1
|
Yan B, Fritsche AK, Haußner E, Inamdar TV, Laumen H, Boettcher M, Gericke M, Michl P, Rosendahl J. From Genes to Environment: Elucidating Pancreatic Carcinogenesis Through Genetically Engineered and Risk Factor-Integrated Mouse Models. Cancers (Basel) 2025; 17:1676. [PMID: 40427173 DOI: 10.3390/cancers17101676] [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: 03/28/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic cancer is characterized by late diagnosis, therapy resistance, and poor prognosis, necessitating the exploration of early carcinogenesis and prevention methods. Preclinical mouse models have evolved from cell line-based to human tumor tissue- or organoid-derived xenografts, now to humanized mouse models and genetically engineered mouse models (GEMMs). GEMMs, primarily driven by oncogenic Kras mutations and tumor suppressor gene alterations, offer a realistic platform for investigating pancreatic cancer initiation, progression, and metastasis. The incorporation of inducible somatic mutations and CRISPR-Cas9 screening methods has expanded their utility. To better recapitulate tumor initiation triggered by inflammatory cues, common pancreatic risk factors are being integrated into model designs. This approach aims to decipher the role of environmental factors as secondary or parallel triggers of tumor initiation alongside oncogenic burdens. Emerging models exploring pancreatitis, obesity, diabetes, and other risk factors offer significant translational potential. This review describes current mouse models for studying pancreatic carcinogenesis, their combination with inflammatory factors, and their utility in evaluating pathogenesis, providing guidance for selecting the most suitable models for pancreatic cancer research.
Collapse
Affiliation(s)
- Bin Yan
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Erik Haußner
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Tanvi Vikrant Inamdar
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Helmut Laumen
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Michael Boettcher
- Institute of Molecular Medicine, Section for Molecular Medicine of Signal Transduction, Faculty of Medicine, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Patrick Michl
- Department of Internal Medicine IV, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Jonas Rosendahl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
2
|
Lin K, Wei L, Wang R, Li L, Song S, Wang F, He M, Pu W, Wang J, Wazir J, Cao W, Yang X, Treuter E, Fan R, Wang Y, Huang Z, Wang H. Disrupted methionine cycle triggers muscle atrophy in cancer cachexia through epigenetic regulation of REDD1. Cell Metab 2025; 37:460-476.e8. [PMID: 39729999 DOI: 10.1016/j.cmet.2024.10.017] [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: 04/18/2024] [Revised: 09/19/2024] [Accepted: 10/19/2024] [Indexed: 12/29/2024]
Abstract
The essential amino acid methionine plays a pivotal role in one-carbon metabolism, facilitating the production of S-adenosylmethionine (SAM), a critical supplier for DNA methylation and thereby a modulator of gene expression. Here, we report that the methionine cycle is disrupted in skeletal muscle during cancer cachexia, leading to endoplasmic reticulum stress and DNA hypomethylation-induced expression of the DNA damage inducible transcript 4 (Ddit4) gene, encoding the regulated in development and DNA damage response 1 (REDD1) protein. Targeting DNA methylation by depletion or pharmacological inhibition of DNA methyltransferase 3A (DNMT3A) exacerbates cachexia, while restoring DNMT3A expression or REDD1 knockout alleviates cancer cachexia-induced skeletal muscle atrophy in mice. Methionine supplementation restores DNA methylation of the Ddit4 promoter in a DNMT3A-dependent manner, thereby inhibiting activating transcription factor 4 (ATF4)-mediated Ddit4 transcription. Thus, with the identification of the methionine/SAM-DNMT3A/DNA hypomethylation-Ddit4/REDD1 axis, our study provides molecular insights into an epigenetic mechanism underlying cancer cachexia, and it suggests nutrient supplementation as a promising therapeutic strategy to prevent or reverse cachectic muscle atrophy.
Collapse
Affiliation(s)
- Kai Lin
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China; Department of Pathology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an 223399, China
| | - Ranran Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Shiyu Song
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China; Nanjing Lupine (YuShanDou) Biomedical Research Institute Co. Ltd, Nanjing 210046, China
| | - Fei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Meiwei He
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210093, Jiangsu, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Wangsen Cao
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China
| | - Xiaozhong Yang
- Department of Gastroenterology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an 223399, China
| | - Eckardt Treuter
- Department of Medicine Huddinge, Biosciences and Nutrition Unit, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Rongrong Fan
- Department of Medicine Huddinge, Biosciences and Nutrition Unit, Karolinska Institutet, 14183 Huddinge, Sweden
| | - Yongxiang Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, 98 West Nantong Road, Yangzhou 225001, China.
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
3
|
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
|
4
|
Morena F, Cabrera AR, Jones RG, Schrems ER, Muhyudin R, Washington TA, Murach KA, Greene NP. Transcriptional analysis of cancer cachexia: conserved and unique features across preclinical models and biological sex. Am J Physiol Cell Physiol 2024; 327:C1514-C1531. [PMID: 39466180 PMCID: PMC11684872 DOI: 10.1152/ajpcell.00647.2024] [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: 09/04/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Studies suggest heterogeneity in cancer cachexia (CC) among models and biological sexes, yet examinations comparing models and sexes are scarce. We compared the transcriptional landscape of skeletal muscle across murine CC models and biological sexes during early and late CC. Global gene expression analyses were performed on gastrocnemius [Lewis lung carcinoma (LLC)], quadriceps (KPC-pancreatic), and tibialis anterior [Colon-26 (C26)-colorectal and ApcMin/+] muscles across biological sexes. Differentially expressed genes (DEGs) were identified using an adj-P value of <0.05, followed by pathway and computational cistrome analyses. Integrating all controls, early and late stages of all models and sexes revealed up to 68% of DEGs and pathways were enriched at early and late CC, indicating a conserved transcriptional profile during CC development. Comparing DEGs and pathways within sexes and across models, in early CC, the transcriptional response was highly heterogeneous. At late stage, 11.5% of upregulated and 10% of downregulated genes were shared between models in males, whereas 18.9% of upregulated and 7% of downregulated DEGs were shared in females. Shared DEGs were enriched in proteasome and mitophagy/autophagy pathways (upregulated), and downregulation of energy metabolism pathways in males only. Between sexes, though the proportion of shared DEGs was low (<16%), similar pathway enrichment was observed, including proteasome and mitophagy at late-stage CC. In early CC, oncostatin M receptor (Osmr) upregulation was the only commonality across all models and sexes, whereas CLOCK and ARNTL/BMAL1 were predicted transcriptional factors associated with dysregulations in all three male models. This study highlights sex and model differences in CC progression and suggests conserved transcriptional changes as potential therapeutic targets.NEW & NOTEWORTHY This study is among the first to integrate and compare the skeletal muscle transcriptional landscape across multiple preclinical models and biological sexes. We highlight that 1) early CC transcriptional changes are two-thirds conserved at late stages, 2) DEGs are largely model and sex specific, and 3) transcriptional factors including CLOCK and ARNTL/BMAL1, which influence early CC gene expression, might represent a global therapeutic target with a chance of efficacy across various cancer types.
Collapse
Affiliation(s)
- Francielly Morena
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ronald G Jones
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ruqaiza Muhyudin
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Kevin A Murach
- Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
5
|
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
|
6
|
Snoke DB, van der Velden JL, Bellafleur ER, Dearborn JS, Lenahan SM, Heininger SCJ, Ather JL, Sarausky H, Stephenson D, Reisz JA, D'Alessandro A, Majumdar D, Ahern TP, Sandler KL, Landman BA, Janssen-Heininger YMW, Poynter ME, Seward DJ, Toth MJ. Early adipose tissue wasting in a novel preclinical model of human lung cancer cachexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615385. [PMID: 39651308 PMCID: PMC11623500 DOI: 10.1101/2024.09.27.615385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Cancer cachexia (CC), a syndrome of skeletal muscle and adipose tissue wasting, reduces responsiveness to therapies and increases mortality. There are no approved treatments for CC, which may relate to discordance between pre-clinical models and human CC. To address the need for clinically relevant models, we generated tamoxifen-inducible, epithelial cell specific Kras G12D/+ ( G12D ) mice. G12D mice develop CC over a protracted time course and phenocopy tissue, cellular, mutational, transcriptomic, and metabolic characteristics of human lung CC. CC in G12D mice is characterized by early loss of adipose tissue, a phenotype confirmed in a large cohort of patients with lung cancer. Tumor-released factors promote adipocyte lipolysis, a driver of adipose wasting in human CC, and adipose tissue wasting was inversely related to tumor burden. Thus, G12D mice model key features of human lung CC and suggest a novel role for early adipose tissue wasting in CC.
Collapse
|
7
|
Pryce BR, Oles A, Talbert EE, Romeo MJ, Vaena S, Sharma S, Spadafora V, Tolliver L, Mahvi DA, Morgan KA, Lancaster WP, Beal E, Koren N, Watts B, Overstreet M, Berto S, Subramanian S, Calisir K, Crawford A, Neelon B, Ostrowski MC, Zimmers TA, Tidball JG, Wang DJ, Guttridge DC. Muscle inflammation is regulated by NF-κB from multiple cells to control distinct states of wasting in cancer cachexia. Cell Rep 2024; 43:114925. [PMID: 39475511 PMCID: PMC11774514 DOI: 10.1016/j.celrep.2024.114925] [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: 10/14/2023] [Revised: 07/01/2024] [Accepted: 10/14/2024] [Indexed: 12/01/2024] Open
Abstract
Although cancer cachexia is classically characterized as a systemic inflammatory disorder, emerging evidence indicates that weight loss also associates with local tissue inflammation. We queried the regulation of this inflammation and its causality to cachexia by exploring skeletal muscle, whose atrophy strongly associates with poor outcomes. Using multiple mouse models and patient samples, we show that cachectic muscle is marked by enhanced innate immunity. Nuclear factor κB (NF-κB) activity in multiple cells, including satellite cells, myofibers, and fibro-adipogenic progenitors, promotes macrophage expansion equally derived from infiltrating monocytes and resident cells. Moreover, NF-κB-activated cells and macrophages undergo crosstalk; NF-κB+ cells recruit macrophages to inhibit regeneration and promote atrophy but, interestingly, also protect myofibers, while macrophages stimulate NF-κB+ cells to sustain an inflammatory feedforward loop. Together, we propose that NF-κB functions in multiple cells in the muscle microenvironment to stimulate macrophages that both promote and protect against muscle wasting in cancer.
Collapse
Affiliation(s)
- Benjamin R Pryce
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alexander Oles
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Erin E Talbert
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Martin J Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sudarshana Sharma
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Victoria Spadafora
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Lauren Tolliver
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - David A Mahvi
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Katherine A Morgan
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - William P Lancaster
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Eryn Beal
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Natlie Koren
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Bailey Watts
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Morgan Overstreet
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Suganya Subramanian
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Kubra Calisir
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anna Crawford
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brian Neelon
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael C Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Teresa A Zimmers
- Department of Cell, Developmental, and Cancer Biology, Knight Cancer Institute, Portland, Oregon Health Science University, Portland, OR 97239, USA
| | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David J Wang
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
8
|
Hasegawa T, Kawahara K, Sato K, Asano Y, Maeda T. Characterization of a Cancer-Induced Bone Pain Model for Use as a Model of Cancer Cachexia. Curr Issues Mol Biol 2024; 46:13364-13382. [PMID: 39727925 PMCID: PMC11726747 DOI: 10.3390/cimb46120797] [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: 10/23/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Cancer cachexia is a debilitating syndrome characterized by progressive weight loss, muscle wasting, and systemic inflammation. Despite the prevalence and severe consequences of cancer cachexia, effective treatments for this syndrome remain elusive. Therefore, there is a greater need for well-characterized animal models to identify novel therapeutic targets. Certain manifestations of cachexia, such as pain and depression, have been extensively studied using animal models of cancer-induced bone pain (CIBP). In contrast, other aspects of cachexia have received less attention in these models. To address this issue, we established the CIBP model by injecting Lewis lung carcinoma into the intramedullary cavity of the femur, observed cachexia-related symptoms, and demonstrated the utility of this model as a preclinical platform to study cancer cachexia. This model accurately recapitulates key features of cancer cachexia, including weight loss, muscle atrophy, adipose tissue depletion, CIBP, and anxiety. These findings suggest that psychological factors, in addition to physiological and metabolic factors, play significant roles in cancer cachexia development. Our model offers a valuable resource for investigating the underlying mechanisms of cancer cachexia and for developing innovative therapeutic strategies that target physical and psychological components.
