1
|
Zhang L, Li Q, Wu M, Feng X, Dai W, Chen P, Chen D, Zheng Z, Lin X, Wei G. TRIM22 governs tumorigenesis and protects against endometrial cancer-associated cachexia by inhibiting inflammatory response and adipose thermogenic activity. Cancer Metab 2025; 13:17. [PMID: 40200303 PMCID: PMC11980105 DOI: 10.1186/s40170-025-00386-2] [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: 12/03/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Endometrial cancer (EC) is one of the most common cancers in women, with a short overall survival and poor prognosis. Besides the biologically aggressive EC properties, Cancer-associated cachexia is the main factor. However, the detailed mechanism underlying EC-related cachexia and its harmful effects on EC progression and patient prognosis remains unclear. METHODS For clinical specimen and the vitro experiment, we detected TRIM22 expression level, EC patients' survival time, EC cell functional change, and adipose thermogenic changes to identify the function of TRIM22 in EC progression, EC-associated cachexia, and their molecular mechanisms. Then, for the vivo experiment, we exploited the xenografts in mice to identify the function of TRIM22 again, and to screen the drug therapeutic schedule. RESULTS Herein, we demonstrated that TRIM22 inhibited EC cell growth, invasion, and migration. Interleukin (IL)-6 mediated brown adipose tissue activation and white adipose tissue browning which induced EC-related cachexia. TRIM22 suppressed the EC cells' secretion of IL-6, and IL-6 mediated EC-related cachexia. Mechanistically, TRIM22 inhibited EC progression by suppressing the nucleotide-binding oligomerization domain 2(NOD2)/nuclear factor-kappaB (NF-κB) signaling pathway, with the purpose of impeding the production of IL-6. Moreover, we revealed that TRIM22 inhibited EC-associated cachexia by suppressing the IL-6/IL-6 receptor (IL-6R) signaling pathway. Therapeutically, we demonstrated that combination treatment with a TRIM22 inducer (progesterone) and a thermogenic inhibitor (IL-6R antibody) synergistically augmented the antitumor efficacy of carbotaxol (carboplatin and paclitaxel), in vivo. CONCLUSION Our data reveals that TRIM22-EC-IL-6-cachexia cross-communication has important clinical relevance and that the use of combined therapy holds great promise for enhancing the efficacy of anti-ECs. (Fig. graphical abstract).
Collapse
Affiliation(s)
- Liping Zhang
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Quanrong Li
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Meiting Wu
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiushan Feng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Weichao Dai
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Peifang Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Dezhao Chen
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Zhiqun Zheng
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001, Fujian Province, China.
| | - Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
2
|
Wyss MT, Heuer C, Herwerth M. The bumpy road of purinergic inhibitors to clinical application in immune-mediated diseases. Neural Regen Res 2024; 19:1206-1211. [PMID: 37905866 PMCID: PMC11467927 DOI: 10.4103/1673-5374.386405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Purinergic signaling plays important roles throughout the body in the regulation of organ functions during and following the disruption of homeostasis. This is also reflected by the widespread expression of two families of purinergic receptors (P1 and P2) with numerous subtypes. In the last few decades, there has been increasing evidence that purinergic signaling plays an important role in the regulation of immune functions. Mainly, signals mediated by P2 receptors have been shown to contribute to immune system-mediated pathologies. Thus, interference with P2 receptors may be a promising strategy for the modulation of immune responses. Although only a few clinical studies have been conducted in isolated entities with limited success, preclinical work suggests that the use of P2 receptor inhibitors may bear some promise in various autoimmune diseases. Despite the association of P2 receptors with several disorders from this field, the use of P2 receptor antagonists in clinical therapy is still very scarce. In this narrative review, we briefly review the involvement of the purinergic system in immunological responses and clinical studies on the effect of purinergic inhibition on autoimmune processes. We then open the aperture a bit and show some preclinical studies demonstrating a potential effect of purinergic blockade on autoimmune events. Using suramin, a non-specific purinergic inhibitor, as an example, we further show that off-target effects could be responsible for observed effects in immunological settings, which may have interesting implications. Overall, we believe that it is worthwhile to further investigate this hitherto underexplored area.
Collapse
Affiliation(s)
- Matthias T. Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
| | - Christine Heuer
- Neurology Department, University Hospital of Zurich, Zürich, Switzerland
| | - Marina Herwerth
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Neurology Department, University Hospital of Zurich, Zürich, Switzerland
| |
Collapse
|
3
|
Cancer Cachexia: Signaling and Transcriptional Regulation of Muscle Catabolic Genes. Cancers (Basel) 2022; 14:cancers14174258. [PMID: 36077789 PMCID: PMC9454911 DOI: 10.3390/cancers14174258] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary An uncontrollable loss in the skeletal muscle of cancer patients which leads to a significant reduction in body weight is clinically referred to as cancer cachexia (CC). While factors derived from the tumor environment which trigger various signaling pathways have been identified, not much progress has been made clinically to effectively prevent muscle loss. Deeper insights into the transcriptional and epigenetic regulation of muscle catabolic genes may shed light on key regulators which can be targeted to develop new therapeutic avenues. Abstract Cancer cachexia (CC) is a multifactorial syndrome characterized by a significant reduction in body weight that is predominantly caused by the loss of skeletal muscle and adipose tissue. Although the ill effects of cachexia are well known, the condition has been largely overlooked, in part due to its complex etiology, heterogeneity in mediators, and the involvement of diverse signaling pathways. For a long time, inflammatory factors have been the focus when developing therapeutics for the treatment of CC. Despite promising pre-clinical results, they have not yet advanced to the clinic. Developing new therapies requires a comprehensive understanding of how deregulated signaling leads to catabolic gene expression that underlies muscle wasting. Here, we review CC-associated signaling pathways and the transcriptional cascade triggered by inflammatory cytokines. Further, we highlight epigenetic factors involved in the transcription of catabolic genes in muscle wasting. We conclude with reflections on the directions that might pave the way for new therapeutic approaches to treat CC.
Collapse
|
4
|
Kouzu K, Tsujimoto H, Ishibashi Y, Shinada H, Oikawa I, Kishi Y, Shinomiya N, Ueno H. Preoperative Fall Risk Assessment Score as a Prognostic Factor in Esophageal Cancer Patients after Esophagectomy. J Clin Med 2021; 10:jcm10245966. [PMID: 34945262 PMCID: PMC8709201 DOI: 10.3390/jcm10245966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 11/17/2022] Open
Abstract
The current study investigated the impact of preoperative fall risk assessment score (FRAS) on long-term prognoses in patients with esophageal cancer (EC). A total of 161 patients with EC who underwent curative surgery were classified into a high-risk (95, 41.0%) and low-risk (66, 41.0%) groups according to their FRAS. This study investigated the relationships between the FRAS and clinicopathological findings and prognoses. Accordingly, patients in the high-risk group were significantly older and had a significantly higher Charlson comorbidity index than those in the low-risk group. No significant difference was found in pathological findings between both groups. The high-risk group had significantly lower overall survival (OS) and relapse-free survival (RFS) rates than the low-risk group (p = 0.004 and 0.001, respectively). Multivariate analysis identified high FRAS as an independent prognostic factor for poor OS, with a hazard ratio of 1.75 (p = 0.033). Moreover, re-analysis of the data after excluding age as a category showed that the high-risk group had significantly worse OS (p = 0.004) and RFS (p = 0.003) than the low-risk group. The FRAS can, therefore, be considered a useful method for assessing frailty and a potential prognostic factor for EC.
Collapse
Affiliation(s)
- Keita Kouzu
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
- Correspondence: ; Tel.: +81-4-2995-1637
| | - Yusuke Ishibashi
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| | - Hanae Shinada
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| | - Isawo Oikawa
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, Saitama 359-0042, Japan;
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Saitama 359-0042, Japan; (K.K.); (Y.I.); (H.S.); (I.O.); (Y.K.); (H.U.)
| |
Collapse
|
5
|
Freire PP, Cury SS, Lopes LO, Fernandez GJ, Liu J, de Moraes LN, de Oliveira G, Oliveira JS, de Moraes D, Cabral-Marques O, Dal-Pai-Silva M, Hu X, Wang DZ, Carvalho RF. Decreased miR-497-5p Suppresses IL-6 Induced Atrophy in Muscle Cells. Cells 2021; 10:3527. [PMID: 34944037 PMCID: PMC8700610 DOI: 10.3390/cells10123527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Interleukin-6 (IL-6) is a pro-inflammatory cytokine associated with skeletal muscle wasting in cancer cachexia. The control of gene expression by microRNAs (miRNAs) in muscle wasting involves the regulation of thousands of target transcripts. However, the miRNA-target networks associated with IL6-induced muscle atrophy remain to be characterized. Here, we show that IL-6 promotes the atrophy of C2C12 myotubes and changes the expression of 20 miRNAs (5 up-regulated and 15 down-regulated). Gene Ontology analysis of predicted miRNAs targets revealed post-transcriptional regulation of genes involved in cell differentiation, apoptosis, migration, and catabolic processes. Next, we performed a meta-analysis of miRNA-published data that identified miR-497-5p, a down-regulated miRNAs induced by IL-6, also down-regulated in other muscle-wasting conditions. We used miR-497-5p mimics and inhibitors to explore the function of miR-497-5p in C2C12 myoblasts and myotubes. We found that miR-497-5p can regulate the expression of the cell cycle genes CcnD2 and CcnE1 without affecting the rate of myoblast cellular proliferation. Notably, miR-497-5p mimics induced myotube atrophy and reduced Insr expression. Treatment with miR-497-5p inhibitors did not change the diameter of the myotubes but increased the expression of its target genes Insr and Igf1r. These genes are known to regulate skeletal muscle regeneration and hypertrophy via insulin-like growth factor pathway and were up-regulated in cachectic muscle samples. Our miRNA-regulated network analysis revealed a potential role for miR-497-5p during IL6-induced muscle cell atrophy and suggests that miR-497-5p is likely involved in a compensatory mechanism of muscle atrophy in response to IL-6.
