1
|
Cancel M, Crottes D, Bellanger D, Bruyère F, Mousset C, Pinault M, Mahéo K, Fromont G. Variable effects of periprostatic adipose tissue on prostate cancer cells: Role of adipose tissue lipid composition and cancer cells related factors. Prostate 2024; 84:358-367. [PMID: 38112233 DOI: 10.1002/pros.24655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Periprostatic adipose tissue (PPAT) is likely to modulate prostate cancer (PCa) progression. We analyzed the variations in the effect of PPAT on cancer cells, according to its fatty acid (FA) composition and tumor characteristics. METHODS The expression of markers of aggressiveness Ki67 and Zeb1, and epigenetic marks that could be modified during PCa progression, was analyzed by immunohistochemistry on a tissue-micro-array containing 59 pT3 PCa, including intra-prostatic areas and extra-prostatic foci in contact with PPAT belonging to the same tumor. In addition, we cocultivated PC3 and LNCaP cell lines with PPAT, which were then analyzed for FA composition. RESULTS Although the contact between PPAT and cancer cells led overall to an increase in Ki67 and Zeb1, and a decrease in the epigenetic marks 5MC, 5HMC, and H3K27ac, these effects were highly heterogeneous. Increased proliferation in extra-prostatic areas was associated with the international society of uropathology score. PC3 and LNCaP cocultures with PPAT led to increased Ki67, Zeb1 and H3K27me3, but only for PPAT associated with aggressive PCa. PC3 proliferation was correlated with high 20.2 n-6 and low 20.5n-3 in PPAT. CONCLUSIONS These results suggest that the effects of PPAT on cancer cells may depend on both PCa characteristics and PPAT composition, and could lead to propose nutritional supplementation.
Collapse
Affiliation(s)
- Mathilde Cancel
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Medical Oncology, CHU Tours, Tours, France
| | - David Crottes
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Dorine Bellanger
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | | | - Coralie Mousset
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Pathology, CHU Tours, Tours, France
| | - Michelle Pinault
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Karine Mahéo
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
| | - Gaëlle Fromont
- Faculté de Médecine, Inserm UMR1069 "Nutrition, Croissance et Cancer" Université François Rabelais, Tours, France
- Department of Pathology, CHU Tours, Tours, France
| |
Collapse
|
2
|
Mylod E, O'Connell F, Donlon NE, Davern M, Marion C, Butler C, Reynolds JV, Lysaght J, Conroy MJ. Real-time ex vivo monitoring of NK cell migration toward obesity-associated oesophageal adenocarcinoma following modulation of CX3CR1. Sci Rep 2024; 14:4017. [PMID: 38369570 PMCID: PMC10874956 DOI: 10.1038/s41598-024-54390-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
Oesophagogastric adenocarcinomas (OAC) are poor prognosis, obesity-associated cancers which may benefit from natural killer (NK) cell-based immunotherapies. Cellular immunotherapies encounter two key challenges to their success in OAC, namely recruitment to extratumoural tissues such as the omentum at the expense of the tumour and an immunosuppressive tumour microenvironment (TME) which can hamper NK cell function. Herein, we examined approaches to overcome the detrimental impact of obesity on NK cells and NK cell-based immunotherapies. We have demonstrated that NK cells migrate preferentially to the chemotactic signals of OAC patient-derived omentum over tumour in an ex vivo model of immune cell migration. We have identified CX3CR1 modulation and/or tumour chemokine profile remodelling as approaches to skew NK cell migration towards tumour. We also report targetable immunosuppressive facets of the obese OAC TME which dampen NK cell function, in particular cytotoxic capabilities. These data provide insights into approaches to therapeutically overcome key challenges presented by obesity and will inform superior design of NK cell-based immunotherapies for OAC.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology Research Group, Department of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Fiona O'Connell
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Caroline Marion
- Cancer Immunology Research Group, Department of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Christine Butler
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - John V Reynolds
- Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, School of Medicine, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Melissa J Conroy
- Cancer Immunology Research Group, Department of Anatomy, School of Medicine, Trinity Biomedical Sciences Institute and Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
3
|
Karaçelik T, Kaya B, Korkmaz M, Karaağaç M, Araz M, Eryılmaz MK, Bozcuk HŞ, Artaç M. Prognostic Significance of Adipose Tissue Distribution and Metabolic Activity in PET/CT in Patients with Metastatic Colorectal Cancer. J Gastrointest Cancer 2023; 54:456-466. [PMID: 35404000 DOI: 10.1007/s12029-022-00819-x] [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] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE In this study, we aimed to evaluate the prognostic significance of adipose tissue distribution and metabolic activity in PET/CT to predict survival in patients with metastatic colorectal cancer (mCRC). METHODS The volume, density (HU), and FDG uptake (standardized uptake value (SUV)) of visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) and maximum FDG uptake of the tumor tissue were measured. Subcutaneous adipose tissue of volume-to-density ratio (SAT ratio) was calculated. RESULTS The median OS for the patients with SAT ratio value < -1.1 and ≥ -1.1 were 38.5 (95% CI 31.54-45.58) and 24.5 (95% CI 14.13-34.93) months, respectively (p = 0.05). During follow-up, 69 patients experienced disease progression. The median progression-free survival (PFS) was 11.03 months (95% CI: 9.11-12.95). Median PFS for patients with tumor SUV max value < 11.5 and ≥ 11.5 were 9.2 (95% CI 7.25-11.27) and 12.6 (95% CI 10.02-15.27) months, respectively (p = 0.14). Forty-eight patients received bevacizumab therapy. VAT SUV mean (HR: 0.09; 95% CI 0.01-0.52, p = 0.008) was significantly associated with PFS in patients receiving bevacizumab. SAT ratio was the significant parameter for the OS (HR: 0.58; 95% CI 0.33-1.01, p = 0.05) and PFS (HR: 1.99; 95% CI 1.02-3.91, p = 0.043). CONCLUSIONS SAT ratio was an independent prognostic factor for survival in patients with mCRC. Higher SAT volume is correlated with longer survival in mCRC patients.
Collapse
Affiliation(s)
- Tuba Karaçelik
- Department of Internal Medicine, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Buğra Kaya
- Department of Nuclear Medicine, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Mustafa Korkmaz
- Department of Medical Oncology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey.
| | - Mustafa Karaağaç
- Department of Medical Oncology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Murat Araz
- Department of Medical Oncology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Melek Karakurt Eryılmaz
- Department of Medical Oncology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hakan Şat Bozcuk
- Department of Medical Oncology, Medical Park Antalya Hospital, Antalya, Turkey
| | - Mehmet Artaç
- Department of Medical Oncology, Meram Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| |
Collapse
|
4
|
Xiang S, Yang YK, Wang TY, Yang ZT, Lu Y, Liu SL. Development and validation of a nomogram to predict anastomotic leakage in colorectal cancer based on CT body composition. Front Nutr 2022; 9:974903. [PMID: 36159450 PMCID: PMC9490075 DOI: 10.3389/fnut.2022.974903] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Background Anastomotic leakage (AL) is one of the most serious postoperative complications. This study aimed to investigate the predictive value of preoperative body composition for AL in patients with colorectal cancer (CRC). Methods We first reviewed data from 3,681 patients who underwent radical CRC resection 2013–2021 in our hospital, and 60 patients were diagnosed with AL after surgery. We designed a nested case-control study and two controls were randomly selected for each case according to the time and position of surgery. Body composition was measured at the level of the third lumbar vertebra based on computed tomography (CT) images. The risk factors of AL were analyzed by univariate and multivariate analysis. Nomogram was built using binary regression analysis and assessed for clinical usefulness, calibration, and discrimination. Results In the multivariate analysis, gender, blood glucose, nutrition risk screening (NRS), skeletal muscle area (SMA) and visceral fat area (VFA) were independent risk factors for developing anastomotic leakage after surgery. The prognostic model had an area under the receiver operating characteristic curve of 0.848 (95% CI, 0.781–0.914). The calibration curve showed good consistency between the predicted and observed outcomes. Decision curve analysis indicated that patients with colorectal cancer can benefit from the prediction model. Conclusions The nomogram that combined with gender, blood glucose, NRS, SMA, and VFA had good predictive accuracy and reliability to AL. It may be conveniently for clinicians to predict AL preoperatively and be useful for guiding treatment decisions.
Collapse
Affiliation(s)
- Shuai Xiang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yong-Kang Yang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong-Yu Wang
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhi-Tao Yang
- Department of Radiology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yun Lu
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yun Lu
| | - Shang-Long Liu
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
- Shang-Long Liu
| |
Collapse
|
5
|
Saha A, Hamilton-Reeves J, DiGiovanni J. White adipose tissue-derived factors and prostate cancer progression: mechanisms and targets for interventions. Cancer Metastasis Rev 2022; 41:649-671. [PMID: 35927363 PMCID: PMC9474694 DOI: 10.1007/s10555-022-10056-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022]
Abstract
Obesity represents an important risk factor for prostate cancer, driving more aggressive disease, chemoresistance, and increased mortality. White adipose tissue (WAT) overgrowth in obesity is central to the mechanisms that lead to these clinical observations. Adipose stromal cells (ASCs), the progenitors to mature adipocytes and other cell types in WAT, play a vital role in driving PCa aggressiveness. ASCs produce numerous factors, especially chemokines, including the chemokine CXCL12, which is involved in driving EMT and chemoresistance in PCa. A greater understanding of the impact of WAT in obesity-induced progression of PCa and the underlying mechanisms has begun to provide opportunities for developing interventional strategies for preventing or offsetting these critical events. These include weight loss regimens, therapeutic targeting of ASCs, use of calorie restriction mimetic compounds, and combinations of compounds as well as specific receptor targeting strategies.
Collapse
Affiliation(s)
- Achinto Saha
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Jill Hamilton-Reeves
- Departments of Urology and Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78723, USA.
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX, 78723, USA.
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, 78723, USA.
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA.
| |
Collapse
|
6
|
Luo X, Li RR, Li YQ, Yu HP, Yu HN, Jiang WG, Li YN. Reducing VEGFB expression regulates the balance of glucose and lipid metabolism in mice via VEGFR1. Mol Med Rep 2022; 26:285. [PMID: 35894135 PMCID: PMC9366154 DOI: 10.3892/mmr.2022.12801] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/17/2022] [Indexed: 11/05/2022] Open
Abstract
In recent years, studies have demonstrated that vascular endothelial growth factor B (VEGFB) can affect the metabolism of fatty acids and glucose, and it is expected to become a target for the diagnosis and treatment of metabolic diseases such as obesity and diabetes. At present, the specific mechanism that VEGFB regulates lipid and glucose metabolism balance is not completely understood. The present study used systemic VEGFB gene-knockout mice to investigate the effects of downregulation of the VEGFB gene on lipid metabolism and insulin secretion, and to explore the mechanism of the VEGFB pathway involved in the regulation of glucose and lipid metabolism. The morphological changes in the liver and pancreas of mice after VEGFB gene deletion were observed under a light microscope and a scanning electron microscope, and the effects of VEGFB gene deletion on lipid metabolism and blood glucose balance were detected by a serological technique. The detection indexes included total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol. Simultaneously, fasting blood glucose, glycosylated hemoglobin A1c (HbA1c), fasting insulin and glucagon were measured. Insulin sensitivity was assessed by using the insulin tolerance tests and glucose tolerance tests, and function of β-cell islets was evaluated by using the insulin resistance index (HOMA-IR) and pancreatic β-cell secretion index (HOMA-β). Τhe protein expression changes of vascular endothelial growth factor receptor 1 (VEGFR1) and vascular endothelial growth factor receptor 2 (VEGFR2) in mouse islets were detected by western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) after the VEGFB gene was knocked down to analyze the mechanism of VEGFB that may be involved in glucose and lipid metabolism. It was observed that after VEGFB was knocked down, mouse hepatocytes exhibited steatosis and increased secretory vesicles in islet cells. The lipid metabolism indexes such as TG, TC and LDL increased significantly; however, the levels of FBS, postprandial blood glucose and HbA1c decreased, whereas the glucose tolerance increased. Serum insulin secretion increased and HOMA-IR decreased since VEGFB was knocked down. Western blotting and RT-qPCR results revealed that the expression levels of VEGFR1 and neuropilin-1 decreased after the VEGFB gene was knocked down, while the expression levels of VEGFA and VEGFR2 increased. The absence of VEGFB may be involved in the regulation of glucose and lipid metabolism in mice by activating the VEGFA/VEGFR2 signaling pathway. VEGFB is expected to become a new target for the treatment of metabolic diseases such as obesity and diabetes. At present, the mechanism of VEGFB involved in regulating lipid metabolism and glucose metabolism is not completely clear. It was identified that downregulating VEGFB improved lipid metabolism and insulin resistance. The role of VEGFB/VEGFR1 pathway and other family members in regulating glucose and lipid metabolism was detected, which provided a theoretical and experimental basis for VEGFB to affect the regulation of glucose and lipid metabolism balance.
