Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell Metab ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39111307

ABSTRACT

Choline is an essential nutrient for the biosynthesis of phospholipids, neurotransmitters, and one-carbon metabolism with a critical step being its import into mitochondria. However, the underlying mechanisms and biological significance remain poorly understood. Here, we report that SLC25A48, a previously uncharacterized mitochondrial inner-membrane carrier protein, controls mitochondrial choline transport and the synthesis of choline-derived methyl donors. We found that SLC25A48 was required for brown fat thermogenesis, mitochondrial respiration, and mitochondrial membrane integrity. Choline uptake into the mitochondrial matrix via SLC25A48 facilitated the synthesis of betaine and purine nucleotides, whereas loss of SLC25A48 resulted in increased production of mitochondrial reactive oxygen species and imbalanced mitochondrial lipids. Notably, human cells carrying a single nucleotide polymorphism on the SLC25A48 gene and cancer cells lacking SLC25A48 exhibited decreased mitochondrial choline import, increased oxidative stress, and impaired cell proliferation. Together, this study demonstrates that SLC25A48 regulates mitochondrial choline catabolism, bioenergetics, and cell survival.

2.
Cell ; 187(10): 2359-2374.e18, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38653240

ABSTRACT

Brown adipose tissue (BAT) is best known for thermogenesis. Rodent studies demonstrated that enhanced BAT thermogenesis is tightly associated with increased energy expenditure, reduced body weight, and improved glucose homeostasis. However, human BAT is protective against type 2 diabetes, independent of body weight. The mechanism underlying this dissociation remains unclear. Here, we report that impaired mitochondrial catabolism of branched-chain amino acids (BCAAs) in BAT, by deleting mitochondrial BCAA carriers (MBCs), caused systemic insulin resistance without affecting energy expenditure and body weight. Brown adipocytes catabolized BCAA in the mitochondria as nitrogen donors for the biosynthesis of non-essential amino acids and glutathione. Impaired mitochondrial BCAA-nitrogen flux in BAT resulted in increased oxidative stress, decreased hepatic insulin signaling, and decreased circulating BCAA-derived metabolites. A high-fat diet attenuated BCAA-nitrogen flux and metabolite synthesis in BAT, whereas cold-activated BAT enhanced the synthesis. This work uncovers a metabolite-mediated pathway through which BAT controls metabolic health beyond thermogenesis.


Subject(s)
Adipose Tissue, Brown , Amino Acids, Branched-Chain , Insulin Resistance , Mitochondria , Nitrogen , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Amino Acids, Branched-Chain/metabolism , Mice , Nitrogen/metabolism , Mitochondria/metabolism , Male , Humans , Energy Metabolism , Mice, Inbred C57BL , Oxidative Stress , Insulin/metabolism , Diet, High-Fat , Adipocytes, Brown/metabolism , Signal Transduction
3.
Cell Rep ; 43(1): 113584, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38117653

ABSTRACT

Severe burns induce a chronic hypermetabolic state that persists well past wound closure, indicating that additional internal mechanisms must be involved. Adipose tissue is suggested to be a central regulator in perpetuating hypermetabolism, although this has not been directly tested. Here, we show that thermogenic adipose tissues are activated in parallel to increases in hypermetabolism independent of cold stress. Using an adipose tissue transplantation model, we discover that burn-derived subcutaneous white adipose tissue alone is sufficient to invoke a hypermetabolic response in a healthy recipient mouse. Concomitantly, transplantation of healthy adipose tissue alleviates metabolic dysfunction in a burn recipient. We further show that the nicotinic acetylcholine receptor signaling pathway may mediate an immune-adipose crosstalk to regulate adipose tissue remodeling post-injury. Targeting this pathway could lead to innovative therapeutic interventions to counteract hypermetabolic pathologies.


