1
|
Parry TL, Gilmore LA, Khamoui AV. Pan-cancer secreted proteome and skeletal muscle regulation: insight from a proteogenomic data-driven knowledge base. Funct Integr Genomics 2025; 25:14. [PMID: 39812750 DOI: 10.1007/s10142-024-01524-7] [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: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
Large-scale, pan-cancer analysis is enabled by data driven knowledge bases that link tumor molecular profiles with phenotypes. A debilitating cancer-related phenotype is skeletal muscle loss, or cachexia, which occurs partly from tumor products secreted into circulation. Using the LinkedOmicsKB knowledge base assembled from the Clinical Proteomics Tumor Analysis Consortium proteogenomic analysis, along with catalogs of human secretome proteins, ligand-receptor pairs and molecular signatures, we sought to identify candidate pan-cancer proteins secreted to blood that could regulate skeletal muscle phenotypes in multiple solid cancers. Tumor proteins having significant pan-cancer associations with muscle were referenced against secretome proteins secreted to blood from the Human Protein Atlas, then verified as increased in paired tumor vs. normal tissues in pan-cancer manner. This workflow revealed seven secreted proteins from cancers afflicting kidneys, head and neck, lungs and pancreas that classified as protein-binding activity modulator, extracellular matrix protein or intercellular signaling molecule. Concordance of these biomarkers with validated molecular signatures of cachexia and senescence supported relevance to muscle and cachexia disease biology, and high tumor expression of the biomarker set associated with lower overall survival. In this article, we discuss avenues by which skeletal muscle and cachexia may be regulated by these candidate pan-cancer proteins secreted to blood, and conceptualize a strategy that considers them collectively as a biomarker signature with potential for refinement by data analytics and radiogenomics for predictive testing of future risk in a non-invasive, blood-based panel amenable to broad uptake and early management.
Collapse
Affiliation(s)
- Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA.
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
2
|
Wyart E, Carrà G, Angelino E, Penna F, Porporato PE. Systemic metabolic crosstalk as driver of cancer cachexia. Trends Endocrinol Metab 2025:S1043-2760(24)00327-8. [PMID: 39757061 DOI: 10.1016/j.tem.2024.12.005] [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: 09/25/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025]
Abstract
Cachexia is a complex metabolic disorder characterized by negative energy balance due to increased consumption and lowered intake, leading to progressive tissue wasting and inefficient energy distribution. Once considered as passive bystander, metabolism is now acknowledged as a regulator of biological functions and disease progression. This shift in perspective mirrors the evolving understanding of cachexia itself, no longer viewed merely as a secondary consequence of cancer but as an active process. However, metabolic dysregulations in cachexia are currently studied in an organ-specific manner, failing to be fully integrated into a comprehensive framework that explains their functional roles in disease progression. Thus, in this review, we aim to provide a general overview of the various metabolic alterations with a potential systemic impact.
Collapse
Affiliation(s)
- Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center 'Guido Tarone', University of Torino, 10126 Torino, Italy.
| | - Giovanna Carrà
- San Luigi Gonzaga Hospital, Orbassano, Italy; Department of Clinical and Biological Science, University of Torino, Orbassano, Italy
| | - Elia Angelino
- Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Fabio Penna
- Department of Clinical and Biological Science, University of Torino, Orbassano, Italy
| | - Paolo E Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center 'Guido Tarone', University of Torino, 10126 Torino, Italy.
| |
Collapse
|
3
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2025; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
4
|
Arneson-Wissink PC, Pelz K, Worley B, Mendez H, Pham P, McCarthy G, Chitsazan A, Brody JR, Grossberg AJ. The RNA-binding protein HuR impairs adipose tissue anabolism in pancreatic cancer cachexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630549. [PMID: 39763867 PMCID: PMC11703191 DOI: 10.1101/2024.12.27.630549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Background Cachexia is defined by chronic loss of fat and muscle, is a frequent complication of pancreatic ductal adenocarcinoma (PDAC), and negatively impacts patient outcomes. Nutritional supplementation cannot fully reverse tissue wasting, and the mechanisms underlying this phenotype are unclear. This work aims to define the relative contributions of catabolism and anabolism to adipose wasting in PDAC-bearing mice. Human antigen R (HuR) is an RNA-binding protein recently shown to suppress adipogenesis. We hypothesize that fat wasting results from a loss of adipose anabolism driven by increased HuR activity in adipocytes of PDAC-bearing mice. Methods Adult C57BL/6J mice received orthotopic PDAC cell (Kras G12D ; p53 R172H/+ ; Pdx1-cre) (OT-PDAC) or PBS (sham) injections. Mice exhibiting moderate cachexia (9 days after injection) were fasted for 24h, or fasted 24h and refed 24h before euthanasia. A separate cohort of PDAC mice were treated with an established HuR inhibitor (KH-3, 100 mg/kg) and subjected to the fast/refeed paradigm. We analyzed body mass, gross fat pad mass, and adipose tissue mRNA expression. We quantified lipolytic rate as the normalized quantity of glycerol released from 3T3-L1 adipocytes in vitro, and gonadal fat pads (gWAT) ex vivo. Results 3T3-L1 adipocytes treated with PDAC cell conditioned media (CM) liberated less triglyceride into the culture media than control-treated adipocytes (-28.1%) and had lower expression of lipolysis and lipogenesis genes than control cells. PDAC gWAT cultured ex vivo displayed decreased lipolysis compared to sham gWAT (-54.7%). PDAC and sham mice lost equivalent fat mass after a 24h fast, however, PDAC mice could not restore inguinal fat pads (iWAT) (-40.5%) or gWAT (-31.8%) mass after refeeding. RNAseq revealed 572 differentially expressed genes in gWAT from PDAC compared to sham mice. Downregulated genes (n=126) were associated with adipogenesis (adj p=0.05), and expression of adipogenesis master regulators Pparg and Cebpa were reduced in gWAT from PDAC mice. Immunohistochemistry revealed increased HuR staining in gWAT (+74.9%) and iWAT (+41.2%) from PDAC mice. Inhibiting HuR binding restored lipogenesis in refed animals with a concomitant increase in iWAT mass (+131.7%) and genes regulating adipogenesis (Pparγ, Cebpa, Retn, Adipoq, Fasn). Conclusions Our work highlights deficient adipose anabolism as a driver of wasting in 3T3-L1 adipocytes treated with PDAC conditioned media and OT-PDAC mice. The small molecule KH3, which disrupts HuR binding, was sufficient to restore adipogenic and lipogenic gene expression and prevent adipose wasting. This highlights HuR as a potentially targetable regulatory node for adipose anabolism in cancer cachexia.
Collapse
Affiliation(s)
- Paige C. Arneson-Wissink
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
| | - Katherine Pelz
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Surgery, Oregon Health & Science University, Portland, OR
| | - Beth Worley
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
| | - Heike Mendez
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
| | - Peter Pham
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
| | - Grace McCarthy
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Surgery, Oregon Health & Science University, Portland, OR
| | - Alex Chitsazan
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR
| | - Jonathan R. Brody
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Surgery, Oregon Health & Science University, Portland, OR
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| | - Aaron J. Grossberg
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| |
Collapse
|
5
|
Kim S, Park H, Hong HJ, Lee S, Kim S, Lee YK, Shong M, Kim YC. Liposomal Codelivery of Doxorubicin and Curcumin Sensitizes Antitumor Activity and Reduces Tumor Metastasis. ACS APPLIED BIO MATERIALS 2024; 7:8367-8376. [PMID: 39591430 DOI: 10.1021/acsabm.4c01146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Multidrug resistance (MDR) is a major obstacle to traditional cancer treatment using chemotherapeutic agents like doxorubicin (DOX). MDR affects drug dosage regimens and enables the recurrence and metastasis of cancer. Because DOX causes severe side effects at high dosages, it is important to use an MDR modulator to make cancer cells sensitive to DOX. This work focused on a liposome-based codelivery system containing curcumin (CUR) and DOX, focusing on CUR as an MDR modulator. The synergistic effect was maximized when the ratio of DOX and CUR was 1:1, and the synthesis of liposomal drugs was successfully verified. In addition, a successful MDR reversal effect was demonstrated through rhodamine 123 assay, Western blotting, and immunofluorescence. Compared to the conventional DOX treatment, the dual-drug treatment exhibits a significantly improved anticancer effect. In the murine metastasis 4T1 IP tumor model, the dual-drug-encapsulating liposomes successfully suppressed tumor growth and reversed the tumoral effect (omental tumor metastasis, fat, and muscle weight loss) into a normal state.
Collapse
Affiliation(s)
- Suyeon Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Heewon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Hyun Jung Hong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
- Life Science Research Institute, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Susam Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Sejin Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, Chung-Buk 27469, Republic of Korea
| | - Minho Shong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| |
Collapse
|
6
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
7
|
Huang L, Zhu L, Zhao Z, Jiang S. Hyperactive browning and hypermetabolism: potentially dangerous element in critical illness. Front Endocrinol (Lausanne) 2024; 15:1484524. [PMID: 39640882 PMCID: PMC11617193 DOI: 10.3389/fendo.2024.1484524] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Brown/beige adipose tissue has attracted much attention in previous studies because it can improve metabolism and combat obesity through non-shivering thermogenesis. However, recent studies have also indicated that especially in critical illness, overactivated brown adipose tissue or extensive browning of white adipose tissue may bring damage to individuals mainly by exacerbating hypermetabolism. In this review, the phenomenon of fat browning in critical illness will be discussed, along with the potential harm, possible regulatory mechanism and corresponding clinical treatment options of the induction of fat browning. The current research on fat browning in critical illness will offer more comprehensive understanding of its biological characteristics, and inspire researchers to develop new complementary treatments for the hypermetabolic state that occurs in critically ill patients.
Collapse
Affiliation(s)
- Lu Huang
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lili Zhu
- Department of Plastic and Reconstructive Surgery, Taizhou Enze Hospital, Taizhou, China
| | - Zhenxiong Zhao
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Shenglu Jiang
- Department of Basic Medical Sciences, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
8
|
Yan G, Liu L, Liu M, Jiang X, Chen P, Li M, Ma Q, Li Y, Duan S, You R, Huang Y, Li Z, You D. Bidirectional association between perioperative skeletal muscle and subcutaneous fat in colorectal cancer patients and their prognostic significance. Front Nutr 2024; 11:1381995. [PMID: 39360277 PMCID: PMC11445023 DOI: 10.3389/fnut.2024.1381995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Low skeletal muscle mass and high adipose tissue coexist across the body weight spectrum and independently predict the survival ratio of colorectal cancer (CRC) patients. This combination may lead to a mutually exacerbating vicious cycle. Tumor-associated metabolic conditions primarily affect subcutaneous adipose tissue, but the nature and direction of its relationship with skeletal muscle are unclear. This study aims to examine the bidirectional causal relationship between skeletal muscle index (SMI) and subcutaneous fat index (SFI) during the perioperative period in CRC patients; as well as to validate the association between perioperative SMI, SFI, and CRC prognosis. Methods This population-based retrospective cohort study included patients with stage I-III colorectal cancer who underwent radical resection at the Third Affiliated Hospital of Kunming Medical University between September 2012 and February 2019. Based on inclusion and exclusion criteria, 1,448 patients were analyzed. Preoperative (P1), 2 months postoperative (P2), and 5 months postoperative (P3) CT scans were collected to evaluate the skeletal muscle index (SMI; muscle area at the third lumbar vertebra divided by height squared) and subcutaneous fat index (SFI; subcutaneous fat area at the third lumbar vertebra divided by height squared). A random intercept cross-lagged panel model (RI-CLPM) was used to examine the intra-individual relationship between SMI and SFI, and Cox regression was employed to assess the association between SMI, SFI, recurrence-free survival (RFS), and overall survival (OS). Results The median age at diagnosis was 59.00 years (IQR: 51.00-66.00), and 587 patients (40.54%) were female. RI-CLPM analysis revealed a negative correlation between SFI and subsequent SMI at the individual level: P1-P2 (β = -0.372, p = 0.038) and P2-P3 (β = -0.363, p = 0.001). SMI and SFI showed a negative correlation during P1-P2 (β = -0.363, p = 0.001) but a positive correlation during P2-P3 (β = 0.357, p = 0.006). No significant correlation was found between the random intercepts of SFI and SMI at the between-person level (r = 0.157, p = 0.603). The Cox proportional hazards multivariate regression model identified that patients with elevated SFI had poorer recurrence-free survival (HR, 1.24; 95% CI: 1.00-1.55). Compared to patients with normal preoperative SMI and SFI, those with low SMI or high SFI had poorer recurrence-free survival (HR, 1.26; 95% CI: 1.03-1.55) and overall survival (HR, 1.39; 95% CI: 1.04-1.87). However, no significant association between SMI and SFI and the prognosis of colorectal cancer patients was observed postoperatively. Conclusion In CRC patients, preoperative muscle loss leads to postoperative fat accumulation, exacerbating muscle loss in a feedback loop. Elevated preoperative SFI predicts poorer survival outcomes. Monitoring SMI and SFI is crucial as prognostic indicators, despite non-significant postoperative associations. Further research is needed to improve patient outcomes.
