1
|
Davis MP, Bader N, Basting J, Vanenkevort E, Koppenhaver N, Patel A, Gupta M, Lagerman B, Wojtowicz M. Are Muscle and Fat Loss Predictive of Clinical Events in Pancreatic Cancer? The Importance of Precision Metrics. J Pain Symptom Manage 2025; 69:141-151. [PMID: 39461674 DOI: 10.1016/j.jpainsymman.2024.10.004] [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: 05/06/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024]
Abstract
CONTEXT Muscle and fat loss from cancer may have prognostic significance. Skeletal muscle and fat areas measured at L3 on a CT scan correlate with body muscle and fat mass. We wished to know if reduced skeletal muscle area or fat on diagnostic CT scans or changes from initial CT scans in patients with pancreatic cancer who died in 2018 and 2019 predicted mortality. METHOD Electronic records of 112 patients with locally advanced or metastatic pancreatic cancer were used to extract stage, age, gender, comorbidities, weight, and height at the time of the first CT scan. Survival (in days) was defined from the first CT scan to the death date. Patients had at least one CT scan of the abdomen. I. Two trained medical students read scans independently using TeraRecon software (Durham, NC). Results were averaged, and the differences determined precision. Interclass correlation coefficient (ICC), coefficient of variation, and least significant change determined the precision between readers. Independent prognostic modeling included age and BMI. RESULTS An evaluable sample of 104 with an average age of 67, 56 were male. Nearly half had a TNM Stage of IV (45%). The average Charlson Comorbidity index is 7.2. In those undergoing repeat scans, most were in the timeframe of 60-120 days. Changes in visceral fat in men in the unadjusted Cox proportional hazard model and reduced skeletal muscle area in the age-adjusted model of men predicted mortality. In contrast, myosteatosis in women marginally predicted improved survival. ICC's precision between readers was adequate but by least significant change would have missed subtle, clinically important changes. DISCUSSION Muscle loss during chemotherapy in men predicted mortality in men but not women. Precision is an important metric when measuring body composition. CONCLUSION Muscle loss in men during chemotherapy of pancreatic cancer predicts mortality.
Collapse
Affiliation(s)
- Mellar P Davis
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA.
| | - Nada Bader
- Geisinger Commonwealth School of Medicine (N.B., J.B.), Scranton, PA
| | - James Basting
- Geisinger Commonwealth School of Medicine (N.B., J.B.), Scranton, PA
| | - Erin Vanenkevort
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA
| | | | - Aalpen Patel
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA
| | - Mudit Gupta
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA
| | - Braxton Lagerman
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA
| | - Mark Wojtowicz
- Geisinger Health System (M.P.D., N.K., A.P., M.G., B.L.), Danville, PA
| |
Collapse
|
2
|
Onyema MC, Oštarijaš E, Zair Z, Roy A, Minhas R, Lajeunesse-Trempe F, Kearney J, Drakou EE, Grossman AB, Aylwin SJ, Canecki-Varžić S, Dimitriadis GK. The Role of Active Brown Adipose Tissue in Patients With Pheochromocytoma or Paraganglioma. Endocr Pract 2024:S1530-891X(24)00828-0. [PMID: 39557120 DOI: 10.1016/j.eprac.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/24/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVES Metabolically-active brown adipose tissue (aBAT) is a common finding on 18fluorodeoxyglucose positron emission tomography (FDG-PET) imaging in patients with pheochromocytoma or paraganglioma (PPGL). In addition to its clinical significance, we aimed to explore the prevalence of this finding on FDG-PET imaging in patients with PPGL. METHODS We conducted a systematic review and meta-analysis of prospective and retrospective studies. Publications were identified through searches in MEDLINE/PubMed, Embase, and SCOPUS from inception until 2022-11-26, with an update check performed on 2024-05-02. Eligible studies included patients with PPGL who had completed FDG-PET imaging. Data on catecholamine levels stratified by the presence of aBAT were extracted and pooled using the random-effects model with the inverse variance method. For the quantitative synthesis, we used standardized mean differences and meta-analysis of proportions. A risk of bias assessment was performed using the Quality in Prognostic Studies tool. RESULTS Our search yielded 6 studies suitable for inclusion. Pooled data showed a statistically significant positive difference in isolated demethylated catecholamine levels in aBAT positive groups compared to aBAT negative. No significant differences were found in multiple domains, including tumor size, tumor burden, germline mutations, or location. The proportion of patients with PPGL who present with aBAT stands at approximately 25%. CONCLUSIONS The demethylated metabolite levels could have potential use in predicting the presence of active brown adipose tissue in patients with PPGL. There is no convincing evidence of increased aBAT prevalence in patients with PPGL and germline mutations. There was, however, evidence suggesting that the presence of aBAT may confer poorer outcomes and decreased life expectancy.
Collapse
Affiliation(s)
- Michael C Onyema
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Eduard Oštarijaš
- Doctoral School of Clinical Medical Sciences, Medical School, University of Pecs, Pecs, Hungary
| | - Zoulikha Zair
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Aparajita Roy
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Raisa Minhas
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Fannie Lajeunesse-Trempe
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom; Department of Nutrition, Laval University, Quebec City, Canada; Faculty of Pharmacy, Laval University, Quebec City, Canada
| | - Jessica Kearney
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Eftychia E Drakou
- Department of Clinical Oncology, Guy's Cancer Centre - Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ashley B Grossman
- Green Templeton College, University of Oxford, Oxford, UK; Centre for Endocrinology, William Harvey Institute, Barts and the London School of Medicine, London, UK; Neuroendocrine Tumour Unit, Royal Free Hospital, London, UK
| | - Simon Jb Aylwin
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Silvija Canecki-Varžić
- Doctoral School of Clinical Medical Sciences, Medical School, University of Pecs, Pecs, Hungary; J. J. Strossmayer University of Osijek, Faculty of Medicine, Osijek, Croatia
| | - Georgios K Dimitriadis
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, United Kingdom; Faculty of Life Sciences and Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Obesity, Type 2 Diabetes and Immunometabolism Research Group, King's College London, London, United Kingdom.
| |
Collapse
|
3
|
Ho PT, Park E, Luong QXT, Hakim MD, Hoang PT, Vo TTB, Kawalin K, Kang H, Lee TK, Lee S. Amelioration of Cancer Cachexia by Dalbergia odorifera Extract Through AKT Signaling Pathway Regulation. Nutrients 2024; 16:3671. [PMID: 39519503 PMCID: PMC11547832 DOI: 10.3390/nu16213671] [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: 10/10/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Cancer cachexia is a multifactorial syndrome characterized by the progressive loss of skeletal muscle mass and adipose tissue. Dalbergia odorifer is widely used in traditional medicine in Korea and China to treat various diseases. However, its exact role and underlying mechanism in regulating cancer cachexia have not been elucidated yet. This research was conducted to investigate the effect of D. odorifer extract (DOE) in preventing the development of cancer-induced cachexia symptoms and figure out the relevant mechanisms. Methods: A cancer cachexia model was established in Balb/c mice using the CT26 colon carcinoma cell line. To evaluate the anti-cachexia effect of Dalbergia odorifer extract (DOE), CT26-bearing mice were orally administered with DOE at concentrations of 50 and 100 mg/kg BW for 14 days. C2C12 myotubes and 3T3L1 adipocytes were treated with 80% CT26 conditioned medium, DOE, and wortmannin, a particular AKT inhibitor to determine the influence of DOE in the AKT signaling pathway. Mice body weight, food intake, myofiber cross-sectional area, adipocyte size, myotube diameter, lipid accumulation, and relevant gene expression were analyzed. Results: The oral administration of DOE at doses of 50 and 100 mg/kg body weight to CT26 tumor-bearing mice resulted in a significant reduction in body weight loss, an increase in food intake, and a decrease in serum glycerol levels. Furthermore, DOE treatment led to an increase in muscle mass, larger muscle fiber diameter, and elevated expression levels of MyH2 and Igf1, while simultaneously reducing the expression of Atrogin1 and MuRF1. DOE also attenuated adipose tissue wasting, as evidenced by increased epididymal fat mass, enlarged adipocyte size, and upregulated Pparγ expression, alongside a reduction in Ucp1 and IL6 levels. In cachectic C2C12 myotubes and 3T3-L1 adipocytes induced by the CT26 conditioned medium, DOE significantly inhibited muscle wasting and lipolysis by activating the AKT signaling pathway. The treatment of wortmannin, a specific AKT inhibitor, effectively neutralized DOE's impact on the AKT pathway, myotube diameter, and lipid accumulation. Conclusions: DOE ameliorates cancer cachexia through the expression of genes involved in protein synthesis and lipogenesis, while suppressing those related to protein degradation, suggesting its potential as a plant-derived therapeutic agent in combating cancer cachexia.
Collapse
Affiliation(s)
- Phuong T. Ho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Eulyong Park
- R&D Center, Easthill Corporation, Suwon 16642, Republic of Korea;
| | - Quynh Xuan Thi Luong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Meutia Diva Hakim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Phuong T. Hoang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Thuy T. B. Vo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Kantawong Kawalin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| | - Hee Kang
- Humanitas College, Kyung Hee University, 1732 Deogyeongdae-ro, Yongin 17104, Republic of Korea;
| | - Taek-Kyun Lee
- Ecological Risk Research Department, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (P.T.H.); (Q.X.T.L.); (M.D.H.); (P.T.H.); (T.T.B.V.); (K.K.)
| |
Collapse
|
4
|
Zakic T, Pekovic-Vaughan V, Cvoro A, Korac A, Jankovic A, Korac B. Redox and metabolic reprogramming in breast cancer and cancer-associated adipose tissue. FEBS Lett 2024; 598:2106-2134. [PMID: 38140817 DOI: 10.1002/1873-3468.14794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Redox and metabolic processes are tightly coupled in both physiological and pathological conditions. In cancer, their integration occurs at multiple levels and is characterized by synchronized reprogramming both in the tumor tissue and its specific but heterogeneous microenvironment. In breast cancer, the principal microenvironment is the cancer-associated adipose tissue (CAAT). Understanding how the redox-metabolic reprogramming becomes coordinated in human breast cancer is imperative both for cancer prevention and for the establishment of new therapeutic approaches. This review aims to provide an overview of the current knowledge of the redox profiles and regulation of intermediary metabolism in breast cancer while considering the tumor and CAAT of breast cancer as a unique Warburg's pseudo-organ. As cancer is now recognized as a systemic metabolic disease, we have paid particular attention to the cell-specific redox-metabolic reprogramming and the roles of estrogen receptors and circadian rhythms, as well as their crosstalk in the development, growth, progression, and prognosis of breast cancer.
Collapse
Affiliation(s)
- Tamara Zakic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Vanja Pekovic-Vaughan
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, William Henry Duncan Building, University of Liverpool, UK
| | | | | | - Aleksandra Jankovic
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
| | - Bato Korac
- Institute for Biological Research "Sinisa Stankovic"-National Institute of Republic of Serbia, University of Belgrade, Serbia
- Faculty of Biology, University of Belgrade, Serbia
| |
Collapse
|
5
|
Han J, Liu X, Wang J, Tang M, Xu J, Tan S, Liu X, Wu G. Prognostic value of body composition in patients with digestive tract cancers: A prospective cohort study of 8,267 adults from China. Clin Nutr ESPEN 2024; 62:192-198. [PMID: 38901941 DOI: 10.1016/j.clnesp.2024.04.017] [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: 08/10/2023] [Revised: 03/05/2024] [Accepted: 04/27/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND & AIMS The characterization and prognostic value of body composition parameter/phenotype based on computed tomography (CT) in patients with digestive tract cancers remain incomplete. This study aimed to investigate the relationship between parameter/phenotype and clinical outcomes in patients with digestive tract cancers. METHODS In this prospective cohort study, 8267 patients with digestive tract cancers were assessed using CT scans to determine body composition. Body composition data, including areas of skeletal muscle (SM), subcutaneous adipose tissue (SAT), and visceral adipose tissue (VAT), were collected at the third lumbar level on CT images obtained within 30 days before surgery. Body composition phenotypes (sarcopenia, cancer cachexia, sarcopenic obesity) were determined based on SM, SAT, and VAT areas. The primary endpoint was overall survival, obtained from electronic medical records and telephone follow-up surveys. Kaplan-Meier and log-rank analyses were employed to compare unadjusted survival, while multivariate survival analyses were conducted using a proportional hazards model adjusted for age, gender, and cancer-node-metastasis (TNM) stages. RESULTS Adjusted hazard ratios (HRs) for all-cause mortality were calculated for the second (Q2), third (Q3), and fourth (Q4) quantiles relative to the first quantile (Q1) for SM areas, revealing adjusted summary HRs of 0.575 (95% CI, 0.361-0.916), 0.419 (95% CI, 0.241-0.729), and 0.384 (95% CI, 0.203-0.726), respectively. Sarcopenia-adjusted summary HRs were 1.795 (95% CI: 1.012-3.181) for male patients and 1.925 (95% CI: 1.065-3.478) for female patients. Cancer cachexia-adjusted summary HRs were 1.542 (95% CI: 1.023-2.324) for male patients and 1.569 (95% CI: 0.820-3.001) for female patients. Sarcopenic obesity-adjusted summary HRs were 1.122 (95% CI: 0.759-1.657) for male patients and 1.303 (95% CI: 0.623-2.725) for female patients. Subgroup analyses indicated varying prognostic values of body composition parameter/phenotype among different cancer types. CONCLUSIONS Our findings suggest a large SM area is a favorable prognostic indicator, while cancer cachexia and sarcopenia signify poor prognosis in patients with digestive tract cancers. These findings have important implications for the personalized preoperative assessment of body composition in patients with digestive tract cancers.
