1
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
2
|
Kingren MS, Hall JS, Ross TJ, Barre MC, Barlow A, Morales M, Treas LD, Maxson RT, Teo E, Porter C. HOUSING TEMPERATURE ALTERS BURN-INDUCED HYPERMETABOLISM IN MICE. Shock 2025; 63:118-131. [PMID: 39450911 DOI: 10.1097/shk.0000000000002476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
ABSTRACT Mice used in biomedical research are typically housed at ambient temperatures (22°C-24°C) below thermoneutrality (26°C-31°C). This chronic cold stress triggers a hypermetabolic response that may limit the utility of mice in modeling hypermetabolism in response to burns. To evaluate the effect of housing temperature on burn-induced hypermetabolism, mice were randomly assigned to receive sham, small, or large scald burns. Mice recovered for 21 days in metabolic phenotyping cages at 24°C or 30°C. Regardless of sex or sham/burn treatment, mice housed at 24°C had greater total energy expenditure ( P < 0.001), which was largely attributable to greater basal energy expenditure when compared to mice housed at 30°C ( P < 0.001). Thermoneutral housing (30°C) altered adipose tissue mass in a sex-dependent manner. Compared to sham and small burn groups, large burns resulted in greater water vapor loss, regardless of housing temperature ( P < 0.01). Compared to sham, large burns resulted in greater basal energy expenditure and total energy expenditure in mice housed at 24°C; however, this hypermetabolic response to large burns was blunted in female mice housed at 30°C, and absent in male mice housed at 30°C. Locomotion was significantly reduced in mice with large burns compared to sham and small burn groups, irrespective of sex or housing temperature ( P < 0.05). Housing at 30°C revealed sexual dimorphism in terms of the impact of burns on body mass and composition, where males with large burns displayed marked cachexia, whereas females did not. Collectively, this study demonstrates a sex-dependent role for housing temperature in influencing energetics and body composition in a rodent model of burn trauma.
Collapse
Affiliation(s)
| | | | | | | | - Abigail Barlow
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Martin Morales
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | | | | | | | | |
Collapse
|
3
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
4
|
Bieerkehazhi S, Barayan D, Khalaf F, de Brito Monteiro L, Aijaz A, Volk C, Matveev A, Saldanha S, Faour S, Jeschke MG. BURNS INDUCE ALTERATIONS IN THE ACYL PROTEOME OF MICE AND HUMANS. Shock 2024; 61:877-884. [PMID: 38661185 DOI: 10.1097/shk.0000000000002355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
ABSTRACT Hypermetabolic reprogramming triggered by thermal injury causes substantial morbidity and mortality. Despite the therapeutic potential of targeting this response, the underlying mechanisms remain poorly understood. Interestingly, protein S-acylation is a reversible posttranslational modification induced by metabolic alterations via DHHC acyltransferases. While this modification aids in the regulation of cellular functions, deregulated S-acylation contributes to various diseases by altering protein structure, stability, and localization. However, whether and how S-acylation may impact morbidity and mortality during postburn hypermetabolism is unknown. In this study, we discovered that alterations in the acyl proteome play a key role in mediating adverse outcomes that occur after burn injury. Using a murine model, we show that burn injury induces profound changes in the expression of various DHHC isoforms in metabolic organs central to regulating postburn hypermetabolism, the adipose tissue, and liver. This was accompanied by increased levels of S-acylated proteins in several pathways involved in mediating the adverse hypermetabolic response, including ER stress, lipolysis, and browning. In fact, similar results were also observed in adipose tissue from severely burned patients, as reflected by increased S-acylation of ERK1/2, eIF2a, ATGL, FGF21, and UCP1 relative to nonburn controls. Importantly, pharmacologically targeting this posttranslational modification using a nonselective DHHC inhibitor effectively attenuated burn-induced ER stress, lipolysis, and browning induction in an ex vivo explant model. Together, these findings suggest that S-acylation may facilitate the protein activation profile that drives burn-induced hypermetabolism and that targeting it could potentially be an effective strategy to restore metabolic function and improve outcomes after injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sara Faour