Collapse
Affiliation(s)
- Paula P. Freire
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Sarah S. Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Letícia O. Lopes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Geysson J. Fernandez
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
- Faculty of Medicine, University of Antioquia, UdeA, Medellín 050010, Colombia
| | - Jianming Liu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
| | - Leonardo Nazario de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Grasieli de Oliveira
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Jakeline S. Oliveira
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Diogo de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo 05508-000, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Robson F. Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| |
Collapse
|
6
|
Yang LT, Zhou L, Chen L, Liang SX, Huang JQ, Zhu XD. Establishment and Verification of a Prediction Model for Symptomatic Radiation Pneumonitis in Patients with Esophageal Cancer Receiving Radiotherapy. Med Sci Monit 2021; 27:e930515. [PMID: 33953150 PMCID: PMC8112075 DOI: 10.12659/msm.930515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This study aimed to determine the value of the significant index in predicting symptomatic radiation pneumonitis (RP) in esophageal cancer patients, establish a nomogram prediction model, and verify the model. MATERIAL AND METHODS The patients enrolled were divided into 2 groups: a model group and a validation group. According to the logistic regression analysis, the independent predictors for symptomatic RP were obtained, and the nomogram prediction model was established according to these independent predictors. The consistency index (C-index) and calibration curve were used to evaluate the accuracy of the model, and the prediction ability of the model was verified in the validation group. Recursive partitioning analysis (RPA) was used for the risk stratification analysis. RESULTS The ratio of change regarding the pre-albumin at the end of treatment (P=0.001), platelet-to-lymphocyte ratio during treatment (P=0.027), and neutrophil-to-lymphocyte ratio at the end of treatment (P=0.001) were the independent predictors for symptomatic RP. The C-index of the nomogram model was 0.811. According to the risk stratification of RPA, the whole group was divided into 3 groups: a low-risk group, a medium-risk group, and a high-risk group. The incidence of symptomatic RP was 0%, 16.9%, and 57.6%, respectively. The receiver operating characteristic curve also revealed that the nomogram model has good accuracy in the validation group. CONCLUSIONS The developed nomogram and corresponding risk classification system have superior prediction ability for symptomatic RP and can predict the occurrence of RP in the early stage.
Collapse
Affiliation(s)
- Liu-Ting Yang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Lei Zhou
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Long Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Shi-Xiong Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Jiang-Qiong Huang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Xiao-Dong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland).,Department of Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
7
|
Lithium Chloride Protects against Sepsis-Induced Skeletal Muscle Atrophy and Cancer Cachexia. Cells 2021; 10:cells10051017. [PMID: 33925786 PMCID: PMC8146089 DOI: 10.3390/cells10051017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022] Open
Abstract
Inflammation-mediated skeletal muscle wasting occurs in patients with sepsis and cancer cachexia. Both conditions severely affect patient morbidity and mortality. Lithium chloride has previously been shown to enhance myogenesis and prevent certain forms of muscular dystrophy. However, to our knowledge, the effect of lithium chloride treatment on sepsis-induced muscle atrophy and cancer cachexia has not yet been investigated. In this study, we aimed to examine the effects of lithium chloride using in vitro and in vivo models of cancer cachexia and sepsis. Lithium chloride prevented wasting in myotubes cultured with cancer cell-conditioned media, maintained the expression of the muscle fiber contractile protein, myosin heavy chain 2, and inhibited the upregulation of the E3 ubiquitin ligase, Atrogin-1. In addition, it inhibited the upregulation of inflammation-associated cytokines in macrophages treated with lipopolysaccharide. In the animal model of sepsis, lithium chloride treatment improved body weight, increased muscle mass, preserved the survival of larger fibers, and decreased the expression of muscle-wasting effector genes. In a model of cancer cachexia, lithium chloride increased muscle mass, enhanced muscle strength, and increased fiber cross-sectional area, with no significant effect on tumor mass. These results indicate that lithium chloride exerts therapeutic effects on inflammation-mediated skeletal muscle wasting, such as sepsis-induced muscle atrophy and cancer cachexia.
Collapse
|
8
|
Yu J, Choi S, Park A, Do J, Nam D, Kim Y, Noh J, Lee KY, Maeng CH, Park KS. Bone Marrow Homeostasis Is Impaired via JAK/STAT and Glucocorticoid Signaling in Cancer Cachexia Model. Cancers (Basel) 2021; 13:1059. [PMID: 33801569 PMCID: PMC7958949 DOI: 10.3390/cancers13051059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 01/08/2023] Open
Abstract
Cancer cachexia is a multifactorial systemic inflammation disease caused by complex interactions between the tumor and host tissues via soluble factors. However, whether cancer cachexia affects the bone marrow, in particular the hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs), remains unclear. Here, we investigated the bone marrow and bone in a cancer cachexia animal model generated by transplanting Lewis lung carcinoma cells. The number of bone marrow mononuclear cells (BM-MNCs) started to significantly decrease in the cancer cachectic animal model prior to the discernable loss of muscle and fat. This decrease in BM-MNCs was associated with myeloid skewing in the circulation and the expansion of hematopoietic progenitors in the bone marrow. Bone loss occurred in the cancer cachexia animal model and accompanied the decrease in the bone marrow MSCs that play important roles in both supporting HSCs and maintaining bone homeostasis. Glucocorticoid signaling mediated the decrease in bone marrow MSCs in the cancer cachectic environment. The cancer cachexia environment also skewed the differentiation of the bone marrow MSCs toward adipogenic fate via JAK/STAT as well as glucocorticoid signaling. Our results suggest that the bone loss induced in cancer cachexia is associated with the depletion and the impaired differentiation capacity of the bone marrow MSCs.
Collapse
Affiliation(s)
- Jinyeong Yu
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (J.Y.); (S.C.); (A.P.)
| | - Sanghyuk Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (J.Y.); (S.C.); (A.P.)
| | - Aran Park
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Korea; (J.Y.); (S.C.); (A.P.)
| | - Jungbeom Do
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.D.); (D.N.); (Y.K.); (J.N.)
| | - Donghyun Nam
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.D.); (D.N.); (Y.K.); (J.N.)
| | - Youngjae Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.D.); (D.N.); (Y.K.); (J.N.)
| | - Jinok Noh
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.D.); (D.N.); (Y.K.); (J.N.)
| | - Kil Yeon Lee
- Department of Surgery, College of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Chi Hoon Maeng
- Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Ki-Sook Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.D.); (D.N.); (Y.K.); (J.N.)
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
9
|
Armstrong VS, Fitzgerald LW, Bathe OF. Cancer-Associated Muscle Wasting-Candidate Mechanisms and Molecular Pathways. Int J Mol Sci 2020; 21:ijms21239268. [PMID: 33291708 PMCID: PMC7729509 DOI: 10.3390/ijms21239268] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive muscle loss is commonly observed in cancer patients and its association with poor prognosis has been well-established. Cancer-associated sarcopenia differs from age-related wasting in that it is not responsive to nutritional intervention and exercise. This is related to its unique pathogenesis, a result of diverse and interconnected mechanisms including inflammation, disordered metabolism, proteolysis and autophagy. There is a growing body of evidence that suggests that the tumor is the driver of muscle wasting by its elaboration of mediators that influence each of these pro-sarcopenic pathways. In this review, evidence for these tumor-derived factors and putative mechanisms for inducing muscle wasting will be reviewed. Potential targets for future research and therapeutic interventions will also be reviewed.
Collapse
Affiliation(s)
- Victoria S. Armstrong
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Liam W. Fitzgerald
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Oliver F. Bathe
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Departments of Surgery and Oncology, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Correspondence: ; Tel.: +1-403-521-3275
| |
Collapse
|
10
|
Abstract
Cachexia is common in end-stage renal disease (ESRD) patients, and it is an important risk factor for poor quality of life and increased mortality and morbidity. Chronic inflammation is an important cause of cachexia in ESRD patients. In the present review, we examine recent evidence suggesting that adipokines or adipocytokines such as leptin, adiponectin, resistin, tumor necrosis factor α, interleukin-6, and interleukin-1β may play important roles in uremic cachexia. We also review the physiology and the potential roles of gut hormones, including ghrelin, peptide YY, and cholecystokinin in ESRD. Understanding the molecular pathophysiology of these novel hormones in ESRD may lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Robert H. Mak
- Division of Pediatric Nephrology, Oregon Health and Science University, Portland, Oregon
- Division of Pediatric Nephrology, University of California at San Diego, La Jolla, California, U.S.A
| | - Wai Cheung
- Division of Pediatric Nephrology, Oregon Health and Science University, Portland, Oregon
- Division of Pediatric Nephrology, University of California at San Diego, La Jolla, California, U.S.A
| |
Collapse
|
11
|
Marceca GP, Londhe P, Calore F. Management of Cancer Cachexia: Attempting to Develop New Pharmacological Agents for New Effective Therapeutic Options. Front Oncol 2020; 10:298. [PMID: 32195193 PMCID: PMC7064558 DOI: 10.3389/fonc.2020.00298] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/20/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer cachexia (CC) is a multifactorial syndrome characterized by systemic inflammation, uncontrolled weight loss and dramatic metabolic alterations. This includes myofibrillar protein breakdown, increased lipolysis, insulin resistance, elevated energy expediture, and reduced food intake, hence impairing the patient's response to anti-cancer therapies and quality of life. While a decade ago the syndrome was considered incurable, over the most recent years much efforts have been put into the study of such disease, leading to the development of potential therapeutic strategies. Several important improvements have been reached in the management of CC from both the diagnostic-prognostic and the pharmacological viewpoint. However, given the heterogeneity of the disease, it is impossible to rely only on single variables to properly treat patients presenting this metabolic syndrome. Moreover, the cachexia symptoms are strictly dependent on the type of tumor, stage and the specific patient's response to cancer therapy. Thus, the attempt to translate experimentally effective therapies into the clinical practice results in a great challenge. For this reason, it is of crucial importance to further improve our understanding on the interplay of molecular mechanisms implicated in the onset and progression of CC, giving the opportunity to develop new effective, safe pharmacological treatments. In this review we outline the recent knowledge regarding cachexia mediators and pathways involved in skeletal muscle (SM) and adipose tissue (AT) loss, mainly from the experimental cachexia standpoint, then retracing the unimodal treatment options that have been developed to the present day.
