首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 109 毫秒
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
Prnp−/− mice lack the prion protein PrPC and are resistant to prion infections, but variable phenotypes have been reported in Prnp−/− mice and the physiological function of PrPC remains poorly understood. Here we examined a cell-autonomous phenotype, inhibition of macrophage phagocytosis of apoptotic cells, previously reported in Prnp−/− mice. Using formal genetic, genomic, and immunological analyses, we found that the regulation of phagocytosis previously ascribed to PrPC is instead controlled by a linked locus encoding the signal regulatory protein α (Sirpa). These findings indicate that control of phagocytosis was previously misattributed to the prion protein and illustrate the requirement for stringent approaches to eliminate confounding effects of flanking genes in studies modeling human disease in gene-targeted mice. The plethora of seemingly unrelated functions attributed to PrPC suggests that additional phenotypes reported in Prnp−/− mice may actually relate to Sirpa or other genetic confounders.The cellular prion protein PrPC, encoded by the Prnp gene, is tethered to the membrane of most mammalian cells by a glycosylphosphatidylinositol anchor. Conversion and aggregation of PrPC into a misfolded conformer (termed PrPSc) triggers transmissible spongiform encephalopathies, also termed prion diseases (Aguzzi and Calella, 2009). Disparate functions have been ascribed to PrPC on the basis of phenotypes described in Prnp−/− mice (Steele et al., 2007; Linden et al., 2008), yet none of these functions has been clarified mechanistically, and their validity was frequently challenged.All currently available Prnp−/− lines were generated using embryonic stem (ES) cells derived from the 129 strain of Mus musculus. Typically, chimeric founder mice were crossed with WT (Prnpwt/wt) mice of the C57BL/6 strain (B6; Sparkes et al., 1986). Consequently, congenic Prnpwt/wt and Prnp−/− mice may differ at additional polymorphic loci (Smithies and Maeda, 1995; Gerlai, 1996). We hypothesized that co-segregation of linked genes may have confounded the attribution of functions to PrPC based on phenotypes observed in Prnp−/− mice (Collinge et al., 1994; Lledo et al., 1996; Walz et al., 1999; Rangel et al., 2007; Laurén et al., 2009; Calella et al., 2010; Gimbel et al., 2010; Ratté et al., 2011; Striebel et al., 2013).

Table 1.

Prnp KO mouse lines analyzed in this study
Prnp KO mouse lineES cellsOrigin of ES cellsStrain of partner of chimeric mouseLocation of colonyReference
PrnpZrchI/ZrchIAB1129S7/SvEvBrdB6Zurich, SwitzerlandBüeler et al. (1992)
PrnpNgsk/NgskJ1129S4/SvJaeB6Nagasaki, JapanSakaguchi et al. (1995)
PrnpEdbg/EdbgE14129/Ola129/OlaEdinburgh, Scotland, UKManson et al. (1994)
PrnpGFP/GFPHM-1129/OlaB6Cambridge, MAHeikenwalder et al. (2008)
PrnpRkn/RknE14129P2/OlaHsdB6Wako-shi, JapanYokoyama et al. (2001)
PrnpZrchII/ZrchIIE14.1129/OlaHsdB6Zurich, SwitzerlandRossi et al. (2001)
PrnpRcm0/Rcm0HM-1129/Ola129/OlaEdinburgh, Scotland, UKMoore et al. (1995)
Open in a separate windowThe present study is based on the analysis of mice carrying seven independently generated Prnp-null alleles. PrnpEdbg/Edbg and PrnpRcm0/Rcm0 were always crossed to isogenic 129/Ola mice, whereas all other Prnp−/− mice were crossed to B6 mice and then kept on a mixed B6 and 129 background or further backcrossed to B6 or other strains.Here we selected a cell-autonomous phenotype previously reported in congenic B6.129-PrnpZrchI/ZrchI mice (Büeler et al., 1992): inhibition of phagocytosis of apoptotic cells (de Almeida et al., 2005). We used RNA sequencing to identify genes linked to Prnp and expressed in macrophages that may influence this phenotype. We report genetic and functional evidence that the regulation of phagocytosis previously ascribed to Prnp−/− is instead controlled by the closely linked gene signal regulatory protein α (Sirpa; Matozaki et al., 2009).  相似文献   

11.
12.
13.
Pathogen- and damage-associated molecular patterns are sensed by the immune system’s pattern recognition receptors (PRRs) upon contact with a microbe or damaged tissue. In situations such as contact with commensals or during physiological cell death, the immune system should not respond to these patterns. Hence, immune responses need to be context dependent, but it is not clear how context for molecular pattern recognition is provided. We discuss inhibitory receptors as potential counterparts to activating pattern recognition receptors. We propose a group of inhibitory pattern recognition receptors (iPRRs) that recognize endogenous and microbial patterns associated with danger, homeostasis, or both. We propose that recognition of molecular patterns by iPRRs provides context, helps mediate tolerance to microbes, and helps balance responses to danger signals.

Pattern recognition receptors (PRRs) recognize molecular patternsThe immune system needs to recognize and correct deviations from normal physiology, such as harmful contact with a microbe, disruption and damage of healthy tissue, and malignant transformation of cells. To sense the presence of microbes, the immune system employs a set of PRRs (Janeway, 1989). At present, five classes of PRRs have been defined: the TLRs and the C-type lectin receptors, which are both localized to cell or endosomal membranes; the cytoplasmic NOD-like receptors and RIG-I–like receptors; and additional cytoplasmic DNA sensors, such as cyclic GMP-AMP synthase (Gong et al., 2020; Takeuchi and Akira, 2010). PRRs recognize highly conserved components of microbes, termed pathogen-associated molecular patterns (PAMPs; Akira et al., 2006; Medzhitov and Janeway, 2002). In addition, PRRs sense endogenous molecules associated with damaged and dying cells termed danger- or damage-associated molecular patterns (DAMPs). Many factors are currently considered DAMPs, among which are S100 proteins, heat shock proteins (Hsps), high mobility group box 1 protein (HMGB1), and different glycans such as heparan sulfate (Chen and Nuñez, 2010; Matzinger, 1994; Matzinger, 2002).The self–nonself model of microbe recognition, first introduced by Frank Macfarlane Burnet and later refined by Charles Janeway, explains how the innate immune system recognizes pathogens through molecular patterns (Burnet, 1959; Janeway, 1989). Because pathogens constantly evolve, they cannot be recognized individually, as this would require an infinite number of receptors. To circumvent this problem, the immune system recognizes components of microbial cells that are highly conserved (but not identical) among microbes and cannot be subject to quick change or removal by the microbe because they are essential for its survival (Bianchi and Manfredi, 2009). These groups of structurally similar molecules are called PAMPs. One of the first PAMPs to be discovered was LPS of Gram-negative bacteria, which is detected by TLR4, providing activating signals that drive adaptive immunity (Medzhitov et al., 1997; Poltorak et al., 1998). Soon after, many additional PAMPs were discovered, such as the lipoteichoic acid (LTA) of Gram-positive bacteria (Schwandner et al., 1999). Later, Polly Matzinger extended the family of “molecular patterns” by presenting the danger theory of immunity, introducing DAMPs. The term DAMP has since been used in the literature to denote both damage- and danger-associated molecular patterns. Unlike PAMPs, DAMPs are not defined structurally, and there is (following Janeway’s argument) little need for that: there are only a finite number of host molecules. Instead, DAMPs are defined contextually: they signal danger, and what is dangerous in one place is not necessarily dangerous in another. Such a model is not easily addressed experimentally because of this elusive definition of danger (Pradeu and Cooper, 2012). As highlighted by Pradeu and Cooper (2012), Matzinger later clarified that while the model is theoretical, the idea behind it is that the immune system responds to damage (Matzinger, 2002), and damage signals are much easier to define than danger signals. Since then, many more groups of molecular patterns have been put forward, among which are resolution-, metabolism-, commensal-, and homeostasis-associated molecular patterns (HAMPs; Cario et al., 2002; Greslehner, 2020; Li et al., 2019; Shields et al., 2011; Wang et al., 2020). Under the term molecular pattern, we now classify groups of molecules that signal the occurrence of a particular event, that elicit similar effects, and that may share common structural features.Immune responses are context dependentThe same molecular pattern does not always evoke the same response. Different microbes inevitably colonize barrier tissues such as the skin and gastrointestinal tract, and most of them are not harmful or even provide benefit to the host, yet still express PAMPs. Similarly, while tissue damage and cell death can be pathologic, cell death can also be part of normal physiology and tissue renewal. To distinguish harmless from potentially harmful circumstances, the immune system must correctly interpret the activating signals molecular patterns are delivering, and therefore the threshold for immune system activation needs to vary by context. Tissues that are highly exposed to microbes, such as the gut and skin, require a high activation threshold to tolerate most microbes, whereas in the circulation, a low activation threshold is required to respond to all microbes (Fig. 1). Furthermore, not all tissues can tolerate tissue damage to the same extent. In situations where inflammatory responses result in more damage to the organism than the disturbance itself, not responding to disturbances is the best strategy (Medzhitov et al., 2012). Following this argument, the threshold for immune activation needs to be higher in organs with low regenerative capacity, such as the heart or brain, where an inflammatory response can lead to detrimental consequences, versus organs with a high regenerative capacity, such as the liver (Fig. 1). Hence, the immune response needs to be context dependent, and it is not clear how context for molecular pattern recognition is provided.Open in a separate windowFigure 1.The optimal threshold for activation is context dependent. The required threshold for activation of immune cells differs per location and depends on (1) the tolerance of the organ for immune pathology and (2) the tolerance to microbial exposure. Organs with a high regenerative capacity, such as the liver, are more able to deal with immunopathology than organs with low regenerative capacity, such as the heart or the brain. The gut and skin are constantly exposed to microbes, most of which are harmless or beneficial and should be tolerated. The eye can tolerate a certain amount of microbial exposure, and the cost of responding to a microbial stimulus will be high, so a high threshold will ensure the response occurs only when needed. In different organs, either tolerance for microbes or tolerance for immunopathology may be more important in determining the optimal threshold for activation.Immune inhibitory receptors dampen immune system activationImmune inhibitory receptors are germline-encoded innate receptors relaying inhibitory signals to immune cells. Much about their functioning has been learned by studying programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte protein 4 (CTLA-4), and killer cell Ig-like inhibitory receptors on NK cells (Long, 2008; Ravetch and Lanier, 2000; Rowshanravan et al., 2018). Inhibitory receptors attenuate activating signals coming from activating receptors and fine-tune the level of activation of an immune cell. Most of them relay the inhibitory signals via one or more immunoreceptor tyrosine-based inhibitory motifs (ITIMs) present in their cytoplasmic tails. ITIMs have the consensus sequence V/L/I/SxYxxV/L/I (Vivier and Daëron, 1997). When immune inhibitory receptors are activated by their ligands, the ITIMs recruit tyrosine phosphatases, which dephosphorylate the cytoplasmic tails of activating receptors