Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Front Immunol ; 12: 699563, 2021.
Article in English | MEDLINE | ID: mdl-34745089

ABSTRACT

The ability to remember a previous encounter with pathogens was long thought to be a key feature of the adaptive immune system enabling the host to mount a faster, more specific and more effective immune response upon the reencounter, reducing the severity of infectious diseases. Over the last 15 years, an increasing amount of evidence has accumulated showing that the innate immune system also has features of a memory. In contrast to the memory of adaptive immunity, innate immune memory is mediated by restructuration of the active chromatin landscape and imprinted by persisting adaptations of myelopoiesis. While originally described to occur in response to pathogen-associated molecular patterns, recent data indicate that host-derived damage-associated molecular patterns, i.e. alarmins, can also induce an innate immune memory. Potentially this is mediated by the same pattern recognition receptors and downstream signaling transduction pathways responsible for pathogen-associated innate immune training. Here, we summarize the available experimental data underlying innate immune memory in response to damage-associated molecular patterns. Further, we expound that trained immunity is a general component of innate immunity and outline several open questions for the rising field of pathogen-independent trained immunity.


Subject(s)
Alarmins/physiology , Immunity, Innate/immunology , Animals , Humans , Immunologic Memory , Pathogen-Associated Molecular Pattern Molecules
2.
Cancer Lett ; 523: 149-161, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34606928

ABSTRACT

Radiation is an integral part of cancer therapy. With the emergence of oncolytic vaccinia virus immunotherapy, it is important to study the combination of radiation and vaccinia virus in cancer therapy. In this study, we investigated the anti-tumor effect of and immune mechanisms underlying the combination of high-dose hypofractionated stereotactic body radiotherapy (SBRT) and oncolytic vaccinia virus in preclinical murine models. The combination enhanced the in vivo anti-tumor effect and increased the numbers of splenic CD4+Ki-67+ helper T lymphocytes and CD8+Ki-67+ cytotoxic T lymphocytes. Combinational therapy also increased tumor-infiltrating CD3+CD4+ helper T lymphocytes and CD3+CD8+ cytotoxic T lymphocytes, but decreased tumor-infiltrating regulatory T cells. In addition, SBRT combined with oncolytic vaccinia virus enhanced in vitro cell death, partly through necroptosis, and subsequent release of damage-associated molecular patterns (DAMPs), and shifted the macrophage M1/M2 ratio. We concluded that SBRT combined with oncolytic vaccinia virus can trigger tumor cell necroptosis and modify macrophages through the release of DAMPs, and then generate potent anti-tumor immunity and effects. Thus, combined therapy is potentially an important strategy for clinical cancer therapy.


Subject(s)
Alarmins/physiology , Necroptosis/physiology , Neoplasms, Experimental/therapy , Oncolytic Virotherapy , Radiosurgery , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Combined Modality Therapy , Female , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Tumor Microenvironment , Vaccinia virus
3.
Front Immunol ; 12: 705361, 2021.
Article in English | MEDLINE | ID: mdl-34489957

ABSTRACT

Immunogenic cell death (ICD) is a form of regulated cell death (RCD) induced by various stresses and produces antitumor immunity via damage-associated molecular patterns (DAMPs) release or exposure, mainly including high mobility group box 1 (HMGB1), calreticulin (CRT), adenosine triphosphate (ATP), and heat shock proteins (HSPs). Emerging evidence has suggested that ionizing radiation (IR) can induce ICD, and the dose, type, and fractionation of irradiation influence the induction of ICD. At present, IR-induced ICD is mainly verified in vitro in mice and there is few clinical evidence about it. To boost the induction of ICD by IR, some strategies have shown synergy with IR to enhance antitumor immune response, such as hyperthermia, nanoparticles, and chemotherapy. In this review, we focus on the molecular mechanisms of ICD, ICD-promoting factors associated with irradiation, the clinical evidence of ICD, and immunogenic forms of cell death. Finally, we summarize various methods of improving ICD induced by IR.


