1
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
2
|
Rashid MU, Lorzadeh S, Gao A, Ghavami S, Coombs KM. PSMA2 knockdown impacts expression of proteins involved in immune and cellular stress responses in human lung cells. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166617. [PMID: 36481484 DOI: 10.1016/j.bbadis.2022.166617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Proteasome subunit alpha type-2 (PSMA2) is a critical component of the 20S proteasome, which is the core particle of the 26S proteasome complex and is involved in cellular protein quality control by recognizing and recycling defective proteins. PSMA2 expression dysregulation has been detected in different human diseases and viral infections. No study yet has reported PSMA2 knockdown (KD) effects on the cellular proteome. METHODS We used SOMAScan, an aptamer-based multiplexed technique, to measure >1300 human proteins to determine the impact of PSMA2 KD on A549 human lung epithelial cells. RESULTS PSMA2 KD resulted in significant dysregulation of 52 cellular proteins involved in different bio-functions, including cellular movement and development, cell death and survival, and cancer. The immune system and signal transduction were the most affected cellular functions. PSMA2 KD caused dysregulation of several signaling pathways involved in immune response, cytokine signaling, organismal growth and development, cellular stress and injury (including autophagy and unfolded protein response), and cancer responses. CONCLUSIONS In summary, this study helps us better understand the importance of PSMA2 in different cellular functions, signaling pathways, and human diseases.
Collapse
Affiliation(s)
- Mahamud-Ur Rashid
- University of Manitoba, Department of Medical Microbiology & Infectious Diseases, Room 543 Basic Medical Sciences Building, 745 Bannatyne Ave., Winnipeg, MB R3E 0J9, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Ang Gao
- Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Kevin M Coombs
- University of Manitoba, Department of Medical Microbiology & Infectious Diseases, Room 543 Basic Medical Sciences Building, 745 Bannatyne Ave., Winnipeg, MB R3E 0J9, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada; Children's Hospital Research Institute of Manitoba, Room 513, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada.
| |
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
|
Ying L, Yao Y, Lv H, Lu G, Zhang Q, Yang Y, Zhou J. IL-17A contributes skeletal muscle atrophy in lung cancer-induced cachexia via JAK2/STAT3 pathway. Am J Physiol Cell Physiol 2022; 322:C814-C824. [PMID: 35319902 DOI: 10.1152/ajpcell.00463.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cachexia is a complex metabolic syndrome that occurs in approximately 50% of patients with cancer. Skeletal muscle atrophy is the primary clinical feature. Interleukin (IL)-17A, a pro-inflammatory factor, plays an important role in many chronic inflammatory diseases. Here, we describe a novel signaling pathway through which IL-17A induced muscle atrophy.We conducted a retrospective clinical study to investigate the relationship between IL-17A and the skeletal muscle index in patients with lung adenocarcinoma. We also investigated the involvement of JAK2/STAT3 signaling pathway regarding the main features of cachexia by injecting Lewis lung carcinoma (LLC) cells into C57BL/6 mice as a model to replicate cancer-induced cachexia. In vitro, C2C12 myotubes were treated with recombinant IL-17A, anti-IL-17A monoclonal antibody, STAT3 inhibitor AG490, and LLC-conditioned medium. Cell viability and aging was also evaluated. We found in cancer conditions, increased serum levels of IL-17A were related to muscle wasting. JAK2/STAT3 phosphorylation was observed in the muscle of LLC tumor-bearing mice, accompanied by decreased MHC/Myog levels and increased MuRF1/Atrogin-1 levels. Administration of anti-IL-17A monoclonal antibody and AG490 slowed muscle atrophy development. Consistent with the in vivo findings, C2C12 myotubes treated with IL-17A and LLC-conditioned medium demonstrated phosphorylated JAK2/STAT3 signaling, resulting in MHC loss and myotube atrophy. IL-17A also inhibited C2C12 cell proliferation, cell cycle breaking, and cellular senescence. Our results identify phosphorylation of IL-17A/JAK2/STAT3 signaling pathway appears to be an important component in the pathogenesis of LLC tumor-induced cachexia. Targeted therapy of IL-17A may be a promising approach to reduce skeletal muscle loss of patients with cancer.
Collapse
Affiliation(s)
- Lin Ying
- Department of Geriatrics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinan Yao
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Handi Lv
- Department of Respiratory Disease, Zhuji People's Hospital, Zhuji, China
| | - Guohua Lu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qin Zhang
- Department of Geriatrics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunmei Yang
- Department of Geriatrics, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Wilburn D, Ismaeel A, Machek S, Fletcher E, Koutakis P. Shared and distinct mechanisms of skeletal muscle atrophy: A narrative review. Ageing Res Rev 2021; 71:101463. [PMID: 34534682 DOI: 10.1016/j.arr.2021.101463] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 12/15/2022]
Abstract
Maintenance of skeletal muscle mass and function is an incredibly nuanced balance of anabolism and catabolism that can become distorted within different pathological conditions. In this paper we intend to discuss the distinct intracellular signaling events that regulate muscle protein atrophy for a given clinical occurrence. Aside from the common outcome of muscle deterioration, several conditions have at least one or more distinct mechanisms that creates unique intracellular environments that facilitate muscle loss. The subtle individuality to each of these given pathologies can provide both researchers and clinicians with specific targets of interest to further identify and increase the efficacy of medical treatments and interventions.
Collapse
Affiliation(s)
- Dylan Wilburn
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Ahmed Ismaeel
- Department of Biology, Baylor University, Waco, TX 76706, USA
| | - Steven Machek
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA
| | - Emma Fletcher
- Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; Department of Biology, Baylor University, Waco, TX 76706, USA
| | | |
Collapse
|
6
|
Mallard J, Hucteau E, Hureau TJ, Pagano AF. Skeletal Muscle Deconditioning in Breast Cancer Patients Undergoing Chemotherapy: Current Knowledge and Insights From Other Cancers. Front Cell Dev Biol 2021; 9:719643. [PMID: 34595171 PMCID: PMC8476809 DOI: 10.3389/fcell.2021.719643] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/10/2021] [Indexed: 01/18/2023] Open
Abstract
Breast cancer represents the most commonly diagnosed cancer while neoadjuvant and adjuvant chemotherapies are extensively used in order to reduce tumor development and improve disease-free survival. However, chemotherapy also leads to severe off-target side-effects resulting, together with the tumor itself, in major skeletal muscle deconditioning. This review first focuses on recent advances in both macroscopic changes and cellular mechanisms implicated in skeletal muscle deconditioning of breast cancer patients, particularly as a consequence of the chemotherapy treatment. To date, only six clinical studies used muscle biopsies in breast cancer patients and highlighted several important aspects of muscle deconditioning such as a decrease in muscle fibers cross-sectional area, a dysregulation of protein turnover balance and mitochondrial alterations. However, in comparison with the knowledge accumulated through decades of intensive research with many different animal and human models of muscle atrophy, more studies are necessary to obtain a comprehensive understanding of the cellular processes implicated in breast cancer-mediated muscle deconditioning. This understanding is indeed essential to ultimately lead to the implementation of efficient preventive strategies such as exercise, nutrition or pharmacological treatments. We therefore also discuss potential mechanisms implicated in muscle deconditioning by drawing a parallel with other cancer cachexia models of muscle wasting, both at the pre-clinical and clinical levels.
Collapse
Affiliation(s)
- Joris Mallard
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France.,Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Elyse Hucteau
- Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France.,Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Thomas J Hureau
- Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| | - Allan F Pagano
- Centre de Recherche en Biomédecine de Strasbourg (CRBS), Fédération de Médecine Translationnelle, UR 3072, Université de Strasbourg, Strasbourg, France.,Faculté des Sciences du Sport, Centre Européen d'Enseignement de Recherche et d'Innovation en Physiologie de l'Exercice (CEERIPE), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
7
|
Smeets JSJ, Horstman AMH, van Dijk DPJ, van Boxtel AGM, Ter Woorst JF, Damink SWMO, Schijns OEMG, van Loon LJC. Basal protein synthesis rates differ between vastus lateralis and rectus abdominis muscle. J Cachexia Sarcopenia Muscle 2021; 12:769-778. [PMID: 33951313 PMCID: PMC8200451 DOI: 10.1002/jcsm.12701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In vivo muscle protein synthesis rates are typically assessed by measuring the incorporation rate of stable isotope labelled amino acids in skeletal muscle tissue collected from vastus lateralis muscle. It remains to be established whether muscle protein synthesis rates in the vastus lateralis are representative of muscle protein synthesis rates of other muscle groups. We hypothesized that post-absorptive muscle protein synthesis rates differ between vastus lateralis and rectus abdominis, pectoralis major, or temporalis muscle in vivo in humans. METHODS Twenty-four patients (62 ± 3 years, 42% female), scheduled to undergo surgery, participated in this study and underwent primed continuous intravenous infusions with l-[ring-13 C6 ]-phenylalanine. During the surgical procedures, serum samples were collected, and muscle tissue was obtained from the vastus lateralis as well as from the rectus abdominis, pectoralis major, or temporalis muscle. Fractional mixed muscle protein synthesis rates (%/h) were assessed by measuring the incorporation of l-[ring-13 C6 ]-phenylalanine into muscle tissue protein. RESULTS Serum l-[ring-13 C6 ]-phenylalanine enrichments did not change throughout the infusion period. Post-absorptive muscle protein synthesis rates calculated based upon serum l-[ring-13 C6 ]-phenylalanine enrichments did not differ between vastus lateralis and rectus abdominis (0.032 ± 0.004 vs. 0.038 ± 0.003%/h), vastus lateralis and pectoralis major, (0.025 ± 0.003 vs. 0.022 ± 0.005%/h) or vastus lateralis and temporalis (0.047 ± 0.005 vs. 0.043 ± 0.005%/h) muscle, respectively (P > 0.05). When fractional muscle protein synthesis rates were calculated based upon tissue-free l-[ring-13 C6 ]-phenylalanine enrichments as the preferred precursor pool, muscle protein synthesis rates were significantly higher in rectus abdominis (0.089 ± 0.008%/h) compared with vastus lateralis (0.054 ± 0.005%/h) muscle (P < 0.01). No differences were observed between fractional muscle protein synthesis rates in vastus lateralis and pectoralis major (0.046 ± 0.003 vs. 0.041 ± 0.008%/h) or vastus lateralis and temporalis (0.073 ± 0.008 vs. 0.083 ± 0.011%/h) muscle, respectively. CONCLUSIONS Post-absorptive muscle protein synthesis rates are higher in rectus abdominis when compared with vastus lateralis muscle. Post-absorptive muscle protein synthesis rates do not differ between vastus lateralis and pectoralis major or temporalis muscle. Protein synthesis rates in muscle tissue samples obtained during surgery do not necessarily represent a good proxy for appendicular skeletal muscle protein synthesis rates.