Collapse
Affiliation(s)
- Xu Luo
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Rong-Rong Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yu-Qi Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Han-Pu Yu
- Clinical Medicine, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Hai-Ning Yu
- Department of Stomatology Medicine, School of Oral Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wen-Guo Jiang
- Department of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ya-Na Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
7
|
Properties of Leukemic Stem Cells in Regulating Drug Resistance in Acute and Chronic Myeloid Leukemias. Biomedicines 2022; 10:biomedicines10081841. [PMID: 36009388 PMCID: PMC9405586 DOI: 10.3390/biomedicines10081841] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Notoriously known for their capacity to reconstitute hematological malignancies in vivo, leukemic stem cells (LSCs) represent key drivers of therapeutic resistance and disease relapse, posing as a major medical dilemma. Despite having low abundance in the bulk leukemic population, LSCs have developed unique molecular dependencies and intricate signaling networks to enable self-renewal, quiescence, and drug resistance. To illustrate the multi-dimensional landscape of LSC-mediated leukemogenesis, in this review, we present phenotypical characteristics of LSCs, address the LSC-associated leukemic stromal microenvironment, highlight molecular aberrations that occur in the transcriptome, epigenome, proteome, and metabolome of LSCs, and showcase promising novel therapeutic strategies that potentially target the molecular vulnerabilities of LSCs.
Collapse
|
8
|
Abstract
Two decades of research have established that Nuclear Factor-κB (NF-κB) signaling plays a critical role in reprogramming the fat cell transcriptome towards inflammation in response to overnutrition and metabolic stress. Several groups have suggested that inhibition of NF-κB signaling could have metabolic benefits for obesity-associated adipose tissue inflammation. However, two significant problems arise with this approach. The first is how to deliver general NF-κB inhibitors into adipocytes without allowing these compounds to disrupt normal functioning in cells of the immune system. The second issue is that general inhibition of canonical NF-κB signaling in adipocytes will likely lead to a massive increase in adipocyte apoptosis under conditions of metabolic stress, leading full circle into a secondary inflammation (However, this problem may not be true for non-canonical NF-κB signaling.). This review will focus on the research that has examined canonical and non-canonical NF-κB signaling in adipocytes, focusing on genetic studies that examine loss-of-function of NF-κB specifically in fat cells. Although the development of general inhibitors of canonical NF-κB signaling seems unlikely to succeed in alleviating adipose tissue inflammation in humans, the door remains open for more targeted therapeutics. In principle, these would include compounds that interrogate NF-κB DNA binding, protein-protein interactions, or post-translational modifications that partition NF-κB activity towards some genes and away from others in adipocytes. I also discuss the possibility for inhibitors of non-canonical NF-κB signaling to realize success in mitigating fat cell dysfunction in obesity. To plant the seeds for such approaches, much biochemical “digging” in adipocytes remains; this includes identifying—in an unbiased manner–NF-κB direct and indirect targets, genomic DNA binding sites for all five NF-κB subunits, NF-κB protein-protein interactions, and post-translational modifications of NF-κB in fat cells.
Collapse
|
9
|
Bai C, Wang Y, Niu Z, Guan Y, Huang J, Nian X, Zuo F, Zhao J, Kazumi T, Wu B. Exenatide improves hepatocyte insulin resistance induced by different regional adipose tissue. Front Endocrinol (Lausanne) 2022; 13:1012904. [PMID: 36246878 PMCID: PMC9558273 DOI: 10.3389/fendo.2022.1012904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is resulted from energy surplus and is characterized by abnormal adipose tissue accumulation and/or distribution. Adipokines secreted by different regional adipose tissue can induce changes in key proteins of the insulin signaling pathway in hepatocytes and result in impaired hepatic glucose metabolism. This study aimed to investigate whether exenatide affects key proteins of IRS2/PI3K/Akt2 signaling pathway in hepatocytes altered by the different regional fat depots. Six non-obese patients without endocrine diseases were selected as the research subjects. Their subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT)were co-cultured with HepG2 cells in the transwell chamber. In the presence or absence of exenatide, adipokines content in the supernatant of each experimental group was detected by ELISA. In addition, HepG2 cells in each co-culture group with and without insulin were collected, and the expression of key proteins IRS2, p-IRS2(S731), PI3K-p85, Akt2, and p-Akt2(S473) was detected by western blotting (WB). The results showed that the adipokines IL-8, MCP-1, VEGF, and sTNFR2 in the supernatant of HepG2 cells induced by different regional adipose tissue were significantly higher than those in the HepG2 group, and VAT released more adipokines than SAT. Furthermore, these adipokines were significantly inhibited by exenatide. Importantly, the different regional fat depot affects the IRS2/PI3K/Akt2 insulin signaling pathway of hepatocytes. Exenatide can up-regulate the expression of hepatocyte proteins IRS2, PI3K-p85, p-Akt2(S731) inhibited by adipose tissue, and down-regulate the expression of hepatocyte proteins p-IRS2(S731) promoted by adipose tissue. The effect of VAT on the expression of these key proteins in hepatocytes is more significant than that of SAT. But there was no statistical difference in the expression of Akt2 protein among each experimental group, suggesting that exenatide has no influence on the expression of Akt2 protein in hepatocytes. In conclusion, exenatide may improve hepatic insulin resistance (IR) by inhibiting adipokines and regulating the expression of key proteins in the IRS2/PI3K/Akt2 pathway.
Collapse
Affiliation(s)
- Chuanmin Bai
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yujun Wang
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Zhi Niu
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yaxin Guan
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jingshan Huang
- School of Computing, University of South Alabama, Mobile, AL, United States
| | - Xin Nian
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Fan Zuo
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Juan Zhao
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Tsutomu Kazumi
- Open Research Center for Studying of Lifestyle−Related Diseases, Mukogawa Women’s University, Nishinomiya, Japan
- Research Institute for Nutrition Sciences, Mukogawa Women’s University, Nishinomiya, Japan
- Department of Medicine, Kohnan Kakogawa Hospital, Kakogawa, Japan
| | - Bin Wu
- Department of Endocrinology, First Affiliated Hospital, Kunming Medical University, Kunming, China
- *Correspondence: Bin Wu,
| |
Collapse
|
10
|
Mylod E, O’Connell F, Donlon NE, Butler C, Reynolds JV, Lysaght J, Conroy MJ. The Omentum in Obesity-Associated Cancer: A Hindrance to Effective Natural Killer Cell Migration towards Tumour Which Can Be Overcome by CX3CR1 Antagonism. Cancers (Basel) 2021; 14:64. [PMID: 35008227 PMCID: PMC8750072 DOI: 10.3390/cancers14010064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 01/16/2023] Open
Abstract
Oesophagogastric adenocarcinomas (OAC) are obesity-associated malignancies, underpinned by severe immune dysregulation. We have previously shown that natural killer (NK) cells preferentially migrate to OAC omentum, where they undergo phenotypic and functional alterations and apoptosis. Furthermore, we have identified the CX3CR1:fractalkine (CX3CL1) pathway as pivotal in their recruitment to omentum. Here, we elucidate whether exposure to the soluble microenvironment of OAC omentum, and in particular fractalkine and IL-15 affects NK cell homing capacity towards oesophageal tumour. Our data uncover diminished NK cell migration towards OAC tumour tissue conditioned media (TCM) following exposure to omental adipose tissue conditioned media (ACM) and reveal that this migration can be rescued with CX3CR1 antagonist E6130. Furthermore, we show that fractalkine has opposing effects on NK cell migration towards TCM, when used alone or in combination with IL-15 and uncover its inhibitory effects on IL-15-mediated stimulation of death receptor ligand expression. Interestingly, treatment with fractalkine and/or IL-15 do not significantly affect NK cell adhesion to MAdCAM-1, despite changes they elicit to the expression of integrin α4β7. This study provides further evidence that CX3CR1 antagonism has therapeutic utility in rescuing NK cells from the deleterious effects of the omentum and fractalkine in OAC, thus limiting their dysfunction.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Fiona O’Connell
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (F.O.); (C.B.)
| | - Noel E. Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Christine Butler
- Department of Surgery, Trinity St. James’s Cancer Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (F.O.); (C.B.)
| | - John V. Reynolds
- Gastro-Intestinal Medicine and Surgery, St. James’s Hospital, Dublin 8, Ireland;
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
| | - Melissa J. Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St James’s Hospital, Dublin 8, Ireland; (E.M.); (N.E.D.); (J.L.)
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
11
|
Heeran AB, McCready J, Dunne MR, Donlon NE, Nugent TS, Bhardwaj A, Mitchelson KAJ, Buckley AM, Ravi N, Roche HM, Reynolds JV, Lynam-Lennon N, O’Sullivan J. Opposing Immune-Metabolic Signature in Visceral Versus Subcutaneous Adipose Tissue in Patients with Adenocarcinoma of the Oesophagus and the Oesophagogastric Junction. Metabolites 2021; 11:768. [PMID: 34822426 PMCID: PMC8624269 DOI: 10.3390/metabo11110768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/24/2022] Open
Abstract
Oesophageal adenocarcinoma (OAC) is an exemplar model of obesity-associated cancer. Previous work in our group has demonstrated that overweight/obese OAC patients have better responses to neoadjuvant therapy, but the underlying mechanisms are unknown. Unravelling the immune-metabolic signatures of adipose tissue may provide insight for this observation. We hypothesised that different metabolic pathways predominate in visceral (VAT) and subcutaneous adipose tissue (SAT) and inflammatory secretions will differ between the fat depots. Real-time ex vivo metabolic profiles of VAT and SAT from 12 OAC patients were analysed. These samples were screened for the secretion of 54 inflammatory mediators, and data were correlated with patient body composition. Oxidative phosphorylation (OXPHOS) was significantly higher in VAT when compared to SAT. OXPHOS was significantly higher in the SAT of patients receiving neoadjuvant treatment. VEGF-A, VEGF-C, P1GF, Flt-1, bFGF, IL-15, IL-16, IL-17A, CRP, SAA, ICAM-1, VCAM-1, IL-2, IL-13, IFN-γ, and MIP-1β secretions were significantly higher from VAT than SAT. Higher levels of bFGF, Eotaxin-3, and TNF-α were secreted from the VAT of obese patients, while higher levels of IL-23 and TARC were secreted from the SAT of obese patients. The angiogenic factors, bFGF and VEGF-C, correlated with visceral fat area. Levels of OXPHOS are higher in VAT than SAT. Angiogenic, vascular injury and inflammatory cytokines are elevated in VAT versus SAT, indicating that VAT may promote inflammation, linked to regulating treatment response.