Collapse
Affiliation(s)
- Guanghong Yan
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Lizhu Liu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Mengmei Liu
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Xinyue Jiang
- Second Ward of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ping Chen
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Min Li
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Qingyan Ma
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Yani Li
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Sifan Duan
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| | - Ruimin You
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yanni Huang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Dingyun You
- Yunnan Provincial Key Laboratory of Public Health and Biosafety, School of Public Health, Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Wang J, Wang R, Li Y, Huang J, Liu Y, Wang J, Xian P, Zhang Y, Yang Y, Zhang H, Li J. Lipolysis engages CD36 to promote ZBP1-mediated necroptosis-impairing lung regeneration in COPD. Cell Rep Med 2024; 5:101732. [PMID: 39255796 PMCID: PMC11525022 DOI: 10.1016/j.xcrm.2024.101732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 06/05/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Lung parenchyma destruction represents a severe condition commonly found in chronic obstructive pulmonary disease (COPD), a leading cause of morbidity and mortality worldwide. Promoting lung regeneration is crucial for achieving clinical improvement. However, no therapeutic drugs are approved to improve the regeneration capacity due to incomplete understanding of the underlying pathogenic mechanisms. Here, we identify a positive feedback loop formed between adipose triglyceride lipase (ATGL)-mediated lipolysis and overexpression of CD36 specific to lung epithelial cells, contributing to disease progression. Genetic deletion of CD36 in lung epithelial cells and pharmacological inhibition of either ATGL or CD36 effectively reduce COPD pathogenesis and promote lung regeneration in mice. Mechanistically, disruption of the ATGL-CD36 loop rescues Z-DNA binding protein 1 (ZBP1)-induced cell necroptosis and restores WNT/β-catenin signaling. Thus, we uncover a crosstalk between lipolysis and lung epithelial cells, suggesting the regenerative potential for therapeutic intervention by targeting the ATGL-CD36-ZBP1 axis in COPD.
Collapse
Affiliation(s)
- Jiazhen Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ru Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yicun Li
- Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jiahui Huang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiayi Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Xian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuanhang Zhang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanmei Yang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haojian Zhang
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
10
|
Hayashi Y, Kamimura-Aoyagi Y, Nishikawa S, Noka R, Iwata R, Iwabuchi A, Watanabe Y, Matsunuma N, Yuki K, Kobayashi H, Harada Y, Harada H. IL36G-producing neutrophil-like monocytes promote cachexia in cancer. Nat Commun 2024; 15:7662. [PMID: 39266531 PMCID: PMC11393454 DOI: 10.1038/s41467-024-51873-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 08/19/2024] [Indexed: 09/14/2024] Open
Abstract
Most patients with advanced cancer develop cachexia, a multifactorial syndrome characterized by progressive skeletal muscle wasting. Despite its catastrophic impact on survival, the critical mediators responsible for cancer cachexia development remain poorly defined. Here, we show that a distinct subset of neutrophil-like monocytes, which we term cachexia-inducible monocytes (CiMs), emerges in the advanced cancer milieu and promotes skeletal muscle loss. Unbiased transcriptome analysis reveals that interleukin 36 gamma (IL36G)-producing CD38+ CiMs are induced in chronic monocytic blood cancer characterized by prominent cachexia. Notably, the emergence of CiMs and the activation of CiM-related gene signatures in monocytes are confirmed in various advanced solid cancers. Stimuli of toll-like receptor 4 signaling are responsible for the induction of CiMs. Genetic inhibition of IL36G-mediated signaling attenuates skeletal muscle loss and rescues cachexia phenotypes in advanced cancer models. These findings indicate that the IL36G-producing subset of neutrophil-like monocytes could be a potential therapeutic target in cancer cachexia.
Collapse
Affiliation(s)
- Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
- Laboratory of Cancer Pathobiology and Therapeutics, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan.
| | - Yasushige Kamimura-Aoyagi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Sayuri Nishikawa
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Rena Noka
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Rika Iwata
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Asami Iwabuchi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yushin Watanabe
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Natsumi Matsunuma
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kanako Yuki
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hiroki Kobayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuka Harada
- Clinical Research Support Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| |
Collapse
|
11
|
Geppert J, Rohm M. Cancer cachexia: biomarkers and the influence of age. Mol Oncol 2024; 18:2070-2086. [PMID: 38414161 PMCID: PMC11467804 DOI: 10.1002/1878-0261.13590] [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: 04/04/2023] [Revised: 12/01/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
Cancer cachexia (Ccx) is a complex metabolic condition characterized by pronounced muscle and fat wasting, systemic inflammation, weakness and fatigue. Up to 30% of cancer patients succumb directly to Ccx, yet therapies that effectively address this perturbed metabolic state are rare. In recent decades, several characteristics of Ccx have been established in mice and humans, of which we here highlight adipose tissue dysfunction, muscle wasting and systemic inflammation, as they are directly linked to biomarker discovery. To counteract cachexia pathogenesis as early as possible and mitigate its detrimental impact on anti-cancer treatments, identification and validation of clinically endorsed biomarkers assume paramount importance. Ageing was recently shown to affect both the validity of Ccx biomarkers and Ccx development, but the underlying mechanisms are still unknown. Thus, unravelling the intricate interplay between ageing and Ccx can help to counteract Ccx pathogenesis and tailor diagnostic and treatment strategies to individual needs.
Collapse
Affiliation(s)
- Julia Geppert
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Maria Rohm
- Institute for Diabetes and CancerHelmholtz MunichNeuherbergGermany
- Joint Heidelberg‐IDC Translational Diabetes Program, Inner Medicine 1Heidelberg University HospitalGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
12
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
13
|
Zhang Y, Dos Santos M, Huang H, Chen K, Iyengar P, Infante R, Polanco PM, Brekken RA, Cai C, Caijgas A, Cano Hernandez K, Xu L, Bassel-Duby R, Liu N, Olson EN. A molecular pathway for cancer cachexia-induced muscle atrophy revealed at single-nucleus resolution. Cell Rep 2024; 43:114587. [PMID: 39116208 PMCID: PMC11472345 DOI: 10.1016/j.celrep.2024.114587] [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: 02/14/2024] [Revised: 05/14/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer cachexia is a prevalent and often fatal wasting condition that cannot be fully reversed with nutritional interventions. Muscle atrophy is a central component of the syndrome, but the mechanisms whereby cancer leads to skeletal muscle atrophy are not well understood. We performed single-nucleus multi-omics on skeletal muscles from a mouse model of cancer cachexia and profiled the molecular changes in cachexic muscle. Our results revealed the activation of a denervation-dependent gene program that upregulates the transcription factor myogenin. Further studies showed that a myogenin-myostatin pathway promotes muscle atrophy in response to cancer cachexia. Short hairpin RNA inhibition of myogenin or inhibition of myostatin through overexpression of its endogenous inhibitor follistatin prevented cancer cachexia-induced muscle atrophy in mice. Our findings uncover a molecular basis of muscle atrophy associated with cancer cachexia and highlight potential therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Yichi Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthieu Dos Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Huocong Huang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kenian Chen
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Puneeth Iyengar
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rodney Infante
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Patricio M Polanco
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunyu Cai
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ambar Caijgas
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Karla Cano Hernandez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Zhou Y, Chu P, Wang Y, Li N, Gao Q, Wang S, Wei J, Xue G, Zhao Y, Jia H, Song J, Zhang Y, Pang Y, Zhu H, Sun J, Ma S, Su C, Hu B, Zhao Z, Zhang H, Lu J, Wang J, Wang H, Sun Z, Fang D. Epinephrine promotes breast cancer metastasis through a ubiquitin-specific peptidase 22-mediated lipolysis circuit. SCIENCE ADVANCES 2024; 10:eado1533. [PMID: 39151008 PMCID: PMC11328899 DOI: 10.1126/sciadv.ado1533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/10/2024] [Indexed: 08/18/2024]
Abstract
Chronic stress-induced epinephrine (EPI) accelerates breast cancer progression and metastasis, but the molecular mechanisms remain unclear. Herein, we found a strong positive correlation between circulating EPI levels and the tumoral expression of ubiquitin-specific peptidase 22 (USP22) in patients with breast cancer. USP22 facilitated EPI-induced breast cancer progression and metastasis by enhancing adipose triglyceride lipase (ATGL)-mediated lipolysis. Targeted USP22 deletion decreased ATGL expression and lipolysis, subsequently inhibiting EPI-mediated breast cancer lung metastasis. USP22 acts as a bona fide deubiquitinase for the Atgl gene transcription factor FOXO1, and EPI architects a lipolysis signaling pathway to stabilize USP22 through AKT-mediated phosphorylation. Notably, USP22 phosphorylation levels are positively associated with EPI and with downstream pathways involving both FOXO1 and ATGL in breast cancers. Pharmacological USP22 inhibition synergized with β-blockers in treating preclinical xenograft breast cancer models. This study reveals a molecular pathway behind EPI's tumor-promoting effects and provides a strong rationale for combining USP22 inhibition with β-blockers to treat aggressive breast cancer.