Collapse
Affiliation(s)
- Joey S J Smeets
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Astrid M H Horstman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - David P J van Dijk
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Astrid G M van Boxtel
- Department of Cardiothoracic Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Joost F Ter Woorst
- Department of Cardiothoracic Surgery, Catharina Hospital, Eindhoven, The Netherlands
| | - Steven W M Olde Damink
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Department of General, Visceral and Transplantation Surgery, RWTH University Hospital Aachen, Aachen, Germany
| | - Olaf E M G Schijns
- Department of Neurosurgery, Maastricht University Medical Centre+, Maastricht, The Netherlands.,School of Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.,Academic Center for Epileptology, location Maastricht, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
8
|
Cancer cachexia: molecular mechanism and pharmacological management. Biochem J 2021; 478:1663-1688. [PMID: 33970218 DOI: 10.1042/bcj20201009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022]
Abstract
Cancer cachexia often occurs in malignant tumors and is a multifactorial and complex symptom characterized by wasting of skeletal muscle and adipose tissue, resulting in weight loss, poor life quality and shorter survival. The pathogenic mechanism of cancer cachexia is complex, involving a variety of molecular substrates and signal pathways. Advancements in understanding the molecular mechanisms of cancer cachexia have provided a platform for the development of new targeted therapies. Although recent outcomes of early-phase trials have showed that several drugs presented an ideal curative effect, monotherapy cannot be entirely satisfactory in the treatment of cachexia-associated symptoms due to its complex and multifactorial pathogenesis. Therefore, the lack of definitive therapeutic strategies for cancer cachexia emphasizes the need to develop a better understanding of the underlying mechanisms. Increasing evidences show that the progression of cachexia is associated with metabolic alternations, which mainly include excessive energy expenditure, increased proteolysis and mitochondrial dysfunction. In this review, we provided an overview of the key mechanisms of cancer cachexia, with a major focus on muscle atrophy, adipose tissue wasting, anorexia and fatigue and updated the latest progress of pharmacological management of cancer cachexia, thereby further advancing the interventions that can counteract cancer cachexia.
Collapse
|
9
|
Pham Van B, Nguyen Thi Thanh H, Le Thi H, Nguyen Le Tuan A, Dang Thi Thu H, Dang Viet D. Nutritional Status and Feeding Regimen of Patients with Esophagus Cancer-A Study from Vietnam. Healthcare (Basel) 2021; 9:healthcare9030289. [PMID: 33800823 PMCID: PMC8000479 DOI: 10.3390/healthcare9030289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/09/2022] Open
Abstract
Background: Esophagus cancer patients are at high risk of malnutrition. This study was performed to assess the nutritional status and dietary intake of newly diagnosed esophageal cancer patients in Vietnam National Cancer Hospital (NCH). Methods: A cross-sectional study was conducted on 206 early esophageal cancer inpatients after gastrostomy from September 2017 to June 2018. The chi-squared test, Fisher exact test, and Mann–Whitney test were performed. The software of the Vietnam National Institute of Nutrition was used to evaluate the dietary intake of patients. Results: All the participants were male with a mean age of 57.1 ± 8.5 years. Overall, 87.4% of patients had dysphagia. Furthermore, 82.5% and 90.8% of patients reported weight loss one and six months pre-diagnosis, respectively. Moreover, 52.9% of patients suffered from mild/moderate malnutrition and 29.6% of patients had severe malnutrition according to the Patient-Generated Subjective Global Assessment (PG-SGA). The body mass index (BMI) and mid upper arm circumference (MUAC) measurement revealed 47.6% and 50% of undernourished patients, respectively. The proportions of patients having malnutrition were 10.7%, 55.8%, and 27.2% according to albumin, prealbumin, and total lymphocyte counts, respectively. The means of energy, protein, lipid, and carbohydrate in the patients’ 24 h preoperative diets were 973.6 ± 443.0 kcal/day, 42.4 ± 21.6 g/day, 31.0 ± 15.5 g/day, and 130.0 ± 64.5 g/day. The total energy, total protein, animal protein, total lipid, and plant lipid in the dietary intake of patients were strongly correlated with age, economic classification, and PG-SGA (each p < 0.05). The total energy intake increased day by day, with the average energy intake of 1343.9 ± 521.3 kcal on the seventh day. Energy and protein response rates increased day by day and were highest at 7 days post-operation at 18.0% and 19.4%. Conclusion: Malnutrition and insufficient intake are noteworthy in esophageal cancer patients. The PG-SGA is strongly correlated with the dietary intake of patients. The results from this study will help medical staff to prevent malnutrition and improve the nutritional status of esophageal cancer inpatients. Furthermore, public awareness should be raised on recognizing weight loss as an early symptom of esophageal cancer and the utilization of preoperative assessment tools for nutritional assessment and malnutrition management.
Collapse
Affiliation(s)
- Binh Pham Van
- Abdominal Surgery Department 1 and Robotic Surgery Center, Vietnam National Cancer Hospital, Hanoi 12511, Vietnam
- Correspondence: or (B.P.V.); (H.L.T.)
| | - Hoa Nguyen Thi Thanh
- Clinical Nutrition Center, Vietnam National Cancer Hospital, Hanoi 12511, Vietnam; (H.N.T.T.); (H.D.T.T.)
| | - Huong Le Thi
- Clinical Nutrition Center, Vietnam National Cancer Hospital, Hanoi 12511, Vietnam; (H.N.T.T.); (H.D.T.T.)
- Institute of Preventive Medicine and Public Health, Hanoi Medical University, Hanoi 11521, Vietnam;
- Correspondence: or (B.P.V.); (H.L.T.)
| | - Anh Nguyen Le Tuan
- Institute of Preventive Medicine and Public Health, Hanoi Medical University, Hanoi 11521, Vietnam;
| | - Hang Dang Thi Thu
- Clinical Nutrition Center, Vietnam National Cancer Hospital, Hanoi 12511, Vietnam; (H.N.T.T.); (H.D.T.T.)
| | - Dung Dang Viet
- Gastrointestinal Surgery Center, 103 Military Hospital, Military Medical Academy, Hanoi 12109, Vietnam;
| |
Collapse
|
10
|
Dolly A, Dumas J, Servais S. Cancer cachexia and skeletal muscle atrophy in clinical studies: what do we really know? J Cachexia Sarcopenia Muscle 2020; 11:1413-1428. [PMID: 33053604 PMCID: PMC7749617 DOI: 10.1002/jcsm.12633] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/24/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Research investigators have shown a growing interest in investigating alterations underlying skeletal muscle wasting in patients with cancer. However, skeletal muscle dysfunctions associated with cancer cachexia have mainly been studied in preclinical models. In the present review, we summarize the results of clinical studies in which skeletal muscle biopsies were collected from cachectic vs. non-cachectic cancer patients. Most of these studies suggest the presence of significant physiological alterations in skeletal muscle from cachectic cancer patients. We suggest a hypothesis, which connects structural and metabolic parameters that may, at least in part, be responsible for the skeletal muscle atrophy characteristic of cancer cachexia. Finally, we discuss the importance of a better standardization of the diagnostic criteria for cancer cachexia, as well as the requirement for additional clinical studies to improve the robustness of these conclusions.
Collapse
Affiliation(s)
- Adeline Dolly
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Jean‐François Dumas
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| | - Stéphane Servais
- INSERM UMR 1069, Nutrition Croissance et CancerUniversité de ToursToursFrance
| |
Collapse
|
11
|
Iwasaki S, Zhu A, Hanley M, Venkatakrishnan K, Xia C. A Translational Physiologically Based Pharmacokinetics/Pharmacodynamics Framework of Target-Mediated Disposition, Target Inhibition and Drug-Drug Interactions of Bortezomib. AAPS JOURNAL 2020; 22:66. [PMID: 32291610 DOI: 10.1208/s12248-020-00448-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/11/2020] [Indexed: 11/30/2022]
Abstract
Bortezomib is a potent 20S proteasome inhibitor approved for the treatment of multiple myeloma and mantle cell lymphoma. Despite the extensive clinical use of bortezomib, the mechanism of the complex time-dependent pharmacokinetics of bortezomib has not been fully investigated in context of its pharmacodynamics (PD) and drug-drug interaction (DDI) profiles. Here, we aimed to develop a mechanistic physiologically based (PB) PK/PD model to project PK, blood target inhibition and DDI of bortezomib in patients. A minimal PBPK/PD model consisting of six compartments was constructed using a bottom-up approach with pre-clinical data and human physiological parameters. Specifically, the target-mediated drug disposition (TMDD) of bortezomib in red blood cells (RBC), which determines target inhibition in blood, was characterized by incorporating the proteasome binding affinity of bortezomib and the proteasome concentration in RBC. The hepatic clearance and fraction metabolized by different CYP isoforms were estimated from in vitro metabolism and phenotyping experiments. The established model adequately characterized the multi-exponential and time-dependent plasma pharmacokinetics, target binding and blood proteasome inhibition of bortezomib. Further, the model was able to accurately predict the impact of a strong CYP3A inducer (rifampicin) and inhibitor (ketoconazole) on bortezomib exposure. In conclusion, the mechanistic PBPK/PD model successfully described the complex pharmacokinetics, target inhibition and DDIs of bortezomib in patients. This study illustrates the importance of incorporating target biology, drug-target interactions and in vitro clearance parameters into mechanistic PBPK/PD models and the utility of such models for pharmacokinetic, pharmacodynamic and DDI predictions.