Subject(s)
Burns , Subcutaneous Fat , Animals , Mice , Subcutaneous Fat/metabolism , Adipose Tissue, White/metabolism , Obesity/metabolism , Energy Metabolism/physiology , Burns/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue/metabolism
4.
Ann Surg ; 278(4): 519-529, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37389480

ABSTRACT

OBJECTIVE AND BACKGROUND: Propranolol, a nonselective beta-receptor blocker, improves outcomes of severely burned patients. While the clinical and physiological benefits of beta-blockade are well characterized, the underlying metabolic mechanisms are less well defined. We hypothesized that propranolol improves outcomes after burn injury by profoundly modulating metabolic pathways. METHODS: In this phase II randomized controlled trial, patients with burns ≥20% of total body surface area were randomly assigned to control or propranolol (dose given to decrease heart rate <100 bpm). Outcomes included clinical markers, inflammatory and lipidomic profiles, untargeted metabolomics, and molecular pathways. RESULTS: Fifty-two severely burned patients were enrolled in this trial (propranolol, n=23 and controls, n=29). There were no significant differences in demographics or injury severity between groups. Metabolomic pathway analyses of the adipose tissue showed that propranolol substantially alters several essential metabolic pathways involved in energy and nucleotide metabolism, as well as catecholamine degradation ( P <0.05). Lipidomic analysis revealed that propranolol-treated patients had lower levels of proinflammatory palmitic acid ( P <0.05) and saturated fatty acids ( P <0.05) with an increased ratio of polyunsaturated fatty acids ( P <0.05), thus shifting the lipidomic profile towards an anti-inflammatory phenotype after burn ( P <0.05). These metabolic effects were mediated by decreased activation of hormone-sensitive lipase at serine 660 ( P <0.05) and significantly reduced endoplasmic reticulum stress by decreasing phospho-JNK ( P <0.05). CONCLUSION: Propranolol's ability to mitigate pathophysiological changes to essential metabolic pathways results in significantly improved stress responses.


Subject(s)
Burns , Propranolol , Humans , Propranolol/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Metabolomics , Adipose Tissue
5.
Proc Natl Acad Sci U S A ; 120(9): e2216810120, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36812201

ABSTRACT

Mitochondria provide essential metabolites and adenosine triphosphate (ATP) for the regulation of energy homeostasis. For instance, liver mitochondria are a vital source of gluconeogenic precursors under a fasted state. However, the regulatory mechanisms at the level of mitochondrial membrane transport are not fully understood. Here, we report that a liver-specific mitochondrial inner-membrane carrier SLC25A47 is required for hepatic gluconeogenesis and energy homeostasis. Genome-wide association studies found significant associations between SLC25A47 and fasting glucose, HbA1c, and cholesterol levels in humans. In mice, we demonstrated that liver-specific depletion of SLC25A47 impaired hepatic gluconeogenesis selectively from lactate, while significantly enhancing whole-body energy expenditure and the hepatic expression of FGF21. These metabolic changes were not a consequence of general liver dysfunction because acute SLC25A47 depletion in adult mice was sufficient to enhance hepatic FGF21 production, pyruvate tolerance, and insulin tolerance independent of liver damage and mitochondrial dysfunction. Mechanistically, SLC25A47 depletion leads to impaired hepatic pyruvate flux and malate accumulation in the mitochondria, thereby restricting hepatic gluconeogenesis. Together, the present study identified a crucial node in the liver mitochondria that regulates fasting-induced gluconeogenesis and energy homeostasis.