Collapse
Affiliation(s)
- Jun Han
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Center, Shanghai, China
| | - Xinyang Liu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junjie Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Tang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahao Xu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shanjun Tan
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Center, Shanghai, China
| | - Xin Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Center, Shanghai, China.
| |
Collapse
|
6
|
Mota INR, Satari S, Marques IS, Santos JMO, Medeiros R. Adipose tissue rearrangement in cancer cachexia: The involvement of β3-adrenergic receptor associated pathways. Biochim Biophys Acta Rev Cancer 2024; 1879:189103. [PMID: 38679401 DOI: 10.1016/j.bbcan.2024.189103] [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: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Cancer-associated cachexia (CAC) is a complex multiple organ syndrome that significantly contributes to reduced quality of life and increased mortality among many cancer patients. Its multifactorial nature makes its early diagnosis and effective therapeutic interventions challenging. Adipose tissue is particularly impacted by cachexia, typically through increased lipolysis, browning and thermogenesis, mainly at the onset of the disease. These processes lead to depletion of fat mass and contribute to the dysfunction of other organs. The β-adrenergic signalling pathways are classical players in the regulation of adipose tissue metabolism. They are activated upon sympathetic stimulation inducing lipolysis, browning and thermogenesis, therefore contributing to energy expenditure. Despite accumulating evidence suggesting that β3-adrenergic receptor stimulation may be crucial to the adipose tissue remodelling during cachexia, the literature remains controversial. Moreover, there is limited knowledge regarding sexual dimorphism of adipose tissue in the context of cachexia. This review paper aims to present the current knowledge regarding adipose tissue wasting during CAC, with a specific focus on the role of the β3-adrenergic receptor, placing it as a potential therapeutic target against cachexia.
Collapse
Affiliation(s)
- Inês N R Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal.
| | - Setareh Satari
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal.
| | - Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal.
| | - Joana M O Santos
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Research Department of the Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), 4200-172 Porto, Portugal.
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Research Department of the Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), 4200-172 Porto, Portugal; Virology Service, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal.
| |
Collapse
|
7
|
Zhai S, Li X, Lin T. Obese Mouse Fat Cell-derived Extracellular Vesicles Transport miR-99a-5p to Mitigate the Proliferation and Migration of Non-small Cell Lung Cancer Cells. Comb Chem High Throughput Screen 2024; 27:214-226. [PMID: 36927435 DOI: 10.2174/1386207326666230316103604] [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: 08/17/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 03/18/2023]
Abstract
OBJECTIVE Fat cells-derived extracellular vesicles (FC-EVs) play a role in regulating the tumor microenvironment in cancers by transporting RNAs. MicroRNAs (miRNAs) are vital regulators of cancer development. This study was conducted to explore the role of FC-EVs in the proliferation and migration of non-small cell lung cancer (NSCLC) cells, providing targets for NSCLC treatment. METHODS The obese mouse model was established via high-fat diet (HFD), followed by separation and characterization of FC-EVs (HFD-EVs). The levels of miR-99a-5p, precursor-miR-99a-5p, and heparan sulfate-glucosamine 3-sulfotransferase 3B1 (HS3ST3B1) were measured by RT-qPCR or Western blot assay. Cell proliferation and migration were evaluated by 3-(4, 5-dimethylthiazol- 2-yl)-2, 5-diphenyltetrazolium bromide and wound healing assays. The expression of Cy3-labeled miR-99a-5p in A549 cells (one NSCLC cell line) was observed via confocal microscopy. The binding of miR-99a-5p to HS3ST3B1 was analyzed by the dual luciferase assay. Rescue experiments were performed to confirm the role of HS3ST3B1 in NSCLC cells. RESULTS miR-99a-5p was upregulated in adipose tissues, FCs, and HFD-EVs. HFD-EVs mitigated the proliferation and migration of NSCLC cells. HFD-EVs transported miR-99a-5p into A549 cells, which upregulated miR-99a-5p expression and inhibited HS3ST3B1 expression in A549 cells. HS3ST3B1 overexpression reversed the inhibition of HFD-EVs on the proliferation and migration of NSCLC cells. CONCLUSION HFD-EVs transported miR-99a-5p into NSCLC cells and inhibited HS3ST3B1, thereby inhibiting proliferation and migration of NSCLC cells.
Collapse
Affiliation(s)
- Shengping Zhai
- Department of General Practice, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xiaoping Li
- Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Tiantian Lin
- Respiratory Intensive Care Unit, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| |
Collapse
|
8
|
Grigoraș A, Amalinei C. Multi-Faceted Role of Cancer-Associated Adipocytes in Colorectal Cancer. Biomedicines 2023; 11:2401. [PMID: 37760840 PMCID: PMC10525260 DOI: 10.3390/biomedicines11092401] [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: 07/17/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed types of cancer, especially in obese patients, and the second cause of cancer-related death worldwide. Based on these data, extensive research has been performed over the last decades to decipher the pivotal role of the tumor microenvironment (TME) and its cellular and molecular components in CRC development and progression. In this regard, substantial progress has been made in the identification of cancer-associated adipocytes' (CAAs) characteristics, considering their active role in the CCR tumor niche, by releasing a panel of metabolites, growth factors, and inflammatory adipokines, which assist the cancer cells' development. Disposed in the tumor invasion front, CAAs exhibit a fibroblastic-like phenotype and establish a bidirectional molecular dialogue with colorectal tumor cells, which leads to functional changes in both cell types and contributes to tumor progression. CAAs also modulate the antitumor immune cells' response and promote metabolic reprogramming and chemotherapeutic resistance in colon cancer cells. This review aims to report recent cumulative data regarding the molecular mechanisms of CAAs' differentiation and their activity spectrum in the TME of CRC. A better understanding of CAAs and the molecular interplay between CAAs and tumor cells will provide insights into tumor biology and may open the perspective of new therapeutic opportunities in CRC patients.
Collapse
Affiliation(s)
- Adriana Grigoraș
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| | - Cornelia Amalinei
- Department of Morphofunctional Sciences I, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Histopathology, Institute of Legal Medicine, 700455 Iasi, Romania
| |
Collapse
|
9
|
Eljalby M, Huang X, Becher T, Wibmer AG, Jiang CS, Vaughan R, Schöder H, Cohen P. Brown adipose tissue is not associated with cachexia or increased mortality in a retrospective study of patients with cancer. Am J Physiol Endocrinol Metab 2023; 324:E144-E153. [PMID: 36576355 PMCID: PMC9902220 DOI: 10.1152/ajpendo.00187.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/28/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Although brown fat is strongly associated with a constellation of cardiometabolic benefits in animal models and humans, it has also been tied to cancer cachexia. In humans, cancer-associated cachexia increases mortality, raising the possibility that brown fat in this context may be associated with increased cancer death. However, the effect of brown fat on cancer-associated cachexia and survival in humans remains unclear. Here, we retrospectively identify patients with and without brown fat on fluorodeoxyglucose (18F-FDG) positron-emission tomography (PET) scans obtained as part of routine cancer care and assemble a cohort to address these questions. We did not find an association between brown fat status and cachexia. Furthermore, we did not observe an association between brown fat and increased mortality in patients with cachexia. Our analyses controlled for confounding factors including age at cancer diagnosis, sex, body mass index, cancer site, cancer stage, outdoor temperature, comorbid conditions (heart failure, type 2 diabetes mellitus, coronary artery disease, hypertension, dyslipidemia, cerebrovascular disease), and β-blocker use. Taken together, our results suggest that brown fat is not linked to cancer-associated cachexia and does not worsen overall survival in patients with cachexia.NEW & NOTEWORTHY This study finds that brown fat is not linked to cancer-associated cachexia. Moreover, this work shows that brown fat does not worsen overall survival in patients with cachexia.
Collapse
Affiliation(s)
- Mahmoud Eljalby
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Xiaojing Huang
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tobias Becher
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
- First Department of Medicine (Division of Cardiology), University Medical Center Mannheim, Mannheim, Germany
| | - Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Caroline S Jiang
- Center for Clinical and Translational Science, The Rockefeller University, New York City, New York
| | - Roger Vaughan
- Center for Clinical and Translational Science, The Rockefeller University, New York City, New York
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York City, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, New York
| |
Collapse
|
10
|
Warrier M, Paules EM, Silva-Gomez J, Friday WB, Bramlett F, Kim H, Zhang K, Trujillo-Gonzalez I. Homocysteine-induced endoplasmic reticulum stress activates FGF21 and is associated with browning and atrophy of white adipose tissue in Bhmt knockout mice. Heliyon 2023; 9:e13216. [PMID: 36755585 PMCID: PMC9900266 DOI: 10.1016/j.heliyon.2023.e13216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023] Open
Abstract
Betaine-homocysteine methyltransferase (BHMT) catalyzes the transfer of methyl groups from betaine to homocysteine (Hcy), producing methionine and dimethylglycine. In this work, we characterize Bhmt wild type (Bhmt-WT) and knockout (Bhmt-KO) mice that were fully backcrossed to a C57Bl6/J background. Consistent with our previous findings, Bhmt-KO mice had decreased body weight, fat mass, and adipose tissue weight compared to WT. Histological analyses and gene expression profiling indicate that adipose browning was activated in KO mice and contributed to the adipose atrophy observed. BHMT is not expressed in adipose tissue but is abundant in liver; thus, a signal must originate from the liver that modulates adipose tissue. We found that, in Bhmt-KO mice, homocysteine-induced endoplasmic reticulum (ER) stress is associated with activation of the hepatic transcription factor cyclic AMP response element binding protein (CREBH), and an increase in hepatic and plasma concentrations of fibroblast growth factor 21 (FGF21), which is known to induce adipose browning. Our data indicate that the deletion of a single gene in one-carbon metabolism modifies adipose biology and energy metabolism. Future studies could focus on identifying if functional polymorphisms in BHMT result in a similar adipose atrophy phenotype.
