Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Aging Dis ; 13(2): 540-551, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35371611

ABSTRACT

Impaired wound healing is a major issue in the elderly population and is associated with substantial health and economic burden, which is exponentially increasing with the growing aging population. While the underlying pathobiology of disturbed skin healing by aging is linked to several genetic and epigenetic factors, little is known about the cell-cell interaction during the wound healing process in aged individuals, particularly the mesenchymal stem cell (MSCs)-macrophages axis. In this study, by using a thermal injury animal model in which we compared the wound healing process of adult and young mice, we found that the insufficient pool of MSCs in adult animals are deficient in migrating to the wound bed and instead are restricted to the wound edge. We identified a deficiency of a CD90-positive MSC subpopulation in the wounds of adult animals, which is positively correlated with the number of F4/80+ macrophages. In vitro, we found that CD90+ cells preferentially adhere to the myeloid cells forming doublet cells. Thus, our findings highlight that in adult mice subjected to a thermal injury, impaired wound healing is likely mediated by a disturbed cellular interplay between myeloid cells and mesenchymal cells.

2.
Methods Cell Biol ; 168: 191-219, 2022.
Article in English | MEDLINE | ID: mdl-35366983

ABSTRACT

Burn injury results in a triad of inter-related adaptive responses: a systemic inflammatory response, a stress response, and a consequent hypermetabolic state which supports the former two. These pathological responses extend beyond the site of injury to affect distant organs and influence long-term outcomes in the patient. Animal models have proven valuable in advancing our understanding of mechanisms underlying the multifactorial manifestations of burn injury. While rodent models have been unprecedented in providing insights into signaling pathways, metabolic responses, protein turnover, cellular and molecular changes; small animal models do not replicate hypermetabolism, hyperinflammation, and wound healing after a burn injury as seen in humans. Herein, we provide a concise review of preferred large animal models utilized to understand burn pathophysiology based on organ systems and associated dysfunction. Additionally, we present a detailed protocol of contact burn injury in the Yorkshire pig model with a focus on preoperative care, anesthesia, analgesia, wound excision and grafting, dressing application, and frequency of dressing changes.


Subject(s)
Burns , Animals , Burns/metabolism , Burns/pathology , Burns/therapy , Humans , Models, Animal , Swine , Wound Healing/physiology
3.
Methods Cell Biol ; 168: 161-189, 2022.
Article in English | MEDLINE | ID: mdl-35366981

ABSTRACT

Burns are a severe form of trauma that account for 1.1 million cases necessitating medical attention and 4500 mortalities annually in the United States alone. Importantly, the initial trauma is succeeded by extensive, prolonged physiological alterations that detrimentally impact multiple organ systems. Given the complexity of post-burn pathophysiology, in vitro experiments are insufficient to model thermal injuries. Therefore, compatible animal burn models are essential for studying burn-related phenomena. In this chapter, we discuss commonly employed small animal burn models and their comparability and applicability to human studies. In particular, we compare post-burn wound healing between the species as well as relevant hypermetabolic and inflammatory characteristics, providing a better understanding of the pros and cons of utilizing a small animal surrogate for human burns. We further provide an overview of the rodent scald burn model methodology as well as a comparison between elderly, aged and young animals, providing a guide for tailoring animal model choice based on the relevant research question.


Subject(s)
Wound Healing , Animals , Disease Models, Animal , Wound Healing/physiology
4.
Immunology ; 165(2): 195-205, 2022 02.
Article in English | MEDLINE | ID: mdl-34773253