Collapse
Affiliation(s)
- Takuya Hasegawa
- Department of Pharmacology, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-8603, Japan;
| | - Kohichi Kawahara
- Department of Bio-Analytical Chemistry, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-8603, Japan;
| | - Koji Sato
- Laboratory of Health Chemistry, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-8603, Japan;
| | - Yoshihisa Asano
- Department of Pharmacology, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-8603, Japan;
| | - Takehiko Maeda
- Department of Pharmacology, Faculty of Pharmacy, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata 956-8603, Japan;
| |
Collapse
|
9
|
Li L, Wazir J, Huang Z, Wang Y, Wang H. A comprehensive review of animal models for cancer cachexia: Implications for translational research. Genes Dis 2024; 11:101080. [PMID: 39220755 PMCID: PMC11364047 DOI: 10.1016/j.gendis.2023.101080] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 09/04/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by progressive weight loss and a disease process that nutritional support cannot reverse. Although progress has been made in preclinical research, there is still a long way to go in translating research findings into clinical practice. One of the main reasons for this is that existing preclinical models do not fully replicate the conditions seen in clinical patients. Therefore, it is important to understand the characteristics of existing preclinical models of cancer cachexia and pay close attention to the latest developments in preclinical models. The main models of cancer cachexia used in current research are allogeneic and xenograft models, genetically engineered mouse models, chemotherapy drug-induced models, Chinese medicine spleen deficiency models, zebrafish and Drosophila models, and cellular models. This review aims to revisit and summarize the commonly used animal models of cancer cachexia by evaluating existing preclinical models, to provide tools and support for translational medicine research.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
10
|
Nasehi F, Rylance C, Schnell E, Greene MA, Conway C, Hough Z, Duckett S, Muise-Helmericks RC, Foley AC. Analysis of potential TAK1/Map3k7 phosphorylation targets in hypertrophy and cachexia models of skeletal muscle. Biol Open 2024; 13:bio060487. [PMID: 39211992 PMCID: PMC11449438 DOI: 10.1242/bio.060487] [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: 04/19/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
TGFβ-activated kinase-1 (TAK1) is phosphorylated during both muscle growth and muscle wasting. To understand how this can lead to such opposite effects, we first performed multiplex kinase array of mouse embryonic stem cells with and without stimulation of TAK1 to determine its potential downstream targets. The phosphorylation of these targets was then compared in three different models: hypertrophic longissimus muscle of Texel sheep, tibialis anterior muscle of mice with cancer-induced cachexia and C2C12-derived myofibers, with and without blockade of TAK1 phosphorylation. In both Texel sheep and in cancer-induced cachexia, phosphorylation of both TAK1 and p38 was increased. Whereas p90RSK was increased in Texel sheep but not cachexia and the phosphorylation of HSP27 and total Jnk were increased in cachexia but not Texel. To understand this further, we examined the expression of these proteins in C2C12 cells as they differentiated into myotubes, with and without blockade of TAK1 phosphorylation. In C2C12 cells, decreased phosphorylation of TAK1 leads to reduced phosphorylation of p38, JNK, and HSP27 after 16 h and muscle fiber hypertrophy after 3 days. However, continuous blockade of this pathway leads to muscle fiber failure, suggesting that the timing of TAK1 activation controls the expression of context-dependent targets.
Collapse
Affiliation(s)
- Fatemeh Nasehi
- Department of Bioengineering, Clemson University, 68 President Street, Charleston, SC 29425, USA
| | - Cameron Rylance
- Department of Bioengineering, Clemson University, 68 President Street, Charleston, SC 29425, USA
| | - Erin Schnell
- University of South Carolina School of Medicine, 6311 Garners Ferry Road, Columbia, SC 29209, USA
| | - Maslyn Ann Greene
- Department of Animal and Veterinary Science, Clemson University, Lane #129, Clemson, SC 29634, USA
| | - Caroline Conway
- Dartmouth's Department of Cognitive Science, 5 Maynard St, Hanover, NH 03755, USA
| | - Zachary Hough
- University of Maryland, Baltimore School of Medicine, Baltimore, MD 21201, USA
| | - Susan Duckett
- Department of Animal and Veterinary Science, Clemson University, Lane #129, Clemson, SC 29634, USA
| | - Robin C. Muise-Helmericks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Ann Catherine Foley
- Department of Bioengineering, Clemson University, 68 President Street, Charleston, SC 29425, USA
| |
Collapse
|
11
|
Roy I, Binder-Markey B, Willbanks A, Sychowski D, Phipps T, Barber A, McAllister D, D'Andrea D, Franz CK, Pichika R, Dwinell MB, Lieber RL. Low-dose orthotopic cancer implantation permits measurement of longitudinal functional changes associated with cachexia. J Appl Physiol (1985) 2024; 137:705-717. [PMID: 39052773 PMCID: PMC11424168 DOI: 10.1152/japplphysiol.00173.2024] [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: 03/08/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
Progressive functional decline is a key element of cancer-associated cachexia. Major barriers to translating preclinical therapies into the clinic include lack of cancer models that accurately mimic functional decline, which develops over time, and use of nonspecific measures, like grip strength, as surrogates for physical function. In this study, we aimed to extend the survival and longevity of a cancer model, to investigate cachexia-related function at the basic science level. Survival extension studies were performed by testing multiple cell lines, dilutions, and vehicle-types in orthotopic implantation of K-rasLSL.G12D/+; Trp53R172H/+; Pdx-1-Cre (KPC)-derived cells. One hundred twenty-eight animals in this new model were assessed for cachexia syndrome phenotype using a battery of anatomical, biochemical, and behavioral techniques. We extended the survival of the KPC orthotopic model to 8-9 wk postimplantation using a relatively low 100-cell dose of DT10022 KPC cells (P < 0.001). In this low-dose orthotopic (LO) model, progressive muscle wasting was detected in parallel to systemic inflammation; skeletal muscle atrophy at the fiber level was detected as early as 3 wk postimplantation compared with controls (P < 0.001). Gait speed in LO animals declined as early as 2 wk postimplantation, whereas grip strength change was a late event. Principal component and regression analyses revealed distinct cachectic and noncachectic animal populations, which we leveraged to show that the gait speed decline was specific to cachexia (P < 0.01), whereas grip strength decline was not (P = 0.19). Gait speed represents an accurate surrogate for cachexia-related physical function as opposed to grip strength.NEW & NOTEWORTHY Previous studies of cancer-induced cachexia have been confounded by the relatively rapid death of animal subjects. Using a lower dose of cancer cells in combination with a battery of behavioral, structural, histological, and biochemical techniques, we show that gait speed is actually the best indicator of functional decline due to cachexia. Future studies are required to define the underlying physiological basis of these findings.
Collapse
Affiliation(s)
- Ishan Roy
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States
- Robert H. Lurie Cancer Center of Northwestern University, Chicago, Illinois, United States
| | - Benjamin Binder-Markey
- Department of Physical Therapy and Rehabilitation Sciences, Drexel, University, Philadelphia, Pennsylvania, United States
| | | | | | - Tenisha Phipps
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
| | - Addison Barber
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
| | - Donna McAllister
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | | | - Colin K Franz
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States
| | - Rajeswari Pichika
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States
- Hines VA Medical Center, Maywood, Illinois, United States
| | - Michael B Dwinell
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Richard L Lieber
- Shirley Ryan AbilityLab, Chicago, Illinois, United States
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States
- Hines VA Medical Center, Maywood, Illinois, United States
| |
Collapse
|
12
|
Morena F, Cabrera AR, Greene NP. Exploring heterogeneity: a dive into preclinical models of cancer cachexia. Am J Physiol Cell Physiol 2024; 327:C310-C328. [PMID: 38853648 PMCID: PMC11427020 DOI: 10.1152/ajpcell.00317.2024] [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: 05/12/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/11/2024]
Abstract
Cancer cachexia (CC) is a multifactorial and complex syndrome experienced by up to 80% of patients with cancer and implicated in ∼40% of cancer-related deaths. Given its significant impact on patients' quality of life and prognosis, there has been a growing emphasis on elucidating the underlying mechanisms of CC using preclinical models. However, the mechanisms of cachexia appear to differ across several variables including tumor type and model and biologic variables such as sex. These differences may be exacerbated by variance in experimental approaches and data reporting. This review examines literature spanning from 2011 to March 2024, focusing on common preclinical models of CC, including Lewis Lung Carcinoma, pancreatic KPC, and colorectal colon-26 and Apcmin/+ models. Our analysis reveals considerable heterogeneity in phenotypic outcomes, and investigated mechanisms within each model, with particular attention to sex differences that may be exacerbated through methodological differences. Although searching for unified mechanisms is critical, we posit that effective treatment approaches are likely to leverage the heterogeneity presented by the tumor and pertinent biological variables to direct specific interventions. In exploring this heterogeneity, it becomes critical to consider methodological and data reporting approaches to best inform further research.
Collapse
Affiliation(s)
- Francielly Morena
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
13
|
Park MA, Whelan CJ, Ahmed S, Boeringer T, Brown J, Carson TL, Crowder SL, Gage K, Gregg C, Jeong DK, Jim HSL, Judge AR, Mason TM, Parker N, Pillai S, Qayyum A, Rajasekhara S, Rasool G, Tinsley SM, Schabath MB, Stewart P, West J, McDonald P, Permuth JB. Defining and Addressing Research Priorities in Cancer Cachexia through Transdisciplinary Collaboration. Cancers (Basel) 2024; 16:2364. [PMID: 39001427 PMCID: PMC11240731 DOI: 10.3390/cancers16132364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
For many patients, the cancer continuum includes a syndrome known as cancer-associated cachexia (CAC), which encompasses the unintended loss of body weight and muscle mass, and is often associated with fat loss, decreased appetite, lower tolerance and poorer response to treatment, poor quality of life, and reduced survival. Unfortunately, there are no effective therapeutic interventions to completely reverse cancer cachexia and no FDA-approved pharmacologic agents; hence, new approaches are urgently needed. In May of 2022, researchers and clinicians from Moffitt Cancer Center held an inaugural retreat on CAC that aimed to review the state of the science, identify knowledge gaps and research priorities, and foster transdisciplinary collaborative research projects. This review summarizes research priorities that emerged from the retreat, examples of ongoing collaborations, and opportunities to move science forward. The highest priorities identified include the need to (1) evaluate patient-reported outcome (PRO) measures obtained in clinical practice and assess their use in improving CAC-related outcomes; (2) identify biomarkers (imaging, molecular, and/or behavioral) and novel analytic approaches to accurately predict the early onset of CAC and its progression; and (3) develop and test interventions (pharmacologic, nutritional, exercise-based, and through mathematical modeling) to prevent CAC progression and improve associated symptoms and outcomes.
Collapse
Affiliation(s)
- Margaret A. Park
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Christopher J. Whelan
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Sabeen Ahmed
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Tabitha Boeringer
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Joel Brown
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Tiffany L. Carson
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.L.C.); (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Sylvia L. Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.L.C.); (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Kenneth Gage
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Christopher Gregg
- School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| | - Daniel K. Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Heather S. L. Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.L.C.); (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Andrew R. Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA;
| | - Tina M. Mason
- Department of Nursing Research, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nathan Parker
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.L.C.); (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Smitha Pillai
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Aliya Qayyum
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Sahana Rajasekhara
- Department of Supportive Care Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghulam Rasool
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Sara M. Tinsley
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.L.C.); (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Paul Stewart
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Jeffrey West
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Patricia McDonald
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Lexicon Pharmaceuticals, Inc., Woodlands, TX 77381, USA
| | - Jennifer B. Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| |
Collapse
|
14
|
Arneson‐Wissink PC, Mendez H, Pelz K, Dickie J, Bartlett AQ, Worley BL, Krasnow SM, Eil R, Grossberg AJ. Hepatic signal transducer and activator of transcription-3 signalling drives early-stage pancreatic cancer cachexia via suppressed ketogenesis. J Cachexia Sarcopenia Muscle 2024; 15:975-988. [PMID: 38632714 PMCID: PMC11154744 DOI: 10.1002/jcsm.13466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Patients with pancreatic ductal adenocarcinoma (PDAC) often suffer from cachexia, a wasting syndrome that significantly reduces both quality of life and survival. Although advanced cachexia is associated with inflammatory signalling and elevated muscle catabolism, the early events driving wasting are poorly defined. During periods of nutritional scarcity, the body relies on hepatic ketogenesis to generate ketone bodies, and lipid metabolism via ketogenesis is thought to protect muscle from catabolizing during nutritional scarcity. METHODS We developed an orthotopic mouse model of early PDAC cachexia in 12-week-old C57BL/6J mice. Murine pancreatic cancer cells (KPC) were orthotopically implanted into the pancreas of wild-type, IL-6-/-, and hepatocyte STAT3-/- male and female mice. Mice were subject to fasting, 50% food restriction, ad libitum feeding or ketogenic diet interventions. We measured longitudinal body composition by EchoMRI, body mass and food intake. At the endpoint, we measured tissue mass, tissue gene expression by quantitative real-time polymerase chain reaction, whole-body calorimetry, circulating hormone levels, faecal protein and lipid content, hepatic lipid content and ketogenic response to medium-chain fatty acid bolus. We assessed muscle atrophy in vivo and C2C12 myotube atrophy in vitro. RESULTS Pre-cachectic PDAC mice did not preserve gastrocnemius muscle mass during 3-day food restriction (-13.1 ± 7.7% relative to food-restricted sham, P = 0.0117) and displayed impaired fatty acid oxidation during fasting, resulting in a hypoketotic state (ketogenic response to octanoate bolus, -83.0 ± 17.3%, P = 0.0328; Hmgcs2 expression, -28.3 ± 7.6%, P = 0.0004). PDAC human patients display impaired fasting ketones (-46.9 ± 7.1%, P < 0.0001) and elevated circulating interleukin-6 (IL-6) (12.4 ± 16.5-fold increase, P = 0.0001). IL-6-/- PDAC mice had improved muscle mass (+35.0 ± 3.9%, P = 0.0031) and ketogenic response (+129.4 ± 44.4%, P = 0.0033) relative to wild-type PDAC mice. Hepatocyte-specific signal transducer and activator of transcription 3 (STAT3) deletion prevented muscle loss (+9.3 ± 4.0%, P = 0.009) and improved fasting ketone levels (+52.0 ± 43.3%, P = 0.018) in PDAC mice. Without affecting tumour growth, a carbohydrate-free diet improved tibialis anterior myofibre diameter (+16.5 ± 3.5%, P = 0.0089), circulating ketone bodies (+333.0 ± 117.6%, P < 0.0001) and Hmgcs2 expression (+106.5 ± 36.1%, P < 0.0001) in PDAC mice. Ketone supplementation protected muscle against PDAC-induced atrophy in vitro (+111.0 ± 17.6%, P < 0.0001 myofibre diameter). CONCLUSIONS In early PDAC cachexia, muscle vulnerability to wasting is dependent on inflammation-driven metabolic reprogramming in the liver. PDAC suppresses lipid β-oxidation and impairs ketogenesis in the liver, which is reversed in genetically modified mouse models deficient in IL-6/STAT3 signalling or through ketogenic diet supplementation. This work establishes a direct link between skeletal muscle homeostasis and hepatic metabolism. Dietary and anti-inflammatory interventions that restore ketogenesis may be a viable preventative approach for pre-cachectic patients with pancreatic cancer.