Subject(s)
Immunogenic Cell Death/radiation effects , Alarmins/physiology , Animals , Antigens, Neoplasm/immunology , Biomarkers , Combined Modality Therapy , Cytokines/physiology , Dose-Response Relationship, Radiation , Ferroptosis/radiation effects , HMGB1 Protein/physiology , Humans , Hyperthermia, Induced , Mice , Morpholines/therapeutic use , Necroptosis/radiation effects , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/radiotherapy , Piperazines/therapeutic use , Pyrroles/therapeutic use , Radiation Tolerance , Radiation, Ionizing
4.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34343135

ABSTRACT

Epithelial cells are charged with protection at barrier sites, but whether this normally beneficial response might sometimes become dysfunctional still needs definition. Here, we recognized a pattern of imbalance marked by basal epithelial cell growth and differentiation that replaced normal airspaces in a mouse model of progressive postviral lung disease due to the Sendai virus. Single-cell and lineage-tracing technologies identified a distinct subset of basal epithelial stem cells (basal ESCs) that extended into gas-exchange tissue to form long-term bronchiolar-alveolar remodeling regions. Moreover, this cell subset was selectively expanded by crossing a cell-growth and survival checkpoint linked to the nuclear-localized alarmin IL-33 that was independent of IL-33 receptor signaling and instead connected to autocrine chromatin accessibility. This mechanism creates an activated stem-progenitor cell lineage with potential for physiological or pathological function. Thus, conditional loss of Il33 gene function in basal epithelial cells disrupted the homeostasis of the epithelial barrier at skin and gut sites but also markedly attenuated postviral disease in the lung based on the downregulation of remodeling and inflammation. Thus, we define a basal ESC strategy to deploy innate immune machinery that appears to overshoot the primordial goal of self-defense. Our findings reveal new targets to stratify and correct chronic and often deadly postviral disease.


Subject(s)
Alarmins/physiology , Epithelial Cells/physiology , Interleukin-33/physiology , Lung Diseases/physiopathology , Respirovirus Infections/complications , Sendai virus , Stem Cells/physiology , Animals , Cell Differentiation , Interleukin-33/genetics , Mice , Single-Cell Analysis , Stem Cells/cytology
5.
Front Immunol ; 12: 711939, 2021.
Article in English | MEDLINE | ID: mdl-34305952

ABSTRACT

Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Recently was been found that pyroptosis is a unique form of proinflammatory programmed death, that is different from apoptosis. A growing number of studies have investigated pyroptosis and its relationship with sepsis, including the mechanisms, role, and relevant targets of pyroptosis in sepsis. While moderate pyroptosis in sepsis can control pathogen infection, excessive pyroptosis can lead to a dysregulated host immune response and even organ dysfunction. This review provides an overview of the mechanisms and potential therapeutic targets underlying pyroptosis in sepsis identified in recent decades, looking forward to the future direction of treatment for sepsis.


Subject(s)
Pyroptosis/physiology , Sepsis/immunology , Alarmins/physiology , Apoptosis/physiology , Caspases/metabolism , Cytokines/physiology , Disseminated Intravascular Coagulation , Drugs, Chinese Herbal/pharmacology , Heart/physiopathology , Humans , Lung/physiopathology , Pyroptosis/drug effects , Pyroptosis/immunology , Sepsis/blood , Sepsis/drug therapy , Sepsis/physiopathology
6.
J Invest Dermatol ; 141(11): 2602-2610.e3, 2021 11.
Article in English | MEDLINE | ID: mdl-33965403

ABSTRACT

The skin epidermis is attached to the underlying dermis by a laminin 332 (Lm332)-rich basement membrane. Consequently, loss of Lm332 leads to the severe blistering disorder epidermolysis bullosa junctionalis in humans and animals. Owing to the indispensable role of Lm332 in keratinocyte adhesion in vivo, the severity of the disease has limited research into other functions of the protein. We have conditionally disrupted Lm332 expression in basal keratinocytes of adult mice. Although blisters develop along the interfollicular epidermis, hair follicle basal cells provide sufficient anchorage of the epidermis to the dermis, making inducible deletion of the Lama3 gene compatible with life. Loss of Lm332 promoted the thickening of the epidermis and exaggerated desquamation. Global RNA expression analysis revealed major changes in the expression of keratins, cornified envelope proteins, and cellular stress markers. These modifications of the keratinocyte genetic program are accompanied by changes in cell shape and disorganization of the actin cytoskeleton. These data indicate that loss of Lm332-mediated progenitor cell adhesion alters cell fate and disturbs epidermal homeostasis.