Collapse
Affiliation(s)
- Gioacchino P Marceca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Priya Londhe
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
12
|
Pettersen K, Andersen S, van der Veen A, Nonstad U, Hatakeyama S, Lambert C, Lach-Trifilieff E, Moestue S, Kim J, Grønberg BH, Schilb A, Jacobi C, Bjørkøy G. Autocrine activin A signalling in ovarian cancer cells regulates secretion of interleukin 6, autophagy, and cachexia. J Cachexia Sarcopenia Muscle 2020; 11:195-207. [PMID: 31436048 PMCID: PMC7015233 DOI: 10.1002/jcsm.12489] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The majority of patients with advanced cancer develop cachexia, a weight loss syndrome that severely reduces quality of life and limits survival. Our understanding of the underlying mechanisms that cause the condition is limited, and there are currently no treatment options that can completely reverse cachexia. Several tumour-derived factors and inflammatory mediators have been suggested to contribute to weight loss in cachectic patients. However, inconsistencies between studies are recurrent. Activin A and interleukin 6 (IL-6) are among the best studied factors that seem to be important, and several studies support their individual role in cachexia development. METHODS We investigated the interplay between activin A and IL-6 in the cachexia-inducing TOV21G cell line, both in culture and in tumours in mice. We previously found that the human TOV21G cells secrete IL-6 that induces autophagy in reporter cells and cachexia in mice. Using this established cachexia cell model, we targeted autocrine activin A by genetic, chemical, and biological approaches. The secretion of IL-6 from the cancer cells was determined in both culture and tumour-bearing mice by a species-specific ELISA. Autophagy reporter cells were used to monitor the culture medium for autophagy-inducing activities, and muscle mass changes were evaluated in tumour-bearing mice. RESULTS We show that activin A acts in an autocrine manner to promote the synthesis and secretion of IL-6 from cancer cells. By inhibiting activin A signalling, the production of IL-6 from the cancer cells is reduced by 40-50% (up to 42% reduction on protein level, P = 0.0048, and 48% reduction on mRNA level, P = 0.0308). Significantly reduced IL-6 secretion (P < 0.05) from the cancer cells is consistently observed when using biological, chemical, and genetic approaches to interfere with the autocrine activin A loop. Inhibiting activin signalling also reduces the ability of the cancer cells to accelerate autophagy in non-cancerous cells (up to 43% reduced autophagy flux, P = 0.0006). Coherent to the in vitro data, the use of an anti-activin receptor 2 antibody in cachectic tumour-bearing mice reduces serum levels of cancer cell-derived IL-6 by 62% (from 417 to 159 pg/mL, P = 0.03), and, importantly, it reverses cachexia and counteracts loss of all measured muscle groups (P < 0.0005). CONCLUSIONS Our data support a functional link between activin A and IL-6 signalling pathways and indicate that interference with activin A-induced IL-6 secretion from the tumour has therapeutic potential for cancer-induced cachexia.
Collapse
Affiliation(s)
- Kristine Pettersen
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Sonja Andersen
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna van der Veen
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Unni Nonstad
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Shinji Hatakeyama
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, Basel, Switzerland
| | - Christian Lambert
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, Basel, Switzerland
| | - Estelle Lach-Trifilieff
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, Basel, Switzerland
| | - Siver Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Jana Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Bjørn Henning Grønberg
- Department of Cancer Research and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Clinic of Oncology, St. Olavs Hospital - Trondheim University Hospital, Trondheim, Norway
| | - Alain Schilb
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, Basel, Switzerland
| | - Carsten Jacobi
- Novartis Institutes for BioMedical Research Basel, Musculoskeletal Disease Area, Novartis Pharma AG, Basel, Switzerland
| | - Geir Bjørkøy
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
13
|
Wu C, Tang L, Ni X, Xu T, Fang Q, Xu L, Ma W, Yang X, Sun H. Salidroside Attenuates Denervation-Induced Skeletal Muscle Atrophy Through Negative Regulation of Pro-inflammatory Cytokine. Front Physiol 2019; 10:665. [PMID: 31293430 PMCID: PMC6604664 DOI: 10.3389/fphys.2019.00665] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle atrophy is associated with pro-inflammatory cytokines. Salidroside is a biologically active ingredient of Rhodiola rosea, which exhibits anti-inflammatory property. However, there is little known about the effect of salidroside on denervation-induced muscle atrophy. Therefore, the present study aimed to determine whether salidroside could protect against denervation-induced muscle atrophy and to clarify potential molecular mechanisms. Denervation caused progressive accumulation of inflammatory factors in skeletal muscle, especially interleukin 6 (IL6) and its receptor, and recombinant murine IL6 (rmIL6) local infusion could induce target muscle atrophy, suggesting that denervation induced inflammation in target muscles and the inflammation may trigger muscle wasting. Salidroside alleviated denervation-induced muscle atrophy and inhibited the production of IL6. Furthermore, the inhibition of phosphorylation of signal transducer and activator of transcription 3 (STAT3), and the decreased levels of suppressor of cytokine signaling (SOCS3), muscle RING finger protein-1 (MuRF1), atrophy F-box (atrogin-1), microtubule-associated protein light chain 3 beta (LC3B) and PTEN-induced putative kinase (PINK1) were observed in denervated muscles that were treated with salidroside. Finally, all of these responses to salidroside were replicated in neutralizing antibody against IL6. Taken together, these results suggest that salidroside alleviates denervation-induced inflammation response, thereby inhibits muscle proteolysis and muscle atrophy. Therefore, it was assumed that salidroside might be a potential therapeutic candidate to prevent muscle wasting.
Collapse
Affiliation(s)
- Changyue Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,School of Medicine, Nantong University, Nantong, China
| | - Longhai Tang
- Departments of Blood Component Preparation, Suzhou Blood Center, Suzhou, China
| | - Xuejun Ni
- Departments of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Tongtong Xu
- School of Medicine, Nantong University, Nantong, China
| | - Qingqing Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Wenjing Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
14
|
Fibroblasts as Modulators of Local and Systemic Cancer Metabolism. Cancers (Basel) 2019; 11:cancers11050619. [PMID: 31058816 PMCID: PMC6562905 DOI: 10.3390/cancers11050619] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 01/05/2023] Open
Abstract
Fibroblast activation is an accompanying feature of solid tumor progression, resembling a conserved host response to tissue damage. Cancer-associated fibroblasts (CAFs) comprise a heterogeneous and plastic population with increasingly appreciated roles in tumor growth, metastatic capacity, and response to therapy. Classical features of fibroblasts in a wound-healing response, including profound extracellular matrix production and cytokine release, are recapitulated in cancer. Emerging evidence suggests that fibroblastic cells in the microenvironments of solid tumors also critically modulate cellular metabolism in the neoplastic compartment through mechanisms including paracrine transfer of metabolites or non-cell-autonomous regulation of metabolic signaling pathways. These metabolic functions may represent common mechanisms by which fibroblasts stimulate growth of the regenerating epithelium during a wound-healing reaction, or may reflect unique co-evolution of cancer cells and surrounding stroma within the tumor microenvironment. Here we review the recent literature supporting an important role for CAFs in regulation of cancer cell metabolism, and relevant pathways that may serve as targets for therapeutic intervention.
Collapse
|
15
|
Cramer Z, Sadek J, Vazquez GG, Di Marco S, Pause A, Pelletier J, Gallouzi IE. eIF4A inhibition prevents the onset of cytokine-induced muscle wasting by blocking the STAT3 and iNOS pathways. Sci Rep 2018; 8:8414. [PMID: 29849089 PMCID: PMC5976662 DOI: 10.1038/s41598-018-26625-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/15/2018] [Indexed: 02/07/2023] Open
Abstract
Cachexia is a deadly muscle wasting syndrome that arises under conditions linked to chronic inflammation, such as cancer. Cytokines, including interferon γ (IFNγ), tumor necrosis factor α (TNFα) and interleukin-6 (IL-6), and their downstream effectors such as Signal Transducer and Activator of Transcription 3 (STAT3), have been shown to play a prominent role in muscle wasting. Previously, we demonstrated that Pateamine A (PatA), a compound that targets eukaryotic initiation factor 4A (eIF4A), could prevent muscle wasting by modulating the translation of the inducible Nitric Oxide Synthase (iNOS) mRNA. Here we show that hippuristanol, a compound that impedes eIF4A in a manner distinct from PatA, similarly inhibits the iNOS/NO pathway and cytokine-induced muscle wasting. Furthermore, we show that hippuristanol perturbs the activation of the STAT3 pathway and expression of STAT3-gene targets such as IL-6. The decreased activation of STAT3, which resulted from a decrease in STAT3 protein expression, was due to the inhibition of STAT3 translation as there were no changes in STAT3 mRNA levels. These effects are likely dependent on the inhibition of eIF4A activity since we observed similar results using PatA. Our results identify the inhibition of eIF4A-responsive transcripts, such as STAT3, as a viable approach to alleviate cachexia.
Collapse
Affiliation(s)
- Zvi Cramer
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Jason Sadek
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Gabriela Galicia Vazquez
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Sergio Di Marco
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Arnim Pause
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Jerry Pelletier
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Imed-Eddine Gallouzi
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada. .,Hamad Bin Khalifa University (HBKU), College of Science and Engineering, Life Sciences Division, Education City, Doha, PB, 5825, Qatar.
| |
Collapse
|
16
|
Bindels LB, Neyrinck AM, Loumaye A, Catry E, Walgrave H, Cherbuy C, Leclercq S, Van Hul M, Plovier H, Pachikian B, Bermúdez-Humarán LG, Langella P, Cani PD, Thissen JP, Delzenne NM. Increased gut permeability in cancer cachexia: mechanisms and clinical relevance. Oncotarget 2018; 9:18224-18238. [PMID: 29719601 PMCID: PMC5915068 DOI: 10.18632/oncotarget.24804] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/26/2018] [Indexed: 12/20/2022] Open
Abstract
Intestinal disorders often occur in cancer patients, in association with body weight loss, and this alteration is commonly attributed to the chemotherapy. Here, using a mouse model of cancer cachexia induced by ectopic transplantation of C26 cancer cells, we discovered a profound alteration in the gut functions (gut permeability, epithelial turnover, gut immunity, microbial dysbiosis) independently of any chemotherapy. These alterations occurred independently of anorexia and were driven by interleukin 6. Gut dysfunction was found to be resistant to treatments with an anti-inflammatory bacterium (Faecalibacterium prausnitzii) or with gut peptides involved in intestinal cell renewal (teduglutide, a glucagon-like peptide 2 analogue). The translational value of our findings was evaluated in 152 colorectal and lung cancer patients with or without cachexia. The serum level of the lipopolysaccharide-binding protein, often presented as a reflection of the bacterial antigen load, was not only increased in cachectic mice and cancer patients, but also strongly correlated with the serum IL-6 level and predictive of death and cachexia occurrence in these patients. Altogether, our data highlight profound alterations of the intestinal homeostasis in cancer cachexia occurring independently of any chemotherapy and food intake reduction, with potential relevance in humans. In addition, we point out the lipopolysaccharide-binding protein as a new biomarker of cancer cachexia related to gut dysbiosis.