Collapse
Affiliation(s)
- Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International, Co., 35 Landsdowne Street, Cambridge, Massachusetts, USA.
| | - Andy Zhu
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International, Co., 35 Landsdowne Street, Cambridge, Massachusetts, USA
| | - Michael Hanley
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals International, Co., 35 Landsdowne Street, Cambridge, Massachusetts, USA
| | - Karthik Venkatakrishnan
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals International, Co., 35 Landsdowne Street, Cambridge, Massachusetts, USA
| | - Cindy Xia
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International, Co., 35 Landsdowne Street, Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
da Fonseca GWP, Farkas J, Dora E, von Haehling S, Lainscak M. Cancer Cachexia and Related Metabolic Dysfunction. Int J Mol Sci 2020; 21:ijms21072321. [PMID: 32230855 PMCID: PMC7177950 DOI: 10.3390/ijms21072321] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer cachexia is a complex multifactorial syndrome marked by a continuous depletion of skeletal muscle mass associated, in some cases, with a reduction in fat mass. It is irreversible by nutritional support alone and affects up to 74% of patients with cancer-dependent on the underlying type of cancer-and is associated with physical function impairment, reduced response to cancer-related therapy, and higher mortality. Organs, like muscle, adipose tissue, and liver, play an important role in the progression of cancer cachexia by exacerbating the pro- and anti-inflammatory response initially activated by the tumor and the immune system of the host. Moreover, this metabolic dysfunction is produced by alterations in glucose, lipids, and protein metabolism that, when maintained chronically, may lead to the loss of skeletal muscle and adipose tissue. Although a couple of drugs have yielded positive results in increasing lean body mass with limited impact on physical function, a single therapy has not lead to effective treatment of this condition. Therefore, a multimodal intervention, including pharmacological agents, nutritional support, and physical exercise, may be a reasonable approach for future studies to better understand and prevent the wasting of body compartments in patients with cancer cachexia.
Collapse
Affiliation(s)
- Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo SP 05403-900, Brazil or
- Department of Cardiology and Pneumology, University Medicine Göttingen (UMG), DE-37075 Goettingen, Germany
| | - Jerneja Farkas
- Research Unit, General Hospital Murska Sobota, SI-9000 Murska Sobota, Slovenia;
- National Institute of Public Health, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Eva Dora
- Division of Cardiology, General Hospital Murska Sobota, SI-9000 Murska Sobota, Slovenia;
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University Medicine Göttingen (UMG), DE-37075 Goettingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Goettingen, DE-37099 Goettingen, Germany
- Correspondence: (S.v.H.); (M.L.); Tel.: +49-551-3920-911 (S.v.H.); +386-251-23-733 (M.L.); Fax: +49-551-3920-918 (S.v.H.); Fax: +386-252-11-007 (M.L.)
| | - Mitja Lainscak
- Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
- Division of Cardiology, General Hospital Murska Sobota, SI-9000 Murska Sobota, Slovenia;
- Correspondence: (S.v.H.); (M.L.); Tel.: +49-551-3920-911 (S.v.H.); +386-251-23-733 (M.L.); Fax: +49-551-3920-918 (S.v.H.); Fax: +386-252-11-007 (M.L.)
| |
Collapse
|
13
|
Sato S, Gao S, Puppa MJ, Kostek MC, Wilson LB, Carson JA. High-Frequency Stimulation on Skeletal Muscle Maintenance in Female Cachectic Mice. Med Sci Sports Exerc 2020; 51:1828-1837. [PMID: 30933004 DOI: 10.1249/mss.0000000000001991] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer cachexia, an unintentional body weight loss due to cancer, affects patients' survival, quality of life, and response to chemotherapy. Although exercise training is a promising intervention to prevent and treat cancer cachexia, our mechanistic understanding of cachexia's effect on contraction-induced muscle adaptation has been limited to the examination of male mice. Because sex can affect muscle regeneration and response to contraction in humans and mice, the effect of cachexia on the female response to eccentric contraction warrants further investigation. PURPOSE The purpose of this study was to determine whether high-frequency electric stimulation (HFES) could attenuate muscle mass loss during the progression of cancer cachexia in female tumor-bearing mice. METHODS Female wild-type (WT) and Apc (Min) mice (16-18 wk old) performed either repeated bouts or a single bout of HFES (10 sets of 6 repetitions, ~22 min), which eccentrically contracts the tibialis anterior (TA) muscle. TA myofiber size, oxidative capacity, anabolic signaling, and catabolic signaling were examined. RESULTS Min had reduced TA muscle mass and type IIa and type IIb fiber sizes compared with WT. HFES increased the muscle weight and the mean cross-sectional area of type IIa and type IIb fibers in WT and Min mice. HFES increased mTOR signaling and myofibrillar protein synthesis and attenuated cachexia-induced AMPK activity. HFES attenuated the cachexia-associated decrease in skeletal muscle oxidative capacity. CONCLUSION HFES in female mice can activate muscle protein synthesis through mTOR signaling and repeated bouts of contraction can attenuate cancer-induced muscle mass loss.
Collapse
Affiliation(s)
- Shuichi Sato
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC.,School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA
| | - Song Gao
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Melissa J Puppa
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - Matthew C Kostek
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| | - L Britt Wilson
- Department of Pharmacology, Physiology, and Neuroscience, School of Medicine, University of South Carolina, Columbia, SC
| | - James A Carson
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC
| |
Collapse
|
14
|
Anoveros‐Barrera A, Bhullar AS, Stretch C, Esfandiari N, Dunichand‐Hoedl AR, Martins KJ, Bigam D, Khadaroo RG, McMullen T, Bathe OF, Damaraju S, Skipworth RJ, Putman CT, Baracos VE, Mazurak VC. Clinical and biological characterization of skeletal muscle tissue biopsies of surgical cancer patients. J Cachexia Sarcopenia Muscle 2019; 10:1356-1377. [PMID: 31307124 PMCID: PMC9536086 DOI: 10.1002/jcsm.12466] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Researchers increasingly use intraoperative muscle biopsy to investigate mechanisms of skeletal muscle atrophy in patients with cancer. Muscles have been assessed for morphological, cellular, and biochemical features. The aim of this study was to conduct a state-of-the-science review of this literature and, secondly, to evaluate clinical and biological variation in biopsies of rectus abdominis (RA) muscle from a cohort of patients with malignancies. METHODS Literature was searched for reports on muscle biopsies from patients with a cancer diagnosis. Quality of reports and risk of bias were assessed. Data abstracted included patient characteristics and diagnoses, sample size, tissue collection and biobanking procedures, and results. A cohort of cancer patients (n = 190, 88% gastrointestinal malignancies), who underwent open abdominal surgery as part of their clinical care, consented to RA biopsy from the site of incision. Computed tomography (CT) scans were used to quantify total abdominal muscle and RA cross-sectional areas and radiodensity. Biopsies were assessed for muscle fibre area (μm2 ), fibre types, myosin heavy chain isoforms, and expression of genes selected for their involvement in catabolic pathways of muscle. RESULTS Muscle biopsy occurred in 59 studies (total N = 1585 participants). RA was biopsied intraoperatively in 40 studies (67%), followed by quadriceps (26%; percutaneous biopsy) and other muscles (7%). Cancer site and stage, % of male participants, and age were highly variable between studies. Details regarding patient medical history and biopsy procedures were frequently absent. Lack of description of the population(s) sampled and low sample size contributed to low quality and risk of bias. Weight-losing cases were compared with weight stable cancer or healthy controls without considering a measure of muscle mass in 21 out of 44 studies. In the cohort of patients providing biopsy for this study, 78% of patients had preoperative CT scans and a high proportion (64%) met published criteria for sarcopenia. Fibre type distribution in RA was type I (46% ± 13), hybrid type I/IIA (1% ± 1), type IIA (36% ± 10), hybrid type IIA/D (15% ± 14), and type IID (2% ± 5). Sexual dimorphism was prominent in RA CT cross-sectional area, mean fibre cross-sectional area, and in expression of genes associated with muscle growth, apoptosis, and inflammation (P < 0.05). Medical history revealed multiple co-morbid conditions and medications. CONCLUSIONS Continued collaboration between researchers and cancer surgeons enables a more complete understanding of mechanisms of cancer-associated muscle atrophy. Standardization of biobanking practices, tissue manipulation, patient characterization, and classification will enhance the consistency, reliability, and comparability of future studies.