Collapse
Affiliation(s)
- Manya Warrier
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA
| | - Evan M Paules
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA.,Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Jorge Silva-Gomez
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA
| | - Walter B Friday
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA
| | - Frances Bramlett
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA
| | - Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Isis Trujillo-Gonzalez
- Department of Nutrition, UNC Nutrition Research Institute, UNC-Chapel Hill, Kannapolis, NC, USA.,Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| |
Collapse
|
11
|
Beneficial effects of buspirone in endothelin-1 induced stroke cachexia in rats. Mol Cell Biochem 2023:10.1007/s11010-022-04653-4. [PMID: 36609633 DOI: 10.1007/s11010-022-04653-4] [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: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/09/2023]
Abstract
Stroke cachexia is associated with prolonged inflammation, muscle loss, poor prognosis, and early death of stroke patients. No particular treatment is available to cure the symptoms or disease. The present study aimed to evaluate the effect of a 5-HT1a agonist, buspirone on stroke cachexia. Wistar rats were injected with endothelin-1 to the bregma region of the brain to induce ischemic stroke followed by induction of cachexia after 4 days. Treatment with buspirone (3 mg/kg p.o) was given for 4 weeks after confirmation of cachexia in animals. Disease control animals exhibited decrease in wire hanging time and increase in foot fault numbers compared to normal animals. Disease control animals also showed weight loss, decrease in food intake, increased serum glucose and lipid profile along with high serum levels of inflammatory cytokines-TNF-α, IL-6 and decrease in weight of skeletal muscle and adipose tissues. Treatment with buspirone improves behavioural parameters along with increases food intake and body weight, decreased inflammatory cytokines IL-6 and TNF-α and serum glucose levels with increase in lipid profile. Buspirone also increased the weight of adipose tissue and maintain the skeletal muscle architecture and function as depicted in histopathological studies. Our study suggests that buspirone produces beneficial role in stroke cachexia by increasing body weight, food intake and adipose tissue depots by activating on 5-HT receptors. Buspirone decreases inflammatory markers in stroke cachexia although mechanism behind it was not fully understood. Buspirone decreases circulating blood glucose by stimulating glucose uptake in skeletal muscle via 5-HT receptors and maintained lipid profile. Buspirone was found to be effective in ameliorating cachectic conditions in stroke.
Collapse
|
12
|
Wang R, Wei L, Wazir J, Li L, Song S, Lin K, Pu W, Zhao C, Su Z, Zhao Q, Wang H. Curcumin treatment suppresses cachexia-associated adipose wasting in mice by blocking the cAMP/PKA/CREB signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154563. [PMID: 36610148 DOI: 10.1016/j.phymed.2022.154563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/24/2022] [Accepted: 11/19/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Cachexia is a multifactorial debilitating syndrome that is responsible for 22% of mortality among cancer patients, and there are no effective therapeutic agents available. Curcumin, a polyphenolic compound derived from the plant turmeric, has been shown to have anti-inflammatory, antioxidant, anti-autophagic, and antitumor activities. However, its function in cancer cachexia remains largely unexplored. PURPOSE This study aimed to elucidate the mechanisms by which curcumin improves adipose atrophy in cancer cachexia. METHODS C26 tumor-bearing BALB/c mice and β3-adrenoceptor agonist CL316243 stimulated BALB/c mice were used to observe the therapeutic effects of curcumin on the lipid degradation of cancer cachexia in vivo. The effects of curcumin in vitro were examined using mature 3T3-L1 adipocytes treated with a conditioned medium of C26 tumor cells or CL316243. RESULTS Mice with C26 tumors and cachexia were protected from weight loss and adipose atrophy by curcumin (50 mg/kg, i.g.). Curcumin significantly reduced serum levels of free fatty acids and increased triglyceride levels. In addition, curcumin significantly inhibited PKA and CREB activation in the adipose tissue of cancer cachectic mice. Curcumin also ameliorated CL316243-induced adipose atrophy and inhibited hormone-mediated PKA and CREB activation in mice. Moreover, the lipid droplet degradation induced by C26 tumor cell conditioned medium in mature 3T3-L1 adipocytes was ameliorated by curcumin (20 µM) treatment. Curcumin also improved the lipid droplet degradation of mature 3T3-L1 adipocytes induced by CL316243. CONCLUSION Curcumin might be expected to be a therapeutic supplement for cancer cachexia patients, primarily through inhibiting adipose tissue loss via the cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Ranran Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Kai Lin
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Zhonglan Su
- Department of Dermatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Quan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
13
|
Wang Y, An Z, Lin D, Jin W. Targeting cancer cachexia: Molecular mechanisms and clinical study. MedComm (Beijing) 2022; 3:e164. [PMID: 36105371 PMCID: PMC9464063 DOI: 10.1002/mco2.164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022] Open
Abstract
Cancer cachexia is a complex systemic catabolism syndrome characterized by muscle wasting. It affects multiple distant organs and their crosstalk with cancer constitute cancer cachexia environment. During the occurrence and progression of cancer cachexia, interactions of aberrant organs with cancer cells or other organs in a cancer cachexia environment initiate a cascade of stress reactions and destroy multiple organs including the liver, heart, pancreas, intestine, brain, bone, and spleen in metabolism, neural, and immune homeostasis. The role of involved organs turned from inhibiting tumor growth into promoting cancer cachexia in cancer progression. In this review, we depicted the complicated relationship of cancer cachexia with the metabolism, neural, and immune homeostasis imbalance in multiple organs in a cancer cachexia environment and summarized the treatment progress in recent years. And we discussed the molecular mechanism and clinical study of cancer cachexia from the perspective of multiple organs metabolic, neurological, and immunological abnormalities. Updated understanding of cancer cachexia might facilitate the exploration of biomarkers and novel therapeutic targets of cancer cachexia.
Collapse
Affiliation(s)
- Yong‐Fei Wang
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Zi‐Yi An
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Dong‐Hai Lin
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| | - Wei‐Lin Jin
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
14
|
Cheng R, Fujinaga M, Yang J, Rong J, Haider A, Ogasawara D, Van RS, Shao T, Chen Z, Zhang X, Calderon Leon ER, Zhang Y, Mori W, Kumata K, Yamasaki T, Xie L, Sun S, Wang L, Ran C, Shao Y, Cravatt B, Josephson L, Zhang MR, Liang SH. A novel monoacylglycerol lipase-targeted 18F-labeled probe for positron emission tomography imaging of brown adipose tissue in the energy network. Acta Pharmacol Sin 2022; 43:3002-3010. [PMID: 35513432 PMCID: PMC9622914 DOI: 10.1038/s41401-022-00912-8] [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: 01/10/2022] [Accepted: 04/13/2022] [Indexed: 11/09/2022] Open
Abstract
Monoacylglycerol lipase (MAGL) constitutes a serine hydrolase that orchestrates endocannabinoid homeostasis and exerts its function by catalyzing the degradation of 2-arachidonoylglycerol (2-AG) to arachidonic acid (AA). As such, selective inhibition of MAGL represents a potential therapeutic and diagnostic approach to various pathologies including neurodegenerative disorders, metabolic diseases and cancers. Based on a unique 4-piperidinyl azetidine diamide scaffold, we developed a reversible and peripheral-specific radiofluorinated MAGL PET ligand [18F]FEPAD. Pharmacokinetics and binding studies on [18F]FEPAD revealed its outstanding specificity and selectivity towards MAGL in brown adipose tissue (BAT) - a tissue that is known to be metabolically active. We employed [18F]FEPAD in PET studies to assess the abundancy of MAGL in BAT deposits of mice and found a remarkable degree of specific tracer binding in the BAT, which was confirmed by post-mortem tissue analysis. Given the negative regulation of endocannabinoids on the metabolic BAT activity, our study supports the concept that dysregulation of MAGL is likely linked to metabolic disorders. Further, we now provide a suitable imaging tool that allows non-invasive assessment of MAGL in BAT deposits, thereby paving the way for detailed mechanistic studies on the role of BAT in endocannabinoid system (ECS)-related pathologies.
Collapse
Affiliation(s)
- Ran Cheng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Jing Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02125, USA
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Daisuke Ogasawara
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Richard S Van
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Xiaofei Zhang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Erick R Calderon Leon
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Wakana Mori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Shaofa Sun
- Hubei Collaborative Innovation Centre for Non-power Nuclear Technology, College of Nuclear Technology & Chemistry and Biology, Hubei University of Science and Technology, Xianning, Hubei Province, 437100, China
| | - Lu Wang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02125, USA
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Benjamin Cravatt
- The Skaggs Institute for Chemical Biology and Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan.
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
15
|
Metabolic Advantage of 25(OH)D3 versus 1,25(OH)2D3 Supplementation in Infantile Nephropathic Cystinosis-Associated Adipose Tissue Browning and Muscle Wasting. Cells 2022; 11:cells11203264. [PMID: 36291130 PMCID: PMC9600749 DOI: 10.3390/cells11203264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 12/03/2022] Open
Abstract
Manifestations of infantile nephropathic cystinosis (INC) often include cachexia and deficiency of circulating vitamin D metabolites. We examined the impact of 25(OH)D3 versus 1,25(OH)2D3 repletion in Ctns null mice, a mouse model of INC. Six weeks of intraperitoneal administration of 25(OH)D3 (75 μg/kg/day) or 1,25(OH)2D3 (60 ng/kg/day) resulted in Ctns−/− mice corrected low circulating 25(OH)D3 or 1,25(OH)2D3 concentrations. While 25(OH)D3 administration in Ctns−/− mice normalized several metabolic parameters characteristic of cachexia as well as muscle function in vivo, 1,25(OH)2D3 did not. Administration of 25(OH)D3 in Ctns−/− mice increased muscle fiber size and decreased fat infiltration of skeletal muscle, which was accompanied by a reduction of abnormal muscle signaling pathways. 1,25(OH)2D3 administration was not as effective. In conclusion, 25(OH)D3 supplementation exerts metabolic advantages over 1,25(OH)2D3 supplementation by amelioration of muscle atrophy and fat browning in Ctns−/− mice.
Collapse
|
16
|
Huemer F, Hecht S, Scharinger B, Schlintl V, Rinnerthaler G, Schlick K, Heregger R, Melchardt T, Wimmer A, Mühlbacher I, Koch OO, Neureiter D, Klieser E, Seyedinia S, Beheshti M, Greil R, Weiss L. Body composition dynamics and impact on clinical outcome in gastric and gastro-esophageal junction cancer patients undergoing perioperative chemotherapy with the FLOT protocol. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04096-w. [PMID: 35864270 DOI: 10.1007/s00432-022-04096-w] [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: 04/03/2022] [Accepted: 05/27/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE Perioperative chemotherapy with FLOT constitutes a standard of care approach for locally advanced, resectable gastric or gastro-esophageal junction (GEJ) cancer. We aimed at investigating anthropometric, CT-based and FDG-PET-based body composition parameters and dynamics during this multidisciplinary approach and the impact on clinical outcomes. METHODS This retrospective, single-center study was based on medical records and (FDG-PET)-CT images among gastric/GEJ cancer patients undergoing perioperative FLOT chemotherapy. RESULTS Between 2016 and 2021, 46 gastric/GEJ cancer patients started perioperative FLOT at our tertiary cancer center (Salzburg, Austria). At a median follow-up of 32 months median PFS was 47.4 months and median OS was not reached. The skeletal muscle index (SMI, cm2/m2) turned out to be the only body composition parameter with a statistically significant decrease during pre-operative FLOT (51.3 versus 48.8 cm2/m2, p = 0.02). Neither pre-FLOT body mass index (BMI), nor SMI had an impact on the duration of pre-operative FLOT, the time interval from pre-operative FLOT initiation to surgery, the necessity of pre-operative or post-operative FLOT de-escalation or the likelihood of the start of postoperative chemotherapy. Pre-FLOT BMI (overweight versus normal, HR: 0.11, 95% CI: 0.02-0.65, p = 0.02) and pre-FLOT SMI (sarcopenia versus no sarcopenia, HR: 5.08, 95% CI: 1.27-20.31, p = 0.02) were statistically significantly associated with PFS in the multivariable analysis. CONCLUSION The statistically significant SMI loss during pre-operative FLOT and the meaningful impact of baseline SMI and BMI on PFS argue for the implementation of a nutritional screening and support program prior to the initiation of pre-operative FLOT in clinical routine.
Collapse
Affiliation(s)
- Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Stefan Hecht
- Department of Radiology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Bernhard Scharinger
- Department of Radiology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Verena Schlintl
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Gabriel Rinnerthaler
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Konstantin Schlick
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ronald Heregger
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Thomas Melchardt
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Angela Wimmer
- Department of Surgery, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Iris Mühlbacher
- Department of Surgery, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Oliver Owen Koch
- Department of Surgery, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Sara Seyedinia
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Lukas Weiss
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute , Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Paracelsus Medical University Salzburg, Salzburg, Austria.