Collapse
Affiliation(s)
| | - Heike Mendez
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
| | - Katherine Pelz
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
| | - Jessica Dickie
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
| | - Alexandra Q. Bartlett
- Division of Surgical Oncology, Department of Surgery, Knight Cancer InstituteOregon Health & Science UniversityPortlandORUSA
| | - Beth L. Worley
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
| | - Stephanie M. Krasnow
- Division of Oncological Sciences, Knight Cancer InstituteOregon Health & Science UniversityPortlandORUSA
| | - Robert Eil
- Division of Surgical Oncology, Department of Surgery, Knight Cancer InstituteOregon Health & Science UniversityPortlandORUSA
| | - Aaron J. Grossberg
- Brenden‐Colson Center for Pancreatic CareOregon Health & Science UniversityPortlandORUSA
- Department of Radiation MedicineOregon Health & Science UniversityPortlandORUSA
- Cancer Early Detection Advanced Research CenterOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
15
|
Yue M, Qin Z, Hu L, Ji H. Understanding cachexia and its impact on lung cancer and beyond. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:95-105. [PMID: 39169934 PMCID: PMC11332896 DOI: 10.1016/j.pccm.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 08/23/2024]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by loss of body weight secondary to skeletal muscle atrophy and adipose tissue wasting. It not only has a significant impact on patients' quality of life but also reduces the effectiveness and tolerability of anticancer therapy, leading to poor clinical outcomes. Lung cancer is a prominent global health concern, and the prevalence of cachexia is high among patients with lung cancer. In this review, we integrate findings from studies of lung cancer and other types of cancer to provide an overview of recent advances in cancer cachexia. Our focus includes topics such as the clinical criteria for diagnosis and staging, the function and mechanism of selected mediators, and potential therapeutic strategies for clinical application. A comprehensive summary of current studies will improve our understanding of the mechanisms underlying cachexia and contribute to the identification of high-risk patients, the development of effective treatment strategies, and the design of appropriate therapeutic regimens for patients at different disease stages.
Collapse
Affiliation(s)
- Meiting Yue
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Qin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang Hu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
16
|
Spadafora V, Pryce BR, Oles A, Talbert EE, Romeo M, Vaena S, Berto S, Ostrowski MC, Wang DJ, Guttridge DC. Optimization of a mouse model of pancreatic cancer to simulate the human phenotypes of metastasis and cachexia. BMC Cancer 2024; 24:414. [PMID: 38570770 PMCID: PMC10993462 DOI: 10.1186/s12885-024-12104-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) presents with a high mortality rate. Two important features of PDAC contribute to this poor outcome. The first is metastasis which occurs in ~ 80% of PDAC patients. The second is cachexia, which compromises treatment tolerance for patients and reduces their quality of life. Although various mouse models of PDAC exist, recapitulating both metastatic and cachectic features have been challenging. METHODS Here, we optimize an orthotopic mouse model of PDAC by altering several conditions, including the subcloning of parental murine PDAC cells, implantation site, number of transplanted cells, and age of recipient mice. We perform spatial profiling to compare primary and metastatic immune microenvironments and RNA sequencing to gain insight into the mechanisms of muscle wasting in PDAC-induced cachexia, comparing non-metastatic to metastatic conditions. RESULTS These modifications extend the time course of the disease and concurrently increase the rate of metastasis to approximately 70%. Furthermore, reliable cachexia endpoints are achieved in both PDAC mice with and without metastases, which is reminiscent of patients. We also find that cachectic muscles from PDAC mice with metastasis exhibit a similar transcriptional profile to muscles derived from mice and patients without metastasis. CONCLUSION Together, this model is likely to be advantageous in both advancing our understanding of the mechanism of PDAC cachexia, as well as in the evaluation of novel therapeutics.
Collapse
Affiliation(s)
- Victoria Spadafora
- Department of Pediatrics, Darby Children's Research Institute, 416, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Benjamin R Pryce
- Department of Pediatrics, Darby Children's Research Institute, 416, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Alexander Oles
- Department of Pediatrics, Darby Children's Research Institute, 416, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Erin E Talbert
- Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University of Iowa, Iowa, 52242, USA
| | - Martin Romeo
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Silvia Vaena
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Stefano Berto
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Michael C Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - David J Wang
- Department of Pediatrics, Darby Children's Research Institute, 416, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, 416, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
17
|
Zhao K, Ebrahimie E, Mohammadi-Dehcheshmeh M, Lewsey MG, Zheng L, Hoogenraad NJ. Transcriptomic signature of cancer cachexia by integration of machine learning, literature mining and meta-analysis. Comput Biol Med 2024; 172:108233. [PMID: 38452471 DOI: 10.1016/j.compbiomed.2024.108233] [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: 09/10/2023] [Revised: 01/23/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Cancer cachexia is a severe metabolic syndrome marked by skeletal muscle atrophy. A successful clinical intervention for cancer cachexia is currently lacking. The study of cachexia mechanisms is largely based on preclinical animal models and the availability of high-throughput transcriptomic datasets of cachectic mouse muscles is increasing through the extensive use of next generation sequencing technologies. METHODS Cachectic mouse muscle transcriptomic datasets of ten different studies were combined and mined by seven attribute weighting models, which analysed both categorical variables and numerical variables. The transcriptomic signature of cancer cachexia was identified by attribute weighting algorithms and was used to evaluate the performance of eleven pattern discovery models. The signature was employed to find the best combination of drugs (drug repurposing) for developing cancer cachexia treatment strategies, as well as to evaluate currently used cachexia drugs by literature mining. RESULTS Attribute weighting algorithms ranked 26 genes as the transcriptomic signature of muscle from mice with cancer cachexia. Deep Learning and Random Forest models performed better in differentiating cancer cachexia cases based on muscle transcriptomic data. Literature mining revealed that a combination of melatonin and infliximab has negative interactions with 2 key genes (Rorc and Fbxo32) upregulated in the transcriptomic signature of cancer cachexia in muscle. CONCLUSIONS The integration of machine learning, meta-analysis and literature mining was found to be an efficient approach to identifying a robust transcriptomic signature for cancer cachexia, with implications for improving clinical diagnosis and management of this condition.
Collapse
Affiliation(s)
- Kening Zhao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Esmaeil Ebrahimie
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, 3086, Australia; School of Animal and Veterinary Science, The University of Adelaide, Adelaide, SA 5371, Australia; School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Manijeh Mohammadi-Dehcheshmeh
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, 3086, Australia; School of Animal and Veterinary Science, The University of Adelaide, Adelaide, SA 5371, Australia.
| | - Mathew G Lewsey
- Australian Research Council Research Hub for Medicinal Agriculture, La Trobe University, AgriBio Building, Bundoora, VIC, 3086, Australia; La Trobe Institute for Sustainable Agriculture and Food, Department of Plant, Animal and Soil Sciences, La Trobe University, AgriBio Building, Bundoora, VIC, 3086, Australia; Australian Research Council Centre of Excellence in Plants for Space, AgriBio Building, La Trobe University, Bundoora, VIC, 3086, Australia.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Nick J Hoogenraad
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia; Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3084, Australia.
| |
Collapse
|
18
|
Rampioni Vinciguerra GL, Capece M, Reggiani Bonetti L, Nigita G, Calore F, Rentsch S, Magistri P, Ballarin R, Di Benedetto F, Distefano R, Cirombella R, Vecchione A, Belletti B, Baldassarre G, Lovat F, Croce CM. Nutrient restriction-activated Fra-2 promotes tumor progression via IGF1R in miR-15a downmodulated pancreatic ductal adenocarcinoma. Signal Transduct Target Ther 2024; 9:31. [PMID: 38342897 PMCID: PMC10859382 DOI: 10.1038/s41392-024-01740-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/07/2023] [Accepted: 01/03/2024] [Indexed: 02/13/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease, characterized by an intense desmoplastic reaction that compresses blood vessels and limits nutrient supplies. PDAC aggressiveness largely relies on its extraordinary capability to thrive and progress in a challenging tumor microenvironment. Dysregulation of the onco-suppressor miR-15a has been extensively documented in PDAC. Here, we identified the transcription factor Fos-related antigen-2 (Fra-2) as a miR-15a target mediating the adaptive mechanism of PDAC to nutrient deprivation. We report that the IGF1 signaling pathway was enhanced in nutrient deprived PDAC cells and that Fra-2 and IGF1R were significantly overexpressed in miR-15a downmodulated PDAC patients. Mechanistically, we discovered that miR-15a repressed IGF1R expression via Fra-2 targeting. In miR-15a-low context, IGF1R hyperactivated mTOR, modulated the autophagic flux and sustained PDAC growth in nutrient deprivation. In a genetic mouse model, Mir15aKO PDAC showed Fra-2 and Igf1r upregulation and mTOR activation in response to diet restriction. Consistently, nutrient restriction improved the efficacy of IGF1R inhibition in a Fra-2 dependent manner. Overall, our results point to a crucial role of Fra-2 in the cellular stress response due to nutrient restriction typical of pancreatic cancer and support IGF1R as a promising and vulnerable target in miR-15a downmodulated PDAC.
Collapse
Affiliation(s)
- Gian Luca Rampioni Vinciguerra
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Marina Capece
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
| | - Luca Reggiani Bonetti
- Department of Diagnostic, Clinic and Public Health Medicine, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
| | - Federica Calore
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
| | - Sydney Rentsch
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
| | - Paolo Magistri
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Roberto Ballarin
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Fabrizio Di Benedetto
- Hepato-pancreato-biliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, 41100, Italy
| | - Rosario Distefano
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA
| | - Roberto Cirombella
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, University of Rome "Sapienza", Rome, 00189, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), National Cancer Institute, Aviano, 33081, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), National Cancer Institute, Aviano, 33081, Italy
| | - Francesca Lovat
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, 43210, OH, USA.
| |
Collapse
|
19
|
Vu TT, Kim K, Manna M, Thomas J, Remaily BC, Montgomery EJ, Costa T, Granchie L, Xie Z, Guo Y, Chen M, Castillo AMM, Kulp SK, Mo X, Nimmagadda S, Gregorevic P, Owen DH, Ganesan LP, Mace TA, Coss CC, Phelps MA. Decoupling FcRn and tumor contributions to elevated immune checkpoint inhibitor clearance in cancer cachexia. Pharmacol Res 2024; 199:107048. [PMID: 38145833 PMCID: PMC10798214 DOI: 10.1016/j.phrs.2023.107048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023]
Abstract
High baseline clearance of immune checkpoint inhibitors (ICIs), independent of dose or systemic exposure, is associated with cachexia and poor outcomes in cancer patients. Mechanisms linking ICI clearance, cachexia and ICI therapy failure are unknown. Here, we evaluate in four murine models and across multiple antibodies whether altered baseline catabolic clearance of administered antibody requires a tumor and/or cachexia and whether medical reversal of cachexia phenotype can alleviate altered clearance. Key findings include mild cachexia phenotype and lack of elevated pembrolizumab clearance in the MC38 tumor-bearing model. We also observed severe cachexia and decreased, instead of increased, baseline pembrolizumab clearance in the tumor-free cisplatin-induced cachexia model. Liver Fcgrt expression correlated with altered baseline catabolic clearance, though elevated clearance was still observed with antibodies having no (human IgA) or reduced (human H310Q IgG1) FcRn binding. We conclude cachexia phenotype coincides with altered antibody clearance, though tumor presence is neither sufficient nor necessary for altered clearance in immunocompetent mice. Magnitude and direction of clearance alteration correlated with hepatic Fcgrt, suggesting changes in FcRn expression and/or recycling function may be partially responsible, though factors beyond FcRn also contribute to altered clearance in cachexia.
