1
|
Papavassiliou KA, Papavassiliou AG. Exploiting the tumor microenvironment and tumor mechanobiology for the treatment of cancer cachexia. Ann Oncol 2024; 35:914-915. [PMID: 39053768 DOI: 10.1016/j.annonc.2024.07.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Affiliation(s)
- K A Papavassiliou
- First University Department of Respiratory Medicine, 'Sotiria' Hospital, Medical School, National and Kapodistrian University of Athens, Athens
| | - A G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
2
|
Chen X, Wu Q, Gong W, Ju S, Fan J, Gao X, Liu X, Lei X, Liu S, Ming X, Wang Q, Fu M, Song Y, Wang Y, Zhan Q. GRP75 triggers white adipose tissue browning to promote cancer-associated cachexia. Signal Transduct Target Ther 2024; 9:253. [PMID: 39327432 PMCID: PMC11427701 DOI: 10.1038/s41392-024-01950-w] [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: 01/06/2024] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
Cachexia, which affects 50-80% of cancer patients, is a debilitating syndrome that leads to 20% of cancer-related deaths. A key feature of cachexia is adipose tissue atrophy, but how it contributes to the development of cachexia is poorly understood. Here, we demonstrate in mouse models of cancer cachexia that white adipose tissue browning, which can be a characteristic early-onset manifestation, occurs prior to the loss of body weight and skeletal muscle wasting. By analysing the proteins differentially expressed in extracellular vesicles derived from cachexia-inducing tumours, we identified a molecular chaperone, Glucose-regulated protein 75 (GRP75), as a critical mediator of adipocyte browning. Mechanistically, GRP75 binds adenine nucleotide translocase 2 (ANT2) to form a GRP75-ANT2 complex. Strikingly, stabilized ANT2 enhances its interaction with uncoupling protein 1, leading to elevated expression of the latter, which, in turn, promotes adipocyte browning. Treatment with withanone, a GRP75 inhibitor, can reverse this browning and alleviate cachectic phenotypes in vivo. Overall, our findings reveal a novel mechanism by which tumour-derived GRP75 regulates white adipose tissue browning during cachexia development and suggest a potential white adipose tissue-centred targeting approach for early cachexia intervention.
Collapse
Affiliation(s)
- Xu Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Wei Gong
- Peking University-Yunnan Baiyao International Medical Research Center, 100191, Beijing, China
| | - Shaolong Ju
- Center for Infection and Immunity, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jiawen Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiaohan Gao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xingyang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiao Lei
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Siqi Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiangdong Ming
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Qianyu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Ming Fu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China.
- Soochow University Cancer Institute, Suzhou, 215000, China.
| |
Collapse
|
3
|
Belev B, Vičić I, Sedlić F, Prtorić M, Soče M, Prejac J, Potočki S, Silovski T, Herceg D, Kulić A. Salivary Interleukin-13 and Transforming Growth Factor Beta as Potential Biomarkers of Cancer Cachexia. Cancers (Basel) 2024; 16:3035. [PMID: 39272892 PMCID: PMC11394294 DOI: 10.3390/cancers16173035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer cachexia is a syndrome characterized by weight and muscle loss and functional impairment, strongly influencing survival in cancer patients. In this study, we aimed to establish the role of saliva cytokine measurement in cancer cachexia investigation and define two potential independent salivary biomarkers of the condition. METHODS serum and saliva specimens were obtained from 78 patients. Forty-six patients were non-cachectic, and 32 patients were cachectic (per SCRINIO group criteria), all with metastatic solid tumors. Commercial ELISA kits were used to determine the salivary and serum concentrations of interleukin 13 (IL-13) and transforming growth factor beta (TGF-β) in two patient groups and healthy controls. Laboratory values were obtained from the hospital information system, and weight and height were measured at the time of sampling. RESULTS A statistically significant difference was observed between the groups in saliva IL-13 concentrations but no difference in serum concentrations. Statistically significant differences were also observed between the groups in saliva and serum concentrations of TGF-β. Logistic regression analysis has identified salivary IL-13 and TGF-β as independent factors for cancer cachexia. CONCLUSIONS We demonstrated saliva as a valuable specimen for cachexia investigation and established IL-13 and TGF-β as potential cancer cachexia biomarkers. Further research is needed to evaluate these findings.
Collapse
Affiliation(s)
- Borislav Belev
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ivan Vičić
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Filip Sedlić
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Matko Prtorić
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Majana Soče
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Juraj Prejac
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Slavica Potočki
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tajana Silovski
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Davorin Herceg
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Dental Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ana Kulić
- Department of Oncology, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
4
|
Pannkuk EL, Moore MS, Bansal S, Kumar K, Suman S, Howell D, Kath JA, Kurta A, Reeder DM, Field KA. White adipose tissue remodeling in Little Brown Myotis (Myotis lucifugus) with white-nose syndrome. Metabolomics 2024; 20:100. [PMID: 39190217 DOI: 10.1007/s11306-024-02165-4] [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: 02/28/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
White-nose syndrome (WNS) is a fungal wildlife disease of bats that has caused precipitous declines in certain Nearctic bat species. A key driver of mortality is premature exhaustion of fat reserves, primarily white adipose tissue (WAT), that bats rely on to meet their metabolic needs during winter. However, the pathophysiological and metabolic effects of WNS have remained ill-defined. To elucidate metabolic mechanisms associated with WNS mortality, we infected a WNS susceptible species, the Little Brown Myotis (Myotis lucifugus), with Pseudogymnoascus destructans (Pd) and collected WAT biopsies for histology and targeted lipidomics. These results were compared to the WNS-resistant Big Brown Bat (Eptesicus fuscus). A similar distribution in broad lipid class was observed in both species, with total WAT primarily consisting of triacylglycerides. Baseline differences in WAT chemical composition between species showed that higher glycerophospholipids (GPs) levels in E. fuscus were dominated by unsaturated or monounsaturated moieties and n-6 (18:2, 20:2, 20:3, 20:4) fatty acids. Conversely, higher GP levels in M. lucifugus WAT were primarily compounds containing n-3 (20:5 and 22:5) fatty acids. Following Pd-infection, we found that perturbation to WAT reserves occurs in M. lucifugus, but not in the resistant E. fuscus. A total of 66 GPs (primarily glycerophosphocholines and glycerophosphoethanolamines) were higher in Pd-infected M. lucifugus, indicating perturbation to the WAT structural component. In addition to changes in lipid chemistry, smaller adipocyte sizes and increased extracellular matrix deposition was observed in Pd-infected M. lucifugus. This is the first study to describe WAT GP composition of bats with different susceptibilities to WNS and highlights that recovery from WNS may require repair from adipose remodeling in addition to replenishing depot fat during spring emergence.
Collapse
Affiliation(s)
- Evan L Pannkuk
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, 3970 Reservoir Road, NW, New Research Building, Room E504, Washington, DC, 20057, USA.
- Center for Metabolomic Studies, Georgetown University, Washington, DC, USA.
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| | - Marianne S Moore
- Department of Biological Sciences, University of the Virgin Islands, St. Thomas, USA
| | - Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Kamendra Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Daryl Howell
- Iowa Department of Natural Resources, Des Moines, IA, USA
| | - Joseph A Kath
- Illinois Department of Natural Resources, Springfield, IL, USA
| | - Allen Kurta
- Department of Biology, Eastern Michigan University, Ypsilanti, MI, USA
| | - DeeAnn M Reeder
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - Kenneth A Field
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| |
Collapse
|
5
|
Compton SLE, Heymsfield SB, Brown JC. Nutritional Mechanisms of Cancer Cachexia. Annu Rev Nutr 2024; 44:77-98. [PMID: 39207878 DOI: 10.1146/annurev-nutr-062122-015646] [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] [Indexed: 09/04/2024]
Abstract
Cancer cachexia is a complex systemic wasting syndrome. Nutritional mechanisms that span energy intake, nutrient metabolism, body composition, and energy balance may be impacted by, and may contribute to, the development of cachexia. To date, clinical management of cachexia remains elusive. Leaning on discoveries and novel methodologies from other fields of research may bolster new breakthroughs that improve nutritional management and clinical outcomes. Characteristics that compare and contrast cachexia and obesity may reveal opportunities for cachexia research to adopt methodology from the well-established field of obesity research. This review outlines the known nutritional mechanisms and gaps in the knowledge surrounding cancer cachexia. In parallel, we present how obesity may be a different side of the same coin and how obesity research has tackled similar research questions. We present insights into how cachexia research may utilize nutritional methodology to expand our understanding of cachexia to improve definitions and clinical care in future directions for the field.
Collapse
Affiliation(s)
- Stephanie L E Compton
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| | - Steven B Heymsfield
- Metabolism and Body Composition Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Justin C Brown
- Cancer Energetics Unit, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA;
| |
Collapse
|
6
|
Ahmad Y, Seo DS, Jang Y. Metabolic Effects of Ketogenic Diets: Exploring Whole-Body Metabolism in Connection with Adipose Tissue and Other Metabolic Organs. Int J Mol Sci 2024; 25:7076. [PMID: 39000187 PMCID: PMC11241756 DOI: 10.3390/ijms25137076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The ketogenic diet (KD) is characterized by minimal carbohydrate, moderate protein, and high fat intake, leading to ketosis. It is recognized for its efficiency in weight loss, metabolic health improvement, and various therapeutic interventions. The KD enhances glucose and lipid metabolism, reducing triglycerides and total cholesterol while increasing high-density lipoprotein levels and alleviating dyslipidemia. It significantly influences adipose tissue hormones, key contributors to systemic metabolism. Brown adipose tissue, essential for thermogenesis and lipid combustion, encounters modified UCP1 levels due to dietary factors, including the KD. UCP1 generates heat by uncoupling electron transport during ATP synthesis. Browning of the white adipose tissue elevates UCP1 levels in both white and brown adipose tissues, a phenomenon encouraged by the KD. Ketone oxidation depletes intermediates in the Krebs cycle, requiring anaplerotic substances, including glucose, glycogen, or amino acids, for metabolic efficiency. Methylation is essential in adipogenesis and the body's dietary responses, with DNA methylation of several genes linked to weight loss and ketosis. The KD stimulates FGF21, influencing metabolic stability via the UCP1 pathways. The KD induces a reduction in muscle mass, potentially involving anti-lipolytic effects and attenuating proteolysis in skeletal muscles. Additionally, the KD contributes to neuroprotection, possesses anti-inflammatory properties, and alters epigenetics. This review encapsulates the metabolic effects and signaling induced by the KD in adipose tissue and major metabolic organs.
Collapse
Affiliation(s)
- Yusra Ahmad
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, Republic of Korea
| | - Dong Soo Seo
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, Republic of Korea
| | - Younghoon Jang
- Department of Biology and Chemistry, Changwon National University, Changwon 51140, Republic of Korea
| |
Collapse
|
7
|
Yue M, Qin Z, Hu L, Ji H. Understanding cachexia and its impact on lung cancer and beyond. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:95-105. [PMID: 39169934 PMCID: PMC11332896 DOI: 10.1016/j.pccm.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 08/23/2024]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by loss of body weight secondary to skeletal muscle atrophy and adipose tissue wasting. It not only has a significant impact on patients' quality of life but also reduces the effectiveness and tolerability of anticancer therapy, leading to poor clinical outcomes. Lung cancer is a prominent global health concern, and the prevalence of cachexia is high among patients with lung cancer. In this review, we integrate findings from studies of lung cancer and other types of cancer to provide an overview of recent advances in cancer cachexia. Our focus includes topics such as the clinical criteria for diagnosis and staging, the function and mechanism of selected mediators, and potential therapeutic strategies for clinical application. A comprehensive summary of current studies will improve our understanding of the mechanisms underlying cachexia and contribute to the identification of high-risk patients, the development of effective treatment strategies, and the design of appropriate therapeutic regimens for patients at different disease stages.
Collapse
Affiliation(s)
- Meiting Yue
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Qin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
8
|
Zhang L, Bonomi PD. Immune System Disorder and Cancer-Associated Cachexia. Cancers (Basel) 2024; 16:1709. [PMID: 38730660 PMCID: PMC11083538 DOI: 10.3390/cancers16091709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer-associated cachexia (CAC) is a debilitating condition marked by muscle and fat loss, that is unresponsive to nutritional support and contributes significantly to morbidity and mortality in patients with cancer. Immune dysfunction, driven by cytokine imbalance, contributes to CAC progression. This review explores the potential relationship between CAC and anti-cancer immune response in pre-clinical and clinical studies. Pre-clinical studies showcase the involvement of cytokines like IL-1β, IL-6, IL-8, IFN-γ, TNF-α, and TGF-β, in CAC. IL-6 and TNF-α, interacting with muscle and adipose tissues, induce wasting through JAK/STAT and NF-κB pathways. Myeloid-derived suppressor cells (MDSCs) exacerbate CAC by promoting inflammation. Clinical studies confirm elevated pro-inflammatory cytokines (IL-6, IL-8, TNFα) and immune markers like the neutrophil-to-lymphocyte ratio (NLR) in patients with CAC. Thus, immunomodulatory mechanisms involved in CAC may impact the anti-neoplastic immune response. Inhibiting CAC mechanisms could enhance anti-cancer therapies, notably immunotherapy. R-ketorolac, a new immunomodulator, reversed the weight loss and increased survival in mice. Combining these agents with immunotherapy may benefit patients with cancer experiencing CAC. Further research is vital to understand the complex interplay between tumor-induced immune dysregulation and CAC during immunotherapy.