Collapse
Affiliation(s)
- Yuanzhang Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Peng Chu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ya Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Na Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Qiong Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shengnan Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juncheng Wei
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guoqing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yue Zhao
- Department of Clinical Laboratory, Dalian Municipal Central Hospital, Dalian 116000, China
| | - Huijun Jia
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Jiankun Song
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yue Zhang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Yujie Pang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Houyu Zhu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Jia Sun
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Suxian Ma
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Chen Su
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Bingjin Hu
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Zhuoyue Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Hui Zhang
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Janice Lu
- Department of Medicine & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jian Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Hongjiang Wang
- Department of Breast Surgery, First Affiliated Hospital of Dalian Medical University, Dalian 116044, China
| | - Zhaolin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
- Dalian College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Deyu Fang
- Department of Pathology & Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
15
|
Tien SC, Chang CC, Huang CH, Peng HY, Chang YT, Chang MC, Lee WH, Hu CM. Exosomal miRNA 16-5p/29a-3p from pancreatic cancer induce adipose atrophy by inhibiting adipogenesis and promoting lipolysis. iScience 2024; 27:110346. [PMID: 39055920 PMCID: PMC11269291 DOI: 10.1016/j.isci.2024.110346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/20/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Over 80% of the patients with pancreatic ductal adenocarcinoma (PDAC) have cachexia/wasting syndrome. Cachexia is associated with reduced survival, decreased quality of life, and higher metastasis rates. Here, we demonstrate that fat loss is the earliest feature of PDAC-exosome-induced cachexia. MicroRNA sequencing of exosomal components from normal and cancer-derived exosomes revealed enrichment of miR-16-5p, miR-21-5p, miR-29a-3p, and miR-125b-5p in serum exosomes of mice harboring PDAC and patients with PDAC. Further, miR-16-5p and miR-29a-3p inhibited adipogenesis through decreasing Erlin2 and Cmpk1 expression which downregulates C/EBPβ and PPARγ. Synergistically, miR-29a-3p promotes lipolysis through increasing ATGL expression by suppressing MCT1 expression. Furthermore, PDAC-exosomes deprived of miR-16-5p and miR-29a-3p fail to induce fat loss. Hence, miR-16-5p and miR-29a-3p exosomal miRs are essential for PDAC-induced fat loss. Thus, we unravel that PDAC induces adipose atrophy via exosomal miRs. This knowledge may provide new diagnostic and therapeutic strategies for PDAC-induced cachexia.
Collapse
Affiliation(s)
- Sui-Chih Tien
- Genomics Research Center, Academia Sinica, Taipei 115201, Taiwan
| | - Chin-Chun Chang
- Genomics Research Center, Academia Sinica, Taipei 115201, Taiwan
| | | | - Hsuan-Yu Peng
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100225, Taiwan
| | - Yu-Ting Chang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100225, Taiwan
- National Taiwan University Hospital Hsin-Chu Branch, Zhubei City, Hsinchu County 302058, Taiwan
| | - Ming-Chu Chang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei 100225, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Wen-Hwa Lee
- Genomics Research Center, Academia Sinica, Taipei 115201, Taiwan
- Drug Development Center, China Medical University, Taichung 406040, Taiwan
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Chun-Mei Hu
- Genomics Research Center, Academia Sinica, Taipei 115201, Taiwan
| |
Collapse
|
16
|
Park WY, Song G, Park JY, Jung SJ, Kim S, Ahn KS, Choe SK, Kwak HJ, Park J, Um JY. Epigallocatechin gallate protects against fat and muscle atrophy in B16BL6 melanoma-bearing mice on a high-fat diet. Life Sci 2024; 348:122677. [PMID: 38702026 DOI: 10.1016/j.lfs.2024.122677] [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: 04/12/2023] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
AIMS Epidemiological evidence indicates that there is a substantial association between body mass index (BMI) and at least ten forms of cancer, including melanoma, and BMI imbalance contributes to the poor survival rate of cancer patients before and after therapy. Nevertheless, few pharmacological studies on models of obesity and cancer have been reported. In this study, we administered epigallocatechin gallate (EGCG) to B16BL6 tumor-bearing mice that received a high-fat diet (HFD) to examine its impact. METHODS B16BL6 tumor-bearing mice were fed a HFD. Body weight and food intake were documented every week. We conducted a Western blot analysis to examine the protein levels in the tumor, gastrocnemius (GAS), and tibialis anterior (TA) muscles, as well as the inguinal and epididymal white adipose tissues (iWAT and eWAT). KEY FINDINGS EGCG has been shown to have anti-cancer effects equivalent to those of cisplatin, a chemotherapy drug. Furthermore, EGCG protected against the loss of epidydimal white adipose tissue by regulating protein levels of lipolysis factors of adipose triglyceride lipase and hormone-sensitive lipase as well as WAT browning factors of uncoupling protein 1, as opposed to cisplatin. EGCG was shown to reduce the protein levels of muscular atrophy factors of muscle RING-finger protein-1, whereas cisplatin did not contribute to rescuing the atrophy of TA and GAS muscles. CONCLUSION Taken together, our findings indicate that EGCG has a preventive effect against cachexia symptoms and has anti-cancer effects similar to those of cisplatin in tumor-bearing mice fed a high-fat diet.
Collapse
Affiliation(s)
- Woo Yong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gahee Song
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seokwoo Kim
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Jinbong Park
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae-Young Um
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
17
|
Tian HY, Yang M, Xie HL, Ruan GT, Ge YZ, Zhang XW, Zhang HY, Liu CA, Liu T, Shi HP. The effect of hyperlipidemia on overall survival in patients with cancer was differentiated by BMI and hyperlipidemia type. Nutr Metab (Lond) 2024; 21:37. [PMID: 38914993 PMCID: PMC11197256 DOI: 10.1186/s12986-024-00811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND AND AIMS The impact of lipids on the overall survival (OS) of patients with malignancy has not yet been clarified. This study aimed to evaluate the effect of hyperlipidemia on the OS among Chinese patients based on Body Mass Index (BMI) stratifications and hyperlipidemia types. METHOD The patients in this study were derived from the Investigation of the Nutrition Status and Clinical Outcome of Common Cancers (INSCOC) trial. Kaplan-Meier was used to draw the survival curve, and the log-rank test was used to estimate the survival rates between each group. Cox proportional hazards regression models were used to estimate the hazard ratios (HR) and 95% confidence intervals (CI). RESULTS A total of 9054 patients were included in the final study, with a median age of 59 years, and 55.3% (5004) of them were males. Regarding types of hyperlipidemia, only low high-density lipoprotein was an independent risk factor for the prognosis of all patients (HR = 1.35, 95% CI: 1.25-1.45, P < 0.001), while high total cholesterol (HR = 1.01, 95% CI: 0.90-1.15, P = 0.839) and high low-density lipoprotein (HR = 1.03, 95%CI: 0.91-1.16, P = 0.680) were not. In terms of BMI stratification, the effect of triglycerides on prognosis varied; high triglycerides were an independent risk factor for the prognosis of underweight patients (HR = 1.56, 95% CI:1.05-2.32, P = 0.027) and a protective factor for overweight patients (HR = 0.75, 95% CI: 0.63-0.89, P = 0.001). However, for normal-weight patients, there was no significant statistical difference (HR = 0.88, 95%CI: 0.75-1.03, P = 0.108). CONCLUSIONS The impact of hyperlipidemia on the OS among patients with cancer varied by different BMI and hyperlipidemia types. BMI and hyperlipidemia type ought to be considered in combination to estimate the prognosis of patients with malignancy.
Collapse
Affiliation(s)
- Hai-Ying Tian
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hai-Lun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Guo-Tian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yi-Zhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiao-Wei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - He-Yang Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Chen-An Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tong Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China.
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
18
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Jin C, Wang S, Sui X, Meng Q, Wu G. Low expression of ELOVL6 may be involved in fat loss in white adipose tissue of cancer-associated cachexia. Lipids Health Dis 2024; 23:144. [PMID: 38760797 PMCID: PMC11100253 DOI: 10.1186/s12944-024-02126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Cancer-associated cachexia (CAC) arises from malignant tumors and leads to a debilitating wasting syndrome. In the pathophysiology of CAC, the depletion of fat plays an important role. The mechanisms of CAC-induced fat loss include the enhancement of lipolysis, inhibition of lipogenesis, and browning of white adipose tissue (WAT). However, few lipid-metabolic enzymes have been reported to be involved in CAC. This study hypothesized that ELOVL6, a critical enzyme for the elongation of fatty acids, may be involved in fat loss in CAC. METHODS Transcriptome sequencing technology was used to identify CAC-related genes in the WAT of a CAC rodent model. Then, the expression level of ELOVL6 and the fatty acid composition were analyzed in a large clinical sample. Elovl6 was knocked down by siRNA in 3T3-L1 mouse preadipocytes to compare with wild-type 3T3-L1 cells treated with tumor cell conditioned medium. RESULTS In the WAT of patients with CAC, a significant decrease in the expression of ELOVL6 was found, which was linearly correlated with the extent of body mass reduction. Gas chromatographic analysis revealed an increase in palmitic acid (C16:0) and a decrease in linoleic acid (C18:2n-6) in these tissue samples. After treatment with tumor cell-conditioned medium, 3T3-L1 mouse preadipocytes showed a decrease in Elovl6 expression, and Elovl6-knockdown cells exhibited a reduction in preadipocyte differentiation and lipogenesis. Similarly, the knockdown of Elovl6 in 3T3-L1 cells resulted in a significant increase in palmitic acid (C16:0) and a marked decrease in oleic acid (C18:1n-9) content. CONCLUSION Overall, the expression of ELOVL6 was decreased in the WAT of CAC patients. Decreased expression of ELOVL6 might induce fat loss in CAC patients by potentially altering the fatty acid composition of adipocytes. These findings suggest that ELOVL6 may be used as a valuable biomarker for the early diagnosis of CAC and may hold promise as a target for future therapies.
Collapse
Affiliation(s)
- Chenyang Jin
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Shuangjie Wang
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Xiangyu Sui
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Qingyang Meng
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China.
- Shanghai Clinical Nutrition Research Centre, Shanghai, China.
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China.
- Shanghai Clinical Nutrition Research Centre, Shanghai, China.
| |
Collapse
|
20
|
Lefevre C, Thibaut MM, Loumaye A, Thissen JP, Neyrinck AM, Navez B, Delzenne NM, Feron O, Bindels LB. Tumoral acidosis promotes adipose tissue depletion by fostering adipocyte lipolysis. Mol Metab 2024; 83:101930. [PMID: 38570069 PMCID: PMC11027574 DOI: 10.1016/j.molmet.2024.101930] [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: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE Tumour progression drives profound alterations in host metabolism, such as adipose tissue depletion, an early event of cancer cachexia. As fatty acid consumption by cancer cells increases upon acidosis of the tumour microenvironment, we reasoned that fatty acids derived from distant adipose lipolysis may sustain tumour fatty acid craving, leading to the adipose tissue loss observed in cancer cachexia. METHODS To evaluate the pro-lipolytic capacities of acid-exposed cancer cells, primary mouse adipocytes from subcutaneous and visceral adipose tissue were exposed to pH-matched conditioned medium from human and murine acid-exposed cancer cells (pH 6.5), compared to naive cancer cells (pH 7.4). To further address the role of tumoral acidosis on adipose tissue loss, a pH-low insertion peptide was injected into tumour-bearing mice, and tumoral acidosis was neutralised with a sodium bicarbonate buffer. Prolipolytic mediators were identified by transcriptomic approaches and validated on murine and human adipocytes. RESULTS Here, we reveal that acid-exposed cancer cells promote lipolysis from subcutaneous and visceral adipocytes and that dampening acidosis in vivo inhibits adipose tissue depletion. We further found a set of well-known prolipolytic factors enhanced upon acidosis adaptation and unravelled a role for β-glucuronidase (GUSB) as a promising new actor in adipocyte lipolysis. CONCLUSIONS Tumoral acidosis promotes the mobilization of fatty acids derived from adipocytes via the release of soluble factors by cancer cells. Our work paves the way for therapeutic approaches aimed at tackling cachexia by targeting the tumour acidic compartment.