Subject(s)
Cell Adhesion Molecules/physiology , Homeostasis/physiology , Keratinocytes/cytology , Actin Cytoskeleton/physiology , Alarmins/physiology , Animals , Blister/etiology , Cell Differentiation , Epidermis/pathology , Keratins/analysis , Mice , Receptor, Fibroblast Growth Factor, Type 1/analysis , Kalinin
7.
Transl Res ; 232: 37-48, 2021 06.
Article in English | MEDLINE | ID: mdl-33358868

ABSTRACT

Approximately 15%-20% of patients infected with SARS-CoV-2 coronavirus (COVID-19) progress beyond mild and self-limited disease to require supplemental oxygen for severe pneumonia; 5% of COVID-19-infected patients further develop acute respiratory distress syndrome (ARDS) and multiorgan failure. Despite mortality rates surpassing 40%, key insights into COVID-19-induced ARDS pathology have not been fully elucidated and multiple unmet needs remain. This review focuses on the unmet need for effective therapies that target unchecked innate immunity-driven inflammation which drives unchecked vascular permeability, multiorgan dysfunction and ARDS mortality. Additional unmet needs including the lack of insights into factors predicting pathogenic hyperinflammatory viral host responses, limited approaches to address the vast disease heterogeneity in ARDS, and the absence of clinically-useful ARDS biomarkers. We review unmet needs persisting in COVID-19-induced ARDS in the context of the potential role for damage-associated molecular pattern proteins in lung and systemic hyperinflammatory host responses to SARS-CoV-2 infection that ultimately drive multiorgan dysfunction and ARDS mortality. Insights into promising stratification-enhancing, biomarker-based strategies in COVID-19 and non-COVID ARDS may enable the design of successful clinical trials of promising therapies.


Subject(s)
Alarmins/physiology , COVID-19/complications , Inflammation/etiology , Respiratory Distress Syndrome/etiology , SARS-CoV-2 , Vascular System Injuries/etiology , Blood Coagulation Disorders/etiology , Capillary Permeability , Cytokines/physiology , Humans , Nicotinamide Phosphoribosyltransferase/physiology , SARS-CoV-2/pathogenicity
9.
J Hematol Oncol ; 13(1): 110, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32778143

ABSTRACT

In recent years, cancer immunotherapy based on immune checkpoint inhibitors (ICIs) has achieved considerable success in the clinic. However, ICIs are significantly limited by the fact that only one third of patients with most types of cancer respond to these agents. The induction of cell death mechanisms other than apoptosis has gradually emerged as a new cancer treatment strategy because most tumors harbor innate resistance to apoptosis. However, to date, the possibility of combining these two modalities has not been discussed systematically. Recently, a few studies revealed crosstalk between distinct cell death mechanisms and antitumor immunity. The induction of pyroptosis, ferroptosis, and necroptosis combined with ICIs showed synergistically enhanced antitumor activity, even in ICI-resistant tumors. Immunotherapy-activated CD8+ T cells are traditionally believed to induce tumor cell death via the following two main pathways: (i) perforin-granzyme and (ii) Fas-FasL. However, recent studies identified a new mechanism by which CD8+ T cells suppress tumor growth by inducing ferroptosis and pyroptosis, which provoked a review of the relationship between tumor cell death mechanisms and immune system activation. Hence, in this review, we summarize knowledge of the reciprocal interaction between antitumor immunity and distinct cell death mechanisms, particularly necroptosis, ferroptosis, and pyroptosis, which are the three potentially novel mechanisms of immunogenic cell death. Because most evidence is derived from studies using animal and cell models, we also reviewed related bioinformatics data available for human tissues in public databases, which partially confirmed the presence of interactions between tumor cell death and the activation of antitumor immunity.