- Cancer Cluster Salzburg, Salzburg, Austria.
| |
Collapse
|
17
|
Ngono Ayissi K, Gorwood J, Le Pelletier L, Bourgeois C, Beaupère C, Auclair M, Foresti R, Motterlini R, Atlan M, Barrail-Tran A, Le Grand R, Desjardins D, Fève B, Lambotte O, Capeau J, Béréziat V, Lagathu C. Inhibition of Adipose Tissue Beiging by HIV Integrase Inhibitors, Dolutegravir and Bictegravir, Is Associated with Adipocyte Hypertrophy, Hypoxia, Elevated Fibrosis, and Insulin Resistance in Simian Adipose Tissue and Human Adipocytes. Cells 2022; 11:cells11111841. [PMID: 35681536 PMCID: PMC9180037 DOI: 10.3390/cells11111841] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/23/2022] [Accepted: 06/02/2022] [Indexed: 01/13/2023] Open
Abstract
For people living with HIV, treatment with integrase-strand-transfer-inhibitors (INSTIs) can promote adipose tissue (AT) gain. We previously demonstrated that INSTIs can induce hypertrophy and fibrosis in AT of macaques and humans. By promoting energy expenditure, the emergence of beige adipocytes in white AT (beiging) could play an important role by limiting excess lipid storage and associated adipocyte dysfunction. We hypothesized that INSTIs could alter AT via beiging inhibition. Fibrosis and gene expression were measured in subcutaneous (SCAT) and visceral AT (VAT) from SIV-infected, dolutegravir-treated (SIVART) macaques. Beiging capacity was assessed in human adipose stromal cells (ASCs) undergoing differentiation and being exposed to dolutegravir, bictegravir, or raltegravir. Expression of beige markers, such as positive-regulatory-domain-containing-16 (PRDM16), were lower in AT of SIVART as compared to control macaques, whereas fibrosis-related genes were higher. Dolutegravir and bictegravir inhibited beige differentiation in ASCs, as shown by lower expression of beige markers and lower cell respiration. INSTIs also induced a hypertrophic insulin-resistant state associated with a pro-fibrotic phenotype. Our results indicate that adipocyte hypertrophy induced by INSTIs is involved via hypoxia (revealed by a greater hypoxia-inducible-factor-1-alpha gene expression) in fat fibrosis, beiging inhibition, and thus (via positive feedback), probably, further hypertrophy and associated insulin resistance.
Collapse
Affiliation(s)
- Kenza Ngono Ayissi
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Jennifer Gorwood
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Laura Le Pelletier
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Christine Bourgeois
- UMR1184 Inserm, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Saclay, 92032 Fontenay-aux-Roses, France; (C.B.); (A.B.-T.); (R.L.G.); (D.D.); (O.L.)
| | - Carine Beaupère
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Martine Auclair
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Roberta Foresti
- INSERM UMR_S955, IMRB, Université Paris-Est Créteil, 94000 Créteil, France; (R.F.); (R.M.)
| | - Roberto Motterlini
- INSERM UMR_S955, IMRB, Université Paris-Est Créteil, 94000 Créteil, France; (R.F.); (R.M.)
| | - Michael Atlan
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
- Service de Chirurgie Plastique et Esthétique, Hôpital Tenon, AP-HP, 75020 Paris, France
| | - Aurélie Barrail-Tran
- UMR1184 Inserm, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Saclay, 92032 Fontenay-aux-Roses, France; (C.B.); (A.B.-T.); (R.L.G.); (D.D.); (O.L.)
| | - Roger Le Grand
- UMR1184 Inserm, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Saclay, 92032 Fontenay-aux-Roses, France; (C.B.); (A.B.-T.); (R.L.G.); (D.D.); (O.L.)
| | - Delphine Desjardins
- UMR1184 Inserm, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Saclay, 92032 Fontenay-aux-Roses, France; (C.B.); (A.B.-T.); (R.L.G.); (D.D.); (O.L.)
| | - Bruno Fève
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
- Service d’Endocrinologie, Diabétologie et Reproduction, Hôpital Saint-Antoine, CRMR, PRISIS, AP-HP, 75012 Paris, France
| | - Olivier Lambotte
- UMR1184 Inserm, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, CEA, Université Paris Saclay, 92032 Fontenay-aux-Roses, France; (C.B.); (A.B.-T.); (R.L.G.); (D.D.); (O.L.)
- Service de Médecine Interne et Immunologie Clinique, Hôpital Bicêtre, AP-HP, 94270 Kremlin-Bicêtre, France
| | - Jacqueline Capeau
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
| | - Véronique Béréziat
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.)
| | - Claire Lagathu
- Inserm UMR_S938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-Métabolisme et Nutrition (ICAN), Sorbonne Université, 75012 Paris, France; (K.N.A.); (J.G.); (L.L.P.); (C.B.); (M.A.); (M.A.); (B.F.); (J.C.)
- Correspondence: (V.B.); (C.L.)
| |
Collapse
|
18
|
Divella R, Gadaleta Caldarola G, Mazzocca A. Chronic Inflammation in Obesity and Cancer Cachexia. J Clin Med 2022; 11:2191. [PMID: 35456284 PMCID: PMC9027625 DOI: 10.3390/jcm11082191] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation has long been linked to obesity and related conditions such as type 2 diabetes and metabolic syndrome. According to current research, the increased risk of cancer in people with certain metabolic diseases may be due to chronic inflammation. Adipocytokines, which are pro-inflammatory cytokines secreted in excess, are elevated in many chronic metabolic diseases. Cytokines and inflammatory mediators, which are not directly linked to DNA, are important in tumorigenesis. Cachexia, a type of metabolic syndrome linked to the disease, is associated with a dysregulation of metabolic pathways. Obesity and cachexia have distinct metabolic characteristics, such as insulin resistance, increased lipolysis, elevated free fatty acids (FFA), and ceramide levels, which are discussed in this section. The goal of this research project is to create a framework for bringing together our knowledge of inflammation-mediated insulin resistance.
Collapse
Affiliation(s)
- Rosa Divella
- ASD Nordic Walking Apulia Lifestyle, Corso Giuseppe Di Vittorio 14, 70024 Gravina in Puglia, Italy
| | | | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Piazza G. Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
19
|
Mak RH, Querfeld U, Gonzalez A, Gunta S, Cheung WW. Differential Effects of 25-Hydroxyvitamin D 3 versus 1α 25-Dihydroxyvitamin D 3 on Adipose Tissue Browning in CKD-Associated Cachexia. Cells 2021; 10:3382. [PMID: 34943890 PMCID: PMC8699879 DOI: 10.3390/cells10123382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with chronic kidney disease (CKD) often have low serum concentrations of 25(OH)D3 and 1,25(OH)2D3. We investigated the differential effects of 25(OH)D3 versus 1,25(OH)2D3 repletion in mice with surgically induced CKD. Intraperitoneal supplementation of 25(OH)D3 (75 μg/kg/day) or 1,25(OH)2D3 (60 ng/kg/day) for 6 weeks normalized serum 25(OH)D3 or 1,25(OH)2D3 concentrations in CKD mice, respectively. Repletion of 25(OH)D3 normalized appetite, significantly improved weight gain, increased fat and lean mass content and in vivo muscle function, as well as attenuated elevated resting metabolic rate relative to repletion of 1,25(OH)2D3 in CKD mice. Repletion of 25(OH)D3 in CKD mice attenuated adipose tissue browning as well as ameliorated perturbations of energy homeostasis in adipose tissue and skeletal muscle, whereas repletion of 1,25(OH)2D3 did not. Significant improvement of muscle fiber size and normalization of fat infiltration of gastrocnemius was apparent with repletion of 25(OH)D3 but not with 1,25(OH)2D3 in CKD mice. This was accompanied by attenuation of the aberrant gene expression of muscle mass regulatory signaling, molecular pathways related to muscle fibrosis as well as muscle expression profile associated with skeletal muscle wasting in CKD mice. Our findings provide evidence that repletion of 25(OH)D3 exerts metabolic advantages over repletion of 1,25(OH)2D3 by attenuating adipose tissue browning and muscle wasting in CKD mice.
Collapse
Affiliation(s)
- Robert H. Mak
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| | - Uwe Querfeld
- Department of Paediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Alex Gonzalez
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| | - Sujana Gunta
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
- Pediatric Services, Vista Community Clinic, Vista, CA 92084, USA
| | - Wai W. Cheung
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093, USA; (A.G.); (S.G.); (W.W.C.)
| |
Collapse
|
20
|
Towards Drug Repurposing in Cancer Cachexia: Potential Targets and Candidates. Pharmaceuticals (Basel) 2021; 14:ph14111084. [PMID: 34832866 PMCID: PMC8618795 DOI: 10.3390/ph14111084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
As a multifactorial and multiorgan syndrome, cancer cachexia is associated with decreased tolerance to antitumor treatments and increased morbidity and mortality rates. The current approaches for the treatment of this syndrome are not always effective and well established. Drug repurposing or repositioning consists of the investigation of pharmacological components that are already available or in clinical trials for certain diseases and explores if they can be used for new indications. Its advantages comparing to de novo drugs development are the reduced amount of time spent and costs. In this paper, we selected drugs already available or in clinical trials for non-cachexia indications and that are related to the pathways and molecular components involved in the different phenotypes of cancer cachexia syndrome. Thus, we introduce known drugs as possible candidates for drug repurposing in the treatment of cancer-induced cachexia.
Collapse
|
21
|
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
|
22
|
Mcclement S. Adipose Tissue and Cancer Cachexia: What Nurses Need to Know. Asia Pac J Oncol Nurs 2021; 8:445-449. [PMID: 34527774 PMCID: PMC8420924 DOI: 10.4103/apjon.apjon-2134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/12/2022] Open
Abstract
The purpose of this article is to discuss the different types of adipose tissue involved in cachexia and describe their role in contributing to increased energy expenditure and negative energy balance. Armed with this knowledge, nurses will be better positioned to understand the clinical picture of cachexia, appreciate the rationale for proposed therapeutic interventions, and confidently dialogue with patients, families, and members of interdisciplinary health care teams about this prevalent condition.
Collapse
Affiliation(s)
- Susan Mcclement
- College of Nursing, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
23
|
Han J, Tang M, Lu C, Shen L, She J, Wu G. Subcutaneous, but not visceral, adipose tissue as a marker for prognosis in gastric cancer patients with cachexia. Clin Nutr 2021; 40:5156-5161. [PMID: 34461589 DOI: 10.1016/j.clnu.2021.08.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Adipose tissue loss is one of the features in patients with cancer cachexia. However, whether subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) contribute differently to the progress of cancer cachexia in gastric cancer patients with cachexia remains unclear. This study aim to investigate the effect of SAT and VAT in gastric cancer patients with cachexia. METHODS Gastric cancer patients who underwent surgery were divided into cancer cachexia group and non-cachexia group. A new deep learning system was developed to segment SAT and VAT from the computed tomography images at the third lumbar vertebra. Indexes of SAT (SATI) and VAT (VATI) were compared between cachexia and non-cachexia groups. The prognostic values of SATI and VATI for patients with gastric cancer cachexia were analyzed by Kaplan-Meier method and Cox regression. RESULTS A total of 1627 gastric cancer patients (411 cachexia and 1216 non-cachexia) were included in this study. A new V-Net-Based segmentation deep learning system was developed to quickly (0.02 s/image) and accurately segment SAT (dice scores = 0.96) and VAT (dice scores = 0.98). The SATI of gastric cancer patients with cachexia were significantly lower than non-cachexia patients (44.91 ± 0.90 vs. 50.92 ± 0.71 cm2/m2, P < 0.001), whereas no significant difference was detected in VATI (35.98 ± 0.84 VS. 37.90 ± 0.45 cm2/m2, P = 0.076). Cachexia patients with low SATI showed poor survival than those with high SATI (HR = 1.35; 95% CI = 1.06-1.74). In contrast, VATI did not show close correlation with survival in patients with cachexia (HR = 1.18; 95% CI = 0.92-1.51). CONCLUSION SAT and VAT showed different effects on gastric cancer patients with cachexia. More attention should be paid to the loss of SAT during the progress of cancer cachexia.