Collapse
Affiliation(s)
- Trang T Vu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Millennium Manna
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Bryan C Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Emma J Montgomery
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Travis Costa
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Lauren Granchie
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Zhiliang Xie
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Min Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Alyssa Marie M Castillo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Samuel K Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, Ohio State University, Columbus, OH, USA; Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA
| | - Sridhar Nimmagadda
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Gregorevic
- Department of Anatomy & Physiology and Centre for Muscle Research, The University of Melbourne, Parkville, VIC, Australia
| | - Dwight H Owen
- Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Latha P Ganesan
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Thomas A Mace
- Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Christopher C Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA; Pelotonia Institute for Immuno-Oncology, OSUCCC - James, The Ohio State University, Columbus, OH , USA; The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
20
|
Roy I, Binder-Markey B, Sychowski D, Willbanks A, Phipps T, McAllister D, Bhakta A, Marquez E, D'Andrea D, Franz C, Pichika R, Dwinell MB, Jayabalan P, Lieber RL. Gait speed is a biomarker of cancer-associated cachexia decline and recovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566852. [PMID: 38014165 PMCID: PMC10680669 DOI: 10.1101/2023.11.13.566852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background Progressive functional decline is a key element of cancer-associated cachexia. No therapies have successfully translated to the clinic due to an inability to measure and improve physical function in cachectic patients. Major barriers to translating pre-clinical therapies to the clinic include lack of cancer models that accurately mimic functional decline and use of non-specific outcome measures of function, like grip strength. New approaches are needed to investigate cachexia-related function at both the basic and clinical science levels. Methods Survival extension studies were performed by testing multiple cell lines, dilutions, and vehicle-types in orthotopic implantation of K-ras LSL.G12D/+ ; Trp53 R172H/+ ; Pdx-1-Cre (KPC) derived cells. 128 animals in this new model were then assessed for muscle wasting, inflammation, and functional decline using a battery of biochemical, physiologic, and behavioral techniques. In parallel, we analyzed a 156-subject cohort of cancer patients with a range of cachexia severity, and who required rehabilitation, to determine the relationship between gait speed via six-minute walk test (6MWT), grip strength (hGS), and functional independence measures (FIM). Cachectic patients were identified using the Weight Loss Grading Scale (WLGS), Fearon consensus criteria, and the Prognostic Nutritional Index (PNI). Results Using a 100-cell dose of DT10022 KPC cells, we extended the survival of the KPC orthotopic model to 8-9 weeks post-implantation compared to higher doses used (p<0.001). In this Low-dose Orthotopic (LO) model, both progressive skeletal and cardiac muscle wasting were detected in parallel to systemic inflammation; skeletal muscle atrophy at the fiber level was detected as early as 3 weeks post-implantation compared to controls (p<0.001). Gait speed in LO animals declined as early 2 week post-implantation whereas grip strength change was a late event and related to end of life. Principle component analysis (PCA) revealed distinct cachectic and non-cachectic animal populations, which we leveraged to show that gait speed decline was specific to cachexia (p<0.01) while grip strength decline was not (p=0.19). These data paralleled our observations in cancer patients with cachexia who required rehabilitation. In cachectic patients (identified by WLGS, Fearon criteria, or PNI, change in 6MWT correlated with motor FIM score changes while hGS did not (r 2 =0.18, p<0.001). This relationship between 6MWT and FIM in cachectic patients was further confirmed through multivariate regression (r 2 =0.30, p<0.001) controlling for age and cancer burden. Conclusion Outcome measures linked to gait are better associated with cachexia related function and preferred for future pre-clinical and clinical cachexia studies.
Collapse
|
21
|
Robinson TP, Hamidi T, Counts B, Guttridge DC, Ostrowski MC, Zimmers TA, Koniaris LG. The impact of inflammation and acute phase activation in cancer cachexia. Front Immunol 2023; 14:1207746. [PMID: 38022578 PMCID: PMC10644737 DOI: 10.3389/fimmu.2023.1207746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
The development of cachexia in the setting of cancer or other chronic diseases is a significant detriment for patients. Cachexia is associated with a decreased ability to tolerate therapies, reduction in ambulation, reduced quality of life, and increased mortality. Cachexia appears intricately linked to the activation of the acute phase response and is a drain on metabolic resources. Work has begun to focus on the important inflammatory factors associated with the acute phase response and their role in the immune activation of cachexia. Furthermore, data supporting the liver, lung, skeletal muscle, and tumor as all playing a role in activation of the acute phase are emerging. Although the acute phase is increasingly being recognized as being involved in cachexia, work in understanding underlying mechanisms of cachexia associated with the acute phase response remains an active area of investigation and still lack a holistic understanding and a clear causal link. Studies to date are largely correlative in nature, nonetheless suggesting the possibility for a role for various acute phase reactants. Herein, we examine the current literature regarding the acute phase response proteins, the evidence these proteins play in the promotion and exacerbation of cachexia, and current evidence of a therapeutic potential for patients.
Collapse
Affiliation(s)
- Tyler P. Robinson
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tewfik Hamidi
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Brittany Counts
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Denis C. Guttridge
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Michael C. Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Leonidas G. Koniaris
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| |
Collapse
|
22
|
Directo D, Lee SR. Cancer Cachexia: Underlying Mechanisms and Potential Therapeutic Interventions. Metabolites 2023; 13:1024. [PMID: 37755304 PMCID: PMC10538050 DOI: 10.3390/metabo13091024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer cachexia, a multifactorial metabolic syndrome developed during malignant tumor growth, is characterized by an accelerated loss of body weight accompanied by the depletion of skeletal muscle mass. This debilitating condition is associated with muscle degradation, impaired immune function, reduced functional capacity, compromised quality of life, and diminished survival in cancer patients. Despite the lack of the known capability of fully reversing or ameliorating this condition, ongoing research is shedding light on promising preclinical approaches that target the disrupted mechanisms in the pathophysiology of cancer cachexia. This comprehensive review delves into critical aspects of cancer cachexia, including its underlying pathophysiological mechanisms, preclinical models for studying the progression of cancer cachexia, methods for clinical assessment, relevant biomarkers, and potential therapeutic strategies. These discussions collectively aim to contribute to the evolving foundation for effective, multifaceted counteractive strategies against this challenging condition.
Collapse
Affiliation(s)
| | - Sang-Rok Lee
- Department of Kinesiology, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
23
|
Guo T, Wei Q. Cell Reprogramming Techniques: Contributions to Cancer Therapy. Cell Reprogram 2023; 25:142-153. [PMID: 37530737 DOI: 10.1089/cell.2023.0011] [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: 08/03/2023] Open
Abstract
The reprogramming of terminally differentiated cells over the past few years has become important for induced pluripotent stem cells (iPSCs) in the field of regenerative medicine and disease drug modeling. At the same time, iPSCs have also played an important role in human cancer research. iPSCs derived from cancer patients can be used to simulate the early progression of cancer, for drug testing, and to study the molecular mechanism of cancer occurrence. In recent years, with the application of cellular immunotherapy in cancer therapy, patient-derived iPSC-induced immune cells (T, natural killer, and macrophage cells) solve the problem of immune rejection and have higher immunogenicity, which greatly improves the therapeutic efficiency of immune cell therapy. With the continuous progress of cancer differentiation therapy, iPSC technology can reprogram cancer cells to a more primitive pluripotent undifferentiated state, and successfully reverse cancer cells to a benign phenotype by changing the epigenetic inheritance of cancer cells. This article reviews the recent progress of cell reprogramming technology in human cancer research, focuses on the application of reprogramming technology in cancer immunotherapy and the problems solved, and summarizes the malignant phenotype changes of cancer cells in the process of reprogramming and subsequent differentiation.
Collapse
Affiliation(s)
- Tongtong Guo
- College of Life Science, Northwest University, Xi'an, China
| | - Qi Wei
- Wuhan Institute of Technology, Wuhan, China
| |
Collapse
|
24
|
Yamakawa T, Zhang G, Najjar LB, Li C, Itakura K. The uncharacterized transcript KIAA0930 confers a cachexic phenotype on cancer cells. Oncotarget 2023; 14:723-737. [PMID: 37477523 PMCID: PMC10360925 DOI: 10.18632/oncotarget.28476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
Patients with cancer cachexia have a poor prognosis and impaired quality of life. Numerous studies using preclinical models have shown that inflammatory cytokines play an important role in the development of cancer cachexia; however, no clinical trial targeting cytokines has been successful. Therefore, it is essential to identify molecular mechanisms to develop anti-cachexia therapies. Here we identified the uncharacterized transcript KIAA0930 as a candidate cachexic factor based on analyses of microarray datasets and an in vitro muscle atrophy assay. While conditioned media from pancreatic, colorectal, gastric, and tongue cancer cells caused muscle atrophy in vitro, conditioned medium from KIAA0930 knockdown cells did not. The PANC-1 orthotopic xenograft study showed that the tibialis anterior muscle weight and cross-sectional area were increased in mice bearing KIAA0930 knockdown cells compared to control mice. Interestingly, KIAA0930 knockdown did not cause consistent changes in the secretion of inflammatory cytokines/chemokines from a variety of cancer cell lines. An initial characterization experiment showed that KIAA0930 is localized in the cytosol and not secreted from cells. These data suggest that the action of KIAA0930 is independent of the expression of cytokines/chemokines and that KIAA0930 could be a novel therapeutic target for cachexia.
Collapse
Affiliation(s)
- Takahiro Yamakawa
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Guoxiang Zhang
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Liza Bengrine Najjar
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Chun Li
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Keiichi Itakura
- Center for RNA Biology and Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
25
|
Kerr HL, Krumm K, Lee I(I, Anderson B, Christiani A, Strait L, Breckheimer BA, Irwin B, Jiang A(S, Rybachok A, Chen A, Caeiro L, Dacek E, Hall DB, Kostyla CH, Hales LM, Soliman TM, Garcia JM. EXT418, a novel long-acting ghrelin, mitigates Lewis lung carcinoma induced cachexia in mice. J Cachexia Sarcopenia Muscle 2023; 14:1337-1348. [PMID: 36942661 PMCID: PMC10235874 DOI: 10.1002/jcsm.13211] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Ghrelin is a potential therapy for cachexia due to its orexigenic properties and anabolic effects on muscle and fat. However, its clinical use is limited by the short half-life of active (acylated) ghrelin (~11 min in humans). EXT418 is a novel long-acting, constitutively active ghrelin analog created by covalently linking it to a vitamin D derivative. Here, we evaluated the effects and mechanisms of action of EXT418 on Lewis lung carcinoma (LLC)-induced cachexia in mice. METHODS Male C57BL/6J mice (5- to 7-month-old) were implanted with 1 × 106 heat-killed (HK) or live LLC cells. When the tumour was palpable, mice were injected with vehicle (T + V) or EXT418 daily (T + 418 Daily, 0.25 mg/kg/day) or every other day (T + 418 EOD, 0.5 mg/kg/EOD) for up to 14 days, whereas HK-treated mice were given vehicle (HK + V). Subsets of T + 418 Daily or EOD-treated mice were pair-fed to the T + V group. Body composition and grip strength were evaluated before tumour implantation and at the end of the experiment. Molecular markers were probed in muscles upon termination. RESULTS In tumour-bearing mice, administration of EXT418 daily or EOD partially prevented weight loss (T + V vs. T + 418 Daily, P = 0.030; and vs. T + 418 EOD, P = 0.020). Similar effects were observed in whole body fat and lean body mass. Grip strength in tumour-bearing mice was improved by EXT418 daily (P = 0.010) or EOD (P = 0.008) administration compared with vehicle-treated mice. These effects of EXT418 on weight and grip strength were partially independent of food intake. EXT418 daily administration also improved type IIA (P = 0.015), IIB (P = 0.037) and IIX (P = 0.050) fibre cross-sectional area (CSA) in tibialis anterior (TA) and EXT418 EOD improved CSA of IIB fibres in red gastrocnemius (GAS; P = 0.005). In skeletal muscles, tumour-induced increases in atrogenes Fbxo32 and Trim63 were ameliorated by EXT418 treatments (TA and GAS/plantaris, PL), which were independent of food intake. EXT418 administration decreased expression of the mitophagy marker Bnip3 (GAS/PL; P ≤ 0.010). Similar effects of EXT418 EOD were observed in p62 (GAS/PL; P = 0.039). In addition, EXT418 treatments ameliorated the tumour-induced elevation in muscle Il6 transcript levels (TA and GAS/PL), independently of food intake. Il-6 transcript levels in adipose tissue and circulating IL-10 were elevated in response to the tumour but these increases were not significant with EXT418 administration. Tumour mass was not altered by EXT418. CONCLUSIONS EXT418 mitigates LLC-induced cachexia by attenuating skeletal muscle inflammation, proteolysis, and mitophagy, without affecting tumour mass and partially independent of food intake.