Subject(s)
Ferroptosis/immunology , Immune Checkpoint Inhibitors/pharmacology , Necroptosis/immunology , Neoplasms/immunology , Pyroptosis/immunology , Adaptive Immunity , Alarmins/physiology , Animals , Antigens, Neoplasm/immunology , Autophagy , Ferroptosis/drug effects , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lymphocyte Subsets/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Models, Immunological , NF-kappa B/metabolism , Necroptosis/drug effects , Neoplasm Proteins/physiology , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinases/physiology , Pyroptosis/drug effects
10.
Mol Hum Reprod ; 26(9): 712-726, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32647859

ABSTRACT

Sterile intra-amniotic inflammation is a clinical condition frequently observed in women with preterm labor and birth, the leading cause of neonatal morbidity and mortality worldwide. Growing evidence suggests that alarmins found in amniotic fluid, such as interleukin (IL)-1α, are central initiators of sterile intra-amniotic inflammation. However, the causal link between elevated intra-amniotic concentrations of IL-1α and preterm birth has yet to be established. Herein, using an animal model of ultrasound-guided intra-amniotic injection of IL-1α, we show that elevated concentrations of IL-1α cause preterm birth and neonatal mortality. Additionally, using immunoblotting techniques and a specific immunoassay, we report that the intra-amniotic administration of IL-1α induces activation of the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in the fetal membranes, but not in the decidua, as evidenced by a concomitant increase in the protein levels of NLRP3, active caspase-1, and IL-1ß. Lastly, using Nlrp3-/- mice, we demonstrate that the deficiency of this inflammasome sensor molecule reduces the rates of preterm birth and neonatal mortality caused by the intra-amniotic injection of IL-1α. Collectively, these results demonstrate a causal link between elevated IL-1α concentrations in the amniotic cavity and preterm birth as well as adverse neonatal outcomes, a pathological process that is mediated by the NLRP3 inflammasome. These findings shed light on the mechanisms underlying sterile intra-amniotic inflammation and provide further evidence that this clinical condition can potentially be treated by targeting the NLRP3 inflammasome.


Subject(s)
Inflammasomes/physiology , Interleukin-1alpha/physiology , Premature Birth/metabolism , Alarmins/physiology , Amniotic Fluid/drug effects , Amniotic Fluid/metabolism , Animals , Animals, Newborn , Female , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1alpha/administration & dosage , Interleukin-1alpha/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pregnancy , Premature Birth/chemically induced , Premature Birth/genetics
11.
Front Immunol ; 11: 688, 2020.
Article in English | MEDLINE | ID: mdl-32425933

ABSTRACT

The restricted capacity of newborn infants to mount inflammatory responses toward microbial challenges has traditionally been linked to the high risk of septic diseases during the neonatal period. In recent years, substantial evidence has been provided that this characteristic of the neonatal immune system is actually a meaningful physiologic state that is based on specific transiently active cellular and molecular mechanisms and required for a favorable course of postnatal immune adaptation. The identification of physiologically high amounts of S100-alarmins in neonates has been one of the crucial pieces in the puzzle that contributed to the change of concept. In this context, innate immune immaturity could be redefined and assigned to the epigenetic silence of adult-like cell-autonomous regulation at the beginning of life. S100-alarmins represent an alternative age-specific mechanism of immune regulation that protects neonates from hyperinflammatory immune responses. Here, we summarize how infants are provided with S100-alarmins and why these allow an uneventful clash between the innate immune system and the extrauterine world. The mode of action of S100-alarmins is highlighted including their tuning functions at multiple levels for establishing a state of homeostasis with the environment in the newborn individual.