- David Braley Research Institute, Hamilton, Ontario, Canada
| | | |
Collapse
|
5
|
Knuth CM, Ricciuti Z, Barayan D, Rehou S, Abdullahi A, Monteiro LDB, Jeschke MG. Single-nuclei RNA Profiling Reveals Disruption of Adipokine and Inflammatory Signaling in Adipose Tissue of Burn Patients. Ann Surg 2023; 278:e1267-e1276. [PMID: 37057618 PMCID: PMC10928875 DOI: 10.1097/sla.0000000000005880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
OBJECTIVE We conducted a large-scale investigation of the systemic and adipose tissue-specific alterations in a clinical population of burn patients to identify factors that may influence hypermetabolism. BACKGROUND Previous research has identified chronic disturbances in adipose tissue inflammation, lipolysis, and browning, which may drive the perpetuation of hypermetabolism following the severe adrenergic stress of a burn injury. Given that adipose tissue is thought to be a central node in the regulation of systemic metabolism, we believe that systematically delineating the pathologic role of adipose tissue postburn, will lead to the identification of novel interventions to mitigate morbidity and mortality from severe burns. METHODS This was a single-institution cohort study, which obtained plasma and subcutaneous adipose tissue samples from severely burn adult patients over various time points during acute hospitalization. Whole-body clinical, metabolic, and inflammatory mediators were assessed in plasma, while genetic analyses through RT-qPCR and single-nuclei RNA sequencing were conducted in adipose tissue. RESULTS Systemic inflammation and adrenergic stress increase IL-6 signaling, lipolysis, browning, and adipokine dysfunction in the adipose tissue of adult burn patients, which may further propagate the long-term hypermetabolic response. Moreover, using single-nuclei RNA sequencing, we provide the first comprehensive characterization of alterations in the adipose tissue microenvironment occurring at acute and chronic stages postburn. CONCLUSION We provide novel insight toward the effect of burns on adipokine release, inflammatory signaling pathways, and adipose heterogeneity over the trajectory of acute and chronic stages.
Collapse
Affiliation(s)
- Carly M. Knuth
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| | | | - Dalia Barayan
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| | - Sarah Rehou
- Sunnybrook Research Institute, Toronto, Canada
- David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada
| | - Abdikarim Abdullahi
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
| | | | - Marc G. Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
- David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Canada
- Hamilton General Hospital, Hamilton Health Sciences, Hamilton, Canada
- Department of Surgery, McMaster University, Hamilton, Canada
| |
Collapse
|
6
|
Abstract
Rather than serving as a mere onlooker, adipose tissue is a complex endocrine organ and active participant in disease initiation and progression. Disruptions of biological processes operating within adipose can disturb healthy systemic physiology, the sequelae of which include metabolic disorders such as obesity and type 2 diabetes. A burgeoning interest in the field of adipose research has allowed for the elucidation of regulatory networks underlying both adipose tissue function and dysfunction. Despite this progress, few diseases are treated by targeting maladaptation in the adipose, an oft-overlooked organ. In this review, we elaborate on the distinct subtypes of adipocytes, their developmental origins and secretory roles, and the dynamic interplay at work within the tissue itself. Central to this discussion is the relationship between adipose and disease states, including obesity, cachexia, and infectious diseases, as we aim to leverage our wealth of knowledge for the development of novel and targeted therapeutics.