Collapse
Affiliation(s)
- Camille Lefevre
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Morgane M Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey Loumaye
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Jean-Paul Thissen
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Department of Endocrinology, Diabetology and Nutrition, IREC, UCLouvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Benoit Navez
- Department of Abdominal Surgery and Transplantation, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
21
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
22
|
Ma D, Qu Y, Wu T, Liu X, Cai L, Wang Y. Excessive fat expenditure in MCT-induced heart failure rats is associated with BMAL1/REV-ERBα circadian rhythmic loop disruption. Sci Rep 2024; 14:8128. [PMID: 38584196 PMCID: PMC10999456 DOI: 10.1038/s41598-024-58577-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/01/2024] [Indexed: 04/09/2024] Open
Abstract
Fat loss predicts adverse outcomes in advanced heart failure (HF). Disrupted circadian clocks are a primary cause of lipid metabolic issues, but it's unclear if this disruption affects fat expenditure in HF. To address this issue, we investigated the effects of disruption of the BMAL1/REV-ERBα circadian rhythmic loop on adipose tissue metabolism in HF.50 Wistar rats were initially divided into control (n = 10) and model (n = 40) groups. The model rats were induced with HF via monocrotaline (MCT) injections, while the control group received equivalent solvent injections. After establishing the HF model, the model group was further subdivided into four groups: normal rhythm (LD), inverted rhythm (DL), lentivirus vector carrying Bmal1 short hairpin RNA (LV-Bmal1 shRNA), and empty lentivirus vector control (LV-Control shRNA) groups, each with 10 rats. The DL subgroup was exposed to a reversed light-dark cycle of 8 h: 16 h (dark: light), while the rest adhered to normal light-dark conditions (light: dark 12 h: 12 h). Histological analyses were conducted using H&E, Oil Red O, and Picrosirius red stains to examine adipose and liver tissues. Immunohistochemical staining, RT-qPCR, and Western blotting were performed to detect markers of lipolysis, lipogenesis, and beiging of white adipose tissue (WAT), while thermogenesis indicators were detected in brown adipose tissue (BAT). The LD group rats exhibited decreased levels of BMAL1 protein, increased levels of REV-ERBα protein, and disrupted circadian circuits in adipose tissue compared to controls. Additionally, HF rats showed reduced adipose mass and increased ectopic lipid deposition, along with smaller adipocytes containing lower lipid content and fibrotic adipose tissue. In the LD group WAT, expression of ATGL, HSL, PKA, and p-PKA proteins increased, alongside elevated mRNA levels of lipase genes (Hsl, Atgl, Peripilin) and FFA β-oxidation genes (Cpt1, acyl-CoA). Conversely, lipogenic gene expression (Scd1, Fas, Mgat, Dgat2) decreased, while beige adipocyte markers (Cd137, Tbx-1, Ucp-1, Zic-1) and UCP-1 protein expression increased. In BAT, HF rats exhibited elevated levels of PKA, p-PKA, and UCP-1 proteins, along with increased expression of thermogenic genes (Ucp-1, Pparγ, Pgc-1α) and lipid transportation genes (Cd36, Fatp-1, Cpt-1). Plasma NT-proBNP levels were higher in LD rats, accompanied by elevated NE and IL-6 levels in adipose tissue. Remarkably, morphologically, the adipocytes in the DL and LV-Bmal1 shRNA groups showed reduced size and lower lipid content, while lipid deposition in the liver was more pronounced in these groups compared to the LD group. At the gene/protein level, the BMAL1/REV-ERBα circadian loop exhibited severe disruption in LV-Bmal1 shRNA rats compared to LD rats. Additionally, there was increased expression of lipase genes, FFA β oxidation genes, and beige adipocyte markers in WAT, as well as higher expression of thermogenic genes and lipid transportation genes in BAT. Furthermore, plasma NT-proBNP levels and adipose tissue levels of NE and IL-6 were elevated in LV-Bmal1 shRNA rats compared with LD rats. The present study demonstrates that disruption of the BMAL1/REV-ERBα circadian rhythmic loop is associated with fat expenditure in HF. This result suggests that restoring circadian rhythms in adipose tissue may help counteract disorders of adipose metabolism and reduce fat loss in HF.
Collapse
Affiliation(s)
- Dufang Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yiwei Qu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Tao Wu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Xue Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Lu Cai
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China
| | - Yong Wang
- Department of Cardiology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, Shandong, China.
| |
Collapse
|
23
|
Liu X, Li S, Cui Q, Guo B, Ding W, Liu J, Quan L, Li X, Xie P, Jin L, Sheng Y, Chen W, Wang K, Zeng F, Qiu Y, Liu C, Zhang Y, Lv F, Hu X, Xiao RP. Activation of GPR81 by lactate drives tumour-induced cachexia. Nat Metab 2024; 6:708-723. [PMID: 38499763 PMCID: PMC11052724 DOI: 10.1038/s42255-024-01011-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Cachexia affects 50-80% of patients with cancer and accounts for 20% of cancer-related death, but the underlying mechanism driving cachexia remains elusive. Here we show that circulating lactate levels positively correlate with the degree of body weight loss in male and female patients suffering from cancer cachexia, as well as in clinically relevant mouse models. Lactate infusion per se is sufficient to trigger a cachectic phenotype in tumour-free mice in a dose-dependent manner. Furthermore, we demonstrate that adipose-specific G-protein-coupled receptor (GPR)81 ablation, similarly to global GPR81 deficiency, ameliorates lactate-induced or tumour-induced adipose and muscle wasting in male mice, revealing adipose GPR81 as the major mediator of the catabolic effects of lactate. Mechanistically, lactate/GPR81-induced cachexia occurs independently of the well-established protein kinase A catabolic pathway, but it is mediated by a signalling cascade sequentially activating Gi-Gβγ-RhoA/ROCK1-p38. These findings highlight the therapeutic potential of targeting GPR81 for the treatment of this life-threatening complication of cancer.
Collapse
Affiliation(s)
- Xidan Liu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Shijin Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Qionghua Cui
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Bujing Guo
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Wanqiu Ding
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jie Liu
- Dazhou Central Hospital, Sichuan, China
| | - Li Quan
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xiaochuan Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Peng Xie
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Li Jin
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Ye Sheng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Wenxin Chen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Kai Wang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | | | - Yifu Qiu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Changlu Liu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Yan Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Fengxiang Lv
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Xinli Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China.
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China.
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China.
| |
Collapse
|
24
|
Bastin J. [Cancer-associated cachexia: an unresolved disease]. Med Sci (Paris) 2024; 40:361-368. [PMID: 38651961 DOI: 10.1051/medsci/2024039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Cachexia is a systemic wasting condition associated to advanced phases of many cancers, which contributes to significant morbidity and mortality. It is mainly characterized by involuntary weight loss due to muscle wasting often associated with loss of adipose tissue, possibly leading to inanition and death, without treatment to date. Symptomatology covers a complex array of disorders (fatigue, inflammation, pain, anorexia, depression) related to multisystemic impairments progressively affecting numerous organs and tissues (muscle, adipose tissue, brain, immune system, gastrointestinal tract). The mechanisms of induction and progression of the disease, still poorly understood, involve inflammatory, metabolic, and neuroendocrine drivers, triggered by a variety of mediators originating from tumor, tumor-host interactions, and inter-organ crosstalk.
Collapse
Affiliation(s)
- Jean Bastin
- Centre de recherche des Cordeliers, Inserm U1138, Sorbonne Université, Université Paris-Cité, Paris, France
| |
Collapse
|
25
|
Kim H, Lee DW, Hwang JK. Curcuma xanthorrhiza extract and xanthorrhizol ameliorate cancer-induced adipose wasting in CT26-bearing mice by regulating lipid metabolism and adipose tissue browning. Integr Med Res 2024; 13:101020. [PMID: 38298864 PMCID: PMC10826318 DOI: 10.1016/j.imr.2023.101020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/03/2023] [Accepted: 12/21/2023] [Indexed: 02/02/2024] Open
Abstract
Background Cancer cachexia-characterized by anorexia, body weight loss, skeletal muscle atrophy, and fat loss-affects nearly 80% of cancer patients and accounts for 20% of cancer deaths. Curcuma xanthorrhiza, known as Java turmeric, and its active compound xanthorrhizol (XAN) exhibit anticancer, anti-inflammatory, and antioxidant properties. However, the ameliorative effects of C. xanthorrhiza extract (CXE) and XAN on cancer-associated adipose atrophy remain unexplored. This study aimed to evaluate the therapeutic effects of CXE and XAN on cancer cachexia-induced adipose tissue wasting in CT26 tumor-bearing mice. Methods CT26 cells were injected subcutaneously into the right flank of BALB/c mice to establish a cancer cachexia model. To evaluate the inhibitory effects of CXE and XAN on cancer cachexia, 50 and 100 mg/kg CXE and 15 mg/kg XAN were administered orally every day for 1 week. Results CXE and XAN administration significantly attenuated the loss of body weight and epidydimal fat mass by cancer cachexia. In epididymal adipose tissues, administration of CXE or XAN inhibited white adipose tissue browning by repressing expression of the thermogenic genes. Simultaneously, CXE or XAN attenuated fat catabolism through the downregulation of lipolytic genes. The administration of CXE or XAN induced the expression of genes associated with adipogenesis and lipogenesis-related genes. Moreover, CXE or XAN treatment was associated with maintaining metabolic homeostasis; regulating the expression of adipokines and AMP-activated protein kinase (AMPK). Conclusions CXE and XAN mitigate cancer-induced adipose tissue atrophy, primarily by modulating lipid metabolism and WAT browning, indicating their therapeutic potential for cachectic cancer patients.
Collapse
Affiliation(s)
- Haeun Kim
- Graduate School of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Dong-Woo Lee
- Graduate School of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jae-Kwan Hwang
- Graduate School of Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Li W, Zhang Z, Berik E, Liu Y, Pei W, Chen S, Wu W, Wang Z, Kong X, Long H, Lei M, Wang JY, Li Z, Liu L, Hou J, Wu W, Guo DA. Energy preservation for skeletal muscles: Shenqi Fuzheng injection prevents tissue wasting and restores bioenergetic profiles in a mouse model of chemotherapy-induced cachexia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155269. [PMID: 38237510 DOI: 10.1016/j.phymed.2023.155269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/28/2023] [Accepted: 12/07/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND Energy deficiency is the characteristic of chemotherapy-induced cachexia (CIC) which is manifested by muscle wasting. glycolysis, tricarboxylic acid (TCA) cycle, and lipid metabolism are central to muscle bioenergy production, which is vulnerable to chemotherapy during cancer treatment. Recent investigations have spotlighted the potential of Shenqi Fuzheng injection (SQ), a Chinese proprietary medicine comprising Radix Codonopsis and Radix Astragali, in alleviating CIC. However, the specific effects of SQ on muscle energy metabolism remains less explored. PURPOSE AND METHODS Here, we integrated transcriptomics, spatial metabolomics, gas chromatography-mass spectrometry targeted quantitative analysis, and transmission electron microscopy techniques, combined with Seahorse live-cell metabolic analysis to reveal the changes in genes and pathways related to energy metabolism in the CIC model and SQ's protective effects at molecular and functional levels. RESULTS Our data showed that chemotherapeutic agents caused glycolysis imbalance, which further leads to metabolic derangements of TCA cycle intermediates. SQ maintained glycolysis balance by facilitating pyruvate fluxing to mitochondria for more efficient bioenergy production, which involved a dual effect on promoting functions of mitochondrial pyruvate dehydrogenase complexes and inhibiting lactate dehydrogenase for lactate production. As a result of the sustained pyruvate level achieved by SQ administration, glycolysis balance was maintained, which further led to the preservation of mitochondrial integrity and function of electron transport chain, thereby, ensuring the normal operation of the TCA cycle and the proper synthesis of adenosine triphosphate (ATP). The above results were further validated using the Seahorse live-cell assay. CONCLUSION In conclusion, our study highlights SQ as a promising strategy for CIC management, emphasizing its ability to harmonize the homeostasis of the muscle bioenergetic profile. Beyond its therapeutic implications, this study also offers a novel perspective for the development of innovative treatments in the realm of herbal medicine.