Subject(s)
Alarmins/physiology , Calgranulin A/physiology , Calgranulin B/physiology , Immunity, Innate , Infant, Newborn/immunology , Adaptation, Physiological , Humans , Signal Transduction/physiology
12.
Mediators Inflamm ; 2020: 5919150, 2020.
Article in English | MEDLINE | ID: mdl-32377162

ABSTRACT

TNIP1 protein is a widely expressed, cytoplasmic inhibitor of inflammatory signaling initiated by membrane receptors such as TLRs which recognize pathogen-associated and damage-associated molecular patterns (PAMPs and DAMPs). Keratinocyte TNIP1 deficiency sensitizes cells to PAMPs and DAMPs promoting hyperresponsive expression and secretion of cytokine markers (e.g., IL-8 and IL-6) relevant to cases of chronic inflammation, like psoriasis, where TNIP1 deficiency has been reported. Here, we examined the impact of TNIP1 deficiency on gene expression and cellular responses (migration and viability) relevant to acute inflammation as typically occurs in wound healing. Using siRNA-mediated TNIP1 expression knockdown in cultured HaCaT keratinocytes, we investigated TNIP1 deficiency effects on signaling downstream of TLR3 agonism with low-concentration poly (I:C), a representative PAMP/DAMP. The combination of TNIP1 knockdown and PAMP/DAMP signaling disrupted expression of specific keratinocyte differentiation markers (e.g., transglutaminase 1 and involucrin). These same conditions promoted synergistically increased expression of wound-associated markers (e.g., S100A8, TGFß, and CCN2) suggesting potential benefit of increased inflammatory response from reduced TNIP1 protein. Unexpectedly, poly (I:C) challenge of TNIP1-deficient cells restricted reepithelialization and reduced cell viability. In these cells, there was not only increased expression for genes associated with inflammasome assembly (e.g., ASC, procaspase 1) but also for A20, a TNIP1 partner protein that represses cell-death signaling. Despite this possibly compensatory increase in A20 mRNA, there was a decrease in phospho-A20 protein, the form necessary for quenching inflammation. Hyperresponsiveness to poly (I:C) in TNIP1-deficient keratinocytes was in part mediated through p38 and JNK pathways. Taken together, we conclude that TNIP1 deficiency promotes enhanced expression of factors associated with promoting wound healing. However, the coupled, increased potential priming of the inflammasome and reduced compensatory activity of A20 has a net negative effect on overall cell recovery potential manifested by poor reepithelialization and viability. These findings suggest a previously unrecognized role for TNIP1 protein in limiting inflammation during successful progression through early wound healing stages.


Subject(s)
DNA-Binding Proteins/physiology , Epithelium/physiology , Inflammasomes/physiology , Keratinocytes/physiology , Wound Healing/physiology , Alarmins/physiology , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/deficiency , Gene Expression , Humans , Keratinocytes/cytology , Pathogen-Associated Molecular Pattern Molecules , Poly I-C/pharmacology , Signal Transduction/physiology , Toll-Like Receptor 3/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
13.
J Zhejiang Univ Sci B ; 21(3): 192-203, 2020.
Article in English | MEDLINE | ID: mdl-32133797

ABSTRACT

Diabetic nephropathy (DN) is currently the most common complication of diabetes. It is considered to be one of the leading causes of end-stage renal disease (ESRD) and affects many diabetic patients. The pathogenesis of DN is extremely complex and has not yet been clarified; however, in recent years, increasing evidence has shown the important role of innate immunity in DN pathogenesis. Pattern recognition receptors (PRRs) are important components of the innate immune system and have a significant impact on the occurrence and development of DN. In this review, we classify PRRs into secretory, endocytic, and signal transduction PRRs according to the relationship between the PRRs and subcellular compartments. PRRs can recognize related pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), thus triggering a series of inflammatory responses, promoting renal fibrosis, and finally causing renal impairment. In this review, we describe the proposed role of each type of PRRs in the development and progression of DN.