Collapse
Affiliation(s)
- Jun Han
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Center, Shanghai, China
| | - Min Tang
- Department of Radiology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Chaocheng Lu
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Lei Shen
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jiaqi She
- Department of Radiology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Center, Shanghai, China.
| |
Collapse
|
24
|
Gonzalez A, Cheung WW, Perens EA, Oliveira EA, Gertler A, Mak RH. A Leptin Receptor Antagonist Attenuates Adipose Tissue Browning and Muscle Wasting in Infantile Nephropathic Cystinosis-Associated Cachexia. Cells 2021; 10:1954. [PMID: 34440723 PMCID: PMC8393983 DOI: 10.3390/cells10081954] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mice lacking the functional cystinosin gene (Ctns-/-), a model of infantile nephropathic cystinosis (INC), exhibit the cachexia phenotype with adipose tissue browning and muscle wasting. Elevated leptin signaling is an important cause of chronic kidney disease-associated cachexia. The pegylated leptin receptor antagonist (PLA) binds to but does not activate the leptin receptor. We tested the efficacy of this PLA in Ctns-/- mice. We treated 12-month-old Ctns-/- mice and control mice with PLA (7 mg/kg/day, IP) or saline as a vehicle for 28 days. PLA normalized food intake and weight gain, increased fat and lean mass, decreased metabolic rate and improved muscle function. It also attenuated perturbations of energy homeostasis in adipose tissue and muscle in Ctns-/- mice. PLA attenuated adipose tissue browning in Ctns-/- mice. PLA increased gastrocnemius weight and fiber size as well as attenuated muscle fat infiltration in Ctns-/- mice. This was accompanied by correcting the increased expression of muscle wasting signaling while promoting the decreased expression of myogenesis in gastrocnemius of Ctns-/- mice. PLA attenuated aberrant expressed muscle genes that have been associated with muscle atrophy, increased energy expenditure and lipolysis in Ctns-/- mice. Leptin antagonism may represent a viable therapeutic strategy for adipose tissue browning and muscle wasting in INC.
Collapse
MESH Headings
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Amino Acid Transport Systems, Neutral/genetics
- Amino Acid Transport Systems, Neutral/metabolism
- Animals
- Body Composition/drug effects
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Cachexia/prevention & control
- Cystinosis/complications
- Cystinosis/drug therapy
- Cystinosis/metabolism
- Cystinosis/pathology
- Disease Models, Animal
- Hormone Antagonists/pharmacology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/etiology
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Atrophy/prevention & control
- Receptors, Leptin/antagonists & inhibitors
- Receptors, Leptin/metabolism
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Alex Gonzalez
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Wai W. Cheung
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Elliot A. Perens
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| | - Eduardo A. Oliveira
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
- Health Sciences Postgraduate Program, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte 30130-100, MG, Brazil
| | - Arieh Gertler
- Institute of Biochemistry, Food Science and Nutrition, Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Robert H. Mak
- Division of Pediatric Nephrology, Rady Children’s Hospital, University of California, San Diego, CA 92093-0831, USA; (A.G.); (W.W.C.); (E.A.P.); (E.A.O.)
| |
Collapse
|
25
|
Siff T, Parajuli P, Razzaque MS, Atfi A. Cancer-Mediated Muscle Cachexia: Etiology and Clinical Management. Trends Endocrinol Metab 2021; 32:382-402. [PMID: 33888422 PMCID: PMC8102392 DOI: 10.1016/j.tem.2021.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
Muscle cachexia has a major detrimental impact on cancer patients, being responsible for 30% of all cancer deaths. It is characterized by a debilitating loss in muscle mass and function, which ultimately deteriorates patients' quality of life and dampens therapeutic treatment efficacy. Muscle cachexia stems from widespread alterations in whole-body metabolism as well as immunity and neuroendocrine functions and these global defects often culminate in aberrant signaling within skeletal muscle, causing muscle protein breakdown and attendant muscle atrophy. This review summarizes recent landmark discoveries that significantly enhance our understanding of the molecular etiology of cancer-driven muscle cachexia and further discuss emerging therapeutic approaches seeking to simultaneously target those newly discovered mechanisms to efficiently curb this lethal syndrome.
Collapse
Affiliation(s)
- Thomas Siff
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Parash Parajuli
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Azeddine Atfi
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA; Sorbonne Universités, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France.
| |
Collapse
|
26
|
Álvarez-Artime A, García-Soler B, Sainz RM, Mayo JC. Emerging Roles for Browning of White Adipose Tissue in Prostate Cancer Malignant Behaviour. Int J Mol Sci 2021; 22:5560. [PMID: 34074045 PMCID: PMC8197327 DOI: 10.3390/ijms22115560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
In addition to its well-known role as an energy repository, adipose tissue is one of the largest endocrine organs in the organism due to its ability to synthesize and release different bioactive molecules. Two main types of adipose tissue have been described, namely white adipose tissue (WAT) with a classical energy storage function, and brown adipose tissue (BAT) with thermogenic activity. The prostate, an exocrine gland present in the reproductive system of most mammals, is surrounded by periprostatic adipose tissue (PPAT) that contributes to maintaining glandular homeostasis in conjunction with other cell types of the microenvironment. In pathological conditions such as the development and progression of prostate cancer, adipose tissue plays a key role through paracrine and endocrine signaling. In this context, the role of WAT has been thoroughly studied. However, the influence of BAT on prostate tumor development and progression is unclear and has received much less attention. This review tries to bring an update on the role of different factors released by WAT which may participate in the initiation, progression and metastasis, as well as to compile the available information on BAT to discuss and open a new field of knowledge about the possible protective role of BAT in prostate cancer.
Collapse
Affiliation(s)
- Alejandro Álvarez-Artime
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Belén García-Soler
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
| | - Rosa María Sainz
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| | - Juan Carlos Mayo
- Departamento de Morfología y Biología Celular, Redox Biology Unit, University of Oviedo, Facultad de Medicina, Julián Clavería 6, 33006 Oviedo, Spain; (A.Á.-A.); (B.G.-S.); (R.M.S.)
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Santiago Gascón Building, Fernando Bongera s/n, 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario s/n, 33011 Oviedo, Spain
| |
Collapse
|
27
|
Narasimhan A, Zhong X, Au EP, Ceppa EP, Nakeeb A, House MG, Zyromski NJ, Schmidt CM, Schloss KNH, Schloss DEI, Liu Y, Jiang G, Hancock BA, Radovich M, Kays JK, Shahda S, Couch ME, Koniaris LG, Zimmers TA. Profiling of Adipose and Skeletal Muscle in Human Pancreatic Cancer Cachexia Reveals Distinct Gene Profiles with Convergent Pathways. Cancers (Basel) 2021; 13:1975. [PMID: 33923976 PMCID: PMC8073275 DOI: 10.3390/cancers13081975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 01/06/2023] Open
Abstract
The vast majority of patients with pancreatic ductal adenocarcinoma (PDAC) suffer cachexia. Although cachexia results from concurrent loss of adipose and muscle tissue, most studies focus on muscle alone. Emerging data demonstrate the prognostic value of fat loss in cachexia. Here we sought to identify the muscle and adipose gene profiles and pathways regulated in cachexia. Matched rectus abdominis muscle and subcutaneous adipose tissue were obtained at surgery from patients with benign conditions (n = 11) and patients with PDAC (n = 24). Self-reported weight loss and body composition measurements defined cachexia status. Gene profiling was done using ion proton sequencing. Results were queried against external datasets for validation. 961 DE genes were identified from muscle and 2000 from adipose tissue, demonstrating greater response of adipose than muscle. In addition to known cachexia genes such as FOXO1, novel genes from muscle, including PPP1R8 and AEN correlated with cancer weight loss. All the adipose correlated genes including SCGN and EDR17 are novel for PDAC cachexia. Pathway analysis demonstrated shared pathways but largely non-overlapping genes in both tissues. Age related muscle loss predominantly had a distinct gene profiles compared to cachexia. This analysis of matched, externally validate gene expression points to novel targets in cachexia.
Collapse
Affiliation(s)
- Ashok Narasimhan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Xiaoling Zhong
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
| | - Ernie P. Au
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eugene P. Ceppa
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Atilla Nakeeb
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Michael G. House
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Nicholas J. Zyromski
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - C. Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Katheryn N. H. Schloss
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Daniel E. I. Schloss
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Yunlong Liu
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Bradley A. Hancock
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Milan Radovich
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Joshua K. Kays
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Safi Shahda
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marion E. Couch
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Otolaryngology—Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leonidas G. Koniaris
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
- Department of Otolaryngology—Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
28
|
Thermogenic adipocytes: lineage, function and therapeutic potential. Biochem J 2020; 477:2071-2093. [PMID: 32539124 PMCID: PMC7293110 DOI: 10.1042/bcj20200298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Metabolic inflexibility, defined as the inability to respond or adapt to metabolic demand, is now recognised as a driving factor behind many pathologies associated with obesity and the metabolic syndrome. Adipose tissue plays a pivotal role in the ability of an organism to sense, adapt to and counteract environmental changes. It provides a buffer in times of nutrient excess, a fuel reserve during starvation and the ability to resist cold-stress through non-shivering thermogenesis. Recent advances in single-cell RNA sequencing combined with lineage tracing, transcriptomic and proteomic analyses have identified novel adipocyte progenitors that give rise to specialised adipocytes with diverse functions, some of which have the potential to be exploited therapeutically. This review will highlight the common and distinct functions of well-known adipocyte populations with respect to their lineage and plasticity, as well as introducing the most recent members of the adipocyte family and their roles in whole organism energy homeostasis. Finally, this article will outline some of the more preliminary findings from large data sets generated by single-cell transcriptomics of mouse and human adipose tissue and their implications for the field, both for discovery and for therapy.
Collapse
|
29
|
Trivanović D, Vignjević Petrinović S, Okić Djordjević I, Kukolj T, Bugarski D, Jauković A. Adipogenesis in Different Body Depots and Tumor Development. Front Cell Dev Biol 2020; 8:571648. [PMID: 33072753 PMCID: PMC7536553 DOI: 10.3389/fcell.2020.571648] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue (AT) forms depots at different anatomical locations throughout the body, being in subcutaneous and visceral regions, as well as the bone marrow. These ATs differ in the adipocyte functional profile, their insulin sensitivity, adipokines’ production, lipolysis, and response to pathologic conditions. Despite the recent advances in lineage tracing, which have demonstrated that individual adipose depots are composed of adipocytes derived from distinct progenitor populations, the cellular and molecular dissection of the adipose clonogenic stem cell niche is still a great challenge. Additional complexity in AT regulation is associated with tumor-induced changes that affect adipocyte phenotype. As an integrative unit of cell differentiation, AT microenvironment regulates various phenotype outcomes of differentiating adipogenic lineages, which consequently may contribute to the neoplastic phenotype manifestations. Particularly interesting is the capacity of AT to impose and support the aberrant potency of stem cells that accompanies tumor development. In this review, we summarize the current findings on the communication between adipocytes and their progenitors with tumor cells, pointing out to the co-existence of healthy and neoplastic stem cell niches developed during tumor evolution. We also discuss tumor-induced adaptations in mature adipocytes and the involvement of alternative differentiation programs.
Collapse
Affiliation(s)
- Drenka Trivanović
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Clinics, Wuerzburg, Germany.,Bernhard-Heine Center for Locomotion Research, University of Wuerzburg, Wuerzburg, Germany
| | - Sanja Vignjević Petrinović
- Laboratory for Neuroendocrinology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Ivana Okić Djordjević
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Tamara Kukolj
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
30
|
The Extract of Arctium lappa L. Fruit (Arctii Fructus) Improves Cancer-Induced Cachexia by Inhibiting Weight Loss of Skeletal Muscle and Adipose Tissue. Nutrients 2020; 12:nu12103195. [PMID: 33086629 PMCID: PMC7603378 DOI: 10.3390/nu12103195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Cachexia induced by cancer is a systemic wasting syndrome and it accompanies continuous body weight loss with the exhaustion of skeletal muscle and adipose tissue. Cancer cachexia is not only a problem in itself, but it also reduces the effectiveness of treatments and deteriorates quality of life. However, effective treatments have not been found yet. Although Arctii Fructus (AF) has been studied about several pharmacological effects, there were no reports on its use in cancer cachexia. Methods: To induce cancer cachexia in mice, we inoculated CT-26 cells to BALB/c mice through subcutaneous injection and intraperitoneal injection. To mimic cancer cachexia in vitro, we used conditioned media (CM), which was CT-26 colon cancer cells cultured medium. Results: In in vivo experiments, AF suppressed expression of interleukin (IL)-6 and atrophy of skeletal muscle and adipose tissue. As a result, the administration of AF decreased mortality by preventing weight loss. In adipose tissue, AF decreased expression of uncoupling protein 1 (UCP1) by restoring AMP-activated protein kinase (AMPK) activation. In in vitro model, CM increased muscle degradation factors and decreased adipocytes differentiation factors. However, these tendencies were ameliorated by AF treatment in C2C12 myoblasts and 3T3-L1 cells. Conclusion: Taken together, our study demonstrated that AF could be a therapeutic supplement for patients suffering from cancer cachexia.