Collapse
Affiliation(s)
- Haiming L. Kerr
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Kora Krumm
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Ian (In‐gi) Lee
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Anthony Christiani
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Lena Strait
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Beatrice A. Breckheimer
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Brynn Irwin
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Alice (Siyi) Jiang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Artur Rybachok
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Amanda Chen
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Lucas Caeiro
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Elizabeth Dacek
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | | | - Caroline H. Kostyla
- Extend Biosciences, Inc.St. NewtonMassachusettsUSA
- Present address:
Atalanta TherapeuticsBostonMassachusettsUSA
| | | | | | - Jose M. Garcia
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| |
Collapse
|
26
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
27
|
van de Worp WR, Theys J, González AS, van der Heyden B, Verhaegen F, Hauser D, Caiment F, Smeets HJ, Schols AM, van Helvoort A, Langen RC. A novel orthotopic mouse model replicates human lung cancer cachexia. J Cachexia Sarcopenia Muscle 2023; 14:1410-1423. [PMID: 37025071 PMCID: PMC10235890 DOI: 10.1002/jcsm.13222] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 04/08/2023] Open
Abstract
INTRODUCTION Cancer cachexia, highly prevalent in lung cancer, is a debilitating syndrome characterized by involuntary loss of skeletal muscle mass and is associated with poor clinical outcome, decreased survival and negative impact on tumour therapy. Various lung tumour-bearing animal models have been used to explore underlying mechanisms of cancer cachexia. However, these models do not simulate anatomical and immunological features key to lung cancer and associated muscle wasting. Overcoming these shortcomings is essential to translate experimental findings into the clinic. We therefore evaluated whether a syngeneic, orthotopic lung cancer mouse model replicates systemic and muscle-specific alterations associated with human lung cancer cachexia. METHODS Immune competent, 11 weeks old male 129S2/Sv mice, were randomly allocated to either (1) sham control group or (2) tumour-bearing group. Syngeneic lung epithelium-derived adenocarcinoma cells (K-rasG12D ; p53R172HΔG ) were inoculated intrapulmonary into the left lung lobe of the mice. Body weight and food intake were measured daily. At baseline and weekly after surgery, grip strength was measured and tumour growth and muscle volume were assessed using micro cone beam CT imaging. After reaching predefined surrogate survival endpoint, animals were euthanized, and skeletal muscles of the lower hind limbs were collected for biochemical analysis. RESULTS Two-third of the tumour-bearing mice developed cachexia based on predefined criteria. Final body weight (-13.7 ± 5.7%; P < 0.01), muscle mass (-13.8 ± 8.1%; P < 0.01) and muscle strength (-25.5 ± 10.5%; P < 0.001) were reduced in cachectic mice compared with sham controls and median survival time post-surgery was 33.5 days until humane endpoint. Markers for proteolysis, both ubiquitin proteasome system (Fbxo32 and Trim63) and autophagy-lysosomal pathway (Gabarapl1 and Bnip3), were significantly upregulated, whereas markers for protein synthesis (relative phosphorylation of Akt, S6 and 4E-BP1) were significantly decreased in the skeletal muscle of cachectic mice compared with control. The cachectic mice exhibited increased pentraxin-2 (P < 0.001) and CXCL1/KC (P < 0.01) expression levels in blood plasma and increased mRNA expression of IκBα (P < 0.05) in skeletal muscle, indicative for the presence of systemic inflammation. Strikingly, RNA sequencing, pathway enrichment and miRNA expression analyses of mouse skeletal muscle strongly mirrored alterations observed in muscle biopsies of patients with lung cancer cachexia. CONCLUSIONS We developed an orthotopic model of lung cancer cachexia in immune competent mice. Because this model simulates key aspects specific to cachexia in lung cancer patients, it is highly suitable to further investigate the underlying mechanisms of lung cancer cachexia and to test the efficacy of novel intervention strategies.
Collapse
Affiliation(s)
- Wouter R.P.H. van de Worp
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Jan Theys
- Department of Precision Medicine, GROW – School for Oncology and Developmental BiologyMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Alba Sanz González
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Brent van der Heyden
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental BiologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology and Developmental BiologyMaastricht University Medical Centre+MaastrichtThe Netherlands
| | - Duncan Hauser
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Hubertus J.M. Smeets
- Department of Toxicogenomics, GROW – School for Oncology and Developmental Biology, MHeNs – School for Mental Health and NeurosciencesMaastricht UniversityMaastrichtThe Netherlands
| | - Annemie M.W.J. Schols
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| | - Ardy van Helvoort
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
- Danone Nutricia ResearchUtrechtThe Netherlands
| | - Ramon C.J. Langen
- Department of Respiratory Medicine, NUTRIM – School of Nutrition and Translational Research in MetabolismMaastricht University Medical Center+MaastrichtThe Netherlands
| |
Collapse
|
28
|
Neyroud D, Laitano O, Dasgupta A, Lopez C, Schmitt RE, Schneider JZ, Hammers DW, Sweeney HL, Walter GA, Doles J, Judge SM, Judge AR. Blocking muscle wasting via deletion of the muscle-specific E3 ligase MuRF1 impedes pancreatic tumor growth. Commun Biol 2023; 6:519. [PMID: 37179425 PMCID: PMC10183033 DOI: 10.1038/s42003-023-04902-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Cancer-induced muscle wasting reduces quality of life, complicates or precludes cancer treatments, and predicts early mortality. Herein, we investigate the requirement of the muscle-specific E3 ubiquitin ligase, MuRF1, for muscle wasting induced by pancreatic cancer. Murine pancreatic cancer (KPC) cells, or saline, were injected into the pancreas of WT and MuRF1-/- mice, and tissues analyzed throughout tumor progression. KPC tumors induces progressive wasting of skeletal muscle and systemic metabolic reprogramming in WT mice, but not MuRF1-/- mice. KPC tumors from MuRF1-/- mice also grow slower, and show an accumulation of metabolites normally depleted by rapidly growing tumors. Mechanistically, MuRF1 is necessary for the KPC-induced increases in cytoskeletal and muscle contractile protein ubiquitination, and the depression of proteins that support protein synthesis. Together, these data demonstrate that MuRF1 is required for KPC-induced skeletal muscle wasting, whose deletion reprograms the systemic and tumor metabolome and delays tumor growth.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
- Institute of Sports Sciences, University of Lausanne, Lausanne, Switzerland
| | - Orlando Laitano
- Myology Institute, University of Florida, Gainesville, FL, USA
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, USA
| | - Aneesha Dasgupta
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Christopher Lopez
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Rebecca E Schmitt
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jessica Z Schneider
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - David W Hammers
- Myology Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - H Lee Sweeney
- Myology Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Glenn A Walter
- Myology Institute, University of Florida, Gainesville, FL, USA
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Jason Doles
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Sarah M Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
- Myology Institute, University of Florida, Gainesville, FL, USA
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA.
- Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
29
|
Gilmore LA, Parry TL, Thomas GA, Khamoui AV. Skeletal muscle omics signatures in cancer cachexia: perspectives and opportunities. J Natl Cancer Inst Monogr 2023; 2023:30-42. [PMID: 37139970 PMCID: PMC10157770 DOI: 10.1093/jncimonographs/lgad006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 05/05/2023] Open
Abstract
Cachexia is a life-threatening complication of cancer that occurs in up to 80% of patients with advanced cancer. Cachexia reflects the systemic consequences of cancer and prominently features unintended weight loss and skeletal muscle wasting. Cachexia impairs cancer treatment tolerance, lowers quality of life, and contributes to cancer-related mortality. Effective treatments for cancer cachexia are lacking despite decades of research. High-throughput omics technologies are increasingly implemented in many fields including cancer cachexia to stimulate discovery of disease biology and inform therapy choice. In this paper, we present selected applications of omics technologies as tools to study skeletal muscle alterations in cancer cachexia. We discuss how comprehensive, omics-derived molecular profiles were used to discern muscle loss in cancer cachexia compared with other muscle-wasting conditions, to distinguish cancer cachexia from treatment-related muscle alterations, and to reveal severity-specific mechanisms during the progression of cancer cachexia from early toward severe disease.
Collapse
Affiliation(s)
- 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
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Gwendolyn A Thomas
- Department of Kinesiology, Pennsylvania State University, University Park, PA, 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
| |
Collapse
|
30
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Understanding the relationship between cancer associated cachexia and hypoxia-inducible factor-1. Biomed Pharmacother 2023; 163:114802. [PMID: 37146421 DOI: 10.1016/j.biopha.2023.114802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a multifactorial disorder characterized by an unrestricted loss of body weight as a result of muscle and adipose tissue atrophy. Cachexia is influenced by several factors, including decreased metabolic activity and food intake, an imbalance between energy uptake and expenditure, excessive catabolism, and inflammation. Cachexia is highly associated with all types of cancers responsible for more than half of cancer-related mortalities worldwide. In healthy individuals, adipose tissue significantly regulates energy balance and glucose homeostasis. However, in metastatic cancer patients, CAC occurs mainly because of an imbalance between muscle protein synthesis and degradation which are organized by certain extracellular ligands and associated signaling pathways. Under hypoxic conditions, hypoxia-inducible factor-1 (HIF-1α) accumulated and translocated to the nucleus and activate numerous genes involved in cell survival, invasion, angiogenesis, metastasis, metabolic reprogramming, and cancer stemness. On the other hand, the ubiquitination proteasome pathway is inhibited during low O2 levels which promote muscle wasting in cancer patients. Therefore, understanding the mechanism of the HIF-1 pathway and its metabolic adaptation to biomolecules is important for developing a novel therapeutic method for cancer and cachexia therapy. Even though many HIF inhibitors are already in a clinical trial, their mechanism of action remains unknown. With this background, this review summarizes the basic concepts of cachexia, the role of inflammatory cytokines, pathways connected with cachexia with special reference to the HIF-1 pathway and its regulation, metabolic changes, and inhibitors of HIFs.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
31
|
Nicotinamide Adenine Dinucleotide Precursor Suppresses Hepatocellular Cancer Progression in Mice. Nutrients 2023; 15:nu15061447. [PMID: 36986177 PMCID: PMC10055624 DOI: 10.3390/nu15061447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/02/2023] [Accepted: 03/12/2023] [Indexed: 03/19/2023] Open
Abstract
Targeting Nicotinamide adenine dinucleotide (NAD) metabolism has emerged as a promising anti-cancer strategy; we aimed to explore the health benefits of boosting NAD levels with nicotinamide riboside (NR) on hepatocellular carcinoma (HCC). We established three in vivo tumor models, including subcutaneous transplantation tumor model in both Balb/c nude mice (xenograft), C57BL/6J mice (allograft), and hematogenous metastatic neoplasm in nude mice. NR (400 mg/kg bw) was supplied daily in gavage. In-situ tumor growth or noninvasive bioluminescence were measured to evaluate the effect of NR on the HCC process. HepG2 cells were treated with transforming growth factor-β (TGF-β) in the absence/presence of NR in vitro. We found that NR supplementation alleviated malignancy-induced weight loss and metastasis to lung in nude mice in both subcutaneous xenograft and hematogenous metastasis models. NR supplementation decreased metastasis to the bone and liver in the hematogenous metastasis model. NR supplementation also significantly decreased the size of allografted tumors and extended the survival time in C57BL/6J mice. In vitro experiments showed that NR intervention inhibited the migration and invasion of HepG2 cells triggered by TGF-β. In summary, our results supply evidence that boosting NAD levels by supplementing NR alleviates HCC progression and metastasis, which may serve as an effective treatment for the suppression of HCC progression.