Collapse
Affiliation(s)
- Ana Anoveros‐Barrera
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - Amritpal S. Bhullar
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | | | - Nina Esfandiari
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Abha R. Dunichand‐Hoedl
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - Karen J.B. Martins
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - David Bigam
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Rachel G. Khadaroo
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Todd McMullen
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Oliver F. Bathe
- Department of OncologyUniversity of CalgaryCalgaryABCanada
- Department of SurgeryUniversity of CalgaryCalgaryABCanada
| | - Sambasivarao Damaraju
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonABCanada
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | | | - Charles T. Putman
- Faculty of Kinesiology, Sport, and Recreation, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Vickie E. Baracos
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Vera C. Mazurak
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
15
|
Li Y, Zhang F, Modrak S, Little A, Zhang H. Chronic Alcohol Consumption Enhances Skeletal Muscle Wasting in Mice Bearing Cachectic Cancers: The Role of TNFα/Myostatin Axis. Alcohol Clin Exp Res 2019; 44:66-77. [PMID: 31657476 DOI: 10.1111/acer.14221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/03/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic alcohol consumption enhances cancer-associated cachexia, which is one of the major causes of decreased survival. The precise molecular mechanism of how alcohol consumption enhances cancer-associated cachexia, especially skeletal muscle loss, remains to be elucidated. METHODS We used a mouse model of chronic alcohol consumption, in which 20% (w/v) alcohol was provided as sole drinking fluid, and Lewis lung carcinoma to study the underlying mechanisms. RESULTS We found that alcohol consumption up-regulated the expression of MAFbx, MuRF-1, and LC3 in skeletal muscle, suggesting that alcohol enhanced ubiquitin-mediated proteolysis and LC3-mediated autophagy. Alcohol consumption enhanced phosphorylation of Smad2/3, p38, and ERK and decreased the phosphorylation of FOXO1. These are the signaling molecules governing protein degradation pathways. Moreover, alcohol consumption slightly up-regulated the expression of insulin receptor substrate-1, did not affect phosphatidylinositol-3 kinase, but decreased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), and down-regulated the expression of Raptor and p70 ribosomal kinase S6 kinase, suggesting that alcohol impaired protein synthesis signaling pathway in skeletal muscle of tumor-bearing mice. Alcohol consumption enhanced the expression of myostatin in skeletal muscle, plasma, and tumor, but did not affect the expression of myostatin in non-tumor-bearing mice. In TNFα knockout mice, the effects of alcohol-enhanced expression of myostatin and protein degradation-related signaling molecules, and decreased protein synthesis signaling in skeletal muscle were abolished. Consequently, alcohol consumption neither affected cancer-associated cachexia nor decreased the survival of TNFα KO mice bearing cachectic cancer. CONCLUSIONS Chronic alcohol consumption enhances cancer-associated skeletal muscle loss through suppressing Akt/mTOR-mediated protein synthesis pathway and enhancing protein degradation pathways. This process is initiated by TNFα and mediated by myostatin.
Collapse
Affiliation(s)
- Yuanfei Li
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.,Department of Oncology, (YL), The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Faya Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Samantha Modrak
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Alex Little
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Hui Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
16
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
17
|
Zamanian Azodi M, Rezaei Tavirani M, Rostami-Nejad M, Rezaei-Tavirani M. Comparative Bioinformatics Characteristic of Bladder Cancer Stage 2 from Stage 4 Expression Profile: A Network-Based Study. Galen Med J 2018; 7:e1279. [PMID: 34466446 PMCID: PMC8343782 DOI: 10.22086/gmj.v0i0.1279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/09/2018] [Accepted: 07/22/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Bladder cancer (BC) has remained as one of the most challenging issues in medicine. The aim of this study was to investigate the differential network analysis of stages 2 and 4 of BC to better understand the molecular pathology of these states. MATERIALS AND METHODS We chose gene expression data of GSE52519 from Gene Expression Omnibus (GEO) database analyzed by the GEO2R online tool. Cytoscape version 3.6.1 and its algorithms are the methods applied for the network construction and investigation of differentially expressed genes (DEG) in these states. RESULT Our result revealed that the analysis DEGs provides useful information about a common molecular feature of stages 2 and 4 of BC. CONCLUSION Consequently, the network finding revealed that more investigation about stage 2 is required to achieve an effective therapeutic protocol to block the transition from stage 2 to stage 4.
Collapse
Affiliation(s)
- Mona Zamanian Azodi
- Student Research Committee, Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
18
|
Bouchè M, Lozanoska-Ochser B, Proietti D, Madaro L. Do neurogenic and cancer-induced muscle atrophy follow common or divergent paths? Eur J Transl Myol 2018; 28:7931. [PMID: 30662704 PMCID: PMC6317130 DOI: 10.4081/ejtm.2018.7931] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/05/2018] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle is a dynamic tissue capable of responding to a large variety of physiological stimuli by adjusting muscle fiber size, metabolism and function. However, in pathological conditions such as cancer and neural disorders, this finely regulated homeostasis is impaired leading to severe muscle wasting, reduced muscle fiber size (atrophy), and impaired function. These disease features develop due to enhanced protein breakdown, which relies on two major degradation systems: the ubiquitin-proteasome and the autophagy-lysosome. These systems are independently regulated by different signalling pathways, which in physiological conditions, determine protein and organelle turnover. However, alterations in one or both systems, as it happens in several disorders, leads to enhanced protein breakdown and muscle atrophy. Although this is a common feature in the different types of muscle atrophy, the relative contribution of each of these systems is still under debate. Here, we will briefly describe the regulation and the activity of the ubiquitin-proteasome and the autophagy-lysosome systems during muscle wasting. We will then discuss what we know regarding how these pathways are involved in cancer induced and in neurogenic muscle atrophy, highlighting common and divergent paths. It is now clear that there is no one unifying common mechanism that can be applied to all models of muscle loss. Detailed understanding of the pathways and proteolysis mechanisms involved in each model will hopefully help the development of drugs to counteract muscle wasting in specific conditions.
Collapse
Affiliation(s)
- Marina Bouchè
- DAHFMO, Unit of Histology, Sapienza University of Rome, 00161 Rome, Italy.,Interuniversity Institute of Myology, Italy
| | | | - Daisy Proietti
- DAHFMO, Unit of Histology, Sapienza University of Rome, 00161 Rome, Italy.,IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Luca Madaro
- IRCCS, Fondazione Santa Lucia, Rome, Italy.,Interuniversity Institute of Myology, Italy
| |
Collapse
|
19
|
|
20
|
Aversa Z, Costelli P, Muscaritoli M. Cancer-induced muscle wasting: latest findings in prevention and treatment. Ther Adv Med Oncol 2017; 9:369-382. [PMID: 28529552 PMCID: PMC5424865 DOI: 10.1177/1758834017698643] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/14/2017] [Indexed: 12/17/2022] Open
Abstract
Cancer cachexia is a severe and disabling clinical condition that frequently accompanies the development of many types of cancer. Muscle wasting is the hallmark of cancer cachexia and is associated with serious clinical consequences such as physical impairment, poor quality of life, reduced tolerance to treatments and shorter survival. Cancer cachexia may evolve through different stages of clinical relevance, namely pre-cachexia, cachexia and refractory cachexia. Given its detrimental clinical consequences, it appears mandatory to prevent and/or delay the progression of cancer cachexia to its refractory stage by implementing the early recognition and treatment of the nutritional and metabolic alterations occurring during cancer. Research on the molecular mechanisms underlying muscle wasting during cancer cachexia has expanded in the last few years, allowing the identification of several potential therapeutic targets and the development of many promising drugs. Several of these agents have already reached the clinical evaluation, but it is becoming increasingly evident that a single therapy may not be completely successful in the treatment of cancer-related muscle wasting, given its multifactorial and complex pathogenesis. This suggests that early and structured multimodal interventions (including targeted nutritional supplementation, physical exercise and pharmacological interventions) are necessary to prevent and/or treat the devastating consequences of this cancer comorbidity, and future research should focus on this approach.
Collapse
Affiliation(s)
- Zaira Aversa
- Department of Clinical Medicine, Sapienza University of Rome, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Maurizio Muscaritoli
- Department of Clinical Medicine, Sapienza, University of Rome, Viale dell’Università 37, 00185 Rome, Italy
| |
Collapse
|
21
|
Sakuma K, Aoi W, Yamaguchi A. Molecular mechanism of sarcopenia and cachexia: recent research advances. Pflugers Arch 2017; 469:573-591. [PMID: 28101649 DOI: 10.1007/s00424-016-1933-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle provides a fundamental basis for human function, enabling locomotion and respiration. Muscle loss occurs as a consequence of several chronic diseases (cachexia) and normal aging (sarcopenia). Although many negative regulators (atrogin-1, muscle ring finger-1, nuclear factor-kappaB (NF-κB), myostatin, etc.) have been proposed to enhance protein degradation during both sarcopenia and cachexia, the adaptation of these mediators markedly differs within both conditions. Sarcopenia and cachectic muscles have been demonstrated to be abundant in myostatin-linked molecules. The ubiquitin-proteasome system (UPS) is activated during rapid atrophy model (cancer cachexia), but few mediators of the UPS change during sarcopenia. NF-κB signaling is activated in cachectic, but not in sarcopenic, muscle. Recent studies have indicated the age-related defect of autophagy signaling in skeletal muscle, whereas autophagic activation occurs in cachectic muscle. This review provides recent research advances dealing with molecular mediators modulating muscle mass in both sarcopenia and cachexia.