Collapse
Affiliation(s)
- Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; .,Howard Hughes Medical Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
7
|
Muacevic A, Adler JR, Torres R, Maita K, Garcia J, Serrano L, Ho O, Forte AJ. Modulation of Burn Hypermetabolism in Preclinical Models. Cureus 2023; 15:e33518. [PMID: 36779088 PMCID: PMC9904913 DOI: 10.7759/cureus.33518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2023] [Indexed: 01/11/2023] Open
Abstract
Severe burns elicit a state of physiological stress and increased metabolism to help the body compensate for the changes associated with the traumatic injury. However, this hypermetabolic state is associated with increased insulin resistance, cardiovascular dysfunction, skeletal muscle catabolism, impaired wound healing, and delayed recovery. Several interventions were attempted to modulate burn hypermetabolism, including nutritional support, early excision and grafting, and growth hormone application. However, burn hypermetabolism still imposes significant morbidity and mortality in burn patients. Due to the limitations of in vitro models, animal models are indispensable in burn research. Animal models provide researchers with invaluable tools to test the safety and efficacy of novel treatments or advance our knowledge of previously utilized agents. Several animal studies evaluated novel therapies to modulate burn hypermetabolism in the last few years, including recombinant human growth hormone, erythropoietin, acipimox, apelin, anti-interleukin-6 monoclonal antibody, and ghrelin therapies. Results from these studies are promising and may be effectively translated into human studies. In addition, other studies revisited drugs previously used in clinical practice, such as insulin and metformin, to further investigate their underlying mechanisms as modulators of burn hypermetabolism. This review aims to update burn experts with the novel therapies under investigation in burn hypermetabolism with a focus on applicability and translation. Furthermore, we aim to guide researchers in selecting the correct animal model for their experiments by providing a summary of the methodology and the rationale of the latest studies.
Collapse
|
8
|
Shen X, Li G, Wang L, Yu H, Zhou L, Deng H, Wang N, Lai C, Zhou W, Gao Y. Quantification of Acipimox in Plasma and Tissues by LC-MS/MS: Application to Pharmacokinetic Comparison between Normoxia and Hypoxia. Molecules 2022; 27:6413. [PMID: 36234950 PMCID: PMC9573116 DOI: 10.3390/molecules27196413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 12/02/2022] Open
Abstract
This study aimed to evaluate the pharmacokinetics of acipimox in rats under simulated high altitude hypoxia conditions. A sensitive and reliable LC-MS/MS method has been established for the quantitation of acipimox in rat plasma and tissue homogenate and validated according to the guidelines of the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA). Western blotting and enzyme linked immunosorbent assay (ELISA) were used to investigate the expression of lipid metabolism-related proteins and free fatty acid (FFA) levels, respectively. Cell viability was detected using a Cell Counting kit-8 assay (CCK-8). The method was then successfully applied in a pharmacokinetic comparison between normoxic and hypoxic rats. The results indicated that there were significant differences in the main pharmacokinetics parameters of acipimox between normoxic and hypoxic rats. HCAR2 expression in the hypoxia group was upregulated compared to that in the normoxia group and the levels of FFA decreased more in the hypoxia group. Under the hypoxia condition, the proliferation of HK2 cells was inhibited with increasing concentrations of acipimox. The results provide important and valuable information for the safety and efficacy of acipimox, which indicated that the dosage of acipimox might be adjusted appropriately during clinical medication in hypoxia.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Libin Wang
- School of Medicine, Shaanxi Energy Institute, Xianyang 712000, China
| | - Huijin Yu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huifang Deng
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chengcai Lai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
9
|
Angiotensin II Promotes White Adipose Tissue Browning and Lipolysis in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6022601. [PMID: 35799891 PMCID: PMC9253869 DOI: 10.1155/2022/6022601] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/27/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022]
Abstract
Emerging evidence has revealed that all components of the renin-angiotensin system (RAS) are present in adipose tissue. Angiotensin II (Ang II), the major bioactive component of the RAS, has been recognized as an adipokine involved in regulating energy homeostasis. However, the precise role of Ang II in white adipose tissue (WAT) remodeling remains to be elucidated. In this present study, C57BL/C male mice were continuously infused with different doses of Ang II (1.44 mg/kg/d or 2.5 mg/kg/d) or saline for 2 weeks and treated with or without the Ang II type 1 receptor blocker valsartan. H&E staining and immunohistochemistry were conducted to investigate the white-to-brown fat conversion. The level of serum total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) was measured. RNA sequencing was employed to explore the differentially expressed genes and their enriched pathways between control and Ang II groups. Our results showed that Ang II substantially resulted in loss of body weight and fat mass. Most importantly, Ang II treatment induced WAT browning in mice, which was partially attenuated by valsartan treatment. Furthermore, Ang II perturbed the serum lipid profiles. Ang II treatment elevated serum levels of TC, TG, LDL-C, and HDL-C in mice. Mechanistically, thermogenesis, cell respiration, and lipid metabolism-associated mRNAs showed significantly increased expression profiling in Ang II-treated WATs compared with control WATs. Moreover, we found that Ang II treatment enhanced AMPK phosphorylation in adipocytes. Therefore, Ang II promotes WAT browning and lipolysis via activating the AMPK signaling pathway.