Collapse
Affiliation(s)
- Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Audrey Loumaye
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Emilie Catry
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Hannah Walgrave
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Claire Cherbuy
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Sophie Leclercq
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium.,Pôle Clinique, Psychiatrie, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Hubert Plovier
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Barbara Pachikian
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Paul Thissen
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, Université Catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
17
|
Z-505 hydrochloride, an orally active ghrelin agonist, attenuates the progression of cancer cachexia via anabolic hormones in Colon 26 tumor-bearing mice. Eur J Pharmacol 2017; 811:30-37. [DOI: 10.1016/j.ejphar.2017.05.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 11/20/2022]
|
18
|
Saucedo-Cárdenas O, García-Garza R, Ramírez-Durón R, Muñoz-Maldonado GE, Villanueva-Olivo A, Montes-de-Oca-Luna R, Soto-Domínguez A. Invasion of TC-1 cells to skeletal muscle fibers protect them from peroxisomicine A1 (T-514) treatment in a murine model of cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8062-8071. [PMID: 31966659 PMCID: PMC6965213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 04/20/2017] [Indexed: 06/10/2023]
Abstract
T-514 or Peroxisomicine A1 (PA1) is a toxin isolated from plants of genus Karwinskia. In vitro studies described selective toxicity of PA1 on malignant cell lines. A toxic effect of PA1 on TC-1 cells in vivo was reported. The objective was to evaluate the effect of PA1 over invasion of TC-1 cells to muscle fibers in vivo. TC-1 cells were implanted in 36 mice divided in two groups (n: 18): treated with PA1 or with vehicle, a control group was included. At 10 days, nine mice of each group were euthanized. TC-1 implant site was analyzed by light and electron microscopy, a morphometric study was also performed. Remaining mice were used to evaluate tumor growth and survival time. Results show tumor cells between muscle fibers, with diminution in diameter, change in the staining pattern, loss of continuity of external lamina, and sarcoplasm with tumor cells. Statistically difference was observed between treated group vs control group. PA1 decreased tumor growth and increased the survival time in treated mice. The degree of resistant activity, aggressiveness, and invasiveness of TC-1 cells described in present work; should be taken into account in studies that evaluate effectiveness of therapies using this cancer model.
Collapse
Affiliation(s)
- Odila Saucedo-Cárdenas
- Departamento de Histología, Universidad Autónoma de Nuevo León. Av. Madero y E. Aguirre-PequeñoMonterrey, N.L., AP 1563, México
- Centro de Investigación Biomédica del Noreste (CIBIN), IMSSSan Luis y 2 de Abril. Col. Independencia. Monterrey, N.L., CP 64720, México
| | - Rubén García-Garza
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Coahuila, Unidad Torreón. Av. Gregorio A. García 198 SurTorreón, CP 27000, Coah, México
| | - Rosalba Ramírez-Durón
- Departamento de Química Analítica Facultad de Medicina, Universidad Autónoma de Nuevo León. Av. Madero y E. Aguirre-PequeñoMonterrey, N.L., AP 1563, México
| | - Gerardo E Muñoz-Maldonado
- Servicio de Cirugía General, Hospital Universitario “Dr. José Eleuterio González”, Universidad Autónoma de Nuevo LeónMonterrey N.L. A.P. 1583, México
| | - Arnulfo Villanueva-Olivo
- Departamento de Histología, Universidad Autónoma de Nuevo León. Av. Madero y E. Aguirre-PequeñoMonterrey, N.L., AP 1563, México
| | - Roberto Montes-de-Oca-Luna
- Departamento de Histología, Universidad Autónoma de Nuevo León. Av. Madero y E. Aguirre-PequeñoMonterrey, N.L., AP 1563, México
| | - Adolfo Soto-Domínguez
- Departamento de Histología, Universidad Autónoma de Nuevo León. Av. Madero y E. Aguirre-PequeñoMonterrey, N.L., AP 1563, México
| |
Collapse
|
19
|
Pettersen K, Andersen S, Degen S, Tadini V, Grosjean J, Hatakeyama S, Tesfahun AN, Moestue S, Kim J, Nonstad U, Romundstad PR, Skorpen F, Sørhaug S, Amundsen T, Grønberg BH, Strasser F, Stephens N, Hoem D, Molven A, Kaasa S, Fearon K, Jacobi C, Bjørkøy G. Cancer cachexia associates with a systemic autophagy-inducing activity mimicked by cancer cell-derived IL-6 trans-signaling. Sci Rep 2017; 7:2046. [PMID: 28515477 PMCID: PMC5435723 DOI: 10.1038/s41598-017-02088-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/05/2017] [Indexed: 12/20/2022] Open
Abstract
The majority of cancer patients with advanced disease experience weight loss, including loss of lean body mass. Severe weight loss is characteristic for cancer cachexia, a condition that significantly impairs functional status and survival. The underlying causes of cachexia are incompletely understood, and currently no therapeutic approach can completely reverse the condition. Autophagy coordinates lysosomal destruction of cytosolic constituents and is systemically induced by starvation. We hypothesized that starvation-mimicking signaling compounds secreted from tumor cells may cause a systemic acceleration of autophagy during cachexia. We found that IL-6 secreted by tumor cells accelerates autophagy in myotubes when complexed with soluble IL-6 receptor (trans-signaling). In lung cancer patients, were cachexia is prevalent, there was a significant correlation between elevated IL-6 expression in the tumor and poor prognosis of the patients. We found evidence for an autophagy-inducing bioactivity in serum from cancer patients and that this is clearly associated with weight loss. Importantly, the autophagy-inducing bioactivity was reduced by interference with IL-6 trans-signaling. Together, our findings suggest that IL-6 trans-signaling may be targeted in cancer cachexia.
Collapse
Affiliation(s)
- Kristine Pettersen
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Sonja Andersen
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Simone Degen
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Valentina Tadini
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Joël Grosjean
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Shinji Hatakeyama
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland
| | - Almaz N Tesfahun
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Siver Moestue
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Jana Kim
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Unni Nonstad
- Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Pål R Romundstad
- Department of Public Health and General Practice, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway
| | - Frank Skorpen
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway
| | - Sveinung Sørhaug
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Thoracic Medicine, St.Olavs Hospital - Trondheim University Hospital, 7006, Trondheim, Norway
| | - Tore Amundsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway.,Department of Thoracic Medicine, St.Olavs Hospital - Trondheim University Hospital, 7006, Trondheim, Norway
| | - Bjørn H Grønberg
- The Cancer Clinic, St.Olavs Hospital - Trondheim University Hospital, 7030, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Florian Strasser
- Oncological Palliative Medicine, Division ofClinic Oncology/Hematology, Department of Internal Medicine and Palliative Care Center, Cantonal Hospital, St. Gallen, Switzerland
| | - Nathan Stephens
- Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, The University of Edinburgh, Royal Infirmary, N-5021, Edinburgh, UK
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, N-5020, Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, N-5021, Bergen, Norway
| | - Stein Kaasa
- European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.,The Cancer Clinic, St.Olavs Hospital - Trondheim University Hospital, 7030, Trondheim, Norway
| | - Kenneth Fearon
- European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.,Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, The University of Edinburgh, Royal Infirmary, N-5021, Edinburgh, UK
| | - Carsten Jacobi
- Musculoskeletal Disease Area, Novartis Institutes for BioMedical Research Basel, Novartis Pharma AG, 4056, Basel, Switzerland.
| | - Geir Bjørkøy
- Department of Medical Laboratory Technology, Faculty of Natural Sciences, NTNU - Norwegian University of Science and Technology, 7491, Trondheim, Norway. .,Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7030, Trondheim, Norway.
| |
Collapse
|
20
|
Shyh-Chang N. Metabolic Changes During Cancer Cachexia Pathogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:233-249. [PMID: 29282687 DOI: 10.1007/978-981-10-6020-5_11] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Wasting of adipose tissue and skeletal muscle is a hallmark of metastatic cancer and a major cause of death. Like patients with cachexia caused by other chronic infections or inflammatory diseases, the cancer subject manifests both malnutrition and metabolic stress. Both carbohydrate utilization and amino acid incorporation are decreased in the muscles of cancer cachexia patients. Cancer cells affect host metabolism in two ways: (a) their own metabolism of nutrients into other metabolites and (b) circulating factors they secrete or induce the host to secrete. Accelerated glycolysis and lactate production, i.e., the Warburg effect and the resultant increase in Cori cycle activity, are the most widely discussed metabolic effects. Meanwhile, although a large number of pro-cachexia circulating factors have been found, such as TNFa, IL-6, myostatin, and PTHrp, none have been shown to be a dominant factor that can be targeted singly to treat cancer cachexia in humans. It is possible that given the complex multifactorial nature of the cachexia secretome, and the personalized differences between cancer patients, targeting any single circulating factor would always be insufficient to treat cachexia for all patients. Here we review the metabolic changes that occur in response to tumor growth and tumor-secreted factors during cachexia.