Collapse
Affiliation(s)
- Kunihiro Sakuma
- Institute for Liberal Arts, Environment and Society, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
| | - Wataru Aoi
- Laboratory of Health Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, 1-5 Hangi-cho Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Akihiko Yamaguchi
- Department of Physical Therapy, Health Sciences University of Hokkaido, Kanazawa, Ishikari-Tobetsu, Hokkaido, 061-0293, Japan
| |
Collapse
|
22
|
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
|
23
|
Patel HJ, Patel BM. TNF-α and cancer cachexia: Molecular insights and clinical implications. Life Sci 2016; 170:56-63. [PMID: 27919820 DOI: 10.1016/j.lfs.2016.11.033] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/27/2016] [Accepted: 11/30/2016] [Indexed: 12/20/2022]
Abstract
Cancer cachexia characterized by a chronic wasting syndrome, involves skeletal muscle loss and adipose tissue loss and resistance to conventional nutritional support. Cachexia is responsible for the reduction in quality and length of life of cancer patients. It also decreases the muscle strength of the patients. The pro-inflammatory and pro-cachectic factors produced by the tumor cells have important role in genesis of cachexia. A number of pro-inflammatory cytokines, like interleukin-1 (IL-1), IL-6, tumor necrosis factor- alpha (TNF-α) may have important role in the pathological mechanisms of cachexia in cancer. Particularly, TNF-α has a direct catabolic effect on skeletal muscle and causes wasting of muscle by the induction of the ubiquitin-proteasome system (UPS). In cancer cachexia condition, there is alteration in carbohydrate, protein and fat metabolism. TNF-α is responsible for the increase in gluconeogenesis, loss of adipose tissue and proteolysis, while causing decrease in protein, lipid and glycogen synthesis. It has been associated with the formation of IL-1 and increases the uncoupling protein-2 (UCP2) and UCP3 expression in skeletal muscle in cachectic state. The main aim of the present review is to evaluate and discuss the role of TNF-α in different metabolic alterations and muscle wasting in cancer cachexia.
Collapse
|
24
|
Rom O, Reznick AZ. The role of E3 ubiquitin-ligases MuRF-1 and MAFbx in loss of skeletal muscle mass. Free Radic Biol Med 2016; 98:218-230. [PMID: 26738803 DOI: 10.1016/j.freeradbiomed.2015.12.031] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/30/2015] [Accepted: 12/25/2015] [Indexed: 12/21/2022]
Abstract
The ubiquitin-proteasome system (UPS) is the main regulatory mechanism of protein degradation in skeletal muscle. The ubiquitin-ligase enzymes (E3s) have a central role in determining the selectivity and specificity of the UPS. Since their identification in 2001, the muscle specific E3s, muscle RING finger-1 (MuRF-1) and muscle atrophy F-box (MAFbx), have been shown to be implicated in the regulation of skeletal muscle atrophy in various pathological and physiological conditions. This review aims to explore the involvement of MuRF-1 and MAFbx in catabolism of skeletal muscle during various pathologies, such as cancer cachexia, sarcopenia of aging, chronic kidney disease (CKD), diabetes, and chronic obstructive pulmonary disease (COPD). In addition, the effects of various lifestyle and modifiable factors (e.g. nutrition, exercise, cigarette smoking, and alcohol) on MuRF-1 and MAFbx regulation will be discussed. Finally, evidence of potential strategies to protect against skeletal muscle wasting through inhibition of MuRF-1 and MAFbx expression will be explored.
Collapse
Affiliation(s)
- Oren Rom
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, P.O. Box 9649, Haifa, Israel.
| | - Abraham Z Reznick
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, P.O. Box 9649, Haifa, Israel
| |
Collapse
|
25
|
The Janus-Faced Role of Antioxidants in Cancer Cachexia: New Insights on the Established Concepts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:9579868. [PMID: 27642498 PMCID: PMC5013212 DOI: 10.1155/2016/9579868] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/28/2016] [Accepted: 07/17/2016] [Indexed: 12/14/2022]
Abstract
Chronic inflammation and excessive loss of skeletal muscle usually occur during cancer cachexia, leading to functional impairment and delaying the cure of cancer. The release of cytokines by tumor promotes the formation of reactive oxygen species (ROS), which in turn regulate catabolic pathways involved in muscle atrophy. ROS also exert a dual role within tumor itself, as they can either promote proliferation and vascularization or induce senescence and apoptosis. Accordingly, previous studies that used antioxidants to modulate these ROS-dependent mechanisms, in cancer and cancer cachexia, have obtained contradictory results, hence the need to gather the main findings of these studies and draw global conclusions in order to stimulate more oriented research in this field. Based on the literature reviewed in this paper, it appears that antioxidant supplementation is (1) beneficial in cancer cachectic patients with antioxidant deficiencies, (2) most likely harmful in cancer patients with adequate antioxidant status (i.e., lung, gastrointestinal, head and neck, and esophageal), and (3) not recommended when undergoing radiotherapy. At the moment, measuring the blood levels of antioxidants may help to identify patients with systemic deficiencies. This approach is simple to realize but could not be a gold standard method for cachexia, as it does not necessarily reflect the redox state in other organs, like muscle.
Collapse
|
26
|
Abstract
Metabolic dysfunction contributes to the clinical deterioration observed in advanced cancer patients and is characterized by weight loss, skeletal muscle wasting, and atrophy of the adipose tissue. This systemic syndrome, termed cancer-associated cachexia (CAC), is a major cause of morbidity and mortality. While once attributed solely to decreased food intake, the present description of cancer cachexia is a disorder of multiorgan energy imbalance. Here we review the molecules and pathways responsible for metabolic dysfunction in CAC and the ideas that led to the current understanding.
Collapse
Affiliation(s)
- Michele Petruzzelli
- Department of Oncology, The Medical Research Council Cancer Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdon
| | - Erwin F Wagner
- Genes, Development, and Disease Group, Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid 28029, Spain
| |
Collapse
|
27
|
Oliveira AG, Gomes-Marcondes MCC. Metformin treatment modulates the tumour-induced wasting effects in muscle protein metabolism minimising the cachexia in tumour-bearing rats. BMC Cancer 2016; 16:418. [PMID: 27388367 PMCID: PMC4936094 DOI: 10.1186/s12885-016-2424-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 06/15/2016] [Indexed: 01/10/2023] Open
Abstract
Background Cancer-cachexia state frequently induces both fat and protein wasting, leading to death. In this way, the knowledge of the mechanism of drugs and their side effects can be a new feature to treat and to have success, contributing to a better life quality for these patients. Metformin is an oral drug used in type 2 diabetes mellitus, showing inhibitory effect on proliferation in some neoplastic cells. For this reason, we evaluated its modulatory effect on Walker-256 tumour evolution and also on protein metabolism in gastrocnemius muscle and body composition. Methods Wistar rats received or not tumour implant and metformin treatment and were distributed into four groups, as followed: control (C), Walker 256 tumour-bearing (W), metformin-treated (M) and tumour-bearing treated with metformin (WM). Animals were weighed three times a week, and after cachexia state has been detected, the rats were euthanised and muscle and tumour excised and analysed by biochemical and molecular assays. Results Tumour growth promoted some deleterious effects on chemical body composition, increasing water and decreasing fat percentage, and reducing lean body mass. In muscle tissue, tumour led to a decreased protein synthesis and an increased proteolysis, showing the higher activity of the ubiquitin-proteasome pathway. On the other hand, the metformin treatment likely minimised the tumour-induced wasting state; in this way, this treatment ameliorated chemical body composition, reduced the higher activities of proteolytic enzymes and decreased the protein waste. Conclusion Metformin treatment not only decreases the tumour growth but also improves the protein metabolism in gastrocnemius muscle in tumour-bearing rats.
Collapse
Affiliation(s)
- André G Oliveira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, UNICAMP, Rua Monteiro Lobato, 255, CP 6109, Campinas, São Paulo, 13083862, Brazil
| | - Maria Cristina C Gomes-Marcondes
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, UNICAMP, Rua Monteiro Lobato, 255, CP 6109, Campinas, São Paulo, 13083862, Brazil.
| |
Collapse
|
28
|
Murton AJ, Maddocks M, Stephens FB, Marimuthu K, England R, Wilcock A. Consequences of Late-Stage Non-Small-Cell Lung Cancer Cachexia on Muscle Metabolic Processes. Clin Lung Cancer 2016; 18:e1-e11. [PMID: 27461772 DOI: 10.1016/j.cllc.2016.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/07/2016] [Accepted: 06/13/2016] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The loss of muscle is common in patients with advanced non-small-cell lung cancer (NSCLC) and contributes to the high morbidity and mortality of this group. The exact mechanisms behind the muscle loss are unclear. PATIENTS AND METHODS To investigate this, 4 patients with stage IV NSCLC who met the clinical definitions for sarcopenia and cachexia were recruited, along with 4 age-matched healthy volunteers. After an overnight fast, biopsy specimens were obtained from the vastus lateralis, and the key components associated with inflammation and the control of muscle protein, carbohydrate, and fat metabolism were assessed. RESULTS Compared with the healthy volunteers, significant increases in mRNA levels for interleukin-6 and NF-κB signaling and lower intramyocellular lipid content in slow-twitch fibers were observed in NSCLC patients. Although a significant decrease in phosphorylation of the mechanistic target of rapamycin (mTOR) signaling protein 4E-BP1 (Ser65) was observed, along with a trend toward reduced p70 S6K (Thr389) phosphorylation (P = .06), no difference was found between groups for the mRNA levels of MAFbx (muscle atrophy F box) and MuRF1 (muscle ring finger protein 1), chymotrypsin-like activity of the proteasome, or protein levels of multiple proteasome subunits. Moreover, despite decreases in intramyocellular lipid content, no robust changes in mRNA levels for key proteins involved in insulin signaling, glycolysis, oxidative metabolism, or fat metabolism were observed. CONCLUSION These findings suggest that examining the contribution of suppressed mTOR signaling in the loss of muscle mass in late-stage NSCLC patients is warranted and reinforces our need to understand the potential contribution of impaired fat metabolism and muscle protein synthesis in the etiology of cancer cachexia.