Collapse
Affiliation(s)
- Wei Li
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijia Zhang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Entezar Berik
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Yawen Liu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenqiang Pei
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sihan Chen
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenyong Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Zhaojun Wang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinqin Kong
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huali Long
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Lei
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jennifer Yiyang Wang
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhaoxia Li
- Beijing Key Lab of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Liangfeng Liu
- Limin Pharmaceutical Factory, Livzon Group Limited, Shaoguan 512028, China; Guangdong Corporate Key Laboratory of High-End Liquid Medicine R&D, Industrilization, Shaoguan 512028, China
| | - Jinjun Hou
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Wanying Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210029, China.
| | - De-An Guo
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
Nakamura Y, Saldajeno DP, Kawaguchi K, Kawaoka S. Progressive, multi-organ, and multi-layered nature of cancer cachexia. Cancer Sci 2024; 115:715-722. [PMID: 38254286 PMCID: PMC10921013 DOI: 10.1111/cas.16078] [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: 10/27/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer cachexia is a complex, multifaceted condition that negatively impacts the health, treatment efficacy, and economic status of cancer patients. The management of cancer cachexia is an essential clinical need. Cancer cachexia is currently defined mainly according to the severity of weight loss and sarcopenia (i.e., macrosymptoms). However, such macrosymptoms may be insufficient to give clinicians clues on how to manage this condition as these symptoms appear at the late stage of cancer. We need to understand earlier events during the progression of cancer cachexia so as not to miss a clinical opportunity to control this complex syndrome. Recent research indicates that cancer-induced changes in the host are much wider than previously recognized, including disruption of liver function and the immune system. Furthermore, such changes are observed before the occurrence of visible distant metastases (i.e., in early, localized cancers). In light of these findings, we propose to expand the definition of cancer cachexia to include all cancer-induced changes to host physiology, including changes caused by early, localized cancers. This new definition of cancer cachexia can provide a new perspective on this topic, which can stimulate the research and development of novel cancer cachexia therapies.
Collapse
Affiliation(s)
- Yuki Nakamura
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Don Pietro Saldajeno
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Mathematical Informatics Laboratory, Division of Information ScienceNara Institute of Science and TechnologyIkomaNaraJapan
| | - Kosuke Kawaguchi
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Shinpei Kawaoka
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer (IDAC)Tohoku UniversitySendaiJapan
| |
Collapse
|
28
|
Li S, Yuan J, Cheng Z, Li Y, Cheng S, Liu X, Huang S, Xu Z, Wu A, Liu L, Dong J. Hsa_circ_0021205 enhances lipolysis via regulating miR-195-5p/HSL axis and drives malignant progression of glioblastoma. Cell Death Discov 2024; 10:71. [PMID: 38341418 DOI: 10.1038/s41420-024-01841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Abnormal lipid metabolism is an essential hallmark of glioblastoma. Hormone sensitive lipase (HSL), an important rate-limiting enzyme contributed to lipolysis, which was involved in aberrant lipolysis of glioblastoma, however, its definite roles and the relevant regulatory pathway have not been fully elucidated. Our investigations disclosed high expression of HSL in glioblastoma. Knock-down of HSL restrained proliferation, migration, and invasion of glioblastoma cells while adding to FAs could significantly rescue the inhibitory effect of si-HSL on tumor cells. Overexpression of HSL further promoted tumor cell proliferation and invasion. Bioinformatics analysis and dual-luciferase reporter assay were performed to predict and verify the regulatory role of ncRNAs on HSL. Mechanistically, hsa_circ_0021205 regulated HSL expression by sponging miR-195-5p, which further promoted lipolysis and drove the malignant progression of glioblastoma. Besides, hsa_circ_0021205/miR-195-5p/HSL axis activated the epithelial-mesenchymal transition (EMT) signaling pathway. These findings suggested that hsa_circ_0021205 promoted tumorigenesis of glioblastoma through regulation of HSL, and targeting hsa_circ_0021205/miR-195-5p/HSL axis can serve as a promising new strategy against glioblastoma.
Collapse
Affiliation(s)
- Suwen Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaqi Yuan
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Zhangjiagang Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Zhe Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, the Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yongdong Li
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shan Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinglei Liu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shilu Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhipeng Xu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Anyi Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Dong
- Department of Neurosurgery, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
29
|
Sun D, Yin H, Liu X, Ding Z, Shen L, Sah S, Han J, Wu G. Elevated 18F-FDG uptake in subcutaneous adipose tissue correlates negatively with nutritional status and prognostic survival in cachexia patients with gastric cancer. Clin Nutr 2024; 43:567-574. [PMID: 38242034 DOI: 10.1016/j.clnu.2024.01.006] [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: 09/21/2023] [Revised: 12/16/2023] [Accepted: 01/04/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Browning of white adipose tissue is a crucial factor contributing to adipose loss in cachexia patients, detectable via 18F-Fluorodeoxyglucose (18F-FDG) uptake. The present study elucidates the clinical relevance of 18F-FDG uptake in the subcutaneous adipose tissue of gastric cancer patients, specifically focusing on adipose browning and its implications on patient clinical parameters and prognosis. METHODS This investigation encompassed 770 gastric cancer patients, with PET-CT imaging and clinical data meticulously combined. The 18F-FDG uptake in subcutaneous adipose tissue at the third lumbar layer was quantified, and its correlation with clinical parameters, particularly those related to nutritional status and fat metabolism, was examined. Kaplan-Meier curves were subsequently employed to probe the relationship between 18F-FDG uptake and overall survival. RESULTS Of the 770 gastric cancer patients, 252 exhibited cancer-associated cachexia, while 518 did not. Cachectic patients demonstrated elevated 18F-FDG uptake in subcutaneous adipose tissue relative to non-cachectic patients (P < 0.001). Increased 18F-FDG uptake was also correlated with reduced plasma concentrations of albumin, prealbumin, hemoglobin, platelets, cholesterol, apolipoprotein A, low-density lipoprotein, and elevated IL-6 concentrations (all P < 0.05). A significant inverse correlation was observed between 18F-FDG uptake and BMI, albumin, low-density lipoprotein, cholesterol, and apolipoprotein A (all P < 0.05). Patients with higher 18F-FDG uptake exhibited diminished overall survival rates compared to those with lower 18F-FDG uptake (P = 0.0065). Furthermore, 18F-FDG uptake in subcutaneous adipose tissue was an independent prognostic indicator in gastric cancer patients (P = 0.028). CONCLUSIONS Browning of subcutaneous adipose tissue was markedly elevated in cachectic gastric cancer patients compared to non-cachectic counterparts. Increased 18F-FDG uptake in subcutaneous adipose tissue in cachectic gastric cancer patients was inversely correlated with nutritional status and survival prognosis.
Collapse
Affiliation(s)
- Diya Sun
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Hongyan Yin
- Department of Nuclear Medicine, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Xiao Liu
- Department of Nursing, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuoyou Ding
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Lei Shen
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Szechun Sah
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Jun Han
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Centre, Shanghai, China.
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital of Fudan University, Shanghai, China; Shanghai Clinical Nutrition Research Centre, Shanghai, China.
| |
Collapse
|
30
|
Diba P, Sattler AL, Korzun T, Habecker BA, Marks DL. Unraveling the lost balance: Adrenergic dysfunction in cancer cachexia. Auton Neurosci 2024; 251:103136. [PMID: 38071925 PMCID: PMC10883135 DOI: 10.1016/j.autneu.2023.103136] [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: 08/21/2023] [Revised: 11/05/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024]
Abstract
Cancer cachexia, characterized by muscle wasting and widespread inflammation, poses a significant challenge for patients with cancer, profoundly impacting both their quality of life and treatment management. However, existing treatment modalities remain very limited, accentuating the necessity for innovative therapeutic interventions. Many recent studies demonstrated that changes in autonomic balance is a key driver of cancer cachexia. This review consolidates research findings from investigations into autonomic dysfunction across cancer cachexia, spanning animal models and patient cohorts. Moreover, we explore therapeutic strategies involving adrenergic receptor modulation through receptor blockers and agonists. Mechanisms underlying adrenergic hyperactivity in cardiac and adipose tissues, influencing tissue remodeling, are also examined. Looking ahead, we present a perspective for future research that delves into autonomic dysregulation in cancer cachexia. This comprehensive review highlights the urgency of advancing research to unveil innovative avenues for combatting cancer cachexia and improving patient well-being.
Collapse
Affiliation(s)
- Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481 Portland, OR 97239, USA
| | - Ariana L Sattler
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481 Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, OR 97201, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR 97201, USA
| | - Tetiana Korzun
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481 Portland, OR 97239, USA
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR 97239, USA; Department of Medicine, Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, SW Sam Jackson Park Rd, Mail Code L481 Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 2720 S Moody Avenue, Portland, OR 97201, USA; Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 2730 S Moody Avenue, Portland, OR 97201, USA.
| |
Collapse
|
31
|
Yang X, Wang J, Chang CY, Zhou F, Liu J, Xu H, Ibrahim M, Gomez M, Guo GL, Liu H, Zong WX, Wondisford FE, Su X, White E, Feng Z, Hu W. Leukemia inhibitory factor suppresses hepatic de novo lipogenesis and induces cachexia in mice. Nat Commun 2024; 15:627. [PMID: 38245529 PMCID: PMC10799847 DOI: 10.1038/s41467-024-44924-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Cancer cachexia is a systemic metabolic syndrome characterized by involuntary weight loss, and muscle and adipose tissue wasting. Mechanisms underlying cachexia remain poorly understood. Leukemia inhibitory factor (LIF), a multi-functional cytokine, has been suggested as a cachexia-inducing factor. In a transgenic mouse model with conditional LIF expression, systemic elevation of LIF induces cachexia. LIF overexpression decreases de novo lipogenesis and disrupts lipid homeostasis in the liver. Liver-specific LIF receptor knockout attenuates LIF-induced cachexia, suggesting that LIF-induced functional changes in the liver contribute to cachexia. Mechanistically, LIF overexpression activates STAT3 to downregulate PPARα, a master regulator of lipid metabolism, leading to the downregulation of a group of PPARα target genes involved in lipogenesis and decreased lipogenesis in the liver. Activating PPARα by fenofibrate, a PPARα agonist, restores lipid homeostasis in the liver and inhibits LIF-induced cachexia. These results provide valuable insights into cachexia, which may help develop strategies to treat cancer cachexia.
Collapse
Affiliation(s)
- Xue Yang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Chun-Yuan Chang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Fan Zhou
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Huiting Xu
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Maria Ibrahim
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Maria Gomez
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
- Department of Veterans Affairs New Jersey Health Care System, East Orange, NJ, USA
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
- Biostatistics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Wei-Xing Zong
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Fredric E Wondisford
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Xiaoyang Su
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
- Metabolomics Core Facility, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
32
|
Tambaro F, Imbimbo G, Ferraro E, Andreini M, Belli R, Amabile MI, Ramaccini C, Lauteri G, Nigri G, Muscaritoli M, Molfino A. Assessment of lipolysis biomarkers in adipose tissue of patients with gastrointestinal cancer. Cancer Metab 2024; 12:1. [PMID: 38167536 PMCID: PMC10762976 DOI: 10.1186/s40170-023-00329-9] [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: 11/29/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Adipose tissue metabolism may be impaired in patients with cancer. In particular, increased lipolysis was described in cancer-promoting adipose tissue atrophy. For this reason, we assessed the expression of the lipolysis-associated genes and proteins in subcutaneous adipose tissue (SAT) of gastrointestinal (GI) cancer patients compared to controls to verify their involvement in cancer, among different types of GI cancers, and in cachexia. METHODS We considered patients with GI cancer (gastric, pancreatic, and colorectal) at their first diagnosis, with/without cachexia, and controls with benign diseases. We collected SAT and total RNA was extracted and ATGL, HSL, PPARα, and MCP1 were analyzed by qRT-PCR. Western blot was performed to evaluate CGI-58, PLIN1 and PLIN5. RESULTS We found higher expression of ATGL and HSL in GI cancer patients with respect to controls (p ≤ 0.008) and a trend of increase for PPARα (p = 0.055). We found an upregulation of ATGL in GI cancer patients with cachexia (p = 0.033) and without cachexia (p = 0.017) vs controls. HSL was higher in patients with cachexia (p = 0.020) and without cachexia (p = 0.021), compared to controls. ATGL was upregulated in gastric cancer vs controls (p = 0.014) and higher HSL was found in gastric (p = 0.008) and in pancreatic cancer (p = 0.033) vs controls. At the protein level, we found higher CGI-58 in cancer vs controls (p = 0.019) and in cachectic vs controls (p = 0.029), as well as in gastric cancer vs controls (p = 0.027). CONCLUSION In our cohort of GI cancer patients, we found a modulation in the expression of genes and proteins involved in lipolysis, and differences were interestingly detected according to cancer type.