Collapse
|
32
|
Zhou Y, Xia J, Xu S, She T, Zhang Y, Sun Y, Wen M, Jiang T, Xiong Y, Lei J. Experimental mouse models for translational human cancer research. Front Immunol 2023; 14:1095388. [PMID: 36969176 PMCID: PMC10036357 DOI: 10.3389/fimmu.2023.1095388] [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: 11/11/2022] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
The development and growth of tumors remains an important and ongoing threat to human life around the world. While advanced therapeutic strategies such as immune checkpoint therapy and CAR-T have achieved astonishing progress in the treatment of both solid and hematological malignancies, the malignant initiation and progression of cancer remains a controversial issue, and further research is urgently required. The experimental animal model not only has great advantages in simulating the occurrence, development, and malignant transformation mechanisms of tumors, but also can be used to evaluate the therapeutic effects of a diverse array of clinical interventions, gradually becoming an indispensable method for cancer research. In this paper, we have reviewed recent research progress in relation to mouse and rat models, focusing on spontaneous, induced, transgenic, and transplantable tumor models, to help guide the future study of malignant mechanisms and tumor prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Tao Jiang
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| | - Yanlu Xiong
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| | - Jie Lei
- *Correspondence: Jie Lei, ; Yanlu Xiong, ; Tao Jiang,
| |
Collapse
|
33
|
Neyroud D, Laitano O, Daguspta A, Lopez C, Schmitt RE, Schneider JZ, Hammers DW, Sweeney HL, Walter GA, Doles J, Judge SM, Judge AR. Blocking muscle wasting via deletion of the muscle-specific E3 ubiquitin ligase MuRF1 impedes pancreatic tumor growth. RESEARCH SQUARE 2023:rs.3.rs-2524562. [PMID: 36798266 PMCID: PMC9934780 DOI: 10.21203/rs.3.rs-2524562/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cancer-induced muscle wasting reduces quality of life, complicates or precludes cancer treatments, and predicts early mortality. Herein, we investigated the requirement of the muscle-specific E3 ubiquitin ligase, MuRF1, for muscle wasting induced by pancreatic cancer. Murine pancreatic cancer (KPC) cells, or saline, were injected into the pancreas of WT and MuRF1-/- mice, and tissues analyzed throughout tumor progression. KPC tumors induced progressive wasting of skeletal muscle and systemic metabolic reprogramming in WT mice, but not MuRF1-/- mice. KPC tumors from MuRF1-/- mice also grew slower, and showed an accumulation of metabolites normally depleted by rapidly growing tumors. Mechanistically, MuRF1 was necessary for the KPC-induced increases in cytoskeletal and muscle contractile protein ubiquitination, and the depression of proteins that support protein synthesis. Together, these data demonstrate that MuRF1 is required for KPC-induced skeletal muscle wasting, whose deletion reprograms the systemic and tumor metabolome and delays tumor growth.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical Therapy, University of Florida, Gainesville, USA
- Myology Institute, University of Florida, Gainesville, USA
- Institute of Sports Sciences, University of Lausanne, Lausanne, Switzerland
| | - Orlando Laitano
- Myology Institute, University of Florida, Gainesville, USA
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, USA
| | - Aneesha Daguspta
- Department of Anatomy, Cell Biology and Physiology, Indiana university school of medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Christopher Lopez
- Department of Physical Therapy, University of Florida, Gainesville, USA
- Myology Institute, University of Florida, Gainesville, USA
| | - Rebecca E. Schmitt
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Jessica Z. Schneider
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - David W. Hammers
- Myology Institute, University of Florida, Gainesville, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, USA
| | - H. Lee Sweeney
- Myology Institute, University of Florida, Gainesville, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, USA
| | - Glenn A Walter
- Myology Institute, University of Florida, Gainesville, USA
- Department of Physiology and Aging, University of Florida, Gainesville, USA
| | - Jason Doles
- Department of Anatomy, Cell Biology and Physiology, Indiana university school of medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Sarah M. Judge
- Department of Physical Therapy, University of Florida, Gainesville, USA
- Myology Institute, University of Florida, Gainesville, USA
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida, Gainesville, USA
- Myology Institute, University of Florida, Gainesville, USA
| |
Collapse
|
34
|
Yuan L, Springer J, Palus S, Busquets S, Jové Q, Alves de Lima Junior E, Anker MS, von Haehling S, Álvarez Ladrón N, Millman O, Oosterlee A, Szymczyk A, López-Soriano FJ, Anker SD, Coats AJS, Argiles JM. The atypical β-blocker S-oxprenolol reduces cachexia and improves survival in a rat cancer cachexia model. J Cachexia Sarcopenia Muscle 2023; 14:653-660. [PMID: 36346141 PMCID: PMC9891926 DOI: 10.1002/jcsm.13116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Beta-blockers and selected stereoisomers of beta-blockers, like bisoprolol and S-pindolol (ACM-001), have been shown to be effective in preclinical cancer cachexia models. Here, we tested the efficacy of stereoisomers of oxprenolol in two preclinical models of cancer cachexia-the Yoshida AH-130 rat model and the Lewis lung carcinoma (LLC) mouse model. METHODS AND RESULTS In the Yoshida AH130 hepatoma rat cancer cachexia model and compared with placebo, 50 mg/kg/d S-oxprenolol (HR: 0.49, 95% CI: 0.28-0.85, P = 0.012) was superior to 50 mg/kg/d R-oxprenolol (HR: 0.83, 95% CI 0.38-1.45, P = 0.51) in reducing mortality (= reaching ethical endpoints). Combination of the three doses (12.5, 25 and 50 mg/kg/d) that had a significant effect on body weight loss in the S-oxprenolol groups vs the same combination of the R-oxprenolol groups lead to a significantly improved survival of S-oxprenolol vs R-oxprenolol (HR: 1.61, 95% CI: 1.08-2.39, P = 0.0185). Interestingly, there is a clear dose dependency in S-oxprenolol-treated (5, 12.5, 25 and 50 mg/kg/d) groups, which was not observed in groups treated with R-oxprenolol. A dose-dependent attenuation of weight and lean mass loss by S-oxprenolol was seen in the Yoshida rat model, whereas R-oxprenolol had only had a significant effect on fat mass. S-oxprenolol also non-significantly reduced weight loss in the LLC model and also improved muscle function (grip strength 428 ± 25 and 539 ± 37 g/100 g body weight for placebo and S-oxprenolol, respectively). However, there was only a minor effect on quality of life indicators food intake and spontaneous activity in the Yoshida model (25 mg/kg/S-oxprenolol: 11.9 ± 2.5 g vs placebo: 4.9 ± 0.8 g, P = 0.013 and also vs 25 mg/kg/d R-oxprenolol: 7.5 ± 2.6 g, P = 0.025). Both enantiomers had no effects on cardiac dimensions and function at the doses used in this study. Western blotting of proteins involved in the anabolic/catabolic homoeostasis suggest that anabolic signalling is persevered (IGF-1 receptor, Akt) and catabolic signalling is inhibited (FXBO-10, TRAF-6) by S-pindolol, but not he R-enantiomer. Expression of glucose transporters Glut1 and Glut 4 was similar in all groups, as was AMPK. CONCLUSIONS S-oxprenolol is superior to R-oxprenolol in cancer cachexia animal models and shows promise for a human application in cancer cachexia.
Collapse
Affiliation(s)
- Luping Yuan
- BIH Center for Regenerative Therapies, Charité-University Medical Center Berlin, Berlin, Germany
| | - Jochen Springer
- BIH Center for Regenerative Therapies, Charité-University Medical Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Palus
- BIH Center for Regenerative Therapies, Charité-University Medical Center Berlin, Berlin, Germany
| | - Silvia Busquets
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Queralt Jové
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Edson Alves de Lima Junior
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Markus S Anker
- BIH Center for Regenerative Therapies, Charité-University Medical Center Berlin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Cardiology (CBF), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medicine Goettingen (UMG), Goettingen, Germany.,German Center for Cardiovascular Research (DZHK), partner site Göttingen, Goettingen, Germany
| | - Natalia Álvarez Ladrón
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Oliver Millman
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Annemijn Oosterlee
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Agata Szymczyk
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Francisco Javier López-Soriano
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Stefan D Anker
- German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Berlin, Germany.,Department of Cardiology (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT) German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | - Josep M Argiles
- Departament de Bioquímica i Biomedicina Molecular, Cancer Research Group, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
35
|
Yao QY, Zhou J, Yao Y, Xue JS, Guo YC, Jian WZ, Zhang RW, Qiu XY, Zhou TY. An integrated PK/PD model investigating the impact of tumor size and systemic safety on animal survival in SW1990 pancreatic cancer xenograft. Acta Pharmacol Sin 2023; 44:465-474. [PMID: 35953645 PMCID: PMC9889390 DOI: 10.1038/s41401-022-00960-0] [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: 03/21/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023]
Abstract
Survival is one of the most important endpoints in cancer therapy, and parametric survival analysis could comprehensively reveal the overall result of disease progression, drug efficacy, toxicity as well as their interactions. In this study we investigated the efficacy and toxicity of dexamethasone (DEX) combined with gemcitabine (GEM) in pancreatic cancer xenograft. Nude mice bearing SW1990 pancreatic cancer cells derived tumor were treated with DEX (4 mg/kg, i.g.) and GEM (15 mg/kg, i.v.) alone or in combination repeatedly (QD, Q3D, Q7D) until the death of animal or the end of study. Tumor volumes and net body weight (NBW) were assessed every other day. Taking NBW as a systemic safety indicator, an integrated pharmacokinetic/pharmacodynamic (PK/PD) model was developed to quantitatively describe the impact of tumor size and systemic safety on animal survival. The PK/PD models with time course data for tumor size and NBW were established, respectively, in a sequential manner; a parametric time-to-event (TTE) model was also developed based on the longitudinal PK/PD models to describe the survival results of the SW1990 tumor-bearing mice. These models were evaluated and externally validated. Only the mice with good tumor growth inhibition and relatively stable NBW had an improved survival result after DEX and GEM combination therapy, and the simulations based on the parametric TTE model showed that NBW played more important role in animals' survival compared with tumor size. The established model in this study demonstrates that tumor size was not always the most important reason for cancer-related death, and parametric survival analysis together with safety issues was also important in the evaluation of oncology therapies in preclinical studies.
Collapse
Affiliation(s)
- Qing-Yu Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Ye Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jun-Sheng Xue
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Chen Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Wei-Zhe Jian
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ren-Wei Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiao-Yan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Tian-Yan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
36
|
Progressive development of melanoma-induced cachexia differentially impacts organ systems in mice. Cell Rep 2023; 42:111934. [PMID: 36640353 PMCID: PMC9983329 DOI: 10.1016/j.celrep.2022.111934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/12/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
Cachexia is a systemic wasting syndrome that increases cancer-associated mortality. How cachexia progressively and differentially impacts distinct tissues is largely unknown. Here, we find that the heart and skeletal muscle undergo wasting at early stages and are the tissues transcriptionally most impacted by cachexia. We also identify general and organ-specific transcriptional changes that indicate functional derangement by cachexia even in tissues that do not undergo wasting, such as the brain. Secreted factors constitute a top category of cancer-regulated genes in host tissues, and these changes include upregulation of the angiotensin-converting enzyme (ACE). ACE inhibition with the drug lisinopril improves muscle force and partially impedes cachexia-induced transcriptional changes, although wasting is not prevented, suggesting that cancer-induced host-secreted factors can regulate tissue function during cachexia. Altogether, by defining prevalent and temporal and tissue-specific responses to cachexia, this resource highlights biomarkers and possible targets for general and tissue-tailored anti-cachexia therapies.
Collapse
|
37
|
Di Girolamo D, Tajbakhsh S. Pathological features of tissues and cell populations during cancer cachexia. CELL REGENERATION 2022; 11:15. [PMID: 35441960 PMCID: PMC9021355 DOI: 10.1186/s13619-022-00108-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022]
Abstract
Cancers remain among the most devastating diseases in the human population in spite of considerable advances in limiting their impact on lifespan and healthspan. The multifactorial nature of cancers, as well as the number of tissues and organs that are affected, have exposed a considerable diversity in mechanistic features that are reflected in the wide array of therapeutic strategies that have been adopted. Cachexia is manifested in a number of diseases ranging from cancers to diabetes and ageing. In the context of cancers, a majority of patients experience cachexia and succumb to death due to the indirect effects of tumorigenesis that drain the energy reserves of different organs. Considerable information is available on the pathophysiological features of cancer cachexia, however limited knowledge has been acquired on the resident stem cell populations, and their function in the context of these diseases. Here we review current knowledge on cancer cachexia and focus on how tissues and their resident stem and progenitor cell populations are individually affected.
Collapse
|
38
|
Murphy BT, Mackrill JJ, O'Halloran KD. Impact of cancer cachexia on respiratory muscle function and the therapeutic potential of exercise. J Physiol 2022; 600:4979-5004. [PMID: 36251564 PMCID: PMC10091733 DOI: 10.1113/jp283569] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer cachexia is defined as a multi-factorial syndrome characterised by an ongoing loss of skeletal muscle mass and progressive functional impairment, estimated to affect 50-80% of patients and responsible for 20% of cancer deaths. Elevations in the morbidity and mortality rates of cachectic cancer patients has been linked to respiratory failure due to atrophy and dysfunction of the ventilatory muscles. Despite this, there is a distinct scarcity of research investigating the structural and functional condition of the respiratory musculature in cancer, with the majority of studies exclusively focusing on limb muscle. Treatment strategies are largely ineffective in mitigating the cachectic state. It is now widely accepted that an efficacious intervention will likely combine elements of pharmacology, nutrition and exercise. However, of these approaches, exercise has received comparatively little attention. Therefore, it is unlikely to be implemented optimally, whether in isolation or combination. In consideration of these limitations, the current review describes the mechanistic basis of cancer cachexia and subsequently explores the available respiratory- and exercise-focused literature within this context. The molecular basis of cachexia is thoroughly reviewed. The pivotal role of inflammatory mediators is described. Unravelling the mechanisms of exercise-induced support of muscle via antioxidant and anti-inflammatory effects in addition to promoting efficient energy metabolism via increased mitochondrial biogenesis, mitochondrial function and muscle glucose uptake provide avenues for interventional studies. Currently available pre-clinical mouse models including novel transgenic animals provide a platform for the development of multi-modal therapeutic strategies to protect respiratory muscles in people with cancer.
Collapse
Affiliation(s)
- Ben T. Murphy
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - John J. Mackrill
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| | - Ken D. O'Halloran
- Department of PhysiologySchool of MedicineCollege of Medicine and HealthUniversity College CorkCorkIreland
| |
Collapse
|
39
|
Baraldo M, Zorzato S, Dondjang AHT, Geremia A, Nogara L, Dumitras AG, Canato M, Marcucci L, Nolte H, Blaauw B. Inducible deletion of raptor and mTOR from adult skeletal muscle impairs muscle contractility and relaxation. J Physiol 2022; 600:5055-5075. [PMID: 36255030 DOI: 10.1113/jp283686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 01/05/2023] Open
Abstract
Skeletal muscle weakness has been associated with different pathological conditions, including sarcopenia and muscular dystrophy, and is accompanied by altered mammalian target of rapamycin (mTOR) signalling. We wanted to elucidate the functional role of mTOR in muscle contractility. Most loss-of-function studies for mTOR signalling have used the drug rapamycin to inhibit some of the signalling downstream of mTOR. However, given that rapamycin does not inhibit all mTOR signalling completely, we generated a double knockout for mTOR and for the scaffold protein of mTORC1, raptor, in skeletal muscle. We found that double knockout in mice results in a more severe phenotype compared with deletion of raptor or mTOR alone. Indeed, these animals display muscle weakness, increased fibre denervation and a slower muscle relaxation following tetanic stimulation. This is accompanied by a shift towards slow-twitch fibres and changes in the expression levels of calcium-related genes, such as Serca1 and Casq1. Double knockout mice show a decrease in calcium decay kinetics after tetanus in vivo, suggestive of a reduced calcium reuptake. In addition, RNA sequencing analysis revealed that many downregulated genes, such as Tcap and Fhod3, are linked to sarcomere organization. These results suggest a key role for mTOR signalling in maintaining proper fibre relaxation in skeletal muscle. KEY POINTS: Skeletal muscle wasting and weakness have been associated with different pathological conditions, including sarcopenia and muscular dystrophy, and are accompanied by altered mammalian target of rapamycin (mTOR) signalling. Mammalian target of rapamycin plays a crucial role in the maintenance of muscle mass and functionality. We found that the loss of both mTOR and raptor results in contractile abnormalities, with severe muscle weakness and delayed relaxation following tetanic stimulation. These results are associated with alterations in the expression of genes involved in sarcomere organization and calcium handling and with an impairment in calcium reuptake after contraction. Taken together, these results provide a mechanistic insight into the role of mTOR in muscle contractility.