Collapse
|
31
|
Becker AS, Zellweger C, Bacanovic S, Franckenberg S, Nagel HW, Frick L, Schawkat K, Eberhard M, Blüthgen C, Volbracht J, Moos R, Wolfrum C, Burger IA. Brown fat does not cause cachexia in cancer patients: A large retrospective longitudinal FDG-PET/CT cohort study. PLoS One 2020; 15:e0239990. [PMID: 33031379 PMCID: PMC7544086 DOI: 10.1371/journal.pone.0239990] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background Brown adipose tissue (BAT) is a specialized form of adipose tissue, able to increase energy expenditure by heat generation in response to various stimuli. Recently, its pathological activation has been implicated in the pathogenesis of cancer cachexia. To establish a causal relationship, we retrospectively investigated the longitudinal changes in BAT and cancer in a large FDG-PET/CT cohort. Methods We retrospectively analyzed 13 461 FDG-PET/CT examinations of n = 8 409 patients at our institution from the winter months of 2007–2015. We graded the activation strength of BAT based on the anatomical location of the most caudally activated BAT depot into three tiers, and the stage of the cancer into five general grades. We validated the cancer grading by an interreader analysis and correlation with histopathological stage. Ambient temperature data (seven-day average before the examination) was obtained from a meteorological station close to the hospital. Changes of BAT, cancer, body mass index (BMI) and temperature between the different examinations were examined with Spearman’s test and a mixed linear model for correlation, and with a causal inference algorithm for causality. Results We found n = 283 patients with at least two examinations and active BAT in at least one of them. There was no significant interaction between the changes in BAT activation, cancer burden or BMI. Temperature changes exhibited a strong negative correlation with BAT activity (ϱ = -0.57, p<0.00001). These results were confirmed with the mixed linear model. Causal inference revealed a link of Temperature ➜ BAT in all subjects and also of BMI ➜ BAT in subjects who had lost weight and increased cancer burden, but no role of cancer and no causal links of BAT ➜ BMI. Conclusions Our data did not confirm the hypothesis that BAT plays a major role in cancer-mediated weight loss. Temperature changes are the main driver of incidental BAT activity on FDG-PET scans.
Collapse
Affiliation(s)
- Anton S. Becker
- Department of Health Sciences and Technology; Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- * E-mail:
| | - Caroline Zellweger
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Sara Bacanovic
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Sabine Franckenberg
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Hannes W. Nagel
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Lukas Frick
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Khoschy Schawkat
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Matthias Eberhard
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Christian Blüthgen
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Jörk Volbracht
- Division of Controlling and Data Management, University Hospital Zurich, Zurich, Switzerland
| | - Rudolf Moos
- Division of Controlling and Data Management, University Hospital Zurich, Zurich, Switzerland
| | - Christian Wolfrum
- Department of Health Sciences and Technology; Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Ose J, Holowatyj AN, Nattenmüller J, Gigic B, Lin T, Himbert C, Habermann N, Achaintre D, Scalbert A, Keski-Rahkonen P, Böhm J, Schrotz-King P, Schneider M, Ulrich A, Kampman E, Weijenberg M, Gsur A, Ueland PM, Kauczor HU, Ulrich CM. Metabolomics profiling of visceral and abdominal subcutaneous adipose tissue in colorectal cancer patients: results from the ColoCare study. Cancer Causes Control 2020; 31:723-735. [PMID: 32430684 PMCID: PMC7425810 DOI: 10.1007/s10552-020-01312-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 05/04/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Underlying mechanisms of the relationship between body fatness and colorectal cancer remain unclear. This study investigated associations of circulating metabolites with visceral (VFA), abdominal subcutaneous (SFA), and total fat area (TFA) in colorectal cancer patients. METHODS Pre-surgery plasma samples from 212 patients (stage I-IV) from the ColoCare Study were used to perform targeted metabolomics. VFA, SFA, and TFA were quantified by computed tomography scans. Partial correlation and linear regression analyses of VFA, SFA, and TFA with metabolites were computed and corrected for multiple testing. Cox proportional hazards were used to assess 2-year survival. RESULTS In patients with metastatic tumors, SFA and TFA were statistically significantly inversely associated with 16 glycerophospholipids (SFA: pFDR range 0.017-0.049; TFA: pFDR range 0.029-0.048), while VFA was not. Doubling of ten of the aforementioned glycerophospholipids was associated with increased risk of death in patients with metastatic tumors, but not in patients with non-metastatic tumors (phet range: 0.00044-0.049). Doubling of PC ae C34:0 was associated with ninefold increased risk of death in metastatic tumors (Hazard Ratio [HR], 9.05; 95% confidence interval [CI] 2.17-37.80); an inverse association was observed in non-metastatic tumors (HR 0.17; 95% CI 0.04-0.87; phet = 0.00044). CONCLUSION These data provide initial evidence that glycerophospholipids in metastatic colorectal cancer are uniquely associated with subcutaneous adiposity, and may impact overall survival.
Collapse
Affiliation(s)
- Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, UT, USA.
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Andreana N Holowatyj
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Johanna Nattenmüller
- Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Tengda Lin
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Caroline Himbert
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Nina Habermann
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - David Achaintre
- International Agency Research on Cancer (IARC), Lyon, France
| | | | | | - Jürgen Böhm
- Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Petra Schrotz-King
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Ellen Kampman
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Matty Weijenberg
- Department of Epidemiology, GROW - School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Andrea Gsur
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | | | - Hans-Ulrich Kauczor
- Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT, USA.
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
33
|
Peixoto da Silva S, Santos JMO, Costa E Silva MP, Gil da Costa RM, Medeiros R. Cancer cachexia and its pathophysiology: links with sarcopenia, anorexia and asthenia. J Cachexia Sarcopenia Muscle 2020; 11:619-635. [PMID: 32142217 PMCID: PMC7296264 DOI: 10.1002/jcsm.12528] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass, along with adipose tissue wasting, systemic inflammation and other metabolic abnormalities leading to functional impairment. Cancer cachexia has long been recognized as a direct cause of complications in cancer patients, reducing quality of life and worsening disease outcomes. Some related conditions, like sarcopenia (age-related muscle wasting), anorexia (appetite loss) and asthenia (reduced muscular strength and fatigue), share some key features with cancer cachexia, such as weakness and systemic inflammation. Understanding the interplay and the differences between these conditions is critical to advance basic and translational research in this field, improving the accuracy of diagnosis and contributing to finally achieve effective therapies for affected patients.
Collapse
Affiliation(s)
- Sara Peixoto da Silva
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Joana M O Santos
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Maria Paula Costa E Silva
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.,Palliative Care Service, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Rui M Gil da Costa
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.,Postgraduate Programme in Adult Health (PPGSAD) and Tumour Biobank, Federal University of Maranhão (UFMA), São Luís, Brazil
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.,Virology Service, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, Porto, Portugal.,Research Department, Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), Porto, Portugal
| |
Collapse
|
34
|
Poulia KA, Sarantis P, Antoniadou D, Koustas E, Papadimitropoulou A, Papavassiliou AG, Karamouzis MV. Pancreatic Cancer and Cachexia-Metabolic Mechanisms and Novel Insights. Nutrients 2020; 12:E1543. [PMID: 32466362 PMCID: PMC7352917 DOI: 10.3390/nu12061543] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/15/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a major characteristic of multiple non-malignant diseases, advanced and metastatic cancers and it is highly prevalent in pancreatic cancer, affecting almost 70-80% of the patients. Cancer cachexia is a multifactorial condition accompanied by compromised appetite and changes in body composition, i.e., loss of fat. It is associated with lower effectiveness of treatment, compromised quality of life, and higher mortality. Understanding the complex pathways underlying the pathophysiology of cancer cachexia, new therapeutic targets will be unraveled. The interplay between tumor and host factors, such as cytokines, holds a central role in cachexia pathophysiology. Cytokines are possibly responsible for anorexia, hypermetabolism, muscle proteolysis, and apoptosis. In particular, cachexia in pancreatic cancer might be the result of the surgical removal of pancreas parts. In recent years, many studies have been carried out to identify an effective treatment algorithm for cachexia. Choosing the most appropriate treatment, the clinical effect and the risk of adverse effects should be taken under consideration. The purpose of this review is to highlight the pathophysiological mechanisms as well as the current ways of cachexia treatment in the pharmaceutical and the nutrition field.
Collapse
Affiliation(s)
- Kalliopi Anna Poulia
- Department of Nutrition and Dietetics, Laiko General Hospital, 11527 Athens, Greece;
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.); (A.G.P.)
| | - Dimitra Antoniadou
- Oncology Department of Daily Hospitality, Laiko General Hospital, 11527 Athens, Greece;
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.); (A.G.P.)
| | - Adriana Papadimitropoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.); (A.G.P.)
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.); (A.G.P.)
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
35
|
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: 9.4] [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
|
36
|
Han J, Lu C, Meng Q, Halim A, Yean TJ, Wu G. Plasma concentration of interleukin-6 was upregulated in cancer cachexia patients and was positively correlated with plasma free fatty acid in female patients. Nutr Metab (Lond) 2019; 16:80. [PMID: 31788012 PMCID: PMC6858650 DOI: 10.1186/s12986-019-0409-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/06/2019] [Indexed: 01/05/2023] Open
Abstract
Background Cancer cachexia is a clinical manifestation in various advanced cancers that characterized by muscle atrophy and fat loss as its main features; it is frequently associated with systemic inflammatory response. However, the differences in inflammatory response and lipid metabolism of different genders remain unclear. This study explores the difference between cachexic and non-cachexic patients in different genders and cancer types and focus on the plasma inflammation factors levels and lipid metabolism parameters in different genders. Methods We first analyzed the general characteristics in 311 cancer patients between cachexic and non-cachexic patients, with an emphasis on expression levels related to inflammatory factors and lipid metabolism parameters. We then further analyzed these characteristics in different genders and cancer types. Lastly, the correlations between plasma interleukin-6 (IL-6) and lipid metabolism parameters in cachexia patients of different genders were analyzed. Results Among 311 patients, there were 74 cancer cachexia patients (50 males and 24 females) and 237non-cachexia patients (150 males and 87 females). Body mass index (BMI), TNM stage, plasma concentration of hemoglobin, platelet, lymphocyte count, total protein, albumin, prealbumin, total cholesterol, apolipoprotein E (ApoE), free fatty acid (FFA) and IL-6 were significantly different between cachexic and non-cachexic patients (all p < 0.05). In addition, these characteristics were different in different cancer types. When compared to male non-cachexic patients, male cachexic patients showed a significant increase in plasma levels of IL-6 and platelet, later TNM stage, with marked decrease in their plasma total protein, albumin, prealbumin, ApoE as well as their lymphocyte counts and hemoglobin levels (all p < 0.05). In comparison with female non-cachexic patients, female cachexic patients' IL-6 levels and FFA were significantly elevated with noticeable decrease in their BMI, total cholesterol, ApoE and prealbumin, as well as later TNM stage (all p < 0.05). Correlation analysis revealed that IL-6 levels in female cachexic patients had a significant positive correlation with FFA expression, but this correlation not reflected in male patients. Conclusion This study demonstrates the different metabolic characteristics of male and female cancer cachexia patients. Future study about cancer cachexia should pay attention to different genders and cancer types.