Collapse
Affiliation(s)
- Ng Shyh-Chang
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore.
| |
Collapse
|
21
|
|
22
|
Chen JL, Walton KL, Qian H, Colgan TD, Hagg A, Watt MJ, Harrison CA, Gregorevic P. Differential Effects of IL6 and Activin A in the Development of Cancer-Associated Cachexia. Cancer Res 2016; 76:5372-82. [PMID: 27328730 DOI: 10.1158/0008-5472.can-15-3152] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/13/2016] [Indexed: 11/16/2022]
Abstract
Cachexia is a life-threatening wasting syndrome lacking effective treatment, which arises in many cancer patients. Although ostensibly induced by multiple tumor-produced cytokines (tumorkines), their functional contribution to initiation and progression of this syndrome has proven difficult to determine. In this study, we used adeno-associated viral vectors to elevate circulating levels of the tumorkines IL6 and/or activin A in animals in the absence of tumors as a tactic to evaluate hypothesized roles in cachexia development. Mice with elevated levels of IL6 exhibited 8.1% weight loss after 9 weeks, whereas mice with elevated levels of activin A lost 11% of their body weight. Co-elevation of both tumorkines to levels approximating those observed in cancer cachexia models induced a more rapid and profound body weight loss of 15.4%. Analysis of body composition revealed that activin A primarily triggered loss of lean mass, whereas IL6 was a major mediator of fat loss. Histologic and transcriptional analysis of affected organs/tissues (skeletal muscle, fat, and liver) identified interactions between the activin A and IL6 signaling pathways. For example, IL6 exacerbated the detrimental effects of activin A in skeletal muscle, whereas activin A curbed the IL6-induced acute-phase response in liver. This study presents a useful model to deconstruct cachexia, opening a pathway to determining which tumorkines are best targeted to slow/reverse this devastating condition in cancer patients. Cancer Res; 76(18); 5372-82. ©2016 AACR.
Collapse
Affiliation(s)
- Justin L Chen
- Hudson Institute of Medical Research, Clayton, Australia. Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Kelly L Walton
- Hudson Institute of Medical Research, Clayton, Australia
| | - Hongwei Qian
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Timothy D Colgan
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia. Department of Physiology, The University of Melbourne, Melbourne, Australia
| | - Adam Hagg
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Matthew J Watt
- The Obesity and Metabolism Program of the Biomedicine Discovery Institute, Monash University, Clayton, Australia. Department of Physiology, Monash University, Clayton, Australia
| | - Craig A Harrison
- Hudson Institute of Medical Research, Clayton, Australia. Department of Physiology, Monash University, Clayton, Australia. Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Paul Gregorevic
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia. Department of Physiology, The University of Melbourne, Melbourne, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia. Department of Neurology, The University of Washington School of Medicine, Seattle, Washington.
| |
Collapse
|
23
|
Assi M, Derbré F, Lefeuvre-Orfila L, Rébillard A. Antioxidant supplementation accelerates cachexia development by promoting tumor growth in C26 tumor-bearing mice. Free Radic Biol Med 2016; 91:204-14. [PMID: 26708754 DOI: 10.1016/j.freeradbiomed.2015.12.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 12/14/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
More than 50% of patients with advanced stages of colon cancer suffer from progressive loss of skeletal muscle, called cachexia, resulting in reduced quality of life and shortened survival. It is becoming evident that reactive oxygen species (ROS) regulate pathways controlling skeletal muscle atrophy. Herein we tested the hypothesis that antioxidant supplementation could prevent skeletal muscle atrophy in a model of cachectic Colon 26 (C26) tumor-bearing mice. Seven-week-old BALB/c mice were subcutaneously inoculated with colon 26 (C26) cancer cells or PBS. Then C26-mice were daily gavaged during 22 days either with PBS (vehicle) or an antioxidant cocktail whose composition is close to that of commercial dietary antioxidant supplements (rich in catechins, quercetin and vitamin C). We found that antioxidants enhanced weight loss and caused premature death of mice. Antioxidants supplementation failed to prevent (i) the increase in plasma TNF-α levels and systemic oxidative damage, (ii) skeletal muscle atrophy and (iii) activation of the ubiquitin-proteasome system (MuRF-1, MAFbx and polyubiquitinated proteins). Accordingly, immunohistological staining for Ki-67 and the expression of cell cycle inhibitors demonstrated that tumor of supplemented mice developed faster with a concomitant decrease in oxidative damage. Previous studies have shown that the use of catechins and quercetin separately can improve the musculoskeletal function in cachectic animals. However, our results indicate that the combination of these antioxidants reduced survival and enhanced cachexia in C26-mice.
Collapse
Affiliation(s)
- Mohamad Assi
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, University of Rennes 2-ENS Rennes, Bruz, France
| | - Frédéric Derbré
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, University of Rennes 2-ENS Rennes, Bruz, France
| | - Luz Lefeuvre-Orfila
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, University of Rennes 2-ENS Rennes, Bruz, France
| | - Amélie Rébillard
- EA1274 Laboratory "Movement, Sport and Health Sciences" M2S, University of Rennes 2-ENS Rennes, Bruz, France.
| |
Collapse
|
24
|
Role of Inflammation in Muscle Homeostasis and Myogenesis. Mediators Inflamm 2015; 2015:805172. [PMID: 26508819 PMCID: PMC4609834 DOI: 10.1155/2015/805172] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/05/2015] [Accepted: 03/10/2015] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle mass is subject to rapid changes according to growth stimuli inducing both hypertrophy, through increased protein synthesis, and hyperplasia, activating the myogenic program. Muscle wasting, characteristic of several pathological states associated with local or systemic inflammation, has been for long considered to rely on the alteration of myofiber intracellular pathways regulated by both hormones and cytokines, eventually leading to impaired anabolism and increased protein breakdown. However, there are increasing evidences that even alterations of the myogenic/regenerative program play a role in the onset of muscle wasting, even though the precise mechanisms involved are far from being fully elucidated. The comprehension of the links potentially occurring between impaired myogenesis and increased catabolism would allow the definition of effective strategies aimed at counteracting muscle wasting. The first part of this review gives an overview of skeletal muscle intracellular pathways determining fiber size, while the second part considers the cells and the regulatory pathways involved in the myogenic program. In both parts are discussed the evidences supporting the role of inflammation in impairing muscle homeostasis and myogenesis, potentially determining muscle atrophy.
Collapse
|
25
|
Inflammation based regulation of cancer cachexia. BIOMED RESEARCH INTERNATIONAL 2014; 2014:168407. [PMID: 24877061 PMCID: PMC4022077 DOI: 10.1155/2014/168407] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/10/2014] [Indexed: 02/08/2023]
Abstract
Cancer cachexia, consisting of significant skeletal muscle wasting independent of nutritional intake, is a major concern for patients with solid tumors that affects surgical, therapeutic, and quality of life outcomes. This review summarizes the clinical implications, background of inflammatory cytokines, and the origin and sources of procachectic factors including TNF-α, IL-6, IL-1, INF-γ, and PIF. Molecular mechanisms and pathways are described to elucidate the link between the immune response caused by the presence of the tumor and the final result of skeletal muscle wasting.
Collapse
|
26
|
Tsoli M, Schweiger M, Vanniasinghe AS, Painter A, Zechner R, Clarke S, Robertson G. Depletion of white adipose tissue in cancer cachexia syndrome is associated with inflammatory signaling and disrupted circadian regulation. PLoS One 2014; 9:e92966. [PMID: 24667661 PMCID: PMC3965507 DOI: 10.1371/journal.pone.0092966] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/27/2014] [Indexed: 11/19/2022] Open
Abstract
Involuntary weight loss in patients with cancer is the hallmark of cancer cachexia. The etiology of cachexia is multifactorial involving loss of skeletal muscle and adipose tissue associated with high systemic levels of acute phase proteins and inflammatory cytokines. While muscle wasting overtly impacts on cancer patient quality of life, loss of lipid depots represents a sustained energy imbalance. In this study fat depletion was examined in Colon-26 model of cancer cachexia, which is a widely used rodent model of this syndrome. We investigated diurnal expression of circadian rhythm regulators as well as key mediators of energy metabolism and cytokine signaling. Mice bearing the C26 tumour exhibited reduced adipose mass, elevated adipose tissue lipolysis and a 5-fold increase in plasma levels of free fatty acids. These changes were associated with activated IL-6 signaling in WAT through a 3-fold increase in phosphorylated STAT3 and high SOCS3 gene expression levels. In addition perturbations in circadian regulation of lipid metabolism were also observed. Lipid catabolism did not appear to be influenced by the classical PKA pathway activating the lipase HSL. ATGL protein levels were elevated 2-fold in cachectic mice while 4-fold increase phosphorylated ACC and a 2-fold decrease in phosphorylated 4EBP1 was observed indicating that lipid metabolism is modulated by the ATGL & AMPK/mTOR pathways. This study provides evidence for activation of cytokine signaling and concomitant alterations in circadian rhythm and regulators of lipid metabolism in WAT of cachectic animals.
Collapse
Affiliation(s)
- Maria Tsoli
- Experimental Therapeutics, Children’s Cancer Institute for Medical Research, Randwick, NSW, Australia
- Cancer Pharmacology Unit, Anzac Research Institute, Concord Repatriation General Hospital, Concord West, NSW, Australia
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anne S. Vanniasinghe
- Experimental Therapeutics, Children’s Cancer Institute for Medical Research, Randwick, NSW, Australia
- Cancer Pharmacology Unit, Anzac Research Institute, Concord Repatriation General Hospital, Concord West, NSW, Australia
| | - Arran Painter
- Cancer Pharmacology Unit, Anzac Research Institute, Concord Repatriation General Hospital, Concord West, NSW, Australia
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Stephen Clarke
- Northern Clinical School, University of Sydney, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Graham Robertson
- Cancer Pharmacology Unit, Anzac Research Institute, Concord Repatriation General Hospital, Concord West, NSW, Australia
- Tumorkine Research, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- * E-mail:
| |
Collapse
|
27
|
Johns N, Stephens NA, Fearon KCH. Muscle wasting in cancer. Int J Biochem Cell Biol 2013; 45:2215-29. [PMID: 23770121 DOI: 10.1016/j.biocel.2013.05.032] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/23/2013] [Accepted: 05/27/2013] [Indexed: 01/06/2023]
Abstract
Skeletal muscle loss appears to be the most significant clinical event in cancer cachexia and is associated with a poor outcome. With regard to such muscle loss, despite extensive study in a range of models, there is ongoing debate as to whether a reduction in protein synthesis, an increase in degradation or a combination of both is the more relevant. Each model differs in terms of key mediators and the pathways activated in skeletal muscle. Certain models do suggest that decreased synthesis accompanied by enhanced protein degradation via the ubiquitin proteasome pathway (UPP) is important. Murine models tend to involve rapid development of cachexia and may represent more acute muscle atrophy rather than the chronic wasting observed in humans. There is a paucity of human data both at a basic descriptive level and at a molecular/mechanism level. Progress in treating the human form of cancer cachexia can only move forwards through carefully designed large randomised controlled clinical trials of specific therapies with validated biomarkers of relevance to underlying mechanisms. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- N Johns
- Department of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | | | | |
Collapse
|
28
|
|
29
|
Oraldi M, Maggiora M, Paiuzzi E, Canuto RA, Muzio G. CLA Reduces Inflammatory Mediators from A427 Human Lung Cancer Cells and A427 Conditioned Medium Promotes Differentiation of C2C12 Murine Muscle Cells. Lipids 2012; 48:29-38. [DOI: 10.1007/s11745-012-3734-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 10/09/2012] [Indexed: 11/28/2022]
|
30
|
Abstract
Anorexia and cachexia frequently complicate the late stages of malignancy and can be a prominent feature of early disease. The resulting weight loss significantly affects the morbidity and mortality of the cancer patient. A fundamental understanding of nutrition and the pathophysiology of cancer cachexia will aid in diligent treatment decisions to achieve optimal results. The pathophysiology of cancer cachexia is discussed, together with methods of nutritional assessment, nutritional requirements, and postoperative nutritional support. The advantages and disadvantages of the various modes of parenteral and enteral feeding are presented, together with information about enteral feeding in the home.