Collapse
Affiliation(s)
- Martina Baraldo
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Sabrina Zorzato
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Achille Homère Tchampda Dondjang
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Alessia Geremia
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Leonardo Nogara
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ana Georgia Dumitras
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marta Canato
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Center for Biosystems Dynamics Research, RIKEN, Suita, Japan
| | - Hendrik Nolte
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Bert Blaauw
- Venetian Institute of Molecular Medicine (VIMM), Padova, Italy.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
40
|
Yu YC, Ahmed A, Lai HC, Cheng WC, Yang JC, Chang WC, Chen LM, Shan YS, Ma WL. Review of the endocrine organ-like tumor hypothesis of cancer cachexia in pancreatic ductal adenocarcinoma. Front Oncol 2022; 12:1057930. [PMID: 36465353 PMCID: PMC9713001 DOI: 10.3389/fonc.2022.1057930] [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: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 08/30/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal types of solid tumors, associated with a high prevalence of cachexia (~80%). PDAC-derived cachexia (PDAC-CC) is a systemic disease involving the complex interplay between the tumor and multiple organs. The endocrine organ-like tumor (EOLT) hypothesis may explain the systemic crosstalk underlying the deleterious homeostatic shifts that occur in PDAC-CC. Several studies have reported a markedly heterogeneous collection of cachectic mediators, signaling mechanisms, and metabolic pathways, including exocrine pancreatic insufficiency, hormonal disturbance, pro-inflammatory cytokine storm, digestive and tumor-derived factors, and PDAC progression. The complexities of PDAC-CC necessitate a careful review of recent literature summarizing cachectic mediators, corresponding metabolic functions, and the collateral impacts on wasting organs. The EOLT hypothesis suggests that metabolites, genetic instability, and epigenetic changes (microRNAs) are involved in cachexia development. Both tumors and host tissues can secrete multiple cachectic factors (beyond only inflammatory mediators). Some regulatory molecules, metabolites, and microRNAs are tissue-specific, resulting in insufficient energy production to support tumor/cachexia development. Due to these complexities, changes in a single factor can trigger bi-directional feedback circuits that exacerbate PDAC and result in the development of irreversible cachexia. We provide an integrated review based on 267 papers and 20 clinical trials from PubMed and ClinicalTrials.gov database proposed under the EOLT hypothesis that may provide a fundamental understanding of cachexia development and response to current treatments.
Collapse
Affiliation(s)
- Ying-Chun Yu
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, Center for Tumor Biology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Azaj Ahmed
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, Center for Tumor Biology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Juan-Chern Yang
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chang
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, Center for Tumor Biology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Lu-Min Chen
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Yan-Shen Shan
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Chen Kung University, Tainan, Taiwan
| | - Wen-Lung Ma
- Department of Medical Research, Department of Obstetrics and Gynecology, Department of Gastroenterology, and Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, Center for Tumor Biology, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
41
|
Identification of Potential Biomarkers for Cancer Cachexia and Anti-Fn14 Therapy. Cancers (Basel) 2022; 14:cancers14225533. [PMID: 36428623 PMCID: PMC9688504 DOI: 10.3390/cancers14225533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Developing therapies for cancer cachexia has not been successful to date, in part due to the challenges of achieving robust quantitative measures as a readout of patient treatment. Hence, identifying biomarkers to assess the outcomes of treatments for cancer cachexia is of great interest and important for accelerating future clinical trials. METHODS We established a novel xenograft model for cancer cachexia with a cachectic human PC3* cell line, which was responsive to anti-Fn14 mAb treatment. Using RNA-seq and secretomic analysis, genes differentially expressed in cachectic and non-cachectic tumors were identified and validated by digital droplet PCR (ddPCR). Correlation analysis was performed to investigate their impact on survival in cancer patients. RESULTS A total of 46 genes were highly expressed in cachectic PC3* tumors, which were downregulated by anti-Fn14 mAb treatment. High expression of the top 10 candidates was correlated with low survival and high cachexia risk in different cancer types. Elevated levels of LCN2 were observed in serum samples from cachectic patients compared with non-cachectic cancer patients. CONCLUSION The top 10 candidates identified in this study are candidates as potential biomarkers for cancer cachexia. The diagnostic value of LCN2 in detecting cancer cachexia is confirmed in patient samples.
Collapse
|
42
|
Ferrara M, Samaden M, Ruggieri E, Vénéreau E. Cancer cachexia as a multiorgan failure: Reconstruction of the crime scene. Front Cell Dev Biol 2022; 10:960341. [PMID: 36158184 PMCID: PMC9493094 DOI: 10.3389/fcell.2022.960341] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cachexia is a devastating syndrome associated with the end-stage of several diseases, including cancer, and characterized by body weight loss and severe muscle and adipose tissue wasting. Although different cancer types are affected to diverse extents by cachexia, about 80% of all cancer patients experience this comorbidity, which highly reduces quality of life and response to therapy, and worsens prognosis, accounting for more than 25% of all cancer deaths. Cachexia represents an urgent medical need because, despite several molecular mechanisms have been identified, no effective therapy is currently available for this devastating syndrome. Most studies focus on skeletal muscle, which is indeed the main affected and clinically relevant organ, but cancer cachexia is characterized by a multiorgan failure. In this review, we focus on the current knowledge on the multiple tissues affected by cachexia and on the biomarkers with the attempt to define a chronological pathway, which might be useful for the early identification of patients who will undergo cachexia. Indeed, it is likely that the inefficiency of current therapies might be attributed, at least in part, to their administration in patients at the late stages of cachexia.
Collapse
Affiliation(s)
- Michele Ferrara
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Samaden
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Ruggieri
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emilie Vénéreau
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
43
|
Wu KC, Chu PC, Cheng YJ, Li CI, Tian J, Wu HY, Wu SH, Lai YC, Kao HH, Hsu AL, Lin HW, Lin CH. Development of a traditional Chinese medicine-based agent for the treatment of cancer cachexia. J Cachexia Sarcopenia Muscle 2022; 13:2073-2087. [PMID: 35718751 PMCID: PMC9397559 DOI: 10.1002/jcsm.13028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Despite recent advances in understanding the pathophysiology of cancer cachexia, prevention/treatment of this debilitating disease remains an unmet medical need. METHODS We developed an integrated, multi-tiered strategy involving both in vitro and in vivo muscle atrophy platforms to identify traditional Chinese medicine (TCM)-based anti-cachectic agents. In the initial screening, we used inflammatory cytokine-induced atrophy of C2C12 myotubes as a phenotypic screening platform to assess the protective effects of TCMs. The selected TCMs were then evaluated for their abilities to protect Caenorhabditis elegans from age-related reduction of mobility and contractility, followed by the C-26 colon adenocarcinoma mouse model of cachexia to confirm the anti-muscle atrophy effects (body/skeletal muscle weights, fibre size distribution, grip strengths, and serum IL-6). Transcriptome analysis, quantitative real-time polymerase chain reaction, and immunoblotting were performed to gain understanding of the potential mechanism(s) by which effective TCM protected against C26 tumour-induced muscle atrophy. RESULTS Of 29 widely used TCMs, Dioscorea radix (DR) and Mu Dan Pi (MDP) showed a complete protection (all P values, 0.0002) vis-à-vis C26 conditioned medium control in the myotube atrophy platform. MDP exhibited a unique ability to ameliorate age-associated decreases in worm mobility, accompanied by improved total body contractions, relative to control (P < 0.0001 and <0.01, respectively), which, however, was not noted with DR. This differential in vivo protective effect between MDP and DR was also confirmed in the C-26 mouse model. MDP at 1000 mg/kg (MDP-H) was effective in protecting body weight loss (P < 0.05) in C-26 tumour-bearing mice without changing food or water intake, accompanied by the restoration of the fibre size distribution of hindleg skeletal muscles (P < 0.0001) and the forelimb grip strength (P < 0.05). MDP-treated C-26-tumour-bearing mice were alert, showed normal posture and better body conditions, and exhibited lower serum IL-6 levels (P = 0.06) relative to vehicle control. This decreased serum IL-6 was associated with the in vitro suppressive effect of MDP (25 and 50 μg/mL) on IL-6 secretion into culture medium by C26 cells. RNA-seq analysis, followed by quantitative real-time polymerase chain reaction and/or immunoblotting, shows that MDP's anti-cachectic effect was attributable to its ability to reverse the C-26 tumour-induced re-programming of muscle homoeostasis-associated gene expression, including that of two cachexia drivers (MuRF1 and Atrogin-1), in skeletal muscles. CONCLUSIONS All these findings suggest the translational potential of MDP to foster new strategies for the prevention and/or treatment of cachexia. The protective effect of MDP on other types of muscle atrophy such as sarcopenia might warrant investigations.
Collapse
Affiliation(s)
- Kun-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Po-Chen Chu
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Rehabilitation, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Ing Li
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jingkui Tian
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Hsing-Yu Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Szu-Hsien Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Yi-Chun Lai
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan
| | - Hsiang-Han Kao
- Department of Family Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ao-Lin Hsu
- Research Center for Healthy Aging, China Medical University, Taichung, Taiwan.,PhD Program for Aging, China Medical University, Taichung, Taiwan.,Department of Internal Medicine, Division of Geriatrics & Palliative Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hsiang-Wen Lin
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan.,Department of Pharmacy, China Medical University Hospital, Taichung, Taiwan.,Department of Pharmacy System, Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Chih-Hsueh Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Geriatric Medicine, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
44
|
Chakedis JM, Dillhoff ME, Schmidt CR, Rajasekera PV, Evans DC, Williams TM, Guttridge DC, Talbert EE. Identification of circulating plasma ceramides as a potential sexually dimorphic biomarker of pancreatic cancer-induced cachexia. JCSM RAPID COMMUNICATIONS 2022; 5:254-265. [PMID: 36591536 PMCID: PMC9797184 DOI: 10.1002/rco2.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 06/01/2022] [Indexed: 06/17/2023]
Abstract
Background Cancer patients who exhibit cachexia lose weight and have low treatment tolerance and poor outcomes compared to cancer patients without weight loss. Despite the clear increased risk for patients, diagnosing cachexia still often relies on self-reported weight loss. A reliable biomarker to identify patients with cancer cachexia would be a valuable tool to improve clinical decision making and identification of patients at risk of adverse outcomes. Methods Targeted metabolomics, that included panels of amino acids, tricarboxylic acids, fatty acids, acylcarnitines, and sphingolipids, were conducted on plasma samples from patients with confirmed pancreatic ductal adenocarcinoma (PDAC) with and without cachexia and control patients without cancer (n=10/group, equally divided by sex). Additional patient samples were analyzed (total n=95) and Receiver Operating Characteristic (ROC) analyses were performed to establish if any metabolite could effectively serve as a biomarker of cachexia. Results Targeted profiling revealed that cachectic patients had decreased circulating levels of three sphingolipids compared to either non-cachectic PDAC patients or patients without cancer. The ratio of C18-ceramide to C24-ceramide (C18:C24) outperformed a number of other previously proposed biomarkers of cachexia (area under ROC = 0.810). It was notable that some biomarkers, including C18:C24, were only altered in cachectic males. Conclusions Our findings identify C18:C24 as a potentially new biomarker of PDAC-induced cachexia that also highlight a previously unappreciated sexual dimorphism in cancer cachexia.
Collapse
Affiliation(s)
- Jeffery M. Chakedis
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
- Present Address: Department of General Surgery, The Permanente Medical Group, Kaiser Permanente Walnut Creek Medical Center, Walnut Creek, CA 94596, USA
| | - Mary E. Dillhoff
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Carl R. Schmidt
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, The Ohio State University, Columbus, OH 43210, USA
- Present Address: Department of Surgery, West Virginia University, Morgantown, WV 26506
| | - Priyani V. Rajasekera
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - David C. Evans
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Division of Trauma, Critical Care, and Burn, The Ohio State University, Columbus, OH 43210, USA
- Present Address: OhioHealth Trauma Services, Columbus, OH 43215, USA
| | - Terence M. Williams
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, USA
- Present Address: Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, 91010 USA
| | - Denis C. Guttridge
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Erin E. Talbert
- Arthur G. James Comprehensive Cancer Center Cancer Cachexia Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Present Address: Department of Health and Human Physiology and Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
45
|
Which anthropometric measurement is better for predicting survival of patients with cancer cachexia? Br J Nutr 2022; 127:1849-1857. [PMID: 34325763 DOI: 10.1017/s0007114521002853] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
No relevant studies have yet been conducted to explore which measurement can best predict the survival time of patients with cancer cachexia. This study aimed to identify an anthropometric measurement that could predict the 1-year survival of patients with cancer cachexia. We conducted a nested case-control study using data from a multicentre clinical investigation of cancer from 2013 to 2020. Cachexia was defined using the Fearon criteria. A total of 262 patients who survived less than 1 year and 262 patients who survived more than 1 year were included in this study. Six candidate variables were selected based on clinical experience and previous studies. Five variables, BMI, mid-arm circumference, mid-arm muscle circumference, calf circumference and triceps skin fold (TSF), were selected for inclusion in the multivariable model. In the conditional logistic regression analysis, TSF (P = 0·014) was identified as a significant independent protective factor. A similar result was observed in all patients with cancer cachexia (n 3084). In addition, a significantly stronger positive association between TSF and the 1-year survival of patients with cancer cachexia was observed in participants aged > 65 years (OR: 0·94; 95 % CI 0·89, 0·99) than in those aged ≤ 65 years (OR: 0·96; 95 % CI 0·93, 0·99; Pinteraction = 0·013) and in participants with no chronic disease (OR: 0·92; 95 % CI 0·87, 0·97) than in those with chronic disease (OR: 0·97; 95 % CI 0·94, 1·00; Pinteraction = 0·049). According to this study, TSF might be a good anthropometric measurement for predicting 1-year survival in patients with cancer cachexia.