Subject(s)
Genome-Wide Association Study , Gluconeogenesis , Humans , Mice , Animals , Gluconeogenesis/physiology , Glucose/metabolism , Liver/metabolism , Energy Metabolism/physiology , Pyruvates/metabolism
6.
Annu Rev Pathol ; 18: 71-93, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36070562

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.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/metabolism , Adipose Tissue/metabolism , Adipocytes/metabolism , Obesity/metabolism , Cachexia/metabolism
7.
Dev Cell ; 57(23): 2623-2637.e8, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36473459

ABSTRACT

De novo beige adipocyte biogenesis involves the proliferation of progenitor cells in white adipose tissue (WAT); however, what regulates this process remains unclear. Here, we report that in mouse models but also in human tissues, WAT lipolysis-derived linoleic acid triggers beige progenitor cell proliferation following cold acclimation, ß3-adrenoceptor activation, and burn injury. A subset of adipocyte progenitors, as marked by cell surface markers PDGFRα or Sca1 and CD81, harbored cristae-rich mitochondria and actively imported linoleic acid via a fatty acid transporter CD36. Linoleic acid not only was oxidized as fuel in the mitochondria but also was utilized for the synthesis of arachidonic acid-derived signaling entities such as prostaglandin D2. Oral supplementation of linoleic acid was sufficient to stimulate beige progenitor cell proliferation, even under thermoneutral conditions, in a CD36-dependent manner. Together, this study provides mechanistic insights into how diverse pathophysiological stimuli, such as cold and burn injury, promote de novo beige fat biogenesis.


Subject(s)
Adipose Tissue, Beige , Linoleic Acid , Humans , Animals , Mice , Linoleic Acid/pharmacology , Cell Proliferation
8.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34423787

ABSTRACT

BACKGROUNDThe incidence of burn injuries in older patients is dramatically increasing as the population of older people grows. Despite the increased demand for elderly burn care, the mechanisms that mediate increased morbidity and mortality in older trauma patients are unknown. We recently showed that a burn injury invokes white adipose tissue browning that leads to a substantially increased hypermetabolic response associated with poor outcomes. Therefore, the aim of this study was to determine the effect of age on the metabolic adipose response of browning after a burn injury.METHODOne hundred and seventy patients with burn injury admitted to the Ross Tilley Burn Centre were prospectively enrolled and grouped by age as older (≥50 years) and young (≤35 years). Adipose tissue and sera were collected and analyzed for browning markers and metabolic state via histology, gene expression, and resting energy expenditure assays.RESULTSWe found that older patients with burn injury lacked the adipose browning response, as they showed significant reductions in uncoupling protein 1 (UCP1) expression. This failure of the browning response was associated with reduced whole-body metabolism and decreased survival in older patients with burn injury. Mechanistically, we found that the adipose of both aged patients after burn trauma and aged mice after a burn showed impairments in macrophage infiltration and IL-6, key immunological regulators of the browning process after a severe trauma.CONCLUSIONTargeting pathways that activate the browning response represents a potential therapeutic approach to improve outcomes after burn trauma for elderly patients.FUNDINGNIH (R01-GM087285-01), Canadian Institutes of Health Research (grant no. 123336), and Canada Foundation for Innovation Leaders Opportunity Fund (no. 25407).


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Aging/metabolism , Burns/pathology , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Adult , Age Factors , Aged , Animals , Basal Metabolism , Burns/diagnosis , Burns/metabolism , Burns/mortality , Canada , Disease Models, Animal , Female , Humans , Injury Severity Score , Kaplan-Meier Estimate , Male , Mice , Middle Aged , Uncoupling Protein 1/analysis , Uncoupling Protein 1/metabolism
10.
Clin Transl Med ; 11(6): e417, 2021 06.
Article in English | MEDLINE | ID: mdl-34185433

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.


Subject(s)
Acyltransferases/physiology , Adipocytes, Beige/metabolism , Adipose Tissue, White/metabolism , Burns/complications , Energy Metabolism , Hepatomegaly/prevention & control , Lipolysis , Adipocytes, Beige/pathology , Adipose Tissue, White/pathology , Animals , Hepatomegaly/etiology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Am J Physiol Cell Physiol ; 321(1): C58-C71, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33909503

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.