Collapse
|
31
|
Abstract
Cancer cachexia is characterized by a significant reduction in body weight resulting predominantly from loss of adipose tissue and skeletal muscle. Cachexia causes reduced cancer treatment tolerance and reduced quality and length of life, and remains an unmet medical need. Therapeutic progress has been impeded, in part, by the marked heterogeneity of mediators, signaling, and metabolic pathways both within and between model systems and the clinical syndrome. Recent progress in understanding conserved, molecular mechanisms of skeletal muscle atrophy/hypertrophy has provided a downstream platform for circumventing the variations and redundancy in upstream mediators and may ultimately translate into new targeted therapies.
Collapse
|
32
|
Bonetto A, Aydogdu T, Jin X, Zhang Z, Zhan R, Puzis L, Koniaris LG, Zimmers TA. JAK/STAT3 pathway inhibition blocks skeletal muscle wasting downstream of IL-6 and in experimental cancer cachexia. Am J Physiol Endocrinol Metab 2012; 303:E410-21. [PMID: 22669242 PMCID: PMC3423125 DOI: 10.1152/ajpendo.00039.2012] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cachexia, the metabolic dysregulation leading to sustained loss of muscle and adipose tissue, is a devastating complication of cancer and other chronic diseases. Interleukin-6 and related cytokines are associated with muscle wasting in clinical and experimental cachexia, although the mechanisms by which they might induce muscle wasting are unknown. One pathway activated strongly by IL-6 family ligands is the JAK/STAT3 pathway, the function of which has not been evaluated in regulation of skeletal muscle mass. Recently, we showed that skeletal muscle STAT3 phosphorylation, nuclear localization, and target gene expression are activated in C26 cancer cachexia, a model with high IL-6 family ligands. Here, we report that STAT3 activation is a common feature of muscle wasting, activated in muscle by IL-6 in vivo and in vitro and by different types of cancer and sterile sepsis. Moreover, STAT3 activation proved both necessary and sufficient for muscle wasting. In C(2)C(12) myotubes and in mouse muscle, mutant constitutively activated STAT3-induced muscle fiber atrophy and exacerbated wasting in cachexia. Conversely, inhibiting STAT3 pharmacologically with JAK or STAT3 inhibitors or genetically with dominant negative STAT3 and short hairpin STAT3 reduced muscle atrophy downstream of IL-6 or cancer. These results indicate that STAT3 is a primary mediator of muscle wasting in cancer cachexia and other conditions of high IL-6 family signaling. Thus STAT3 could represent a novel therapeutic target for the preservation of skeletal muscle in cachexia.
Collapse
MESH Headings
- Animals
- CHO Cells
- Cachexia/etiology
- Cachexia/genetics
- Cachexia/pathology
- Cachexia/prevention & control
- Cells, Cultured
- Cricetinae
- Cricetulus
- Disease Models, Animal
- Female
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Interleukin-6/physiology
- Janus Kinases/antagonists & inhibitors
- Janus Kinases/genetics
- Janus Kinases/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Mutant Proteins/administration & dosage
- Mutant Proteins/genetics
- Neoplasms/complications
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/metabolism
- Nitriles
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/pharmacology
- Pyrazoles/administration & dosage
- Pyrazoles/pharmacology
- Pyrimidines
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/pharmacology
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Wasting Syndrome/genetics
- Wasting Syndrome/metabolism
- Wasting Syndrome/pathology
- Wasting Syndrome/prevention & control
Collapse
Affiliation(s)
- Andrea Bonetto
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Yamaki T, Wu CL, Gustin M, Lim J, Jackman RW, Kandarian SC. Rel A/p65 is required for cytokine-induced myotube atrophy. Am J Physiol Cell Physiol 2012; 303:C135-42. [PMID: 22592403 DOI: 10.1152/ajpcell.00111.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle atrophy can be triggered by systemic illnesses that are associated with elevated proinflammatory/catabolic cytokines, which, in turn, are thought to contribute to muscle wasting. In this study, we found that the prototypical NF-κB transcription factor, Rel A (p65), is required for NF-κB activation in C2C12 and L6 myotubes due to treatment with exogenous TNF-α, IL-1α, IL-1β, TNF-related weak inducer of apoptosis, but not IL-6. All five cytokines induced atrophy in C2C12 myotubes, and inhibition of p65 reversed atrophy due to TNF-α, IL-1α, IL-1β, TNF-related weak inducer of apoptosis, but not IL-6 treatment. p65 was also required for TNF-α-induced increase in atrophy and inflammatory gene expression. TNF-α- and IL-1β-treated myotubes increased IL-6 protein expression, but use of an IL-6 blocking antibody showed that the IL-6 production did not contribute to atrophy. These data show that p65 is a required transcription factor mediating the catabolic effects of four different cytokines in cultured myotubes, but IL-6 works by a different mechanism.
Collapse
Affiliation(s)
- Takuo Yamaki
- Department of Health Sciences, Boston University, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
34
|
Suramin ameliorates collagen induced arthritis. Int Immunopharmacol 2012; 12:288-93. [DOI: 10.1016/j.intimp.2011.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 11/28/2011] [Accepted: 12/01/2011] [Indexed: 01/04/2023]
|
35
|
Robert F, Mills JR, Agenor A, Wang D, DiMarco S, Cencic R, Tremblay ML, Gallouzi IE, Hekimi S, Wing SS, Pelletier J. Targeting protein synthesis in a Myc/mTOR-driven model of anorexia-cachexia syndrome delays its onset and prolongs survival. Cancer Res 2011; 72:747-56. [PMID: 22158946 DOI: 10.1158/0008-5472.can-11-2739] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Anorexia-cachexia syndrome (ACS) is a major determinant of cancer-related death that causes progressive body weight loss due to depletion of skeletal muscle mass and body fat. Here, we report the development of a novel preclinical murine model of ACS in which lymphomas harbor elevated Myc and activated mTOR signaling. The ACS phenotype in this model correlated with deregulated expression of a number of cytokines, including elevated levels of interleukin-10 which was under the direct translational control of mTOR. Notably, pharmacologic intervention to impair protein synthesis restored cytokine production to near-normal levels, delayed ACS progression, and extended host survival. Together, our findings suggest a new paradigm to treat ACS by strategies which target protein synthesis to block the production of procachexic factors.
Collapse
Affiliation(s)
- Francis Robert
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen MF, Lu MS, Chen PT, Chen WC, Lin PY, Lee KD. Role of interleukin 1 beta in esophageal squamous cell carcinoma. J Mol Med (Berl) 2011; 90:89-100. [PMID: 21912958 DOI: 10.1007/s00109-011-0809-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 07/18/2011] [Accepted: 08/02/2011] [Indexed: 11/26/2022]
Abstract
Interleukin (IL)-1 beta has been reported to be a marker of shorter survival in gastric and colorectal adenocarcinoma. In the present study, we examined the potential role and prognostic value of IL-1 beta in esophageal squamous cell carcinoma (SCC). Human esophageal SCC cell line, CE81T, was selected for cellular and animal experiments, in which biological changes after experimental manipulation of IL-1 beta signaling were explored, including tumor growth, invasion capacity, and the sensitivity to treatment. Moreover, 147 esophageal SCC samples were analyzed using immunohistochemical staining to correlate the expression of IL-1 beta with clinical outcome. Our data revealed that IL-1 beta was significantly overexpressed both at mRNA and protein levels in cancer specimens compared to nonmalignant tissues. When IL-1 beta signaling was blocked, tumor growth, invasion ability, and treatment resistance were attenuated. Activation of NF-kappa B, increase of E2-EPF ubiquitin carrier protein and subsequent epithelial-mesenchymal transition might be the underlying mechanisms of the more aggressive tumor growth in IL-1 beta-positive esophageal cancer. The immunochemistry findings indicate that positivity staining of IL-1 beta correlated significantly with higher clinical stage, lower response rate to concurrent chemoradiotherapy (CCRT), and higher recurrence rate after curative treatment. Moreover, IL-1 beta was a significant predictor of survival in patients undergoing surgical intervention or definite CCRT. In conclusion, IL-1 beta is significantly linked to poor prognosis for patients with esophageal cancer and may be a promising molecular target for therapeutic intervention for esophageal SCC.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chia-Yi, Chia-Yi, Taiwan
| | | | | | | | | | | |
Collapse
|
37
|
Bonetto A, Aydogdu T, Kunzevitzky N, Guttridge DC, Khuri S, Koniaris LG, Zimmers TA. STAT3 activation in skeletal muscle links muscle wasting and the acute phase response in cancer cachexia. PLoS One 2011; 6:e22538. [PMID: 21799891 PMCID: PMC3140523 DOI: 10.1371/journal.pone.0022538] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Accepted: 06/29/2011] [Indexed: 01/05/2023] Open
Abstract
Background Cachexia, or weight loss despite adequate nutrition, significantly impairs quality of life and response to therapy in cancer patients. In cancer patients, skeletal muscle wasting, weight loss and mortality are all positively associated with increased serum cytokines, particularly Interleukin-6 (IL-6), and the presence of the acute phase response. Acute phase proteins, including fibrinogen and serum amyloid A (SAA) are synthesized by hepatocytes in response to IL-6 as part of the innate immune response. To gain insight into the relationships among these observations, we studied mice with moderate and severe Colon-26 (C26)-carcinoma cachexia. Methodology/Principal Findings Moderate and severe C26 cachexia was associated with high serum IL-6 and IL-6 family cytokines and highly similar patterns of skeletal muscle gene expression. The top canonical pathways up-regulated in both were the complement/coagulation cascade, proteasome, MAPK signaling, and the IL-6 and STAT3 pathways. Cachexia was associated with increased muscle pY705-STAT3 and increased STAT3 localization in myonuclei. STAT3 target genes, including SOCS3 mRNA and acute phase response proteins, were highly induced in cachectic muscle. IL-6 treatment and STAT3 activation both also induced fibrinogen in cultured C2C12 myotubes. Quantitation of muscle versus liver fibrinogen and SAA protein levels indicates that muscle contributes a large fraction of serum acute phase proteins in cancer. Conclusions/Significance These results suggest that the STAT3 transcriptome is a major mechanism for wasting in cancer. Through IL-6/STAT3 activation, skeletal muscle is induced to synthesize acute phase proteins, thus establishing a molecular link between the observations of high IL-6, increased acute phase response proteins and muscle wasting in cancer. These results suggest a mechanism by which STAT3 might causally influence muscle wasting by altering the profile of genes expressed and translated in muscle such that amino acids liberated by increased proteolysis in cachexia are synthesized into acute phase proteins and exported into the blood.