ABSTRACT

Although sepsis in burn patients is a major contributor to mortality, treatments are not always effective and underlying mechanisms have yet to be completely elucidated. NLRP3 inflammasome orchestrates burn-induced, inflammatory-driven pathophysiologic processes. Here, we determined the mechanism of NLRP3 inflammasome activation on bacterial clearance and mortality in burn sepsis. We obtained tissue and blood from 30 wild-type and 30 Nlrp3-/- mice. Mice were subjected to a two-hit model of 25-30% TBSA scald burn followed by Pseudomonas aeruginosa wound infection 72 hours after injury. We also obtained tissue from 34 adult burn patients (≥18 years of age) with early (0-11 days post-burn) and later (≥12 days post-burn) surgical time-points and ten healthy controls. Murine studies indicated that Nlrp3-/- had 30% improved survival and bacterial clearance at the site of injury and is systemically relative to burn sepsis wild type. Greater macrophage and neutrophil infiltration occurred acutely after infection (12 hours) to the site of injury and adipose tissue. This was followed by increased macrophage and neutrophil infiltration to lymphoid organs and liver beyond the acute phase (24 and 72 hours). Interestingly, Nlrp3 ablation increased acute systemic inflammation (IL-6, TNF-α, IL-1ß). Septic burn patients had persistently increased adipose NLRP3 by-product expression beyond the acute phase that was more pronounced in late-onset sepsis. Our findings suggest that Nlrp3 genetic ablation enhanced acute tissue-specific inflammatory responsiveness. Likely, this occurs by paradoxically increasing acute immune infiltration and inflammation with a non-persistent response. Clinically, persistent NLRP3-mediated inflammation occurs in septic versus normal burn patients and potentially detrimentally impacts patient outcomes.


Subject(s)
Burns/complications , Disease Susceptibility , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Sepsis/etiology , Sepsis/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Organ Specificity , Prognosis , Sepsis/mortality , Sepsis/therapy
6.
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
7.
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
8.
J Cell Mol Med ; 24(17): 9764-9773, 2020 09.
Article in English | MEDLINE | ID: mdl-32810382

ABSTRACT

The endoplasmic reticulum (ER) adapts to stress by activating a signalling cascade known as the ER stress response. While ER stress signalling is a central component of the cellular defence against environmental insult, persistent activation is thought to contribute to the progression of various metabolic complications via loss of protein function and cell death. Despite its importance however, whether and how ER stress impacts morbidity and mortality in conditions of hypermetabolism remain unclear. In this study, we discovered that chronic ER stress response plays a role in mediating adverse outcomes that occur after major trauma. Using a murine model of thermal injury, we show that induction of ER stress with Tunicamycin not only increased mortality but also resulted in hepatic damage and hepatic steatosis. Importantly, post-burn treatment with chaperone ER stress inhibitors attenuated hepatic ER stress and improved organ function following injury. Our study identifies ER stress as a potential hub of the signalling network affecting multiple aspects of metabolism after major trauma and as a novel potential molecular target to improve the clinical outcomes of severely burned patients.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Liver Diseases/genetics , Liver/metabolism , Wounds and Injuries/genetics , Animals , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Fatty Liver/metabolism , Fatty Liver/pathology , Humans , Liver/pathology , Liver Diseases/mortality , Liver Diseases/pathology , Mice , Signal Transduction/genetics , Wounds and Injuries/mortality , Wounds and Injuries/pathology
9.
Endocrinology ; 161(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32790834

ABSTRACT

Inflammasomes are multiprotein complexes that respond to pathogen or host associated damage markers, leading to caspase-1 maturation and processing of pro-inflammatory cytokines. Initially, inflammasomes were implicated primarily in inflammatory and infectious conditions. However, increasing evidence demonstrates broader roles beyond inflammation, including regulation of adipose tissue metabolism after burns. Here, we conducted a search for articles on PubMed, Web of Science, Embase, Scopus, and UpToDate with applied search strategies including a combination of "burns," "trauma," "(NLRP3) inflammasome," "metabolic conditions," "white adipose tissue," "macrophages," "browning," and "lipolysis" and included papers from 2000 to 2020. We discuss unexpected roles for NLRP3, the most characterized inflammasome to date, as a key metabolic driver in a variety of conditions. In particular, we highlight the function of NLRP3 inflammasome in burn trauma, which is characterized by both hyperinflammation and hypermetabolism. We identify a critical part for NLRP3 activation in macrophage dynamics and delineate a novel role in postburn white adipose tissue remodeling, a pathological response associated with hypermetabolism and poor clinical outcomes. Mechanistically, how inflammation and inflammasome activation is linked to postburn hypermetabolism is a novel concept to contemplate, and herein we provide evidence of an immunometabolic crosstalk between adipocytes and infiltrating macrophages.


