn increases cancer cell proliferation. The interplay between leptin and estrogen is one of the mechanisms through which leptin influences breast carcinogenesis. An unbalanced estrogen metabolism increases the formations of catechol estrogen quinones, DNA adducts, and cancer mutations. This study aims to investigate the effect of leptin on some estrogen metabolic enzymes and DNA adduction in breast cancer cells. Methods: High performance liquid chromatography (HPLC) was performed to analyze the DNA adducts 4-OHE1[E2]-1-N3 adenine and 4-OHE1[E2]-1-N7 guanine. Reporter gene assay, real time reverse transcription polymerase chain reaction (real time RT-PCR), and Western blot were used to assess the expression of estrogen metabolizing genes and enzymes: Cytochrome P-450 1B1 (CYP1B1), Nicotinamide adenine dinucleotide phosphate-quinone oxidoreductase1 (NQO1), and Catechol-O-methyl transferase (COMT). Results: Leptin significantly increased the DNA adducts 4-OHE1[E2]-1-N3 adenine and 4-OHE1[E2]-1-N7 guanine. Furthermore, leptin significantly upregulated CYP1B1 promoter activity and protein expression. The luciferase promoter activities of NQO1 and mRNA levels were significantly reduced. Moreover, leptin greatly reduced the reporter activities of the COMT-P1 and COMT-P2 promoters and diminished the protein expression of COMT. Conclusions: Leptin increases DNA adduct levels in breast cancer cells partly by affecting key genes and enzymes involved in estrogen metabolism. Thus, increased focus should be directed toward leptin and its effects on the estrogen metabolic pathway as an effective approach against breast cancer.
Collapse
Affiliation(s)
- Samia Shouman
- Department of Cancer Biology, Unit of Pharmacology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Mohamed Wagih
- Department of Pathology, Faculty of Medicine, Beni-Suef University, Benisuef 62511, Egypt
| | - Marwa Kamel
- Department of Cancer Biology, Unit of Pharmacology, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| |
Collapse
|
44
|
Swisher AK, Abraham J, Bonner D, Gilleland D, Hobbs G, Kurian S, Yanosik MA, Vona-Davis L. Exercise and dietary advice intervention for survivors of triple-negative breast cancer: effects on body fat, physical function, quality of life, and adipokine profile. Support Care Cancer 2015; 23:2995-3003. [PMID: 25724409 PMCID: PMC4624214 DOI: 10.1007/s00520-015-2667-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 02/15/2015] [Indexed: 01/19/2023]
Abstract
PURPOSE Regular exercise and healthy eating are routinely recommended for breast cancer survivors, and past studies show benefits in quality of life and decreased inflammation. However, this has not been tested specifically in triple-negative breast cancer survivors. Increasing physical activity and losing body fat are thought to positively affect inflammatory biomarkers that have been associated with breast cancer. Therefore, the primary purpose of this study was to determine if participation in an exercise and dietary counseling program can improve body fat, physical function, and quality of life in survivors of this aggressive breast cancer. Secondarily, we sought to determine if participation in the program had beneficial effects on obesity-related markers of the adipokine profile. METHODS Sixty-six survivors of triple-negative breast cancer with BMI >25 were invited to participate. Twenty-eight enrolled and 23 completed the randomized, controlled trial (13 intervention, 10 control). Moderate-intensity aerobic exercise (150 min per week, for 12 weeks) and diet counseling were compared to usual care, education only. The primary outcome of interest was weight loss (body mass, BMI, % fat), and secondary outcomes included physical function (exercise capacity), quality of life (Function After Cancer Therapy-Breast (FACT-B)), cytokines (C-reactive protein (CRP), TNF-α, IL-6), and adipokine profile (leptin, adiponectin, insulin). RESULTS Participants in the program lost more body fat (2.4 % loss vs. 0.4 % gain, p < 0.05) than the control group. The intervention group also improved quality of life (FACT-B total score +14 pts) and decreased sedentary time but did not improve peak exercise capacity. The intervention had no effect on serum cytokines and adipokines after 12 weeks in the program. However, serum leptin and adiponectin and their ratio were significantly correlated with BMI in the intervention group (p < 0.05). CONCLUSIONS Exercise and dietary counseling led to loss of body fat and improved quality of life in survivors of triple-negative breast cancer. BMI was associated with favorable changes in leptin and adiponectin which may reflect a change in adiposity with intervention. Exercise and healthy eating may be equally effective in this high-risk population as in other breast cancer survivors and should be encouraged as a part of a cancer survivorship program.