Collapse
|
10
|
Knuth CM, Auger C, Chi L, Barayan D, Abdullahi A, Jeschke MG. Thermal Stress Induces Long-Term Remodeling of Adipose Tissue and Is Associated with Systemic Dysfunction. Shock 2021; 56:744-754. [PMID: 33534398 PMCID: PMC8316494 DOI: 10.1097/shk.0000000000001743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ABSTRACT Severe burns are characterized by the magnitude and duration of the hypermetabolic response thereafter, and demarcated by the loss of lean body mass and catabolism of fat stores. The aim of the present study was to delineate the temporal and location-specific physiological changes to adipose depots and downstream consequences post-burn in a murine model of thermal injury. C57BL/6 mice were subjected to a 30% total body surface area burn and body mass, food intake, and tissue mass were monitored for various time points up until 60 days postinjury. Mitochondrial respirometry was performed using a Seahorse XF96 analyzer. Lipolytic markers and browning markers were analyzed via Western blotting and histology. A severe burn results in a futile cycle of lipolysis and white adipose tissue (WAT) browning, the sequelae of which include fat catabolism, hepatomegaly, and loss of body mass despite increased food intake. A dynamic remodeling of epididymal WAT was observed with acute and chronic increases in lipolysis. Moreover, we demonstrate that pathological browning of inguinal WAT persists up to 60 days post-burn, highlighting the magnitude of the β-adrenergic response to thermal injury. Our data suggests that adipose depots have a heterogeneous response to burns and that therapeutic interventions targeting these physiological changes can improve outcomes. These data may also have implications for treating catabolic conditions such as cancer cachexia as well as developing treatments for obesity and type II diabetes.
Collapse
Affiliation(s)
- Carly M. Knuth
- Institute of Medical Science, University of Toronto, Canada
| | | | - Leon Chi
- Sunnybrook Research Institute, Toronto, Canada
| | - Dalia Barayan
- Institute of Medical Science, University of Toronto, Canada
| | | | - Marc G. Jeschke
- Institute of Medical Science, University of Toronto, Canada
- Department of Immunology, University of Toronto, Canada
- Sunnybrook Research Institute, Toronto, Canada
- Department of Surgery, Division of Plastic Surgery, University of Toronto, Canada
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
11
|
Kaur S, Auger C, Barayan D, Shah P, Matveev A, Knuth CM, Harris TE, Jeschke MG. Adipose-specific ATGL ablation reduces burn injury-induced metabolic derangements in mice. Clin Transl Med 2021; 11:e417. [PMID: 34185433 PMCID: PMC8181198 DOI: 10.1002/ctm2.417] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/19/2021] [Accepted: 04/25/2021] [Indexed: 12/25/2022] Open
Abstract
Hypermetabolism following severe burn injuries is associated with adipocyte dysfunction, elevated beige adipocyte formation, and increased energy expenditure. The resulting catabolism of adipose leads to detrimental sequelae such as fatty liver, increased risk of infections, sepsis, and even death. While the phenomenon of pathological white adipose tissue (WAT) browning is well-documented in cachexia and burn models, the molecular mechanisms are essentially unknown. Here, we report that adipose triglyceride lipase (ATGL) plays a central role in burn-induced WAT dysfunction and systemic outcomes. Targeting adipose-specific ATGL in a murine (AKO) model resulted in diminished browning, decreased circulating fatty acids, and mitigation of burn-induced hepatomegaly. To assess the clinical applicability of targeting ATGL, we demonstrate that the selective ATGL inhibitor atglistatin mimics the AKO results, suggesting a path forward for improving patient outcomes.