Subject(s)
Burns/complications , Inflammasomes/physiology , Inflammation/etiology , Lipid Metabolism Disorders/etiology , Metabolism/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Burns/metabolism , Humans , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/metabolism , Lipid Metabolism Disorders/immunology , Lipid Metabolism Disorders/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
10.
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
11.
Shock ; 53(2): 137-145, 2020 02.
Article in English | MEDLINE | ID: mdl-31425403

ABSTRACT

Extensive burn injuries promote an increase in the lipolysis of white adipose tissue (WAT), a complication that enhances postburn hypermetabolism contributing to hyperlipidemia and hepatic steatosis. The systemic increase of free fatty acids (FFAs) due to burn-induced lipolysis and subsequent organ fatty infiltration may culminate in multiple organ dysfunction and, ultimately, death. Thus, reducing WAT lipolysis to diminish the mobilization of FFAs may render an effective means to improve outcomes postburn. Here, we investigated the metabolic effects of Acipimox, a clinically approved drug that suppresses lipolysis via inhibition of hormone-sensitive lipase (HSL). Using a murine model of thermal injury, we show that specific inhibition of HSL with Acipimox effectively suppresses burn-induced lipolysis in the inguinal WAT leading to lower levels of circulating FFAs at 7 days postburn (P < 0.05). The FFA substrate shortage indirectly repressed the thermogenic activation of adipose tissue after injury, reflected by the decrease in protein expression of key browning markers, UCP-1 (P < 0.001) and PGC-1α (P < 0.01). Importantly, reduction of FFA mobilization by Acipimox significantly decreased liver weight and intracellular fat accumulation (P < 0.05), suggesting that it may also improve organ function postburn. Our data validate the pharmacological inhibition of lipolysis as a potentially powerful therapeutic strategy to counteract the detrimental metabolic effects induced by burn.


Subject(s)
Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Lipolysis/drug effects , Liver/drug effects , Liver/metabolism , Pyrazines/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Blotting, Western , Burns/metabolism , Fatty Acids, Nonesterified/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Sterol Esterase/metabolism , Weight Loss/drug effects
12.
Transl Res ; 217: 47-60, 2020 03.
Article in English | MEDLINE | ID: mdl-31843468

ABSTRACT

Survival of burn patients is contingent on effective wound healing, a complex process that requires coordinated responses of myeloid cells and inflammatory pathways. NLRP3, which serves as a platform for secretion of proinflammatory cytokines, is implicated as a central regulator of wound healing. However, its role during the acute dermal and epidermal regeneration in the context of burns is unknown. Wild-type (WT) and NLRP3-/- mice were exposed to a 30% TBSA scald burn. Gene expression was conducted via real-time polymerase chain reaction. Trichrome staining was used to assess collagen deposition and granulation tissue formation. F4/80 immunostaining compared macrophage infiltration. Flow cytometric analysis was used to characterize skin macrophage distribution and profile. NLRP3, IL1ß and IL18 expression was upregulated in skin after burn, and these changes were nonexistent in NLRP3-/-. NLRP3-/- had decreased expression of proinflammatory cytokines, chemokines, inflammatory markers, and growth factors at 3 days (P < 0.05). NLRP3-/- burn skin demonstrated significantly less macrophage infiltration and higher expression of anti-inflammatory markers Arg1 and Fizz1 (P < 0.05) compared to WT. Trichrome staining showed decreased collagen deposition compared to WT. We show that NLRP3 is protective in burn wound healing, primarily through production of inflammatory mediators, macrophage recruitment, and polarization to a proinflammatory phenotype. Our findings highlight a central role of NLRP3 in wound healing through regulation of inflammation and macrophage polarization after burns.


Subject(s)
Burns/physiopathology , Inflammasomes/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Wound Healing/physiology , Animals , Cytokines/analysis , Female , Glyburide/pharmacology , Humans , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Wound Healing/drug effects
13.
Am J Physiol Endocrinol Metab ; 317(5): E751-E759, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31453709