Collapse
Affiliation(s)
- Jun Han
- 1Department of General Surgery, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Chaocheng Lu
- 1Department of General Surgery, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Qingyang Meng
- 1Department of General Surgery, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| | - Alice Halim
- 2Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 China
| | - Thong Jia Yean
- 2Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai, 200032 China
| | - Guohao Wu
- 1Department of General Surgery, Zhongshan Hospital of Fudan University, 180 Fenglin Road, Shanghai, 200032 China
| |
Collapse
|
37
|
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: 3.5] [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
|
38
|
Liu H, Li L, Zou J, Zhou T, Wang B, Sun H, Yu S. Coix seed oil ameliorates cancer cachexia by counteracting muscle loss and fat lipolysis. Altern Ther Health Med 2019; 19:267. [PMID: 31615487 PMCID: PMC6792186 DOI: 10.1186/s12906-019-2684-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 09/12/2019] [Indexed: 01/06/2023]
Abstract
Background Cancer cachexia is a cancer-induced multifactorial debilitating syndrome directly accounting for 20% of cancer deaths without effective therapeutic approaches. It is extremely urgent to explore effective anti-cachexia drugs to ameliorate muscle and fat loss in cachexia patients. Methods Lewis lung carcinoma bearing C57BL/6 mice were applied as the animal model to examine the therapeutic effect of Coix seed oil (CSO) on cancer cachexia. The food intake and body weight change were monitored every 3 days throughout the experiment. The IL-6 and TNF-α levels in serum were detected by ELISA assay. Several key proteins involved in muscle wasting and fat lipolysis were tested by Western blot to identify the potential mechanism of CSO. Results Administration of CSO through gavage significantly prevented body weight loss and ameliorated systemic inflammation without affecting food intake and tumor size. The weight and histological morphology of gastrocnemius muscle and epididymal adipose tissue in CSO-treated mice were also improved. In mechanism, we found that CSO decreased the expression of MuRF1 and the ratio of phospho-p65 (Ser536) to p65 in muscle tissue. Meanwhile, cancer-induced activation of HSL and AMPK was also inhibited by CSO administration. Conclusion Coix seed oil exerts an anti-cachexia pharmaceutical effect by counteracting muscle and adipose tissue loss most likely through regulating NF-κB-MuRF1 and AMPK-HSL pathway.
Collapse
|
39
|
Lima JD, Simoes E, de Castro G, Morais MRP, de Matos‐Neto EM, Alves MJ, Pinto NI, Figueredo RG, Zorn TM, Felipe‐Silva AS, Tokeshi F, Otoch JP, Alcantara P, Cabral FJ, Ferro ES, Laviano A, Seelaender M. Tumour-derived transforming growth factor-β signalling contributes to fibrosis in patients with cancer cachexia. J Cachexia Sarcopenia Muscle 2019; 10:1045-1059. [PMID: 31273954 PMCID: PMC6818454 DOI: 10.1002/jcsm.12441] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 04/05/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cachexia is a paraneoplastic syndrome related with poor prognosis. The tumour micro-environment contributes to systemic inflammation and increased oxidative stress as well as to fibrosis. The aim of the present study was to characterise the inflammatory circulating factors and tumour micro-environment profile, as potentially contributing to tumour fibrosis in cachectic cancer patients. METHODS 74 patients (weight stable cancer n = 31; cachectic cancer n = 43) diagnosed with colorectal cancer were recruited, and tumour biopsies were collected during surgery. Multiplex assay was performed to study inflammatory cytokines and growth factors. Immunohistochemistry analysis was carried out to study extracellular matrix components. RESULTS Higher protein expression of inflammatory cytokines and growth factors such as epidermal growth factor, granulocyte-macrophage colony-stimulating factor, interferon-α, and interleukin (IL)-8 was observed in the tumour and serum of cachectic cancer patients in comparison with weight-stable counterparts. Also, IL-8 was positively correlated with weight loss in cachectic patients (P = 0.04; r = 0.627). Immunohistochemistry staining showed intense collagen deposition (P = 0.0006) and increased presence of α-smooth muscle actin (P < 0.0001) in tumours of cachectic cancer patients, characterizing fibrosis. In addition, higher transforming growth factor (TGF)-β1, TGF-β2, and TGF-β3 expression (P = 0.003, P = 0.05, and P = 0.047, respectively) was found in the tumour of cachectic patients, parallel to p38 mitogen-activated protein kinase alteration. Hypoxia-inducible factor-1α mRNA content was significantly increased in the tumour of cachectic patients, when compared with weight-stable group (P = 0.005). CONCLUSIONS Our results demonstrate TGF-β pathway activation in the tumour in cachexia, through the (non-canonical) mitogen-activated protein kinase pathway. The results show that during cachexia, intratumoural inflammatory response contributes to the onset of fibrosis. Tumour remodelling, probably by TGF-β-induced transdifferentiation of fibroblasts to myofibroblasts, induces unbalanced inflammatory cytokine profile, angiogenesis, and elevation of extracellular matrix components (EMC). We speculate that these changes may affect tumour aggressiveness and present consequences in peripheral organs.
Collapse
Affiliation(s)
- Joanna D.C.C. Lima
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | - Estefania Simoes
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | - Gabriela de Castro
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | - Mychel Raony P.T. Morais
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | | | - Michele J. Alves
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
- Department of PathologyOhio State UniversityColumbusOHUSA
| | - Nelson I. Pinto
- Department of PhysiologyFederal University of São PauloSão PauloBrazil
| | - Raquel G. Figueredo
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | - Telma M.T. Zorn
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
| | | | - Flavio Tokeshi
- Department of Clinical SurgeryUniversity of São PauloSão PauloBrazil
| | - José P. Otoch
- Department of Clinical SurgeryUniversity of São PauloSão PauloBrazil
| | - Paulo Alcantara
- Department of Clinical SurgeryUniversity of São PauloSão PauloBrazil
| | | | - Emer S. Ferro
- Department of PharmacologyUniversity of São PauloSão PauloBrazil
| | | | - Marilia Seelaender
- Cancer Metabolism Research Group, Institute of Biomedical SciencesUniversity of São PauloSão PauloBrazil
- Department of Clinical SurgeryUniversity of São PauloSão PauloBrazil
| |
Collapse
|
40
|
de Sousa MV, da Silva Soares DB, Caraça ER, Cardoso R. Dietary protein and exercise for preservation of lean mass and perspectives on type 2 diabetes prevention. Exp Biol Med (Maywood) 2019; 244:992-1004. [PMID: 31307203 PMCID: PMC6879776 DOI: 10.1177/1535370219861910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Sedentary lifestyle and aging favor the increasing prevalence of obesity and type 2 diabetes and their comorbidities. The loss of lean body mass reduces muscle strength, resulting in impaired functional capacity and leading to increased risks of chronic diseases with advancing age. Besides aging, conditions such as inappetence, social isolation, and inadequate dietary intake cause the loss of lean body mass and increased abdominal fatty mass, resulting in sarcopenic obesity and predisposition to type 2 diabetes. Compared to younger people, this condition is more common in the elderly owing to natural changes in body composition associated with aging. Lifestyle changes such as increased physical activity and improved dietary behaviors are effective for preventing the occurrence of comorbidities. Regarding muscle nutrition, besides caloric adequacy, meeting the requirements for the consumption of dietary amino acids and proteins is important for treating sarcopenia and sarcopenic obesity because muscle tissue mainly consists of proteins and is, therefore, the largest reservoir of amino acids in the body. Thus, this review discusses the effects of dietary protein on the preservation of lean body mass, improvements in the functional capacity of muscle tissue, and prevention of chronic diseases such as type 2 diabetes. In addition, we address the effects of regular physical training associated with dietary protein strategies on lean body mass, body fat loss, and muscle strength in the elderly at a risk for type 2 diabetes development.
Collapse
Affiliation(s)
- Maysa Vieira de Sousa
- Endocrinology Division, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | | | - Elaine Reis Caraça
- Organização Social de Saúde, Santa Marcelina de Itaquaquecetuba, SP 08599-280, Brazil
| | - Ronaldo Cardoso
- Endocrinology Division, School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| |
Collapse
|
41
|
Auger C, Knuth CM, Abdullahi A, Samadi O, Parousis A, Jeschke MG. Metformin prevents the pathological browning of subcutaneous white adipose tissue. Mol Metab 2019; 29:12-23. [PMID: 31668383 PMCID: PMC6728757 DOI: 10.1016/j.molmet.2019.08.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/13/2022] Open
Abstract
Objective Browning, the conversion of white adipose tissue (WAT) to a beige phenotype, has gained interest as a strategy to induce weight loss and improve insulin resistance in metabolic disorders. However, for hypermetabolic conditions stemming from burn trauma or cancer cachexia, browning is thought to contribute to energy wasting and supraphysiological nutritional requirements. Metformin's impact on this phenomenon and underlying mechanisms have not been explored. Methods We used both a murine burn model and human ex vivo adipose explants to assess metformin and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR)'s effects on the development of subcutaneous beige adipose. Enzymes involved in fat homeostasis and browning, as well as mitochondrial dynamics, were assessed to determine metformin's effects. Results Treatment with the biguanide metformin lowers lipolysis in beige fat by inducing protein phosphatase 2A (PP2A) independently of adenosine monophosphate kinase (AMPK) activation. Increased PP2A activity catalyzes the dephosphorylation of acetyl-CoA carboxylase (Ser 79) and hormone sensitive lipase (Ser 660), thus promoting fat storage and the “whitening” of otherwise lipolytic beige adipocytes. Moreover, co-incubation of metformin with the PP2A inhibitor okadaic acid countered the anti-lipolytic effects of this biguanide in human adipose. Additionally, we show that metformin does not activate this pathway in the WAT of control mice and that AICAR sustains the browning of white adipose, offering further evidence that metformin acts independently of this cellular energy sensor. Conclusions This work provides novel insights into the mechanistic underpinnings of metformin's therapeutic benefits and potential as an agent to reduce the lipotoxicity associated with hypermetabolism and adipose browning. Metformin prevents the catabolism of murine iWAT tissue post-burn injury. Mitochondrial respiration and uncoupling in adipose are decreased by metformin. Metformin, independently of AMPK, reduces adipose lipolysis and β-oxidation via PP2A. AICAR treatment activates AMPK in peripheral adipose leading to sustained browning. PP2A is directly induced by metformin in scWAT, lowering ACC/HSL phosphorylation.
Collapse
Affiliation(s)
- Christopher Auger
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, M4N 3M5, Canada
| | - Carly M Knuth
- University of Toronto, Toronto, Ontario, M5S 1A1, Canada
| | | | - Osai Samadi
- University of Toronto, Toronto, Ontario, M5S 1A1, Canada
| | - Alexandra Parousis
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, M4N 3M5, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, M4N 3M5, Canada; University of Toronto, Toronto, Ontario, M5S 1A1, Canada.
| |
Collapse
|
42
|
Pearce JV, Farrar JS, Lownik JC, Ni B, Chen S, Kan TW, Celi FS. E0771 and 4T1 murine breast cancer cells and interleukin 6 alter gene expression patterns but do not induce browning in cultured white adipocytes. Biochem Biophys Rep 2019; 18:100624. [PMID: 31193642 PMCID: PMC6536889 DOI: 10.1016/j.bbrep.2019.100624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 03/05/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer remains a substantial clinical problem worldwide, and cancer-associated cachexia is a condition associated with poor prognosis in this and other malignancies. Adipose tissue is involved in the development and progression of cancer-associated cachexia, but its various roles and mechanisms of action are not completely defined, especially as it relates to breast cancer. Interleukin 6 has been implicated in several mechanisms contributing to increased breast cancer tumorigenesis, as well as a net-negative energy balance and cancer-associated cachexia via adipose tissue remodeling in other models of cancer; however, its potential role in breast cancer-associated white adipose browning has not been explored. In this study, we demonstrate localized white adipose tissue browning in a spontaneous model of murine mammary cancer. We then used an in vitro murine adipocyte culture system with the E0771 and 4T1 cell lines as models of breast cancer. We demonstrate that while the E0771 and 4T1 secretomes and cross-talk with white adipocytes alter white adipocyte mRNA expression, they do not directly induce white adipocyte browning. Additionally, we show that neither exogenous administration of interleukin 6 alone or with its soluble receptor directly induce white adipocyte browning. Together, these results demonstrate that neither the E0771 or 4T1 murine breast cancer cell lines, nor interleukin 6, directly cause browning of cultured white adipocytes. This suggests that their roles in adipose tissue remodeling are more complex and indirect in nature.