Collapse
Affiliation(s)
- Aisling B. Heeran
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Jessica McCready
- Department of Biological and Physical Sciences, Assumption University, Worcester, MA 01609, USA;
| | - Margaret R. Dunne
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Noel E. Donlon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Timothy S. Nugent
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Anshul Bhardwaj
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Kathleen A. J. Mitchelson
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Conway Institute, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (K.A.J.M.); (H.M.R.)
| | - Amy M. Buckley
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Narayanasamy Ravi
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Helen M. Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Conway Institute, University College Dublin, Belfield, D04 V1W8 Dublin, Ireland; (K.A.J.M.); (H.M.R.)
- Institute for Global Food Security, Queens University Belfast, Belfast BT9 5DL, UK
| | - John V. Reynolds
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Niamh Lynam-Lennon
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| | - Jacintha O’Sullivan
- Trinity St. James’s Cancer Institute, Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James’s Hospital, D08 W9RT Dublin, Ireland; (A.B.H.); (M.R.D.); (N.E.D.); (T.S.N.); (A.B.); (A.M.B.); (N.R.); (J.V.R.); (N.L.-L.)
| |
Collapse
|
12
|
Li S, Qiu R, Yuan G, Wang Q, Li Z, Li Q, Zhang N. Body composition in relation to postoperative anastomotic leakage and overall survival in patients with esophageal cancer. Nutrition 2021; 94:111534. [PMID: 34952360 DOI: 10.1016/j.nut.2021.111534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Body composition was reported to be related to the prognosis of patients with cancer. This study aimed to investigate the influence of preoperative body composition on anastomotic leakage and overall survival in patients with esophageal cancer. METHODS In this retrospective study, 93 patients with esophageal cancers were evaluated. Skeletal muscle area, intermuscular adipose tissue, visceral adipose tissue (VAT), and subcutaneous adipose tissue were measured on computed tomography images at the level of the third lumbar vertebra. Subsequently, each body composition index was also calculated by dividing the body composition by the square of the height. The cut-off values of body compositions were defined using X-tile software (version 3.6.1; Yale University, New Haven, CTA). Univariate and multivariate analyses were performed to evaluate the risk factors of anastomotic leakage. Kaplan-Meier method and Cox regression analysis were used to evaluate the risk factors of overall survival. RESULTS VAT and visceral fat index (VFI) were higher in patients with anastomotic fistula than in those without anastomotic fistula, but none of them were independent risk factors. Patients with higher body mass index (BMI), higher VFI, and higher subcutaneous fat index (SFI) had better overall survival. By multivariate analysis, SFI >27.6 cm2/m2 was still significantly associated with overall survival. CONCLUSION Patients with higher VAT and VFI were prone to have an anastomotic leakage. Lower BMI, VFI, and SFI were associated with a reduction in overall survival.
Collapse
Affiliation(s)
- Shichao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruixing Qiu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guanjie Yuan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
13
|
Obesity and Pancreatic Cancer: Insight into Mechanisms. Cancers (Basel) 2021; 13:cancers13205067. [PMID: 34680216 PMCID: PMC8534007 DOI: 10.3390/cancers13205067] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Obesity is recognized as a chronic progressive disease and risk factor for many human diseases. The high and increasing number of obese people may underlie the expected increase in pancreatic cancer cases in the United States. There are several pathways discussed that link obesity with pancreatic cancer. Adipose tissue and adipose tissue-released factors may thereby play an important role. This review discusses selected mechanisms that may accelerate pancreatic cancer development in obesity. Abstract The prevalence of obesity in adults and children has dramatically increased over the past decades. Obesity has been declared a chronic progressive disease and is a risk factor for a number of metabolic, inflammatory, and neoplastic diseases. There is clear epidemiologic and preclinical evidence that obesity is a risk factor for pancreatic cancer. Among various potential mechanisms linking obesity with pancreatic cancer, the adipose tissue and obesity-associated adipose tissue inflammation play a central role. The current review discusses selected topics and mechanisms that attracted recent interest and that may underlie the promoting effects of obesity in pancreatic cancer. These topics include the impact of obesity on KRAS activity, the role of visceral adipose tissue, intrapancreatic fat, adipose tissue inflammation, and adipokines on pancreatic cancer development. Current research on lipocalin-2, fibroblast growth factor 21, and Wnt5a is discussed. Furthermore, the significance of obesity-associated insulin resistance with hyperinsulinemia and obesity-induced gut dysbiosis with metabolic endotoxemia is reviewed. Given the central role that is occupied by the adipose tissue in obesity-promoted pancreatic cancer development, preventive and interceptive strategies should be aimed at attenuating obesity-associated adipose tissue inflammation and/or at targeting specific molecules that mechanistically link adipose tissue with pancreatic cancer in obese patients.
Collapse
|
14
|
Leiter A, Carroll E, De Alwis S, Brooks D, Shimol JB, Eisenberg E, Wisnivesky JP, Galsky MD, Gallagher EJ. Metabolic disease and adverse events from immune checkpoint inhibitors. Eur J Endocrinol 2021; 184:857-865. [PMID: 34552304 PMCID: PMC8451971 DOI: 10.1530/eje-20-1362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Obese and overweight body mass index (BMI) categories have been associated with increased immune-related adverse events (irAEs) in patients with cancer receiving immune checkpoint inhibitors (ICIs); however, the impact of being overweight in conjunction with related metabolic syndrome-associated factors on irAEs have not been investigated. We aimed to evaluate the impact of overweight and obese BMI according to metabolic disease burden on the development of irAEs. DESIGN AND METHODS We conducted a retrospective observational study of patients receiving ICIs at a cancer center. Our main study outcome was development of ≥grade 2 (moderate) irAEs. Our main predictor was weight/metabolic disease risk category: (1) normal weight (BMI 18.5-24.9 kg/m2)/low metabolic risk (<2 metabolic diseases [diabetes, dyslipidemia, hypertension]), (2) normal weight/high metabolic risk (≥2 metabolic diseases), (3) overweight (BMI ≥25 kg/m2)/low metabolic risk, and (4) overweight/high metabolic risk. RESULTS Of 411 patients in our cohort, 374 were eligible for analysis. Overall, 111 (30%) patients developed ≥grade 2 irAEs. In Cox analysis, overweight/low metabolic risk was significantly associated with ≥grade 2 irAEs (hazard ratio [HR]: 2.0, 95% confidence interval [95% CI]: 1.2-3.4) when compared to normal weight/low metabolic risk, while overweight/high metabolic risk (HR: 1.3, 95% CI: 0.7-2.2) and normal weight/high metabolic risk (HR: 1.5, 95% CI: 0.7-3.0) were not. CONCLUSIONS Overweight patients with fewer metabolic comorbidities were at increased risk for irAEs. This study provides an important insight that BMI should be evaluated in the context of associated metabolic comorbidities in assessing risk of irAE development and ICI immune response.
Collapse
Affiliation(s)
- Amanda Leiter
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emily Carroll
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sonia De Alwis
- Division of Endocrinology, Department of Medicine, New York University Langone Medical Center
| | - Danielle Brooks
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jennifer Ben Shimol
- Department of Rheumatology, Edith Wolfson Medical Center, Holon, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elliot Eisenberg
- Division of Pulmonology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Juan P. Wisnivesky
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Matthew D. Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emily Jane Gallagher
- Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
15
|
Skeletal Muscle-Adipose Tissue-Tumor Axis: Molecular Mechanisms Linking Exercise Training in Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094469. [PMID: 33922898 PMCID: PMC8123194 DOI: 10.3390/ijms22094469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Increased visceral adiposity may influence the development of prostate cancer (PCa) aggressive tumors and cancer mortality. White adipose tissue (WAT), usually referred to as periprostatic adipose tissue (PPAT), surrounds the prostatic gland and has emerged as a potential mediator of the tumor microenvironment. Exercise training (ET) induces several adaptations in both skeletal muscle and WAT. Some of these effects are mediated by ET-induced synthesis and secretion of several proteins, known as myo- and adipokines. Together, myokines and adipokines may act in an endocrine-like manner to favor communication between skeletal muscle and WAT, as they may work together to improve whole-body metabolic health. This crosstalk may constitute a potential mechanism by which ET exerts its beneficial role in the prevention and treatment of PCa-related disorders; however, this has not yet been explored. Therefore, we reviewed the current evidence on the effects of skeletal muscle–WAT–tumor crosstalk in PCa, and the potential mediators of this process to provide a better understanding of underlying ET-related mechanisms in cancer.
Collapse
|
16
|
Peripancreatic Adipose Tissue Remodeling and Inflammation during High Fat Intake of Palm Oils or Lard in Rats. Nutrients 2021; 13:nu13041134. [PMID: 33808251 PMCID: PMC8065769 DOI: 10.3390/nu13041134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 12/25/2022] Open
Abstract
Excessive fat consumption leads to the development of ectopic adipose tissues, affecting the organs they surround. Peripancreatic adipose tissue is implicated in glucose homeostasis regulation and can be impaired in obesity. High palm oil consumption's effects on health are still debated. We hypothesised that crude and refined palm oil high-fat feeding may have contrasting effects on peripancreatic adipocyte hypertrophy, inflammation and lipid oxidation compound production in obese rats. In Wistar rats, morphological changes, inflammation and isoprostanoid production following oxidative stress were assessed in peripancreatic adipose tissue after 12 weeks of diets enriched in crude or refined palm oil or lard (56% energy from fat in each case) versus a standard chow diet (11% energy from fat). Epididymal white and periaortic brown adipose tissues were also included in the study. A refined palm oil diet disturbed glucose homeostasis and promoted lipid deposition in periaortic locations, as well as adipocyte hypertrophy, macrophage infiltration and isoprostanoid (5-F2c-isoprostane and 7(RS)-ST-Δ8-11-dihomo-isofuran) production in peripancreatic adipose tissue. Crude palm oil induced a lower impact on adipose deposits than its refined form and lard. Our results show that the antioxidant composition of crude palm oil may have a protective effect on ectopic adipose tissues under the condition of excessive fat intake.
Collapse
|
17
|
Elliott JA, Reynolds JV. Visceral Obesity, Metabolic Syndrome, and Esophageal Adenocarcinoma. Front Oncol 2021; 11:627270. [PMID: 33777773 PMCID: PMC7994523 DOI: 10.3389/fonc.2021.627270] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) represents an exemplar of obesity-associated carcinogenesis, with a progressive increase in EAC risk with increased body mass index. In this context, there is increased focus on visceral adipose tissue and associated metabolic dysfunction, including hypertension, diabetes mellitus and hyperlipidemia, or combinations of these in the metabolic syndrome. Visceral obesity (VO) may promote EAC via both directly impacting on gastro-esophageal reflux disease and Barrett's esophagus, as well as via reflux-independent effects, involving adipokines, growth factors, insulin resistance, and the microbiome. In this review these pathways are explored, including the impact of VO on the tumor microenvironment, and on cancer outcomes. The current evidence-based literature regarding the role of dietary, lifestyle, pharmacologic and surgical interventions to modulate the risk of EAC is explored.
Collapse
Affiliation(s)
- Jessie A Elliott
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
18
|
Wang L, Sun P, Wu Y, Wang L. Metabolic tissue-resident CD8 + T cells: A key player in obesity-related diseases. Obes Rev 2021; 22:e13133. [PMID: 32935464 DOI: 10.1111/obr.13133] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/15/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023]
Abstract
Obesity-induced low-grade chronic inflammation in the metabolic tissues, such as adipose tissue (AT) and liver tissue, in individuals with obesity is a major etiological factor for several diseases, such as insulin resistance, type 2 diabetes, fatty liver disease, atherosclerosis and cardiovascular problems, as well as cancer and autoimmune diseases. Previous studies have revealed that tissue-resident macrophages play a crucial role in this process. However, the mechanisms responsible for recruiting and activating macrophages and initiating chronic inflammation in the metabolic tissues have not yet been clearly elucidated. In the most recent decade, there has been a growing emphasis on the critical role of the adaptive CD8+ T cells in obesity-induced chronic inflammation and related metabolic diseases. In this review, we will summarize the relevant studies in both mice and human regarding the role of metabolic tissue-resident CD8+ T cells in obesity-related inflammation and diseases, as well as the possible mechanisms underlying the regulation of CD8+ T cell recruitment, activation and function in the metabolic tissues, and discuss their potential as therapeutic targets for obesity-related diseases.