ABSTRACT

A hallmark after burn is the stress and inflammatory-induced hypermetabolic response. Recently, we and others found that browning of white adipose tissue (WAT) is a critical component of this complex detrimental response. Although browning and inflammation have been independently delineated to occur after injury, their interaction is currently not well defined. One of the master regulators of inflammation and adipose tissue remodeling after burns is nucleotide-binding and oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) inflammasome. The aim of this this study was to determine whether NLRP3 modulates and activates WAT browning after burn. To obtain molecular and mechanistic insights, we used an NLRP3 knockout (NLRP3-/-) murine burn model. We demonstrated that genetic deletion of NLRP3 promoted persistent and augmented browning in adipocytes, evidenced by increased gene expression of peroxisome proliferator-activated receptor γ and CIDEA at 3 days (5.74 vs. 0.29, P < 0.05; 26.0 vs. 0.71, P < 0.05) and uncoupling protein 1 (UCP1) and PGC1α at 7 days (7,406 vs. 3,894, P < 0.05; 20.6 vs. 2.52, P < 0.01) and enhanced UCP1 staining and multilocularity. Additionally, the main regulator of postburn WAT browning, IL-6, was elevated in the plasma acutely after burn in NLRP3-/- compared with wild-type counterparts (478.9 vs. 67.1 pg/mL, P < 0.05 at 3 days). These results suggest that NLRP3 has antibrowning effects and that blocking NLRP3 increases thermogenesis and augments browning via increased levels of IL-6. Our findings provide insights into targeting innate inflammatory systems for regulation of adaptive thermogenesis, a critical response after burns and other hypermetabolic conditions.


Subject(s)
Adipose Tissue, White/physiopathology , Burns/physiopathology , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Adult , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Female , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , PPAR gamma/genetics , PPAR gamma/metabolism , Thermogenesis , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
14.
Expert Rev Anti Infect Ther ; 17(8): 607-619, 2019 08.
Article in English | MEDLINE | ID: mdl-31353976

ABSTRACT

Introduction: Despite modern advances, the primary cause of death after burns remains infection and sepsis. A key factor in determining outcomes is colonization with multi-drug resistant (MDR) organisms. Infections secondary to MDR organisms are challenging due to lack of adequate antibiotic treatment, subsequently prolonging hospital stay and increasing risk of adverse outcomes. Areas covered: This review highlights the most frequent organisms colonizing burn wounds as well as the most common MDR bacterial infections. Additionally, we discuss different treatment modalities and MDR infection prevention strategies as their appropriate management would minimize morbidity and mortality in this population. We conducted a search for articles on PubMed, Web of Science, Embase, Cochrane, Scopus and UpToDate with applied search strategies including a combination of: "burns, 'thermal injury,' 'infections,' 'sepsis,' 'drug resistance,' and 'antimicrobials.' Expert opinion: Management and prevention of MDR infections in burns is an ongoing challenge. We highlight the importance of preventative over therapeutic strategies, which are easy to implement and cost-effective. Additionally, targeted, limited use of antimicrobials can be beneficial in burn patients. A promising future area of investigation within this field is post-trauma microbiome profiling. Currently, the best treatment strategy for MDR in burn patients is prevention.


Subject(s)
Anti-Infective Agents/administration & dosage , Burns/complications , Wound Infection/epidemiology , Anti-Infective Agents/pharmacology , Bacterial Infections/drug therapy , Bacterial Infections/epidemiology , Bacterial Infections/microbiology , Drug Resistance, Microbial , Drug Resistance, Multiple, Bacterial , Humans , Sepsis/drug therapy , Sepsis/epidemiology , Sepsis/microbiology , Wound Infection/drug therapy , Wound Infection/microbiology
15.
Sci Rep ; 8(1): 15197, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30315247

ABSTRACT

Burns result in generalized catabolism, lipolysis, and hyperinflammation. NLRP3 inflammasome, a mediator of hyperinflammation, is upregulated in burn patients' adipose tissue within 7 days post-burn. However, its role during the acute phase is unknown. Here, wild-type (WT) and NLRP3 knockout (NLRP3-/-) mice were exposed to 25% TBSA scald burn. Flow cytometric analysis demonstrated greater liver macrophage infiltration in NLRP3-/- yet decreased protein expression of NLRP3 components, ER stress, and apoptosis. NLRP3-/- had increased circulating free fatty acids (FFA), fatty deposition and liver weight 1 hour post-burn. Alterations in adipose fatty acid synthase (Fasn) expression affects FFA levels post-burn; WT have an early peak in Fasn gene and protein expression that is lost in NLRP3-/-, resulting in increased lipolysis and hepatic fatty deposition. In summary, our findings reveal that NLRP3 inflammasome activation is a double-edged sword. While prolonged inflammation and long-term effects of macrophage activation are associated with poor outcomes, acute inflammation may be beneficial. These results highlight the important metabolic role that NLRP3 inflammasome plays in the acute phase, ultimately affecting survival post-burn.