Collapse
Affiliation(s)
- Supreet Kaur
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Christopher Auger
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Dalia Barayan
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
- Institute of Medical SciencesUniversity of TorontoTorontoOntarioCanada
| | - Priyal Shah
- Institute of Medical SciencesUniversity of TorontoTorontoOntarioCanada
| | - Anna Matveev
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Carly M. Knuth
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
- Institute of Medical SciencesUniversity of TorontoTorontoOntarioCanada
| | - Thurl E. Harris
- Department of PharmacologyUniversity of Virginia School of MedicineCharlottesville VAUSA
| | - Marc G. Jeschke
- Ross Tilley Burn CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
- Institute of Medical SciencesUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
12
|
Asarian L. The Good, the Bad, and NLRP3-characterizing Novel Roles for the Inflammasome in Adipose-Tissue Biology and Burn Trauma. Endocrinology 2021; 162:6159687. [PMID: 33751110 PMCID: PMC8063676 DOI: 10.1210/endocr/bqab048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Lori Asarian
- University of Vermont, Department of Medicine, Burlington, VT 05405, USA
- Correspondence: Lori Asarian, Department of Medicine, University of Vermont, 89 Beaumont Avenue, Given C352, Burlington, VT 05405, USA. E-mail:
| |
Collapse
|
13
|
Barayan D, Abdullahi A, Vinaik R, Knuth CM, Auger C, Jeschke MG. Interleukin-6 blockade, a potential adjunct therapy for post-burn hypermetabolism. FASEB J 2021; 35:e21596. [PMID: 33871073 PMCID: PMC8982752 DOI: 10.1096/fj.202100388r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/04/2023]
Abstract
Severe burns remain a leading cause of death and disability worldwide. Despite advances in patient care, the excessive and uncontrolled hypermetabolic stress response induced by this trauma inevitably affects every organ system causing substantial morbidity and mortality. Recent evidence suggests interleukin-6 (IL-6) is a major culprit underlying post-burn hypermetabolism. Indeed, genetic deletion of IL-6 alleviates various complications associated with poor clinical outcomes including the adverse remodeling of adipose tissue, cachexia and hepatic steatosis. Thus, pharmacological blockade of IL-6 may be a more favorable treatment option to fully restore metabolic function after injury. To test this, we investigated the safety and effectiveness of blocking IL-6 for post-burn hypermetabolism using a validated anti-IL-6 monoclonal antibody (mAb) in our experimental murine model. Here, we show daily anti-IL-6 mAb administration protects against burn-induced weight loss (P < .0001) without any adverse effect on mortality. At the organ level, post-burn treatment with the IL-6 blocker suppressed the thermogenic activation of adipose tissue (P < .01) and its associated wasting (P < .05). The reduction of browning-induced lipolysis (P < .0001) indirectly decreased hepatic lipotoxicity (P < .01) which improved liver dysfunction (P < .05). Importantly, the beneficial effects of this anti-IL-6 agent extended to the skin, reflected by the decrease in excessive collagen deposition (P < .001) and genes involved in pathologic fibrosis and scarring (P < .05). Together, our results indicate that post-burn IL-6 blockade leads to significant improvements in systemic hypermetabolism by inhibiting pathological alterations in key immunometabolic organs. These findings support the therapeutic potential of anti-IL-6 interventions to improve care, quality of life, and survival in burned patients.
Collapse
Affiliation(s)
| | | | - Roohi Vinaik
- Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | - Marc G. Jeschke
- Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
14
|
Knuth CM, Auger C, Jeschke MG. Burn-induced hypermetabolism and skeletal muscle dysfunction. Am J Physiol Cell Physiol 2021; 321:C58-C71. [PMID: 33909503 DOI: 10.1152/ajpcell.00106.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Critical illnesses, including sepsis, cancer cachexia, and burn injury, invoke a milieu of systemic metabolic and inflammatory derangements that ultimately results in increased energy expenditure leading to fat and lean mass catabolism. Burn injuries present a unique clinical challenge given the magnitude and duration of the hypermetabolic response compared with other forms of critical illness, which drastically increase the risk of morbidity and mortality. Skeletal muscle metabolism is particularly altered as a consequence of burn-induced hypermetabolism, as it primarily provides a main source of fuel in support of wound healing. Interestingly, muscle catabolism is sustained long after the wound has healed, indicating that additional mechanisms beyond wound healing are involved. In this review, we discuss the distinctive pathophysiological response to burn injury with a focus on skeletal muscle function and metabolism. We first examine the diverse consequences on skeletal muscle dysfunction between thermal, electrical, and chemical burns. We then provide a comprehensive overview of the known mechanisms underlying skeletal muscle dysfunction that may be attributed to hypermetabolism. Finally, we review the most promising current treatment options to mitigate muscle catabolism, and by extension improve morbidity and mortality, and end with future directions that have the potential to significantly improve patient care.