Subject(s)
Diabetic Nephropathies/etiology , Receptors, Pattern Recognition/physiology , Alarmins/physiology , C-Reactive Protein/analysis , C-Reactive Protein/physiology , Endocytosis , Humans , Immunity, Innate , Mannose-Binding Lectin/physiology , Pathogen-Associated Molecular Pattern Molecules , Serum Amyloid P-Component/physiology , Signal Transduction
15.
Annu Rev Pathol ; 15: 493-518, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31675482

ABSTRACT

Recognizing the importance of leukocyte trafficking in inflammation led to some therapeutic breakthroughs. However, many inflammatory pathologies remain without specific therapy. This review discusses leukocytes in the context of sterile inflammation, a process caused by sterile (non-microbial) molecules, comprising damage-associated molecular patterns (DAMPs). DAMPs bind specific receptors to activate inflammation and start a highly optimized sequence of immune cell recruitment of neutrophils and monocytes to initiate effective tissue repair. When DAMPs are cleared, the recruited leukocytes change from a proinflammatory to a reparative program, a switch that is locally supervised by invariant natural killer T cells. In addition, neutrophils exit the inflammatory site and reverse transmigrate back to the bloodstream. Inflammation persists when the program switch or reverse transmigration fails, or when the coordinated leukocyte effort cannot clear the immunostimulatory molecules. The latter causes inappropriate leukocyte activation, a driver of many pathologies associated with poor lifestyle choices. We discuss lifestyle-associated inflammatory diseases and their corresponding immunostimulatory lifestyle-associated molecular patterns (LAMPs) and distinguish them from DAMPs.


Subject(s)
Alarmins/physiology , Environmental Biomarkers/physiology , Inflammation/etiology , Life Style , Pathogen-Associated Molecular Pattern Molecules , Animals , Biological Factors/physiology , Humans , Immunity/physiology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Neutrophils/physiology , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism
16.
Article in English | WPRIM (Western Pacific) | ID: wpr-1010527

ABSTRACT

Diabetic nephropathy (DN) is currently the most common complication of diabetes. It is considered to be one of the leading causes of end-stage renal disease (ESRD) and affects many diabetic patients. The pathogenesis of DN is extremely complex and has not yet been clarified; however, in recent years, increasing evidence has shown the important role of innate immunity in DN pathogenesis. Pattern recognition receptors (PRRs) are important components of the innate immune system and have a significant impact on the occurrence and development of DN. In this review, we classify PRRs into secretory, endocytic, and signal transduction PRRs according to the relationship between the PRRs and subcellular compartments. PRRs can recognize related pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), thus triggering a series of inflammatory responses, promoting renal fibrosis, and finally causing renal impairment. In this review, we describe the proposed role of each type of PRRs in the development and progression of DN.


Subject(s)
Humans , Alarmins/physiology , C-Reactive Protein/physiology , Diabetic Nephropathies/etiology , Endocytosis , Immunity, Innate , Mannose-Binding Lectin/physiology , Pathogen-Associated Molecular Pattern Molecules , Receptors, Pattern Recognition/physiology , Serum Amyloid P-Component/physiology , Signal Transduction
17.
JCI Insight ; 4(16)2019 08 22.
Article in English | MEDLINE | ID: mdl-31434802

ABSTRACT

Multiple organ failure (MOF) is the leading cause of late mortality and morbidity in patients who are admitted to intensive care units (ICUs). However, there is an epidemiologic discrepancy in the mechanism of underlying immunologic derangement dependent on etiology between sepsis and trauma patients in MOF. We hypothesized that damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), while both involved in the development of MOF, contribute differently to the systemic innate immune derangement and coagulopathic changes. We found that DAMPs not only produce weaker innate immune activation than counterpart PAMPs, but also induce less TLR signal desensitization, contribute to less innate immune cell death, and propagate more robust systemic coagulopathic effects than PAMPs. This differential contribution to MOF provides further insight into the contributing factors to late mortality in critically ill trauma and sepsis patients. These findings will help to better prognosticate patients at risk of MOF and may provide future therapeutic molecular targets in this disease process.