Collapse
Affiliation(s)
- Federica Tambaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Martina Andreini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Roberta Belli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Ida Amabile
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Cesarina Ramaccini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulia Lauteri
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Nigri
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
33
|
Bakopoulos D, Golenkina S, Dark C, Christie EL, Sánchez-Sánchez BJ, Stramer BM, Cheng LY. Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model. EMBO Rep 2023; 24:e57695. [PMID: 38014610 DOI: 10.15252/embr.202357695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.
Collapse
Affiliation(s)
- Daniel Bakopoulos
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Brian M Stramer
- Kings College London, Randall Centre for Cell & Molecular Biophysics, London, UK
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
34
|
Kim DH, Song NY, Yim H. Targeting dysregulated lipid metabolism in the tumor microenvironment. Arch Pharm Res 2023; 46:855-881. [PMID: 38060103 PMCID: PMC10725365 DOI: 10.1007/s12272-023-01473-y] [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: 09/27/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
The reprogramming of lipid metabolism and its association with oncogenic signaling pathways within the tumor microenvironment (TME) have emerged as significant hallmarks of cancer. Lipid metabolism is defined as a complex set of molecular processes including lipid uptake, synthesis, transport, and degradation. The dysregulation of lipid metabolism is affected by enzymes and signaling molecules directly or indirectly involved in the lipid metabolic process. Regulation of lipid metabolizing enzymes has been shown to modulate cancer development and to avoid resistance to anticancer drugs in tumors and the TME. Because of this, understanding the metabolic reprogramming associated with oncogenic progression is important to develop strategies for cancer treatment. Recent advances provide insight into fundamental mechanisms and the connections between altered lipid metabolism and tumorigenesis. In this review, we explore alterations to lipid metabolism and the pivotal factors driving lipid metabolic reprogramming, which exacerbate cancer progression. We also shed light on the latest insights and current therapeutic approaches based on small molecular inhibitors and phytochemicals targeting lipid metabolism for cancer treatment. Further investigations are worthwhile to fully understand the underlying mechanisms and the correlation between altered lipid metabolism and carcinogenesis.
Collapse
Affiliation(s)
- Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, 16227, Korea
| | - Na-Young Song
- Department of Applied Life Science, The Graduate School, BK21 Four Project, Yonsei University, Seoul, 03722, Korea
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, 03722, Korea
| | - Hyungshin Yim
- Department of Pharmacy, College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 15588, Korea.
| |
Collapse
|
35
|
Xu W, Li G, Chen Y, Ye X, Song W. A novel antidiuretic hormone governs tumour-induced renal dysfunction. Nature 2023; 624:425-432. [PMID: 38057665 DOI: 10.1038/s41586-023-06833-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Maintenance of renal function and fluid transport are essential for vertebrates and invertebrates to adapt to physiological and pathological challenges. Human patients with malignant tumours frequently develop detrimental renal dysfunction and oliguria, and previous studies suggest the involvement of chemotherapeutic toxicity and tumour-associated inflammation1,2. However, how tumours might directly modulate renal functions remains largely unclear. Here, using conserved tumour models in Drosophila melanogaster3, we characterized isoform F of ion transport peptide (ITPF) as a fly antidiuretic hormone that is secreted by a subset of yki3SA gut tumour cells, impairs renal function and causes severe abdomen bloating and fluid accumulation. Mechanistically, tumour-derived ITPF targets the G-protein-coupled receptor TkR99D in stellate cells of Malpighian tubules-an excretory organ that is equivalent to renal tubules4-to activate nitric oxide synthase-cGMP signalling and inhibit fluid excretion. We further uncovered antidiuretic functions of mammalian neurokinin 3 receptor (NK3R), the homologue of fly TkR99D, as pharmaceutical blockade of NK3R efficiently alleviates renal tubular dysfunction in mice bearing different malignant tumours. Together, our results demonstrate a novel antidiuretic pathway mediating tumour-renal crosstalk across species and offer therapeutic opportunities for the treatment of cancer-associated renal dysfunction.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Gerui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Yuan Chen
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
| | - Wei Song
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
36
|
Dave S, Patel BM. Deliberation on debilitating condition of cancer cachexia: Skeletal muscle wasting. Fundam Clin Pharmacol 2023; 37:1079-1091. [PMID: 37474262 DOI: 10.1111/fcp.12931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Cancer cachexia is a debilitating syndrome associated with marked body loss because of muscular atrophy and fat loss. There are several mechanisms contributing to the pathogenesis of cachexia. The presence of the tumor releases cytokines from inflammatory and immune cells, which play a significant role in activating and deactivating certain pathways associated with protein, carbohydrate, and lipid metabolism. This review focuses on various cascades involving an imbalance between protein synthesis and degradation in the skeletal muscles. OBJECTIVES This study aimed to elucidate the mechanisms involved in skeletal muscle wasting phenomenon over the last few years. METHODS This article briefly overviews different pathways responsible for muscle atrophy in cancer cachexia. Studies published up to April 2023 were included. Important findings and study contributions were chosen and compiled using several databases including PubMed, Google Scholar, Science Direct, and ClinicalTrials.gov using relevant keywords. RESULTS Cancer cachexia is a complex disease involving multiple factors resulting in atrophy of skeletal muscles. Systemic inflammation, altered energy balance and carbohydrate metabolism, altered lipid and protein metabolism, and adipose tissue browning are some of the major culprits in cancer cachexia. Increased protein degradation and decreased protein synthesis lead to muscle atrophy. Changes in signaling pathway like ubiquitin-proteasome, autophagy, mTOR, AMPK, and IGF-1 also lead to muscle wasting. Physical exercise, nutritional supplementation, steroids, myostatin inhibitors, and interventions targeting on inflammation have been investigated to treat cancer cachexia. Some therapy showed positive results in preclinical and clinical settings, although more research on the efficacy and safety of the treatment should be done. CONCLUSION Muscle atrophy in cancer cachexia is the result of multiple complex mechanisms; as a result, a lot more research has been done to describe the pathophysiology of the disease. Targeted therapy and multimodal interventions can improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Srusti Dave
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Bhoomika M Patel
- School of Medico-legal Studies, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
37
|
Mannelli M, Bartoloni B, Cantini G, Nencioni E, Magherini F, Luconi M, Modesti A, Gamberi T, Fiaschi T. STAT3 Signalling Drives LDH Up-Regulation and Adiponectin Down-Regulation in Cachectic Adipocytes. Int J Mol Sci 2023; 24:16343. [PMID: 38003534 PMCID: PMC10671608 DOI: 10.3390/ijms242216343] [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: 10/15/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Cachexia is a devastating pathology that worsens the quality of life and antineoplastic treatment outcomes of oncologic patients. Herein, we report that the secretome from murine colon carcinoma CT26 induces cachectic features in both murine and human adipocytes that are associated with metabolic alterations such as enhanced lactate production and decreased oxygen consumption. The use of oxamate, which inhibits lactate dehydrogenase activity, hinders the effects induced by CT26 secretome. Interestingly, the CT26 secretome elicits an increased level of lactate dehydrogenase and decreased expression of adiponectin. These modifications are driven by the STAT3 signalling cascade since the inhibition of STAT3 with WP1066 impedes the formation of the cachectic condition and the alteration of lactate dehydrogenase and adiponectin levels. Collectively, these findings show that STAT3 is responsible for the altered lactate dehydrogenase and adiponectin levels that, in turn, could participate in the worsening of this pathology and highlight a step forward in the comprehension of the mechanisms underlying the onset of the cachectic condition in adipocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche “M. Serio”, Università degli studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy; (M.M.); (G.C.); (F.M.); (M.L.); (A.M.); (T.G.)
| |
Collapse
|
38
|
Pan X, Zhang G, Wei K, Gu X, Dan J, Zhao Y, Liu X, Cheng C, Zhang X. Carnosol analogue WK-63 alleviated cancer cachexia by inhibiting NF-κB and activating AKT pathways in muscle while inhibiting NF-κB and AMPK pathways in adipocyte. Toxicol Appl Pharmacol 2023; 479:116729. [PMID: 37863360 DOI: 10.1016/j.taap.2023.116729] [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: 06/15/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Cancer cachexia is a systemic metabolic disorder syndrome characterized by severe wasting of muscle and adipose tissues while is lack of effective therapeutic approaches. Carnosol (CS) was found in our previous study to exhibit ameliorating effects on cancer cachexia. In the present study, we designed and synthesized 49 CS analogues by structural modification of CS. Results of activity screening revealed that, among the analogues, WK-63 exhibited better effects than CS in ameliorating atrophy of C2C12 myotubes induced by conditioned medium of C26 tumor cells. WK-63 could also dose-dependently alleviate adipocyte lipolysis of mature 3 T3-L1 cells induced by C26 tumor cell conditioned medium. WK-63 alleviated myotube atrophy by inhibiting Nuclear Factor kappa-B (NF-κB) and activating the Protein Kinase B (AKT) signaling pathway, and also alleviated fat loss by inhibiting NF-κB and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathways. Results of pharmacokinetic (PK) assay showed that, compared with other analogues, WK-63 exhibited longer half-life (T1/2) and mean residence time (MRTs), as well as a larger concentration curve area (AUC0-t). These findings suggested that WK-63 might exert optimal effects in vivo. In the C26 tumor-bearing mice model, administration of WK-63 ameliorated the body weight loss and also improved the weight loss of epididymal adipose tissue. WK-63 is expected to be a novel therapeutic option for the treatment of cancer cachexia.
Collapse
Affiliation(s)
- Xiaojuan Pan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Gang Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Kun Wei
- College of Chemical Engineering, Key Laboratory of Green Chemistry of Sichuan Institutes of Higher Education, Sichuan University of Science & Engineering, Zigong 643000, Sichuan, PR China
| | - Xiaofan Gu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Jiahuan Dan
- College of Chemical Engineering, Key Laboratory of Green Chemistry of Sichuan Institutes of Higher Education, Sichuan University of Science & Engineering, Zigong 643000, Sichuan, PR China
| | - Yun Zhao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201003, China.
| | - Chunru Cheng
- College of Chemical Engineering, Key Laboratory of Green Chemistry of Sichuan Institutes of Higher Education, Sichuan University of Science & Engineering, Zigong 643000, Sichuan, PR China.