or key molecules in their signaling pathways (Coxon et al., 2017; Gergely et al., 1999). The ligands for many inhibitory receptors are still unknown, while some single-molecule ligands have been identified for others. We previously argued that immune inhibitory receptors regulate immune responses in different ways. They may set a threshold for immune cell activation by preventing activating receptor signaling in certain contexts or dampen activating receptor signaling after it has already happened. The mode of action of any inhibitory receptor depends on the expression pattern of the receptor and the availability of its ligand (Rumpret et al., 2020). By providing an inhibitory signal, inhibitory receptors give additional information on the context in which an activating signal is sensed, thereby adjusting the immune response to the specific situation.Some activating PRRs, under specific circumstances, can also demonstrate inhibitory functions. For example, TLR4 signaling from the cell membrane typically evokes proinflammatory responses, while TLR4 signaling from the endosome also triggers antiinflammatory responses (Kagan, 2012; Siegemund and Sauer, 2012). Here, we discuss the concept of inhibitory pattern recognition receptors (iPRRs). We specifically focus on canonical inhibitory receptors that use ITIM-dependent inhibitory signaling pathways to relay their signals, resulting in inhibitory functions. We define a group of immune inhibitory receptors that recognize DAMPs, HAMPs, and PAMPs and classify these inhibitory receptors as iPRRs. We propose that, just like most activating PRRs (Gong et al., 2020), most iPRRs recognize both microbial and endogenous patterns (Fig. 2). We propose that iPRRs constitute the inhibitory counterparts of activating PRRs and provide context to the activating signals coming from activating PRRs.Open in a separate windowFigure 2.iPRRs and their endogenous and microbial ligands. The currently known group of iPRRs consist of CD300a/f, Siglecs 2, 3, and 5–11, CEACAM1, LILRB1 and LILRB3, TIGIT, poliovirus receptor (PVR), LAIR-1, and SIRL-1. The upper part of the figure displays endogenous ligands, and the bottom part displays the microbial ligands for iPRRs. For most receptors, both endogenous and exogenous ligands have been identified. Protein ligands are depicted as rectangles, lipids as circles, and carbohydrates as hexagons. All inhibitory receptors depicted are composed of Ig domains, and the number of Ig domains is schematically depicted for each receptor. In humans, most of these receptors are located in the chromosomal region 19q13, except CD300a/f (17q25) and TIGIT (3q13). *, LTA is a ligand for the mouse orthologue of the human LILRB3. PSM, phenol-soluble modulin; S100s, S100 proteins; SIA, sialic acid.iPRRs recognize DAMPsUpon the occurrence of damaged or dying cells, different DAMPs can arise and promote inflammation, leading to tissue repair but also immunopathology (Gong et al., 2020). Multiple inhibitory receptors could potentially tune DAMP-induced inflammatory responses (Fig. 2; Arnold et al., 2013; Brewer et al., 2019; Carlin et al., 2007; Chang et al., 2014; Chen et al., 2009; Choi et al., 2011; Conners et al., 2008; Fong et al., 2015; Gur et al., 2015; Gur et al., 2019; Jones et al., 2016; Klaile et al., 2017; Königer et al., 2016; Korotkova et al., 2008; Kumawat et al., 2019; Lebbink et al., 2009; Macauley et al., 2014; Nakayama et al., 2012; Rumpret et al., 2021a; Rumpret et al., 2021b; Simhadri et al., 2012; van Sorge et al., 2021; Virji et al., 1996; Yu et al., 2009). The sialic acid–binding Ig-like lectin (Siglec)-10–CD24 complex recognizes HMGB1, Hsp70, and Hsp90 and limits the immune response to damaged cells (Chen et al., 2009). It thereby limits harmful inflammatory responses in conditions such as sepsis (Chen et al., 2011), infection (Chen et al., 2013), and liver damage. Indeed, CD24−/− mice die of sublethal doses of acetaminophen-induced liver injury (Chen et al., 2009). Siglec-5 recognizes Hsp70 and delivers antiinflammatory signals to monocytes, which results in decreased production of TNFα and IL-8 in cells stimulated with LPS (Fong et al., 2015). Similarly, CD85j (leukocyte Ig-like receptor subfamily B member 1 [LILRB1]; Arnold et al., 2013) and signal inhibitory receptor on leukocytes 1 (SIRL-1; Rumpret et al., 2021a) recognize S100 proteins, another group of prototypical DAMPs. Blocking SIRL-1 enhances S100-induced release of reactive oxygen species in human neutrophils (Rumpret et al., 2021a). SIRL-1 additionally recognizes another DAMP, the antimicrobial peptide LL-37 (Rumpret et al., 2021b). LILRB3 recognizes a cytokeratin-associated protein, a cytoskeleton protein that is exposed in the extracellular environment after necrotic cell death and is recognized by the activating receptor LILRA6 (Jones et al., 2016). Thus, several iPRRs recognize DAMPs.Die. Where? How?Cells can die in either an immunologically silent manner (apoptosis) or an immunogenic and proinflammatory manner; the latter can be a controlled process (such as necroptosis and pyroptosis) or an uncontrolled process (necrosis). Apoptotic cells are recognized, engulfed by phagocytes, and degraded intracellularly. In contrast, membranes of cells that die via immunogenic cell death (ICD) are ruptured, and intracellular components are released into the local microenvironment, many of which are regarded as DAMPs by neighboring cells (Bedoui et al., 2020). Interestingly, the type of ICD may determine which type of DAMP is released. This is illustrated by the finding that HMGB1 release can occur after both necroptosis and pyroptosis, while release of S100, Hsp70, and Hsp90 only occurs upon necrosis and/or necroptosis, but not in the context of pyroptosis (Frank and Vince, 2019). Thus, ICD results in the release of DAMPs and sets off a chain reaction, since DAMPs themselves induce ICD in cells that recognize them. This inflammatory chain reaction can be unwanted and highly dangerous, particularly in locations with low regenerative capacity (Fig. 1). Mechanical stress, such as brain trauma, can induce both apoptosis and ICD via necrosis (Vourc’h et al., 2018). The balance between these two types of cell death in cases of mechanical stress varies between tissues and seems to shift more toward necrosis upon increased levels of stress and duration of stress (Takao et al., 2019; Valon and Levayer, 2019; Vourc’h et al., 2018). A recent review posits that a certain level of plasticity exists between apoptosis and ICD: inflammasomes, multiprotein oligomers that form intracellularly upon recognition of PAMPs or DAMPs and usually activate ICD, can drive apoptosis when specific molecules (caspase 1 or gasdermin D) are inhibited (Bedoui et al., 2020). iPRR could provide this inhibitory signal upon recognition of DAMPs, resulting in the immediate dampening of an inflammatory chain reaction by steering the response away from ICD and toward apoptosis. Consequently, one can imagine that if inhibitory signaling occurs swiftly in sterile stress conditions, such as ischemia–reperfusion injury or trauma, inflammatory responses can be avoided. Importantly, sterile stress conditions do not always result in measurable inflammatory responses, and it is conceivable that cells in specific essential tissues do not respond to the initial release of DAMPs altogether. Since dependence on a rapid switch from ICD toward apoptosis is a risky bet for essential tissues, a more rapid alternative would be if DAMPs that bind iPRRs directly rendered the cells unresponsive.iPRRs recognize molecules associated with homeostasisAs opposed to DAMPs, which typically are associated with danger and damage, HAMPs have previously been proposed to inhibit immune activation (Li et al., 2019; Sun et al., 2018; Wang et al., 2016). HAMPs have various properties and mechanisms of action; for example, lysophospholipids bind G protein–coupled receptors (Wang et al., 2016), and IL-35 binds cytokine receptors (Li et al., 2019). Already before the introduction of the concept of HAMPs, the guard theory of immunity was established in plants. The guard theory proposes that rather than sensing insults such as pathogens directly, the immune system recognizes the consequences of these insults for the organism. This is reflected by changes in the levels of the guard proteins, triggering immune responses (Dangl and Jones, 2001). Multiple lines of evidence suggest that the foundations of the guard theory also apply to the animal immune system (Medzhitov, 2009). Thus, HAMPs in animals and humans may be seen as a parallel to the preceding guard theory. Here, we discuss HAMPs that ligate immune inhibitory receptors.When cells undergo apoptosis, lipids such as phosphatidylserine (PS) and phosphatidylethanolamine (PE) are exposed on the cell surface and signal tissue-resident immune cells to find and dispose of the dying cells without triggering inflammation (Arandjelovic and Ravichandran, 2015; Gordon and Plüddemann, 2018; Segawa and Nagata, 2015). PS and PE are sensed by inhibitory members of the CD300 family of immune receptors, CD300a and CD300f (Choi et al., 2011; Simhadri et al., 2012). These interactions primarily result in dampening of mast cell activation by apoptotic cells, preventing inflammatory responses (Nakahashi-Oda et al., 2012). In line with this, CD300a/ mice develop exacerbated joint inflammation in an antigen-induced arthritis model (Valiate et al., 2019). In addition to apoptotic cells, viable cells can also transiently expose PS and PE, which may occur under inflammatory conditions (Arandjelovic and Ravichandran, 2015; Gong et al., 2020; Ravichandran, 2010), suggesting that additional layers of regulation may be needed to prevent phagocytosis of nonapoptotic cells. Indeed, it has been shown that CD300a/f ligation by PS and PE also negatively regulates phagocytosis of apoptotic cells (Ju et al., 2008; Simhadri et al., 2012). It is possible that a similar regulatory circuit is in place to prevent phagocytosis of PS- or PE-bearing nonapoptotic cells. Furthermore, all host cells express diverse sialylated glycan structures, and these sialic acids are effectively a molecular pattern associated with self and homeostasis. Sialylated glycans are sensed by immune receptors of the Siglec family (reviewed in Macauley et al., 2014). Most Siglecs (human Siglec 2, 3, and 5–11) harbor an ITIM motif and are inhibitory receptors. Each Siglec exhibits preferential recognition of a different sialylated glycan. Siglecs participate in immune surveillance and provide the immune system with inhibitory signals to prevent reactivity against self. It has recently been shown that, in addition to cell surface proteins and lipids, small RNAs can be modified with glycans and tethered to the cell membrane of diverse cells under homeostatic conditions, emphasizing the role glycans play in the maintenance of homeostasis (Flynn et al., 2021). In line with this, the lack of Siglec signaling is associated with autoimmune disease. Mice double-deficient for Siglec-G and Siglec-2 spontaneously develop systemic lupus erythematosus–like systemic autoimmune disease upon aging (Jellusova et al., 2010). Other mechanisms of the host’s own molecules preventing activation of the immune system have recently been demonstrated: for example, the inhibitory properties of select endogenous lipids on interactions between CD1a and TCR, effectively preventing T cell responses (Cotton et al., 2021). It remains to be determined whether similar molecules can also deliver inhibitory signals to immune cells via inhibitory receptors.Some molecular patterns elicit activating and inhibitory signalsSeveral molecular patterns can be recognized by both inhibitory and activating receptors. The inhibitory receptor leukocyte-associated Ig-like receptor 1 (LAIR-1) recognizes a HAMP present in different transmembrane and extracellular matrix–associated collagens as well as collectins, leading to negative regulation of inflammatory responses, such as airway inflammation during viral infection (Kumawat et al., 2019; Lebbink et al., 2009). Collagens are also recognized by the activating receptor osteoclast-associated Ig-like receptor (OSCAR), through which they can promote inflammation (Barrow et al., 2011; Schultz et al., 2016). Further, a few Siglec receptors are activating (Macauley et al., 2014), indicating there may be instances where sialylated glycans instigate immune activation. The relative expression of activating and inhibitory receptors on immune cells in a given situation, together with other potential environmental cues, will thus determine to what extent a cell becomes activated by these molecular patterns.iPRRs can deliver potent inhibitory signals to immune cells and attenuate or halt immune system activation. Therefore, they are often exploited by tumors to evade the immune system. For instance, many tumors highly express diverse collagens, dampening antitumor immune responses through LAIR-1 activation on immune cells (Peng et al., 2020; Rygiel et al., 2011). Similarly, various tumor types up-regulate sialylated ligands for inhibitory Siglec receptors, resulting in a dampened antitumor immune response (Fraschilla and Pillai, 2017; Jandus et al., 2014; van de Wall et al., 2020). CD155, the ligand for inhibitory receptor T cell immunoreceptor with Ig and ITIM domains (TIGIT), is also up-regulated on tumor cells and inhibits T cell antitumor immune responses (Braun et al., 2020; Dougall et al., 2017). Up-regulation of inhibitory receptor ligands in tumor tissues thus appears to be a strategy of immune evasion in cancer.iPRRs recognize microbial molecular patternsSimilar to how the occurrence of DAMPs does not always result in inflammation, microbial PAMPs do not always relay inflammation-promoting signals. Most microbes do not behave as either strictly pathogens or strictly commensals. Microbes with high pathogenic potential can also exist as harmless colonizers of the host, and commensal microbes can cause disease when they behave in an atypical way. Activating PRRs alone cannot differentiate between these situations, and it has thus been suggested that the immune system makes distinctions between pathogenic and nonpathogenic microbes through an integrated system of signals rather than one particular signal (Greslehner, 2020; Swiatczak et al., 2011). We argue that iPRRs may provide these additional signals.Immune inhibitory receptors have been shown to interact with microbes, but since these interactions have been predominantly studied in experimental models of infection, it is commonly thought that iPRR–microbe interactions mediate immune evasion by the microbe (Van Avondt et al., 2015). Since most microbes are not strictly pathogens, it is reasonable to think that the interaction of microbial ligands with inhibitory receptors could contribute to symbiosis. Multiple iPRRs recognize microbial ligands (Fig. 2). Staphylococcus aureus, a bacterium that commonly colonizes the human skin and nasal mucosa, interacts with the mouse paired Ig-like receptor B (PIR-B, orthologue of human LILRB3) through LTA, thereby limiting proinflammatory cytokine production. Indeed, PIR-B−/− mice infected with S. aureus show decreased survival compared with wild-type mice (Nakayama et al., 2012). LTA is a PAMP and an essential component of the cell wall universally expressed not only by S. aureus, but also by other related, less pathogenic species. The inhibitory receptor PIR-B/LILRB3 could thus regulate the host interaction with S. aureus in a noninflammatory context through recognition of PAMPs.As discussed above, endogenous sialic acids are a molecular pattern associated with self and homeostasis, and they interact with different inhibitory Siglec receptors. Sialic acids present on the surface of group B streptococcus (GBS) likewise interact with inhibitory Siglecs (Carlin et al., 2007; Chang et al., 2014). The sialic acid is common to all GBSs, which is not a strict pathogen but rather an opportunist. CD33 Siglecs are expressed in skin-resident Langerhans cells, which could allow for interaction between Langerhans cells and GBS, resulting in an inhibitory signal and thus promoting the colonizing lifestyle of GBS. Other inhibitory receptors interacting with bacteria are SIRL-1, which recognizes staphylococcal phenol-soluble modulins (Rumpret et al., 2021b), and TIGIT, which recognizes a ligand expressed by the oral commensal bacterium Fusobacterium nucleatum (Gur et al., 2015). The functional roles of these interactions are yet to be fully explored.A particularly prominent binder of microbial ligands is the inhibitory receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1). On immune cells, CEACAM1 is restrictively expressed on activated cells, whereas it is constitutively expressed by epithelial cells (Gray-Owen and Blumberg, 2006; Huang et al., 2015). It binds many different microbial ligands, such as bacterial Dr adhesins of Escherichia coli (Korotkova et al., 2008), the Opa protein of Neisseria meningitidis, Neisseria gonorrhoeae (Virji et al., 1996) and commensal Neisseria species (Toleman et al., 2001), adhesin UspA1 of Moraxella catarrhalis (Conners et al., 2008), the HopQ adhesin of Helicobacter pylori (Königer et al., 2016), CbpF adhesion of Fusobacterium sp. (Brewer et al., 2019; Gur et al., 2019), the streptococcal β protein (van Sorge et al., 2021), and an unidentified ligand in the fungus Candida sp. (Klaile et al., 2017). Although most of these microbes can be pathogenic, they do not always cause disease. Moreover, the absence of CEACAM1 has been shown in mouse models to predispose to colitis (Jin et al., 2016; Nagaishi et al., 2006). Together, these data indicate that CEACAM1 may have a tolerizing function in host–microbe interactions rather than serving only as a means for immune evasion.Concluding remarks and future perspectivesHere, we define a group of inhibitory receptors that can be classified as iPRRs. We argue that iPRRs, like their activating counterparts, recognize molecular patterns (Akira et al., 2006; Alvarez et al., 2008; An and Brodsky, 2016; Angata et al., 2002; Arakawa et al., 2018; Arnold et al., 2013; Brewer et al., 2019; Brown and Crocker, 2016; Carlin et al., 2007; Chang et al., 2014; Chen et al., 2009; Choi et al., 2011; Conners et al., 2008; Dougall et al., 2017; Fong et al., 2015; Gray-Owen and Blumberg, 2006; Gur et al., 2015; Gur et al., 2019; Han et al., 2005; Jones et al., 2016; Klaile et al., 2017; Königer et al., 2016; Korotkova et al., 2008; Kretschmer et al., 2010; Kumawat et al., 2019; Lebbink et al., 2009; Lewis Marffy and McCarthy, 2020; Liu et al., 2014; Macauley et al., 2014; Nakayama et al., 2012; Nakayama et al., 2007; Nuñez et al., 2018; Pende et al., 2006; Pérez-Oliva et al., 2011; Prantner et al., 2020; Rumpret et al., 2021a; Rumpret et al., 2021b; Segawa and Nagata, 2015; Simhadri et al., 2012; Sims et al., 2010; Steevels et al., 2013; van Sorge et al., 2021; Virji et al., 1996; Young et al., 2008; Yu et al., 2009; Zenarruzabeitia et al., 2015). This recognition provides context- and location-dependent signals to help shape the immune response. We indicate that most of the iPRRs discussed here are able to recognize both endogenous and microbial patterns (Fig. 2). The relative expression of activating and inhibitory PRRs and the integration of their signals ultimately determines the strength of an immune response to microbes or damage. This allows a differential response to tissue damage in different organs, depending on their susceptibility to immunopathology (Fig. 1). For example, in tissues that have low regenerative capacity, such as the brain, increased expression of iPRRs could provide a higher activation threshold and prevent the release of DAMPs that leads to inflammation and further tissue damage (Ashour et al., 2021). We also point out that endogenous patterns can signal “safety” via iPRRs to ensure that commonly occurring events such as apoptosis do not trigger the immune system. Similarly, there may be microbial patterns ensuring that harmless microbes colonizing the host do not bring about inflammatory responses (Fig. 3). For example, in the blood, microbial patterns such as LTA are recognized by activating PRRs. In contrast, in other anatomic locations such as the skin, iPRRs could also signal in response to these patterns, abrogating their potential to trigger inflammatory responses. We argue that the interactions between iPRRs and their microbial ligands may thus be vital for establishing and maintaining commensal–host homeostasis and suggest that studies in this direction are needed to examine this hypothesis. Further exploration of possible additional iPRRs, their ligands, and their expression patterns will provide a better understanding of the interactions of the host with its microbiota and the contextual regulation of septic and sterile inflammation.Table 1.Overview of different properties of iPRRs
iPRRiPRR expressioniPRR structureSignaling pathwayEndogenous ligandEndogenous ligand expressionMicrobial ligandActivating receptor for the same ligand
CD300a/fBroad on immune cells, upregulated on activationIg-likeITIMPS, PEExposed in programmed cell deathTim4
CEACAM‑1Broad on immune, epithelial, and endothelial cellsIg-likeITIMCEACAM1 and other CEACAMsConstitutiveIg fold proteinsOther CEACAMs
LAIR-1Broad on immune cells; on activation, upregulated on neutrophils and downregulated on T cellsIg-likeITIMCollagenConstitutiveOSCAR
LILRB1 (CD85j)Neutrophil, monocyte, dendritic cell, and NK cell, upregulated on activationIg-likeITIMS100 proteinsUpon cell damageTLR4, RAGE
LILRB3 (CD85a)Neutrophil, monocyte, dendritic cellIg-likeITIMUnknown cytokeratin-associated ligandUpon cell damageUnknown in S. aureus (LTA shown for mice ortholog PIR-B)TLR2/6
PVRDendritic cell, upregulated on activationIg-likeITIMNectin-3ConstitutivePoliovirus
Siglec 2, 3, 5–11Broad on immune cells, differs per receptorIg-likeITIMSialic acidsConstitutiveSialic acidsSiglec 14–16
Siglec 2, 3, 5–11Broad on immune cells, differs per receptorIg-likeITIMHsp70Upon cell damageTLR4, RAGE
Siglec 10B cell, eosinophil, monocyteIg-likeITIMHMGB1, Hsp90Upon cell damageTLR4, RAGE
SIRL-1Neutrophil, monocyte, downregulated on activationIg-likeITIMLL-37, S100 proteinsUpon cell damage and immune activationPhenol-soluble modulins of StaphylococcusTLR4, RAGE, FPR2
TIGITT cell, NK cell, upregulated on activationIg-likeITIMDNAM-1, TIGITTIGIT upregulated on activationUnknown in F. nucleatumDNAM-1
Open in a separate windowOSCAR, osteoclast-associated Ig-like receptor; PVR, poliovirus receptor; RAGE, receptor for advanced glycation end products.Open in a separate windowFigure 3.The integration of activating and inhibitory signals determines the outcome of the immune response. When damage or a dangerous microbe should not be tolerated, DAMPs and PAMPs signal through activating PRRs to mount an immune response. However, when it is more beneficial for the host to tolerate damage or a harmless microbe, then the same DAMP or PAMP, or a different pattern, can concomitantly signal an iPRR to inhibit the immune response. The relative expression of PRRs and iPRRs and their respective ligands determine the strength of the resulting immune response.Finally, iPRRs can be exploited to treat or prevent disease. The increased understanding of the function of inhibitory receptors has led to significant advances in the treatment of cancer. PD-1 and CTLA-4 have proven their potential as therapeutic targets on T cells for cancer immunotherapy (Ribas and Wolchok, 2018). Innate cells such as NK cells, innate lymphoid cells, and different myeloid cell types are also important in anticancer immune responses. These cells can directly contribute to tumor removal and additionally modulate antitumor T cell responses by steering T cell activation. Different iPRRs expressed on these cells, such as TIGIT and CD96, are already being explored as additional therapeutic targets (Dougall et al., 2017). With an increased understanding of the properties of iPRRs and their ligands, we expect that more of these receptors will be used as targets for immunotherapy.  相似文献   

14.
15.
16.
Pregnancy is recognized as a spontaneously acquired state of immunological tolerance by the mother to her semi-allogeneic fetus, but it is a major cause of allosensitization in candidates for organ transplantation. This sensitization, assessed by the presence of anti-HLA IgG, contributes to sex disparity in access to transplantation and increases the risk for rejection and graft loss. Understanding this dual tolerance/sensitization conundrum may lead to new strategies for equalizing access to transplantation among sexes and improving transplant outcomes in parous women. Here, we review the clinical evidence that pregnancy results in humoral sensitization and query whether T cell responses are sensitized. Furthermore, we summarize preclinical evidence on the effects of pregnancy on fetus-specific CD4+ conventional, regulatory, and CD8+ T cells, and humoral responses. We end with a discussion on the impact of the divergent effects that pregnancy has upon alloantigen re-encounter in the context of solid organ transplantation, and how these insights point to a therapeutic roadmap for controlling pregnancy-dependent allosensitization.

IntroductionThe fact that multiple successive pregnancies with the same male partner can be brought to term successfully suggests that the immunological response to a semi-allogeneic fetus is diametrically opposite to the responses elicited by genetically comparable transplanted organs. Peter Medawar in 1953 (Medawar, 1953) discussed this “immunological paradox of pregnancy,” and since then, there have been extensive investigations into how the fetus avoids rejection. A plethora of immune regulatory mechanisms has been uncovered within the uterine environment, including enrichment in regulatory T cells (Tregs), natural killer cells, regulatory macrophages, entrapment of APCs, and chemokine gene silencing of decidual stromal cells (PrabhuDas et al., 2015). Systemic factors that prevent fetal rejection have also been identified, including immune modulation by pregnancy-related hormones and release of tolerogenic placental debris, which may contribute to the preferential systemic expansion of fetus-specific Tregs and acquired dysfunction by conventional T cells (Tconvs) and CD8+ T cells. Since the majority of these mechanisms either act locally or only during pregnancy, it was assumed that T cell tolerance would manifest itself only in the context of subsequent pregnancy, and that encounter with the same alloantigens in the context of a solid organ transplant, in the absence of local or systemic pregnancy-induced immunomodulation, would trigger allograft rejection.The emphasis on T cells as the major mediator of allograft rejection and on T cell tolerance as a means to achieve transplantation tolerance parallels the focus on the constraint of T cells in pregnancy. Thus, despite studies in the 1980s by Bell and Billington (Bell and Billington, 1981; Bell and Billington, 1983; Bell and Billington, 1986) that pregnancy can elicit paternal-reactive antibodies, how pregnancy sensitizes B cell responses while maintaining T cell tolerance to the semi-allogeneic fetus has remained an under-investigated topic in preclinical models (PrabhuDas et al., 2015). In contrast and driven by the ease in quantifying HLA-specific antibodies but difficulty in assessing HLA-specific T cell responses, clinical studies in solid organ transplantation have revealed that pregnancy is a highly sensitizing event that results in the production of fetus-reactive anti-HLA antibodies, and the presence of these antibodies limits access to transplantation and contributes to increased risk of transplant rejection. In this review, we focus on the contrasting effects of pregnancy on these two arms of the adaptive immune system, and on how these pregnancy-shaped responses are recalled by alloantigens that are shared between offspring and transplanted allograft.Clinical impact of pregnancy alloimmunization in organ transplantationHumoral sensitizationThe effect of pregnancy on the immune system was first reported by J.J. Rodd in 1959 when he described peripartum women experiencing an increased number of blood transfusion reactions (Van Rood et al., 1958). It was this observation that allowed for the discovery of anti-HLA antibodies from the sera of pregnant women (Van Rood et al., 1958). Anti-HLA antibodies are produced during the first trimester of a pregnancy and increase in titer over the gestational course and with multiple pregnancies (Lee et al., 2011). During the postpartum phase, antibody levels rise in the first 90 d and gradually disappear in 50% of postpartum women over a 1–2 yr period (Cecka, 2010; Masson et al., 2013). Anti-HLA antibody titers following kidney transplantation increase more robustly in patients having had prior pregnancies than in those having received previous transplantation or transfusion, suggestive of robust pregnancy-induced memory B cells (Higgins et al., 2015). Notably, although pregnancy-induced alloantibodies can diminish with time, alloreactive memory T and B cells can persist (Senn et al., 2021). Thus, anti-HLA antibodies and memory B cells induced by semi-allogeneic pregnancies play a pivotal role prior to and after transplantation, especially for multiparous women.Historically, anti-HLA antibody titers were measured by the panel-reactive antibody (PRA) technique through a complement-dependent cytotoxicity assay; however, the major limitation of this method is its inconsistency and lack of HLA specificity. In 2009, the United Network for Organ Sharing implemented measuring sensitization using single HLA-coated beads, an assay that precisely identifies specific HLA antigen targets (Cecka, 2010). A computer algorithm generates a calculated PRA (cPRA) according to the HLA frequencies derived from the donor population with the goal of providing consistently accurate results on the extent of sensitization of transplant candidates and the chances for a highly sensitized candidate to find a compatible organ donor. Around 30% of pregnant women are sensitized when measured via complement-dependent cytotoxicity assay, whereas 50–75% of women were found to be sensitized by pregnancy when the single HLA bead assay was used (Bromberger et al., 2017). Furthermore, a retrospective analysis of the United Network for Organ Sharing registry’s waitlist pool showed that individuals with a cPRA >98% were over-represented by women by ~60% (Redfield et al., 2016). Cumulatively, these data reveal the detrimental impact of pregnancy in women in need of a transplant and the disparity it creates toward identifying a suitable donor organ and having a successful post-transplantation course.Living donor kidney transplantation has better outcomes compared to kidney transplantation from deceased donors (Roodnat et al., 2003). However, 30% fewer women received living donor kidney transplantation as compared with men despite comparable referrals (Bromberger et al., 2017; Roodnat et al., 2003). Pregnancy was identified as a major contributor to this disparity, as postpartum women were increasingly incompatible with their spouse and offspring compared with men (Bromberger et al., 2017). Furthermore, parous women are at a higher risk of being sensitized to unrelated donors sharing an allele of the partner or offspring (Gibney et al., 2006; Vaidya et al., 2006). Child-specific sensitization measured by single-HLA bead assay was detected at the HLA-A/B/C/DR loci in 28–38% of 301 multiparous women analyzed (Honger et al., 2013), with child-specific HLA-B loci being the most sensitizing followed by HLA-A > HLA-DRB1 > HLA-C (Dankers et al., 2003; Honger et al., 2013). Furthermore, by quantifying mother/child mismatches by the number of mismatched HLA eplets, where an eplet is defined as the cluster of amino acids representing the smallest functional unit of structural epitopes on the HLA molecule targeted by B cell receptor and antibodies, the rate of child-specific sensitization increased with the presence of ≥20 mismatched eplets (Honger et al., 2013). These observations are reminiscent of eplet-load mismatch between the organ donor and the recipient predicting de novo anti-HLA antibody production by the host and reduced graft survival, and thus underscories the detrimental effects of pregnancy-induced humoral sensitization (Philogene et al., 2020; Sapir-Pichhadze et al., 2020).T cell sensitizationIn contrast to the abundant evidence that fetus-specific B cell responses are induced during pregnancy and the barrier they pose to transplantation, the effects of pregnancy-induced effector T cell responses on subsequent transplantation are more opaque. Specifically, although it is clear that maternal T cells acquire tolerance to the semi-allogeneic fetus, it is uncertain whether this T cell tolerance extends to subsequent organ allografts sharing antigens with the fetus. Early observations that fetal-derived stem cells can persist in low numbers in the mother’s circulation for as long as 27 yr, a phenomenon termed peripheral fetal microchimerism (Nelson, 1998), prompted the hypothesis that this microchimerism mediates long-term fetus-specific tolerance in mothers and promotes the acceptance of grafts from their offspring (Starzl et al., 1993). However, several studies testing the correlation between donor/recipient kinship and allograft fate have reported comparable outcomes between groups receiving grafts from offspring versus non-offspring (Cohen et al., 2018; Ghafari, 2008; Mahanty et al., 2001). A recent retrospective analysis performed using the Organ Procurement and Transplant Network living donor liver transplant database revealed that 1-, 5- and 10-yr allografts and patient survival was poorer among mothers who received the organ from their offspring as compared with unrelated living donors (Dagan et al., 2020). A major caveat of such studies is the potential pro-rejection effects of pregnancy-sensitized B cells even when pregnancy-induced antibodies have diminished; as a result, the contribution of pregnancy-primed T cells, either pro-rejection or pro-tolerogenic, may be obscured. Indeed, Senn et al. (2021) reported that women with prior pregnancies receiving kidneys from their husband consistently had a higher rate of antibody-mediated rejection compared with women with prior pregnancies receiving kidneys from other living or deceased donors.A limited number of studies have attempted to directly quantify ex vivo donor-specific T cell responses arising during normal human pregnancy using proliferation, cytokine production, or cellular cytotoxicity as readouts. When IL-4 and IFNγ ELISPOT assays were used to quantify PBMC responses from non-pregnant versus pregnant women to paternal or pooled alloantigens, Mjosberg et al. (2007) reported that pregnancy did not result in increased paternal-specific IL-4 or IFNγ responses. Furthermore, removal of Tregs resulted in non-specific increases in IFNγ responses and paternal-specific augmentation in IL-4 production. Collectively, their study suggested an absence of pregnancy-specific sensitization of T cells, while also hinting at postpartum Tregs controlling fetus-specific IL-4 responses and broadly controlling IFNγ responses. Notably, reduced frequencies of circulating FoxP3+ Tregs were observed with spontaneous preterm birth, preeclampsia, and recurrent spontaneous miscarriages compared to healthy pregnancies suggesting a more systemic effect of Tregs (Dimova et al., 2011; Inada et al., 2015; Inada et al., 2013; Kisielewicz et al., 2010; Koucky et al., 2014; Mjosberg et al., 2010; Nadkarni et al., 2016; Schober et al., 2012; Tilburgs et al., 2008; Tsuda et al., 2018).Pregnancy-induced Tregs are critical for promoting both primary and secondary pregnancies by suppressing T cell proliferation and cytokine production not only in secondary lymphoid organs but also in the placenta (Salvany-Celades et al., 2019). Expansion of Tregs in the decidual tissue has been prostulated to suppress fetus-specific responses locally (Tilburgs et al., 2008; Erlebacher, 2013). Notably, three different Treg populations have been identified at the maternal–fetal interface: CD25HIFOXP3+, PD1HIFOXP3IL-10+, and TIGIT+FOXP3dim Tregs. Decidual CD25HIFOXP3+ Tregs were able to suppress the proliferation and IFNγ and TNFα production by CD4+ and effector CD8+ T cells in vitro, whereas decidual PD1HI Tregs and TIGIT+ Tregs inhibited CD4+ but not effector CD8+ T cells. However, whether pregnancy-induced Tregs are most potent in the decidua or whether they can also dominantly suppress T cell responses to offspring-matched allografts in secondary lymphoid organs is currently unknown.CD8+ T cell responses to fetus-specific minor antigens have been more consistently reported to develop during pregnancy compared to CD4+ T cell responses (Linscheid and Petroff, 2013). Lissauer et al. (2012) assayed fetal-specific CD8+ cytotoxic responses using MHC-peptide dextramer multimers bearing a HY-immunodominant peptide in women pregnant with a male fetus. These CD8+ T cells expanded during pregnancy and persisted in the post-natal period in 50–62% of pregnant women. Furthermore, the fetal-specific CD8+ T cells retained their ability to proliferate, secrete IFNγ, and lyse target cells. These observations corroborated previous studies (Bouma et al., 1996; James et al., 2003; Mommaas et al., 2002; Piper et al., 2007; Verdijk et al., 2004) and suggested that fetal-specific CD8+ T cells expand during pregnancy and persist postpartum. It is tempting to speculate that preservation of fetus-CD8+ T cell responses during pregnancy, especially in the decidua, may have been evolutionarily selected to ensure the development of protective immunity for the developing fetus against viral infections, given that the fetus is haplo-identical to the mother, and thus maternal HLA-restricted CD8+ responses will recognize virally infected fetal cells (Tilburgs and Strominger, 2013; van Egmond et al., 2016). Indeed, observations that the decidua contains a higher percentage of CD8+ T cells and a lower percentage of CD4+ T cells compared with the peripheral blood is consistent with this possibility (Tilburgs et al., 2009; van Egmond et al., 2016).Potentially divergent fates of fetus-specific T cell subsets, together with a paucity of studies examining fetus-specific T cell responses in the extended postpartum period, make it difficult to definitively conclude if pregnancy-primed T cells are functionally tolerant or sensitized to fetal antigens presented in the context of a solid organ transplant. The ex vivo quantification of fetus-specific T cell responses is technically challenging and complicated by the increased frequency of pregnancy-induced Tregs (Salvany-Celades et al., 2019). Furthermore, ex vivo observations may not necessarily predict how these cells will behave in vivo after transplantation with organs sharing HLA antigens with the fetus. In vivo studies in postpartum recipients suggest that poorer outcomes are complicated by pregnancy-induced humoral sensitization (
AuthorNo. of transplantsOutcome
Terasaki et al. (1995) Husband-to-mother: n = 368Comparable allograft survival between spousal donor and unrelated living donor. Pregnancy is a risk factor for loss of allograft
Child-to-mother: n = 1,411
Mahanty et al. (2001) Offspring-to-mother: n = 874Fetal tolerance did not translate to a superior allograft survival from offspring donors. Multiple pregnancy trended towards poor allograft survival
Unrelated living donor to mother: n = 310
Cohen et al. (2003) Offspring-to-parent: n = 3,370Comparable death censored 5-yr allograft survival in offspring-to-parent compared to unrelated living donor
Unrelated living donor: n = 8,351
Deceased donor: n = 44,792
Miles et al. (2008) Offspring to mother: n = 3,124Comparable and poor allograft survival in offspring-to-parent and parent-to-offspring transplants
Parent to offspring: n = 6,076
Ghafari (2008) Offspring-to-mother: n = 12Unrelated living donor allografts survival was significantly higher compared to offspring and husband donor allografts
Husband-to-mother: n = 9
Unrelated living donor: n = 150
Choi et al. (2012) Offspring-to-mother: n = 49Comparable 5- and 10-yr kidney graft survival between offspring-to-mother and offspring-to-father transplant. Mother-to-child had worse outcome
Parent-to-offspring: n = 146
Redfield et al. (2016) Highly sensitized: n = 7,145Increased graft loss by 23% among women with a history of pregnancy and transfusion compared to non-sensitized
Non-sensitized: n = 100,147
Cohen et al. (2018) Offspring-to-mother: n = 1,332Comparable allograft survival between offspring and unrelated living donor transplant to mother
Unrelated living donor: n = 1,435
Dagan et al. (2020) Offspring-to-mother: n = 148Offspring donor allograft survival lower compared to unrelated living donor
Unrelated living donor: n = 93Male offspring donor resulted in poorer survival compared to female offspring donor
Senn et al. (2021) Husband-to-mother: n = 25Poor allograft survival among mothers who received allograft from spouse compared to unrelated living donor or deceased donor
Unrelated living donor: n = 52
Deceased donor: n = 120
Open in a separate windowSemi-allogeneic pregnancy in mice tolerizes T cell responses, but primes fetus-specific B cell responsesRecognition of fetal antigens by T cellsThe identification of fetus-reactive T cells in pregnant mice has relied on either the transfer of a tracer population of TCR-Tg T cells specific for a paternal-derived antigen or the use of fluorescent peptide:MHC multimers. Multimers present a peptide derived from a model antigen such as membrane-bound ovalbumin (mOVA), constitutively expressed by the mating male and present in the seminal fluid and some or all the products of conception (Moldenhauer et al., 2009). During pregnancy, mOVA is expressed in the placental and endovascular trophoblast with access to the decidua and maternal spiral arterioles, respectively, and thus to the gestational mother’s immune system (Erlebacher et al., 2007).Initial encounter of paternal antigens occurs via exposure to seminal fluid (Moldenhauer et al., 2009), and both CD4+ and CD8+ T cells reactive to OVA were found in the para-aortic lymph nodes when TCR-Tg T cells were transferred into female hosts immediately postcoitus with an mOVA-expressing male. Using bone marrow chimeric mice, in which only hematopoietic or only non-hematopoietic cells could present OVA, it was shown that presentation of seminal antigen occurred indirectly in female hematopoietic cells (Moldenhauer et al., 2009). During pregnancy, recognition of placental antigen starts at around E10.5, and presentation of paternal antigens by maternal APCs also occurs systemically in secondary lymphoid organs in addition to the para-aortic lymph nodes (Erlebacher et al., 2007). Presentation of fetal antigen increases over the course of gestation, with some presentation remaining until 3 wk postpartum (Erlebacher et al., 2007; Moldenhauer et al., 2009).Function of fetus-reactive T cellsWhile paternal OVA–reactive TCR-Tg CD8+ T cells proliferate to cognate antigen expressed in seminal fluid and in placenta, and they persist, they acquire little IFNγ production capacity when compared to positive control stimulation (Erlebacher et al., 2007). Phenotypically, they express high levels of the inhibitory receptor PD-1, and appeared to retain cytotoxic potential but failed to re-expand upon secondary pregnancy (Barton et al., 2017), suggesting a state akin to T cell exhaustion. Indeed, Lewis et al. (2022) used mouse models and human transplant registry data to demonstrate that pregnancy induced a sustained exhausted phenotype in CD8 T cells (PD-1, Lag-3, CD38, Eomes, and TOX) that was associated with hypofunctional CD8+ T cells and prolonged allograft survival. A similar profile of high expression of PD-1 and TIM-3 was found in endogenous polyclonal OVA-reactive CD8+ T cells of females pregnant with OVA-expressing fetuses (Kinder et al., 2020). This exhaustion state was further reinforced upon secondary pregnancy (but not upon non-pregnancy secondary encounter of the alloantigen), and blockade of PDL1/TIM-3 unleashed the activation of these fetal-reactive T cells and triggered fetal loss during secondary but not primary pregnancy (Kinder et al., 2020).Analysis of endogenous CD4+ T cells reactive to the model antigen 2W expressed by concepti revealed that pregnancy can induce a state of anergy in paternal-reactive Tconvs, characterized by high expression of the surface receptors FR4 and CD73 and lower production of IL-2 upon restimulation (Kalekar et al., 2016). Importantly, a subset of these anergic Tconvs, contained within the Neuropilin-1+ population, differentiated into induced Tregs (iTregs) that could suppress inflammation caused by anergic Tconvs reinvigorated during T cell lymphopenia (Kalekar et al., 2016). Both anergic Tconvs and Tregs are thought to be important for the maintenance of fetal tolerance, as depletion of Tregs or blockade of negative regulators of T cell activation expressed by both effector Tconvs and Tregs such as PD-1 are known to precipitate fetal loss (Aluvihare et al., 2004; Guleria et al., 2005); whether these signals are important in Tregs or non-Treg subsets has been difficult to parse out (Zhang and Sun, 2020).Importantly, pre-existing memory T cells do not cause fetal loss upon antigen-positive pregnancy, despite their relative resistance to Treg suppression (Yang et al., 2007), reduced dependence on co-stimulation for activation (Croft et al., 1994), and ability to enter target tissues in the absence of secondary lymphoid organ priming (Chalasani et al., 2002). It has been shown that OVA-reactive memory T cells generated by immunization with OVA plus adjuvant were prevented from entering the decidua because of epigenetic silencing of CXCL9, CXCL10, and CCL5 in decidual stromal cells, thus suggesting a mechanism of preserving the semi-allogeneic fetus (Nancy et al., 2012).Fetus-reactive TregsTregs are necessary for the implantation of early pregnancy following allogeneic mating, but not for sustaining late pregnancy (Shima et al., 2010). Using a pregnancy model in which the concepti express the paternal antigen 2W, Rowe et al. found a preferential expansion of 2W-reactive Tregs over Tconvs, resulting in >60% Tregs of 2W-reactive CD4+ T cells by the end of gestation (Rowe et al., 2012b). These Tregs, expanded during primary pregnancy from thymic Tregs and iTregs, persisted after parturition and were recalled rapidly during secondary pregnancy (Rowe et al., 2012b). Indeed, the importance of iTregs in fetal tolerance was confirmed with female mice that lack the FoxP3 CNS1 enhancer element, a necessary region for the differentiation of iTregs. These mice experienced a higher fetal resorption rate when mated with allogeneic compared with syngeneic males (Samstein et al., 2012). In addition, infection with Listeria monocytogenes during pregnancy, which elicits inflammation, reduces the ratio of paternal-specific Treg:Tconv, diminishes Treg suppression, and also triggers fetal wastage after allogeneic mating (Rowe et al., 2012a). This could be prevented if the placental entry of effector T cells was blocked with anti-CXCR3 antibody (Chaturvedi et al., 2015). This loss of fetal tolerance during inflammation is similar to the abrogation of transplantation tolerance we observed in recipients of heart allografts infected with L. monocytogenes after the establishment of cardiac transplantation tolerance (Wang et al., 2010).The mechanisms by which antigens in the semen or shed from the placenta can convert Tconvs into iTregs remain to be fully understood. Semen is known to contain high levels of TGFβ (Sharkey et al., 2012), a cytokine which in synergy with IL-2 can drive iTreg differentiation. A recent study suggests that TLR4 signals are essential in the immediate postcoital period to expand Tregs, an observation that may explain why females lacking TLR4 had impaired reproductive outcomes after allogeneic mating (Chan et al., 2021). Interestingly, alloreactive Tregs can potentially arise long before mating, following exposure in utero to maternal tissue that can establish allogeneic microchimerism in the progeny and sustained exposure to non-inherited maternal antigens (NIMA). Subsequent mating of the female offspring with allogeneic males that share determinants with NIMA further expands these Tregs, which confers a more robust fetal tolerance compared with progeny not exposed to NIMA. The increased NIMA-reactive Treg expansion during pregnancy can result in resistance to infection-triggered fetal loss and improved reproductive fitness (Kinder et al., 2015).The mechanisms by which Tregs prevent fetal wastage are not well understood. Suppression of Tconvs in an antigen-dependent manner likely plays a role, as suggested by the partial loss of 2W+ but not 2W concepti in female mice harboring memory 2W-reactive Th1 cells that are unable to convert into Tregs during subsequent pregnancy (Xin et al., 2014). One mechanism of Treg suppression that may play a role in pregnancy is its ability to induce indolamine 2,3-dioxygenase (IDO) in dendritic cells (Fallarino et al., 2003). IDO causes tryptophan catabolism and kynurenin production that is deleterious to T cell proliferation and survival, respectively. Indeed, pharmacological inhibition of IDO results in fetal loss following allogeneic but not syngeneic mating (Munn et al., 1998). A cautionary observation is that litter sizes of IDO-knockout females mated with allogeneic IDO-knockout males were of normal size, even upon treatment with an IDO inhibitor during gestation (Baban et al., 2004), thus suggesting possible development of compensatory mechanisms when IDO is absent from birth. Finally, Tregs may also prevent fetal loss in a T cell–independent manner, through their control of inflammation, as Treg depletion triggers significant inflammation and fetal wastage, similarly to that observed following injection of LPS (Bizargity et al., 2009). This observation is reminiscent of the loss of cardiac transplantation tolerance in mice with high levels of circulating IFNβ and IL-6 (Wang et al., 2010).Semi-allogeneic pregnancy sensitizes fetus-specific B cell responsesChanges in B cell lymphopoiesis occur during pregnancy and have been demonstrated in mice and humans (Lima et al., 2016; Muzzio et al., 2014). Muzzio et al. (2014) reported that immature B cells are lower in number and mature B cells are higher in the bone marrow during the late phase of pregnancy. In the spleen, B220+ B cells decreased in number as compared to non-pregnant mice but increased by ∼2–2.3-fold in the para-aortic lymph nodes draining the uterus. Billington and colleagues (Bell and Billington, 1981; Bell and Billington, 1983; Bell and Billington, 1986) demonstrated that murine pregnancy induced anti-paternal alloantibodies in some responder strains of mice, which could be eluted from the placenta and detected in the fetus. Importantly, paternal-specific antibodies increased during the final 3 d of pregnancy and reached maximal levels around 1 wk postpartum, while in secondary pregnancies, the antibody response was observed between day 9 and 10 of pregnancy, consistent with a recall response (Roe and Bell, 1982). While only a limited set of responder mouse strains generated anti-paternal antibodies and only after multiple pregnancies, it is notable that the agglutination or hemadsorption assays used to detect antibodies were relatively insensitive and would only be able to detect high titer antibodies. Additionally, the detection of anti-paternal antibodies was hampered by their lack of complement-dependent cytolytic activity, thus precluding the use of hemolytic assays (Bell and Billington, 1980).Recently, Suah et al. (2021) used a mouse model of semi-allogeneic pregnancy that included the 2W1S-OVA as a model paternal antigen to show that 2W-specific T cell responses are tolerized whereas B cell responses are simultaneously elicited during allogeneic pregnancy (Fig. 1). Fetus-specific CD4+ Tconvs expanded and developed a phenotype of exhaustion/anergy with upregulated FR4, CD73, and PD-1, and the ability of fetus-specific CD4+ and CD8+ T cells to produce IFNγ was inhibited as late as postpartum day 45. Importantly, there was a preferential expansion of 2W-specific FoxP3+Tregs over Tconvs, and these Tregs exhibited significant increases in the expression of CTLA-4 and CD73. Interestingly, fetus-specific antibody was detected at the time of parturition and increased further in the first week postpartum for most first-time mothers and remained elevated thereafter; all mothers developed fetus-specific antibodies by day 7 postpartum in secondary pregnancies. Thus murine pregnancies recapitulate the humoral sensitizing effects observed in human pregnancies, with the caveat that the fetus-specific antibody response was significantly reduced compared with skin sensitization. Fetus-specific antibodies were generated independently of germinal center reaction but were nevertheless blocked with CTLA-4Ig administered starting at the last week of pregnancy. These observations suggest that the anti-fetus IgG response is T cell but germinal center–independent, raising several questions including the affinity and specific antigenic targets of antibodies, signals provided by T cells to the development of anti-paternal antibodies, and how such helper T cells can develop in the backdrop of T cell tolerance to the semi-allogeneic fetus.Open in a separate windowFigure 1. Pictorial summary of the impact of semi-allogeneic pregnancy on fetus-specific T and B cell responses and subsequent consequence upon recall by fetus-matched transplanted allografts. Pregnancy-induced T cell tolerance to fetus-matched allografts is overridden by sensitized B cell responsesThe consequences of pregnancy-sensitized B cell responses to transplanted organs expressing alloantigens shared with offspring is well characterized; however, the implications of pregnancy-induced tolerance of T cell responses to the same allografts are less understood. Pioneering studies by Barton et al. (2017) showed that OVA-specific OT1 T cells adoptively transferred into pregnant mice acquired a state of dysfunction that was persistent when the postpartum mice were challenged with OVA-expressing skin grafts. In addition, those OT1 cells expressed elevated PD-1, and did not expand nor develop the ability to produce cytokines. Nevertheless, postpartum mice were able to reject OVA-skin grafts suggesting that pregnancy-tolerized OT-1 or endogenous OVA-specific T cells retained sufficient function to mediate skin graft rejection. More recently, Kinder et al. (2020) investigated the fate of an endogenous population of fetus (OVA)-specific CD8+ T cells in females pregnant after mating with OVA+ males. OVA-specific CD8+ T cells were primed and accumulated during primary pregnancy and persisted as an activated memory pool after parturition. While the dysfunctional state was reinforced during secondary pregnancies, postpartum mice challenged with OVA+ splenocytes in vivo underwent robust expansion and exhibited cytolytic activity. Moreover, in the setting of tumor immunity, Jasti et al. (2017) reported enhanced immune response in post-parous mice bred with OVA-expressing males to subsequent tumors expressing OVA. That endogenous CD8+ T cells primed by pregnancy retained the ability to respond to the same antigens in a non-pregnancy context suggested that CD8+ T cell responses in secondary pregnancies are curtailed locally and, despite features of dysfunction, may not be sufficient to facilitate the spontaneous acceptance of offspring-matched transplanted grafts.The observations of effector/pathogenic T cell responses to fetal antigens encountered in a non-pregnancy context can be explained by an alternative hypothesis, namely that pregnancy-induced T cell tolerance is overridden by pregnancy-sensitized memory B cells and/or fetus-specific antibodies. In support of this hypothesis, postpartum WT females acutely rejected offspring-matched F1 hearts whereas postpartum µMT mice that lacked B cells and antibodies spontaneously accepted F1 hearts. Notably, sIgKO mice that are B cell replete but lacked secreted antibodies and underwent a F1 pregnancy, rejected F1 hearts, while the depletion of B cells restored F1 heart acceptance. Adoptive transfer of pregnancy-primed but not naive B cells from sIgKO mice into postpartum µMT mice precipitated F1 heart rejection (Fig. 2); rejection was associated with the accumulation of fetus-specific Tconvs and restored IFNγ-responses in fetus-specific CD8+ T cells. Finally, we showed that anti-fetus antibodies transferred into postpartum µMT mice also precipitated F1 heart rejection; based on our previous observations (Burns and Chong, 2011) we speculate that anti-F1 antibodies function to opsonize antigens, activate antigen-presenting dendritic cells through engagement of Fc, and complement receptors that are able to override T cell tolerance (Fig. 2). These observations are supported by Lewis et al. (2022) where, in the absence of B cells, postpartum µMT mice exhibited prolonged allograft survival compared with virgin µMT mice.Open in a separate windowFigure 2.Postpartum T cell tolerance is overridden in the presence of pregnancy-sensitized B cells and fetus-specific antibodies. (A–C) Loss of pregnancy-induced T cell tolerance and F1 graft rejection is driven by cognate interactions between pregnancy-primed T and (A) pregnancy-primed B cells but not (B) naive B cells, or (C) with antigen-presenting dendritic cells (DC) activated by donor antigen (Ag) opsonized with pregnancy-primed F1-specific antibodies. The signals overriding T cell tolerance requires definition. PP, postpartum.The observations that fetus-specific T cell tolerance induced in the presence or absence of B cells during pregnancy extends to F1 heart allografts only in the absence of pregnancy-primed B cells and antibodies raise new questions. The signals delivered by pregnancy-primed B cells or fetus-specific IgG remain undefined (Fig. 2). Additionally, that pregnancy-sensitized B cells and fetus-specific antibodies do not prevent subsequent pregnancies underscores mechanisms within the placental microenvironment that allow T tolerance to F1 fetus to be maintained but are absent in allografts. Finally, these observations raise the intriguing possibility that humoral desensitization to eliminate pregnancy-primed B cells and antibodies at the time of graft transplantation may reveal a propensity of postpartum women to become tolerant to offspring-matched allografts. Humoral desensitization protocols identified in preclinical models and tested in the clinic suggest a roadmap for exploring this possibility (Alishetti et al., 2020; Jain et al., 2020; Jordan et al., 2021; Schinstock et al., 2020).ConclusionsSuccessful pregnancies balance the need to develop tolerance to the semi-allogeneic fetus and the need to preserve the ability to develop protective immunity to infections in the mother and pass this immunity to the fetus and the neonate. We speculate that preserving humoral immunity, especially late in pregnancy and in the postpartum period, permits the inadvertent generation of fetus-specific memory B cells and antibodies. That these B cells and antibodies are pathogenic for a subsequent organ transplant but do not prevent subsequent pregnancies underscores the potent mechanisms at the maternal–fetal interface that mitigate the effects of antibodies and memory B cells. Understanding these mechanisms might provide novel insights into attenuating their effects in organ transplantation. Conversely, observations that pregnancy-induced T cell tolerance extends to fetus-matched allografts suggest that potent immunomodulatory mechanisms in the uterine–fetal interface are not necessary to constrain fetus-specific T cells in subsequent pregnancies and the possibility of leveraging these pregnancy-induced tolerance mechanisms for promoting the acceptance of allogeneic transplants. Finally, the ability of pregnancy-sensitized B cells and alloantibodies to override potential donor-specific T cell tolerance reveals an opportunity to target humoral desensitization and cognate T:B cell interactions to allow for pregnancy-induced T cell tolerance to dominate in the setting of allograft transplantation.  相似文献   

17.
Neutralizing antibodies to HIV-1 induced by immunization     
Laura E. McCoy  Robin A. Weiss 《The Journal of experimental medicine》2013,210(2):209-223
  相似文献   

18.
Development of Genomic DNA Reference Materials for Genetic Testing of Disorders Common in People of Ashkenazi Jewish Descent     
Lisa Kalman  Jean Amos Wilson  Arlene Buller  John Dixon  Lisa Edelmann  Louis Geller  William Edward Highsmith  Leonard Holtegaard  Ruth Kornreich  Elizabeth M. Rohlfs  Toby L. Payeur  Tina Sellers  Lorraine Toji  Kasinathan Muralidharan 《The Journal of molecular diagnostics : JMD》2009,11(6):530-536
Many recessive genetic disorders are found at a higher incidence in people of Ashkenazi Jewish (AJ) descent than in the general population. The American College of Medical Genetics and the American College of Obstetricians and Gynecologists have recommended that individuals of AJ descent undergo carrier screening for Tay Sachs disease, Canavan disease, familial dysautonomia, mucolipidosis IV, Niemann-Pick disease type A, Fanconi anemia type C, Bloom syndrome, and Gaucher disease. Although these recommendations have led to increased test volumes and number of laboratories offering AJ screening, well-characterized genomic reference materials are not publicly available. The Centers for Disease Control and Prevention-based Genetic Testing Reference Materials Coordination Program, in collaboration with members of the genetic testing community and Coriell Cell Repositories, have developed a panel of characterized genomic reference materials for AJ genetic testing. DNA from 31 cell lines, representing many of the common alleles for Tay Sachs disease, Canavan disease, familial dysautonomia, mucolipidosis IV, Niemann-Pick disease type A, Fanconi anemia type C, Bloom syndrome, Gaucher disease, and glycogen storage disease, was prepared by the Repository and tested in six clinical laboratories using three different PCR-based assay platforms. A total of 33 disease alleles was assayed and 25 different alleles were identified. These characterized materials are publicly available from Coriell and may be used for quality control, proficiency testing, test development, and research.Many ethnic groups have genetic disorders that are over-represented due to founder effects. Examples include cystic fibrosis and α-1-antitrypsin deficiency in European Caucasians,1,2 and α or β thalassemia in groups living in equatorial regions with endemic malaria (Medical Genetics Information Resource, http://www.genetests.org, last accessed May 4, 2009).3Ashkenazi Jewish (AJ) individuals are the descendents of those belonging to the Hebrew ethnic and religious group that settled in Eastern Europe in the early Middle Ages. Several autosomal recessive disorders are more common in the AJ population than in the general population 4,5,6 An estimated one in 4.8 AJ individuals is a carrier of one of these diseases,8 most of which are severe and cause significant morbidity and mortality. Treatment to reduce symptoms and prolong life is available for some of these disorders, and novel treatments and therapies, including enzyme replacement therapy, have recently become available or are in development.

Table 1

Alleles Included in Clinical Ashkenazi Jewish Testing Panels
DisorderGeneAlleles on clinical AJ panelsAllele frequency in affected AJ populationAJ heterozygote frequency (all alleles)Allele frequency in affected non-AJ populationAllele found in DNA samples studied
BSBLM2281del6/ins7>99%71:107,7 111,13 1578Yes
NM_000057.2:c.2340delATCTGA. insTAGATTC
CDASPAE285A82.9%91:37,8 38,11 65102.5%9Yes
NM_000049.2:c.854A>C
CDASPAY231X14.8%90.0%9Yes
NM_000049.2:c.693C>A
CDASPAA305E0%960%9Yes
NM_000049.2:c.914C>A
CDASPA433(−2)A>G (IVS2-2)1.1%90.0%9No
NM_000049.2:c.433–2A>G
FAFACCIVS4(+4)A>T>99%121:77,8 89,12 9213Yes
NG_011707.1:g.82053A>T
FAFACC322delGYes
NM_000136:c.67delG
FDIKBKAPIVS20+6T>C (2507+6T>C)98%81:29,8 316Yes
NG_008788.1:g.40664T>C
FDIKBKAPR696P2%8No
NP_03631.2:p.R696P
GDGBAN370S85%51:17,8 1810Yes
NM_000157.2:c.1226A>C
GDGBA84GG (1035insG)6%5Yes
NM_000157.2:c,93_94insG
GDGBAL444P3.5%5Yes
NM_000157.2:c.1448T>C
GDGBAIVS2+1G>A1%5Yes
NM_000157.2:c.27+1G>A
GDGBAR496HNo
NM_000157.2:c.1604G>A
GDGBAD409HNo
NM_000157.2:c.1343A>T
GDGBAV394LYes
NM_000157.2:c.1297G>T
GDGBAdel55bpNo
NM_000157.2:c.1263del55
MLIVMCOLN1IVS3-2A>G66%141:67,8 100,15 12714Yes
NM_020533.1:c.406-2A>G
MLIVMCOLN1del6434(ex1-7)30%14Yes
AF_287270:g.511-6943del
NPSMPD1R496L97%16,101:90,16 103,8 12510Yes
NP_000534.3:R496L
NPSMPD1fsP330Yes
NM_00543.2:c.990delC
NPSMPD1L302PYes
NP_000534.3:L302P
NPSMPD1delR608Yes
NP_000534.3:R608del
TSDHEXA1278+TATC81%171:311732%17Yes
M_16411:c.1278.insTATC
TSDHEXAIVS12+1G>C15%17Yes
M_16421:g.200G>C
TSDHEXAG269S2%17Yes
M_16411:c.805G>A
TSDHEXAIVS9(+1)G>A0%1714%17Yes
M_16417:g.149G>A
TSDHEXAR247W (pseudo)0%17Yes
M_16411:c.739C>T
TSDHEXAR249W (pseudo)0%17No
M_16411:c.745C>T
TSDHEXAdel7.6kbNo
NT_010194:g.del70457939-70449986
TSDHEXAIVS7+1G>ANo
M_16417:g.149G>A
GSDG6PCR83C∼100%181:7118Yes
NM_000151.2:c.247C>T
GSDG6PCQ347X
NM_000151.2:c:1039C>TYes
Open in a separate windowAJ, Ashkenazi Jewish; BS, Bloom Syndrome; CD, Canavan Disease; FA, Fanconi Anemia; FD, familial dysautonomia; GD, Gaucher disease; MLIV, mucolipidosis type IV; NP, NP disease type A; TSD, Tay-Sachs disease; GSD, glycogen storage disorder type la.Most cases of these diseases in the AJ population are due to one or a few disease causing alleles. For example, three mutations account for approximately 95% of Niemann Pick (NP) chromosomes in the AJ population, while there is no common mutation associated with NP in the general population 19,20 Molecular testing for many of the disorders common in the AJ population has been developed and is currently in widespread use.The American and Israeli Jewish communities have been highly supportive of population-based carrier testing and reproductive genetic counseling for carriers. Tay-Sachs disease (TSD) is the first genetic condition for which community-based carrier detection was implemented.17,21 In Jewish communities around the world, couples are urged to participate in screening before pregnancy and, in some cases, before marriage. Now 30 years old, carrier screening for TSD is the longest-running, population-based program designed to prevent a lethal genetic disease. Screening has reduced the number of TSD cases in the United States and Canada by 90% (Genome News Network, http://www.genomenewsnetwork.org/articles/08_01/Tay_Sachs_gene_tests.shtml, last accessed March 9, 2009).In 2004, the American College of Obstetricians and Gynecologists recommended that individuals of AJ descent undergo prenatal and preconceptional carrier testing for TSD, Canavan disease (CD), familial dysautonomia (FD), and cystic fibrosis. Testing for mucolipidosis type IV (MLIV), NP disease type A, Fanconi anemia (FA) group C, Bloom syndrome (BS), and Gaucher disease (GD) was also suggested.22 Although glycogen storage disorder type 1a (GSD) testing has not been recommended by American College of Obstetricians and Gynecologists, many laboratories offer this analysis because testing for two alleles, R83C and Q347C, in the G6PC gene can detect almost 100% of affected patients in the AJ population. In addition to the American College of Obstetricians and Gynecologists recommendations, the American College of Medical Genetics has recommended carrier testing for cystic fibrosis, CD, FD, TSD, and suggests that screening should be offered for FA, NP, BS, MLIV and GD.23 Many molecular genetics laboratories currently offer testing for some or all of these disorders and often multiplex them into a single testing panel. More laboratories are expected to offer testing as the demand increases.A variety of assay methods, including commercial analyte specific reagents and laboratory developed tests (LDTs) are in use. As with other genetic diseases, laboratories testing for AJ disorders often find it difficult to obtain reference materials (RMs) or quality control (QC) materials for test development, validation, QC, and proficiency testing/external quality assessment. This is due to the relative rarity of affected patients, paucity of archived samples in research laboratories, and lack of materials available from repositories.To address the lack of RMs for AJ panel testing, the Centers for Disease Control and Prevention-based Genetic Testing Reference Materials Coordination Program (GeT-RM), in collaboration with members of the genetic testing community and the Coriell Cell Repositories, have created a set of 31 genomic DNA materials with confirmed mutations. These RMs were selected to include alleles representing nine disorders (BS, CD, FA, FD, GD, MLIV, NP, TSD and GSD) commonly included in clinical AJ testing panels. The alleles in these samples were confirmed by six volunteer laboratories using a variety of methods. A separate set of materials with characterized cystic fibrosis mutations has been developed and is reported separately.24  相似文献   

19.
Predicting the Future: Opportunities and Challenges for the Chemical Industry to Apply 21st-Century Toxicity Testing     
Raja S Settivari  Nicholas Ball  Lynea Murphy  Reza Rasoulpour  Darrell R Boverhof  Edward W Carney 《Journal of the American Association for Laboratory Animal Science》2015,54(2):214-223
  相似文献   

20.
Low Prevalence of Chagas Parasite Infection in a Nonhuman Primate Colony in Louisiana     
Patricia L Dorn  Megan E Daigle  Crescent L Combe  Ashley H Tate  Lori Stevens  Kathrine M Phillippi-Falkenstein 《Journal of the American Association for Laboratory Animal Science》2012,51(4):443-447
Chagas disease, an important cause of heart disease in Latin America, is caused by the parasite Trypanosoma cruzi, which typically is transmitted to humans by triatomine insects. Although autochthonous transmission of the Chagas parasite to humans is rare in the United States, triatomines are common, and more than 20 species of mammals are infected with the Chagas parasite in the southern United States. Chagas disease has also been detected in colonies of nonhuman primates (NHP) in Georgia and Texas, and heart abnormalities consistent with Chagas disease have occurred at our NHP center in Louisiana. To determine the level of T. cruzi infection, we serologically tested 2157 of the approximately 4200 NHP at the center; 34 of 2157 primates (1.6%) tested positive. Presence of the T. cruzi parasite was confirmed by hemoculture in 4 NHP and PCR of the cultured parasites. These results strongly suggest local transmission of T. cruzi, because most of the infected NHP were born and raised at this site. All 3 species of NHP tested yielded infected animals, with significantly higher infection prevalence in pig-tailed macaques, suggesting possible exploration of this species as a model organism. The local T. cruzi strain isolated during this study would enhance such investigations. The NHP at this center are bred for use in scientific research, and the effects of the Chagas parasite on infected primates could confuse the interpretation of other studies.Abbreviation: NHP, nonhuman primate; TNPRC, Tulane National Primate Research CenterMost nonhuman primates (NHP) used in research in the United States are now raised at 1 of 8 National Primate Research Centers or other institutions in the United States and are shipped as needed for studies, thereby avoiding the importation of pathogens from their native countries. However, knowledge about the infection status of these research animals within colonies in the United States is important with regard to colony health, the outcome of the scientific studies that use these NHP, and the safety of caregivers and laboratory workers.Infection with the hemoflagellate parasitic pathogen Trypanosoma cruzi, the causative agent of Chagas disease, has been reported sporadically in NHP colonies in the United States (33 Of the approximately 8 million people infected,26 20% to 30% will develop chronic disease; most of these persons will die of heart disease (70% to 85%), digestive disorders (15% to 30%), or neurologic disease (less than 5%). Most (80%) transmission occurs through insect vectors, specifically through contact of parasite-containing feces (which are deposited while the insect is taking a blood meal) with mammalian mucous membranes or through a break in the skin. In addition, approximately 1% to 12% of offspring of infected mothers will acquire the parasite by congenital transmission.7 Insect-vector-mediated autochthonous transmission to humans is rare in the United States, with only 7 documented cases.11,18 However, a robust sylvan cycle exists in the United States, including T. cruzi-infected triatomine insects and a variety of mammals.31,36

Table 1.

Reports of T. cruziinfection in nonhuman primates in the United States
SpeciesNo. of NHP infected/ no. testedState where infection identified (and likely acquired)Evidence (method of detection)Reference
Pileated gibbon (Hylobates pileatus)1/1LouisianaAmastigotes in myocardium at necropsy30
Rhesus macaque (Macaca mulatta)1/1Maryland (Texas or Georgia)Blood culture after inadvertently transferred to immunosuppressed NHP; recipient confirmed by blood smears, serology, and xenodiagnosis9
Rhesus macaque20/236 (8.5%)TexasIndex case: amastigotes observed at necropsy; 19 additional by serology17
Squirrel monkey (Saimiri sciureus)2/2LouisianaMicroscopy, hemoculture, and xenodiagnosis12
Yellow baboon (Papio cynocephalus)1/1TexasAmastigotes noted at necropsy13
Crested black macaque (Macaca nigra)1/1Oregon (Texas)Flagellates observed in spinal fluid; amastigotes in brain at necropsy; serology25
Lion-tailed macaque (Macaca silenus)7/11 (64%)GeorgiaHemoculture and PCR27
Ring-tailed lemur (Lemur catta)1/19 (5%)GeorgiaHemoculture and PCR27
Pig-tailed macaque (Macaca nemestrina)1/1Washington (Louisiana)Hemoculture, PCR, and serology29
Ring-tailed lemur (Lemur catta)21/41 (51%)GeorgiaHemoculture, PCR, and serology14
Black-eyed lemur (Eulemur macaco flavifrons)1/5 (20%)GeorgiaSerology14
Black and white ruffed lemur (Varecia variegata variegata)3/4 (75%)GeorgiaSerology14
Chimpanzee (Pan troglodytes)1TexasAmastigotes on necropsy, PCR, and immunohistochemistry5
Open in a separate windowT. cruzi was first identified in the United States in 1916 in the triatomine insect vector Triatoma protracta.19 Eleven species of triatomine insects live in the southern two-thirds of the United States, where they are commonly known as ‘kissing bugs’ (because as night feeders, they often feed on the face) or ‘cone-nosed bugs.’ T. sanguisuga is the species reported most commonly in Louisiana.10 On average, the prevalence of infection in the insect vectors is 25%,36 although much higher prevalence is reported in some areas, including Louisiana (56%).10 T. cruzi has been identified in more than 20 mammalian species across the southern United States; the most important of these mammals are rodents, raccoons, opossums, and armadillos.6,16 Recent studies showed that the highest prevalence of antibodies against T. cruzi occurred in raccoons (0% to 68%, range depends on state) and opossums (17% to 52%).6The first case of T. cruzi in a NHP in the United States occurred at the Delta Regional Primate Research Center (Covington, LA; now called the Tulane National Primate Research Center [TNPRC]), where a gibbon (Hylobates pileatus) from Malaysia died of symptoms of Chagas disease30. This case suggested local transmission because Chagas is endemic only to the Americas (相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号