Collapse
Affiliation(s)
| | - Philip D. Bonomi
- Division of Hematology/Oncology, Rush University Medical Center, Chicago, IL 60612, USA;
| |
Collapse
|
9
|
Zhang Q, Lu C, Lu F, Liao Y, Cai J, Gao J. Challenges and opportunities in obesity: the role of adipocytes during tissue fibrosis. Front Endocrinol (Lausanne) 2024; 15:1365156. [PMID: 38686209 PMCID: PMC11056552 DOI: 10.3389/fendo.2024.1365156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/01/2024] [Indexed: 05/02/2024] Open
Abstract
Obesity is a chronic disease that affects the energy balance of the whole body. In addition to increasing fat mass, tissue fibrosis occurred in white adipose tissue in obese condition. Fibrosis is the over-activation of fibroblasts leading to excessive accumulation of extracellular matrix, which could be caused by various factors, including the status of adipocytes. The morphology of adipocytes responds rapidly and dynamically to nutrient fluctuations. Adaptive hypertrophy of normal adipocytes protects peripheral organs from damage from lipotoxicity. However, the biological behavior of hypertrophic adipocytes in chronic obesity is abnormally altered. Adipocytes lead to fibrotic remodeling of the extracellular matrix by inducing unresolved chronic inflammation, persistent hypoxia, and increasing myofibroblast numbers. Moreover, adipocyte-induced fibrosis not only restricts the flexible expansion and contraction of adipose tissue but also initiates the development of various diseases through cellular autonomic and paracrine effects. Regarding anti-fibrotic therapy, dysregulated intracellular signaling and epigenetic changes represent potential candidate targets. Thus, modulation of adipocytes may provide potential therapeutic avenues for reversing pathological fibrosis in adipose tissue and achieving the anti-obesity purpose.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chongxuan Lu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjun Liao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junrong Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Ma D, Qu Y, Wu T, Liu X, Cai L, Wang Y. Excessive fat expenditure in MCT-induced heart failure rats is associated with BMAL1/REV-ERBα circadian rhythmic loop disruption. Sci Rep 2024; 14:8128. [PMID: 38584196 PMCID: PMC10999456 DOI: 10.1038/s41598-024-58577-8] [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: 11/22/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
Fat loss predicts adverse outcomes in advanced heart failure (HF). Disrupted circadian clocks are a primary cause of lipid metabolic issues, but it's unclear if this disruption affects fat expenditure in HF. To address this issue, we investigated the effects of disruption of the BMAL1/REV-ERBα circadian rhythmic loop on adipose tissue metabolism in HF.50 Wistar rats were initially divided into control (n = 10) and model (n = 40) groups. The model rats were induced with HF via monocrotaline (MCT) injections, while the control group received equivalent solvent injections. After establishing the HF model, the model group was further subdivided into four groups: normal rhythm (LD), inverted rhythm (DL), lentivirus vector carrying Bmal1 short hairpin RNA (LV-Bmal1 shRNA), and empty lentivirus vector control (LV-Control shRNA) groups, each with 10 rats. The DL subgroup was exposed to a reversed light-dark cycle of 8 h: 16 h (dark: light), while the rest adhered to normal light-dark conditions (light: dark 12 h: 12 h). Histological analyses were conducted using H&E, Oil Red O, and Picrosirius red stains to examine adipose and liver tissues. Immunohistochemical staining, RT-qPCR, and Western blotting were performed to detect markers of lipolysis, lipogenesis, and beiging of white adipose tissue (WAT), while thermogenesis indicators were detected in brown adipose tissue (BAT). The LD group rats exhibited decreased levels of BMAL1 protein, increased levels of REV-ERBα protein, and disrupted circadian circuits in adipose tissue compared to controls. Additionally, HF rats showed reduced adipose mass and increased ectopic lipid deposition, along with smaller adipocytes containing lower lipid content and fibrotic adipose tissue. In the LD group WAT, expression of ATGL, HSL, PKA, and p-PKA proteins increased, alongside elevated mRNA levels of lipase genes (Hsl, Atgl, Peripilin) and FFA β-oxidation genes (Cpt1, acyl-CoA). Conversely, lipogenic gene expression (Scd1, Fas, Mgat, Dgat2) decreased, while beige adipocyte markers (Cd137, Tbx-1, Ucp-1, Zic-1) and UCP-1 protein expression increased. In BAT, HF rats exhibited elevated levels of PKA, p-PKA, and UCP-1 proteins, along with increased expression of thermogenic genes (Ucp-1, Pparγ, Pgc-1α) and lipid transportation genes (Cd36, Fatp-1, Cpt-1). Plasma NT-proBNP levels were higher in LD rats, accompanied by elevated NE and IL-6 levels in adipose tissue. Remarkably, morphologically, the adipocytes in the DL and LV-Bmal1 shRNA groups showed reduced size and lower lipid content, while lipid deposition in the liver was more pronounced in these groups compared to the LD group. At the gene/protein level, the BMAL1/REV-ERBα circadian loop exhibited severe disruption in LV-Bmal1 shRNA rats compared to LD rats. Additionally, there was increased expression of lipase genes, FFA β oxidation genes, and beige adipocyte markers in WAT, as well as higher expression of thermogenic genes and lipid transportation genes in BAT. Furthermore, plasma NT-proBNP levels and adipose tissue levels of NE and IL-6 were elevated in LV-Bmal1 shRNA rats compared with LD rats. The present study demonstrates that disruption of the BMAL1/REV-ERBα circadian rhythmic loop is associated with fat expenditure in HF. This result suggests that restoring circadian rhythms in adipose tissue may help counteract disorders of adipose metabolism and reduce fat loss in HF.
Collapse
Affiliation(s)
- Dufang Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yiwei Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Tao Wu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Xue Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Lu Cai
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China.
| |
Collapse
|
11
|
Neshan M, Tsilimigras DI, Han X, Zhu H, Pawlik TM. Molecular Mechanisms of Cachexia: A Review. Cells 2024; 13:252. [PMID: 38334644 PMCID: PMC10854699 DOI: 10.3390/cells13030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
Cachexia is a condition characterized by substantial loss of body weight resulting from the depletion of skeletal muscle and adipose tissue. A considerable fraction of patients with advanced cancer, particularly those who have been diagnosed with pancreatic or gastric cancer, lung cancer, prostate cancer, colon cancer, breast cancer, or leukemias, are impacted by this condition. This syndrome manifests at all stages of cancer and is associated with an unfavorable prognosis. It heightens the susceptibility to surgical complications, chemotherapy toxicity, functional impairments, breathing difficulties, and fatigue. The early detection of patients with cancer cachexia has the potential to enhance both their quality of life and overall survival rates. Regarding this matter, blood biomarkers, although helpful, possess certain limitations and do not exhibit universal application. Additionally, the available treatment options for cachexia are currently limited, and there is a lack of comprehensive understanding of the underlying molecular pathways associated with this condition. Thus, this review aims to provide an overview of molecular mechanisms associated with cachexia and potential therapeutic targets for the development of effective treatments for this devastating condition.
Collapse
Affiliation(s)
- Mahdi Neshan
- Department of General Surgery, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd 8915887857, Iran;
| | - Diamantis I. Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Xu Han
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| |
Collapse
|
12
|
Bakopoulos D, Golenkina S, Dark C, Christie EL, Sánchez-Sánchez BJ, Stramer BM, Cheng LY. Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model. EMBO Rep 2023; 24:e57695. [PMID: 38014610 DOI: 10.15252/embr.202357695] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.
Collapse
Affiliation(s)
- Daniel Bakopoulos
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Brian M Stramer
- Kings College London, Randall Centre for Cell & Molecular Biophysics, London, UK
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
13
|
Qu Y, Wang Y, Wu T, Liu X, Wang H, Ma D. A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes. Lipids Health Dis 2023; 22:211. [PMID: 38041133 PMCID: PMC10691093 DOI: 10.1186/s12944-023-01967-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
Cardiac cachexia is a deadly consequence of advanced heart failure that is characterised by the dysregulation of adipose tissue homeostasis. Once cachexia occurs with heart failure, it prevents the normal treatment of heart failure and increases the risk of death. Targeting adipose tissue is an important approach to treating cardiac cachexia, but the pathogenic mechanisms are still unknown, and there are no effective therapies available. Transcriptomics, metabolomics, and lipidomics were used to examine the underlying mechanisms of cardiac cachexia. Transcriptomics investigation of cardiac cachexia adipose tissue revealed that genes involved in fibrosis and monocyte/macrophage migration were increased and strongly interacted. The ECM-receptor interaction pathway was primarily enriched, as shown by KEGG enrichment analysis. In addition, gene set enrichment analysis revealed that monocyte chemotaxis/macrophage migration and fibrosis gene sets were upregulated in cardiac cachexia. Metabolomics enrichment analysis demonstrated that the sphingolipid signalling pathway is important for adipose tissue remodelling in cardiac cachexia. Lipidomics analysis showed that the adipose tissue of rats with cardiac cachexia had higher levels of sphingolipids, including Cer and S1P. Moreover, combined multiomics analysis suggested that the sphingolipid metabolic pathway was associated with inflammatory-fibrotic changes in adipose tissue. Finally, the key indicators were validated by experiments. In conclusion, this study described a mechanism by which the sphingolipid signalling pathway was involved in adipose tissue remodelling by inducing inflammation and fat fibrosis in cardiac cachexia.
Collapse
Affiliation(s)
- Yiwei Qu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yong Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tao Wu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huaizhe Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dufang Ma
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
14
|
Bondi D, Bevere M, Piccirillo R, Sorci G, Di Felice V, Re Cecconi AD, D'Amico D, Pietrangelo T, Fulle S. Integrated procedures for accelerating, deepening, and leading genetic inquiry: A first application on human muscle secretome. Mol Genet Metab 2023; 140:107705. [PMID: 37837864 DOI: 10.1016/j.ymgme.2023.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/15/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
PURPOSE Beyond classical procedures, bioinformatic-assisted approaches and computational biology offer unprecedented opportunities for scholars. However, these amazing possibilities still need epistemological criticism, as well as standardized procedures. Especially those topics with a huge body of data may benefit from data science (DS)-assisted methods. Therefore, the current study dealt with the combined expert-assisted and DS-assisted approaches to address the broad field of muscle secretome. We aimed to apply DS tools to fix the literature research, suggest investigation targets with a data-driven approach, predict possible scenarios, and define a workflow. METHODS Recognized scholars with expertise on myokines were invited to provide a list of the most important myokines. GeneRecommender, GeneMANIA, HumanNet, and STRING were selected as DS tools. Networks were built on STRING and GeneMANIA. The outcomes of DS tools included the top 5 recommendations. Each expert-led discussion has been then integrated with an DS-led approach to provide further perspectives. RESULTS Among the results, 11 molecules had already been described as bona-fide myokines in literature, and 11 molecules were putative myokines. Most of the myokines and the putative myokines recommended by the DS tools were described as present in the cargo of extracellular vesicles. CONCLUSIONS Including both supervised and unsupervised learning methods, as well as encompassing algorithms focused on both protein interaction and gene represent a comprehensive approach to tackle complex biomedical topics. DS-assisted methods for reviewing existent evidence, recommending targets of interest, and predicting original scenarios are worth exploring as in silico recommendations to be integrated with experts' ideas for optimizing molecular studies.
Collapse
Affiliation(s)
- Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Michele Bevere
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.
| | - Rosanna Piccirillo
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Guglielmo Sorci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Valentina Di Felice
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Andrea David Re Cecconi
- Department of Neurosciences, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy.
| | - Daniela D'Amico
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy.
| | - Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" Chieti - Pescara, Chieti, Italy; Interuniversity Institute of Myology (IIM), Perugia, Italy.
| |
Collapse
|
15
|
Robinson TP, Hamidi T, Counts B, Guttridge DC, Ostrowski MC, Zimmers TA, Koniaris LG. The impact of inflammation and acute phase activation in cancer cachexia. Front Immunol 2023; 14:1207746. [PMID: 38022578 PMCID: PMC10644737 DOI: 10.3389/fimmu.2023.1207746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
The development of cachexia in the setting of cancer or other chronic diseases is a significant detriment for patients. Cachexia is associated with a decreased ability to tolerate therapies, reduction in ambulation, reduced quality of life, and increased mortality. Cachexia appears intricately linked to the activation of the acute phase response and is a drain on metabolic resources. Work has begun to focus on the important inflammatory factors associated with the acute phase response and their role in the immune activation of cachexia. Furthermore, data supporting the liver, lung, skeletal muscle, and tumor as all playing a role in activation of the acute phase are emerging. Although the acute phase is increasingly being recognized as being involved in cachexia, work in understanding underlying mechanisms of cachexia associated with the acute phase response remains an active area of investigation and still lack a holistic understanding and a clear causal link. Studies to date are largely correlative in nature, nonetheless suggesting the possibility for a role for various acute phase reactants. Herein, we examine the current literature regarding the acute phase response proteins, the evidence these proteins play in the promotion and exacerbation of cachexia, and current evidence of a therapeutic potential for patients.