Collapse
Affiliation(s)
- Carly M Knuth
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Auger
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Blears E, Ross E, Ogunbileje JO, Porter C, Murton AJ. The impact of catecholamines on skeletal muscle following massive burns: Friend or foe? Burns 2021; 47:756-764. [PMID: 33568281 DOI: 10.1016/j.burns.2021.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
Profound skeletal muscle wasting in the setting of total body hypermetabolism is a defining characteristic of massive burns, compromising the patient's recovery and necessitating a protracted period of rehabilitation. In recent years, the prolonged use of the non-selective beta-blocker, propranolol, has gained prominence as an effective tool to assist with suppressing epinephrine-dependent burn-induced hypermetabolism and by extension, blunting muscle catabolism. However, synthetic β-adrenergic agonists, such as clenbuterol, are widely associated with the promotion of muscle growth in both animals and humans. Moreover, experimental adrenodemedullation is known to result in muscle catabolism. Therefore, the blunting of muscle β-adrenergic signaling via the use of propranolol would be expected to negatively impair muscle protein homeostasis. This review explores these paradoxical observations and identifies the manner by which propranolol is thought to exert its anti-catabolic effects in burn patients. Moreover, we identify potential avenues by which the use of beta-blocker therapy in the treatment of massive burns could potentially be further refined to promote the recovery of muscle mass in these critically ill patients while continuing to ameliorate total body hypermetabolism.
Collapse
Affiliation(s)
- Elizabeth Blears
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Department of Surgery, Allegheny Health Network, Pittsburgh, PA, USA
| | - Evan Ross
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - John O Ogunbileje
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Craig Porter
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Andrew J Murton
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center of Aging, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
16
|
What's New in Shock, February 2020? Shock 2021; 53:133-136. [PMID: 31934961 DOI: 10.1097/shk.0000000000001483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Kaur S, Auger C, Jeschke MG. Adipose Tissue Metabolic Function and Dysfunction: Impact of Burn Injury. Front Cell Dev Biol 2020; 8:599576. [PMID: 33251224 PMCID: PMC7676399 DOI: 10.3389/fcell.2020.599576] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
For decades, adipose tissue had been considered as merely a storage depot and cushion to protect organs against trauma and injury. However, in recent years, a number of impactful studies have pinpointed the adipose tissue as an endocrine organ mediating systemic dysfunction in not only metabolic disorders such as obesity, but also in the stages following traumatic events such as severe burns. For instance, thermal injury induces a chronic β-adrenergic response associated with drastic increases in adipose lipolysis, macrophage infiltration and IL-6 mediated browning of white adipose tissue (WAT). The downstream consequences of these physiological changes to adipose, such as hepatomegaly and muscle wasting, are only now coming to light and suggest that WAT is both a culprit in and initiator of metabolic disorders after burn injury. To that effect, the aim of this review is to chronicle and critically analyze the scientific advances made in the study of adipose tissue with regards to its role in orchestrating the hypermetabolic response and detrimental effects of burn injury. The topics covered include the magnitude of the lipolytic response following thermal trauma and how WAT browning and inflammation perpetuate this cycle as well as how WAT physiology impacts insulin resistance and hyperglycemia post-burn. To conclude, we discuss how these findings can be translated from bench to bedside in the form of therapeutic interventions which target physiological changes to WAT to restore systemic homeostasis following a severe burn.
Collapse
Affiliation(s)
- Supreet Kaur
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Departments of Surgery and Immunology, University of Toronto, Toronto, ON, Canada
| | - Christopher Auger
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Departments of Surgery and Immunology, University of Toronto, Toronto, ON, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Departments of Surgery and Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|