Collapse
Affiliation(s)
- Andrew J Murton
- Division of Nutritional Sciences, School of Biosciences, The University of Nottingham, Loughborough, United Kingdom; MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, United Kingdom.
| | - Matthew Maddocks
- Department of Palliative Medicine, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom; King's College London, Cicely Saunders Institute, London, United Kingdom
| | - Francis B Stephens
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, United Kingdom
| | - Kanagaraj Marimuthu
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, United Kingdom
| | - Ruth England
- Department of Palliative Medicine, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Andrew Wilcock
- Department of Palliative Medicine, The University of Nottingham, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| |
Collapse
|
29
|
Gallagher IJ, Jacobi C, Tardif N, Rooyackers O, Fearon K. Omics/systems biology and cancer cachexia. Semin Cell Dev Biol 2016; 54:92-103. [DOI: 10.1016/j.semcdb.2015.12.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 10/22/2022]
|
30
|
Mueller TC, Bachmann J, Prokopchuk O, Friess H, Martignoni ME. Molecular pathways leading to loss of skeletal muscle mass in cancer cachexia--can findings from animal models be translated to humans? BMC Cancer 2016; 16:75. [PMID: 26856534 PMCID: PMC4746781 DOI: 10.1186/s12885-016-2121-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/03/2016] [Indexed: 02/06/2023] Open
Abstract
Background Cachexia is a multi-factorial, systemic syndrome that especially affects patients with cancer of the gastrointestinal tract, and leads to reduced treatment response, survival and quality of life. The most important clinical feature of cachexia is the excessive wasting of skeletal muscle mass. Currently, an effective treatment is still lacking and the search for therapeutic targets continues. Even though a substantial number of animal studies have contributed to a better understanding of the underlying mechanisms of the loss of skeletal muscle mass, subsequent clinical trials of potential new drugs have not yet yielded any effective treatment for cancer cachexia. Therefore, we questioned to which degree findings from animal studies can be translated to humans in clinical practice and research. Discussion A substantial amount of animal studies on the molecular mechanisms of muscle wasting in cancer cachexia has been conducted in recent years. This extensive review of the literature showed that most of their observations could not be consistently reproduced in studies on human skeletal muscle samples. However, studies on human material are scarce and limited in patient numbers and homogeneity. Therefore, their results have to be interpreted critically. Summary More research is needed on human tissue samples to clarify the signaling pathways that lead to skeletal muscle loss, and to confirm pre-selected drug targets from animal models in clinical trials. In addition, improved diagnostic tools and standardized clinical criteria for cancer cachexia are needed to conduct standardized, randomized controlled trials of potential drug candidates in the future.
Collapse
Affiliation(s)
- Tara C Mueller
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675, Munich, Germany.
| | - Jeannine Bachmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675, Munich, Germany
| | - Olga Prokopchuk
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675, Munich, Germany
| | - Marc E Martignoni
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, D-81675, Munich, Germany
| |
Collapse
|
31
|
Couch ME, Dittus K, Toth MJ, Willis MS, Guttridge DC, George JR, Chang EY, Gourin CG, Der-Torossian H. Cancer cachexia update in head and neck cancer: Pathophysiology and treatment. Head Neck 2015; 37:1057-72. [PMID: 24634283 DOI: 10.1002/hed.23696] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/10/2023] Open
Abstract
The pathophysiology of cancer cachexia remains complex. A comprehensive literature search was performed up to April 2013 using PubMed, the Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, and the Google search engine. In this review, we focus on the different mediators of impaired anabolism and upregulated catabolism that alter the skeletal muscle homeostasis resulting in the wasting of cancer cachexia. We present recent evidence of targeted treatment modalities from clinical trials along with their potential mechanisms of action. We also report on the most current evidence from randomized clinical trials using multimodal treatments in patients with cancer cachexia, but also the evidence from head and neck cancer-specific trials. A more complete understanding of the pathophysiology of the syndrome may lead to more effective targeted therapies and improved outcomes for patients.
Collapse
Affiliation(s)
- Marion E Couch
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Vermont Cancer Center, University of Vermont, College of Medicine, Burlington, Vermont
| | - Kim Dittus
- Division of Hematology-Oncology, Department of Medicine, Vermont Cancer Center, University of Vermont, College of Medicine, Burlington, Vermont
| | - Michael J Toth
- Department of Molecular Physiology and Biophysics, University of Vermont, College of Medicine, Burlington, Vermont
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology, and Medical Genetics, Ohio State University, Columbus, Ohio
| | - Jonathan R George
- Department of Otolaryngology - Head and Neck Surgery, University of California, San Francisco, California
| | - Eric Y Chang
- University of Vermont, College of Medicine, Burlington, Vermont
| | - Christine G Gourin
- Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins Hospital, Baltimore, Maryland
| | - Hirak Der-Torossian
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Vermont Cancer Center, University of Vermont, College of Medicine, Burlington, Vermont
| |
Collapse
|
32
|
Polge C, Attaix D, Taillandier D. Role of E2-Ub-conjugating enzymes during skeletal muscle atrophy. Front Physiol 2015; 6:59. [PMID: 25805999 PMCID: PMC4354305 DOI: 10.3389/fphys.2015.00059] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/14/2015] [Indexed: 01/05/2023] Open
Abstract
The Ubiquitin Proteasome System (UPS) is a major actor of muscle wasting during various physio-pathological situations. In the past 15 years, increasing amounts of data have depicted a picture, although incomplete, of the mechanisms implicated in myofibrillar protein degradation, from the discovery of muscle-specific E3 ligases to the identification of the signaling pathways involved. The targeting specificity of the UPS relies on the capacity of the system to first recognize and then label the proteins to be degraded with a poly-ubiquitin (Ub) chain. It is fairly assumed that the recognition of the substrate is accomplished by the numerous E3 ligases present in mammalian cells. However, most E3s do not possess any catalytic activity and E2 enzymes may be more than simple Ub-providers for E3s since they are probably important actors in the ubiquitination machinery. Surprisingly, most authors have tried to characterize E3 substrates, but the exact role of E2s in muscle protein degradation is largely unknown. A very limited number of the 35 E2s described in humans have been studied in muscle protein breakdown experiments and the vast majority of studies were only descriptive. We review here the role of E2 enzymes in skeletal muscle and the difficulties linked to their study and provide future directions for the identification of muscle E2s responsible for the ubiquitination of contractile proteins.
Collapse
Affiliation(s)
- Cecile Polge
- UMR 1019 Nutrition Humaine, Institut National de la Recherche Agronomique Saint Genès Champanelle, France
| | - Didier Attaix
- UMR 1019 Nutrition Humaine, Institut National de la Recherche Agronomique Saint Genès Champanelle, France
| | - Daniel Taillandier
- UMR 1019 Nutrition Humaine, Institut National de la Recherche Agronomique Saint Genès Champanelle, France
| |
Collapse
|
33
|
Yuan L, Han J, Meng Q, Xi Q, Zhuang Q, Jiang Y, Han Y, Zhang B, Fang J, Wu G. Muscle-specific E3 ubiquitin ligases are involved in muscle atrophy of cancer cachexia: an in vitro and in vivo study. Oncol Rep 2015; 33:2261-8. [PMID: 25760630 DOI: 10.3892/or.2015.3845] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/09/2015] [Indexed: 11/06/2022] Open
Abstract
Muscle atrophy F-Box (MAFbx)/atrogin-1 and muscle ring-finger-1 (MuRF-1) have been identified as two muscle-specific E3 ubiquitin ligases that are highly expressed in skeletal muscle during muscle atrophy. However, the role of muscle-specific E3 ubiquitin ligases during the process of muscle atrophy of cancer cachexia remains largely unknown. In the present study, we analyzed the expression of atrogin-1 and MuRF-1 in the skeletal muscle of patients with malignant and benign disease. The possible mechanisms were studied both in a colon 26-induced cancer cachexia mouse model and in tumor necrosis factor-α (TNF-α) induced atrophy C2C12 cells. Our results demonstrated that atrogin-1 and MuRF-1 tended to be increased in the skeletal muscle of patients with malignant disease even before weight loss. Non-tumor body weights and gastrocnemius weights were significantly decreased while expression levels of ubiquitin proteasome pathway associated genes (atrogin-1, MuRF-1, ubiquitin and E2-14K) were upregulated in cancer cachexia mice. Significant myotube atrophy with atrogin-1 overexpression was observed in the C2C12 cells treated with TNF-α. Meanwhile, knockdown of atrogin-1 by small interfering RNA (siRNA) protected C2C12 cells from the adverse effect of TNF-α. In conclusion, muscle-specific E3 ubiquitin ligases were upregulated during cancer cachexia, and atrogin-1 may be a potential molecular target for treating muscle atrophy induced by cancer cachexia.
Collapse
Affiliation(s)
- Lei Yuan
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jun Han
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qingyang Meng
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qiulei Xi
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qiulin Zhuang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yi Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yusong Han
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Bo Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jing Fang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Guohao Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
34
|
Viana LR, Gomes-Marcondes MCC. A Leucine-Rich Diet Modulates the Tumor-Induced Down-Regulation of the MAPK/ERK and PI3K/Akt/mTOR Signaling Pathways and Maintains the Expression of the Ubiquitin-Proteasome Pathway in the Placental Tissue of NMRI Mice1. Biol Reprod 2015; 92:49. [DOI: 10.1095/biolreprod.114.123307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
35
|
How Do Skeletal Muscles Die? An Overview. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 861:99-111. [DOI: 10.1007/5584_2015_140] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Kazemzadeh-Narbat M, Annabi N, Tamayol A, Oklu R, Ghanem A, Khademhosseini A. Adenosine-associated delivery systems. J Drug Target 2015; 23:580-96. [PMID: 26453156 PMCID: PMC4863639 DOI: 10.3109/1061186x.2015.1058803] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Adenosine is a naturally occurring purine nucleoside in every cell. Many critical treatments such as modulating irregular heartbeat (arrhythmias), regulation of central nervous system (CNS) activity and inhibiting seizural episodes can be carried out using adenosine. Despite the significant potential therapeutic impact of adenosine and its derivatives, the severe side effects caused by their systemic administration have significantly limited their clinical use. In addition, due to adenosine's extremely short half-life in human blood (<10 s), there is an unmet need for sustained delivery systems to enhance efficacy and reduce side effects. In this article, various adenosine delivery techniques, including encapsulation into biodegradable polymers, cell-based delivery, implantable biomaterials and mechanical-based delivery systems, are critically reviewed and the existing challenges are highlighted.