Subject(s)
Cachexia/drug therapy , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/prevention & control , Protein Biosynthesis , Sepsis/metabolism , Burns/genetics , Burns/metabolism , Burns/pathology , Burns/rehabilitation , Cachexia/genetics , Cachexia/metabolism , Cachexia/pathology , Epigenesis, Genetic , Exercise , Human Growth Hormone/therapeutic use , Humans , Insulin/therapeutic use , Metformin/therapeutic use , Muscle Proteins/biosynthesis , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Oxandrolone/therapeutic use , Propranolol/therapeutic use , Proteolysis , Sepsis/microbiology , Sepsis/pathology , Sepsis/rehabilitation , Signal Transduction , Wound Healing/drug effects , Wound Healing/physiology
14.
FASEB J ; 35(5): e21596, 2021 05.
Article in English | MEDLINE | ID: mdl-33871073

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.


Subject(s)
Adipose Tissue/drug effects , Antibodies, Monoclonal/pharmacology , Burns/complications , Fibrosis/drug therapy , Interleukin-6/antagonists & inhibitors , Metabolic Diseases/drug therapy , Animals , Fibrosis/etiology , Fibrosis/pathology , Lipolysis , Male , Metabolic Diseases/etiology , Metabolic Diseases/pathology , Mice , Mice, Inbred C57BL
15.
Shock ; 56(5): 744-754, 2021 11 01.
Article in English | MEDLINE | ID: mdl-33534398

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 ß-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.


Subject(s)
Adipose Tissue/physiopathology , Burns/physiopathology , Animals , Injury Severity Score , Mice , Mice, Inbred C57BL
16.
Metabolism ; 118: 154733, 2021 05.
Article in English | MEDLINE | ID: mdl-33631145

ABSTRACT

It is well-established that mitochondria are the powerhouses of the cell, producing adenosine triphosphate (ATP), the universal energy currency. However, the most significant strengths of the electron transport chain (ETC), its intricacy and efficiency, are also its greatest downfalls. A reliance on metal complexes (FeS clusters, hemes), lipid moities such as cardiolipin, and cofactors including alpha-lipoic acid and quinones render oxidative phosphorylation vulnerable to environmental toxins, intracellular reactive oxygen species (ROS) and fluctuations in diet. To that effect, it is of interest to note that temporal disruptions in ETC activity in most organisms are rarely fatal, and often a redundant number of failsafes are in place to permit continued ATP production when needed. Here, we highlight the metabolic reconfigurations discovered in organisms ranging from parasitic Entamoeba to bacteria such as pseudomonads and then complex eukaryotic systems that allow these species to adapt to and occasionally thrive in harsh environments. The overarching aim of this review is to demonstrate the plasticity of metabolic networks and recognize that in times of duress, life finds a way.


Subject(s)
Mitochondria/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Citric Acid Cycle , Diphosphates/metabolism , Electron Transport , Glycolysis , Heat-Shock Proteins/metabolism , Humans , Microbiota , Oxidative Stress , Phosphorylation
17.
Stem Cell Res Ther ; 12(1): 137, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597003

ABSTRACT

INTRODUCTION: Burned human skin, which is routinely excised and discarded, contains viable mesenchymal stromal/stem cells (burn-derived mesenchymal stromal/stem cells; BD-MSCs). These cells show promising potential to enable and aid wound regeneration. However, little is known about their cell characteristics and biological function. OBJECTIVES: This study had two aims: first, to assess critical and cellular characteristics of BD-MSCs and, second, to compare those results with multipotent well-characterized MSCs from Wharton's jelly of human umbilical cords (umbilical cord mesenchymal stromal/stem cells, UC-MSCs). METHODS: BD- and UC-MSCs were compared using immunophenotyping, multi-lineage differentiation, seahorse analysis for glycolytic and mitochondrial function, immune surface markers, and cell secretion profile assays. RESULTS: When compared to UC-MSCs, BD-MSCs demonstrated a lower mesenchymal differentiation capacity and altered inflammatory cytokine secretomes at baseline and after stimulation with lipopolysaccharides. No significant differences were found in population doubling time, colony formation, cell proliferation cell cycle, production of reactive oxygen species, glycolytic and mitochondrial function, and in the expression of major histocompatibility complex I and II and toll-like receptor (TLR). IMPORTANCE, TRANSLATION: This study reveals valuable insights about MSCs obtained from burned skin and show comparable cellular characteristics with UC-MSCs, highlighting their potentials in cell therapy and skin regeneration.