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China.
| |
Collapse
|
39
|
Machado H, Hofer P, Zechner R, Smith TK, Figueiredo LM. Adipocyte lipolysis protects mice against Trypanosoma brucei infection. Nat Microbiol 2023; 8:2020-2032. [PMID: 37828246 PMCID: PMC10627827 DOI: 10.1038/s41564-023-01496-7] [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] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 09/11/2023] [Indexed: 10/14/2023]
Abstract
Trypanosoma brucei causes African trypanosomiasis, colonizing adipose tissue and inducing weight loss. Here we investigated the molecular mechanisms responsible for adipose mass loss and its impact on disease pathology. We found that lipolysis is activated early in infection. Mice lacking B and T lymphocytes fail to upregulate adipocyte lipolysis, resulting in higher fat mass retention. Genetic ablation of the rate-limiting adipose triglyceride lipase specifically from adipocytes (AdipoqCre/+-Atglfl/fl) prevented the stimulation of adipocyte lipolysis during infection, reducing fat mass loss. Surprisingly, these mice succumbed earlier and presented a higher parasite burden in the gonadal adipose tissue, indicating that host lipolysis limits parasite growth. Consistently, free fatty acids comparable with those of adipose interstitial fluid induced loss of parasite viability. Adipocyte lipolysis emerges as a mechanism controlling local parasite burden and affecting the loss of fat mass in African trypanosomiasis.
Collapse
Affiliation(s)
- Henrique Machado
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Peter Hofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Terry K Smith
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Luísa M Figueiredo
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
40
|
Affiliation(s)
- Brittany R Counts
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Teresa A Zimmers
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA.
- Knight Cancer Institute, Portland, OR, USA.
- Portland Veterans Administration Medical Center, Portland, OR, USA.
| |
Collapse
|
41
|
Song Y, Bae MI, Han DW, Park EJ, Park S, Ham SY. Prognostic role of body composition in peritoneal carcinomatosis patients undergoing cytoreduction and hyperthermic intraperitoneal chemotherapy. World J Surg Oncol 2023; 21:345. [PMID: 37891626 PMCID: PMC10604686 DOI: 10.1186/s12957-023-03233-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Bioelectric impedance analysis (BIA)-measured body composition and nutritional status have been used as prognostic indicators in various cancer cohorts. This study investigated whether BIA could provide information on prognosis in peritoneal carcinomatosis patients undergoing cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). METHODS We retrospectively analyzed the data of 99 patients with preoperative BIA data among those who underwent CRS and HIPEC. The association between BIA-derived parameters and intraoperative peritoneal cancer index (PCI) score was assessed. Predictive analysis for the occurrence of postoperative morbidities including major complications (Clavien-Dindo classification 3-4) and re-admission within 30 days after surgery as well as 1 year mortality was also performed. RESULTS BIA-derived mineral (r = 0.224, p = 0.027), fat (r = - 0.202, p = 0.048), and total body water (TBW)/fat-free mass (FFM) (r = - 0.280, p = 0.005) showed significant associations with intraoperative PCI score. Lower TBW/FFM was an independent predictor of major postoperative complications (OR 0.047, 95% CI 0.003-0.749, p = 0.031) and re-admission (OR 0.094, 95% CI 0.014-0.657, p = 0.017) within 30 days after surgery. Higher fat mass was also independently associated with a higher risk of major postoperative complications (OR 1.120, 95% CI 1.006-1.248, p = 0.039) and re-admission (OR 1.123, 95% CI 1.024-1.230, p = 0.013). Intraoperative PCI score > 20 (OR 4.489, 95% CI 1.191-16.917, p = 0.027) and re-admission within 30 days after surgery (OR 5.269, 95% CI 1.288-21.547, p = 0.021) independently predicted postoperative 1-year mortality. CONCLUSIONS We demonstrate that preoperative BIA-derived TBW/FFM and fat mass were significantly correlated with metastatic extent, assessed by PCI score, in patients with peritoneal carcinomatosis. In addition, BIA-derived TBW/FFM and fat mass showed independent predictability for postoperative 30-day major complications and re-admission in patients undergoing CRS and HIPEC. Our findings suggest that assessment of BIA may improve discrete risk stratification in patients who are planned to receive CRS and HIPEC.
Collapse
Affiliation(s)
- Young Song
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-Gu, 06273, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myung Il Bae
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-Gu, 06273, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong Woo Han
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-Gu, 06273, Seoul, Republic of Korea
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Park
- Division of Colon and Rectal Surgery, Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sujung Park
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-Gu, 06273, Seoul, Republic of Korea
| | - Sung Yeon Ham
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-Gu, 06273, Seoul, Republic of Korea.
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Sciarretta F, Ceci V, Tiberi M, Zaccaria F, Li H, Zhou ZY, Sun Q, Konja D, Matteocci A, Bhusal A, Verri M, Fresegna D, Balletta S, Ninni A, Di Biagio C, Rosina M, Suk K, Centonze D, Wang Y, Chiurchiù V, Aquilano K, Lettieri-Barbato D. Lipocalin-2 promotes adipose-macrophage interactions to shape peripheral and central inflammatory responses in experimental autoimmune encephalomyelitis. Mol Metab 2023; 76:101783. [PMID: 37517520 PMCID: PMC10448472 DOI: 10.1016/j.molmet.2023.101783] [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: 05/26/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023] Open
Abstract
OBJECTIVE Accumulating evidence suggests that dysfunctional adipose tissue (AT) plays a major role in the risk of developing multiple sclerosis (MS), the most common immune-mediated and demyelinating disease of the central nervous system. However, the contribution of adipose tissue to the etiology and progression of MS is still obscure. This study aimed at deciphering the responses of AT in experimental autoimmune encephalomyelitis (EAE), the best characterized animal model of MS. RESULTS AND METHODS We observed a significant AT loss in EAE mice at the onset of disease, with a significant infiltration of M1-like macrophages and fibrosis in the AT, resembling a cachectic phenotype. Through an integrative and multilayered approach, we identified lipocalin2 (LCN2) as the key molecule released by dysfunctional adipocytes through redox-dependent mechanism. Adipose-derived LCN2 shapes the pro-inflammatory macrophage phenotype, and the genetic deficiency of LCN2 specifically in AT reduced weight loss as well as inflammatory macrophage infiltration in spinal cord in EAE mice. Mature adipocytes downregulating LCN2 reduced lipolytic response to inflammatory stimuli (e.g. TNFα) through an ATGL-mediated mechanism. CONCLUSIONS Overall data highlighted a role LCN2 in exacerbating inflammatory phenotype in EAE model, suggesting a pathogenic role of dysfunctional AT in MS.
Collapse
Affiliation(s)
| | - Veronica Ceci
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marta Tiberi
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Fabio Zaccaria
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Haoyun Li
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhong-Yan Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiyang Sun
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Martina Verri
- Pathology Unit, University Hospital Campus Bio-Medico of Rome, 00128 Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00163 Rome, Italy
| | - Sara Balletta
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Andrea Ninni
- PhD Program in Evolutionary Biology and Ecology, Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Claudia Di Biagio
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marco Rosina
- Neurology Unit, Fondazione PTV Policlinico Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; Unit of Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, IRCCS Santa Lucia Foundation, 00179 Rome, Italy; Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daniele Lettieri-Barbato
- IRCCS, Fondazione Santa Lucia, 00179 Rome, Italy; Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
43
|
McLelland GL, Lopez-Osias M, Verzijl CRC, Ellenbroek BD, Oliveira RA, Boon NJ, Dekker M, van den Hengel LG, Ali R, Janssen H, Song JY, Krimpenfort P, van Zutphen T, Jonker JW, Brummelkamp TR. Identification of an alternative triglyceride biosynthesis pathway. Nature 2023; 621:171-178. [PMID: 37648867 PMCID: PMC10482677 DOI: 10.1038/s41586-023-06497-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Triacylglycerols (TAGs) are the main source of stored energy in the body, providing an important substrate pool for mitochondrial beta-oxidation. Imbalances in the amount of TAGs are associated with obesity, cardiac disease and various other pathologies1,2. In humans, TAGs are synthesized from excess, coenzyme A-conjugated fatty acids by diacylglycerol O-acyltransferases (DGAT1 and DGAT2)3. In other organisms, this activity is complemented by additional enzymes4, but whether such alternative pathways exist in humans remains unknown. Here we disrupt the DGAT pathway in haploid human cells and use iterative genetics to reveal an unrelated TAG-synthesizing system composed of a protein we called DIESL (also known as TMEM68, an acyltransferase of previously unknown function) and its regulator TMX1. Mechanistically, TMX1 binds to and controls DIESL at the endoplasmic reticulum, and loss of TMX1 leads to the unconstrained formation of DIESL-dependent lipid droplets. DIESL is an autonomous TAG synthase, and expression of human DIESL in Escherichia coli endows this organism with the ability to synthesize TAG. Although both DIESL and the DGATs function as diacylglycerol acyltransferases, they contribute to the cellular TAG pool under specific conditions. Functionally, DIESL synthesizes TAG at the expense of membrane phospholipids and maintains mitochondrial function during periods of extracellular lipid starvation. In mice, DIESL deficiency impedes rapid postnatal growth and affects energy homeostasis during changes in nutrient availability. We have therefore identified an alternative TAG biosynthetic pathway driven by DIESL under potent control by TMX1.
Collapse
Affiliation(s)
- Gian-Luca McLelland
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Marta Lopez-Osias
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cristy R C Verzijl
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Brecht D Ellenbroek
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rafaela A Oliveira
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nicolaas J Boon
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marleen Dekker
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lisa G van den Hengel
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rahmen Ali
- Animal Modeling Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Hans Janssen
- Electron Microscope Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Animal Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Paul Krimpenfort
- Animal Modeling Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tim van Zutphen
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Faculty Campus Fryslân, University of Groningen, Leeuwarden, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Thijn R Brummelkamp
- Oncode Institute, Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Tichy L, Parry TL. The pathophysiology of cancer-mediated cardiac cachexia and novel treatment strategies: A narrative review. Cancer Med 2023; 12:17706-17717. [PMID: 37654192 PMCID: PMC10524052 DOI: 10.1002/cam4.6388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/15/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
SIGNIFICANCE Two of the leading causes of death worldwide are cancer and cardiovascular diseases. Most cancer patients suffer from a metabolic wasting syndrome known as cancer-induced cardiac cachexia, resulting in death in up to 30% of cancer patients. Main symptoms of this disease are severe cardiac muscle wasting, cardiac remodeling, and cardiac dysfunction. Metabolic alterations, increased inflammation, and imbalance of protein homeostasis contribute to the progression of this multifactorial syndrome, ultimately resulting in heart failure and death. Cancer-induced cardiac cachexia is associated with decreased quality of life, increased fatiguability, and decreased tolerance to therapeutic interventions. RECENT ADVANCES While molecular mechanisms of this disease are not fully understood, researchers have identified different stages of progression of this disease, as well as potential biomarkers to detect and monitor the development. Preclinical and clinical studies have shown positive results when implementing certain pharmacological and non-pharmacological therapy interventions. CRITICAL ISSUES There are still no clear diagnostic criteria for cancer-mediated cardiac cachexia and the condition remains untreated, leaving cancer patients with irreversible effects of this syndrome. While traditional cardiovascular therapy interventions, such as beta-blockers, have shown some positive results in preclinical and clinical research studies, recent preclinical studies have shown more successful results with certain non-traditional treatment options that have not been further evaluated yet. There is still no clinical standard of care or approved FDA drug to aid in the prevention or treatment of cancer-induced cardiac cachexia. This review aims to revisit the still not fully understood pathophysiological mechanisms of cancer-induced cardiac cachexia and explore recent studies using novel treatment strategies. FUTURE DIRECTIONS While research has progressed, further investigations might provide novel diagnostic techniques, potential biomarkers to monitor the progression of the disease, as well as viable pharmacological and non-pharmacological treatment options to increase quality of life and reduce cancer-induced cardiac cachexia-related mortality.