Collapse
Affiliation(s)
- Andrea Bonetto
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Tufan Aydogdu
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Noelia Kunzevitzky
- Center for Computational Science, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Denis C. Guttridge
- Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University School of Medicine, Columbus, Ohio, United States of America
| | - Sawsan Khuri
- Center for Computational Science, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Leonidas G. Koniaris
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Teresa A. Zimmers
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Surgical Oncology, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Division of Burns, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
38
|
Laird BJA, Scott AC, Colvin LA, McKeon AL, Murray GD, Fearon KCH, Fallon MT. Cancer pain and its relationship to systemic inflammation: an exploratory study. Pain 2011; 152:460-463. [PMID: 21159432 DOI: 10.1016/j.pain.2010.10.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 09/20/2010] [Accepted: 10/25/2010] [Indexed: 01/09/2023]
Abstract
Pain is the commonest symptom in cancer patients, whereas inflammation is implicated in cancer development and progression. The relationship between pain and inflammation in cancer is therefore of interest; however, it is challenging to examine because multiple factors may affect these variables. This study assessed the relationship between cancer pain and systemic inflammation using a retrospective analysis of 2 clinical trial datasets of patients with cancer cachexia. Included patients had gastrointestinal, lung, or pancreatic cancer. Pain was assessed using the pain subscale of the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire C-30. Inflammation was assessed using C-reactive protein (CRP). A regression analysis between pain and logarithmically transformed CRP was run, and Pearson correlation coefficients were calculated. A total of 718 patients entered the trials, of whom 449 had CRP measured. Both trial populations were well matched. Pain positively correlated with CRP. The Pearson correlation coefficients were 0.126 and 0.163 for trials 1 and 2, respectively. This correlation was statistically significant at the P<.05 level. These findings support that pain is related to systemic inflammation in a cohort of cancer patients. Many factors can affect pain and inflammation in cancer, demonstrating that any relationship that exists between pain and inflammation is of interest. This is in keeping with work showing this relationship in nonmalignant pain. Studies targeting inflammation and assessing its effect on pain in cancer would be an important step in the research agenda.
Collapse
Affiliation(s)
- Barry J A Laird
- University of Edinburgh, UK Western General Hospital, Edinburgh, UK
| | | | | | | | | | | | | |
Collapse
|
39
|
Lopez ES, Rizzo MM, Croxatto JO, Mazzolini G, Gallo JE. Suramab, a novel antiangiogenic agent, reduces tumor growth and corneal neovascularization. Cancer Chemother Pharmacol 2010; 67:723-8. [PMID: 20857116 DOI: 10.1007/s00280-010-1457-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 09/01/2010] [Indexed: 12/20/2022]
Abstract
PURPOSE Oncological and ophthalmological diseases are increasingly treated with antiangiogenic agents. These agents have different intensities and duration of effects that should be considered to choose the most suitable therapy. Our purpose was to evaluate the synergistic effect of two drugs, jointly administered as a pharmaceutical compound, in two animal models. METHODS Corneal neovascularization was induced in three groups of nine white New Zealand rabbits, applying a filter paper disk soaked in 1 M NaOH on the central cornea (Ormerod et al., Invest Ophthalmol Vis Sci 30:2148-2153, 1989). Group one was treated immediately after injury with intravenous Suramab, compound of Bevacizumab + Suramin, and group two with intravenous Bevacizumab. A third group of non-treated rabbits was included as control group. Digital photographs were taken at days 9, 15, 21, and 35. Neovessel index (NVI) was calculated using the Image J Program. Neovessels formation was quantified and given a score from 0 to 4 to each quadrant according to the centripetal growth of the longest vessel. Colorectal animal model: 6- to 8-week-old male BALB/c mice were inoculated with cancer cells. Seven days after tumor inoculation, four groups of BALB/c mice were treated with intravenous Bevacizumab (n = 9); intravenous Suramin (n = 10); intravenous Suramab (n = 10); and intravenous saline solution (n = 4). Tumor growth was assessed twice weekly by caliper measurement. RESULTS The NVI was remarkably inferior in the group of rabbits treated with intravenous Suramab compared with controls after 35 days of follow-up. A greater inhibitory effect was obtained with Suramab compared to that obtained with Bevacizumab. Suramab significantly reduced tumor volume and prolonged survival of mice compared to controls. CONCLUSIONS Suramab strongly reduced neovascularization in a rabbit model of corneal angiogenesis and induced a potent antitumoral effect in mice.
Collapse
Affiliation(s)
- Emiliano S Lopez
- Departments of Ophthalmology and Internal Medicine, Austral University Medical School and Austral University Hospital, Pilar, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
40
|
Penna F, Minero VG, Costamagna D, Bonelli G, Baccino FM, Costelli P. Anti-cytokine strategies for the treatment of cancer-related anorexia and cachexia. Expert Opin Biol Ther 2010; 10:1241-50. [DOI: 10.1517/14712598.2010.503773] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Burckart K, Beca S, Urban RJ, Sheffield-Moore M. Pathogenesis of muscle wasting in cancer cachexia: targeted anabolic and anticatabolic therapies. Curr Opin Clin Nutr Metab Care 2010; 13:410-6. [PMID: 20473155 PMCID: PMC4618842 DOI: 10.1097/mco.0b013e328339fdd2] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Cancer-related muscle loss, or cachexia, is the cause of death for approximately 2 million people worldwide and severely reduces quality of life. The degree of cachexia is inversely correlated with survival time; however, the exact mechanisms behind cancer-induced muscle wasting remain under investigation. RECENT FINDINGS Cytokines such as tumor necrosis factor-alpha trigger degradatory pathways through nuclear factor-kappaB signaling that activate the ubiquitin-proteasome system and muscle proteolysis. Androgen treatment has been shown to reduce inflammatory cytokines and even stimulate anti-inflammatory cytokine production. Amino acid supplementation has been shown to induce muscle protein synthesis in ovarian cancer patients. SUMMARY Targeted anabolic therapies aimed at preventing or reversing cancer cachexia might involve the combined use of androgens and amino acids working concurrently to enhance muscle protein synthesis and reduce muscle protein breakdown. Additional focused clinical studies are needed to identify muscle-specific targets or biomarkers for defined therapeutic approaches to slow or prevent cancer cachexia. In this review, we summarize the pathogenesis of cancer-related muscle wasting and discuss potential interventions at reversing or preventing cancer-related muscle loss.
Collapse
Affiliation(s)
- Kimberlee Burckart
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX
| | - Sorin Beca
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX
| | - Randall J. Urban
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX
| | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The present investigation is devoted to uncovering the different signaling pathways - particularly transcriptional factors - involved in muscle wasting. RECENT FINDINGS Although the search for the cachectic factor(s) started a long time ago, and although many scientific and economic efforts have been devoted to its discovery, we are still a long way from knowing the whole truth. In this review we describe recent findings about the tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, TWEAK and myostatin actions in cancer cachexia models. SUMMARY The main aim of the present review is to summarize and evaluate the different molecular mechanisms and catabolic mediators (mainly cytokines) involved in cancer cachexia since they may represent targets for future promising clinical investigations.
Collapse
|
43
|
Argilés JM, Olivan M, Busquets S, López-Soriano FJ. Optimal management of cancer anorexia-cachexia syndrome. Cancer Manag Res 2010; 2:27-38. [PMID: 21188094 PMCID: PMC3004581 DOI: 10.2147/cmar.s7101] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Indexed: 12/30/2022] Open
Abstract
According to a recent consensus, cachexia is a complex metabolic syndrome associated with underlying illness and characterized by loss of muscle with or without loss of fat mass. The prominent clinical feature of cachexia is weight loss. Cachexia occurs in the majority of cancer patients before death and it is responsible for the deaths of 22% of cancer patients. Although bodyweight is the most important endpoint of any cachexia treatment, body composition, physical performance and quality of life should be monitored. From the results presented here, one can speculate that a single therapy may not be completely successful in the treatment of cachexia. From this point of view, treatments involving different combinations are more likely to be successful. The objectives of any therapeutic combination are two-fold: an anticatabolic aim directed towards both fat and muscle catabolism and an anabolic objective leading to the synthesis of macromolecules such as contractile proteins.