Collapse
Affiliation(s)
- Mehdi Kazemzadeh-Narbat
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Nasim Annabi
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, MA, USA
- Department of Chemical Engineering, Northeastern University, Boston 02115, MA, USA
| | - Ali Tamayol
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
| | - Rahmi Oklu
- Massachusetts General Hospital, Harvard Medical School, Division of Interventional Radiology, Boston 02114, MA, USA
| | - Amyl Ghanem
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02139, MA, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge 02139, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, MA, USA
- Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| |
Collapse
|
37
|
Lindholm ME, Huss M, Solnestam BW, Kjellqvist S, Lundeberg J, Sundberg CJ. The human skeletal muscle transcriptome: sex differences, alternative splicing, and tissue homogeneity assessed with RNA sequencing. FASEB J 2014; 28:4571-81. [PMID: 25016029 DOI: 10.1096/fj.14-255000] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Human skeletal muscle health is important for quality of life and several chronic diseases, including type II diabetes, heart disease, and cancer. Skeletal muscle is a tissue widely used to study mechanisms behind different diseases and adaptive effects of controlled interventions. For such mechanistic studies, knowledge about the gene expression profiles in different states is essential. Since the baseline transcriptome has not been analyzed systematically, the purpose of this study was to provide a deep reference profile of female and male skeletal muscle. RNA sequencing data were analyzed from a large set of 45 resting human muscle biopsies. We provide extensive information on the skeletal muscle transcriptome, including 5 previously unannotated protein-coding transcripts. Global transcriptional tissue homogeneity was strikingly high, within both a specific muscle and the contralateral leg. We identified >23,000 known isoforms and found >5000 isoforms that differ between the sexes. The female and male transcriptome was enriched for genes associated with oxidative metabolism and protein catabolic processes, respectively. The data demonstrate remarkably high tissue homogeneity and provide a deep and extensive baseline reference for the human skeletal muscle transcriptome, with regard to alternative splicing, novel transcripts, and sex differences in functional ontology.transcriptome: sex differences, alternative splicing, and tissue homogeneity assessed with RNA sequencing.
Collapse
Affiliation(s)
- Malene E Lindholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden;
| | - Mikael Huss
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden; and
| | - Beata W Solnestam
- Science for Life Laboratory, School of Biotechnology, Royal Institute of Technology, Solna, Sweden
| | - Sanela Kjellqvist
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden; and
| | - Joakim Lundeberg
- Science for Life Laboratory, School of Biotechnology, Royal Institute of Technology, Solna, Sweden
| | - Carl J Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
38
|
Mirza KA, Pereira SL, Voss AC, Tisdale MJ. Comparison of the anticatabolic effects of leucine and Ca-β-hydroxy-β-methylbutyrate in experimental models of cancer cachexia. Nutrition 2014; 30:807-13. [DOI: 10.1016/j.nut.2013.11.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/15/2013] [Accepted: 11/21/2013] [Indexed: 11/16/2022]
|
39
|
Al-Zhoughbi W, Huang J, Paramasivan GS, Till H, Pichler M, Guertl-Lackner B, Hoefler G, Hoefler G. Tumor macroenvironment and metabolism. Semin Oncol 2014; 41:281-95. [PMID: 24787299 PMCID: PMC4012137 DOI: 10.1053/j.seminoncol.2014.02.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this review we introduce the concept of the tumor macroenvironment and explore it in the context of metabolism. Tumor cells interact with the tumor microenvironment including immune cells. Blood and lymph vessels are the critical components that deliver nutrients to the tumor and also connect the tumor to the macroenvironment. Several factors are then released from the tumor itself but potentially also from the tumor microenvironment, influencing the metabolism of distant tissues and organs. Amino acids, and distinct lipid and lipoprotein species can be essential for further tumor growth. The role of glucose in tumor metabolism has been studied extensively. Cancer-associated cachexia is the most important tumor-associated systemic syndrome and not only affects the quality of life of patients with various malignancies but is estimated to be the cause of death in 15%-20% of all cancer patients. On the other hand, systemic metabolic diseases such as obesity and diabetes are known to influence tumor development. Furthermore, the clinical implications of the tumor macroenvironment are explored in the context of the patient's outcome with special consideration for pediatric tumors. Finally, ways to target the tumor macroenvironment that will provide new approaches for therapeutic concepts are described.
Collapse
Affiliation(s)
- Wael Al-Zhoughbi
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jianfeng Huang
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | - Holger Till
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria,Address correspondence to Gerald Hoefler, MD, Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036 Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
40
|
D'Orlando C, Marzetti E, François S, Lorenzi M, Conti V, di Stasio E, Rosa F, Brunelli S, Doglietto GB, Pacelli F, Bossola M. Gastric cancer does not affect the expression of atrophy-related genes in human skeletal muscle. Muscle Nerve 2014; 49:528-33. [PMID: 23835743 DOI: 10.1002/mus.23945] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 11/09/2022]
Abstract
INTRODUCTION We evaluated the gene expression levels of atrogin-1, MuRF1, myostatin, follistatin, activin A, and inhibin alpha in skeletal muscle samples of patients with gastric cancer and controls. METHODS We studied 38 cancer patients and 12 controls who underwent surgery for gastric adenocarcinoma and benign abdominal diseases, respectively. A biopsy specimen was obtained from the rectus abdominis muscle from all participants. The relative gene expression of atrogin-1, MuRF1, myostatin, follistatin, activin A, and inhibin alpha was determined by quantitative real-time polymerase chain reaction analysis. RESULTS Atrogin-1 and MuRF1 mRNA expression was similar between cancer patients and controls and was unaffected by the disease stage or the severity of body weight loss. Transcript levels of myostatin and follistatin did not differ between cases and controls and were similar across disease stages and categories of weight loss. Finally, no differences were detected in activin A and inhibin alpha gene expression between cancer patients and controls. CONCLUSIONS In skeletal muscle, the gene expression of atrogin-1, MuRF1, myostatin, follistatin, activin A, and inhibin alpha is not affected by the presence of cancer. The expression of atrophy-related genes is unaffected by the disease stage and the degree of weight loss.
Collapse
|
41
|
Collins J, Noble S, Chester J, Coles B, Byrne A. The assessment and impact of sarcopenia in lung cancer: a systematic literature review. BMJ Open 2014; 4:e003697. [PMID: 24384894 PMCID: PMC3902311 DOI: 10.1136/bmjopen-2013-003697] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES There is growing awareness of the relationship between sarcopenia (loss of muscle mass and function), and outcomes in cancer, making it a potential target for future therapies. In order to inform future research and practice, we undertook a systematic review of factors associated with loss of muscle mass, and the relationship between muscle function and muscle mass in lung cancer, a common condition associated with poor outcomes. DESIGN We conducted a computerised systematic literature search on five databases. Studies were included if they explored muscle mass as an outcome measure in patients with lung cancer, and were published in English. SETTING Secondary care. PARTICIPANTS Patients with lung cancer. PRIMARY OUTCOME Factors associated with loss of muscle mass and muscle function, or sarcopenia, and the clinical impact thereof in patients with lung cancer. RESULTS We reviewed 5726 citations, and 35 articles were selected for analysis. Sarcopenia, as defined by reduced muscle mass alone, was found to be very prevalent in patients with lung cancer, regardless of body mass index, and where present was associated with poorer functional status and overall survival. There were diverse studies exploring molecular and metabolic factors in the development of loss of muscle mass; however, the precise mechanisms that contribute to sarcopenia and cachexia remain uncertain. The effect of nutritional supplements and ATP infusions on muscle mass showed conflicting results. There are very limited data on the correlation between degree of sarcopenia and muscle function, which has a non-linear relationship in older non-cancer populations. CONCLUSIONS Loss of muscle mass is a significant contributor to morbidity in patients with lung cancer. Loss of muscle mass and function may predate clinically overt cachexia, underlining the importance of evaluating sarcopenia, rather than weight loss alone. Understanding this relationship and its associated factors will provide opportunities for focused intervention to improve clinical outcomes.