Collapse
Affiliation(s)
- Lina Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing, China.,Department of Immunology, Weifang Medical University, Weifang, China
| | - Ping Sun
- Department of Immunology, Weifang Medical University, Weifang, China
| | - Yuzhang Wu
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Wang
- Institute of Immunology PLA, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
19
|
Hamilton-Reeves JM, Johnson CN, Hand LK, Bechtel MD, Robertson HL, Michel C, Metcalf M, Chalise P, Mahan NJ, Mirza M, Lee EK, Sullivan DK, Klemp JR, Befort CA, Parker WP, Gibbs HD, Demark-Wahnefried W, Thrasher JB. Feasibility of a Weight Management Program Tailored for Overweight Men with Localized Prostate Cancer - A Pilot Study. Nutr Cancer 2020; 73:2671-2686. [PMID: 33295204 PMCID: PMC8371995 DOI: 10.1080/01635581.2020.1856890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Overweight men with prostate cancer are more likely to suffer from recurrence and death following prostatectomy compared with healthy weight men. This study tested the feasibility of delivering a comprehensive program to foster weight loss before and weight maintenance after surgery in overweight men with localized prostate cancer. METHODS Twenty overweight men scheduled for prostatectomy elected either the intervention (n = 15) or the nonintervention (n = 5). Anthropometrics, biomarkers, diet quality, nutrition literacy, quality of life, and long-term follow-up were assessed in both groups. RESULTS The intervention led to 5.55 kg of weight loss including 3.88 kg of fat loss from baseline to surgery (mean = 8.3 weeks). The intervention significantly increased fiber, protein, fruit, nut, and vegetable intake; and decreased trans fats intake during weight loss. The intervention significantly reduced insulin, C-peptide, systolic blood pressure, leptin:adiponectin ratio, and visceral adiposity compared to the nonintervention. Post-surgically, weight loss was maintained. Changes in lipid profiles, nutrition literacy, and follow-up were not statistically significant in either group. CONCLUSION Significant weight loss (≥5%) is feasible with a coaching intervention in overweight men preparing for prostatectomy and is associated with favorable cardiometabolic effects. This study is registered under NCT02252484 (www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Jill M Hamilton-Reeves
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Chelsea N Johnson
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Lauren K Hand
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Misty D Bechtel
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hilary L Robertson
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Carrie Michel
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Meredith Metcalf
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Prabhakar Chalise
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Nicholas J Mahan
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Moben Mirza
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Eugene K Lee
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Debra K Sullivan
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jennifer R Klemp
- Breast Cancer Prevention Center, University of Kansas Medical Center, Westwood, Kansas, USA
| | - Christie A Befort
- Department of Preventative Medicine and Public Health, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - William P Parker
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Heather D Gibbs
- Department of Dietetics & Nutrition, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wendy Demark-Wahnefried
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - J Brantley Thrasher
- Department of Urology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
20
|
Haffa M, Holowatyj AN, Kratz M, Toth R, Benner A, Gigic B, Habermann N, Schrotz-King P, Böhm J, Brenner H, Schneider M, Ulrich A, Herpel E, Schirmacher P, Straub BK, Nattenmüller J, Kauczor HU, Lin T, Ball CR, Ulrich CM, Glimm H, Scherer D. Transcriptome Profiling of Adipose Tissue Reveals Depot-Specific Metabolic Alterations Among Patients with Colorectal Cancer. J Clin Endocrinol Metab 2019; 104:5225-5237. [PMID: 31225875 PMCID: PMC6763280 DOI: 10.1210/jc.2019-00461] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
CONTEXT Adipose tissue inflammation and dysregulated energy homeostasis are key mechanisms linking obesity and cancer. Distinct adipose tissue depots strongly differ in their metabolic profiles; however, comprehensive studies of depot-specific perturbations among patients with cancer are lacking. OBJECTIVE We compared transcriptome profiles of visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) from patients with colorectal cancer and assessed the associations of different anthropometric measures with depot-specific gene expression. DESIGN Whole transcriptomes of VAT and SAT were measured in 233 patients from the ColoCare Study, and visceral and subcutaneous fat area were quantified via CT. RESULTS VAT compared with SAT showed elevated gene expression of cytokines, cell adhesion molecules, and key regulators of metabolic homeostasis. Increased fat area was associated with downregulated lipid and small molecule metabolism and upregulated inflammatory pathways in both compartments. Comparing these patterns between depots proved specific and more pronounced gene expression alterations in SAT and identified unique associations of integrins and lipid metabolism-related enzymes. VAT gene expression patterns that were associated with visceral fat area poorly overlapped with patterns associated with self-reported body mass index (BMI). However, subcutaneous fat area and BMI showed similar associations with SAT gene expression. CONCLUSIONS This large-scale human study demonstrates pronounced disparities between distinct adipose tissue depots and reveals that BMI poorly correlates with fat mass-associated changes in VAT. Taken together, these results provide crucial evidence for the necessity to differentiate between distinct adipose tissue depots for a correct characterization of gene expression profiles that may affect metabolic health of patients with colorectal cancer.
Collapse
Affiliation(s)
- Mariam Haffa
- Division of Translational Functional Cancer Genomics, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Division of Translational Medical Oncology, National Center for Tumor Diseases Dresden and German Cancer Research Center, Dresden, Germany
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Andreana N Holowatyj
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Mario Kratz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Reka Toth
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - Biljana Gigic
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Nina Habermann
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Petra Schrotz-King
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | - Jürgen Böhm
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Hermann Brenner
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Herpel
- NCT Tissue Bank, National Center for Tumor Diseases and University Hospital Heidelberg, Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Beate K Straub
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Institute of Pathology, University Medicine Mainz, Mainz, Germany
| | - Johanna Nattenmüller
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Tengda Lin
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Claudia R Ball
- Division of Translational Functional Cancer Genomics, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Division of Translational Medical Oncology, National Center for Tumor Diseases Dresden and German Cancer Research Center, Dresden, Germany
- Center for Personalized Oncology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
| | - Cornelia M Ulrich
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Huntsman Cancer Institute, Salt Lake City, Utah
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Hanno Glimm
- Division of Translational Functional Cancer Genomics, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Division of Translational Medical Oncology, National Center for Tumor Diseases Dresden and German Cancer Research Center, Dresden, Germany
- Center for Personalized Oncology, University Hospital Carl Gustav Carus Dresden, Technische Universität Dresden, Dresden, Germany
- DKTK, Dresden, Germany
| | - Dominique Scherer
- Division of Preventive Oncology, National Center for Tumor Diseases Heidelberg and German Cancer Research Center, Heidelberg, Germany
- Institute of Medical Biometry and Informatics, University Heidelberg, Heidelberg, Germany
- Correspondence and Reprint Requests: Dominique Scherer, PhD, Institute of Medical Biometry and Informatics, University of Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany. E-mail:
| |
Collapse
|
21
|
Obesity-Induced Adipose Tissue Inflammation as a Strong Promotional Factor for Pancreatic Ductal Adenocarcinoma. Cells 2019; 8:cells8070673. [PMID: 31277269 PMCID: PMC6678863 DOI: 10.3390/cells8070673] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to soon become the second leading cause of cancer related deaths in the United States. This may be due to the rising obesity prevalence, which is a recognized risk factor for PDAC. There is great interest in deciphering the underlying driving mechanisms of the obesity–PDAC link. Visceral adiposity has a strong correlation to certain metabolic diseases and gastrointestinal cancers, including PDAC. In fact, our own data strongly suggest that visceral adipose tissue inflammation is a strong promoter for PDAC growth and progression in a genetically engineered mouse model of PDAC and diet-induced obesity. In this review, we will discuss the relationship between obesity-associated adipose tissue inflammation and PDAC development, with a focus on the key molecular and cellular components in the dysfunctional visceral adipose tissue, which provides a tumor permissive environment.
Collapse
|
22
|
Pyrazinib (P3), [(E)-2-(2-Pyrazin-2-yl-vinyl)-phenol], a small molecule pyrazine compound enhances radiosensitivity in oesophageal adenocarcinoma. Cancer Lett 2019; 447:115-129. [DOI: 10.1016/j.canlet.2019.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/13/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023]
|
23
|
Mongan AM, Lynam-Lennon N, Doyle SL, Casey R, Carr E, Cannon A, Conroy MJ, Pidgeon GP, Brennan L, Lysaght J, Reynolds JV, O'Sullivan J. Visceral Adipose Tissue Modulates Radiosensitivity in Oesophageal Adenocarcinoma. Int J Med Sci 2019; 16:519-528. [PMID: 31171903 PMCID: PMC6535661 DOI: 10.7150/ijms.29296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/13/2018] [Indexed: 12/21/2022] Open
Abstract
Oesophageal adenocarcinoma (OAC) is an exemplar model of obesity-associated cancer. Response to neoadjuvant chemoradiotherapy (NA CRT) is a clinical challenge. We examined if visceral adipose tissue and obesity status alter radiosensitivity in OAC. The radioresistant (OE33R) and radioresponsive (OE33P) OAC isogenic model was cultured with adipose tissue conditioned media from three patient cohorts: non-cancer patients, surgery only OAC patients and NA CRT OAC patients. Cell survival was characterised by clonogenic assay, metabolomic profiling by nuclear magnetic resonance spectroscopy and adipokine receptor gene expression by qPCR. A retrospective in vivo study compared tumour response to NA CRT in normal weight (n=53) versus overweight/obese patients (n=148). Adipose conditioned media (ACM) from all patient cohorts significantly increased radiosensitivity in radioresistant OE33R cells. ACM from the NA CRT OAC cohort increased radiosensitivity in OE33P cells. Metabolomic profiling demonstrated separation of the non-cancer and surgery only OAC cohorts and between the non-cancer and NA CRT OAC cohorts. Gene expression profiling of OE33P versus OE33R cells demonstrated differential expression of the adiponectin receptor-1 (AR1), adiponectin receptor-2 (AR2), leptin receptor (LepR) and neuropilin receptor-1 (NRP1) genes. In vivo overweight/obese OAC patients achieved an enhanced tumour response following NA CRT compared to normal weight patients. This study demonstrates that visceral adipose tissue modulates the cellular response to radiation in OAC.
Collapse
Affiliation(s)
- Ann Marie Mongan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Niamh Lynam-Lennon
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Suzanne L Doyle
- School of Biological Sciences, Dublin Institute of Technology, Dublin, Ireland
| | - Rory Casey
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Eibhlin Carr
- School of Agriculture & Food Science, Science Centre-South, Belfield, Dublin 4, Ireland
| | - Aoife Cannon
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Melissa J Conroy
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Graham P Pidgeon
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Lorraine Brennan
- School of Agriculture & Food Science, Science Centre-South, Belfield, Dublin 4, Ireland
| | - Joanne Lysaght
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, St. James's Hospital, Dublin 8, Ireland
| |
Collapse
|
24
|
Neto NIP, Murari ASDP, Oyama LM, Otoch JP, Alcântara PSM, Tokeshi F, Figuerêdo RG, Alves MJ, Lima JDCC, Matos-Neto EMD, Seelaender M, Oller do Nascimento CM. Peritumoural adipose tissue pro-inflammatory cytokines are associated with tumoural growth factors in cancer cachexia patients. J Cachexia Sarcopenia Muscle 2018; 9:1101-1108. [PMID: 30284380 PMCID: PMC6240753 DOI: 10.1002/jcsm.12345] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/24/2018] [Accepted: 08/19/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer cachexia (CC) is a multifactorial syndrome, often irreversible, that affects patients with cancer influenced, in part, by the inflammatory condition. Peritumoural adipose tissue produces adipokines and angiogenic, apoptotic, and growth factors; given the possible crosstalk between the peritumoural adipose tissue and tumour, these may play an important role in cancer biology and carcinogenesis. METHODS The aim of this study was to evaluate the factors produced by peritumoural adipose tissue in a cohort of 16 colorectal cancer patients with either weight-stable cancer (WSC; n = 7) or CC (n = 9). The study was approved by the Ethics Research Committee (972.914). Samples of peritumoural adipose tissue were analysed for concentrations of TNF-α, IL-1β, STAT-1, STAT-3, RANTES, IL-1Ra, IP-10, IL-15, MCP-1, IFN-α, GCSF, FADD, and TGF-β. The cytokines and proteins were measured using Multiplex. Correlations between the proteins and cytokines were evaluated. RESULTS TNF-α, STAT-1, and FADD, a factor involved in apoptosis, were significantly higher in CC group than in the WSC group. In the peritumoural adipose tissue of the CC group, RANTES showed a significant positive correlation with IL-1Ra and IP-10 and a negative correlation with IFN-α; and GCSF showed significant negative correlations with IL-1Ra, IP-10, IL-15, and MCP-1 and a positive correlation with IFN-α. In the peritumoural adipose tissue of the WSC group, no significant correlations were detected between RANTES, GCSF, IL-3, FADD, and STAT-1 and the cytokines/chemokines analysed. CONCLUSIONS These results indicated that inflammatory and tumorigenic pathways were altered in peritumoural adipose tissue in CC. Furthermore, inflammatory cytokines were correlated with growth factors in the peritumoural adipose tissue of cachectic patients, suggesting that inflammatory cytokines modulated the proliferative environment closely linked to the tumour.
Collapse
Affiliation(s)
- Nelson Inácio Pinto Neto
- Escola Paulista de Medicina, Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Lila Missae Oyama
- Escola Paulista de Medicina, Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Pinhata Otoch
- Department of Clinical Surgery, University of São Paulo, São Paulo, Brazil
| | | | - Flavio Tokeshi
- Department of Clinical Surgery, University of São Paulo, São Paulo, Brazil
| | - Raquel Galvão Figuerêdo
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Michele Joana Alves
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Marilia Seelaender
- Department of Clinical Surgery, University of São Paulo, São Paulo, Brazil.,Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
25
|
Del Cornò M, Conti L, Gessani S. Innate Lymphocytes in Adipose Tissue Homeostasis and Their Alterations in Obesity and Colorectal Cancer. Front Immunol 2018; 9:2556. [PMID: 30455701 PMCID: PMC6230679 DOI: 10.3389/fimmu.2018.02556] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and a leading cause of death, with burden expected to increase in the coming years. Enhanced adiposity, particularly visceral fat, is associated with increased cancer incidence representing an important indicator of survival, prognosis, recurrence rates, and response to therapy for several tumors including CRC. Compelling evidence has been achieved that the low-grade chronic inflammation characterizing obesity represents a main factor that can favor carcinogenesis. Adipocytes and adipose tissue (AT) infiltrating immune cells contribute to obesity-related inflammation by releasing soluble factors affecting, both locally and systemically, the function of several cell types, including immune and cancer cells. The unbalanced production of immune mediators as well as the profound changes in the repertoire and activation state of immune cells in AT of obese subjects represent key events in the processes that set the basis for a pro-tumorigenic microenvironment. AT harbors a unique profile of immune cells of different origin that play an important role in tissue homeostasis. Among these, tissue-resident innate lymphocytes are emerging as important AT components whose depletion/aberrant activation occurring in obesity could have an impact on inflammation and immune-surveillance against tumors. However, a direct link between obesity-induced dysfunction and cancer development has not been demonstrated yet. In this review, we provide an overview of human obesity- and CRC-induced alterations of blood and adipose tissue-associated innate lymphocytes, and discuss how the adipose tissue microenvironment in obesity might influence the development of CRC.
Collapse
Affiliation(s)
- Manuela Del Cornò
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Conti
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sandra Gessani
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
26
|
Obesity and gastrointestinal cancer: the interrelationship of adipose and tumour microenvironments. Nat Rev Gastroenterol Hepatol 2018; 15:699-714. [PMID: 30323319 DOI: 10.1038/s41575-018-0069-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Increasing recognition of an association between obesity and many cancer types exists, but how the myriad of local and systemic effects of obesity affect key cellular and non-cellular processes within the tumour microenvironment (TME) relevant to carcinogenesis, tumour progression and response to therapies remains poorly understood. The TME is a complex cellular environment in which the tumour exists along with blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, signalling molecules and the extracellular matrix. Obesity, in particular visceral obesity, might fuel the dysregulation of key pathways relevant to both the adipose microenvironment and the TME, which interact to promote carcinogenesis in at-risk epithelium. The tumour-promoting effects of obesity can occur at the local level as well as systemically via circulating inflammatory, growth factor and metabolic mediators associated with adipose tissue inflammation, as well as paracrine and autocrine effects. This Review explores key pathways linking visceral obesity and gastrointestinal cancer, including inflammation, hypoxia, altered stromal and immune cell function, energy metabolism and angiogenesis.
Collapse
|
27
|
Chakedis J, Beal EW, Sun S, Galo J, Chafitz A, Davidson G, Reardon J, Dillhoff M, Pawlik TM, Abdel-Misih S, Bloomston M, Schmidt CR. Implementation and early outcomes for a surgeon-directed hepatic arterial infusion pump program for colorectal liver metastases. J Surg Oncol 2018; 118:1065-1073. [DOI: 10.1002/jso.25249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Jeffery Chakedis
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Eliza W. Beal
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Steven Sun
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Jason Galo
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Aaron Chafitz
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Gail Davidson
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Joshua Reardon
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Mary Dillhoff
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Timothy M. Pawlik
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | - Sherif Abdel-Misih
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| | | | - Carl R. Schmidt
- Division of Surgical Oncology, Department of Surgery; The Ohio State University Wexner Medical Center, Arthur G. James Cancer Hospital and Solove Research Institute; Columbus Ohio
| |
Collapse
|
28
|
Canter RJ, Le CT, Beerthuijzen JM, Murphy WJ. Obesity as an immune-modifying factor in cancer immunotherapy. J Leukoc Biol 2018; 104:487-497. [PMID: 29762866 PMCID: PMC6113103 DOI: 10.1002/jlb.5ri1017-401rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy has achieved breakthrough status in many advanced stage malignancies and is rapidly becoming the fourth arm of cancer treatment. Although cancer immunotherapy has generated significant excitement because of the potential for complete and sometimes durable responses, there is also the potential for severe and occasionally life-threatening toxicities, including cytokine release syndrome and severe autoimmunity. A large body of work also points to a "metainflammatory" state in obesity associated with impairment of immune responses. Because immune checkpoint blockade (and other cancer immunotherapies) have altered the landscape of immunotherapy in cancer, it is important to understand how immune responses are shaped by obesity and how obesity may modify both immunotherapy responses and potential toxicities.
Collapse
Affiliation(s)
- Robert J. Canter
- University of California, Davis, School of Medicine, Department of Surgery, Division of Surgical Oncology, Sacramento, CA 95817
| | - Catherine T Le
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| | - Johanna M.T. Beerthuijzen
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| | - William J. Murphy
- University of California, Davis, School of Medicine, Departments of Dermatology and Internal Medicine, Sacramento, CA 95817
| |
Collapse
|
29
|
Himbert C, Delphan M, Scherer D, Bowers LW, Hursting S, Ulrich CM. Signals from the Adipose Microenvironment and the Obesity-Cancer Link-A Systematic Review. Cancer Prev Res (Phila) 2018; 10:494-506. [PMID: 28864539 DOI: 10.1158/1940-6207.capr-16-0322] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/06/2017] [Accepted: 07/11/2017] [Indexed: 12/13/2022]
Abstract
Obesity and its associated metabolic dysregulation are established risk factors for many cancers. However, the biologic mechanisms underlying this relationship remain incompletely understood. Given the rising rates of both obesity and cancer worldwide, and the challenges for many people to lose excess adipose tissue, a systematic approach to identify potential molecular and metabolic targets is needed to develop effective mechanism-based strategies for the prevention and control of obesity-driven cancer. Epidemiologic, clinical, and preclinical data suggest that within the growth-promoting, proinflammatory microenvironment accompanying obesity, crosstalk between adipose tissue (comprised of adipocytes, macrophages and other cells) and cancer-prone cells may occur via obesity-associated hormones, cytokines, and other mediators that have been linked to increased cancer risk and/or progression. We report here a systematic review on the direct "crosstalk" between adipose tissue and carcinomas in humans. We identified 4,641 articles with n = 20 human clinical studies, which are summarized as: (i) breast (n = 7); (ii) colorectal (n = 4); (iii) esophageal (n = 2); (iv) esophageal/colorectal (n = 1); (v) endometrial (n = 1); (vi) prostate (n = 4); and (vii) ear-nose-throat (ENT) cancer (n = 1). Findings from these clinical studies reinforce preclinical data and suggest organ-dependent crosstalk between adipose tissue and carcinomas via VEGF, IL6, TNFα, and other mechanisms. Moreover, visceral white adipose tissue plays a more central role, as it is more bioenergetically active and is associated with a more procancer secretome than subcutaneous adipose tissue. Efforts to eavesdrop and ultimately interfere with this cancer-enhancing crosstalk may lead to new targets and strategies for decreasing the burden of obesity-related cancers. Cancer Prev Res; 10(9); 494-506. ©2017 AACR.
Collapse
Affiliation(s)
- Caroline Himbert
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Mahmoud Delphan
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah.,Exercise Immunology, Physical Education and Sport Sciences Department, Tarbiat Modares University, Tehran, Iran
| | - Dominique Scherer
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Laura W Bowers
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Stephen Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah. .,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
30
|
Jin H, Li D, Wang X, Jia J, Chen Y, Yao Y, Zhao C, Lu X, Zhang S, Togo J, Ji Y, Zhang L, Feng X, Zheng Y. VEGF and VEGFB Play Balancing Roles in Adipose Differentiation, Gene Expression, and Function. Endocrinology 2018; 159:2036-2049. [PMID: 29596616 DOI: 10.1210/en.2017-03246] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/14/2018] [Indexed: 01/04/2023]
Abstract
Obesity is the result of abnormal adipose development and energy metabolism. Using vascular endothelial growth factor (VEGF) B-knockout and inducible VEGF downregulation mouse models, we have shown that VEGFB inactivation caused expansion of white adipose, whitening of brown adipose, an increase in fat accumulation, and a reduction in energy consumption. At the same time, expression of the white adipose-associated genes was increased and brown adipose-associated genes decreased. VEGF repression, in contrast, induced brown adipose expansion and brown adipocyte development in white adipose, increased energy expenditure, upregulated brown adipose-associated genes, and downregulated white adipose-associated genes. When VEGFB-knockout and VEGF-repressed mice are crossed together, VEGF and VEGFB can counteractively regulate large numbers of genes and efficiently reverse each other's roles. These genes, under counteractive VEGF and VEGFB regulations, include transcription factors, adhesion molecules, and metabolic enzymes. This balancing role is confirmed by morphologic and functional changes. This study reports that VEGF and VEGFB counteractively regulate adipose development and function in energy metabolism.
Collapse
Affiliation(s)
- Honghong Jin
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Dan Li
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Xutong Wang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
| | - Jia Jia
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yang Chen
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yapeng Yao
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Chunlan Zhao
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Xiaodan Lu
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Shujie Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Jacques Togo
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yan Ji
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston
| | - Luqing Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
| | - Xuechao Feng
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
| | - Yaowu Zheng
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
| |
Collapse
|
31
|
Galvin KC, Conroy MJ, Doyle SL, Dunne MR, Fahey R, Foley E, O'Sullivan KE, Doherty DG, Geoghegan JG, Ravi N, O'Farrelly C, Reynolds JV, Lysaght J. Extratumoral PD-1 blockade does not perpetuate obesity-associated inflammation in esophageal adenocarcinoma. Cancer Lett 2018; 418:230-238. [PMID: 29339209 DOI: 10.1016/j.canlet.2018.01.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/22/2023]
Abstract
Checkpoint inhibitors, such as anti-PD-1 (Programmed death-1), are transforming cancer treatment for inoperable or advanced disease. As the incidence of obesity-associated malignancies, including esophageal adenocarcinoma (EAC) continues to increase and treatment with checkpoint inhibitors are being FDA approved for a broader range of cancers, it is important to assess how anti-PD-1 treatment might exacerbate pre-existing inflammatory processes at other sites. Outside the EAC tumor, the omentum and liver were found to be enriched with substantial populations of PD-1 expressing T cells. Treatment of omental and hepatic T cells with anti-PD-1 (clone EH12.2H7) did not enhance inflammatory cytokine expression or proliferation, but transiently increased CD107a expression by CD8+ T cells. Importantly, PD-1-expressing T cells are significantly lower in EAC tumor post neoadjuvant chemoradiotherapy, suggesting that combination with specific conventional treatments may severely impair the efficacy of anti-PD-1 immunotherapy. This study provides evidence that systemically administered anti-PD-1 treatment is unlikely to exacerbate pre-existing T cell-mediated inflammation outside the tumor in obesity-associated cancers, such as EAC. Furthermore, our data suggests that studies are required to identify the negative impact of concomitant therapies on PD-1 expression in order to boost overall response rates.
Collapse
Affiliation(s)
- Karen C Galvin
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | - Melissa J Conroy
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | - Suzanne L Doyle
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | - Margaret R Dunne
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | - Ronan Fahey
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Emma Foley
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | - Katie E O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland
| | | | - Justin G Geoghegan
- Liver Transplant Unit, St. Vincent's University Hospital, Dublin, Ireland
| | - Narayanasamy Ravi
- National Esophageal and Gastric Center, St. James's Hospital, Dublin, Ireland
| | - Cliona O'Farrelly
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - John V Reynolds
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland; National Esophageal and Gastric Center, St. James's Hospital, Dublin, Ireland
| | - Joanne Lysaght
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin, Ireland.
| |
Collapse
|
32
|
Zoico E, Rizzatti V, Darra E, Budui SL, Franceschetti G, Vinante F, Pedrazzani C, Guglielmi A, De Manzoni G, Mazzali G, Rossi AP, Fantin F, Zamboni M. Morphological and Functional Changes in the Peritumoral Adipose Tissue of Colorectal Cancer Patients. Obesity (Silver Spring) 2017; 25 Suppl 2:S87-S94. [PMID: 29086519 DOI: 10.1002/oby.22008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/06/2017] [Accepted: 08/17/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The role of peritumoral adipose tissue (AT) has not been extensively studied in colorectal cancer (CRC). METHODS This study was conducted in 20 male subjects undergoing elective surgery for CRC. The differences between the peritumoral visceral adipose tissue (P-VAT), visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT) of the patients were described via immunohistochemistry and molecular biology analyses. The interactions between adipocytes and a colon cancer cell line were also investigated by using an in vitro coculture system. RESULTS The analyses revealed that adipocytes near the tumor were significantly smaller than the adipocytes from other sites. The P-VAT was preferentially infiltrated by a CD68+/CD163+/IDO- macrophage subset with a prevalent reparative inflammatory response, while the macrophages identified in VAT and SAT mainly presented inflammatory features. Furthermore, the P-VAT presented a higher expression of adiponectin compared with other sites. Morphological analysis in vitro showed that after a few days of coculture, 3T3-L1 adipocytes were reduced in number and size with an increase in lipolysis rate and dedifferentiation phenomena. CONCLUSIONS This study reveals important morphological and functional changes in the AT surrounding the tumor as an increase in lipolysis and in adiponectin-producing adipocytes, preferentially infiltrated by a macrophage subset, with prevalent reparative inflammatory response.
Collapse
Affiliation(s)
- Elena Zoico
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | - Vanni Rizzatti
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | - Elena Darra
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | | | - Guido Franceschetti
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | - Fabrizio Vinante
- Hematology Section, Department of Medicine, Cancer Research & Cell Biology Laboratory, University of Verona, Verona, Italy
| | - Corrado Pedrazzani
- Division of General and Hepatobiliary Surgery, Department of Surgical Sciences, Dentistry, Gynecology, and Pediatrics, University of Verona, Verona, Italy
| | - Alfredo Guglielmi
- Division of General and Hepatobiliary Surgery, Department of Surgical Sciences, Dentistry, Gynecology, and Pediatrics, University of Verona, Verona, Italy
| | - Giovanni De Manzoni
- Division of General and Upper Gastrointestinal Surgery, Department of Surgical Sciences, Dentistry, Gynecology, and Pediatrics, University of Verona, Verona, Italy
| | - Gloria Mazzali
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | | | - Francesco Fantin
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Zamboni
- Geriatric Section, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
33
|
Fallahian F, Ardestani A, Pranckevicius E, Raut CP, Tavakkoli A, Sheu EG. The impact of lipomatous tumors on type 2 diabetes: are adipose-derived tumors metabolically active? J Surg Res 2017; 222:48-54. [PMID: 29273375 DOI: 10.1016/j.jss.2017.09.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/02/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND The metabolic and immunologic properties of adipose tissue are linked to the pathogenesis of type 2 diabetes mellitus. Lipomatous tumors, such as liposarcomas, are rare but can reach significant size. We hypothesized that some lipomatous tumors are metabolically active and can alter systemic glucose homeostasis. METHODS We performed a retrospective study of patients who underwent resection of a lipomatous tumor at a tertiary cancer referral center (2004-2015). We divided patients into nondiabetics, well-controlled diabetics (hemoglobin A1c [HbA1c] < 7), and poorly controlled diabetics (HbA1c ≥ 7). We compared patient demographics, tumor characteristics, and measures of glycemic control among these groups before and after tumor resection. RESULTS We reviewed 217 operations for lipomatous tumors. No differences were observed in tumor characteristics in patients with and without diabetes. However, tumor characteristics differed significantly between the well-controlled and poorly controlled diabetics groups. Patients with poorly controlled diabetes had larger tumors that were more likely to be malignant, retroperitoneal, and well-differentiated. Tumor resection had no detectable impact on diabetes, as assessed by HbA1c, and requirement for diabetic medications. CONCLUSIONS Poorly controlled diabetes was linked to the presence of large, malignant, and retroperitoneal lipomatous tumors. However, in limited follow-up, no detectable impact of tumor resection was apparent on glycemic control. These data suggest that most lipomatous tumors do not exert a clinically relevant impact on glucose homeostasis.
Collapse
Affiliation(s)
- Fedra Fallahian
- University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Ali Ardestani
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elisha Pranckevicius
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ali Tavakkoli
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eric G Sheu
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
34
|
Cleal L, Aldea T, Chau YY. Fifty shades of white: Understanding heterogeneity in white adipose stem cells. Adipocyte 2017; 6:205-216. [PMID: 28949833 PMCID: PMC5638386 DOI: 10.1080/21623945.2017.1372871] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/21/2017] [Accepted: 08/23/2017] [Indexed: 01/03/2023] Open
Abstract
The excessive expansion of white adipose tissue underlies the global obesity epidemic. However, not all fat is equal, and the impact of heterogeneity on the development and expansion of different adipose depots is becoming increasingly apparent. Two mechanisms are responsible for the growth of adipose tissue: hyperplasia (increasing adipocyte number) and hypertrophy (increasing adipocyte size). The former relies on the differentiation of adipocyte stem cells, which reside within the adipose stromal vascular fraction. Many differences in gene expression, adipogenesis, and the response to obesogenic stimuli have been described when comparing adipose stem cells from different depots. Considering that there is disparity in the pathogenicity of the depots, understanding this heterogeneity has clinically relevant implications. Here we review the current knowledge surrounding such differences, in the context of development, expansion and therapeutics. Moreover, given the importance of these differences, we suggest that careful consideration for the precise methodologies used, is essential if we are to truly understand the physiologically relevant consequences of this heterogeneity.
Collapse
Affiliation(s)
- Louise Cleal
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Teodora Aldea
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - You-Ying Chau
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| |
Collapse
|
35
|
Berriel Diaz M, Herzig S, Schafmeier T. Biological Mechanisms for the Effect of Obesity on Cancer Risk: Experimental Evidence. Recent Results Cancer Res 2017; 208:219-242. [PMID: 27909910 DOI: 10.1007/978-3-319-42542-9_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple epidemiological studies demonstrated that overweight and obesity significantly increase the risk of several types of cancer. As the prevalence of obesity is dramatically rising, it is expected that it will represent one of the major lifestyle-associated risk factors for cancer development in the near future. Numerous recent studies expanded knowledge about key players and pathways, which are deregulated in the obese state and potentially promote cancer initiation, progression and aggressiveness via remote and local effects. These players include (but are not limited to) insulin/IGF, adipokines and inflammatory signaling molecules as well as metabolites. Nevertheless, the detailed mechanisms linking obesity and malignant transformation at the systemic, cellular and molecular level still demand further investigation. Additionally, dysfunctional molecular metabolic pathways appear to be specific for distinct cancer entities, thereby yet precluding definition of a common principle. This chapter will present an overview of the current knowledge of molecular nodes linking obesity and cancer and will briefly touch upon potential therapy options addressing metabolic cancer etiologies.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, University Hospital Heidelberg, Heidelberg, Germany.
- Chair Molecular Metabolic Control, Medical Faculty, Technical University Munich, Munich, Germany.
| | - Tobias Schafmeier
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
36
|
Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Ortega VA, Hernández-Lizoain JL, Baixauli J, Becerril S, Rotellar F, Valentí V, Moncada R, Silva C, Salvador J, Frühbeck G. IL-32α-induced inflammation constitutes a link between obesity and colon cancer. Oncoimmunology 2017; 6:e1328338. [PMID: 28811968 DOI: 10.1080/2162402x.2017.1328338] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/20/2022] Open
Abstract
Growing evidence indicates that adipose tissue inflammation is an important mechanism whereby obesity promotes cancer risk and progression. Since IL-32 is an important inflammatory and remodeling factor in obesity and is also related to colon cancer (CC) development, the aim of this study was to explore whether IL-32 could function as an inflammatory factor in human obesity-associated CC promoting a microenvironment favorable for tumor growth. Samples obtained from 84 subjects [27 lean (LN) and 57 obese (OB)] were used in the study. Enrolled subjects were further subclassified according to the established diagnostic protocol for CC (49 without CC and 35 with CC). We show, for the first time, that obesity (p = 0.009) and CC (p = 0.026) increase circulating concentrations of IL-32α. Consistently, we further showed that gene (p < 0.05) and protein (p < 0.01) expression levels of IL-32α were upregulated in VAT from obese patients with CC. Additionally, we revealed that IL32 expression levels are enhanced by hypoxia and inflammation-related factors in HT-29 CC cells as well as that IL-32α is involved in the upregulation of inflammation (IL8, TNF, and CCL2) and extracellular matrix (ECM) remodeling (SPP1 and MMP9) genes in HT-29 cancer cells. Additionally, we also demonstrate that the adipocyte-conditioned medium obtained from obese patients stimulates (p < 0.05) the expression of IL32 in human CC cells. These findings provide evidence of the potential involvement of IL-32 in the development of obesity-associated CC as a pro-inflammatory and ECM remodeling cytokine.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Victor A Ortega
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Fernando Rotellar
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain.,Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain.,Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain.,Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
37
|
Parallel Profiles of Inflammatory and Effector Memory T Cells in Visceral Fat and Liver of Obesity-Associated Cancer Patients. Inflammation 2017; 39:1729-36. [PMID: 27423204 DOI: 10.1007/s10753-016-0407-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the midst of a worsening obesity epidemic, the incidence of obesity-associated morbidities, including cancer, diabetes, cardiac and liver disease is increasing. Insights into mechanisms underlying pathological obesity-associated inflammation are lacking. Both the omentum, the principal component of visceral fat, and liver of obese individuals are sites of excessive inflammation, but to date the T cell profiles of both compartments have not been assessed or compared in a patient cohort with obesity-associated disease. We have previously identified that omentum is enriched with inflammatory cytokines, chemokines and T cells. Here, we compared the inflammatory profile of T cells in the omentum and liver of patients with the obesity-associated malignancy oesophageal adenocarcinoma (OAC). Furthermore, we assessed the secreted cytokine profile in OAC patient serum, omentum and liver to assess systemic and local inflammation. We observed parallel T cell cytokine profiles and phenotypes in the omentum and liver of OAC patients, in particular CD69(+) and inflammatory effector memory T cells. This study reflects similar processes of inflammation and T cell activation in the omentum and liver, and may suggest common targets to modulate pathological inflammation at these sites.