Collapse
Affiliation(s)
- Louisa Tichy
- Department of KinesiologyUniversity of North Carolina GreensboroGreensboroNorth CarolinaUSA
| | - Traci L. Parry
- Department of KinesiologyUniversity of North Carolina GreensboroGreensboroNorth CarolinaUSA
| |
Collapse
|
45
|
Zhou Y, Lu R, Lin F, Chen S, He QQ, Wu G, Huang C, Lin D. Exploring the Therapeutic Potential of Ethyl 3-Hydroxybutyrate in Alleviating Skeletal Muscle Wasting in Cancer Cachexia. Biomolecules 2023; 13:1330. [PMID: 37759730 PMCID: PMC10527383 DOI: 10.3390/biom13091330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Cachexia (CAC) is a debilitating metabolic syndrome. Although dietary interventions are attractive, long-term adherence to specific diets is difficult to maintain and can lead to systemic side effects. Ethyl 3-hydroxybutyrate (EHB) is a commonly used food additive found in wine and Tribolium castaneum. In this study, we investigated the effects of EHB administration in cachectic mice. After a single intraperitoneal injection of EHB into mice, 3-hydroxybutyrate (3-HB) levels were significantly increased in the serum and gastrocnemius of mice. The administration of EHB alleviated cachexia-related symptoms, ameliorated skeletal muscle atrophy, and improved survival in cachectic mice. In addition, the supplementation of cachectic mice with 3-HB by EHB administration significantly reduced tumor weights, indicating the anti-tumor effects of 3-HB. Remarkably, the addition of 3-HB to the culture medium significantly attenuated the C2C12 myotube atrophy induced by the culture supernatant of CT26 cell lines, highlighting its potential to counteract the destructive effects of tumor-derived elements on muscle tissue. NMR-based metabolomics analysis provided insights into the underlying mechanisms and revealed that the anti-cachexia effects of 3-HB treatment can be attributed to three key mechanisms: the promotion of the TCA cycle and the attenuation of proteolysis, the promotion of protein synthesis and the improvement of metabolic homeostasis, and a reduction in inflammation and an enhancement of the antioxidant capacity. This study provided compelling evidence for the protective effects of 3-HB treatment on the cachectic gastrocnemius and highlighted the efficacy of EHB administration as a ketone supplementation approach to achieve nutritional ketosis without the need for dietary restriction.
Collapse
Affiliation(s)
- Yu Zhou
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.Z.); (R.L.)
| | - Ruohan Lu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.Z.); (R.L.)
| | - Fusheng Lin
- Department of General Surgery, Zhongshan Hospital, Xiamen University, Xiamen 361005, China;
| | - Shu Chen
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.Z.); (R.L.)
| | - Qi-Qing He
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.Z.); (R.L.)
| | - Guoyang Wu
- Department of General Surgery, Zhongshan Hospital, Xiamen University, Xiamen 361005, China;
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361005, China;
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China; (Y.Z.); (R.L.)
| |
Collapse
|
46
|
Richter FC, Bergthaler A. You only waste twice: T cells in parasite-induced organ wasting. Cell Rep 2023; 42:112829. [PMID: 37490906 DOI: 10.1016/j.celrep.2023.112829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023] Open
Abstract
In this issue of Cell Reports, Redford et al.1 uncouple the role of CD4+ and CD8+ T cells in controlling anorexia and wasting of muscle and adipose tissue during chronic parasitic infections. These results shed light on the impact of adaptive immune cells on organ catabolism.
Collapse
Affiliation(s)
- Felix Clemens Richter
- Institute of Hygiene and Applied Immunology, Department of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Andreas Bergthaler
- Institute of Hygiene and Applied Immunology, Department of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna 1090, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| |
Collapse
|
47
|
Redford SE, Varanasi SK, Sanchez KK, Thorup NR, Ayres JS. CD4+ T cells regulate sickness-induced anorexia and fat wasting during a chronic parasitic infection. Cell Rep 2023; 42:112814. [PMID: 37490905 DOI: 10.1016/j.celrep.2023.112814] [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: 12/18/2022] [Revised: 05/14/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023] Open
Abstract
Infections cause catabolism of fat and muscle stores. Traditionally, studies have focused on understanding how the innate immune system contributes to energy stores wasting, while the role of the adaptive immune system remains elusive. In the present study, we examine the role of the adaptive immune response in adipose tissue wasting and cachexia using a murine model of the chronic parasitic infection Trypanosoma brucei, the causative agent of sleeping sickness. We find that the wasting response occurs in two phases, with the first stage involving fat wasting caused by CD4+ T cell-induced anorexia and a second anorexia-independent cachectic stage that is dependent on CD8+ T cells. Fat wasting has no impact on host antibody-mediated resistance defenses or survival, while later-stage muscle wasting contributes to disease-tolerance defenses. Our work reveals a decoupling of adaptive immune-mediated resistance from the catabolic response during infection.
Collapse
Affiliation(s)
- Samuel E Redford
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA; Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Karina K Sanchez
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA; Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Natalia R Thorup
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA; Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Janelle S Ayres
- Molecular and Systems Physiology Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA; Gene Expression Lab, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
48
|
Saavedra P, Dumesic PA, Hu Y, Filine E, Jouandin P, Binari R, Wilensky SE, Rodiger J, Wang H, Chen W, Liu Y, Spiegelman BM, Perrimon N. REPTOR and CREBRF encode key regulators of muscle energy metabolism. Nat Commun 2023; 14:4943. [PMID: 37582831 PMCID: PMC10427696 DOI: 10.1038/s41467-023-40595-1] [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: 03/03/2022] [Accepted: 08/03/2023] [Indexed: 08/17/2023] Open
Abstract
Metabolic flexibility of muscle tissue describes the adaptive capacity to use different energy substrates according to their availability. The disruption of this ability associates with metabolic disease. Here, using a Drosophila model of systemic metabolic dysfunction triggered by yorkie-induced gut tumors, we show that the transcription factor REPTOR is an important regulator of energy metabolism in muscles. We present evidence that REPTOR is activated in muscles of adult flies with gut yorkie-tumors, where it modulates glucose metabolism. Further, in vivo studies indicate that sustained activity of REPTOR is sufficient in wildtype muscles to repress glycolysis and increase tricarboxylic acid (TCA) cycle metabolites. Consistent with the fly studies, higher levels of CREBRF, the mammalian ortholog of REPTOR, reduce glycolysis in mouse myotubes while promoting oxidative metabolism. Altogether, our results define a conserved function for REPTOR and CREBRF as key regulators of muscle energy metabolism.
Collapse
Affiliation(s)
- Pedro Saavedra
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Patrick Jouandin
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Montpellier, France
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Jonathan Rodiger
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Weihang Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
49
|
Kim KH, Jia Z, Snyder M, Chen J, Qiu J, Oprescu SN, Chen X, Syed SA, Yue F, Roseguini BT, Imbalzano AN, Hu C, Kuang S. PRMT5 links lipid metabolism to contractile function of skeletal muscles. EMBO Rep 2023; 24:e57306. [PMID: 37334900 PMCID: PMC10398672 DOI: 10.15252/embr.202357306] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Skeletal muscle plays a key role in systemic energy homeostasis besides its contractile function, but what links these functions is poorly defined. Protein Arginine Methyl Transferase 5 (PRMT5) is a well-known oncoprotein but also expressed in healthy tissues with unclear physiological functions. As adult muscles express high levels of Prmt5, we generated skeletal muscle-specific Prmt5 knockout (Prmt5MKO ) mice. We observe reduced muscle mass, oxidative capacity, force production, and exercise performance in Prmt5MKO mice. The motor deficiency is associated with scarce lipid droplets in myofibers due to defects in lipid biosynthesis and accelerated degradation. Specifically, PRMT5 deletion reduces dimethylation and stability of Sterol Regulatory Element-Binding Transcription Factor 1a (SREBP1a), a master regulator of de novo lipogenesis. Moreover, Prmt5MKO impairs the repressive H4R3 symmetric dimethylation at the Pnpla2 promoter, elevating the level of its encoded protein ATGL, the rate-limiting enzyme catalyzing lipolysis. Accordingly, skeletal muscle-specific double knockout of Pnpla2 and Prmt5 normalizes muscle mass and function. Together, our findings delineate a physiological function of PRMT5 in linking lipid metabolism to contractile function of myofibers.
Collapse
Affiliation(s)
- Kun Ho Kim
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Zhihao Jia
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Madigan Snyder
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
| | - Jingjuan Chen
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Jiamin Qiu
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Stephanie N Oprescu
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
| | - Xiyue Chen
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Sabriya A Syed
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Feng Yue
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Bruno T Roseguini
- Department of Health and KinesiologyPurdue UniversityWest LafayetteINUSA
| | - Anthony N Imbalzano
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Changdeng Hu
- Department of Medicinal Chemistry and Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
- Center for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| | - Shihuan Kuang
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Center for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
50
|
Ji H, Englmaier F, Morigny P, Giroud M, Gräsle P, Brings S, Szendrödi J, Berriel Diaz M, Plettenburg O, Herzig S, Rohm M. Development of a peptide drug restoring AMPK and adipose tissue functionality in cancer cachexia. Mol Ther 2023; 31:2408-2421. [PMID: 37408309 PMCID: PMC10422018 DOI: 10.1016/j.ymthe.2023.06.020] [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: 03/28/2023] [Revised: 06/01/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023] Open
Abstract
Cancer cachexia is a severe systemic wasting disease that negatively affects quality of life and survival in patients with cancer. To date, treating cancer cachexia is still a major unmet clinical need. We recently discovered the destabilization of the AMP-activated protein kinase (AMPK) complex in adipose tissue as a key event in cachexia-related adipose tissue dysfunction and developed an adeno-associated virus (AAV)-based approach to prevent AMPK degradation and prolong cachexia-free survival. Here, we show the development and optimization of a prototypic peptide, Pen-X-ACIP, where the AMPK-stabilizing peptide ACIP is fused to the cell-penetrating peptide moiety penetratin via a propargylic glycine linker to enable late-stage functionalization using click chemistry. Pen-X-ACIP was efficiently taken up by adipocytes, inhibited lipolysis, and restored AMPK signaling. Tissue uptake assays showed a favorable uptake profile into adipose tissue upon intraperitoneal injection. Systemic delivery of Pen-X-ACIP into tumor-bearing animals prevented the progression of cancer cachexia without affecting tumor growth and preserved body weight and adipose tissue mass with no discernable side effects in other peripheral organs, thereby achieving proof of concept. As Pen-X-ACIP also exerted its anti-lipolytic activity in human adipocytes, it now provides a promising platform for further (pre)clinical development toward a novel, first-in-class approach against cancer cachexia.
Collapse
Affiliation(s)
- Honglei Ji
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Felix Englmaier
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute of Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Institute of Organic Chemistry, Center of Biomolecular Research, Leibniz University Hannover, 30167 Hannover, Germany
| | - Pauline Morigny
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Maude Giroud
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Pamina Gräsle
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Sebastian Brings
- Department of Internal Medicine I and Clinical Chemistry, Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; Department of Nuclear Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Julia Szendrödi
- Department of Internal Medicine I and Clinical Chemistry, Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Oliver Plettenburg
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Institute of Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Institute of Organic Chemistry, Center of Biomolecular Research, Leibniz University Hannover, 30167 Hannover, Germany; Laboratory of Nano- and Quantum Engineering (LNQE), Leibniz University Hannover, 30167 Hanover, Germany; Institute of Lung Health (ILH), 35392 Gießen, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany; Chair Molecular Metabolic Control, Technical University Munich, 81675 Munich, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Diabetes Center, Helmholtz Center Munich, 85764 Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Unit, Heidelberg University Hospital, 69120 Heidelberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.
| |
Collapse
|