Subject(s)
Burns , Mesenchymal Stem Cells , Wharton Jelly , Burns/therapy , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Umbilical Cord
18.
Stem Cell Res Ther ; 12(1): 82, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33494813

ABSTRACT

BACKGROUND: Multipotent mesenchymal stromal/stem cell (MSC) therapy is under investigation in promising (pre-)clinical trials for wound healing, which is crucial for survival; however, the optimal cell dosage remains unknown. The aim was to investigate the efficacy of different low-to-high MSC dosages incorporated in a biodegradable collagen-based dermal regeneration template (DRT) Integra®. METHODS: We conducted a porcine study (N = 8 Yorkshire pigs) and seeded between 200 and 2,000,000 cells/cm2 of umbilical cord mesenchymal stromal/stem cells on the DRT and grafted it onto full-thickness burn excised wounds. On day 28, comparisons were made between the different low-to-high cell dose groups, the acellular control, a burn wound, and healthy skin. RESULT: We found that the low dose range between 200 and 40,000 cells/cm2 regenerates the full-thickness burn excised wounds most efficaciously, followed by the middle dose range of 200,000-400,000 cells/cm2 and a high dose of 2,000,000 cells/cm2. The low dose of 40,000 cells/cm2 accelerated reepithelialization, reduced scarring, regenerated epidermal thickness superiorly, enhanced neovascularization, reduced fibrosis, and reduced type 1 and type 2 macrophages compared to other cell dosages and the acellular control. CONCLUSION: This regenerative cell therapy study using MSCs shows efficacy toward a low dose, which changes the paradigm that more cells lead to better wound healing outcome.


Subject(s)
Burns , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Skin , Swine , Umbilical Cord , Wound Healing
19.
Front Cell Dev Biol ; 8: 599576, 2020.
Article in English | MEDLINE | ID: mdl-33251224

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 ß-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.

20.
JCI Insight ; 5(17)2020 09 03.
Article in English | MEDLINE | ID: mdl-32750036

ABSTRACT

One of the most significant adverse postburn responses is abnormal scar formation, such as keloids. Despite its prolificacy, the underlying pathophysiology of keloid development is unknown. We recently demonstrated that NLRP3 inflammasome, the master regulator of inflammatory and metabolic responses (e.g., aerobic glycolysis), is essential for physiological wound healing. Therefore, burn patients who develop keloids may exhibit altered immunometabolic responses at the site of injury, which interferes with normal healing and portends keloid development. Here, we confirmed keloid NLRP3 activation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.05], IL-18 [P < 0.01]) and upregulation in Glut1 (P < 0.001) and glycolytic enzymes. Burn skin similarly displayed enhanced glycolysis and Glut1 expression (P < 0.01). However, Glut1 was significantly higher in keloid compared with nonkeloid burn patients (>2 SD above mean). Targeting aberrant glucose metabolism with shikonin, a pyruvate kinase M2 inhibitor, dampened NLRP3-mediated inflammation (cleaved caspase-1 [P < 0.05], IL-1ß [P < 0.01]) and improved healing in vivo. In summary, burn skin exhibited evidence of Warburg-like metabolism, similar to keloids. Targeting this altered metabolism could change the trajectory toward normal scarring, indicating the clinical possibility of shikonin for abnormal scar prevention.


Subject(s)
Burns/complications , Glycolysis , Inflammation/prevention & control , Keloid/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Skin/drug effects , Wound Healing , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Case-Control Studies , Female , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Inflammasomes , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators , Keloid/etiology , Keloid/metabolism , Keloid/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Naphthoquinones/pharmacology , Pyruvate Kinase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL