Trauma equals danger damage control by the immune system

Size: px
Start display at page:

Download "Trauma equals danger damage control by the immune system"

Transcription

1 Review Trauma equals danger damage control by the immune system Veit M. Stoecklein, Akinori Osuka, and James A. Lederer 1 Department of Surgery, Brigham and Women s Hospital/Harvard Medical School, Boston, Massachusetts, USA RECEIVED FEBRUARY 10, 2012; REVISED MAY 4, 2012; ACCEPTED MAY 7, DOI: /jlb ABSTRACT Traumatic injuries induce a complex host response that disrupts immune system homeostasis and predisposes patients to opportunistic infections and inflammatory complications. The response to injuries varies considerably by type and severity, as well as by individual variables, such as age, sex, and genetics. These variables make studying the impact of trauma on the immune system challenging. Nevertheless, advances have been made in understanding how injuries influence immune system function as well as the immune cells and pathways involved in regulating the response to injuries. This review provides an overview of current knowledge about how traumatic injuries affect immune system phenotype and function. We discuss the current ideas that traumatic injuries induce a unique type of a response that may be triggered by a combination of endogenous danger signals, including alarmins, DAMPs, self-antigens, and cytokines. Additionally, we review and propose strategies for redirecting injury responses to help restore immune system homeostasis. J. Leukoc. Biol. 92: ; OVERVIEW OF TRAUMA EFFECTS ON THE IMMUNE SYSTEM The immune system responds rapidly to traumatic injuries by reacting to tissue damage. Following this initial response to injury, cells and mediators of the innate and adaptive immune systems undergo temporal change that have been categorized into proinflammatory and counterinflammatory immune responses and are commonly referred to as SIRS, CARS, or Abbreviations: / deficient, CARS compensatory anti-inflammatory response syndrome, CDC Centers for Disease Control and Prevention, CLP cecal ligation and puncture, DAMP danger-associated molecular pattern, DTH delayed type hypersensitivity, EAE experimental autoimmune encephalitis, Flt3L fms-like tyrosine kinase-3 ligand, FoxP3 forkhead box P3, HMGB1 high-mobility group box nuclear protein 1, HMGN1 high-mobility group nucleosome-binding protein 1, HSP heat shock protein, MARS mixed anti-inflammatory response syndrome, MOF multiple organ failure, MSC myeloid suppressor cell, mtdna mitochondrial DNA, PD-1 programmed death 1, RAGE receptor for advanced glycation end products, ROS reactive oxygen species, SIRS systemic inflammatory response syndrome, TIR Toll-IL-1R, Treg regulatory T cell, TRIF Toll-IL-1R domain-containing adapter-inducing IFN- MARS [1, 2]. Although these descriptive terminologies for this complex host response were first suggested over 20 years ago, they persist and remain useful as terms for delineating the clinical manifestations of trauma [3]. Research efforts have addressed fundamental reasons for the development of SIRS, CARS, and MARS following trauma. The aim of these efforts was to better understand the cellular and molecular mechanisms responsible for the development of these postinjury syndromes. Studies examining the effects of injury on innate and adaptive immune system phenotypes have shown that traumatic injuries induce inflammatory- and counterinflammatory-type immune responses. We believe that these changes in innate and adaptive immune system phenotype and function ultimately disrupt immune system homeostasis, and it is the loss of homeostasis that predisposes trauma patients to opportunistic infections and complications. We also suggest that injuries induce a specific type of an immune response that may have evolved to protect the injured host from opportunistic infections and excessive reactivity to damaged tissues and cells. Similar to other diseases, there are genetic and individual variables that contribute to the severity of trauma effects on the morbidity and mortality associated with trauma and trauma-induced complications. However, these variables are not well-defined. Future efforts to identify some of these variables will be a formidable task but will surely help advance treatment strategies for trauma. One serious complication of trauma is the development of the two-hit response phenotype, which was first defined by Moore et al. [4] as an amplified response to a secondary insult in patients who survive traumatic injuries. In trauma patients who develop infections, this second hit, which is usually an opportunistic infection, can cause severe septic shock and MOF. Thus, the combination of increased risk of developing infections and the development of the two-hit response is a principal complication of traumatic injuries. Research findings suggest that the two-hit response occurs, as cells of the innate immune system, primarily macrophages and neutrophils, become hyper-reactive to bacteria and bacterial toxins [5 8]. Once primed, macrophages and neutrophils produce height- 1. Correspondence: Dept. of Surgery, Brigham and Women s Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA. jlederer@rics.bwh.harvard.edu /12/ Society for Leukocyte Biology Volume 92, September 2012 Journal of Leukocyte Biology 539

2 ened levels of inflammatory cytokines and ROS, causing systemic inflammation and detrimental cell-mediated tissue destruction. This priming effect of injuries on the immune system may be detrimental or beneficial. The excessive reactivity could trigger excessive inflammatory cascades in injured patients who develop opportunistic infections. However, the primed innate immune system may also be better prepared for rapid and strong antimicrobial immune defense. A report by Maung et al. [9] supports this latter hypothesis. In that study, burninjured mice were found to resist i.p. Escherichia coli infections significantly better than sham-injured control mice. In marked contrast, the detrimental impact of heightened innate immune system reactivity was shown by the high mortality in injured mice given a polymicrobial challenge by the CLP technique [10, 11]. An explanation for these contrasting findings may be that the CLP model more closely mimics clinical sepsis after traumatic injuries with a mixed microbial infection, bacteremia, excessive systemic inflammation, and the development of septic shock. Moreover, the difference between trauma effects on antimicrobial immunity and the two-hit sepsis response phenotype highlights the idea that injury disrupts immune system homeostasis. For example, a compartmentalized or lowlevel infection can be controlled more effectively by the innate immune system following injury, but if the infection is not controlled as a result of other influences, the heightened innate immune response to infection sets in motion the two-hit response, putting the injured patient at risk of secondary SIRS, septic shock, and MOF. Another observed complication of traumatic injuries is the development of a counterinflammatory-type immune response, which is believed to be a compensatory response to injury-induced inflammation. The counterinflammatory response to injury has historically been referred to as trauma-induced immune suppression and is defined clinically as the CARS or MARS phase of the trauma response. It was labeled as an immune-suppressive response, as T cell-mediated responses show anergic-like properties including: low proliferative responses to mitogens and specific antigens; suppressed skin DTH responses; suppressed Th1-type immune reactivity with increased Th2- type cytokine production; and increased Treg activity [12 24]. In addition, there is a specific increase in a subpopulation of macrophages referred to as MSCs that occurs coincident with these counterinflammatory-type T cell responses to trauma [25, 26]. These innate cells also arise in clinical conditions, which involve chronic or unresolved immune responses, such as cancer and parasitic infections [27 30]. Interestingly, the counterinflammatory or CARS phase of the injury response becomes pronounced at later time-points after injury, which suggests that it is a developed response to the cellular and molecular triggers that initiates the injury response. We suggest that a logical reason for the counterinflammatory response to traumatic injuries is to control immune reactivity to tissue damage, to suppress trauma-induced inflammation, and to promote a natural healing response to help restore immune system homeostasis. On the other hand, a primary complication of the counterinflammatory response to injuries is that it can actively suppress antimicrobial immunity and may be responsible for the increased susceptibility of trauma patients to opportunistic infections that often occur during the counterinflammatory phase of the injury response. Thus, there are likely beneficial and detrimental influences of counterinflammatory responses to traumatic injuries. A schematic diagram summarizing these general effects of injuries on the immune system responses to trauma and their relationship to SIRS and CARS is shown in Fig. 1. This diagram illustrates that the inflammatory response to injury is driven by the innate immune system and that the counterinflammatory response is mediated by the adaptive immune system. These opposing responses appear to occur in concert, as evidenced by the kinetics of injury-induced changes in innate and adaptive immune system phenotypes. The end result is that injury disrupts immune system homeostasis represented by SIRS and CARS, shown at the peaks or the footballshaped curve. If opportunistic infections arise at time-points when there is a wide imbalance in immune response homeostasis, the injured host will be at high risk of developing traumaassociated complications, such as sepsis, septic shock, or MOF. Figure 1. Traumatic injuries induce an imbalance in immune system homeostasis. This diagram summarizes generalized effects of injury on the immune system and the relationship to the development of SIRS and CARS phenotypes described in trauma patients. As illustrated, injury induces progressive enhancement of inflammatory and anti-inflammatory immune responses. The inflammatory response is driven by cells and mediators of the innate immune system, and the counterinflammatory response is regulated by the adaptive immune system. These opposing injury responses disrupt immune system homeostasis and lead to the development of SIRS and CARS in trauma patients. If opportunistic infections arise at time-points when there is a wide imbalance in immune homeostasis, the injured host will be at high risk of developing trauma-associated complications such as sepsis, septic shock, or MOF. Resolution of the injury response equals restoration of immune system homeostasis in patients who survive. Pro-Inflamma atory Anti-Inflam mmatory Injury SIRS, Two-Hit Response Innate System Response 7 Days Adaptive System Response CARS, Increased Risk of Infection MOF Immune System Homeostasis 540 Journal of Leukocyte Biology Volume 92, September

3 Stoecklein et al. Immune system responses to trauma We propose that these opposing immune responses to injuries represent a natural healing-type host response aimed at controlling trauma-induced tissue damage and at restoring immune system homeostasis following a traumatic event. INITIATORS OF THE INJURY RESPONSE According to the CDC, accidental injuries tops the list as the leading cause of death in the United States for people under 44 years of age ( db64.pdf). This statistic does not include deaths from complications of injuries, such as sepsis, so this statistic may be higher than calculated by the CDC. Accidental injuries can be organized into general groups to include hemorrhage, crush injuries, blunt-force trauma, burn injury, bone fractures, chemical injury, radiation injury, and combination injuries, such as trauma with hemorrhage or radiation with combined trauma. It is not challenging to pair the above-mentioned types of injury with traumatic scenarios, such as motor vehicle accidents, earthquakes, fires, explosions, shootings, stabbings, major surgeries, and radionuclear accidents or events. War and terrorism contribute significantly to some of these types of injuries, and this has alerted nations to develop emergency preparedness strategies and medical countermeasures to help protect civilians and war fighters in harms way [31]. Although injury can occur in many ways, there are some common features to trauma that allow for a reductionist, scientific approach to study how injury influences the immune system. One common component of trauma is that it causes tissue damage. Although the level and type of tissues that are damaged by injury vary, it is generally agreed that injured tissue initiates the early host response to trauma. Another feature of traumatic injuries is that they occur by accident or without prior warning. Therefore, research into how to pretreat the physiological and immunological consequences of traumatic injuries will not provide clinically useful findings. We also posit that injury is a natural type of an immune response that has undergone evolutionary changes in a manner similar to antimicrobial immune responses or adaptive immune regulation in mammals. Thus, the initiating factors and mediators of injury responses may use the same immune cells and signaling pathways for normal immune responses against pathogens but will also stimulate unique types of reactions. In addition to specific effects of trauma on immune cell responses, there are physiological events associated with the host response to trauma, such as stress, shock, and neurological responses, which add to the complexity of the trauma response. A future challenge will be to work at the interface between the physiology and immunology of trauma to provide insights about how to restore immune homeostasis following traumatic injuries. TRAUMA EQUALS DANGER The substantial tissue destruction and necrotic cell death that is induced by traumatic injuries, lead to the release of previously sequestered antigens and factors. These antigens alert the immune system that tissue damage is present and are appropriately called alarmins [32, 33]. Alarmins are detected by immune cells and initiate an immune response by interacting with PRRs, such as TLRs, which are present on innate or adaptive immune cells [34]. Once detected, alarmins specifically alert immune cells to initiate inflammatory responses, chemotaxis, antimicrobial defenses, and adaptive immune cell responses. The detection of alarmins by cells of the immune system forms the basis for the danger theory, as proposed by Matzinger in 1994 [35]. Danger is clearly a central component of the immune response to trauma. Following trauma, the immune system is exposed to a large amount of tissue damage and endogenous antigens, including alarmins, and cell debris are likely released from necrotic cells and tissue. There may also be significant exposure to microbial molecules that are part of the microbiome [36]. Immune cells would then respond to injury antigens by PRRs that recognize DAMPs of microbial or exogenous origin (PAMPs) or endogenous origin (alarmins) [37, 38]. Whereas PAMPs alert the immune system to activate antimicrobial defenses, alarmins alert the immune system to tissue damage and danger. The shared recognition of danger by PAMPs and alarmins suggests that a primary function for PRRs might be to signal antimicrobial immunity against PAMPs to trigger an antipathogen immune response or against future opportunistic pathogens that could infect damaged tissues. The ability of PRRs to recognize PAMPs and alarmins suggests an evolutionarily conserved function for these receptors as innate immune recognition systems [38]. Among the expanding list of PRRs, TLRs, especially TLR4, have emerged as promiscuous sensors for alarmins following trauma. However, other TLRs and innate immune recognition molecules have also been shown react to alarmins and signal danger responses [39, 40]. We provide an updated list of some of the known trauma-induced alarmins, as well as some basic information about these molecules in Table 1. A more indepth overview of other potential trauma-associated alarmins was published recently by Manson et al. [56]. It is clear from this list of alarmins that TLR4 has emerged as a primary receptor for danger. However, it is important to note that PAMPs, like bacterial LPS, can also bind to TLR4, which has generated controversy over whether DAMPs may carry microbial molecules to PRRs[57]. Examples of this scenario include reports that HMGB1 binds LPS, bacterial DNA, as well as IL-1 [58, 59]. Moreover, some authors have recently called into question whether DAMPs, such as HMGB1 and HSPs, actually have cytokine-like properties [60, 61]. They report that highly purified HMGB1 and HSPs have low biological activity. Nevertheless, whether by indirect or direct mechanisms, HMGB1 and HSPs appear to act as alarmins, as they are released following cell injury and tissue trauma [62, 63]. For example, Cohen et al. [42] detected HMGB1 in the plasma of trauma patients early after the injury and reported that higher plasma levels of HMGB1 were associated with the severity of injury, organ dysfunction, post-trauma coagulopathy, and adverse outcome. Moreover, HMGB1 has been implicated in the pathogenesis of a variety of human inflammatory diseases that also cause substantial tissue injury, such as sepsis, arthritis, cancer, and type I Volume 92, September 2012 Journal of Leukocyte Biology 541

4 TABLE 1. List of Injury-Induced Alarmins Alarmin Receptor Pathways activated Pathophysiological importance References HMGB1 TLR2, TLR4, RAGE MyD88, TRIF Trauma, sepsis, arthritis, [41 46] cancer, type I diabetes HMGN1 TLR4 MyD88 Unknown [47] HSPs TLR2, TLR4 MyD88, TRIF Trauma, cardiovascular [48, 49] diseases Mitochondria/mtDNA TLR9 and FPR1 MyD88, TRIF, G-coupled Trauma [50] receptor signaling pathways Uric acid NLRP3, IL-1R Caspase-1, MyD88 Lung fibrosis, gout [51 53] ATP P2X 7 Caspase-1 Lung fibrosis [54] S100 TLR4, RAGE MyD88 Autoimmune diseases, cancer [55] NLRP3, Nucleotide-binding oligomerization-like receptor family, pryin domain-containing 3. diabetes [41, 44 46]. As HMGB1 signals through TLR4, it is believed that it causes complications in trauma by initiating or amplifying inflammatory cytokine production by macrophages [64]. Other receptors activated by HMGB1 include TLR2 and RAGE [65]. Members of the HSP family also act as alarmins, as they are released extracellularly by damaged or stressed cells or tissues following trauma [49]. Similar to HMGB1, HSPs have been shown to trigger TLR responses, in particular, TLR2 and TLR4 pathways, and HSPs have been shown to mediate detrimental effects in trauma, e.g., in the cardiovascular system [48]. Moreover, like HMGB1, HSPs have also been shown to act as carrier molecules for PAMPs [57, 66 68]. HMGN1 was characterized recently as an alarmin for antigenspecific immune responses, as well as LPS-induced innate immune responses. Like other alarmins, HMGN1 was found to signal through the TLR4/MyD88 pathway [47]. The role of HMGN1 in trauma is yet unknown. Finally, Zhang et al. [50] reported that systemic inflammatory responses to trauma can be initiated by mitochondrial danger molecules formyl peptides and mtdna which are released into the bloodstream following trauma. The recognition of mitochondria debris as alarmins is thought to be a result of molecular similarities between bacteria and mitochondria, which are obligate bacterial endosymbionts. INFLUENCES OF TRAUMA ON INNATE IMMUNE SYSTEM RESPONSES AND FUNCTIONS Our group has shown that TLR4 reactivity is enhanced following trauma and that this increase in TLR4 responsiveness contributes to the development of the two-hit response phenotype in a mouse injury model [8, 69]. The primary complication of the two-hit response is the development of a systemic inflammatory response against sepsis-causing opportunistic pathogens, such as Pseudomonas aeruginosa, E. coli, and Staphylococcal infections. A clinically relevant example of the two-hit response to trauma is the high mortality that occurs in injured mice, given a sepsis-causing infection by the CLP approach at 1 2 weeks after the injury [11, 70, 71]. Importantly, the kinetics of the development of the two-hit response phenotype in mice overlaps with clinical observations in trauma patients [72, 73]. Another feature of the two-hit response to trauma is that macrophages appear to be the primary innate immune cell type showing enhanced innate reactivity [8, 74, 75]. Our group [8] showed that macrophages from burn-injured mice produce and express high levels of IL-1, IL-6, and TNF- following LPS challenge. Schwacha et al. [76] found similar results in a mouse burn injury model and also demonstrated that unresponsiveness to IL-10 by macrophages may be one mechanism responsible for increased inflammatory cytokine production by macrophages following trauma. Moreover, it has been shown recently that burn injury or trauma causes a progressive increase in splenic macrophages with a phenotype similar to MSCs that were first identified in tumor-bearing mice. Myeloid suppressor-type macrophages have also been identified in mice challenged with parasite antigens or cannabinoid receptor activation or in mice subjected to CLP-induced sepsis [26, 29, 30, 77 79]. The trauma-induced macrophages express cell-surface F4/80, CD11b, and GR-1 markers and appear to be the primary source of TLR4-induced TNF- production [25, 80]. Macrophages display heighted inflammatory reactivity in other injury models, including in mouse models for femur fracture, trauma-hemorrhage, and spinal cord injury [81 83]. This proinflammatory behavior of GR- 1 CD11b macrophages may be unique to injury responses. Thus, it appears that trauma-induced changes in macrophage phenotype may be a common response to traumatic injuries. It is important to note that several studies have shown a diminished production of proinflammatory cytokines when whole blood or mononuclear blood cells from trauma patients were stimulated with LPS ex vivo [84, 85]. The findings in these patient studies contradict the priming effects of injury on the innate immune system, which we discussed above. One potential explanation for this controversy could be a result of the cellular compartments that were tested in patients versus in animal studies. Patient studies were performed using circu- 542 Journal of Leukocyte Biology Volume 92, September

5 Stoecklein et al. Immune system responses to trauma lating immune cells, whereas animal studies used cells prepared from peripheral immune organs. A recent publication from the Inflammation and Host Response to Injury Large- Scale Collaborative Research Program reported on gene-expression profiles in blood leukocytes prepared from trauma patients. In this comprehensive study, it showed up-regulation of many proinflammatory innate immune cell response genes, as well as counterinflammatory genes [86]. Thus, there remains controversy over whether trauma induces suppression of innate immune cell responses. Future work should focus on studying specific molecular triggers responsible for the development of the two-hit response phenotype following trauma and should focus primarily on macrophage biology. Alarmins are strong candidates for these molecular triggers. Furthermore, as alarmins, such as HMGB1, signal inflammasome activation, this pathway might also be a part of the signaling network for cellular responses to injury. In agreement with this, a recent report by Osuka et al. [87] showed that burn injury triggered inflammasome activation by 1 day after injury in innate and adaptive immune cell subsets. Interestingly, blocking caspase-1 activation in injured mice caused heightened inflammation and mortality, which suggests a protective role for inflammasome activation as a protective response to injury. A similar protective role for caspase-1 and inflammasome activation in the liver was shown to occur in a mouse model for hemorrhagic shock [88]. Although we do not yet know the molecular signals for increased TLR signaling by macrophages following trauma, the pathways that signal TLR responses are well known and provide some clues about how trauma augments innate immune responses and inflammation. Thirteen different TLRs have been identified in mice and 11 in humans [89]. These receptors share similarity to Drosophila Toll protein containing leucine-rich repeats and TIR domains for recognition of alarmins and intracellular signaling. Two adaptor proteins, also with TIR domains MyD88 and TRIF control the initiation of TLR responses and are adaptors for the MyD88-dependent and -independent (TRIF-dependent) signaling pathways [90]. Activation of the MyD88 pathway leads to immune cell activation and cytokine production through the NF- B pathway, and the TRIF-dependent pathway induces type I IFN production through the transcription factor IFN regulatory factor 3 [90]. We identified that the p38 MAPK signaling pathway was amplified significantly in LPS-stimulated macrophages from injured mice [69]. Other reports confirmed this finding and also suggest a role for the p38 MAPK signaling pathway as mediating the amplified TLR reactivity that has been described to occur after injuries [91 94]. It is not yet known whether the LPSenhanced p38 MAPK signaling depends on the MyD88 or TRIF pathway, and as suggested by Triantafilou et al. [95], receptor crosstalk among CD14, CXCR4, and TLRs could be involved in amplifying TLR signaling. Nevertheless, therapies that specifically target the p38 MAPK signaling pathway in trauma patients showing clinical signs of septic shock or SIRS, might help mollify the two-hit response and its associated complications. DAMAGE CONTROL: THE ADAPTIVE IMMUNE RESPONSE TO TRAUMA Cells and mediators of the innate and adaptive immune system do not function in isolation. Their interaction is needed to initiate specific adaptive immune responses that then feed back on innate immune cells to augment antimicrobial immunity and resolve inflammation. APCs provide one direct link between innate and adaptive immune cell activation during infections and vaccinations. For example, infections caused by bacterial pathogens or viruses evoke a strong antimicrobialtype T cell response, as APCs are activated in a manner to skew CD4 T cell differentiation toward Th1 or Th17 CD4 T cell subsets [96 99]. These CD4 T cell subsets produce cytokines, which then provide activating signals to innate cells, such as neutrophils, macrophages, and NK cells, to boost the antimicrobial effector activity. On the other hand, certain types of immune responses lead to the development of Th2- type or counterinflammatory-type immune responses. Th2 responses occur with infections caused by some parasites or allergic responses and tend to suppress or antagonize Th1- or Th17-type immune responses [96]. The balance between Th1- and Th2-type immune response phenotypes can be an important determinant of infection, disease, and clinical outcomes. It is generally agreed that trauma suppresses CD4 T cell responses [19, ]. For example, Th1-type immune responses have been shown to be reduced markedly following trauma in patients and in mouse injury models [13, 103, 104]. Trauma has also been described to promote Th2-type immune system responses and T cell anergy in trauma patients and in mice [105, 106]. Interestingly, exercise can also skew T cellmediated immune responses, suggesting that injury-induced changes in the immune system can be induced by activities that have health benefits [107, 108]. We [105] and other research groups [109, 110] have focused significant efforts on identifying how trauma might suppress CD4 T cell activation and push the adaptive immune response toward a counterinflammatory phenotype. Early studies suggested that the counterinflammatory behavior of that adaptive immune system may be a result of a shift toward high Th2 and low Th1 responses in mice and in patients. Other investigators [13] argued that as Th1- and Th2-type immune responses were found in trauma patients, T cell anergy may be another mechanism contributing to suppressed CD4 T cell responses following trauma. Our research group [103] performed immunization studies in mice as an attempt to determine whether injury drives higher Th2- type reactivity or suppresses Th1-type responses. This was accomplished by measuring antigen-specific antibody isotype formation in mice immunized with a T cell-dependent antigen at the time of injury and measuring CD4 T cell help for antibody production. We observed that Th1 responses were suppressed dramatically in immunized burn mice. The suppressive effect on injury on Th1 responses occurred even though the immunogen was given in a strong adjuvant, CFA. This adjuvant works by providing TLR-activating signals to APCs, yet Th1 responses remained suppressed for at least 10 days after injury and immunization. This dominant and strong Th1-suppressive activity caused by injury was confirmed in subsequent immuni- Volume 92, September 2012 Journal of Leukocyte Biology 543

6 zation and adoptive transfer studies using CD4 T cells from DO-11 TCR transgenic mice [23]. DO-11 CD4 T cells express TCRs that are specific for OVA peptide (aa sequence ) and can be detected in OVA peptide-immunized mice to track expansion and phenotypic influences of injury on adaptive immune responses. In these studies, burn injury suppressed antigen-driven CD4 T cell expansion and OVA peptide-specific Th1 cytokine production. Taken collectively, the results of these immunization studies suggest that trauma can actively suppress CD4 T cell activation, Th1 differentiation, and proliferation without an increase in Th2-type responses. The biological reasons for trauma effects on adaptive immune responses were not well understood at the time that these studies were published. However, we now propose that trauma-induced changes in adaptive immune responses might occur to protect the injured host from alarmin-induced innate responses and to prevent possible self-antigen reactivity to injury antigens released by tissue damage. Tregs CONTROL INJURY RESPONSES Tregs are a subset of thymic-derived CD4 T cells, which have been shown to be central to the maintenance of immunological tolerance and to controlling inflammation in autoimmune and inflammatory disease mouse models [111, 112]. This unique CD4 T cell subset, which makes up 10% of the peripheral CD4 T cell population, expresses TCR repertoire diversity similar to conventional CD4 T cells [113, 114]. However, in contrast to conventional CD4 T cells, some Tregs are self-reactive, and this self-reactivity plays a role in their immune tolerance function [ ]. Their development is dependent on the FoxP3 transcription factor, as mice lacking the FoxP3 gene do not have detectable Tregs [118]. FoxP3 / mice develop fatal autoimmune disease, providing more support for the concept that Tregs are a distinct CD4 T cell lineage and that they are needed to maintain immune tolerance [ ]. Additional subsets of Tregs, called T regulatory 1, Th3, or induced Tregs, arise in the periphery in response to IL-10 or TGF- signaling and have been characterized as being important in controlling inflammation, especially in a mouse colitis experimental model and EAE [ ]. Tregs are often distinguished from conventional CD4 T cells by the expression of cell-surface CD25 molecules. Although not as definitive a marker of Tregs as FoxP3, CD25 expression on CD4 T cells is widely used as a tool to purify mouse Tregs and to target the depletion of Tregs in mice [112]. Purified Tregs markedly suppress the proliferation of TCR-activated CD4 T cells by cell contact [126]. Tregs also have the capacity to produce TGF- 1, IL-9, and IL-10 and to up-regulate the cell-surface expression of several negative costimulatory molecules, such as CTLA-4, ICOS, and PD-1, as well as TNFR2 [24, ]. As all of these factors have been described as potent suppressors of inflammation or T cell reactivity, it is possible that these factors contribute to Treg function during trauma or other host responses, which involve substantial tissue damage, such as severe infections, transplantation, cancer, and autoimmune reactions. Several biological features of Tregs suggest that they may control the adaptive immune response to injury. First, their capacity to suppress CD4 T cell activation and proliferation suggests that they could drive trauma-induced suppression of Th1 responses and T cell anergy. Second, the tissue damage associated with injuries could activate self-reactive Tregs. The first evidence supporting this idea came from a mouse injury study, showing that by 12 h after burn injury, there was a significant increase in CD4 T cells coexpressing CD25 and CTLA-4 in the LNs draining the site of injury [131]. Recently, our group confirmed that burn injury triggers TCR signaling in CD4 FoxP3 T cells but not CD4 FoxP3 or CD8 T cells in the LN draining the injury site [132]. Finally, as trauma can induce systemic immune suppression in patients, it is possible that overly active Tregs could inadvertently promote or amplify immune suppression by blocking antimicrobial immunity, while attempting to control the inflammatory response to injury. To determine whether injury affects Tregs, we compared the regulatory activities of CD25 CD4 T cells purified from the LNs or spleens of sham- and burn-injured mice at 7 days after injury. We discovered that Tregs from injured mice were significantly more potent at blocking CD4 T cell proliferation than sham Tregs [24]. Moreover, we found that LN but not splenic Tregs expressed higher Treg activity, which provided new evidence to suggest that Treg activation by injury may be compartmentalized to injury-site LNs. Closer investigation into injury effects on Tregs showed that it caused increased expression of costimulatory molecules, indicative of increased Treg function or survival, CTLA-4, PD-1, ICOS, and CD28 [24]. The enhanced expression of these costimulatory molecules could function to amplify Treg activity following traumatic injuries. For instance, the increase in CTLA-4 expression by Tregs at 7 days after injury could help increase Treg potency by suppressing DC APC function through an IDO mechanism or by competitively blocking CD80 and CD86 access to CD28 to suppress T cell activation [133]. ICOS and PD-1 have been shown to play a role in mediating Treg responses and CD28 signaling and also appear to be required to maintain Treg proliferation in peripheral immune tissues [ ]. Finally and most important, conventional CD4 T cells showed no injury-induced changes in costimulatory molecule expression, suggesting that only Tregs are activated in response to injury. Similar type changes in Tregs have been reported to occur in trauma and sepsis patients, which strengthens the clinical importance of these findings reported in mouse injury studies [138, 139]. Taken together, these results provide additional evidence supporting the idea that tissue injury from trauma delivers some type of an activation signal to Tregs and that Treg activation may be a natural element of host response to traumatic injuries. The discovery that injury augments Treg activity prompted experiments to test whether Tregs might regulate trauma-induced changes in innate and adaptive immune functions. Based on what is known about how Tregs modulate immune responses, our research group examined whether Tregs might contribute to the suppression of Th1-type immune reactivity and the control of the two-hit response phenotype in trauma. To address these questions in vivo, we optimized a Treg-depletion protocol using a moderate dose of anti-cd25 antibody, 544 Journal of Leukocyte Biology Volume 92, September

7 Stoecklein et al. Immune system responses to trauma which removed CD25 T cells from the peripheral immune tissues of mice but did not prevent T cell activation. T cell responses were measured in vivo by immunizing Treg-depleted mice with the T cell-dependent antigen, TNP-haptenated OVA, and measuring antibody isotype formation, as well as OVA-induced IFN- production. Our results indicated that sham or burn Treg-depleted mice produced higher titers of the Th1-type antibodies and higher levels of IFN-, which indicated that Tregs were actively suppressing Th1-type reactivity in injured mice [24]. A subsequent report using the DO-11 TCR transgenic adoptive-transfer experimental system showed that injury in Treg-depleted mice greatly enhanced antigenspecific CD4 T cell expansion as well as Th1 reactivity [140]. Importantly, the results of this study showed for the first time that Tregs may be activated by trauma and tissue damage in an antigen-specific manner. The reason for antigen-specific control of antigen-specific T cell responses may be to protect the host from developing potentially harmful Th1-type immune responses to self-antigens released by the tissue damage associated with the injury. Interestingly, findings reported by Zang et al. [141] showed that high-level T cell activation at early time-points after burn injury, induced by bacterial superantigen, led to high mortality, whereas there was no mortality when mice were challenged with superantigen at 1 week after injury a time-point when Treg numbers are increased, and Tregs can potently suppress CD4 T cell activation [142]. Similar immune regulatory functions for Tregs have been suggested by other studies, showing that Tregs protect the host from excessive self-t cell reactivity in inflammatory disease models, such as T cell-mediated colitis and EAE [123, ]. Thus, Tregs appear to control CD4 T cell responses in trauma or related inflammatory-type diseases by reduction Th1-type inflammation and self-antigen reactivity (Fig. 2). The potential that Tregs might also control the inflammatory response to injury was first suggested by the results of experiments examining how Rag1 / mice, which lack T and B cells, respond to injury. When injured, spleen cells from Rag1 / mice showed heightened reactivity to TLR2 and TLR4 stimulation, as compared with spleen cells from WT control mice [11]. This observation suggested that the adaptive immune system must actively suppress or control the inflammatory behavior of the innate immune response to injury. To address which adaptive immune cell type might be needed to suppress injury-induced inflammation, CD4 and CD8 T cell / mice were compared directly with Rag1 / and WT mice for changes in TLR4 responsiveness after burn injury. The results of these experiments demonstrated that CD4 T cell / mice resembled Rag1 / mice, showing higher TLR4 responses, whereas CD8 T cell / mice resembled WT mice [147]. When Rag1 / mice were given WT CD4 T cells by adoptive transfer, the injury-induced increase in TLR4 responsiveness was suppressed. Further separation of CD4 T cells into CD25 and CD25 subsets prior to adoptive transfer into CD4 / mice showed that CD25 CD4 T cells suppressed this injury-induced increase in TLR4 responsiveness [147]. These findings provided strong evidence to indicate that Tregs play an active role in controlling the inflammatory response to injury. As it is know that injured mice and humans can develop heightened systemic inflammatory reactivity after severe injury, which can promote the development of MOF, we tested recently whether Treg deficient mice might be more susceptible to this two-hit response. Using a burn injury, followed by a LPS challenge two-hit mortality model, we directly compared the two-hit response in Treg deficient mice with control non- Treg-depleted mice [69]. We found that Treg deficient burninjured mice were significantly more susceptible to LPS challenge, showing 100% mortality versus 50% mortality for burn WT mice (unpublished results). Importantly, Treg deficient sham mice given LPS did not die, suggesting that it is the combination of Treg deficiency and injury that enhanced the two-hit response. Moreover, LPS-challenged Treg deficient burn-injured mice showed higher levels of TNF- and IL-6 in lungs, liver, and kidney than control mice, further suggesting that Tregs have the capacity to control inflammatory responses to traumatic injuries (Fig. 2). These findings showed that Tregs played a protective role in controlling the magnitude of the injury response to trauma. The clinical significance of this observation is that individuals with lower levels of Tregs or dysfunctional Tregs could be at higher risk for developing inflammatory complications to trauma or other inflammatory-type diseases. For example, Treg numbers are decreased in patients with systemic lupus erythematosus and rheumatoid arthritis [148, 149]. Although a clinical relationship between the development of postinjury sepsis and altered Treg numbers or function is unknown, there is evidence that injury and sepsis can influence Tregs in patients. We reported recently [20] that circulating Injury MacΦ Treg TLR4 Reactivity Anti-Microbial Response GR-1+CD11b+ Macrophages MacΦ Innate Adaptive CD4 T Cell Inflammatory Response Two-Hit Response Macrophage TLR4 Response Th1 Reactivity T Cell Activation Figure 2. Immune cell subsets that react to injury and control the injury response. Tissue damage caused by trauma leads to the release of alarmins and self-antigens into extracellular compartments, which could activate macrophages (Mac ) and Tregs. In macrophages, injury triggers a number of changes in phenotype and function, including enhanced TLR4 reactivity and antimicrobial responses, as well as increased appearance of GR-1 /CD11b macrophages. Tregs are then activated and have been shown to act as master regulators of the injury response by suppressing innate and adaptive cellular responses to trauma. Volume 92, September 2012 Journal of Leukocyte Biology 545

8 Tregs from trauma patients demonstrate enhanced Treg activity by 5 7 days after injury as compared with Tregs prepared from patients at 1 day after injury or from normal individuals. This finding validates the results of experimental studies in our mouse burn injury model. Because of this, we have subsequently examined changes in mouse blood Treg function and found that they also displayed enhanced Treg potency by 7 days after injury (unpublished observation). Another report showed that septic patients develop higher numbers of circulating Tregs, suggesting that enhanced Treg presence or responsiveness may also be part of the host response to sepsis and strong inflammatory responses [150]. Experiments performed in a mouse sepsis model, CLP, support this hypothesis. Two independent reports indicate that CLP induces an increase in Treg numbers and function [151, 152]. Whether Tregs are beneficial or harmful during a sepsis response remains controversial. Two studies indicate that the CLP-induced survival of mice, made Treg deficient by anti-cd25 antibody treatment, was not different than control mice [151, 152]. However, one study showed that transferring CD25 CD4 T cells into CLP-challenged mice was protective [153]. One potential reason for these opposing findings is that mice that lack Tregs may develop enhanced inflammatory reactivity following sepsis and could concomitantly develop a stronger, protective Th1 response to boost T cell-mediated immunity. These offsetting responses might result in no apparent survival differences in CLP-induced survival between mice, with or without functional Tregs. The differences in CLP survival responses between these reports could also be a result of the severity of the CLP challenge. POTENTIAL TREATMENT STRATEGIES TO RESTORE IMMUNE SYSTEM HOMEOSTASIS FOLLOWING TRAUMA The results of basic and clinical studies support the overall hypothesis that traumatic injuries disturb normal immune system homeostasis and that interventional approaches to reduce this imbalance could reduce the morbidity and mortality rates associated with traumatic injuries. One of the roadblocks in developing trauma-directed therapies is the heterogeneity in the injury response among trauma patients. Thus, efforts to classify patients into trauma response phenotypes by genetic or biological markers will greatly augment the potential for developing more effective treatment approaches for this complex disease process. Furthermore, the limited understanding of trauma and sepsis in the past had a large negative impact on advancing drug development and treatments for this complex disease as a result of failed clinical trials. Nevertheless, some studies in preclinical animal models have shown that it is possible to change the clinical trajectory of traumatic injuries using cytokines, sex hormones, and immunonutrition, among others, to restore immune system homeostasis following trauma. The available information gained from these studies suggests that treatments, which reduce the extremes of innate and adaptive immune system responses to injuries, help protect trauma patients from developing infectious and inflammatory complications from trauma. These encouraging preclinical findings, along with the better present day understanding of how trauma influences the immune system, suggest that clinical trials to evaluate new and safer approaches should be explored. Cytokines are made by immune and nonimmune cell types and can transmit local and systemic signals to initiate and regulate immune responses. Early clinical studies tested the potential of blocking inflammatory cytokines, such as TNF and IL-1, as they were shown to be increased in patients who showed worse outcomes in trauma and sepsis. Unfortunately, results of these clinical trials showed that anti-inflammatory approaches were not beneficial [154]. Another cytokine therapy that was tested clinically was GM-CSF treatment for lung injury and sepsis. GM-CSF is produced by activated macrophages, T cells, endothelial cells, and fibroblasts and acts on hematopoietic stem cells to promote development of neutrophils, monocytes, and macrophages. When given to patients with acute lung injury, the treatment showed no significant benefit, but meta-analysis of its effects in treating septic shock showed it reduced infection [155, 156]. This suggests that there may be a use for GM-CSF as an immune-enhancing therapy for traumatic injuries to prevent opportunistic infections. Advances for treating traumatic injuries have been made in preclinical animal studies. One successful approach was to use IL-12 to promote Th1-type immune responses in mice to heighten antimicrobial immunity. In one study, IL-12 treatment, given at early time-points after injury, restored resistance to CLP sepsis challenge in burn-injured mice [157]. Another report indicated that IL-12-treated burn mice were protected from herpes simplex infection when treated early after injury [158]. The clinical relevance of these studies was shown by the observation that PBMCs from trauma patients were found to produce significantly less IL-12 in response to LPS stimulation as compared with healthy controls [159]. As IL-12 is known to potently induce IFN- production and Th1-type immune responses, these findings provide evidence to suggest that redirecting the counterinflammatory T cell response to injuries could be a useful approach to restore immune homeostasis. However, clinical trials for IL-12 treatment in trauma patients have not yet been pursued as a result of concerns over the toxicity observed in human cancer trials in patients given high doses of IL-12 [160]. Regardless, there is ongoing interest in IL-12 in cancer research, and it may be important to revisit IL-12 as a therapy for traumatic injuries [161]. Another cytokine, which has been shown to restore immune function in injured mice, is IL-18, which induces IFN- production by NK cells; this could promote Th1-type responses after traumatic injuries. Treating mice with IL-18 after burn injury protected mice from CLP sepsis and also increased antimicrobial immunity in injured mice [162]. Another report showed that blocking IL-10 activity in vivo by anti-il-10 antibody treatment could enhance Th1 immunity in mice and also protect burn-injured mice from CLP sepsis [163, 164]. A commonality among all of these cytokine-based treatments, which restore immune system function in injured mice, is that they all enhance early inflammatory-type immune reactions. Thus, it seems possible that developing treatments, which boost early inflammatory-type reactions rather than suppressing inflammation after trauma, 546 Journal of Leukocyte Biology Volume 92, September

9 Stoecklein et al. Immune system responses to trauma may help redirect the immune system responses to trauma and restore immune system homeostasis. In support of this hypothesis, we found recently that giving CpG oligodeoxynucleotide at 1 day after injury could restore Th1-type responses in mice and protect them from succumbing to two-hit sepsis at 1 week after burn injury and treatment (unpublished results). Other cytokines, which have been used to treat the immunological complications of trauma, are IL-15 and Flt3L. IL-15 is produced by activated macrophages and stimulates NK cell and CD8 T cell activation. Flt3L is a hematopoietic growth factor, which stimulates DC growth and differentiation [165]. Treating mice with IL-15 was found to restore depressed, splenic DC function in mice following trauma-hemorrhage [166]. Similarly, Flt3L ameliorated burn injury-induced immune defects and increased resistance to infection in mice [ ]. These findings provide evidence supporting a role for cytokines as effective treatments to redirect or restore immune system function in trauma by stimulating hematopoiesis or to reprogram immune cell functions. Gender is a central determinant of clinical outcomes for trauma and sepsis, with male gender having a negative influence on trauma complications [ ]. Therefore, a different approach to treating the detrimental effects of traumatic injury on the immune system is modulating sex hormones. Many studies from the Chaudry group [174, 175] showed that 17 -estradiol is the key gender factor responsible for the finding that proestrus female mice have enhanced immune function following trauma as compared with male mice. Accordingly, treatment with 17 -estradiol has been shown to be beneficial in trauma-hemorrhage and burn injury in mice [176]. In a more recent study, 17 -estradiol effects on restoring immune system function in a trauma-hemorrhage mouse model were shown to be dependent on peroxisome proliferator-activated receptor [177]. Taken together, the results of these and other experimental studies suggest great potential for modulating sex hormones to transiently treat the immune consequences of trauma [178]. Various other approaches, which have been tested for improving immune system function after trauma, include immunonutrition. Examples of immunonutrition as treatment for critically ill patients and trauma include glutamine, arginine, and omega-3 fatty acid feeding [179]. For example, one clinical study supports using glutamine feeding to reduce clinical complications in trauma patients [180]. In summary, the results of experimental studies testing cytokines, sex hormones, and immune-enhancing nutrition as treatments for the clinical complications of traumatic injuries are encouraging. Nevertheless, translating these results from the bench to the bedside remains a challenge for all investigators in the field. CONCLUDING REMARKS Substantial progress has been made in understanding how traumatic injuries influence the immune system, and the information gained supports the hypothesis that trauma shifts normal immune system balance toward proinflammatory and counterinflammatory phenotypes. Data support the idea that these opposing influences of injuries on the immune system can be protective, can occur in concert, and can also contribute to the pathophysiological complications of traumatic injuries. For example, the development of the two-hit response phenotype represents a complication of trauma, whereas this same priming effect of injuries on the innate immune system can boost protective antimicrobial immune function. Moreover, research findings have uncovered the principal immune cell subsets that mediate the inflammatory and counterinflammatory responses to traumatic injuries. We propose that tissue macrophages mediate the innate inflammatory response to injuries, whereas CD4 Tregs control the adaptive immune response. Furthermore, there is significant crosstalk between these immune cells, as injury in the absence of Tregs skews the injury response to a more proinflammatory-type response. The versatile regulatory activities of Tregs make them wellsuited to act as master regulators of the injury response. We believe that alarmins, including DAMPs and PAMPs, will likely emerge as molecules that trigger immune responses to injuries. As such, alarmins could trigger a priming effect on the immune system to promote inflammation, antimicrobial immunity, and Treg activation. Available data support the ideas that Treg activation by alarmins could occur by direct signaling, by indirect activation via alarmin-activated APCs, or by self-antigen recognition. Unraveling these at-present, unknown mechanisms will be a significant advancement for injury research and the development of treatments to restore immune system homeostasis in trauma patients. AUTHORSHIP This review was prepared in collaboration among all of the listed authors (V.M.S., A.O., and J.A.L.). ACKNOWLEDGMENTS The work from our laboratory, which was discussed in this review, was supported by funding from the U.S. National Institutes of Health Grants R01GM035633, RO1GM57664, and R33AI The authors thank Drs. John Mannick and Andrew Lichtman for their helpful comments. REFERENCES 1. Novotny, A. R., Reim, D., Assfalg, V., Altmayr, F., Friess, H. M., Emmanuel, K., Holzmann, B. (2012) Mixed antagonist response and sepsis severity-dependent dysbalance of pro- and anti-inflammatory responses at the onset of postoperative sepsis. Immunobiology 217, Osuchowski, M. F., Welch, K., Siddiqui, J., Remick, D. G. (2006) Circulating cytokine/inhibitor profiles reshape the understanding of the SIRS/CARS continuum in sepsis and predict mortality. J. Immunol. 177, Bone, R. C. (1996) Sir Isaac Newton, sepsis, SIRS, and CARS. Crit. Care Med. 24, Moore, F. A., Moore, E. E., Read, R. A. (1993) Postinjury multiple organ failure: role of extrathoracic injury and sepsis in adult respiratory distress syndrome. New Horiz. 1, Fan, J., Marshall, J. C., Jimenez, M., Shek, P. N., Zagorski, J., Rotstein, O. D. (1998) Hemorrhagic shock primes for increased expression of cytokine-induced neutrophil chemoattractant in the lung: role in pulmonary inflammation following lipopolysaccharide. J. Immunol. 161, Fan, J., Kapus, A., Li, Y. H., Rizoli, S., Marshall, J. C., Rotstein, O. D. (2000) Priming for enhanced alveolar fibrin deposition after hemorrhagic shock: role of tumor necrosis factor. Am. J. Respir. Cell Mol. Biol. 22, Volume 92, September 2012 Journal of Leukocyte Biology 547

Innate immunity. Abul K. Abbas University of California San Francisco. FOCiS

Innate immunity. Abul K. Abbas University of California San Francisco. FOCiS 1 Innate immunity Abul K. Abbas University of California San Francisco FOCiS 2 Lecture outline Components of innate immunity Recognition of microbes and dead cells Toll Like Receptors NOD Like Receptors/Inflammasome

More information

2. Innate immunity 2013

2. Innate immunity 2013 1 Innate Immune Responses 3 Innate immunity Abul K. Abbas University of California San Francisco The initial responses to: 1. Microbes: essential early mechanisms to prevent, control, or eliminate infection;

More information

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF

Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases. Abul K. Abbas UCSF Tolerance, autoimmunity and the pathogenesis of immunemediated inflammatory diseases Abul K. Abbas UCSF Balancing lymphocyte activation and control Activation Effector T cells Tolerance Regulatory T cells

More information

Innate immune regulation of T-helper (Th) cell homeostasis in the intestine

Innate immune regulation of T-helper (Th) cell homeostasis in the intestine Innate immune regulation of T-helper (Th) cell homeostasis in the intestine Masayuki Fukata, MD, Ph.D. Research Scientist II Division of Gastroenterology, Department of Medicine, F. Widjaja Foundation,

More information

Immune Regulation and Tolerance

Immune Regulation and Tolerance Immune Regulation and Tolerance Immunoregulation: A balance between activation and suppression of effector cells to achieve an efficient immune response without damaging the host. Activation (immunity)

More information

Toll-like Receptors (TLRs): Biology, Pathology and Therapeutics

Toll-like Receptors (TLRs): Biology, Pathology and Therapeutics Toll-like Receptors (TLRs): Biology, Pathology and Therapeutics Dr Sarah Sasson SydPATH Registrar 23 rd June 2014 TLRs: Introduction Discovered in 1990s Recognise conserved structures in pathogens Rely

More information

Immunology Lecture 4. Clinical Relevance of the Immune System

Immunology Lecture 4. Clinical Relevance of the Immune System Immunology Lecture 4 The Well Patient: How innate and adaptive immune responses maintain health - 13, pg 169-181, 191-195. Immune Deficiency - 15 Autoimmunity - 16 Transplantation - 17, pg 260-270 Tumor

More information

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION Scott Abrams, Ph.D. Professor of Oncology, x4375 scott.abrams@roswellpark.org Kuby Immunology SEVENTH EDITION CHAPTER 11 T-Cell Activation, Differentiation, and Memory Copyright 2013 by W. H. Freeman and

More information

Effector T Cells and

Effector T Cells and 1 Effector T Cells and Cytokines Andrew Lichtman, MD PhD Brigham and Women's Hospital Harvard Medical School 2 Lecture outline Cytokines Subsets of CD4+ T cells: definitions, functions, development New

More information

Innate Immunity & Inflammation

Innate Immunity & Inflammation Innate Immunity & Inflammation The innate immune system is an evolutionally conserved mechanism that provides an early and effective response against invading microbial pathogens. It relies on a limited

More information

Innate Immunity. Chapter 3. Connection Between Innate and Adaptive Immunity. Know Differences and Provide Examples. Antimicrobial peptide psoriasin

Innate Immunity. Chapter 3. Connection Between Innate and Adaptive Immunity. Know Differences and Provide Examples. Antimicrobial peptide psoriasin Chapter Know Differences and Provide Examples Innate Immunity kin and Epithelial Barriers Antimicrobial peptide psoriasin -Activity against Gram (-) E. coli Connection Between Innate and Adaptive Immunity

More information

DNA vaccine, peripheral T-cell tolerance modulation 185

DNA vaccine, peripheral T-cell tolerance modulation 185 Subject Index Airway hyperresponsiveness (AHR) animal models 41 43 asthma inhibition 45 overview 41 mast cell modulation of T-cells 62 64 respiratory tolerance 40, 41 Tregs inhibition role 44 respiratory

More information

T cell maturation. T-cell Maturation. What allows T cell maturation?

T cell maturation. T-cell Maturation. What allows T cell maturation? T-cell Maturation What allows T cell maturation? Direct contact with thymic epithelial cells Influence of thymic hormones Growth factors (cytokines, CSF) T cell maturation T cell progenitor DN DP SP 2ry

More information

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins,

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins, Cytokines http://highered.mcgraw-hill.com/sites/0072507470/student_view0/chapter22/animation the_immune_response.html Cytokines modulate the functional activities of individual cells and tissues both under

More information

Lecture outline. Immunological tolerance and immune regulation. Central and peripheral tolerance. Inhibitory receptors of T cells. Regulatory T cells

Lecture outline. Immunological tolerance and immune regulation. Central and peripheral tolerance. Inhibitory receptors of T cells. Regulatory T cells 1 Immunological tolerance and immune regulation Abul K. Abbas UCSF 2 Lecture outline Central and peripheral tolerance Inhibitory receptors of T cells Regulatory T cells 1 The immunological equilibrium:

More information

Immunology in the Trauma Patient

Immunology in the Trauma Patient Immunology in the Trauma Patient Christine S. Cocanour, MD, FACS, FCCM I have no disclosures as it pertains to this presentation 1 Basic Immunology Danger Theory Immune system recognizes not just nonself

More information

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes: Interactions between innate immunity & adaptive immunity What happens to T cells after they leave the thymus? Naïve T cells exit the thymus and enter the bloodstream. If they remain in the bloodstream,

More information

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes: Interactions between innate immunity & adaptive immunity What happens to T cells after they leave the thymus? Naïve T cells exit the thymus and enter the bloodstream. If they remain in the bloodstream,

More information

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS

Tolerance 2. Regulatory T cells; why tolerance fails. Abul K. Abbas UCSF. FOCiS 1 Tolerance 2. Regulatory T cells; why tolerance fails Abul K. Abbas UCSF FOCiS 2 Lecture outline Regulatory T cells: functions and clinical relevance Pathogenesis of autoimmunity: why selftolerance fails

More information

Chapter 3 The Induced Responses of Innate Immunity

Chapter 3 The Induced Responses of Innate Immunity Chapter 3 The Induced Responses of Innate Immunity Pattern recognition by cells of the innate immune system Pattern recognition by cells of the innate immune system 4 main pattern recognition receptors

More information

Innate Immunity and the Paradoxes of Viral Pathogens and Tissue Injury in Gene Therapy

Innate Immunity and the Paradoxes of Viral Pathogens and Tissue Injury in Gene Therapy Innate Immunity and the Paradoxes of Viral Pathogens and Tissue Injury in Gene Therapy Daniel R. Salomon, M.D. Department of Molecular and Experimental Medicine The Scripps Research Institute Tissue and

More information

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells ICI Basic Immunology course Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells Abul K. Abbas, MD UCSF Stages in the development of T cell responses: induction

More information

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center General Overview of Immunology Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center Objectives Describe differences between innate and adaptive immune responses

More information

Innate Immunity: (I) Molecules & (II) Cells. Part II: Cells (aka the Sentinels)

Innate Immunity: (I) Molecules & (II) Cells. Part II: Cells (aka the Sentinels) Innate Immunity: (I) Molecules & (II) Cells Stephanie Eisenbarth, M.D., Ph.D. FOCIS Advanced Course 2/19/18 Department of Laboratory Medicine Yale School of Medicine Department of Immunobiology Yale School

More information

Innate Immunity. By Dr. Gouse Mohiddin Shaik

Innate Immunity. By Dr. Gouse Mohiddin Shaik Innate Immunity By Dr. Gouse Mohiddin Shaik Types of immunity Immunity Innate / inborn Non-specific Acquired / adaptive Specific 3rd line of defense Physical barriers Skin, Saliva, Mucous, Stomach acid,

More information

TRAUMA IS ONE of the leading causes of death worldwide.

TRAUMA IS ONE of the leading causes of death worldwide. Acute Medicine & Surgery 2014; 1: 63 69 doi: 10.1002/ams2.17 Mini Review Immune response to traumatic injury: harmony and discordance of immune system homeostasis Akinori Osuka, 1,2,3 Hiroshi Ogura, 2

More information

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS Choompone Sakonwasun, MD (Hons), FRCPT Types of Adaptive Immunity Types of T Cell-mediated Immune Reactions CTLs = cytotoxic T lymphocytes

More information

TOLL-LIKE RECEPTORS AND CYTOKINES IN SEPSIS

TOLL-LIKE RECEPTORS AND CYTOKINES IN SEPSIS TOLL-LIKE RECEPTORS AND CYTOKINES IN SEPSIS A/PROF WILLIAM SEWELL ST VINCENT S CLINICAL SCHOOL, UNSW SYDPATH, ST VINCENT S HOSPITAL SYDNEY GARVAN INSTITUTE INNATE VERSUS ADAPTIVE IMMUNE RESPONSES INNATE

More information

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION

Scott Abrams, Ph.D. Professor of Oncology, x4375 Kuby Immunology SEVENTH EDITION Scott Abrams, Ph.D. Professor of Oncology, x4375 scott.abrams@roswellpark.org Kuby Immunology SEVENTH EDITION CHAPTER 13 Effector Responses: Cell- and Antibody-Mediated Immunity Copyright 2013 by W. H.

More information

Adaptive immune responses: T cell-mediated immunity

Adaptive immune responses: T cell-mediated immunity MICR2209 Adaptive immune responses: T cell-mediated immunity Dr Allison Imrie allison.imrie@uwa.edu.au 1 Synopsis: In this lecture we will discuss the T-cell mediated immune response, how it is activated,

More information

Immunology Part II. Innate Immunity. 18. April 2018, Ruhr-Universität Bochum Marcus Peters,

Immunology Part II. Innate Immunity. 18. April 2018, Ruhr-Universität Bochum Marcus Peters, Immunology Part II Innate Immunity 18. April 2018, Ruhr-Universität Bochum Marcus Peters, marcus.peters@rub.de Conserved structures of pathogens PAMPs are detected by Pattern Recognition Receptors PRRs

More information

Animal Models to Understand Immunity

Animal Models to Understand Immunity Animal Models to Understand Immunity Hussein El Saghire hesaghir@sckcen.be Innate Adaptive immunity Immunity MAPK and NF-kB TLR pathways receptors Fast Slow Non-specific Specific NOD-like receptors T-cell

More information

Innate Immunity. Connection Between Innate and Adaptive Immunity. Know Differences and Provide Examples Chapter 3. Antimicrobial peptide psoriasin

Innate Immunity. Connection Between Innate and Adaptive Immunity. Know Differences and Provide Examples Chapter 3. Antimicrobial peptide psoriasin Know Differences and Provide Examples Chapter * Innate Immunity * kin and Epithelial Barriers * Antimicrobial peptide psoriasin -Activity against Gram (-) E. coli Connection Between Innate and Adaptive

More information

Immunological Tolerance

Immunological Tolerance Immunological Tolerance Introduction Definition: Unresponsiveness to an antigen that is induced by exposure to that antigen Tolerogen = tolerogenic antigen = antigen that induces tolerance Important for

More information

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell? Abbas Chapter 2: Sarah Spriet February 8, 2015 Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell? a. Dendritic cells b. Macrophages c. Monocytes

More information

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline 1 T Lymphocyte Activation and Costimulation Abul K. Abbas, MD UCSF FOCiS 2 Lecture outline T cell activation Costimulation, the B7:CD28 family Inhibitory receptors of T cells Targeting costimulators for

More information

Tolerance 2. Regulatory T cells; why tolerance fails. FOCiS. Lecture outline. Regulatory T cells. Regulatory T cells: functions and clinical relevance

Tolerance 2. Regulatory T cells; why tolerance fails. FOCiS. Lecture outline. Regulatory T cells. Regulatory T cells: functions and clinical relevance 1 Tolerance 2. Regulatory T cells; why tolerance fails Abul K. Abbas UCSF FOCiS 2 Lecture outline Regulatory T cells: functions and clinical relevance Pathogenesis of autoimmunity: why selftolerance fails

More information

Central tolerance. Mechanisms of Immune Tolerance. Regulation of the T cell response

Central tolerance. Mechanisms of Immune Tolerance. Regulation of the T cell response Immunoregulation: A balance between activation and suppression that achieves an efficient immune response without damaging the host. Mechanisms of Immune Tolerance ACTIVATION (immunity) SUPPRESSION (tolerance)

More information

Mechanisms of Immune Tolerance

Mechanisms of Immune Tolerance Immunoregulation: A balance between activation and suppression that achieves an efficient immune response without damaging the host. ACTIVATION (immunity) SUPPRESSION (tolerance) Autoimmunity Immunodeficiency

More information

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco Determinants of Immunogenicity and Tolerance Abul K. Abbas, MD Department of Pathology University of California San Francisco EIP Symposium Feb 2016 Why do some people respond to therapeutic proteins?