Collapse
Affiliation(s)
| | - Simon Noble
- Department of Palliative Medicine, Cardiff University, Cardiff, South Wales, UK
| | - John Chester
- Department of Medical Oncology, Cardiff University, Cardiff, South Wales, UK
| | - Bernadette Coles
- Cancer Research Wales Library, Velindre NHS Trust, Cardiff, South Wales, UK
| | - Anthony Byrne
- Marie Curie Palliative Care Research Centre, Cardiff University, Cardiff, South Wales, UK
| |
Collapse
|
42
|
Tardif N, Klaude M, Lundell L, Thorell A, Rooyackers O. Autophagic-lysosomal pathway is the main proteolytic system modified in the skeletal muscle of esophageal cancer patients. Am J Clin Nutr 2013; 98:1485-92. [PMID: 24108784 DOI: 10.3945/ajcn.113.063859] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND In cancer cachexia, muscle depletion is related to morbidity and mortality. Muscle-wasting mechanisms in cancer patients are not fully understood. OBJECTIVE We investigated the involvement of the proteolytic systems (proteasome, autophagic-lysosomal, calpain, and caspase) in muscle wasting during cancer cachexia. DESIGN Esophageal cancer patients [n = 14; mean ± SD age: 64.1 ± 6.6 y] and weight-stable control patients undergoing reflux surgery (n = 8; age: 57.5 ± 5.8 y) were included. Enzymatic activities were measured in the vastus lateralis and diaphragm. Protein expressions were also measured in the vastus lateralis of control (n = 7) and cancer (n = 8) patients. RESULTS Proteasome, calpain, and caspase 3 activities in the vastus lateralis and diaphragm muscles did not differ between the 2 groups. Cathepsin B and L activities were 90% (± SD) [2.4 ± 0.2 compared with 1.3 ± 0.2 pmol 7-amido-4-methylcoumarin (AMC) · μg protein⁻¹ · min⁻¹; P < 0.001] and 115% (5.3 ± 0.4 compared with 2.5 ± 0.3 pmol AMC · μg protein⁻¹ · min⁻¹; P < 0.001) greater, respectively, in the vastus lateralis of cancer patients than in that of control subjects. We observed (in conjunction with increased lysosomal protease activities) higher microtubule-associated protein 1 light chain 3B-II/I ratios (0.14 ± 0.08 compared with 0.04 ± 0.04) and cathepsin B and L expressions in the vastus lateralis of cancer patients than in that of control subjects (P < 0.05). Protein expression of p62 in the vastus lateralis did not differ between the 2 groups. CONCLUSIONS The autophagic-lysosomal pathway in the skeletal muscle of cancer patients was modified, whereas other proteolytic systems were unchanged. These findings suggest involvement of the autophagic-lysosomal proteolytic system during cancer cachexia development in humans.
Collapse
Affiliation(s)
- Nicolas Tardif
- Departments of Anesthesiology and Intensive Care (NT, MK, and OR) and Surgery (LL), Gastrocentrum, Karolinska University Hospital Huddinge and Karolinska Institutet, Stockholm, Sweden, and the Department of Clinical Science, Danderyds Hospital & Department of Surgery, Ersta Hospital, Stockholm, Sweden (AT)
| | | | | | | | | |
Collapse
|
43
|
Leucine modulates the effect of Walker factor, a proteolysis-inducing factor-like protein from Walker tumours, on gene expression and cellular activity in C2C12 myotubes. Cytokine 2013; 64:343-50. [DOI: 10.1016/j.cyto.2013.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/15/2013] [Accepted: 05/17/2013] [Indexed: 01/26/2023]
|
44
|
Op den Kamp CM, Langen RC, Snepvangers FJ, de Theije CC, Schellekens JM, Laugs F, Dingemans AMC, Schols AM. Nuclear transcription factor κ B activation and protein turnover adaptations in skeletal muscle of patients with progressive stages of lung cancer cachexia. Am J Clin Nutr 2013; 98:738-48. [PMID: 23902785 DOI: 10.3945/ajcn.113.058388] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Experimental models of cancer cachexia have indicated that systemic inflammation induces muscle-protein breakdown and wasting via muscular nuclear transcription factor κB (NF-κB) activation. This process may limit the efficacy of nutritional intervention. OBJECTIVES We assessed muscle NF-κB activity and protein turnover signaling in progressive stages of clinical lung cancer cachexia and assessed whether circulating factors can induce muscular NF-κB activity. DESIGN Patients with lung cancer precachexia (n = 10) and cachexia (n = 16) were cross-sectionally compared with 22 healthy control subjects. mRNA transcripts of muscle proteolytic (ubiquitin proteasome system and autophagy lysosomal pathway) and myogenic markers and protein expression of PI3K/Akt, myostatin, and autophagy signaling were measured. A multiplex analysis showed the systemic inflammatory status, whereas plasma exposure to stable NF-κB-luciferase-reporter muscle cells revealed NF-κB inducibility. RESULTS Compared with healthy control subjects, cachectic patients had reduced (appendicular) muscle mass (-10%), muscle fiber atrophy (-27%), and decreased quadriceps strength (-31%). Subtle alterations in the muscle morphology were also detectable in precachectic patients, without changes in body composition. Despite increased Akt phosphorylation, downstream phosphosubstrates glycogen synthase kinase 3β, mammalian target of rapamycin, and Forkhead box protein were unaltered. The expression of autophagy effectors B cell lymphoma 2/adenovirus E1B 19-kDa protein-interacting protein 3 and microtubule-associated proteins 1A/1B light chain 3B gradually increased from precachectic to cachectic patients, without differences in E3 ubiquitin ligases. Systemic and local inflammation was evident in cachexia and intermediate in precachexia, but the plasma of both patients groups caused ex vivo muscle NF-κB activation. CONCLUSIONS In lung cancer, muscular NF-κB activity is induced by factors contained within the circulation. Autophagy may contribute to increased muscle proteolysis in lung cancer cachexia, whereas the absence of downstream changes in phosphosubstrates despite increased Akt phosphorylation suggests impaired anabolic signaling that may require targeted nutritional intervention.
Collapse
Affiliation(s)
- Céline M Op den Kamp
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre, Maastricht, Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Rosiglitazone and imidapril alone or in combination alleviate muscle and adipose depletion in a murine cancer cachexia model. Tumour Biol 2013; 35:323-32. [DOI: 10.1007/s13277-013-1043-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/18/2013] [Indexed: 01/09/2023] Open
|
46
|
Bonetto A, Penna F, Aversa Z, Mercantini P, Baccino FM, Costelli P, Ziparo V, Lucia S, Fanelli FR, Muscaritoli M. Early changes of muscle insulin-like growth factor-1 and myostatin gene expression in gastric cancer patients. Muscle Nerve 2013; 48:387-92. [DOI: 10.1002/mus.23798] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Andrea Bonetto
- Department of Experimental Medicine and Oncology; University of Turin; Turin; Italy
| | - Fabio Penna
- Department of Experimental Medicine and Oncology; University of Turin; Turin; Italy
| | - Zaira Aversa
- Department of Clinical Medicine, Sapienza; University of Rome; Rome; Italy
| | - Paolo Mercantini
- Department of Surgery, S. Andrea Hospital, Sapienza; University of Rome; Rome; Italy
| | - Francesco M. Baccino
- Department of Experimental Medicine and Oncology; University of Turin; Turin; Italy
| | - Paola Costelli
- Department of Experimental Medicine and Oncology; University of Turin; Turin; Italy
| | - Vincenzo Ziparo
- Department of Surgery, S. Andrea Hospital, Sapienza; University of Rome; Rome; Italy
| | - Simone Lucia
- Department of Clinical Medicine, Sapienza; University of Rome; Rome; Italy
| | | | | |
Collapse
|
47
|
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
|
48
|
Vaughan VC, Martin P, Lewandowski PA. Cancer cachexia: impact, mechanisms and emerging treatments. J Cachexia Sarcopenia Muscle 2013; 4:95-109. [PMID: 23097000 PMCID: PMC3684701 DOI: 10.1007/s13539-012-0087-1] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 09/03/2012] [Indexed: 12/18/2022] Open
Abstract
Many forms of cancer present with a complex metabolic profile characterised by loss of lean body mass known as cancer cachexia. The physical impact of cachexia contributes to decreased patient quality of life, treatment success and survival due to gross alterations in protein metabolism, increased oxidative stress and systemic inflammation. The psychological impact also contributes to decreased quality of life for both patients and their families. Combination therapies that target multiple pathways, such as eicosapentaenoic acid administered in combination with exercise, appetite stimulants, antioxidants or anti-inflammatories, have potential in the treatment of this complex syndrome and require further development.
Collapse
Affiliation(s)
- Vanessa C Vaughan
- School of Medicine, Deakin University, 75 Pigdons Road, Waurn Ponds, Victoria, 3216, Australia
| | | | | |
Collapse
|
49
|
Julienne CM, Dumas JF, Goupille C, Pinault M, Berri C, Collin A, Tesseraud S, Couet C, Servais S. Cancer cachexia is associated with a decrease in skeletal muscle mitochondrial oxidative capacities without alteration of ATP production efficiency. J Cachexia Sarcopenia Muscle 2012; 3:265-75. [PMID: 22648737 PMCID: PMC3505576 DOI: 10.1007/s13539-012-0071-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 04/30/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancer cachexia is a complex syndrome related to a negative energy balance resulting in muscle wasting. Implication of muscle mitochondrial bioenergetics alterations during cancer cachexia was suggested. Therefore, the aim of this study was to explore the efficiency of oxidative phosphorylation in skeletal muscle mitochondria in a preclinical model of cancer cachexia. METHODS Berlin-Druckrey IX rats with peritoneal carcinosis (PC) were used as a model of cancer cachexia with healthy pair-fed rats (PF) as control. Hindlimb muscle morphology and fibre type composition were analysed in parallel with ubiquitin ligases and UCP gene expression. Oxidative phosphorylation was investigated in isolated muscle mitochondria by measuring oxygen consumption and ATP synthesis rate. RESULTS PC rats underwent significant muscle wasting affecting fast glycolytic muscles due to a reduction in fibre cross-sectional area. MuRF1 and MAFbx gene expression were significantly increased (9- and 3.5-fold, respectively) in the muscle of PC compared to PF rats. Oxygen consumption in non-phosphorylating state and the ATP/O were similar in both groups. Muscle UCP2 gene was overexpressed in PC rats. State III and the uncoupled state were significantly lower in muscle mitochondria from PC rats with a parallel reduction in complex IV activity (-30 %). CONCLUSION This study demonstrated that there was neither alteration in ATP synthesis efficiency nor mitochondrial uncoupling in skeletal muscle of cachectic rats despite UCP2 gene overexpression. Muscle mitochondrial oxidative capacities were reduced due to a decrease in complex IV activity. This mitochondrial bioenergetics alteration could participate to insulin resistance, lipid droplet accumulation and lactate production.
Collapse
Affiliation(s)
- Cloé M Julienne
- INSERM U921, Nutrition, Croissance et Cancer, 37032, Tours, France
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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
|