Subject(s)
Alarmins/physiology , Critical Illness/mortality , Multiple Organ Failure/mortality , Pathogen-Associated Molecular Pattern Molecules/metabolism , Adult , Animals , Bacteria/pathogenicity , Blood Coagulation , Cells, Cultured , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Multiple Organ Failure/etiology , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Necrosis , Rats, Sprague-Dawley , Sepsis/immunology , Sepsis/metabolism , Sepsis/mortality , Wounds and Injuries/immunology , Wounds and Injuries/metabolism , Wounds and Injuries/mortality
18.
J Leukoc Biol ; 106(1): 161-169, 2019 07.
Article in English | MEDLINE | ID: mdl-30946496

ABSTRACT

High mobility group box 1 (HMGB1) is a multifunctional nuclear protein, probably known best as a prototypical alarmin or damage-associated molecular pattern (DAMP) molecule when released from cells. However, HMGB1 has multiple functions that depend on its location in the nucleus, in the cytosol, or extracellularly after either active release from cells, or passive release upon lytic cell death. Movement of HMGB1 between cellular compartments is a dynamic process induced by a variety of cell stresses and disease processes, including sepsis, trauma, and hemorrhagic shock. Location of HMGB1 is intricately linked with its function and is regulated by a series of posttranslational modifications. HMGB1 function is also regulated by the redox status of critical cysteine residues within the protein, and is cell-type dependent. This review highlights some of the mechanisms that contribute to location and functions of HMGB1, and focuses on some recent insights on important intracellular effects of HMGB1 during sepsis and trauma.


Subject(s)
HMGB1 Protein/physiology , Inflammation/etiology , Sepsis/etiology , Wounds and Injuries/complications , Alarmins/physiology , Animals , Cell Nucleus/metabolism , Humans , Inflammasomes/physiology
19.
J Leukoc Biol ; 106(1): 95-103, 2019 07.
Article in English | MEDLINE | ID: mdl-30924969

ABSTRACT

Gut immune system homeostasis involves diverse structural interactions among resident microbiota, the protective mucus layer, and a variety of cells (intestinal epithelial, lymphoid, and myeloid). Due to the substantial surface area in direct contact with an "external" environment and the diversity of xenobiotic, abiotic, and self-interactions coordinating to maintain gut homeostasis, there is enhanced potential for the generation of endogenous danger signals when this balance is lost. Here, we focus on the potential generation and reception of damage in the gut resulting from exposure to nanoparticles (NPs), common food and drug additives. Specifically, we describe recent evidence in the literature showing that certain NPs are potential generators of damage-associated molecular patterns, as well as potential immune-stimulating molecular patterns themselves.


Subject(s)
Alarmins/physiology , Intestinal Mucosa/drug effects , Nanoparticles/toxicity , Adenosine Triphosphate/metabolism , Administration, Oral , Animals , DNA, Mitochondrial/physiology , Homeostasis , Humans , Intestinal Mucosa/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nanoparticles/metabolism , Oxidative Stress/drug effects
20.
J Leukoc Biol ; 106(1): 193-200, 2019 07.
Article in English | MEDLINE | ID: mdl-30776147

ABSTRACT

The oral cavity is a unique environment containing teeth juxtaposed with soft tissues, all of which are constantly bathed in microbial products and host-derived factors. While microbial dysbiosis in the oral cavity clearly leads to oral inflammatory disease, recent advances find that endogenous danger-associated molecular patterns (DAMPs) released from oral and salivary tissue also contribute to the progression of inflammatory and autoimmune disease, respectively. In contrast, DAMPs produced during oral fungal infection actually promote the resolution of infection. Here, we present a review of the literature suggesting a role for signaling by DAMPs, which may intersect with pathogen-associated molecular pattern (PAMP) signaling, in diseases that manifest in the oral cavity, specifically periodontal disease, oropharyngeal candidiasis, and Sjögren's syndrome.


Subject(s)
Alarmins/physiology , Candidiasis, Oral/etiology , Periodontal Diseases/etiology , Sjogren's Syndrome/etiology , Candidiasis, Oral/immunology , Extracellular Traps/physiology , Humans , Pathogen-Associated Molecular Pattern Molecules/pharmacology , Periodontal Diseases/immunology , Signal Transduction/physiology , Sjogren's Syndrome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...