Collapse
|
38
|
Farey JE, Fisher OM, Levert-Mignon AJ, Forner PM, Lord RV. Decreased Levels of Circulating Cancer-Associated Protein Biomarkers Following Bariatric Surgery. Obes Surg 2017; 27:578-585. [PMID: 27525640 PMCID: PMC5306243 DOI: 10.1007/s11695-016-2321-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Epidemiological studies have identified obesity as a major risk factor for cancer in humans, and trials have demonstrated a significant reduction in the incidence of cancer after bariatric surgery. The rapidity of weight loss after bariatric surgery provides an opportunity to identify the molecular changes associated with effective obesity treatment. Indirectly, this may provide some insights into the mechanisms that drive the association between obesity and cancer. We sought to measure circulating cancer-associated proteins before and after laparoscopic sleeve gastrectomy (LSG). METHODS We prospectively enrolled 15 patients undergoing LSG. Thirty-four plasma protein biomarkers thought to be associated with cancer processes were analyzed at baseline and following successful weight loss at 12 weeks using a multiplex bead-based assay. RESULTS Mean excess body weight loss was 44 % at 12-week follow-up. After LSG, a significant reduction in circulating plasma levels was observed for half (17/34) of the proteins assessed: VEGF-A, VEGF-C, VEGF-D, endoglin, PLGF, sFASL, IGFBP-1, IL-18, prolactin, EGF, TGFα, sCD40L, IL-18, TNFα, IL-6, HB-EGF, and PAI-1. Nonsignificant decreases were found for the remaining proteins. CONCLUSIONS Circulating cancer-related biomarker levels were reduced by surgical weight loss, and this benefit was achieved as early as 3 months after operation. The observed reduction in cancer biomarkers may be related to the reported decrease in cancer incidence following bariatric surgery.
Collapse
Affiliation(s)
- John Edward Farey
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Oliver M. Fisher
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Angelique J. Levert-Mignon
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Patrice M. Forner
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Reginald V. Lord
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| |
Collapse
|
39
|
Donohoe CL, Lysaght J, O'Sullivan J, Reynolds JV. Emerging Concepts Linking Obesity with the Hallmarks of Cancer. Trends Endocrinol Metab 2017; 28:46-62. [PMID: 27633129 DOI: 10.1016/j.tem.2016.08.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022]
Abstract
There is compelling epidemiological evidence linking obesity to many tumours; however, the molecular mechanisms fuelling this association are not clearly understood. Emerging evidence links changes in the tumour microenvironment with the obese state, and murine and human studies highlight the relevance of adipose stromal cells (ASCs), including immune cells, both at remote fat depots, such as the omentum, as well as in peritumoural tissue. These obesity-associated changes have been implicated in several hallmarks of cancer, including the chronic inflammatory state and associated cell signalling, epithelial-to-mesenchymal transition (EMT), tumour-related fibrosis, angiogenesis, and genomic instability. Here, we present a summary of developments over the past 5 years, with particular focus on the tumour microenvironment in the obese state.
Collapse
Affiliation(s)
- Claire L Donohoe
- Department of Surgery, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin/St James' Hospital, Dublin, Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin/St James' Hospital, Dublin, Ireland
| | - Jacintha O'Sullivan
- Department of Surgery, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin/St James' Hospital, Dublin, Ireland
| | - John V Reynolds
- Department of Surgery, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin/St James' Hospital, Dublin, Ireland.
| |
Collapse
|
40
|
Zhang Y, Kolonin MG. Cytokine signaling regulating adipose stromal cell trafficking. Adipocyte 2016; 5:369-374. [PMID: 27994950 DOI: 10.1080/21623945.2016.1220452] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/31/2023] Open
Abstract
Adipocyte progenitors, known as adipose stromal cells (ASC), can become mobilized, recruited by tumors, and contribute to cancer progression. Mechanisms underlying ASC trafficking have remained obscure. We recently reported that CXCL1 expressed by cancer cells chemoattracts ASC expressing CXCR1 in obesity. As a candidate mechanism of CXCL1 activation, we identified interleukin (IL)-22, systemic circulation of which is increased in obesity. It has been reported that IL-22 signaling through IL-22R is upstream of CXCL1. Here, we provide evidence that IL-22 expression by leukocytes infiltrating WAT and IL-22R expression by tumors is obesity-dependent. We propose that obesity-associated adipocyte death and the resulting recruitment of leukocytes triggers the IL-22 signaling cascade that induces CXCL1 secretion by cancer cells responsible for ASC trafficking to tumors.
Collapse
|
41
|
The Relationship between Obesity, Prostate Tumor Infiltrating Lymphocytes and Macrophages, and Biochemical Failure. PLoS One 2016; 11:e0159109. [PMID: 27487262 PMCID: PMC4972345 DOI: 10.1371/journal.pone.0159109] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/27/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Obesity reflects a chronic inflammatory environment that may contribute to prostate cancer progression and poor treatment outcomes. However, it is not clear which mechanisms drive this association within the tumor microenvironment. The aim of this pilot study was to examine prostatic inflammation via tumor infiltrating lymphocytes and macrophages characterized by obesity and cancer severity. METHODS We studied paraffin-embedded prostatectomy tissue from 99 participants (63 non-obese and 36 obese) from the Study of Clinical Outcomes, Risk and Ethnicity (University of Pennsylvania). Pathologists analyzed the tissue for type and count of lymphocytes and macrophages, including CD3, CD8, FOXP3, and CD68. Pathology data were linked to clinical and demographic variables. Statistical analyses included frequency tables, Kruskal-Wallis tests, Spearman correlations, and multivariable models. RESULTS We observed positive univariate associations between the number of CD68 cells and tumor grade (p = 0.019). In multivariable analysis, CD8 counts were associated with time to biochemical failure (HR = 1.09, 95% CI = 1.004-1.192, p-value = 0.041.) There were no differences in lymphocytes or macrophages by obesity status or BMI. CONCLUSIONS The number of lymphocytes and macrophages in the tumor microenvironment did not differ by obesity status. However, these inflammation markers were associated with poor prostate cancer outcomes. Further examination of underlying mechanisms that influence obesity-related effects on prostate cancer outcomes is warranted. Such research will guide immunotherapy protocols and weight management as they apply to diverse patient populations and phenotypes.
Collapse
|
42
|
Mechanisms underlying the association between obesity and Hodgkin lymphoma. Tumour Biol 2016; 37:13005-13016. [PMID: 27465553 DOI: 10.1007/s13277-016-5198-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022] Open
Abstract
A solid body of knowledge indicates that overweight and obese subjects are prone to develop cancer, aggressive disease, and death more than their lean counterparts. While obesity has been causally associated with various cancers, only a limited number of studies beheld the link with classical Hodgkin lymphoma (HL). Contemporary meta-analysis and prospective studies confirmed the association of body mass index with HL. Besides epidemiological evidence, excess adiposity is known to influence tumor behavior through adipokines, adipose-derived stem cell migration, and metabolism regulation, and by modulating immunoinflammatory response. Nevertheless, the obesity paradox has been described in few cancers. Considering that adipose tissue is an immunomodulatory organ, and that inflammation is the cornerstone of HL pathophysiology, the rationale for being causally related due to endocrine/paracrine interactions cannot be negligible. In this hypothesis-generating review, we explore the biologically plausible links between excess adiposity and HL in light of recent basic and clinical data, in order to create a basis for understanding the underlying mechanisms and foster applied research. The establishment of an association of excess adiposity with HL will determine public health preventive measures to fight obesity and eventually novel therapeutic approaches in HL patients.
Collapse
|
43
|
Arsenii N, Piardi T, Diébold MD, Chenard MP, Drame M, De Mestier L, Hoeffel C, Pessaux P, Sommacale D, Thiéfin G. Impact of Visceral Obesity on Microvascular Invasion in Hepatocellular Carcinoma. Cancer Invest 2016; 34:271-8. [PMID: 27355681 DOI: 10.1080/07357907.2016.1193747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Our objective was to determine whether visceral obesity is associated with increased microvascular invasion (MVI) in patients surgically treated for hepatocellular carcinoma (HCC). METHODS Data were collected retrospectively in a series of 79 patients treated by surgical resection for HCC, using CT-scan for evaluation of visceral obesity. RESULTS There was no significant association between visceral obesity and MVI (OR = 1.20 (0.38-3.75), p = 0.75). Independent predictive factors of MVI were moderate/poor differentiation, tumor size above 50 mm and underlying cirrhosis. CONCLUSION This study did not support the hypothesis that visceral obesity might promote MVI in patients with HCC.
Collapse
Affiliation(s)
- Nicolae Arsenii
- a Department of Hepato-Gastroenterology , Reims University Hospital , Reims , France
| | - Tullio Piardi
- b Department of General , Digestive and Endocrine Surgery, Reims University Hospital , Reims , France
| | | | | | - Moustapha Drame
- e Department of Research and Innovation , Reims University Hospital , Reims , France
| | - Louis De Mestier
- a Department of Hepato-Gastroenterology , Reims University Hospital , Reims , France
| | - Christine Hoeffel
- f Department of Radiology , Reims University Hospital , Reims , France
| | - Patrick Pessaux
- g Department of Digestive Surgery , University Hospitals of Strasbourg , Strasbourg , France
| | - Daniele Sommacale
- b Department of General , Digestive and Endocrine Surgery, Reims University Hospital , Reims , France
| | - Gérard Thiéfin
- a Department of Hepato-Gastroenterology , Reims University Hospital , Reims , France
| |
Collapse
|
44
|
Robust Early Inflammation of the Peripancreatic Visceral Adipose Tissue During Diet-Induced Obesity in the KrasG12D Model of Pancreatic Cancer. Pancreas 2016; 45:458-65. [PMID: 26495779 PMCID: PMC4854638 DOI: 10.1097/mpa.0000000000000497] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Obesity increases the incidence of multiple types of cancer. Our previous work has shown that a high-fat, high-calorie diet (HFCD) leads to visceral obesity, pancreatic inflammation, and accelerated pancreatic neoplasia in KrasG12D (KC) mice. In this study, we aimed to investigate the effects of an HFCD on visceral adipose inflammation with emphasis on potential differences between distinct visceral adipose depots. METHODS We examined the weight and visceral obesity in both wild-type and KC mice on either control diet (CD) or HFCD. After 3 months, mice were killed for histological examination. Multiplex assays were also performed to obtain cytokine profiles between different adipose depots. RESULTS Both wild-type and KC mice on an HFCD exhibited significantly increased inflammation in the visceral adipose tissue, particularly in the peripancreatic fat (PPF), compared with animals on a CD. This was associated with significantly increased inflammation in the pancreas. Cytokine profiles were different between visceral adipose depots and between mice on the HFCD and CD. CONCLUSIONS Our results clearly demonstrate that an HFCD leads to obesity and inflammation in the visceral adipose tissue, particularly the PPF. These data suggest that obesity-associated inflammation in PPF may accelerate pancreatic neoplasia in KC mice.