Collapse
Affiliation(s)
- Josep M Argilés
- Departament de Bioquímica i Biologia Molecular, Universitat de Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
44
|
Benny Klimek ME, Aydogdu T, Link MJ, Pons M, Koniaris LG, Zimmers TA. Acute inhibition of myostatin-family proteins preserves skeletal muscle in mouse models of cancer cachexia. Biochem Biophys Res Commun 2009; 391:1548-54. [PMID: 20036643 DOI: 10.1016/j.bbrc.2009.12.123] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 12/21/2009] [Indexed: 02/07/2023]
Abstract
Cachexia, progressive loss of fat and muscle mass despite adequate nutrition, is a devastating complication of cancer associated with poor quality of life and increased mortality. Myostatin is a potent tonic muscle growth inhibitor. We tested how myostatin inhibition might influence cancer cachexia using genetic and pharmacological approaches. First, hypermuscular myostatin null mice were injected with Lewis lung carcinoma or B16F10 melanoma cells. Myostatin null mice were more sensitive to tumor-induced cachexia, losing more absolute mass and proportionately more muscle mass than wild-type mice. Because myostatin null mice lack expression from development, however, we also sought to manipulate myostatin acutely. The histone deacetylase inhibitor Trichostatin A has been shown to increase muscle mass in normal and dystrophic mice by inducing the myostatin inhibitor, follistatin. Although Trichostatin A administration induced muscle growth in normal mice, it failed to preserve muscle in colon-26 cancer cachexia. Finally we sought to inhibit myostatin and related ligands by administration of the Activin receptor extracellular domain/Fc fusion protein, ACVR2B-Fc. Systemic administration of ACVR2B-Fc potently inhibited muscle wasting and protected adipose stores in both colon-26 and Lewis lung carcinoma cachexia, without affecting tumor growth. Enhanced cachexia in myostatin knockouts indicates that host-derived myostatin is not the sole mediator of muscle wasting in cancer. More importantly, skeletal muscle preservation with ACVR2B-Fc establishes that targeting myostatin-family ligands using ACVR2B-Fc or related molecules is an important and potent therapeutic avenue in cancer cachexia.
Collapse
Affiliation(s)
- Margaret E Benny Klimek
- Department of Cell Biology & Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|
45
|
Murton A, Constantin D, Greenhaff P. The involvement of the ubiquitin proteasome system in human skeletal muscle remodelling and atrophy. Biochim Biophys Acta Mol Basis Dis 2008; 1782:730-43. [DOI: 10.1016/j.bbadis.2008.10.011] [Citation(s) in RCA: 197] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 10/23/2008] [Accepted: 10/24/2008] [Indexed: 12/14/2022]
|
46
|
Ballok DA, Sakic B. Purine receptor antagonist modulates serology and affective behaviors in lupus-prone mice: evidence of autoimmune-induced pain? Brain Behav Immun 2008; 22:1208-16. [PMID: 18601998 PMCID: PMC2783694 DOI: 10.1016/j.bbi.2008.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 06/09/2008] [Accepted: 06/09/2008] [Indexed: 12/31/2022] Open
Abstract
Neurologic and psychiatric (NP) manifestations are severe complications of systemic lupus erythematosus (SLE). As commonly seen in patients, spontaneous disease onset in the MRL/MpJ-Fas(lpr)/J (MRL-lpr) mouse model of NP-SLE is accompanied by increased autoantibodies, pro-inflammatory cytokines and behavioral dysfunction which precede neuroinflammation and structural brain lesions. The role of purinergic receptors in the regulation of immunity and behavior remains largely unexplored in the field of neuropsychiatry. To examine the possibility that purinoception is involved in the development of affective behaviors, the P2X purinoceptor antagonist, suramin, was administered to lupus-prone mice from 5 to 14 weeks of age. In addition to food and water measures, novel object and sucrose preference tests were performed to assess neophobic anxiety- and anhedonic-like behaviors. Enzyme-linked immunosorbant assays for anti-nuclear antibodies (ANA) and pro-inflammatory cytokines were employed in immunopathological analyses. Changes in dendritic morphology in the hippocampal CA1 region were examined by a Golgi impregnation method. Suramin significantly lowered serum ANA and prevented behavioral deficits, but did not prevent neuronal atrophy in MRL-lpr animals. In a new batch of asymptomatic mice, systemic administration of corticosterone was found to induce aberrations in CA1 dendrites, comparable to the "stress" of chronic disease. The precise mechanism(s) through which purine receptor inhibition exerted beneficial effects is not known. The present data supports the hypothesis that activation of the peripheral immune system induces nociceptive-related behavioral symptomatology which is attenuated by the analgesic effects of suramin. Hypercortisolemia may also initiate neuronal damage, and metabolic perturbations may underlie neuro-immuno-endocrine imbalances in MRL-lpr mice.
Collapse
Affiliation(s)
- David A Ballok
- Department of Surgery (Neurosurgery, Neurobiology), McMaster University, Canada L8N 3Z5.
| | | |
Collapse
|
47
|
Zhang Y, Wang S, Li Y, Xiao Z, Hu Z, Zhang J. Sophocarpine and matrine inhibit the production of TNF-alpha and IL-6 in murine macrophages and prevent cachexia-related symptoms induced by colon26 adenocarcinoma in mice. Int Immunopharmacol 2008; 8:1767-72. [PMID: 18775799 DOI: 10.1016/j.intimp.2008.08.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 08/12/2008] [Accepted: 08/18/2008] [Indexed: 12/18/2022]
Abstract
The present study aims to access the effects of sophora alkaloids on the production of pro-inflammatory cytokines and evaluate their therapeutic efficiency on cachexia. The comparative study showed that all sophora alkaloids tested here, including matrine, oxymatrine, sophocarpine, sophoramine, and sophoridine, inhibited TNF-alpha and IL-6 production in both RAW264.7 cells and murine primary macrophages, and sophocarpine showed the most potent inhibitory effect among them. Quantification of TNF-alpha and IL-6 mRNA in RAW264.7 cells by real-time RT-PCR revealed that both sophocarpine and matrine suppressed TNF-alpha and IL-6 expression and sophocarpine has stronger suppressing potency than matrine. Inoculation (s.c.) of colon26 adenocarcinoma cells into BALB/c mice induced cachexia, as evidenced by progressive weight loss, reduction in food intake, wasting of gastrocnemius muscle and epididymal fat, and increase in serum levels of TNF-alpha and IL-6. Administration of 50 mg/kg/d sophocarpine or matrine for 5 days from the onset of cachexia did not inhibit the tumor growth but resulted in attenuation of cachexia symptoms. Furthermore, sophocarpine and matrine decreased the serum levels of TNF-alpha and IL-6, and sophocarpine showed a better therapeutic effect than matrine. These results suggest that sophocarpine and matrine exert anti-cachectic effects probably through inhibition of TNF-alpha and IL-6.
Collapse
Affiliation(s)
- Yuefan Zhang
- Department of Biochemical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Cancer anorexia cachexia syndrome is frequent yet still a not well understood cancer-related problem. The pathophysiology of cancer cachexia is multifactorial. It is suggested to be the result of tumor-host interactions and studies of the disturbances seen during cancer anorexia cachexia syndrome, such as anorexia, hyper-metabolism, tissue wasting, metabolic abnormalities, and hormonal changes, all point to the involvement in one way or another of one key factor: cytokines. The purpose of this review is to summarize the latest developments in the field of cytokines and their role in cancer anorexia cachexia syndrome. The emphasis is on the role of cytokines in anorexia and tissue wasting.
Collapse
Affiliation(s)
- Nabila Bennani-Baiti
- Harry R. Horvitz Center for Palliative Medicine (a World Health Organization demonstration project in palliative medicine), Cleveland Clinic Taussig Cancer Center, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
49
|
Yano CL, Ventrucci G, Field WN, Tisdale MJ, Gomes-Marcondes MCC. Metabolic and morphological alterations induced by proteolysis-inducing factor from Walker tumour-bearing rats in C2C12 myotubes. BMC Cancer 2008; 8:24. [PMID: 18226207 PMCID: PMC2266935 DOI: 10.1186/1471-2407-8-24] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Accepted: 01/28/2008] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Patients with advanced cancer suffer from cachexia, which is characterised by a marked weight loss, and is invariably associated with the presence of tumoral and humoral factors which are mainly responsible for the depletion of fat stores and muscular tissue. METHODS In this work, we used cytotoxicity and enzymatic assays and morphological analysis to examine the effects of a proteolysis-inducing factor (PIF)-like molecule purified from ascitic fluid of Walker tumour-bearing rats (WF), which has been suggested to be responsible for muscle atrophy, on cultured C2C12 muscle cells. RESULTS WF decreased the viability of C2C12 myotubes, especially at concentrations of 20-25 mug.mL-1. There was an increase in the content of the pro-oxidant malondialdehyde, and a decrease in antioxidant enzyme activity. Myotubes protein synthesis decreased and protein degradation increased together with an enhanced in the chymotrypsin-like enzyme activity, a measure of functional proteasome activity, after treatment with WF. Morphological alterations such as cell retraction and the presence of numerous cells in suspension were observed, particularly at high WF concentrations. CONCLUSION These results indicate that WF has similar effects to those of proteolysis-inducing factor, but is less potent than the latter. Further studies are required to determine the precise role of WF in this experimental model.
Collapse
Affiliation(s)
- Claudia L Yano
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), CP 6109, 13083-970, Campinas, São Paulo, Brazil
| | - Gislaine Ventrucci
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), CP 6109, 13083-970, Campinas, São Paulo, Brazil
| | - William N Field
- Cancer Research Laboratory, Pharmaceutical Sciences Research Institute, Aston University, Birmingham, B4 7ET, UK
| | - Michael J Tisdale
- Cancer Research Laboratory, Pharmaceutical Sciences Research Institute, Aston University, Birmingham, B4 7ET, UK
| | - Maria Cristina C Gomes-Marcondes
- Departamento de Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), CP 6109, 13083-970, Campinas, São Paulo, Brazil
| |
Collapse
|
50
|
Argilés JM, López-Soriano FJ, Busquets S. Emerging drugs for cancer cachexia. Expert Opin Emerg Drugs 2007; 12:555-70. [PMID: 17979599 DOI: 10.1517/14728214.12.4.555] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cachexia is a complex syndrome. The main components of this pathological state are anorexia and metabolic abnormalities such as glucose intolerance, fat depletion and muscle protein catabolism among others. The altered metabolic status generates a high degree of energetic inefficiency that results in weight loss, fatigue and a considerable loss of muscle and, therefore, asthenia. The aim of the present article is to review the different therapeutic approaches and emerging drugs that have been designed to fight and counteract cachexia associated with cancer.
Collapse
Affiliation(s)
- Josep M Argilés
- Universitat de Barcelona, Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Diagonal 645, 08071-Barcelona, Spain.
| | | | | |
Collapse
|