Collapse
Affiliation(s)
- Anne K Swisher
- Division of Physical Therapy, West Virginia University, PO Box 9226, Morgantown, WV, 26506-9226, USA,
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Chettouh H, Lequoy M, Fartoux L, Vigouroux C, Desbois-Mouthon C. Hyperinsulinaemia and insulin signalling in the pathogenesis and the clinical course of hepatocellular carcinoma. Liver Int 2015; 35:2203-17. [PMID: 26123841 DOI: 10.1111/liv.12903] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/09/2015] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer and is one of the leading causes of cancer-related death. The risk factors for HCC include cirrhosis, chronic viral hepatitis, heavy alcohol intake and metabolic diseases such as obesity, type 2 diabetes and metabolic syndrome. Insulin resistance is a common denominator of all of these conditions and is tethered to hyperinsulinaemia. Here, we give an overview of the recent advances linking hyperinsulinaemia to HCC development and progression. In particular, we summarise the underlying causes of hyperinsulinaemia in the setting of chronic liver diseases. We present epidemiological evidence linking metabolic diseases to HCC risk and HCC-related mortality, as well as the pathogenic cellular and molecular mechanisms explaining this relation. A better understanding of the mechanisms by which insulin participates in HCC biology might ultimately provide novel opportunities for prevention and treatment.
Collapse
Affiliation(s)
- Hamza Chettouh
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Marie Lequoy
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Laetitia Fartoux
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Corinne Vigouroux
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Laboratoire Commun de Biologie et Génétique Moléculaires AP-HP, Hôpital Saint-Antoine, Paris, France.,ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Christèle Desbois-Mouthon
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| |
Collapse
|
46
|
Cowen S, McLaughlin SL, Hobbs G, Coad J, Martin KH, Olfert IM, Vona-Davis L. High-Fat, High-Calorie Diet Enhances Mammary Carcinogenesis and Local Inflammation in MMTV-PyMT Mouse Model of Breast Cancer. Cancers (Basel) 2015; 7:1125-42. [PMID: 26132316 PMCID: PMC4586761 DOI: 10.3390/cancers7030828] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 06/16/2015] [Accepted: 06/18/2015] [Indexed: 01/26/2023] Open
Abstract
Epidemiological studies provide strong evidence that obesity and the associated adipose tissue inflammation are risk factors for breast cancer; however, the molecular mechanisms are poorly understood. We evaluated the effect of a high-fat/high-calorie diet on mammary carcinogenesis in the immunocompetent MMTV-PyMT murine model. Four-week old female mice (20/group) were randomized to receive either a high-fat (HF; 60% kcal as fat) or a low-fat (LF; 16% kcal) diet for eight weeks. Body weights were determined, and tumor volumes measured by ultrasound, each week. At necropsy, the tumors and abdominal visceral fat were weighed and plasma collected. The primary mammary tumors, adjacent mammary fat, and lungs were preserved for histological and immunohistochemical examination and quantification of infiltrating macrophages, crown-like structure (CLS) formation, and microvessel density. The body weight gains, visceral fat weights, the primary mammary tumor growth rates and terminal weights, were all significantly greater in the HF-fed mice. Adipose tissue inflammation in the HF group was indicated by hepatic steatosis, pronounced macrophage infiltration and CLS formation, and elevations in plasma monocyte chemoattractant protein-1 (MCP-1), leptin and proinflammatory cytokine concentrations. HF intake was also associated with higher tumor-associated microvascular density and the proangiogenic factor MCP-1. This study provides preclinical evidence in a spontaneous model of breast cancer that mammary adipose tissue inflammation induced by diet, enhances the recruitment of macrophages and increases tumor vascular density suggesting a role for obesity in creating a microenvironment favorable for angiogenesis in the progression of breast cancer.
Collapse
Affiliation(s)
- Sarah Cowen
- Department of Surgery, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Sarah L McLaughlin
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Gerald Hobbs
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Department of Statistics, West Virginia University, Morgantown, WV 26506, USA.
| | - James Coad
- Department of Pathology, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Karen H Martin
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Department of Neurobiology and Anatomy, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - I Mark Olfert
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Department of Human Performance and Exercise Physiology, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| | - Linda Vona-Davis
- Department of Surgery, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
- Mary Babb Randolph Cancer Center, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| |
Collapse
|
47
|
|
48
|
Buckland G, Travier N, Agudo A. The role of diet, weight control and physical activity in breast cancer survivors. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.14.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Recent advances in breast cancer (BC) treatment and improved screening have resulted in an increasing number of BC survivors. However, since recurrences are still a relatively common event there is a critical need to investigate modifiable factors that could impact disease recurrence and long-term prognosis. There is substantial evidence from observational studies and increasingly from randomized controlled trials, showing that weight management, increased physical activity and dietary modification may be effective methods to improve BC survival and reduce recurrences, due to their interrelated beneficial effects on systemic inflammation, circulating reproductive hormones and metabolic imbalances. Although ongoing randomized controlled trials should be able to confirm the role of these lifestyle factors on BC prognosis, further research is needed to establish specific lifestyle recommendations.