Collapse
|
45
|
Veiga-Lopez A, Moeller J, Sreedharan R, Singer K, Lumeng C, Ye W, Pease A, Padmanabhan V. Developmental programming: interaction between prenatal BPA exposure and postnatal adiposity on metabolic variables in female sheep. Am J Physiol Endocrinol Metab 2016; 310:E238-47. [PMID: 26646100 PMCID: PMC4888526 DOI: 10.1152/ajpendo.00425.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 01/05/2023]
Abstract
Among potential contributors for the increased incidence of metabolic diseases is the developmental exposure to endocrine-disrupting chemicals such as bisphenol A (BPA). BPA is an estrogenic chemical used in a variety of consumer products. Evidence points to interactions of BPA with the prevailing environment. The aim of this study was to assess the effects of prenatal exposure to BPA on postnatal metabolic outcomes, including insulin resistance, adipose tissue distribution, adipocyte morphometry, and expression of inflammatory markers in adipose tissue as well as to assess whether postnatal overfeeding would exacerbate these effects. Findings indicate that prenatal BPA exposure leads to insulin resistance in adulthood in the first breeder cohort (study 1), but not in the second cohort (study 2), which is suggestive of potential differences in genetic susceptibility. BPA exposure induced adipocyte hypertrophy in the visceral fat depot without an accompanying increase in visceral fat mass or increased CD68, a marker of macrophage infiltration, in the subcutaneous fat depot. Cohens effect size analysis found the ratio of visceral to subcutaneous fat depot in the prenatal BPA-treated overfed group to be higher compared with the control-overfed group. Altogether, these results suggest that exposure to BPA during fetal life at levels found in humans can program metabolic outcomes that lead to insulin resistance, a forerunner of type 2 diabetes, with postnatal obesity failing to manifest any interaction with prenatal BPA relative to insulin resistance and adipocyte hypertrophy.
Collapse
Affiliation(s)
- Almudena Veiga-Lopez
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan; Department of Animal Sciences, Michigan State University, East Lansing, Michigan
| | - Jacob Moeller
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Rohit Sreedharan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | | | - Carey Lumeng
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Wen Ye
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan; and
| | - Anthony Pease
- Department of Small Animal Clinical Sciences, Michigan State University, East Lansing, Michigan
| | | |
Collapse
|
46
|
Abstract
Epidemiological studies have established an association between obesity, insulin resistance, type 2 diabetes and a number of cancer types. Research has focused predominantly on altered endocrine factors, growth factors and signalling pathways, with little known in man about the immune involvement in the relevant pathophysiological processes. Moreover, in an era of exciting new breakthroughs in cancer immunotherapy, there is also a need to study the safety and efficacy of immunotherapeutics in the complex setting of inflammatory-driven obesity-associated cancer. This review addresses key immune cell subsets underpinning obesity-associated inflammation and describes how such immune compartments might be targeted to prevent and treat obesity-associated cancer. We propose that the modulation, metabolism, migration and abundance of pro- and anti-inflammatory cells and tumour-specific T cells might be therapeutically altered to both restore immune balance, alleviating pathological inflammation, and to improve anti-tumour immune responses in obesity-associated cancer.
Collapse
|
47
|
Lu Y, Ness-Jensen E, Hveem K, Martling A. Metabolic predispositions and increased risk of colorectal adenocarcinoma by anatomical location: a large population-based cohort study in Norway. Am J Epidemiol 2015; 182:883-893. [PMID: 26511906 DOI: 10.1093/aje/kwv141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/28/2015] [Indexed: 01/08/2023] Open
Abstract
Whether different definitions of metabolic syndrome (MetS) are differently associated with colorectal adenocarcinoma (CA) by anatomical location is unclear. A population-based cohort study, the Cohort of Norway (CONOR) Study, was conducted in Norway from 1995 to 2010. Anthropometric measurements, blood samples, and lifestyle data were collected at recruitment. CAs were identified through linkage to the Norwegian Cancer Register. A composite index of MetS as defined by the International Diabetes Federation (IDF) or/and the National Cholesterol Education Program's Adult Treatment Panel III (ATP III) and single components of MetS, including anthropometric factors, blood pressure, lipids, triglycerides, and glucose, were analyzed. Cox proportional hazards regression was performed to estimate hazard ratios and 95% confidence intervals. Significant associations between single MetS components and CA, except for reduced high-density lipoprotein cholesterol and nonfasting glucose levels, were observed. MetS defined by 2 criteria separately showed a similar association with CA in general, and MetS defined by both the IDF and ATP III showed consistent results. Stronger associations were observed in the proximal colon among men (IDF: hazard ratio (HR) = 1.51, 95% confidence interval (CI): 1.24, 1.84; ATP III: HR = 1.40, 95% CI: 1.15, 1.70) and in the rectum among women (IDF: HR = 1.42, 95% CI: 1.07, 1.89; ATP III: HR = 1.43, 95% CI: 1.08, 1.90).
Collapse
|
48
|
Mongan AM, Lynam-Lennon N, Casey R, Maher S, Pidgeon G, Reynolds JV, O'Sullivan J. Visceral obesity stimulates anaphase bridge formation and spindle assembly checkpoint dysregulation in radioresistant oesophageal adenocarcinoma. Clin Transl Oncol 2015; 18:632-40. [PMID: 26474871 DOI: 10.1007/s12094-015-1411-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/14/2015] [Indexed: 01/04/2023]
Abstract
PURPOSE Oesophageal adenocarcinoma is an exemplar model of obesity-associated cancer. Locally advanced disease is treated with neoadjuvant chemoradiotherapy, and survival rates are highest in patients demonstrating a pathological response following neoadjuvant therapy. Given that 55 % of oesophageal adenocarcinoma patients are obese, uncovering the effect of adipose tissue on radioresponse is clinically relevant. This study investigates if adipose tissue activates genomic instability events in radioresponsive (OE33P) and radioresistant (OE33R) oesophageal cancer cell lines and tumour samples. METHODS OE33R and OE33P were cultured with adipose-conditioned media derived from oesophageal adenocarcinoma patients (n = 10). Anaphase bridges, a marker of genomic instability, were enumerated in both cell lines following treatment with adipose media, and normalised to cell number. Genomic instability is regulated by the spindle assembly complex. Expression of two spindle assembly complex genes (MAD2L2, BUB1B) was assessed using qPCR, and validated in patient tumour specimens from viscerally obese (n = 46) and nonobese patients (n = 41). RESULTS Adipose-conditioned media increased anaphase bridging in OE33R (p < 0.0001), with a threefold increase in OE33R compared to OE33P (p < 0.01). Levels of anaphase bridges in OE33R cells correlated with visceral obesity status as measured by waist circumference (R = 0.709, p = 0.03) and visceral fat area (R = 0.794, p = 0.006). Adipose tissue altered expression of MAD2L2 in vitro. In vivo, MAD2L2 expression was higher in viscerally obese oesophageal adenocarcinoma patients compared with nonobese patients (p < 0.05). CONCLUSIONS Anaphase bridge levels are influenced by obesity and radiosensitivity status in oesophageal adenocarcinoma. Furthermore, visceral adipose-conditioned media stimulates dysregulation of the spindle assembly complex in oesophageal adenocarcinoma patients.
Collapse
Affiliation(s)
- A M Mongan
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | - N Lynam-Lennon
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | - R Casey
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | - S Maher
- Cancer Biology & Therapeutics Lab, School of Biological, Biomedical & Environmental Sciences, University of Hull, Cottingham road, Hull, HU6 76X, UK
| | - G Pidgeon
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | - J V Reynolds
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland
| | - J O'Sullivan
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
49
|
Ayyildiz T, Dolar E, Ugras N, Eminler AT, Erturk B, Adim SB, Yerci O. Adipo-R1 and adipo-R2 expression in colorectal adenomas and carcinomas. Asian Pac J Cancer Prev 2015; 16:367-72. [PMID: 25640382 DOI: 10.7314/apjcp.2015.16.1.367] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human adiponectin (ApN), a 30 kDa glycoprotein of 244-amino acids which is predominantly produced by adipocytes, exerts its effects via two receptors, namely adiponectin receptor-1 (adipo-R1) and adiponectin receptor-2 (adipo-R2) with differential binding affinity to globular adiponectin. Adiponectin receptor expression has been studied in several cancer tissues. However, there are no studies of colorectal adenomas which are considered to be precursors for colorectal carcinoma (CRC). OBJECTIVES In the present study, the expression of adipo-R1 and adipo-R2 was investigated immunohistochemically in colorectal adenomas and colorectal carcinoma tissues in an attempt to determine associations with these tumors. MATERIALS AND METHODS The study enrolled 50 CRC patients with tumor resection and 82 patients who were diagnosed with adenomatous polyps, classified as negative for neoplasia, low-grade dysplasia (L-GD) or high- grade dysplasia (H-GD). RESULTS Expression of both adipo-R1 and adipo-R2 was found to be significantly lower in the CRCs than in colorectal adenomas (tubular and tubulovillous, p=0.009 and p<0.001, respectively). Adipo-R1 and adipo-R2 expression was also significantly lower in the CRC group when compared with the groups of patients with low grade dysplasia, high-grade dysplasia or no neoplasia (p=0.012 and p<0.001, respectively). In addition, it was observed that adipo-R2 expression was generally positive in the non-neoplastic group irrespective of the adipo-R2 expression. In the L-GD, H-GD and CRC groups, the adipo-R2 result was positive whenever adipo-R1 result was positive but some patients with negative adipo-R1 had positive adipo-R2 (p<0.001, p=0.004, p<0.001, respectively). CONCLUSIONS This study indicated that ApN may play a role in the progression of colorectal adenomatous polyps to carcinoma through actions on adipo-R1 and adipo-R2 receptors.
Collapse
Affiliation(s)
- Talat Ayyildiz
- Department of Gastroenterology, Medical Faculty, Ondokuz Mayis University, Samsun, Turkey E-mail :
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
There has been a substantial increase in the incidence of esophageal adenocarcinoma over the past 40 years. Meta-analyses of large prospective cohorts and population-based case-control studies demonstrate consistent associations between obesity and the development of adenocarcinoma of the esophagus and esophago-gastric junction, with an approximate doubling of risk of esophageal adenocarcinoma among patients who are obese, and an almost five-fold increased risk among those with BMI >40 kg/m2. The pathologic precursor, specialized intestinal metaplasia in Barrett's esophagus, is also associated with increased adiposity. Epidemiologic evidence suggests that this cancer risk is not solely due to increased gastro-esophageal reflux, and that adipose tissue itself, in particular visceral adipose, may fuel carcinogenesis through the production of adipokines, cytokines, growth factors, and increased inflammation. The robust epidemiologic evidence linking obesity with esophageal adenocarcinoma makes it an exemplar model for investigating the molecular mechanisms underpinning obesity-associated malignant progression, which are discussed in this review.
Collapse
Affiliation(s)
- Jessie A Elliott
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
- b 2 Diabetes Complications Research Centre, Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Dublin 4, Ireland
| | - Claire L Donohoe
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
| | - John V Reynolds
- a 1 Department of Surgery, Trinity Centre for Health Sciences, Trinity College Dublin & St. James' Hospital, Dublin 8, Ireland
| |
Collapse
|