Collapse
|
46
|
Hardee JP, Carson JA. Muscular contraction's therapeutic potential for cancer-induced wasting. Am J Physiol Cell Physiol 2022; 323:C378-C384. [PMID: 35704693 PMCID: PMC9359654 DOI: 10.1152/ajpcell.00021.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle atrophy and dysfunction contribute to cancer patient morbidity and mortality. Cachexia pathophysiology is highly complex, given that perturbations to the systemic cancer environment and the interaction with diverse tissues can contribute to wasting processes. Systemic interleukin 6 (IL-6) and glycoprotein 130 (gp130) receptor signaling have established roles in some types of cancer-induced muscle wasting through disruptions to protein turnover and oxidative capacity. While exercise has documented benefits for cancer prevention and patient survival, there are significant gaps in our understanding of muscle adaptation and plasticity during severe cachexia. Preclinical models have provided valuable insight into the adaptive potential of muscle to contraction within the cancer environment. We summarize the current understanding of how resistance-type exercise impacts mechanisms involved in cancer-induced muscle atrophy and dysfunction. Specifically, the role of IL-6 and gp130 receptor in the pathophysiology of muscle wasting and the adaptive response to exercise is explained. The discussion includes current knowledge gaps and future research directions needed to improve preclinical research and accelerate clinical translation in human cancer patients.
Collapse
Affiliation(s)
- Justin P Hardee
- Centre for Muscle Research, Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
| | - James A Carson
- Center for Muscle Metabolism & Neuropathology, Division of Rehabilitation Sciences, University of Tennessee Health Science Center, Memphis, TN, United States.,College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
47
|
Wang R, Kumar B, Doud EH, Mosley AL, Alexander MS, Kunkel LM, Nakshatri H. Skeletal muscle-specific overexpression of miR-486 limits mammary tumor-induced skeletal muscle functional limitations. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:231-248. [PMID: 35402076 PMCID: PMC8971682 DOI: 10.1016/j.omtn.2022.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/12/2022] [Indexed: 11/28/2022]
Abstract
miR-486 is a myogenic microRNA, and its reduced skeletal muscle expression is observed in muscular dystrophy. Transgenic overexpression of miR-486 using muscle creatine kinase promoter (MCK-miR-486) partially rescues muscular dystrophy phenotype. We had previously demonstrated reduced circulating and skeletal muscle miR-486 levels with accompanying skeletal muscle defects in mammary tumor models. To determine whether skeletal muscle miR-486 is functionally similar in dystrophies and cancer, we performed functional limitations and biochemical studies of skeletal muscles of MMTV-Neu mice that mimic HER2+ breast cancer and MMTV-PyMT mice that mimic luminal subtype B breast cancer and these mice crossed to MCK-miR-486 mice. miR-486 significantly prevented tumor-induced reduction in muscle contraction force, grip strength, and rotarod performance in MMTV-Neu mice. In this model, miR-486 reversed cancer-induced skeletal muscle changes, including loss of p53, phospho-AKT, and phospho-laminin alpha 2 (LAMA2) and gain of hnRNPA0 and SRSF10 phosphorylation. LAMA2 is a part of the dystrophin-associated glycoprotein complex, and its loss of function causes congenital muscular dystrophy. Complementing these beneficial effects on muscle, miR-486 indirectly reduced tumor growth and improved survival, which is likely due to systemic effects of miR-486 on production of pro-inflammatory cytokines such as IL-6. Thus, similar to dystrophy, miR-486 has the potential to reverse skeletal muscle defects and cancer burden.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma H. Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber L. Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S. Alexander
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
| | - Louis M. Kunkel
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
48
|
Balnis J, Drake LA, Singer DV, Vincent CE, Korponay TC, D’Armiento J, Lee CG, Elias JA, Singer HA, Jaitovich A. Deaccelerated Myogenesis and Autophagy in Genetically Induced Pulmonary Emphysema. Am J Respir Cell Mol Biol 2022; 66:623-637. [PMID: 35286819 PMCID: PMC9163640 DOI: 10.1165/rcmb.2021-0351oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/31/2022] [Indexed: 01/18/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD)-pulmonary emphysema often develop locomotor muscle dysfunction, which entails reduced muscle mass and force-generation capacity and is associated with worse outcomes, including higher mortality. Myogenesis contributes to adult muscle integrity during injury-repair cycles. Injurious events crucially occur in the skeletal muscles of patients with COPD in the setting of exacerbations and infections, which lead to acute decompensations for limited periods of time, after which patients typically fail to recover the baseline status they had before the acute event. Autophagy, which is dysregulated in muscles from patients with COPD, is a key regulator of muscle stem-satellite- cells activation and myogenesis, yet very little research has so far mechanistically investigated the role of autophagy dysregulation in COPD muscles. Using a genetically inducible interleukin-13-driven pulmonary emphysema model leading to muscle dysfunction, and confirmed with a second genetic animal model, we found a significant myogenic dysfunction associated with the reduced proliferative capacity of satellite cells. Transplantation experiments followed by lineage tracing suggest that an intrinsic defect in satellite cells, and not in the COPD environment, plays a dominant role in the observed myogenic dysfunction. RNA sequencing analysis and direct observation of COPD mice satellite cells suggest dysregulated autophagy. Moreover, while autophagy flux experiments with bafilomycin demonstrated deacceleration of autophagosome turnover in COPD mice satellite cells, spermidine-induced autophagy stimulation leads to a higher replication rate and myogenesis in these animals. Our data suggest that pulmonary emphysema causes disrupted myogenesis, which could be improved with stimulation of autophagy and satellite cells activation, leading to an attenuated muscle dysfunction.
Collapse
Affiliation(s)
- Joseph Balnis
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Lisa A. Drake
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Diane V. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Catherine E. Vincent
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Tanner C. Korponay
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Jeanine D’Armiento
- Departments of Anesthesiology and Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York; and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island
| | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine and
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
49
|
Graca FA, Rai M, Hunt LC, Stephan A, Wang YD, Gordon B, Wang R, Quarato G, Xu B, Fan Y, Labelle M, Demontis F. The myokine Fibcd1 is an endogenous determinant of myofiber size and mitigates cancer-induced myofiber atrophy. Nat Commun 2022; 13:2370. [PMID: 35501350 PMCID: PMC9061726 DOI: 10.1038/s41467-022-30120-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/14/2022] [Indexed: 12/19/2022] Open
Abstract
Decline in skeletal muscle cell size (myofiber atrophy) is a key feature of cancer-induced wasting (cachexia). In particular, atrophy of the diaphragm, the major muscle responsible for breathing, is an important determinant of cancer-associated mortality. However, therapeutic options are limited. Here, we have used Drosophila transgenic screening to identify muscle-secreted factors (myokines) that act as paracrine regulators of myofiber growth. Subsequent testing in mouse myotubes revealed that mouse Fibcd1 is an evolutionary-conserved myokine that preserves myofiber size via ERK signaling. Local administration of recombinant Fibcd1 (rFibcd1) ameliorates cachexia-induced myofiber atrophy in the diaphragm of mice bearing patient-derived melanoma xenografts and LLC carcinomas. Moreover, rFibcd1 impedes cachexia-associated transcriptional changes in the diaphragm. Fibcd1-induced signaling appears to be muscle selective because rFibcd1 increases ERK activity in myotubes but not in several cancer cell lines tested. We propose that rFibcd1 may help reinstate myofiber size in the diaphragm of patients with cancer cachexia.
Collapse
Affiliation(s)
- Flavia A Graca
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Liam C Hunt
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Brittney Gordon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
- Xenograft Core, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Ruishan Wang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Giovanni Quarato
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Beisi Xu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yiping Fan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Myriam Labelle
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States.
- Solid Tumor Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, TN, United States.
| |
Collapse
|
50
|
Fukui K, Nomura M, Kishimoto K, Tanuma N, Kurosawa K, Kanazawa K, Kato H, Sato T, Miura S, Miura K, Sato I, Tsuji H, Yamashita Y, Tamai K, Watanabe T, Yasuda J, Tanaka T, Satoh K, Furukawa T, Jingu K, Shima H. PP6 deficiency in mice with KRAS mutation and Trp53 loss promotes early death by PDAC with cachexia-like features. Cancer Sci 2022; 113:1613-1624. [PMID: 35247012 PMCID: PMC9128171 DOI: 10.1111/cas.15315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022] Open
Abstract
To examine effects of PP6 gene (Ppp6c) deficiency on pancreatic tumor development, we developed pancreas-specific, tamoxifen-inducible Cre-mediated KP (KRAS(G12D) plus Trp53-deficient) mice (cKP mice) and crossed them with Ppp6cflox / flox mice. cKP mice with the homozygous Ppp6c deletion developed pancreatic tumors, became emaciated and required euthanasia within 150 days of mutation induction, phenotypes that were not seen in heterozygous or wild-type (WT) mice. At 30 days, a comparative analysis of genes commonly altered in homozygous versus WT Ppp6c cKP mice revealed enhanced activation of Erk and NFκB pathways in homozygotes. By 80 days, the number and size of tumors and number of precancerous lesions had significantly increased in the pancreas of Ppp6c homozygous relative to heterozygous or WT cKP mice. Ppp6c-/- tumors were pathologically diagnosed as pancreatic ductal adenocarcinoma (PDAC) undergoing the epithelial-mesenchymal transition (EMT), and cancer cells had invaded surrounding tissues in three out of six cases. Transcriptome and metabolome analyses indicated an enhanced cancer-specific glycolytic metabolism in Ppp6c-deficient cKP mice and the increased expression of inflammatory cytokines. Individual Ppp6c-/- cKP mice showed weight loss, decreased skeletal muscle and adipose tissue, and increased circulating tumor necrosis factor (TNF)-α and IL-6 levels, suggestive of systemic inflammation. Overall, Ppp6c deficiency in the presence of K-ras mutations and Trp53 gene deficiency promoted pancreatic tumorigenesis with generalized cachexia and early death. This study provided the first evidence that Ppp6c suppresses mouse pancreatic carcinogenesis and supports the use of Ppp6c-deficient cKP mice as a model for developing treatments for cachexia associated with pancreatic cancer.
Collapse
Affiliation(s)
- Katsuya Fukui
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
- Department of Radiation OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Miyuki Nomura
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Kazuhiro Kishimoto
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
- Department of Head and Neck SurgeryKanazawa Medical UniversityKanazawaJapan
| | - Nobuhiro Tanuma
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
| | - Koreyuki Kurosawa
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
- Department of Plastic and Reconstructive SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Kosuke Kanazawa
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
- Division of SurgeryMiyagi Cancer CenterNatoriJapan
| | - Hiroyuki Kato
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Tomoki Sato
- Laboratory of Nutritional BiochemistryGraduate School of Nutritional and Environmental SciencesUniversity of ShizuokaShizuokaJapan
| | - Shinji Miura
- Laboratory of Nutritional BiochemistryGraduate School of Nutritional and Environmental SciencesUniversity of ShizuokaShizuokaJapan
| | - Koh Miura
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of SurgeryMiyagi Cancer CenterNatoriJapan
| | - Ikuro Sato
- Division of PathologyMiyagi Cancer CenterNatoriJapan
| | - Hiroyuki Tsuji
- Department of Head and Neck SurgeryKanazawa Medical UniversityKanazawaJapan
| | - Yoji Yamashita
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Keiichi Tamai
- Division of Cancer Stem CellMiyagi Cancer Center Research InstituteNatoriJapan
| | - Toshio Watanabe
- Department of Biological ScienceGraduate School of Humanities and SciencesNara Women’s UniversityNaraJapan
| | - Jun Yasuda
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
- Division of Molecular Cellular OncologyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Takuji Tanaka
- Research Center of Diagnostic PathologyGifu Municipal HospitalGifuJapan
| | - Kennichi Satoh
- Division of GastroenterologyTohoku Medical Pharmaceutical UniversitySendaiJapan
| | - Toru Furukawa
- Department of Investigative PathologyTohoku University Graduate School of MedicineSendaiJapan
| | - Keiichi Jingu
- Department of Radiation OncologyTohoku University Graduate School of MedicineSendaiJapan
| | - Hiroshi Shima
- Division of Cancer ChemotherapyMiyagi Cancer Center Research InstituteNatoriJapan
- Division of Cancer Molecular BiologyTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|