Collapse
Affiliation(s)
- Genevieve Buckland
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Noemie Travier
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Agudo
- Unit of Nutrition & Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| |
Collapse
|
49
|
Zhang J, Jin HC, Zhu AK, Ying RC, Wei W, Zhang FJ. Prognostic significance of plasma chemerin levels in patients with gastric cancer. Peptides 2014; 61:7-11. [PMID: 25152503 DOI: 10.1016/j.peptides.2014.08.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 08/14/2014] [Accepted: 08/14/2014] [Indexed: 12/27/2022]
Abstract
Chemerin is a novel adipokine, which is linked to adipogenesis and chemotaxis of the innate immune system. This study aimed to evaluate the relationship between preoperative plasma chemerin level and prognosis of gastric cancers. One hundred ninety-six patients and 196 age- and gender-matched healthy individuals were recruited. Fasting venous blood samples were collected 2 days prior to surgery for the gastric cancer patients and at the physical examination day for the healthy volunteers. Recorded clinicopathological information included invasion depth, lymph node metastasis, distant metastasis, peritoneal dissemination, tumor size and tumor-node metastasis stage. Plasma chemerin levels were determined using enzyme-linked immunosorbent assay. Plasma chemerin levels were statistically significantly in all patients than in healthy controls (53.1 ± 19.0 ng/mL vs. 31.3 ± 11.3 ng/mL; P < 0.001). And it was identified as an independent predictor for 5-year mortality [odds ratio (OR), 2.718; 95% confidence interval (CI), 1.201-4.229; P = 0.005) and adverse event (OR, 2.982; 95% CI, 1.223-4.879; P = 0.003) of gastric cancer, and had high area under receiver operating characteristic curve (AUC) for prediction of 5-year mortality (AUC, 0.808; 95% CI, 0.745-0.860) and adverse event (AUC, 0.787; 95% CI, 0.723-0.842). It also emerged as an independent predictor for overall survival (Hazard ratios, 1.788; 95% CI, 1.200-2.663; P = 0.002) and disease-free survival (Hazard ratios, 2.016; 95% CI, 1.312-3.125; P = 0.004). Thus, our results suggest that high plasma chemerin levels are associated with long-term poor prognosis and survival of gastric cancer as well as may play a role as prognostic biomarker in gastric cancer survival.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China
| | - Hui-Cheng Jin
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China
| | - A-Kao Zhu
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China
| | - Rong-Chao Ying
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China.
| | - Wei Wei
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China
| | - Fang-Jie Zhang
- Department of Gastroenterological Surgery, The Hangzhou First People's Hospital, Nanjing Medical University Affiliated Hangzhou Hospital, 261 Huansha Road, Hangzhou 310006, China
| |
Collapse
|
50
|
Yang WH, Chen JC, Hsu KH, Lin CY, Wang SW, Wang SJ, Chang YS, Tang CH. Leptin increases VEGF expression and enhances angiogenesis in human chondrosarcoma cells. Biochim Biophys Acta Gen Subj 2014; 1840:3483-93. [PMID: 25230157 DOI: 10.1016/j.bbagen.2014.09.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Leptin, 16kDa product of obese gene, is adipocytokine playing critical role in regulation of body weight. In recent years, leptin is also defined as potent angiogenic factor involving in tumorigenesis, angiogenesis, and metastasis. However, it is unknown whether leptin regulates VEGF production in human chondrosarcoma and contributing the tumor-associated angiogenesis. METHODS We analyzed protein level of leptin and VEGF in human chondrosarcoma tissues. Effects of leptin on chondrosarcoma cells were examined by in vitro and in vivo assays. In addition, intracellular signal pathways were investigated by pharmacological and genetic approaches. RESULTS We found that both leptin and VEGF are highly expressed in human chondrosarcoma tissues, and positively correlated with tumor stage. Leptin increases VEGF production by activating OBRl receptor and MAPKs (p38, ERK, and JNK), which in turn enhances binding of AP-1 transcription factor to VEGF promoter, resulting in the transactivation of VEGF expression and subsequently promoting migration and tube formation in endothelial progenitor cells (EPCs). In vivo, knockdown leptin significantly reduces angiogenesis and tumor growth. CONCLUSION Leptin may be a therapeutic target of angiogenesis and metastasis in chondrosarcoma. GENERAL SIGNIFICANCE These findings provide better understanding of pathogenesis of chondrosarcoma and can utilize this knowledge to design new therapeutic strategy.
Collapse
Affiliation(s)
- Wei-Hung Yang
- Department of Orthopedic Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan; School of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Kai-Hsiang Hsu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Shoou-Jyi Wang
- Department of Orthopedic Surgery, Chang-Hua Hospital, Ministry of Health and Welfare, Puhsin Township, Changhua County, Taiwan
| | - Yung-Sen Chang
- Department of Orthopedic Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|