Collapse
Affiliation(s)
- Janina V. Pearce
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Jared S. Farrar
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Joseph C. Lownik
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Bin Ni
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Shanshan Chen
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Tiffany W. Kan
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Francesco S. Celi
- Department of Internal Medicine, Division of Endocrinology, Diabetes and Metabolism, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
43
|
Corrêa LH, Heyn GS, Magalhaes KG. The Impact of the Adipose Organ Plasticity on Inflammation and Cancer Progression. Cells 2019; 8:E662. [PMID: 31262098 PMCID: PMC6679170 DOI: 10.3390/cells8070662] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized by chronic and low-grade systemic inflammation, an increase of adipose tissue, hypertrophy, and hyperplasia of adipocytes. Adipose tissues can be classified into white, brown, beige and pink adipose tissues, which display different regulatory, morphological and functional characteristics of their adipocyte and immune cells. Brown and white adipocytes can play a key role not only in the control of energy homeostasis, or through the balance between energy storage and expenditure, but also by the modulation of immune and inflammatory responses. Therefore, brown and white adipocytes can orchestrate important immunological crosstalk that may deeply impact the tumor microenvironment and be crucial for cancer establishment and progression. Recent works have indicated that white adipose tissues can undergo a process called browning, in which an inducible brown adipocyte develops. In this review, we depict the mechanisms involved in the differential role of brown, white and pink adipocytes, highlighting their structural, morphological, regulatory and functional characteristics and correlation with cancer predisposition, establishment, and progression. We also discuss the impact of the increased adiposity in the inflammatory and immunological modulation. Moreover, we focused on the plasticity of adipocytes, describing the molecules produced and secreted by those cells, the modulation of the signaling pathways involved in the browning phenomena of white adipose tissue and its impact on inflammation and cancer.
Collapse
MESH Headings
- Adipocytes, Brown/immunology
- Adipocytes, Brown/metabolism
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/immunology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Adiposity/immunology
- Animals
- Carcinogenesis/immunology
- Carcinogenesis/pathology
- Disease Models, Animal
- Disease Progression
- Energy Metabolism/immunology
- Humans
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Obesity/complications
- Obesity/immunology
- Obesity/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Gabriella Simões Heyn
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Kelly Grace Magalhaes
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia 70910-900, Brazil.
| |
Collapse
|
44
|
CD8 + T cells induce cachexia during chronic viral infection. Nat Immunol 2019; 20:701-710. [PMID: 31110314 PMCID: PMC6531346 DOI: 10.1038/s41590-019-0397-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Cachexia represents a leading cause of morbidity and mortality in various cancers, chronic inflammation and infections. Understanding of the mechanisms that drive cachexia has remained limited, especially for infection-associated cachexia (IAC). Here we describe a model of reversible cachexia in mice with chronic viral infection and identify an essential role for CD8+ T cells in IAC. Cytokines linked to cancer-associated cachexia did not contribute to IAC. Instead, virus-specific CD8+ T cells caused morphological and molecular changes in the adipose tissue, which led to depletion of lipid stores. These changes occurred at a time point that preceded the peak of the CD8+ T cell response and required T cell–intrinsic type 1 interferon signaling and antigen-specific priming. Our results link systemic antiviral immune responses to adipose-tissue remodeling and reveal an underappreciated role of CD8+ T cells in IAC.
Collapse
|
45
|
Wang YX, Zhu N, Zhang CJ, Wang YK, Wu HT, Li Q, Du K, Liao DF, Qin L. Friend or foe: Multiple roles of adipose tissue in cancer formation and progression. J Cell Physiol 2019; 234:21436-21449. [PMID: 31054175 DOI: 10.1002/jcp.28776] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 12/19/2022]
Abstract
Obesity is well-known as the second factor for tumorigenesis after smoking and is bound up with the malignant progression of several kinds of cancers, including esophageal cancer, liver cancer, colorectal cancer, kidney cancer, and ovarian cancer. The increased morbidity and mortality of obesity-related cancer are mostly attributed to dysfunctional adipose tissue. The possible mechanisms connecting dysfunctional adipose tissue to high cancer risk mainly focus on chronic inflammation, obesity-related microenvironment, adipokine secretion disorder, and browning of adipose tissue, and so forth. The stromal vascular cells in adipose tissue trigger chronic inflammation through secreting inflammatory factors and promote cancer cell proliferation. Hypertrophic adipose tissues lead to metabolic disorders of adipocytes, such as abnormal levels of adipokines that mediate cancer progression and metastasis. Cancer patients often show adipose tissue browning and cancerous cachexia in an advanced stage, which lead to unsatisfied chemotherapy effect and poor prognosis. However, increasing evidence has shown that adipose tissue may display quite opposite effects in cancer development. Therefore, the interaction between cancers and adipose tissue exert a vital role in mediates adipose tissue dysfunction and further leads to cancer progression. In conclusion, targeting the dysfunction of adipose tissue provides a promising strategy for cancer prevention and therapy.
Collapse
Affiliation(s)
- Yu-Xiang Wang
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Neng Zhu
- Department of Urology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chan-Juan Zhang
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yi-Kai Wang
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, Georgia
| | - Hong-Tao Wu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qun Li
- Outpatient Department of Hanpu Campus, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ke Du
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, Key Lab for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Li Qin
- School of Pharmacy, Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Cancer-associated muscle wasting affects many patients and leads to reduced patient function, decreased quality of life and poor responses to surgical and oncological treatments. Despite advancements in the understanding of its pathophysiology, no current treatment or accepted strategy for successful management exists. In this review, we provide an update on potential novel therapeutic targets in cancer cachexia. RECENT FINDINGS Recent research has focused on molecular mechanisms underlying cancer-associated muscle wasting, allowing identification of potential therapeutic targets and the development of several promising drugs. However, due to the multifactorial and patient-specific pathogenesis of cachexia, the demonstration of a measurable and meaningful clinical effect in randomized controlled trials has proven difficult. Potential novel targets such as circulating macrophage inhibitory cytokine 1/growth differentiation factor 15 and ZRT/IRT-like protein 14 have shown relevance in animal models, but their therapeutic manipulation has yet to be translated to patients. Increasing evidence has suggested that a single therapy may not be successful and a targeted, multimodal approach is required. SUMMARY The management of cancer-associated muscle wasting is complex. Future clinical trials should focus on early multimodal therapeutic interventions involving targeted therapies, with careful deliberation of chosen nutritional and functional outcomes.
Collapse
Affiliation(s)
- Janice Miller
- Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
47
|
Yanagihara K, Kubo T, Iino Y, Mihara K, Morimoto C, Seyama T, Kuwata T, Ochiai A, Yokozaki H. Development and characterization of a cancer cachexia model employing a rare human duodenal neuroendocrine carcinoma-originating cell line. Oncotarget 2019; 10:2435-2450. [PMID: 31069007 PMCID: PMC6497432 DOI: 10.18632/oncotarget.26764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer cachexia interferes with therapy and worsens patients' quality of life. Therefore, for a better understanding of cachexia, we aimed to establish a reliable cell line to develop a cachexia model. We recently established and characterized the TCC-NECT-2 cell line, derived from a Japanese patient with poorly differentiated neuroendocrine carcinoma of the duodenum (D-NEC). Subcutaneous xenograft of TCC-NECT-2 cells in mice resulted in tumor formation, angiogenesis, and 20% incidence of body weight (BW)-loss. Subsequently, we isolated a potent cachexia-inducing subline using stepwise selection and designated as AkuNEC. Orthotopic and s.c. implantation of AkuNEC cells into mice led to diminished BW, anorexia, skeletal muscle atrophy, adipose tissue loss, and decreased locomotor activity at 100% incidence. Additionally, orthotopic implantation of AkuNEC cells resulted in metastasis and angiogenesis. Serum IL-8 overproduction was observed, and levels were positively correlated with BW-loss and reduced adipose tissue and muscle volumes in tumor-bearing mice. However, shRNA knockdown of the IL-8 gene did not suppress tumor growth and cachexia in the AkuNEC model, indicating that IL-8 is not directly involved in cachexia induction. In conclusion, AkuNEC cells may serve as a useful model to study cachexia and D-NEC.
Collapse
Affiliation(s)
- Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takanori Kubo
- Department of Life Sciences, Yasuda Women’s University Faculty of Pharmacy, Hiroshima, Japan
| | - Yuki Iino
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Keichiro Mihara
- Department of Hematology/Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Chie Morimoto
- Department of Living Science Nutrition Course, Matsuyama Shinonome Junior College, Matsuyama, Japan
| | - Toshio Seyama
- Department of Life Sciences, Yasuda Women’s University Faculty of Pharmacy, Hiroshima, Japan
| | - Takeshi Kuwata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Chiba, Japan
| | - Atsushi Ochiai
- Division of Biomarker Discovery, Exploratory Oncology and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
48
|
Abstract
Cachexia is a systemic condition that occurs during many neoplastic diseases, such as cancer. Cachexia in cancer is characterized by loss of body weight and muscle and by adipose tissue wasting and systemic inflammation. Cancer cachexia is often associated with anorexia and increased energy expenditure. Even though the cachectic condition severely affects skeletal muscle, a tissue that accounts for ~40% of total body weight, it represents a multi-organ syndrome that involves tissues and organs such as white adipose tissue, brown adipose tissue, bone, brain, liver, gut and heart. Indeed, evidence suggests that non-muscle tissues and organs, as well as tumour tissues, secrete soluble factors that act on skeletal muscle to promote wasting. In addition, muscle tissue also releases various factors that can interact with the metabolism of other tissues during cancer. In this Review, we examine the effect of non-muscle tissues and inter-tissue communication in cancer cachexia and discuss studies aimed at developing novel therapeutic strategies for the condition.
Collapse
Affiliation(s)
- Josep M Argilés
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| | | | - Francisco J López-Soriano
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| | - Silvia Busquets
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
49
|
Dropping in on lipid droplets: insights into cellular stress and cancer. Biosci Rep 2018; 38:BSR20180764. [PMID: 30111611 PMCID: PMC6146295 DOI: 10.1042/bsr20180764] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Lipid droplets (LD) have increasingly become a major topic of research in recent years following its establishment as a highly dynamic organelle. Contrary to the initial view of LDs being passive cytoplasmic structures for lipid storage, studies have provided support on how they act in concert with different organelles to exert functions in various cellular processes. Although lipid dysregulation resulting from aberrant LD homeostasis has been well characterised, how this translates and contributes to cancer progression is poorly understood. This review summarises the different paradigms on how LDs function in the regulation of cellular stress as a contributing factor to cancer progression. Mechanisms employed by a broad range of cancer cell types in differentially utilising LDs for tumourigenesis will also be highlighted. Finally, we discuss the potential of targeting LDs in the context of cancer therapeutics.
Collapse
|
50
|
Carobbio S, Guénantin AC, Samuelson I, Bahri M, Vidal-Puig A. Brown and beige fat: From molecules to physiology and pathophysiology. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:37-50. [PMID: 29852279 DOI: 10.1016/j.bbalip.2018.05.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/31/2018] [Accepted: 05/23/2018] [Indexed: 12/20/2022]
Abstract
The adipose organ portrays adipocytes of diverse tones: white, brown and beige, each type with distinct functions. Adipocytes orchestrate their adaptation and expansion to provide storage to excess nutrients, the quick mobilisation of fuel to supply peripheral functional demands, insulation, and, in their thermogenic form, heat generation to maintain core body temperature. Thermogenic adipocytes could be targets for anti-obesity and anti-diabetic therapeutic approaches aiming to restore adipose tissue functionality and increase energy dissipation. However, for thermogenic adipose tissue to become therapeutically relevant, a better understanding of its development and origins, its progenitors and their characteristics and the composition of its niche, is essential. Also crucial is the identification of stimuli and molecules promoting its specific differentiation and activation. Here we highlight the structural/cellular differences between human and rodent brown adipose tissue and discuss how obesity and metabolic complication affects brown and beige cells as well as how they could be targeted to improve their activation and improve global metabolic homeostasis. Finally, we describe the limitations of current research models and the advantages of new emerging approaches.
Collapse
Affiliation(s)
- Stefania Carobbio
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Anne-Claire Guénantin
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Isabella Samuelson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Myriam Bahri
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK; Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|