More information

Innate Immunity: (I) Molecules & (II) Cells

Innate Immunity: (I) Molecules & (II) Cells Innate Immunity: (I) Molecules & (II) Cells Stephanie Eisenbarth, M.D., Ph.D. FOCIS Advanced Course 2/19/18 Department of Laboratory Medicine Yale School of Medicine Department of Immunobiology Yale School

More information

Writing Effective Grant Proposals

Writing Effective Grant Proposals WritingEffectiveGrantProposals SUPPLEMENTALHANDOUT EXERCISES (toaccompanypowerpointslidepresentation) PamelaDerish ScientificPublicationsManager DepartmentofSurgery,UCSF tel415.885 7686 Pamela.Derish@ucsfmedctr.org

More information

There are 2 major lines of defense: Non-specific (Innate Immunity) and. Specific. (Adaptive Immunity) Photo of macrophage cell

There are 2 major lines of defense: Non-specific (Innate Immunity) and. Specific. (Adaptive Immunity) Photo of macrophage cell There are 2 major lines of defense: Non-specific (Innate Immunity) and Specific (Adaptive Immunity) Photo of macrophage cell Development of the Immune System ery pl neu mφ nk CD8 + CTL CD4 + thy TH1 mye

More information

Inflammation: How to Cool the Fire Inside your Gut? REINVENTING DIAGNOSTICS

Inflammation: How to Cool the Fire Inside your Gut? REINVENTING DIAGNOSTICS Inflammation: How to Cool the Fire Inside your Gut? REINVENTING DIAGNOSTICS Future of Healthcare REINVENTING DIAGNOSTICS Inflammation Gut Inflammation Basis of a Healthy

More information

Autoimmunity. Autoimmunity arises because of defects in central or peripheral tolerance of lymphocytes to selfantigens

Autoimmunity. Autoimmunity arises because of defects in central or peripheral tolerance of lymphocytes to selfantigens Autoimmunity Autoimmunity arises because of defects in central or peripheral tolerance of lymphocytes to selfantigens Autoimmune disease can be caused to primary defects in B cells, T cells and possibly

More information

The Innate Immune Response

The Innate Immune Response The Innate Immune Response FUNCTIONS OF THE IMMUNE SYSTEM: Recognize, destroy and clear a diversity of pathogens. Initiate tissue and wound healing processes. Recognize and clear damaged self components.

More information

1. Overview of Adaptive Immunity

1. Overview of Adaptive Immunity Chapter 17A: Adaptive Immunity Part I 1. Overview of Adaptive Immunity 2. T and B Cell Production 3. Antigens & Antigen Presentation 4. Helper T cells 1. Overview of Adaptive Immunity The Nature of Adaptive

More information

Pathophysiologic Basis of Autoimmune Disorders

Pathophysiologic Basis of Autoimmune Disorders Pathophysiologic Basis of Autoimmune Disorders Linda Felver, Ph.D., R.N. Associate Professor School of Nursing Oregon Health & Science University The immune system has two arms: Adaptive (Acquired) Immune

More information

Journal club. Lama Nazzal

Journal club. Lama Nazzal Journal club Lama Nazzal Background Kidney stone disease affects about 12% of men and 5% of women during their lifetimes in the United States Intrarenal nephrocalcinosis is often asymptomatic, but can

More information

Immunology for the Rheumatologist

Immunology for the Rheumatologist Immunology for the Rheumatologist Rheumatologists frequently deal with the immune system gone awry, rarely studying normal immunology. This program is an overview and discussion of the function of the

More information

Role of Innate Immunity in Control of Adaptive Immunity

Role of Innate Immunity in Control of Adaptive Immunity Role of Innate Immunity in Control of Adaptive Immunity Innate Immunity The burden of pathogen sensing is placed on the innate immune system Danger hypothesis Missing Self Based on the detection of molecular

More information

LESSON 2: THE ADAPTIVE IMMUNITY

LESSON 2: THE ADAPTIVE IMMUNITY Introduction to immunology. LESSON 2: THE ADAPTIVE IMMUNITY Today we will get to know: The adaptive immunity T- and B-cells Antigens and their recognition How T-cells work 1 The adaptive immunity Unlike

More information

FOCiS. Lecture outline. The immunological equilibrium: balancing lymphocyte activation and control. Immunological tolerance and immune regulation -- 1

FOCiS. Lecture outline. The immunological equilibrium: balancing lymphocyte activation and control. Immunological tolerance and immune regulation -- 1 1 Immunological tolerance and immune regulation -- 1 Abul K. Abbas UCSF FOCiS 2 Lecture outline Principles of immune regulation Self-tolerance; mechanisms of central and peripheral tolerance Inhibitory

More information

Objectives. Abbas Chapter 11: Immunological Tolerance. Question 1. Question 2. Question 3. Definitions

Objectives. Abbas Chapter 11: Immunological Tolerance. Question 1. Question 2. Question 3. Definitions Objectives Abbas Chapter 11: Immunological Tolerance Christina Ciaccio, MD Children s Mercy Hospitals and Clinics February 1, 2010 To introduce the concept of immunologic tolerance To understand what factors

More information

T Cell Activation, Costimulation and Regulation

T Cell Activation, Costimulation and Regulation 1 T Cell Activation, Costimulation and Regulation Abul K. Abbas, MD University of California San Francisco 2 Lecture outline T cell antigen recognition and activation Costimulation, the B7:CD28 family

More information

Darwinian selection and Newtonian physics wrapped up in systems biology

Darwinian selection and Newtonian physics wrapped up in systems biology Darwinian selection and Newtonian physics wrapped up in systems biology Concept published in 1957* by Macfarland Burnet (1960 Nobel Laureate for the theory of induced immune tolerance, leading to solid

More information

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS 1 Antigen Presentation and T Lymphocyte Activation Abul K. Abbas UCSF FOCiS 2 Lecture outline Dendritic cells and antigen presentation The role of the MHC T cell activation Costimulation, the B7:CD28 family

More information

This is a free sample of content from Immune Tolerance. Click here for more information or to buy the book.

This is a free sample of content from Immune Tolerance. Click here for more information or to buy the book. A ACPAs. See Antibodies to citrullinated peptide antigens Activation-induced cell death (AICD), 25 AICD. See Activation-induced cell death AIRE, 3, 18 19, 24, 88, 103 104 AKT, 35 Alefacept, 136 ALPS. See

More information

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY

ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY ACTIVATION OF T LYMPHOCYTES AND CELL MEDIATED IMMUNITY The recognition of specific antigen by naïve T cell induces its own activation and effector phases. T helper cells recognize peptide antigens through

More information

T Cell Effector Mechanisms I: B cell Help & DTH

T Cell Effector Mechanisms I: B cell Help & DTH T Cell Effector Mechanisms I: B cell Help & DTH Ned Braunstein, MD The Major T Cell Subsets p56 lck + T cells γ δ ε ζ ζ p56 lck CD8+ T cells γ δ ε ζ ζ Cα Cβ Vα Vβ CD3 CD8 Cα Cβ Vα Vβ CD3 MHC II peptide

More information

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology Code : AS-2246 M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology A. Select one correct option for each of the following questions:- 2X10=10 1. (b)

More information

The Adaptive Immune Responses

The Adaptive Immune Responses The Adaptive Immune Responses The two arms of the immune responses are; 1) the cell mediated, and 2) the humoral responses. In this chapter we will discuss the two responses in detail and we will start

More information

Innate Immunity. Hathairat Thananchai, DPhil Department of Microbiology Faculty of Medicine Chiang Mai University 2 August 2016

Innate Immunity. Hathairat Thananchai, DPhil Department of Microbiology Faculty of Medicine Chiang Mai University 2 August 2016 Innate Immunity Hathairat Thananchai, DPhil Department of Microbiology Faculty of Medicine Chiang Mai University 2 August 2016 Objectives: Explain how innate immune system recognizes foreign substances

More information

Introduction to Immune System

Introduction to Immune System Introduction to Immune System Learning outcome You will be able to understand, at a fundamental level, the STRUCTURES and FUNCTIONS of cell surface and soluble molecules involved in recognition of foreign

More information

Newly Recognized Components of the Innate Immune System

Newly Recognized Components of the Innate Immune System Newly Recognized Components of the Innate Immune System NOD Proteins: Intracellular Peptidoglycan Sensors NOD-1 NOD-2 Nod Protein LRR; Ligand Recognition CARD RICK I-κB p50 p65 NF-κB Polymorphisms in Nod-2

More information

Basis of Immunology and

Basis of Immunology and Basis of Immunology and Immunophysiopathology of Infectious Diseases Jointly organized by Institut Pasteur in Ho Chi Minh City and Institut Pasteur with kind support from ANRS & Université Pierre et Marie

More information

Chapter 13: Cytokines

Chapter 13: Cytokines Chapter 13: Cytokines Definition: secreted, low-molecular-weight proteins that regulate the nature, intensity and duration of the immune response by exerting a variety of effects on lymphocytes and/or

More information

Toll-like Receptor Signaling

Toll-like Receptor Signaling Toll-like Receptor Signaling 1 Professor of Medicine University of Massachusetts Medical School, Worcester, MA, USA Why do we need innate immunity? Pathogens multiply very fast We literally swim in viruses

More information

Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D.

Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D. Regulation of anti-tumor immunity through migration of immune cell subsets within the tumor microenvironment Thomas F. Gajewski, M.D., Ph.D. Professor, Departments of Pathology and Medicine Program Leader,

More information

Advances in Cancer Immunotherapy

Advances in Cancer Immunotherapy Advances in Cancer Immunotherapy Immunology 101 for the Non-Immunologist Arnold H. Zea, PhD azea@lsuhsc.edu Disclosures No relevant financial relationships to disclose This presentation does not contain

More information

Long-term innate immune memory via effects on bone marrow progenitors

Long-term innate immune memory via effects on bone marrow progenitors Long-term innate immune memory via effects on bone marrow progenitors Helen S Goodridge, PhD helen.goodridge@csmc.edu Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, USA Fondation

More information

Innate Immunity II. Integration. Lindsay Nicholson Advanced Immunology L2

Innate Immunity II. Integration. Lindsay Nicholson Advanced Immunology L2 Innate Immunity II Integration Lindsay Nicholson Advanced Immunology L2 l.nicholson@bristol.ac.uk Lecture 1 Defining Innate Immunity Recognition and effector mechanisms (I) Lecture 2 Recognition and effector

More information

Commensal Bacteria, Toll-like Receptors and Intestinal Injury. Journal Club December 16, 2004

Commensal Bacteria, Toll-like Receptors and Intestinal Injury. Journal Club December 16, 2004 Commensal Bacteria, Toll-like Receptors and Intestinal Injury Journal Club December 16, 2004 Gut-Commensal Interactions Nutrient metabolism Tissue development Resistance to colonization with pathogens

More information

Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells

Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells Immunology Basics Relevant to Cancer Immunotherapy: T Cell Activation, Costimulation, and Effector T Cells Andrew H. Lichtman, M.D. Ph.D. Department of Pathology Brigham and Women s Hospital and Harvard

More information

Immunity and Infection. Chapter 17

Immunity and Infection. Chapter 17 Immunity and Infection Chapter 17 The Chain of Infection Transmitted through a chain of infection (six links) Pathogen: Disease causing microorganism Reservoir: Natural environment of the pathogen Portal

More information

Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) Spring 2009 Semester

Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) Spring 2009 Semester Micr-6005, Current Concepts of Immunology (Rutgers course number: 16:681:543) (3 Credits) Spring 2009 Semester Course Director: (732-235-4501, ) Please note that this course is offered once every 2 years.

More information

CELL BIOLOGY - CLUTCH CH THE IMMUNE SYSTEM.

CELL BIOLOGY - CLUTCH CH THE IMMUNE SYSTEM. !! www.clutchprep.com CONCEPT: OVERVIEW OF HOST DEFENSES The human body contains three lines of against infectious agents (pathogens) 1. Mechanical and chemical boundaries (part of the innate immune system)

More information

Immune Tolerance. Kyeong Cheon Jung. Department of Pathology Seoul National University College of Medicine

Immune Tolerance. Kyeong Cheon Jung. Department of Pathology Seoul National University College of Medicine Immune Tolerance Kyeong Cheon Jung Department of Pathology Seoul National University College of Medicine Immune tolerance Unresponsiveness to an antigen that is induced by previous exposure to that antigen

More information

How the Innate Immune System Profiles Pathogens

How the Innate Immune System Profiles Pathogens How the Innate Immune System Profiles Pathogens Receptors on macrophages, neutrophils, dendritic cells for bacteria and viruses Broad specificity - Two main groups of bacteria: gram positive, gram-negative

More information

Allergy and Immunology Review Corner: Chapter 13 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti, MD.

Allergy and Immunology Review Corner: Chapter 13 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti, MD. Allergy and Immunology Review Corner: Chapter 13 of Immunology IV: Clinical Applications in Health and Disease, by Joseph A. Bellanti, MD. Chapter 13: Mechanisms of Immunity to Viral Disease Prepared by

More information

Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel:

Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel: Cytokines (II) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel: 4677363 aalshamsan@ksu.edu.sa Learning Objectives By the end of this lecture you will be able to: 1 Understand the physiological

More information

Chapter 22: The Lymphatic System and Immunity

Chapter 22: The Lymphatic System and Immunity Bio40C schedule Lecture Immune system Lab Quiz 2 this week; bring a scantron! Study guide on my website (see lab assignments) Extra credit Critical thinking questions at end of chapters 5 pts/chapter Due

More information

Chapter 1. Chapter 1 Concepts. MCMP422 Immunology and Biologics Immunology is important personally and professionally!

Chapter 1. Chapter 1 Concepts. MCMP422 Immunology and Biologics Immunology is important personally and professionally! MCMP422 Immunology and Biologics Immunology is important personally and professionally! Learn the language - use the glossary and index RNR - Reading, Note taking, Reviewing All materials in Chapters 1-3

More information

Self-tolerance. Lack of immune responsiveness to an individual s own tissue antigens. Central Tolerance. Peripheral tolerance

Self-tolerance. Lack of immune responsiveness to an individual s own tissue antigens. Central Tolerance. Peripheral tolerance Autoimmunity Self-tolerance Lack of immune responsiveness to an individual s own tissue antigens Central Tolerance Peripheral tolerance Factors Regulating Immune Response Antigen availability Properties

More information

Synergistic effects of indoleamine 2,3-dioxygenase (IDO) inhibitor drugs and chemotherapy in melanoma.

Synergistic effects of indoleamine 2,3-dioxygenase (IDO) inhibitor drugs and chemotherapy in melanoma. Synergistic effects of indoleamine 2,3-dioxygenase (IDO) inhibitor drugs and chemotherapy in melanoma. Madhav D. Sharma Cancer Immunology, Inflammation, and Tolerance Program, Department of Pediatrics,

More information

Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School

Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School CTLs, Natural Killers and NKTs 1 Shiv Pillai Ragon Institute, Massachusetts General Hospital Harvard Medical School CTL inducing tumor apoptosis 3 Lecture outline CD8 + Cytotoxic T lymphocytes (CTL) Activation/differentiation

More information

7/6/2009. The study of the immune system and of diseases that occur as a result of inappropriate or inadequate actions of the immune system.

7/6/2009. The study of the immune system and of diseases that occur as a result of inappropriate or inadequate actions of the immune system. Diseases of Immunity 2009 CL Davis General Pathology Paul W. Snyder, DVM, PhD Purdue University Acknowledgements Pathologic Basis of Veterinary Disease, 4 th Ed Veterinary Immunology, An Introduction 8

More information

The Immune System: Innate and Adaptive Body Defenses Outline PART 1: INNATE DEFENSES 21.1 Surface barriers act as the first line of defense to keep

The Immune System: Innate and Adaptive Body Defenses Outline PART 1: INNATE DEFENSES 21.1 Surface barriers act as the first line of defense to keep The Immune System: Innate and Adaptive Body Defenses Outline PART 1: INNATE DEFENSES 21.1 Surface barriers act as the first line of defense to keep invaders out of the body (pp. 772 773; Fig. 21.1; Table

More information

Immune response to infection

Immune response to infection Immune response to infection Dr. Sandra Nitsche (Sandra.Nitsche@rub.de ) 20.06.2018 1 Course of acute infection Typical acute infection that is cleared by an adaptive immune reaction 1. invasion of pathogen

More information

Adaptive Immunity. Jeffrey K. Actor, Ph.D. MSB 2.214,

Adaptive Immunity. Jeffrey K. Actor, Ph.D. MSB 2.214, Adaptive Immunity Jeffrey K. Actor, Ph.D. MSB 2.214, 500-5344 Lecture Objectives: Understand role of various molecules including cytokines, chemokines, costimulatory and adhesion molecules in the development

More information

All animals have innate immunity, a defense active immediately upon infection Vertebrates also have adaptive immunity

All animals have innate immunity, a defense active immediately upon infection Vertebrates also have adaptive immunity 1 2 3 4 5 6 7 8 9 The Immune System All animals have innate immunity, a defense active immediately upon infection Vertebrates also have adaptive immunity Figure 43.2 In innate immunity, recognition and

More information

Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant

Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant Reporting Period July 1, 2012 June 30, 2013 Formula Grant Overview The Children's Hospital of Pittsburgh received $228,401 in

More information

Cellular Pathology of immunological disorders

Cellular Pathology of immunological disorders Cellular Pathology of immunological disorders SCBM344 Cellular and Molecular Pathology Witchuda Payuhakrit, Ph.D (Pathobiology) witchuda.pay@mahidol.ac.th Objectives Describe the etiology of immunological

More information

Structure and Function of Antigen Recognition Molecules

Structure and Function of Antigen Recognition Molecules MICR2209 Structure and Function of Antigen Recognition Molecules Dr Allison Imrie allison.imrie@uwa.edu.au 1 Synopsis: In this lecture we will examine the major receptors used by cells of the innate and

More information

Lecture on Innate Immunity and Inflammation. Innate Immunity: An Evolutionary View

Lecture on Innate Immunity and Inflammation. Innate Immunity: An Evolutionary View Lecture on Innate Immunity and Inflammation Evolutionary View Epithelial barriers to infection Four main types of innate recognition molecules:tlrs, CLRs, NLRs, RLRs NF-κB, the master transcriptional regulator

More information

Genetics. Environment. You Are Only 10% Human. Pathogenesis of IBD. Advances in the Pathogenesis of IBD: Genetics Leads to Function IBD

Genetics. Environment. You Are Only 10% Human. Pathogenesis of IBD. Advances in the Pathogenesis of IBD: Genetics Leads to Function IBD Advances in the Pathogenesis of IBD: Genetics Leads to Function Pathogenesis of IBD Environmental Factors Microbes Scott Plevy, MD Associate Professor of Medicine, Microbiology & Immunology UNC School

More information

Immunology. T-Lymphocytes. 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters,

Immunology. T-Lymphocytes. 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters, Immunology T-Lymphocytes 16. Oktober 2014, Ruhr-Universität Bochum Karin Peters, karin.peters@rub.de The role of T-effector cells in the immune response against microbes cellular immunity humoral immunity

More information

The development of T cells in the thymus

The development of T cells in the thymus T cells rearrange their receptors in the thymus whereas B cells do so in the bone marrow. The development of T cells in the thymus The lobular/cellular organization of the thymus Immature cells are called

More information

Identification of Microbes

Identification of Microbes Identification of Microbes Recognition by PRR (pattern recognition receptors) Recognize conserved molecular patterns on microbes called pathogen associated molecular patterns (PAMPs) which are not present

More information

SPECIFIC AIMS. II year (1st semester)

SPECIFIC AIMS. II year (1st semester) II year (1st semester) Scientific Field IMMUNOLOGY AND IMMUNOPATHOLOGY TUTOR ECTS MALISAN F. COORDINATOR MED/04 Immunology and Immunopathology Malisan Florence 5 MED/04 Immunology and Immunopathology Testi

More information