Subject(s)
Burns/metabolism , Fatty Acid Synthases/metabolism , Inflammasomes/metabolism , Lipolysis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Animals , Apoptosis , Body Weight , Burns/complications , Burns/etiology , Burns/pathology , Chemotaxis , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Gene Expression , Lipolysis/genetics , Liver/metabolism , Liver/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Non-alcoholic Fatty Liver Disease/etiology , Organ Size
16.
Surg Infect (Larchmt) ; 19(2): 168-175, 2018.
Article in English | MEDLINE | ID: mdl-29327977

ABSTRACT

Burns are a common form of trauma that account for more than 300,000 deaths each year worldwide. Survival rates have improved over the past decades because of improvements in nutritional and fluid support, burn wound care, and infection control practices. Death, however, remains unacceptably high. The primary cause of death has changed over the last decades from anoxic causes to now predominantly infections and sepsis. Sepsis and septic complications are not only major contributors to poor outcomes, but they further result in longer hospital stay and higher healthcare costs. Despite the importance of infections and sepsis, the diagnosis and prediction remain a major challenge. To date, no clear diagnostic criteria or predictive formula exist that can predict reliably the occurrence of sepsis and infections. This review will highlight and discuss current definitions and criteria for diagnosis as well as predictive biomarkers of sepsis in patients with burns. It will also present the diagnostic tools employed, such as procalcitonin, C-reactive protein, and cytokines. We will discuss the benefits and shortcomings of different treatment modalities in the context of sepsis prevention. Last, we identify new therapeutic strategies for sepsis prediction and present future considerations to prevent sepsis in patients with burns. Minimizing and preventing septic complications through early detection would significantly benefit patients and necessitate continued research to unravel new biomarkers and mechanisms. Subsequent studies need to take a fresh perspective and consider the implementation of patient-centered therapeutic strategies.


Subject(s)
Biomarkers/analysis , Burns/complications , Decision Support Techniques , Disease Management , Sepsis/diagnosis , Sepsis/prevention & control , Humans
17.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 69(Pt 5): 499-502, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23695562

ABSTRACT

Attempts to crystallize several mammalian proteins overexpressed in Escherichia coli revealed a common contaminant, triosephosphate isomerase, a protein involved in glucose metabolism. Even with triosephosphate isomerase present in very small amounts, similarly shaped crystals appeared in the crystallization drops in a number of polyethylene glycol-containing conditions. All of the target proteins were His-tagged and their purification involved immobilized metal-affinity chromatography (IMAC), a step that was likely to lead to triosephosphate isomerase contamination. Analysis of the triosephosphate isomerase crystals led to the structure of E. coli triosephosphate isomerase at 1.85 Šresolution, which is a significant improvement over the previous structure.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Triose-Phosphate Isomerase/chemistry , Animals , Crystallization , Escherichia coli Proteins/biosynthesis , Humans , Mice , Protein Structure, Secondary , Solubility , Triose-Phosphate Isomerase/biosynthesis
18.
PLoS One ; 8(4): e62021, 2013.
Article in English | MEDLINE | ID: mdl-23614004

ABSTRACT

Protein disulfide isomerases comprise a large family of enzymes responsible for catalyzing the proper oxidation and folding of newly synthesized proteins in the endoplasmic reticulum (ER). Protein disulfide isomerase-related (PDIR) protein (also known as PDIA5) is a specialized member that participates in the folding of α1-antitrypsin and N-linked glycoproteins. Here, the crystal structure of the non-catalytic domain of PDIR was determined to 1.5 Å resolution. The structure adopts a thioredoxin-like fold stabilized by a structural disulfide bridge with a positively charged binding surface for interactions with the ER chaperones, calreticulin and ERp72. Crystal contacts between molecules potentially mimic the interactions of PDIR with misfolded substrate proteins. The results suggest that the non-catalytic domain of PDIR plays a key role in the recognition of protein partners and substrates.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Catalytic Domain/genetics , Catalytic Domain/physiology , Humans , Protein Binding , Protein Disulfide-Isomerases
SELECTION OF CITATIONS
SEARCH DETAIL
...