Collapse
Affiliation(s)
- Tyler P. Robinson
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tewfik Hamidi
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Brittany Counts
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Denis C. Guttridge
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Michael C. Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Leonidas G. Koniaris
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| |
Collapse
|
16
|
Shivnani P, Shekhawat S, Prajapati A. Cancer Cachexia and breast cancer stem cell signalling - A crosstalk of signalling molecules. Cell Signal 2023; 110:110847. [PMID: 37557973 DOI: 10.1016/j.cellsig.2023.110847] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Cancer Cachexia is a condition characterized by the involuntary loss of lean body mass, a negative protein and energy balance, and systemic inflammation. This syndrome profoundly impacts the patient's quality of life and is linked to poor chemotherapy response and reduced survival. Despite multiple mechanisms being implicated in its development, and various cytokines believed to contribute to the persistent catabolic state, cachexia is still not fully recognized and is often left untreated. Cachexia is caused by altered metabolic adaptation and lack of anticactic therapy due to systemic cytokines promoting and fuelling cancer growth. The exact molecular mechanisms and clinical endpoints remain poorly defined. It has an occurrence rate of 30%-80%, accounting for 20% of total cancer mortality. Tumor cells remodel the microenvironment suitable for their proliferation, wherein they communicate with fibroblast cells to modulate their expression and induce tumor progressive cytokines. Several studies have reported its strong correlation with systemic cytokines that initiate and aggravate the condition. Plenty of studies show the prominent role of cancer-induced cachexia in pancreatic cancer, colon cancer, and lung cancer. However, limited data are available for breast cancer-induced cachexia, highlighting the need for studying it. Breast cancer stem cells (BCSCs) are a prominently explored area in breast cancer research. They are characterized by CD44+/CD24-/ALDH+ expression and are a focus of cancer research. They are a source of renewal and differentiation within the tumor environment and are responsible for progression, and chemotherapeutic resistance. The tumor microenvironment and its cytokines are responsible for maintaining and inducing their differentiation. Cytokines significantly impact BCSC development and self-renewal, stimulating or inhibiting proliferation depending on cytokine and environment. Pro-inflammatory mediators like IL-6, TNF-α, and IL-8 increase proliferation, promoting tumor growth. Experimental models and clinical studies have shown a direct relationship between cytokines and BCSC proliferation. Several of them seem to be interconnected as they initiate signalling down different pathways but converge at BCSC increase and tumor proliferation. This review highlights the common pathways between cachexia and BCSC signalling, to identify potential therapeutic targets that can aid both conditions.
Collapse
Affiliation(s)
- Priyanka Shivnani
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India
| | - Saroj Shekhawat
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India
| | - Akhilesh Prajapati
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India.
| |
Collapse
|
17
|
Santos KBNH, Knobl P, Henriques F, Lopes MA, Franco FO, Bueno LL, Farmer SR, Batista ML. Pathological beige remodeling induced by cancer cachexia depends on the disease severity and involves mainly the trans-differentiation of mature white adipocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558327. [PMID: 37781595 PMCID: PMC10541144 DOI: 10.1101/2023.09.18.558327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
In cancer associated cachexia (CAC), white adipose tissue undergoes morphofunctional and inflammatory changes that lead to tissue dysfunction and remodeling. In addition to metabolic changes in white adipose tissues (WAT), adipose tissue atrophy has been implicated in several clinical complications and poor prognoses associated with cachexia. Adipocyte atrophy may be associated with increased beige remodeling in human CAC as evidenced by the "beige remodeling" observed in preclinical models of CAC. Even though beige remodeling is associated with CAC-induced WAT dysfunction, there are still some open questions regarding their cellular origins. In this study, we investigated the development of beige remodeling in CAC from a broader perspective. In addition, we used a grading system to identify the scAT as being affected by mice weight loss early and intensely. Using different in vitro and ex-vivo techniques, we demonstrated that Lewis LLC1 cells can induce a switch from white to beige adipocytes, which is specific to this type of tumor cell. During the more advanced stages of CAC, beige adipocytes are mainly formed from the transdifferentiation of cells. According to our results, humanizing the CAC classification system is an efficient approach to defining the onset of the syndrome in a more homogeneous manner. Pathological beige remodeling occurred early in the disease course and exhibited phenotypic characteristics specific to LLC cells' secretomes. Developing therapeutic strategies that recruit beige adipocytes in vivo may be better guided by an understanding of the cellular origins of beige adipocytes emitted by CAC.
Collapse
Affiliation(s)
| | - Pamela Knobl
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - Felipe Henriques
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts,USA
| | - Magno A. Lopes
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - Felipe O. Franco
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - Luana L. Bueno
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - Stephen R. Farmer
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA 02215, USA
| | - Miguel L. Batista
- Department of Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
- Department of Biochemistry, School of Medicine, Boston University, Boston, MA 02215, USA
| |
Collapse
|
18
|
Setiawan T, Sari IN, Wijaya YT, Julianto NM, Muhammad JA, Lee H, Chae JH, Kwon HY. Cancer cachexia: molecular mechanisms and treatment strategies. J Hematol Oncol 2023; 16:54. [PMID: 37217930 DOI: 10.1186/s13045-023-01454-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/13/2023] [Indexed: 05/24/2023] Open
Abstract
Muscle wasting is a consequence of physiological changes or a pathology characterized by increased catabolic activity that leads to progressive loss of skeletal muscle mass and strength. Numerous diseases, including cancer, organ failure, infection, and aging-associated diseases, are associated with muscle wasting. Cancer cachexia is a multifactorial syndrome characterized by loss of skeletal muscle mass, with or without the loss of fat mass, resulting in functional impairment and reduced quality of life. It is caused by the upregulation of systemic inflammation and catabolic stimuli, leading to inhibition of protein synthesis and enhancement of muscle catabolism. Here, we summarize the complex molecular networks that regulate muscle mass and function. Moreover, we describe complex multi-organ roles in cancer cachexia. Although cachexia is one of the main causes of cancer-related deaths, there are still no approved drugs for cancer cachexia. Thus, we compiled recent ongoing pre-clinical and clinical trials and further discussed potential therapeutic approaches for cancer cachexia.
Collapse
Affiliation(s)
- Tania Setiawan
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Ita Novita Sari
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore
| | - Yoseph Toni Wijaya
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Nadya Marcelina Julianto
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Jabir Aliyu Muhammad
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Hyeok Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Ji Heon Chae
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea
| | - Hyog Young Kwon
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea.
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, 31151, Republic of Korea.
| |
Collapse
|
19
|
das Neves RX, Yamashita AS, Riccardi DMR, Köhn-Gaone J, Camargo RG, Neto NI, Caetano D, Gomes SP, Santos FH, Lima JDCC, Batista ML, Rosa-Neto JC, Martins De Alcântara PS, Maximiano LF, Otoch JP, Trinchieri G, Tirnitz-Parker JEE, Seelaender M. Cachexia causes time-dependent activation of the inflammasome in the liver. J Cachexia Sarcopenia Muscle 2023. [PMID: 37177862 PMCID: PMC10401524 DOI: 10.1002/jcsm.13236] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Cachexia is a wasting syndrome associated with systemic inflammation and metabolic disruption. Detection of the early signs of the disease may contribute to the effective attenuation of associated symptoms. Despite playing a central role in the control of metabolism and inflammation, the liver has received little attention in cachexia. We previously described relevant disruption of metabolic pathways in the organ in an animal model of cachexia, and herein, we adopt the same model to investigate temporal onset of inflammation in the liver. The aim was thus to study inflammation in rodent liver in the well-characterized cachexia model of Walker 256 carcinosarcoma and, in addition, to describe inflammatory alterations in the liver of one cachectic colon cancer patient, as compared to one control and one weight-stable cancer patient. METHODS Colon cancer patients (one weight stable [WSC] and one cachectic [CC]) and one patient undergoing surgery for cholelithiasis (control, n = 1) were enrolled in the study, after obtainment of fully informed consent. Eight-week-old male rats were subcutaneously inoculated with a Walker 256 carcinosarcoma cell suspension (2 × 107 cells in 1.0 mL; tumour-bearing [T]; or phosphate-buffered saline-controls [C]). The liver was excised on Days 0 (n = 5), 7 (n = 5) and 14 (n = 5) after tumour cell injection. RESULTS In rodent cachexia, we found progressively higher numbers of CD68+ myeloid cells in the liver along cancer-cachexia development. Similar findings are described for CC, whose liver showed infiltration of the same cell type, compared with both WSC and control patient organs. In advanced rodent cachexia, hepatic phosphorylated c-Jun N-terminal kinase protein content and the inflammasome pathway protein expression were increased in relation to baseline (P < 0.05). These changes were accompanied by augmented expression of the active interleukin-1β (IL-1β) form (P < 0.05 for both circulating and hepatic content). CONCLUSIONS The results show that cancer cachexia is associated with an increase in the number of myeloid cells in rodent and human liver and with modulation of hepatic inflammasome pathway. The latter contributes to the aggravation of systemic inflammation, through increased release of IL-1β.
Collapse
Affiliation(s)
- Rodrigo Xavier das Neves
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
- LICI, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alex S Yamashita
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela M R Riccardi
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Julia Köhn-Gaone
- Department of Surgery, School of Veterinary Medicine and Animal Science of University of São Paulo-FMVZ/USP, São Paulo, Brazil
| | - Rodolfo G Camargo
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Nelson I Neto
- Department of Physiology, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Caetano
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Silvio P Gomes
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
- Department of Surgery, School of Veterinary Medicine and Animal Science of University of São Paulo-FMVZ/USP, São Paulo, Brazil
| | - Felipe H Santos
- Laboratory of Adipose Tissue Biology, Center for Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - Joanna D C C Lima
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Miguel L Batista
- Laboratory of Adipose Tissue Biology, Center for Integrated Biotechnology, University of Mogi das Cruzes, São Paulo, Brazil
| | - José Cesar Rosa-Neto
- Immunometabolism Research Group, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paulo Sérgio Martins De Alcântara
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Linda F Maximiano
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - José P Otoch
- LICI, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Giorgio Trinchieri
- LICI, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Janina E E Tirnitz-Parker
- Liver Disease and Regeneration Laboratory, School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Marília Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM26-HCFMUSP Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Understanding the relationship between cancer associated cachexia and hypoxia-inducible factor-1. Biomed Pharmacother 2023; 163:114802. [PMID: 37146421 DOI: 10.1016/j.biopha.2023.114802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a multifactorial disorder characterized by an unrestricted loss of body weight as a result of muscle and adipose tissue atrophy. Cachexia is influenced by several factors, including decreased metabolic activity and food intake, an imbalance between energy uptake and expenditure, excessive catabolism, and inflammation. Cachexia is highly associated with all types of cancers responsible for more than half of cancer-related mortalities worldwide. In healthy individuals, adipose tissue significantly regulates energy balance and glucose homeostasis. However, in metastatic cancer patients, CAC occurs mainly because of an imbalance between muscle protein synthesis and degradation which are organized by certain extracellular ligands and associated signaling pathways. Under hypoxic conditions, hypoxia-inducible factor-1 (HIF-1α) accumulated and translocated to the nucleus and activate numerous genes involved in cell survival, invasion, angiogenesis, metastasis, metabolic reprogramming, and cancer stemness. On the other hand, the ubiquitination proteasome pathway is inhibited during low O2 levels which promote muscle wasting in cancer patients. Therefore, understanding the mechanism of the HIF-1 pathway and its metabolic adaptation to biomolecules is important for developing a novel therapeutic method for cancer and cachexia therapy. Even though many HIF inhibitors are already in a clinical trial, their mechanism of action remains unknown. With this background, this review summarizes the basic concepts of cachexia, the role of inflammatory cytokines, pathways connected with cachexia with special reference to the HIF-1 pathway and its regulation, metabolic changes, and inhibitors of HIFs.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
21
|
Bell S, Young JA, List EO, Basu R, Geitgey DK, Lach G, Lee K, Swegan D, Caggiano LJ, Okada S, Kopchick JJ, Berryman DE. Increased Fibrosis in White Adipose Tissue of Male and Female bGH Transgenic Mice Appears Independent of TGF-β Action. Endocrinology 2023; 164:7069260. [PMID: 36869769 DOI: 10.1210/endocr/bqad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023]
Abstract
Fibrosis is a pathological state caused by excess deposition of extracellular matrix proteins in a tissue. Male bovine growth hormone (bGH) transgenic mice experience metabolic dysfunction with a marked decrease in lifespan and with increased fibrosis in several tissues including white adipose tissue (WAT), which is more pronounced in the subcutaneous (Sc) depot. The current study expanded on these initial findings to evaluate WAT fibrosis in female bGH mice and the role of transforming growth factor (TGF)-β in the development of WAT fibrosis. Our findings established that female bGH mice, like males, experience a depot-dependent increase in WAT fibrosis, and bGH mice of both sexes have elevated circulating levels of several markers of collagen turnover. Using various methods, TGF-β signaling was found unchanged or decreased-as opposed to an expected increase-despite the marked fibrosis in WAT of bGH mice. However, acute GH treatments in vivo, in vitro, or ex vivo did elicit a modest increase in TGF-β signaling in some experimental systems. Finally, single nucleus RNA sequencing confirmed no perturbation in TGF-β or its receptor gene expression in any WAT cell subpopulations of Sc bGH WAT; however, a striking increase in B lymphocyte infiltration in bGH WAT was observed. Overall, these data suggest that bGH WAT fibrosis is independent of the action of TGF-β and reveals an intriguing shift in immune cells in bGH WAT that should be further explored considering the increasing importance of B cell-mediated WAT fibrosis and pathology.
Collapse
Affiliation(s)
- Stephen Bell
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Jonathan A Young
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Edward O List
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | | | - Grace Lach
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Kevin Lee
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Deborah Swegan
- College of Arts and Sciences, Ohio University, Athens, OH 45701, USA
| | | | - Shigeru Okada
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| | - Darlene E Berryman
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
22
|
Ginting RP, Lee JM, Lee MW. The Influence of Ambient Temperature on Adipose Tissue Homeostasis, Metabolic Diseases and Cancers. Cells 2023; 12:cells12060881. [PMID: 36980222 PMCID: PMC10047443 DOI: 10.3390/cells12060881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
Adipose tissue is a recognized energy storage organ during excessive energy intake and an endocrine and thermoregulator, which interacts with other tissues to regulate systemic metabolism. Adipose tissue dysfunction is observed in most obese mouse models and humans. However, most studies using mouse models were conducted at room temperature (RT), where mice were chronically exposed to mild cold. In this condition, energy use is prioritized for thermogenesis to maintain body temperature in mice. It also leads to the activation of the sympathetic nervous system, followed by the activation of β-adrenergic signaling. As humans live primarily in their thermoneutral (TN) zone, RT housing for mice limits the interpretation of disease studies from mouse models to humans. Therefore, housing mice in their TN zone (~28–30 °C) can be considered to mimic humans physiologically. However, factors such as temperature ranges and TN pre-acclimatization periods should be examined to obtain reliable results. In this review, we discuss how adipose tissue responds to housing temperature and the outcomes of the TN zone in metabolic disease studies. This review highlights the critical role of TN housing in mouse models for studying adipose tissue function and human metabolic diseases.
Collapse
Affiliation(s)
- Rehna Paula Ginting
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Ji-Min Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Min-Woo Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
- Correspondence: ; Tel.: +82-41-413-5029
| |
Collapse
|
23
|
Molfino A, Ambrosani F, Tambaro F, Belli R, Imbimbo G, Udali S, Moruzzi S, Pattini P, Ramaccini C, Castagna A, Muscaritoli M, Friso S. Changes of gene expression in peripheral blood mononuclear cells of lung cancer patients with or without anorexia. Clin Nutr 2023; 42:9-17. [PMID: 36473427 DOI: 10.1016/j.clnu.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/07/2022] [Accepted: 11/13/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND & AIMS Anorexia is a disabling symptom in cancer and we aimed at investigating the role of changes in gene expression in lung cancer patients presenting with anorexia. METHODS Genome-wide transcriptomic profiling was assessed in PBMCs RNA from newly diagnosed lung cancer patients and in a control group. RT-qPCR was used for selected genes. RESULTS RNA-Seq analysis revealed among groups a large number of differentially expressed genes mainly implicated in immune system regulation, oxidative stress and cytokine-mediated inflammation signaling pathways. In particular, we identified a total of 983 DEGs (843 up-regulated; 140 down-regulated) in anorexic cancer compared to controls. A selected number of DEGs including ADAM8, SMAD4, CCR4 and CLU were differentially expressed within cancer group according to the presence/absence of anorexia. In terms of RT-qPCR, ADAM8 was less expressed in cancer patients than controls (p < 0.001), and in anorexic patients vs controls (p = 0.001). The expression of SMAD4 was lower in cancer vs controls (p = 0.005), and in anorexic patients vs controls (p = 0.009). We observed lower CCR4 expression in both anorexic and non-anorexic vs control (p = 0.004, p = 0.011, respectively) and a similar trend was present for CLU. CONCLUSIONS Our data shed new light on the role of specific genes and their associated molecular pathways as potential key mechanisms for the development of anorexia and may represent a novel landmark for understanding the complex pathophysiology of impaired appetite in cancer.
Collapse
Affiliation(s)
- Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| | - Francesca Ambrosani
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Federica Tambaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberta Belli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Udali
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Sara Moruzzi
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Patrizia Pattini
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Cesarina Ramaccini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalisa Castagna
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Simonetta Friso
- Department of Medicine, University of Verona School of Medicine, Verona, Italy
| |
Collapse
|
24
|
Obesity-Associated ECM Remodeling in Cancer Progression. Cancers (Basel) 2022; 14:cancers14225684. [PMID: 36428776 PMCID: PMC9688387 DOI: 10.3390/cancers14225684] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Adipose tissue, an energy storage and endocrine organ, is emerging as an essential player for ECM remodeling. Fibrosis is one of the hallmarks of obese adipose tissue, featuring excessive ECM deposition and enhanced collagen alignment. A variety of ECM components and ECM-related enzymes are produced by adipocytes and myofibroblasts in obese adipose tissue. Data from lineage-tracing models and a single-cell analysis indicate that adipocytes can transform or de-differentiate into myofibroblast/fibroblast-like cells. This de-differentiation process has been observed under normal tissue development and pathological conditions such as cutaneous fibrosis, wound healing, and cancer development. Accumulated evidence has demonstrated that adipocyte de-differentiation and myofibroblasts/fibroblasts play crucial roles in obesity-associated ECM remodeling and cancer progression. In this review, we summarize the recent progress in obesity-related ECM remodeling, the mechanism underlying adipocyte de-differentiation, and the function of obesity-associated ECM remodeling in cancer progression.
Collapse
|
25
|
Torregrosa C, Chorin F, Beltran EEM, Neuzillet C, Cardot-Ruffino V. Physical Activity as the Best Supportive Care in Cancer: The Clinician's and the Researcher's Perspectives. Cancers (Basel) 2022; 14:5402. [PMID: 36358820 PMCID: PMC9655932 DOI: 10.3390/cancers14215402] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 08/11/2023] Open
Abstract
Multidisciplinary supportive care, integrating the dimensions of exercise alongside oncological treatments, is now regarded as a new paradigm to improve patient survival and quality of life. Its impact is important on the factors that control tumor development, such as the immune system, inflammation, tissue perfusion, hypoxia, insulin resistance, metabolism, glucocorticoid levels, and cachexia. An increasing amount of research has been published in the last years on the effects of physical activity within the framework of oncology, marking the appearance of a new medical field, commonly known as "exercise oncology". This emerging research field is trying to determine the biological mechanisms by which, aerobic exercise affects the incidence of cancer, the progression and/or the appearance of metastases. We propose an overview of the current state of the art physical exercise interventions in the management of cancer patients, including a pragmatic perspective with tips for routine practice. We then develop the emerging mechanistic views about physical exercise and their potential clinical applications. Moving toward a more personalized, integrated, patient-centered, and multidisciplinary management, by trying to understand the different interactions between the cancer and the host, as well as the impact of the disease and the treatments on the different organs, this seems to be the most promising method to improve the care of cancer patients.
Collapse
Affiliation(s)
- Cécile Torregrosa
- Oncologie Digestive, Département d’Oncologie Médicale Institut Curie, Université Versailles Saint-Quentin—Université Paris Saclay, 35, rue Dailly, 92210 Saint-Cloud, France
- Département de Chirurgie Digestive et Oncologique, Hôpital Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, 9 avenue Charles de Gaulle, 92100 Boulogne Billancourt, France
| | - Frédéric Chorin
- Laboratoire Motricité Humaine, Expertise, Sport, Santé (LAMHESS), HEALTHY Graduate School, Université Côte d’Azur, 06205 Nice, France
- Clinique Gériatrique du Cerveau et du Mouvement, Centre Hospitalier Universitaire de Nice, Université Côte d’Azur, 06205 Nice, France
| | - Eva Ester Molina Beltran
- Oncologie Digestive, Département d’Oncologie Médicale Institut Curie, Université Versailles Saint-Quentin—Université Paris Saclay, 35, rue Dailly, 92210 Saint-Cloud, France
| | - Cindy Neuzillet
- Oncologie Digestive, Département d’Oncologie Médicale Institut Curie, Université Versailles Saint-Quentin—Université Paris Saclay, 35, rue Dailly, 92210 Saint-Cloud, France
- GERCOR, 151 rue du Faubourg Saint-Antoine, 75011 Paris, France
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
26
|
Hu X, Sun A, Chen H, Yan X, Ding F, Zheng P, Li Z, Yan YE. Saponins from Panax japonicus alleviate adipose tissue fibrosis and metabolic dysfunction in high-fat-diet-induced obese mice. Biomarkers 2022; 27:784-794. [DOI: 10.1080/1354750x.2022.2122566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Xiaoqin Hu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Ao Sun
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Huijian Chen
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Xiyue Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Fei Ding
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Peng Zheng
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| | - Zhen Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - You-e Yan
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China
| |
Collapse
|
27
|
Transforming Growth Factor-Beta Signaling in Cancer-Induced Cachexia: From Molecular Pathways to the Clinics. Cells 2022; 11:cells11172671. [PMID: 36078078 PMCID: PMC9454487 DOI: 10.3390/cells11172671] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a metabolic syndrome consisting of massive loss of muscle mass and function that has a severe impact on the quality of life and survival of cancer patients. Up to 20% of lung cancer patients and up to 80% of pancreatic cancer patients are diagnosed with cachexia, leading to death in 20% of them. The main drivers of cachexia are cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), macrophage inhibitory cytokine 1 (MIC-1/GDF15) and transforming growth factor-beta (TGF-β). Besides its double-edged role as a tumor suppressor and activator, TGF-β causes muscle loss through myostatin-based signaling, involved in the reduction in protein synthesis and enhanced protein degradation. Additionally, TGF-β induces inhibin and activin, causing weight loss and muscle depletion, while MIC-1/GDF15, a member of the TGF-β superfamily, leads to anorexia and so, indirectly, to muscle wasting, acting on the hypothalamus center. Against this background, the blockade of TGF-β is tested as a potential mechanism to revert cachexia, and antibodies against TGF-β reduced weight and muscle loss in murine models of pancreatic cancer. This article reviews the role of the TGF-β pathway and to a minor extent of other molecules including microRNA in cancer onset and progression with a special focus on their involvement in cachexia, to enlighten whether TGF-β and such other players could be potential targets for therapy.
Collapse
|
28
|
Xiong S, Tan J, Wang Y, He J, Hu F, Wu X, Liu Z, Lin S, Li X, Chen Z, Mao R. Fibrosis in fat: From other diseases to Crohn’s disease. Front Immunol 2022; 13:935275. [PMID: 36091035 PMCID: PMC9453038 DOI: 10.3389/fimmu.2022.935275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Creeping fat is a specific feature of Crohn’s disease (CD) and is characterized by mesenteric fat wrapping around the intestine. It highly correlates with intestinal transmural inflammation, muscular hypertrophy, fibrosis, and stricture formation. However, the pathogenesis of creeping fat remains unclear. Molecular crosstalk exists between mesenteric fat and the intestine. Indeed, creeping fat contains different types of cells, including adipocytes and immune cells. These cell types can produce various cytokines, fatty acids, and growth factors, which affect the mesenteric fat function and modulate intestinal inflammation and immunity. Moreover, adipocyte progenitors can produce extracellular matrix to adapt to fat expansion. Previous studies have shown that fat fibrosis is an important feature of adipose tissue malfunction and exists in other diseases, including metabolic disorders, cancer, atrial fibrillation, and osteoarthritis. Furthermore, histological sections of CD showed fibrosis in the creeping fat. However, the role of fibrosis in the mesenteric fat of CD is not well understood. In this review, we summarized the possible mechanisms of fat fibrosis and its impact on other diseases. More specifically, we illustrated the role of various cells (adipocyte progenitors, macrophages, mast cells, and group 1 innate lymphoid cells) and molecules (including hypoxia-inducible factor 1-alpha, transforming growth factor-beta, platelet-derived growth factor, and peroxisome proliferator-activated receptor-gamma) in the pathogenesis of fat fibrosis in other diseases to understand the role of creeping fat fibrosis in CD pathogenesis. Future research will provide key information to decipher the role of fat fibrosis in creeping fat formation and intestinal damage, thereby helping us identify novel targets for the diagnosis and treatment of CD.
Collapse
Affiliation(s)
- Shanshan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinyu Tan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jinshen He
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Fan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaomin Wu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zishan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sinan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuehua Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhihui Chen
- Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Ren Mao, ; Zhihui Chen,
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Gastroenterology, Huidong People’s Hospital, Huizhou, China
- *Correspondence: Ren Mao, ; Zhihui Chen,
| |
Collapse
|
29
|
Malla J, Zahra A, Venugopal S, Selvamani TY, Shoukrie SI, Selvaraj R, Dhanoa RK, Hamouda RK, Mostafa J. What Role Do Inflammatory Cytokines Play in Cancer Cachexia? Cureus 2022; 14:e26798. [PMID: 35971351 PMCID: PMC9372379 DOI: 10.7759/cureus.26798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/12/2022] [Indexed: 11/05/2022] Open
|
30
|
de Sousa Neto IV, Durigan JLQ, da Silva ASR, de Cássia Marqueti R. Adipose Tissue Extracellular Matrix Remodeling in Response to Dietary Patterns and Exercise: Molecular Landscape, Mechanistic Insights, and Therapeutic Approaches. BIOLOGY 2022; 11:biology11050765. [PMID: 35625493 PMCID: PMC9138682 DOI: 10.3390/biology11050765] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022]
Abstract
Simple Summary Adipose tissue is considered a metabolic organ that adjusts overall energy homeostasis and critical hormones to the body’s needs. In conditions of caloric intake surpassing energy expenditure, lipid accumulation occurs with constant extracellular matrix deposition. Excess lipids and adipocyte hypertrophy may reduce extracellular matrix flexibility in conjunction with hypoxia and inflammation. These processes induce the development of adipose tissue fibrosis and correlated metabolic dysfunctions, such as insulin resistance. With the increasing rate of chronic diseases worldwide, it is essential to generate a more precise knowledge of fibrotic processes, as well as to create optimal models to study potential therapies to combat the harmful effects of extracellular matrix deposition. In this work, we focused on the physiological processes in the remodeling of adipose tissue fibrosis, along with their relevance to clinical indications. Furthermore, we emphasize understanding how lifestyle can alleviate adipocyte dysfunction. Several studies showed that a nutritionally balanced diet combined with exercise is a remarkable potential strategy for lipolytic activity, preventing rapid extracellular matrix expansion in parallel with insulin and glucose action improvements. Thus, the emerging beneficial role of exercise training and low-calorie diet on adipose tissue ECM remodeling is a topic that deserves attention from health professionals. Abstract The extracellular matrix (ECM) is a 3-dimensional network of molecules that play a central role in differentiation, migration, and survival for maintaining normal homeostasis. It seems that ECM remodeling is required for adipose tissue expansion. Despite evidence indicating that ECM is an essential component of tissue physiology, adipose tissue ECM has received limited attention. Hence, there is great interest in approaches to neutralize the harmful effects of ECM enlargement. This review compiles and discusses the current literature on adipose tissue ECM remodeling in response to different dietary patterns and exercise training. High-calorie diets result in substantial adipose tissue ECM remodeling, which in turn could lead to fibrosis (excess deposition of collagens, elastin, and fibronectin), inflammation, and the onset of metabolic dysfunction. However, combining a nutritionally balanced diet with exercise is a remarkable potential strategy for lipolytic activity, preventing rapid ECM expansion in different adipose tissue depots. Despite the distinct exercise modalities (aerobic or resistance exercise) reversing adipose tissue fibrosis in animal models, the beneficial effect on humans remains controversial. Defining molecular pathways and specific mechanisms that mediate the positive effects on adipose tissue, ECM is essential in developing optimized interventions to improve health and clinical outcomes.
Collapse
Affiliation(s)
- Ivo Vieira de Sousa Neto
- Molecular Analysis Laboratory, Faculty of Ceilândia, Universidade de Brasília, Brasília 70910-900, Brazil; or
- Correspondence:
| | | | - Adelino Sanchez Ramos da Silva
- Graduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto 14040-900, Brazil;
- School of Physical Education and Sport of Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Rita de Cássia Marqueti
- Molecular Analysis Laboratory, Faculty of Ceilândia, Universidade de Brasília, Brasília 70910-900, Brazil; or
- Graduate Program in Rehabilitation Sciences, Universidade de Brasília, Brasília 70910-900, Brazil;
- Graduate Program in Health Sciences and Technology, Universidade de Brasília, Brasília 70910-900, Brazil
| |
Collapse
|
31
|
Xue W, Yu SY, Kuss MA, Kong Y, Shi W, Chung S, Kim SY, Duan B. 3D bioprinted white adipose model for in vitro study of cancer-associated cachexia induced adipose tissue remodeling. Biofabrication 2022; 14. [PMID: 35504266 DOI: 10.1088/1758-5090/ac6c4b] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/03/2022] [Indexed: 11/11/2022]
Abstract
Cancer-associated cachexia (CAC) is a complex metabolic and behavioral syndrome with multiple manifestations that involve systemic inflammation, weight loss, and adipose lipolysis. It impacts the quality of life of patients and is the direct cause of death in 20-30% of cancer patients. The severity of fat loss and adipose tissue remodeling negatively correlate with patients' survival outcomes. To address the mechanism of fat loss and design potential approaches to prevent the process, it will be essential to understand CAC pathophysiology through white adipose tissue models. In the present study, an engineered human white adipose tissue (eWAT) model based on three-dimensional (3D) bioprinting was developed and treated with pancreatic cancer cell-conditioned medium (CM) to mimic the status of CAC in vitro. We found that the CM treatment significantly increased the lipolysis and accumulation of the extracellular matrix (ECM). The 3D eWATs were further vascularized to study the influence of vascularization on lipolysis and CAC progression, which was largely unknown. Results demonstrated that CM treatment improved the angiogenesis of vascularized eWATs (veWATs), and veWATs demonstrated decreased glycerol release but increased Ucp1 expression, compared to eWATs. Many unique inflammatory cytokines (IL-8, CXCL-1, GM-CSF, etc) from the CM were detected and supposed to contribute to eWAT lipolysis, Ucp1 up-regulation, and ECM development. In response to CM treatment, eWATs also secreted inflammatory adipokines related to the metastatic ability of cancer, muscle atrophy, and vascularization (NGAL, CD54, IGFBP-2, etc). Our work demonstrated that the eWAT is a robust model for studying cachectic fat loss and the accompanying remodeling of adipose tissue. It is therefore a useful tool for future research exploring CAC physiologies and developing potential therapies.
Collapse
Affiliation(s)
- Wen Xue
- University of Nebraska Medical Center, DRCII, Omaha, 68198-7400, UNITED STATES
| | - Seok-Yeong Yu
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| | - Mitchell A Kuss
- Regenerative Medicine, University of Nebraska Medical Center, DRCII, Omaha, Nebraska, 68106, UNITED STATES
| | - Yunfan Kong
- University of Nebraska Medical Center, DRCII, Omaha, 68198-7400, UNITED STATES
| | - Wen Shi
- University of Nebraska Medical Center, DRCII, Omaha, Nebraska, 68106, UNITED STATES
| | - Soonkyu Chung
- University of Massachusetts Amherst, UMA, Amherst, Massachusetts, 01003, UNITED STATES
| | - So-Youn Kim
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| | - Bin Duan
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| |
Collapse
|
32
|
Kita A, Saito Y, Miura N, Miyajima M, Yamamoto S, Sato T, Yotsuyanagi T, Fujimiya M, Chikenji TS. Altered regulation of mesenchymal cell senescence in adipose tissue promotes pathological changes associated with diabetic wound healing. Commun Biol 2022; 5:310. [PMID: 35383267 PMCID: PMC8983691 DOI: 10.1038/s42003-022-03266-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 03/14/2022] [Indexed: 01/13/2023] Open
Abstract
Pathologic diabetic wound healing is caused by sequential and progressive deterioration of hemostasis, inflammation, proliferation, and resolution/remodeling. Cellular senescence promotes wound healing; however, diabetic wounds exhibit low levels of senescent factors and accumulate senescent cells, which impair the healing process. Here we show that the number of p15INK4B + PDGFRα + senescent mesenchymal cells in adipose tissue increases transiently during early phases of wound healing in both non-diabetic mice and humans. Transplantation of adipose tissue from diabetic mice into non-diabetic mice results in impaired wound healing and an altered cellular senescence–associated secretory phenotype (SASP), suggesting that insufficient induction of adipose tissue senescence after injury is a pathological mechanism of diabetic wound healing. These results provide insight into how regulation of senescence in adipose tissue contributes to wound healing and could constitute a basis for developing therapeutic treatment for wound healing impairment in diabetes. Type-2 diabetic adipose tissue impairs transient senescence during wound healing with expression of different components of the senescence-associated secretory phenotype (SASP), and this is associated with deteriorated wound healing.
Collapse
Affiliation(s)
- Arisa Kita
- Department of Plastic and Reconstructive Surgery, Sapporo Medical University, Sapporo, Japan
| | - Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Norihiro Miura
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Maki Miyajima
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Sena Yamamoto
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Tsukasa Sato
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Takatoshi Yotsuyanagi
- Department of Plastic and Reconstructive Surgery, Sapporo Medical University, Sapporo, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan. .,Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
33
|
Molfino A, Carletti R, Imbimbo G, Amabile MI, Belli R, di Gioia CRT, Belloni E, Spinelli F, Rizzo V, Catalano C, Nigri G, Muscaritoli M. Histomorphological and inflammatory changes of white adipose tissue in gastrointestinal cancer patients with and without cachexia. J Cachexia Sarcopenia Muscle 2022; 13:333-342. [PMID: 34939367 PMCID: PMC8818610 DOI: 10.1002/jcsm.12893] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND During cancer cachexia, several alterations occur in peripheral tissues, and the adipose tissue may be involved during the catabolic state. We aimed at investigating histological rearrangement and infiltration of inflammatory cells in subcutaneous adipose tissue (SAT) of patients with cancer undergoing surgery, according to the presence/absence of cachexia. METHODS We considered gastrointestinal cancer patients and controls with non-malignant diseases undergoing surgery. We collected SAT samples and performed histomorphological analyses [cross-sectional area (CSA) and per cent of fibrosis] and immunohistochemistry to characterize the inflammatory cells. By computed tomography (CT) scan, we calculated SAT and visceral adipose tissue (VAT). RESULTS We enrolled 51 participants (31 gastrointestinal cancer patients and 20 controls). In cancer patients, cachexia was present in 13/31 (42%). The CSA (μm2 ) of the adipocytes from SAT was reduced in cancer patients vs. controls (3148, inter-quartile range 2574-3755 vs. 4474, inter-quartile range 3654-5183) (P < 0.001), in particular in cachectic patients vs. non-cachectic (median 2518 vs. median 3470) (P = 0.03) and in cachectic vs. controls (P < 0.001), as well as in non-cachectic vs. controls (P = 0.04). The median per cent of fibrosis was higher in cancer patients vs. controls (9 vs. 3) (P = 0.0001), in particular in cachectic vs. non-cachectic (13.35 vs. 7.13) (P = 0.03). We observed a higher number of macrophages (CD68) (P = 0.0001) and T lymphocytes (CD3) (P = 0.002) in SAT of cancer patients vs. controls, and the number of T lymphocytes was higher in cachectic vs. non-cachectic patients (P = 0.025). Anorexic cancer patients showed in SAT a higher number of macrophages and T lymphocytes with respect to controls (P < 0.0001), whereas no difference was present between anorexic and non-anorexic patients. At CT scan, cachectic patients showed lower VAT and SAT vs. non-cachectic (VAT: 97.64 ± 40.79 vs. 212.53 ± 79.24, P = 0.0002; SAT: 126.27 ± 87.92 vs. 206.27 ± 61.93, P = 0.01, respectively). Cancer patients with low CSA, high degree of fibrosis, and high number of T lymphocytes presented with lower body mass index and lower SAT and VAT at CT scan (P ≤ 0.01). CONCLUSIONS We found histological alterations of SAT among gastrointestinal cancer patients and in particular significant changes in CSA, fibrosis, and inflammation when cachexia was present; the changes in histomorphological parameters of the adipocytes reflected alterations in adiposity at body composition analysis.
Collapse
Affiliation(s)
- Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Ida Amabile
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberta Belli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Cira R T di Gioia
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Elena Belloni
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Spinelli
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Veronica Rizzo
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Carlo Catalano
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Nigri
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Xu PC, You M, Yu SY, Luan Y, Eldani M, Caffrey TC, Grandgenett PM, O'Connell KA, Shukla SK, Kattamuri C, Hollingsworth MA, Singh PK, Thompson TB, Chung S, Kim SY. Visceral adipose tissue remodeling in pancreatic ductal adenocarcinoma cachexia: the role of activin A signaling. Sci Rep 2022; 12:1659. [PMID: 35102236 PMCID: PMC8803848 DOI: 10.1038/s41598-022-05660-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) patients display distinct phenotypes of cachexia development, with either adipose tissue loss preceding skeletal muscle wasting or loss of only adipose tissue. Activin A levels were measured in serum and analyzed in tumor specimens of both a cohort of Stage IV PDAC patients and the genetically engineered KPC mouse model. Our data revealed that serum activin A levels were significantly elevated in Stage IV PDAC patients in comparison to age-matched non-cancer patients. Little is known about the role of activin A in adipose tissue wasting in the setting of PDAC cancer cachexia. We established a correlation between elevated activin A and remodeling of visceral adipose tissue. Atrophy and fibrosis of visceral adipose tissue was examined in omental adipose tissue of Stage IV PDAC patients and gonadal adipose tissue of an orthotopic mouse model of PDAC. Remarkably, white visceral adipose tissue from both PDAC patients and mice exhibited decreased adipocyte diameter and increased fibrotic deposition. Strikingly, expression of thermogenic marker UCP1 in visceral adipose tissues of PDAC patients and mice remained unchanged. Thus, we propose that activin A signaling could be relevant to the acceleration of visceral adipose tissue wasting in PDAC-associated cachexia.
Collapse
Affiliation(s)
- Pauline C Xu
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mikyoung You
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts Amherst, 211 Chenoweth Laboratory, 100 Holdsworth Way, Amherst, MA, 01003-9282, USA
| | - Seok-Yeong Yu
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yi Luan
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maya Eldani
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Thomas C Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kelly A O'Connell
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surendra K Shukla
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chandramohan Kattamuri
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 68198, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH, 68198, USA
| | - Soonkyu Chung
- Department of Nutrition, School of Public Health and Health Sciences, University of Massachusetts Amherst, 211 Chenoweth Laboratory, 100 Holdsworth Way, Amherst, MA, 01003-9282, USA.
| | - So-Youn Kim
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, College of Medicine, University of Nebraska Medical Center, 985860 Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
35
|
Abstract
Diverse inflammatory diseases, infections and malignancies are associated with wasting syndromes. In many of these conditions, the standards for diagnosis and treatment are lacking due to our limited understanding of the causative molecular mechanisms. Here, we discuss the complex immunological context of cachexia, a systemic catabolic syndrome that depletes both fat and muscle mass with profound consequences for patient prognosis. We highlight the main cytokine and immune cell-driven pathways that have been linked to weight loss and tissue wasting in the context of cancer-associated and infection-associated cachexia. Moreover, we discuss the potential immunometabolic consequences of cachexia on the basis of newly identified pathways and explore the multilayered area of immunometabolic crosstalk both upstream and downstream of tissue catabolism. Collectively, this Review highlights the intricate relationship of the immune system with cachexia in the context of malignant and infectious diseases.
Collapse
|
36
|
Blaszkiewicz M, Gunsch G, Willows JW, Gardner ML, Sepeda JA, Sas AR, Townsend KL. Adipose Tissue Myeloid-Lineage Neuroimmune Cells Express Genes Important for Neural Plasticity and Regulate Adipose Innervation. Front Endocrinol (Lausanne) 2022; 13:864925. [PMID: 35795142 PMCID: PMC9251313 DOI: 10.3389/fendo.2022.864925] [Citation(s) in RCA: 8] [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: 01/29/2022] [Accepted: 05/05/2022] [Indexed: 12/19/2022] Open
Abstract
Peripheral nerves allow a bidirectional communication between brain and adipose tissues, and many studies have clearly demonstrated that a loss of the adipose nerve supply results in tissue dysfunction and metabolic dysregulation. Neuroimmune cells closely associate with nerves in many tissues, including subcutaneous white adipose tissue (scWAT). However, in scWAT, their functions beyond degrading norepinephrine in an obese state remain largely unexplored. We previously reported that a myeloid-lineage knockout (KO) of brain-derived neurotrophic factor (BDNF) resulted in decreased innervation of scWAT, accompanied by an inability to brown scWAT after cold stimulation, and increased adiposity after a high-fat diet. These data underscored that adipose tissue neuroimmune cells support the peripheral nerve supply to adipose and impact the tissue's metabolic functions. We also reported that a subset of myeloid-lineage monocyte/macrophages (Ly6c+CCR2+Cx3cr1+) is recruited to scWAT in response to cold, a process known to increase neurite density in adipose and promote metabolically healthy processes. These cold-induced neuroimmune cells (CINCs) also expressed BDNF. Here we performed RNAseq on CINCs from cold-exposed and room temperature-housed mice, which revealed a striking and coordinated differential expression of numerous genes involved in neuronal function, including neurotrophin signaling and axonal guidance, further supporting that CINCs fulfill a nerve-supporting role in adipose. The increased expression of leukocyte transendothelial migration genes in cold-stimulated CINCs also confirms prior evidence that they are recruited to scWAT and are not tissue resident. We now provide whole-depot imaging of scWAT from LysM-BDNF KO mice, revealing a striking reduction of innervation across the depot fitting with their reduced energy expenditure phenotype. By contrast, Cx3cr1-BDNF KO mice (a macrophage subset of LysM+ cells) exhibited increased thermogenesis and energy expenditure, with compensatory increased food intake and no change in adiposity or body weight. While these KO mice also exhibit a significantly reduced innervation of scWAT, especially around the subiliac lymph node, they displayed an increase in small fiber sympathetic neurite branching, which may underlie their increased thermogenesis. We propose a homeostatic role of scWAT myeloid-lineage neuroimmune cells together in nerve maintenance and neuro-adipose regulation of energy expenditure.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Gilian Gunsch
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Jake W. Willows
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Miranda L. Gardner
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
- Campus Chemical Instrument Center, Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, United States
| | - Jesse A. Sepeda
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Andrew R. Sas
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Kristy L. Townsend
- Neurobiology and Energy Balance Laboratory, Department of Neurological Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, United States
- *Correspondence: Kristy L. Townsend,
| |
Collapse
|
37
|
de Jesus JDCR, Murari ASDP, Radloff K, de Moraes RCM, Figuerêdo RG, Pessoa AFM, Rosa-Neto JC, Matos-Neto EM, Alcântara PSM, Tokeshi F, Maximiano LF, Bin FC, Formiga FB, Otoch JP, Seelaender M. Activation of the Adipose Tissue NLRP3 Inflammasome Pathway in Cancer Cachexia. Front Immunol 2021; 12:729182. [PMID: 34630405 PMCID: PMC8495409 DOI: 10.3389/fimmu.2021.729182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/03/2021] [Indexed: 12/11/2022] Open
Abstract
Background Cachexia is a paraneoplastic syndrome that accompanies and compromises cancer treatment, especially in advanced stages, affecting the metabolism and function of several organs. The adipose tissue is the first to respond to the presence of the tumor, contributing to the secretion of factors which drive the systemic inflammation, a hallmark of the syndrome. While inflammation is a defensive innate response, the control mechanisms have been reported to be disrupted in cachexia. On the other hand, little is known about the role of NLRP3 inflammasome in this scenario, a multiprotein complex involved in caspase-1 activation and the processing of the cytokines IL-1β and IL-18. Aim based on the evidence from our previous study with a rodent model of cachexia, we examined the activation of the NLRP3 inflammasome pathway in two adipose tissue depots obtained from patients with colorectal cancer and compared with that another inflammatory pathway, NF-κB. Results For CC we found opposite modulation in ScAT and PtAT for the gene expression of TLR4, Caspase-1 (cachectic group) and for NF-κB p50, NF-κB p65, IL-1β. CD36, expression was decreased in both depots while that of NLRP3 and IL-18 was higher in both tissues, as compared with controls and weight stable patients (WSC). Caspase-1 basal protein levels in the ScAT culture supernatant were higher in WSC and (weight stable patients) CC, when compared to controls. Basal ScAT explant culture medium IL-1β and IL-18 protein content in ScAT supernatant was decreased in the WSC and CC as compared to CTL explants. Conclusions The results demonstrate heterogeneous responses in the activation of genes of the NLRP3 inflammasome pathway in the adipose tissue of patients with cancer cachexia, rendering this pathway a potential target for therapy aiming at decreasing chronic inflammation in cancer.
Collapse
Affiliation(s)
- Joyce de Cassia Rosa de Jesus
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ariene Soares de Pinho Murari
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Katrin Radloff
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ruan Carlos Macêdo de Moraes
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Raquel Galvão Figuerêdo
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Flavia Marçal Pessoa
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - José César Rosa-Neto
- Immunometabolism Laboratory, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Emídio Marques Matos-Neto
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo S M Alcântara
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Flavio Tokeshi
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Linda Ferreira Maximiano
- University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Fang Chia Bin
- Department of Coloproctology, Santa Casa de São Paulo, São Paulo, Brazil
| | | | - José P Otoch
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,University Hospital, Department of Surgical Clinic, Universidade de São Paulo, São Paulo, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery Laboratório de Investigação Médica (LIM26), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Tumor-derived MMPs regulate cachexia in a Drosophila cancer model. Dev Cell 2021; 56:2664-2680.e6. [PMID: 34473940 DOI: 10.1016/j.devcel.2021.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 07/09/2021] [Accepted: 08/12/2021] [Indexed: 12/14/2022]
Abstract
Cachexia, the wasting syndrome commonly observed in advanced cancer patients, accounts for up to one-third of cancer-related mortalities. We have established a Drosophila larval model of organ wasting whereby epithelial overgrowth in eye-antennal discs leads to wasting of the adipose tissue and muscles. The wasting is associated with fat-body remodeling and muscle detachment and is dependent on tumor-secreted matrix metalloproteinase 1 (Mmp1). Mmp1 can both modulate TGFβ signaling in the fat body and disrupt basement membrane (BM)/extracellular matrix (ECM) protein localization in both the fat body and the muscle. Inhibition of TGFβ signaling or Mmps in the fat body/muscle using a QF2-QUAS binary expression system rescues muscle wasting in the presence of tumor. Altogether, our study proposes that tumor-derived Mmps are central mediators of organ wasting in cancer cachexia.
Collapse
|
39
|
Longenecker JZ, Petrosino JM, Martens CR, Hinger SA, Royer CJ, Dorn LE, Branch DA, Serrano J, Stanford KI, Kyriazis GA, Baskin KK, Accornero F. Cardiac-derived TGF-β1 confers resistance to diet-induced obesity through the regulation of adipocyte size and function. Mol Metab 2021; 54:101343. [PMID: 34583010 PMCID: PMC8529557 DOI: 10.1016/j.molmet.2021.101343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022] Open
Abstract
Regulation of organismal homeostasis in response to nutrient availability is a vital physiological process that involves inter-organ communication. Understanding the mechanisms controlling systemic cross-talk for the maintenance of metabolic health is critical to counteract diet-induced obesity. Here, we show that cardiac-derived transforming growth factor beta 1 (TGF-β1) protects against weight gain and glucose intolerance in mice subjected to high-fat diet. Secretion of TGF-β1 by cardiomyocytes correlates with the bioavailability of this factor in circulation. TGF-β1 prevents adipose tissue inflammation independent of body mass and glucose metabolism phenotypes, indicating protection from adipocyte dysfunction-driven immune cell recruitment. TGF-β1 alters the gene expression programs in white adipocytes, favoring their fatty acid oxidation and consequently increasing their mitochondrial oxygen consumption rates. Ultimately, subcutaneous and visceral white adipose tissue from cadiac-specific TGF-β1 transgenic mice fail to undergo cellular hypertrophy, leading to reduced overall adiposity during high-fat feeding. Thus, TGF-β1 is a critical mediator of heart-fat communication for the regulation of systemic metabolism. TGFb1 is secreted from the heart to alter systemic metabolism. TGFb1 protects from diet-induced obesity. TGFb1 increases mitochondrial basal respiration in white adipocytes. Cardiac-derived TGFb1 prevents adipose tissue inflammation.
Collapse
Affiliation(s)
- Jacob Z Longenecker
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Jennifer M Petrosino
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Colton R Martens
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Scott A Hinger
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Charlotte J Royer
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Lisa E Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Daniel A Branch
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Joan Serrano
- Department of Biological Chemistry and Pharmacology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - George A Kyriazis
- Department of Biological Chemistry and Pharmacology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Kedryn K Baskin
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
40
|
Lima RS, Mattos RT, Medeiros NI, Kattah FM, Nascimento JRS, Menezes CA, Rios-Santos F, Dutra WO, Gomes JAS, Moreira PR. CXCL8 expression and methylation are correlated with anthropometric and metabolic parameters in childhood obesity. Cytokine 2021; 143:155538. [PMID: 33926776 DOI: 10.1016/j.cyto.2021.155538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/02/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Childhood obesity is a global and increasing health issue. Inflammation and dysregulated adipose tissue secretion are common findings in obesity and have been related to poor metabolic function. Given that DNA methylation impacts gene expression and is responsive to environmental changes, we aimed, in addition to characterize the patients in anthropometric and biochemical terms, to determine the expression of cytokines and adipokines, assess the methylation on regulatory regions of the genes that code for these molecules, and investigate the association of the expression and gene methylation with anthropometric and biochemical parameters in childhood obesity. Obese children present dyslipidemia, dysregulated serum levels of adipokines and their ratios, altered leukocytic expression of cytokines, and higher methylation at the CXCL8 promoter as compared to the control group. However, no significant results were observed in the fasting plasma glucose levels or the methylation of TGFB1, LEP, and the enhancer region of ADIPOQ. We also found negative correlations of CXCL8 expression with anthropometric and biochemical parameters, and positive correlation of CXCL8 promoter methylation and the serum levels of hepatic enzymes. Our results indicate that changes in metabolic parameters observed in childhood obesity are associated with the expression of adipokines and cytokines, and the methylation status at the CXCL8 promoter. CXCL8 may be a key factor for these alterations, as it correlates with many of the parameters assessed in the present study.
Collapse
Affiliation(s)
- Rafael S Lima
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Rafael T Mattos
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Nayara I Medeiros
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Fabiana M Kattah
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Julya R S Nascimento
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Carlos A Menezes
- Department of Genetics, State University of Santa Cruz, Bahia, Brazil; Service of Preventive Medicine - Unimed, Sergipe, Brazil
| | - Fabricio Rios-Santos
- Department of Basic and Health Sciences, Federal University of Mato Grosso, Mato Grosso, Brazil
| | - Walderez O Dutra
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil; National Institute of Science and Technology in Tropical Diseases - INCT-DT, Brazil
| | - Juliana A S Gomes
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Paula R Moreira
- Department of Morphology, Institute of Biological Sciences (ICB), Federal University of Minas Gerais, Minas Gerais, Brazil.
| |
Collapse
|
41
|
Kasprzak A. The Role of Tumor Microenvironment Cells in Colorectal Cancer (CRC) Cachexia. Int J Mol Sci 2021; 22:ijms22041565. [PMID: 33557173 PMCID: PMC7913937 DOI: 10.3390/ijms22041565] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cachexia (CC) is a multifactorial syndrome in patients with advanced cancer characterized by weight loss via skeletal-muscle and adipose-tissue atrophy, catabolic activity, and systemic inflammation. CC is correlated with functional impairment, reduced therapeutic responsiveness, and poor prognosis, and is a major cause of death in cancer patients. In colorectal cancer (CRC), cachexia affects around 50–61% of patients, but remains overlooked, understudied, and uncured. The mechanisms driving CC are not fully understood but are related, at least in part, to the local and systemic immune response to the tumor. Accumulating evidence demonstrates a significant role of tumor microenvironment (TME) cells (e.g., macrophages, neutrophils, and fibroblasts) in both cancer progression and tumor-induced cachexia, through the production of multiple procachectic factors. The most important role in CRC-associated cachexia is played by pro-inflammatory cytokines, including the tumor necrosis factor α (TNFα), originally known as cachectin, Interleukin (IL)-1, IL-6, and certain chemokines (e.g., IL-8). Heterogeneous CRC cells themselves also produce numerous cytokines (including chemokines), as well as novel factors called “cachexokines”. The tumor microenvironment (TME) contributes to systemic inflammation and increased oxidative stress and fibrosis. This review summarizes the current knowledge on the role of TME cellular components in CRC-associated cachexia, as well as discusses the potential role of selected mediators secreted by colorectal cancer cells in cooperation with tumor-associated immune and non-immune cells of tumor microenvironment in inducing or potentiating cancer cachexia. This knowledge serves to aid the understanding of the mechanisms of this process, as well as prevent its consequences.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Święcicki Street 6, 60-781 Poznań, Poland
| |
Collapse
|
42
|
Takahashi Y, Suzuki S, Hamada K, Nakada T, Oya Y, Sakakura N, Matsushita H, Kuroda H. Sarcopenia is poor risk for unfavorable short- and long-term outcomes in stage I non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:325. [PMID: 33708952 PMCID: PMC7944314 DOI: 10.21037/atm-20-4380] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Sarcopenia characterized by skeletal muscle loss may influence postoperative outcomes through physical decline and weakened immunity. We aimed to investigate clinical significance of sarcopenia in resected early-stage non-small cell lung cancer (NSCLC). Methods We retrospectively reviewed 315 consecutive patients with pathologic stage I NSCLC who had undergone lobectomy with systematic nodal dissection. Sarcopenia was defined as the lowest quartile of psoas muscle area on the 3rd vertebra on the high-resolution computed tomography (HRCT) image. Clinicopathological variables were used to investigate the correlation to postoperative complications as well as overall and recurrence-free survival. Results Upon multivariable analysis, male sex [odds ratio (OR) =5.780, 95% confidence interval (CI): 2.681–12.500, P<0.001], and sarcopenia (OR =21.00, 95% CI: 10.30–42.80, P<0.001) were independently associated with postoperative complications. The sarcopenia group showed significantly lower 5-over all survival (84.4% vs. 69.1%, P<0.001) and recurrence-free survival (77.2% vs. 62.0%, P<0.001) comparing with the non-sarcopenia group. In a multivariable analysis, sarcopenia was an independent prognostic factor [hazard ratio (HR) =1.978, 95% CI: 1.177–3.326, P=0.010] together with age ≥70 years (HR =1.956, 95% CI: 1.141–3.351, P=0.015) and non-adenocarcinoma histology (HR =1.958, 95% CI: 1.159–3.301, P=0.016). Conclusions This is the first study which demonstrates that preoperative sarcopenia is significantly associated with unfavorable postoperative complications as well as long-term survival in pathologic stage I NSCLC. This readily available factor on HRCT may provide valuable information to consider possible choice of surgical procedure and perioperative management.
Collapse
Affiliation(s)
- Yusuke Takahashi
- Department of General Thoracic Surgery, Sagamihara Kyodo Hospital, Sagamihara, Kanagawa, Japan.,Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan.,Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Shigeki Suzuki
- Department of General Thoracic Surgery, Sagamihara Kyodo Hospital, Sagamihara, Kanagawa, Japan
| | - Kenichi Hamada
- Department of General Thoracic Surgery, Sagamihara Kyodo Hospital, Sagamihara, Kanagawa, Japan
| | - Takeo Nakada
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Yuko Oya
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan.,Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Noriaki Sakakura
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| |
Collapse
|
43
|
de Castro GS, Correia-Lima J, Simoes E, Orsso CE, Xiao J, Gama LR, Gomes SP, Gonçalves DC, Costa RGF, Radloff K, Lenz U, Taranko AE, Bin FC, Formiga FB, de Godoy LGL, de Souza RP, Nucci LHA, Feitoza M, de Castro CC, Tokeshi F, Alcantara PSM, Otoch JP, Ramos AF, Laviano A, Coletti D, Mazurak VC, Prado CM, Seelaender M. Myokines in treatment-naïve patients with cancer-associated cachexia. Clin Nutr 2020; 40:2443-2455. [PMID: 33190987 DOI: 10.1016/j.clnu.2020.10.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/10/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
Cancer-associated cachexia is a complex metabolic syndrome characterized by weight loss and systemic inflammation. Muscle loss and fatty infiltration into muscle are associated with poor prognosis in cancer patients. Skeletal muscle secretes myokines, factors with autocrine, paracrine and/or endocrine action, which may be modified by or play a role in cachexia. This study examined myokine content in the plasma, skeletal muscle and tumor homogenates from treatment-naïve patients with gastric or colorectal stages I-IV cancer with cachexia (CC, N = 62), or not (weight stable cancer, WSC, N = 32). Myostatin, interleukin (IL) 15, follistatin-like protein 1 (FSTL-1), fatty acid binding protein 3 (FABP3), irisin and brain-derived neurotrophic factor (BDNF) protein content in samples was measured with Multiplex technology; body composition and muscle lipid infiltration were evaluated in computed tomography, and quantification of triacylglycerol (TAG) in the skeletal muscle. Cachectic patients presented lower muscle FSTL-1 expression (p = 0.047), higher FABP3 plasma content (p = 0.0301) and higher tumor tissue expression of FABP3 (p = 0.0182), IL-15 (p = 0.007) and irisin (p = 0.0110), compared to WSC. Neither muscle TAG content, nor muscle attenuation were different between weight stable and cachectic patients. Lumbar adipose tissue (AT) index, visceral AT index and subcutaneous AT index were lower in CC (p = 0.0149, p = 0.0455 and p = 0.0087, respectively), who also presented lower muscularity in the cohort (69.2% of patients; p = 0.0301), compared to WSC. The results indicate the myokine profile in skeletal muscle, plasma and tumor is impacted by cachexia. These findings show that myokines eventually affecting muscle wasting may not solely derive from the muscle itself (as the tumor also may contribute to the systemic scenario), and put forward new perspectives on cachexia treatment targeting myokines and associated receptors and pathways.
Collapse
Affiliation(s)
- Gabriela S de Castro
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil.
| | - Joanna Correia-Lima
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Estefania Simoes
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Camila E Orsso
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Canada
| | - Jingjie Xiao
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Canada; Covenant Health Palliative Institute, Edmonton, Alberta, Canada
| | - Leonardo R Gama
- Departamento de Radiologia e Oncologia & Instituto do Câncer do Estado de São Paulo, Universidade de Sao Paulo, São Paulo, Brazil
| | - Silvio P Gomes
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil; Universidade de Sao Paulo Faculdade de Medicina Veterinaria, Departamento de Cirurgia, Brazil
| | - Daniela Caetano Gonçalves
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil; Universidade Federal de Sao Paulo, Instituto de Biociencias, Santos, Brazil
| | - Raquel G F Costa
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Katrin Radloff
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Ulrike Lenz
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Anna E Taranko
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| | - Fang Chia Bin
- Santa Casa de Misericoria de Sao Paulo, São Paulo, Brazil
| | | | | | | | - Luis H A Nucci
- Instituto do Cancer Arnaldo Vieira de Carvalho, São Paulo, Brazil
| | - Mario Feitoza
- Instituto do Cancer Arnaldo Vieira de Carvalho, São Paulo, Brazil
| | - Claudio C de Castro
- Universidade de Sao Paulo Faculdade de Medicina, Departamento de Radiologia, São Paulo, Brazil; Universidade de Sao Paulo Hospital Universitario, São Paulo, Brazil
| | - Flavio Tokeshi
- Universidade de Sao Paulo Hospital Universitario, São Paulo, Brazil
| | | | - Jose P Otoch
- Universidade de Sao Paulo Hospital Universitario, São Paulo, Brazil
| | - Alexandre F Ramos
- Departamento de Radiologia e Oncologia & Instituto do Câncer do Estado de São Paulo, Universidade de Sao Paulo, São Paulo, Brazil; Escola de Artes, Ciencias e Humanidades, Universidade de Sao Paulo, São Paulo, Brazil
| | - Alessandro Laviano
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Dario Coletti
- Sorbonne Université, Department of Biological Adaptation and Aging, B2A, Paris, France; Department of AHFMO - Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Vera C Mazurak
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Canada
| | - Carla M Prado
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Canada
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Universidade de Sao Paulo Instituto de Ciencias Biomedicas, Departamento de Biologia Celular e do Desenvolvimento and Faculdade de Medicina da Universidade de Sao Paulo, Departamento de Cirurgia, LIM 26-HC, São Paulo, Brazil
| |
Collapse
|
44
|
Gao HC, Huang YZ, Liu YQ, Chen Y, Wang ZH, Yin GH. Role of TG2 and TGF-β 1 in the pathogenesis of human breast cancer. Oncol Lett 2020; 20:221. [PMID: 32963627 PMCID: PMC7491046 DOI: 10.3892/ol.2020.12057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/15/2020] [Indexed: 12/04/2022] Open
Abstract
The present study analyzed the role of transforming growth factor-β1 (TGF-β1) and tissue transglutaminase (TG2) in breast cancer, as well as their protein levels in MCF-7 cells treated with cisplatin. In addition, the present study investigated the effects of TG2 and TGF-β1 in MCF-7 cells following TGF-β1 and TG2 inhibition or TGF-β1 induction. The protein levels of TG2 and TGF-β1 in breast cancer tissues and in MCF-7 cells treated with cisplatin, TG2 and TGF-β1 inhibitors or 10 ng/ml TGF-β1 were analyzed by immunohistochemical staining, immunofluorescence and western blotting. The results revealed that the expression levels of TG2 and TGF-β1 in breast cancer tissues were significantly higher compared with those in paracancerous tissues. The fluorescence intensity of TG2 and TGF-β1 in MCF-7 cells treated with cisplatin was lower compared with that in untreated MCF-7 cells. Using bioinformatics analysis, the present study predicted that TGF-β1 may be associated with TG2. In addition, the expression levels of TGF-β1 and TG2 in MCF-7 cells treated with inhibitors of TGF-β1 and TG2 were lower compared with those in untreated MCF-7 cells. By contrast, the expression levels of TGF-β1 and TG2 in MCF-7 cells treated with TGF-β1 were higher compared with those in untreated MCF-7 cells. Therefore, the present study demonstrated that TGF-β1 and TG2 may serve an important role in breast cancer tissues and in MCF-7 cells. In addition, it was revealed that TG2 and TGF-β1 may have a synergistic role in MCF-7 cells.
Collapse
Affiliation(s)
- Hai-Cheng Gao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan-Zhi Huang
- Department of Respiratory Medicine, Children's Hospital, Changchun, Jilin 130000, P.R. China
| | - Yu-Qi Liu
- Department of Respiratory Medicine, Children's Hospital, Changchun, Jilin 130000, P.R. China
| | - Yan Chen
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhe-Hui Wang
- Department of Surgery, China-Japan Union Hospital, Changchun, Jilin 130031, P.R. China
| | - Guang-Hao Yin
- Department of Breast Surgery, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
45
|
Melchor SJ, Hatter JA, Castillo ÉAL, Saunders CM, Byrnes KA, Sanders I, Abebayehu D, Barker TH, Ewald SE. T. gondii infection induces IL-1R dependent chronic cachexia and perivascular fibrosis in the liver and skeletal muscle. Sci Rep 2020; 10:15724. [PMID: 32973293 PMCID: PMC7515928 DOI: 10.1038/s41598-020-72767-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a progressive muscle wasting disease that contributes to death in a wide range of chronic diseases. Currently, the cachexia field lacks animal models that recapitulate the long-term kinetics of clinical disease, which would provide insight into the pathophysiology of chronic cachexia and a tool to test therapeutics for disease reversal. Toxoplasma gondii (T. gondii) is a protozoan parasite that uses conserved mechanisms to infect rodents and human hosts. Infection is lifelong and has been associated with chronic weight loss and muscle atrophy in mice. We have recently shown that T. gondii-induced muscle atrophy meets the clinical definition of cachexia. Here, the longevity of the T. gondii-induced chronic cachexia model revealed that cachectic mice develop perivascular fibrosis in major metabolic organs, including the adipose tissue, skeletal muscle, and liver by 9 weeks post-infection. Development of cachexia, as well as liver and skeletal muscle fibrosis, is dependent on intact signaling through the type I IL-1R receptor. IL-1α is sufficient to activate cultured fibroblasts and primary hepatic stellate cells (myofibroblast precursors in the liver) in vitro, and IL-1α is elevated in the sera and liver of cachectic, suggesting a mechanism by which chronic IL-1R signaling could be leading to cachexia-associated fibrosis.
Collapse
Affiliation(s)
- Stephanie J Melchor
- Department of Microbiology, Immunology, and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jessica A Hatter
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Claire M Saunders
- Department of Microbiology, Immunology, and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kari A Byrnes
- Department of Microbiology, Immunology, and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Imani Sanders
- Department of Microbiology, Immunology, and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sarah E Ewald
- Department of Microbiology, Immunology, and Cancer Biology and The Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
46
|
Zhu X, Callahan MF, Gruber KA, Szumowski M, Marks DL. Melanocortin-4 receptor antagonist TCMCB07 ameliorates cancer- and chronic kidney disease-associated cachexia. J Clin Invest 2020; 130:4921-4934. [PMID: 32544087 PMCID: PMC7456235 DOI: 10.1172/jci138392] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Cachexia, a devastating wasting syndrome characterized by severe weight loss with specific losses of muscle and adipose tissue, is driven by reduced food intake, increased energy expenditure, excess catabolism, and inflammation. Cachexia is associated with poor prognosis and high mortality and frequently occurs in patients with cancer, chronic kidney disease, infection, and many other illnesses. There is no effective treatment for this condition. Hypothalamic melanocortins have a potent and long-lasting inhibitory effect on feeding and anabolism, and pathophysiological processes increase melanocortin signaling tone, leading to anorexia, metabolic changes, and eventual cachexia. We used 3 rat models of anorexia and cachexia (LPS, methylcholanthrene sarcoma, and 5/6 subtotal nephrectomy) to evaluate efficacy of TCMCB07, a synthetic antagonist of the melanocortin-4 receptor. Our data show that peripheral treatment using TCMCB07 with intraperitoneal, subcutaneous, and oral administration increased food intake and body weight and preserved fat mass and lean mass during cachexia and LPS-induced anorexia. Furthermore, administration of TCMCB07 diminished hypothalamic inflammatory gene expression in cancer cachexia. These results suggest that peripheral TCMCB07 treatment effectively inhibits central melanocortin signaling and therefore stimulates appetite and enhances anabolism, indicating that TCMCB07 is a promising drug candidate for treating cachexia.
Collapse
MESH Headings
- Animals
- Appetite/drug effects
- Cachexia/drug therapy
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Male
- Rats
- Rats, Sprague-Dawley
- Receptor, Melanocortin, Type 4/antagonists & inhibitors
- Receptor, Melanocortin, Type 4/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Sarcoma, Experimental/complications
- Sarcoma, Experimental/drug therapy
- Sarcoma, Experimental/metabolism
- Sarcoma, Experimental/pathology
Collapse
Affiliation(s)
- Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Michael F. Callahan
- Tensive Controls Inc., MU Life Sciences Business Incubator at Monsanto Place, Columbia, Missouri, USA
| | - Kenneth A. Gruber
- Tensive Controls Inc., MU Life Sciences Business Incubator at Monsanto Place, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center and
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Marek Szumowski
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Daniel L. Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, Oregon, USA
- Knight Cancer Institute and
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
47
|
Miller J, Dreczkowski G, Ramage MI, Wigmore SJ, Gallagher IJ, Skipworth RJE. Adipose depot gene expression and intelectin-1 in the metabolic response to cancer and cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1141-1153. [PMID: 32232960 PMCID: PMC7432578 DOI: 10.1002/jcsm.12568] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/21/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancer cachexia is a poorly understood metabolic consequence of cancer. During cachexia, different adipose depots demonstrate differential wasting rates. Animal models suggest adipose tissue may be a key driver of muscle wasting through fat-muscle crosstalk, but human studies in this area are lacking. We performed global gene expression profiling of visceral (VAT) and subcutaneous (SAT) adipose from weight stable and cachectic cancer patients and healthy controls. METHODS Cachexia was defined as >2% weight loss plus low computed tomography-muscularity. Biopsies of SAT and VAT were taken from patients undergoing resection for oesophago-gastric cancer, and healthy controls (n = 16 and 8 respectively). RNA was isolated and reverse transcribed. cDNA was hybridised to the Affymetrix Clariom S microarray and data analysed using R/Bioconductor. Differential expression of genes was assessed using empirical Bayes and moderated-t-statistic approaches. Category enrichment analysis was used with a tissue-specific background to examine the biological context of differentially expressed genes. Selected differentially regulated genes were validated by qPCR. Enzyme-linked immunosorbent assay (ELISA) for intelectin-1 was performed on all VAT samples. The previously-described cohort plus 12 additional patients from each group also had plasma I = intelectin-1 ELISA carried out. RESULTS In VAT vs. SAT comparisons, there were 2101, 1722, and 1659 significantly regulated genes in the cachectic, weight stable, and control groups, respectively. There were 2200 significantly regulated genes from VAT in cachectic patients compared with controls. Genes involving inflammation were enriched in cancer and control VAT vs. SAT, although different genes contributed to enrichment in each group. Energy metabolism, fat browning (e.g. uncoupling protein 1), and adipogenesis genes were down-regulated in cancer VAT (P = 0.043, P = 5.4 × 10-6 and P = 1 × 10-6 respectively). The gene showing the largest difference in expression was ITLN1, the gene that encodes for intelectin-1 (false discovery rate-corrected P = 0.0001), a novel adipocytokine associated with weight loss in other contexts. CONCLUSIONS SAT and VAT have unique gene expression signatures in cancer and cachexia. VAT is metabolically active in cancer, and intelectin-1 may be a target for therapeutic manipulation. VAT may play a fundamental role in cachexia, but the down-regulation of energy metabolism genes implies a limited role for fat browning in cachectic patients, in contrast to pre-clinical models.
Collapse
Affiliation(s)
- Janice Miller
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | | | - Michael I Ramage
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Stephen J Wigmore
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Iain J Gallagher
- Faculty of Health Science and Sport, University of Stirling, Stirling, UK
| | - Richard J E Skipworth
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
48
|
Vohra R, Campbell MD, Park J, Whang S, Gravelle K, Wang YN, Hwang JH, Marcinek DJ, Lee D. Increased tumour burden alters skeletal muscle properties in the KPC mouse model of pancreatic cancer. JCSM RAPID COMMUNICATIONS 2020; 3:44-55. [PMID: 33073264 PMCID: PMC7566781 DOI: 10.1002/rco2.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND Cancer cachexia is a multifactorial wasting syndrome that is characterized by the loss of skeletal muscle mass and weakness, which compromises physical function, reduces quality of life, and ultimately can lead to mortality. Experimental models of cancer cachexia have recapitulated this skeletal muscle atrophy and consequent decline in muscle force generating capacity. We address these issues in a novel transgenic mouse model Kras, Trp53 and Pdx-1-Cre (KPC) of pancreatic ductal adenocarcinoma (PDA) using multi-parametric magnetic resonance (mp-MR) measures. METHODS KPC mice (n = 10) were divided equally into two groups (n = 5/group) depending on the size of the tumor i.e. tumor size <250 mm3 and >250 mm3. Using mp-MR measures, we demonstrated the changes in the gastrocnemius muscle at the microstructural level. In addition, we evaluated skeletal muscle contractile function in KPC mice using an in vivo approach. RESULTS Increase in tumor size resulted in decrease in gastrocnemius maximum cross sectional area, decrease in T2 relaxation time, increase in magnetization transfer ratio, decrease in mean diffusivity, and decrease in radial diffusivity of water across the muscle fibers. Finally, we detected significant decrease in absolute and specific force production of gastrocnemius muscle with increase in tumor size. CONCLUSIONS Our findings indicate that increase in tumor size may cause alterations in structural and functional parameters of skeletal muscles and that MR parameters may be used as sensitive biomarkers to noninvasively detect structural changes in cachectic muscles.
Collapse
Affiliation(s)
- Ravneet Vohra
- Department of Radiology, University of Washington, Seattle,
USA
| | | | - Joshua Park
- Department of Radiology, University of Washington, Seattle,
USA
| | - Stella Whang
- Department of Medicine, University of Washington, Seattle,
USA
| | - Kayla Gravelle
- Department of Medicine, University of Washington, Seattle,
USA
| | - Yak-Nam Wang
- Applied Physics Laboratory, University of Washington,
Seattle, USA
| | - Joo-Ha Hwang
- Division of Gastroenterology and Hepatology, Stanford
University, Stanford, USA
| | | | - Donghoon Lee
- Department of Radiology, University of Washington, Seattle,
USA
| |
Collapse
|
49
|
Adipocytes in Breast Cancer, the Thick and the Thin. Cells 2020; 9:cells9030560. [PMID: 32120856 PMCID: PMC7140407 DOI: 10.3390/cells9030560] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
It is well established that breast cancer development and progression depend not only on tumor-cell intrinsic factors but also on its microenvironment and on the host characteristics. There is growing evidence that adipocytes play a role in breast cancer progression. This is supported by: (i) epidemiological studies reporting the association of obesity with a higher cancer risk and poor prognosis, (ii) recent studies demonstrating the existence of a cross-talk between breast cancer cells and adipocytes locally in the breast that leads to acquisition of an aggressive tumor phenotype, and (iii) evidence showing that cancer cachexia applies also to fat tissue and shares similarities with stromal-carcinoma metabolic synergy. This review summarizes the current knowledge on the epidemiological link between obesity and breast cancer and outlines the results of the tumor-adipocyte crosstalk. We also focus on systemic changes in body fat in patients with cachexia developed in the course of cancer. Moreover, we discuss and compare adipocyte alterations in the three pathological conditions and the mechanisms through which breast cancer progression is induced.
Collapse
|
50
|
Rosa-Caldwell ME, Fix DK, Washington TA, Greene NP. Muscle alterations in the development and progression of cancer-induced muscle atrophy: a review. J Appl Physiol (1985) 2019; 128:25-41. [PMID: 31725360 DOI: 10.1152/japplphysiol.00622.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer cachexia-cancer-associated body weight and muscle loss-is a significant predictor of mortality and morbidity in cancer patients across a variety of cancer types. However, despite the negative prognosis associated with cachexia onset, there are no clinical therapies approved to treat or prevent cachexia. This lack of treatment may be partially due to the relative dearth of literature on mechanisms occurring within the muscle before the onset of muscle wasting. Therefore, the purpose of this review is to compile the current scientific literature on mechanisms contributing to the development and progression of cancer cachexia, including protein turnover, inflammatory signaling, and mitochondrial dysfunction. We define "development" as changes in cell function occurring before the onset of cachexia and "progression" as alterations to cell function that coincide with the exacerbation of muscle wasting. Overall, the current literature suggests that multiple aspects of cellular function, such as protein turnover, inflammatory signaling, and mitochondrial quality, are altered before the onset of muscle loss during cancer cachexia and clearly highlights the need to study more thoroughly the developmental stages of cachexia. The studying of these early aberrations will allow for the development of effective therapeutics to prevent the onset of cachexia and improve health outcomes in cancer patients.
Collapse
Affiliation(s)
- Megan E Rosa-Caldwell
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Dennis K Fix
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Nicholas P Greene
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|