Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 23(8): 1236-1245, 2022 08.
Article in English | MEDLINE | ID: mdl-35882933

ABSTRACT

Tissue-resident memory T cells (TRM cells) provide rapid and superior control of localized infections. While the transcription factor Runx3 is a critical regulator of CD8+ T cell tissue residency, its expression is repressed in CD4+ T cells. Here, we show that, as a direct consequence of this Runx3-deficiency, CD4+ TRM cells lacked the transforming growth factor (TGF)-ß-responsive transcriptional network that underpins the tissue residency of epithelial CD8+ TRM cells. While CD4+ TRM cell formation required Runx1, this, along with the modest expression of Runx3 in CD4+ TRM cells, was insufficient to engage the TGF-ß-driven residency program. Ectopic expression of Runx3 in CD4+ T cells incited this TGF-ß-transcriptional network to promote prolonged survival, decreased tissue egress, a microanatomical redistribution towards epithelial layers and enhanced effector functionality. Thus, our results reveal distinct programming of tissue residency in CD8+ and CD4+ TRM cell subsets that is attributable to divergent Runx3 activity.


Subject(s)
Immunologic Memory , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism
2.
Nat Immunol ; 19(2): 183-191, 2018 02.
Article in English | MEDLINE | ID: mdl-29311695

ABSTRACT

Although tissue-resident memory T cells (TRM cells) are critical in fighting infection, their fate after local pathogen re-encounter is unknown. Here we found that skin TRM cells engaged virus-infected cells, proliferated in situ in response to local antigen encounter and did not migrate out of the epidermis, where they exclusively reside. As a consequence, secondary TRM cells formed from pre-existing TRM cells, as well as from precursors recruited from the circulation. Newly recruited antigen-specific or bystander TRM cells were generated in the skin without displacement of the pre-existing TRM cell pool. Thus, pre-existing skin TRM cell populations are not displaced after subsequent infections, which enables multiple TRM cell specificities to be stably maintained within the tissue.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Skin/immunology , Animals , Cell Proliferation/physiology , Herpes Simplex/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
Immunity ; 45(4): 889-902, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27692609

ABSTRACT

In recent years, various intervention strategies have reduced malaria morbidity and mortality, but further improvements probably depend upon development of a broadly protective vaccine. To better understand immune requirement for protection, we examined liver-stage immunity after vaccination with irradiated sporozoites, an effective though logistically difficult vaccine. We identified a population of memory CD8+ T cells that expressed the gene signature of tissue-resident memory T (Trm) cells and remained permanently within the liver, where they patrolled the sinusoids. Exploring the requirements for liver Trm cell induction, we showed that by combining dendritic cell-targeted priming with liver inflammation and antigen recognition on hepatocytes, high frequencies of Trm cells could be induced and these cells were essential for protection against malaria sporozoite challenge. Our study highlights the immune potential of liver Trm cells and provides approaches for their selective transfer, expansion, or depletion, which may be harnessed to control liver infections or autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Animals , CD8-Positive T-Lymphocytes/parasitology , Culicidae , Dendritic Cells/immunology , Dendritic Cells/parasitology , Hepatocytes/immunology , Hepatocytes/parasitology , Liver/parasitology , Liver Diseases/immunology , Liver Diseases/parasitology , Malaria Vaccines/immunology , Mice , Plasmodium berghei/immunology , Sporozoites/immunology , Sporozoites/parasitology , Vaccination/methods
4.
PLoS Pathog ; 18(2): e1010185, 2022 02.
Article in English | MEDLINE | ID: mdl-35143591

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne viruses that are a major cause of infectious arthropathies worldwide, and recent outbreaks of chikungunya virus and Ross River virus (RRV) infections highlight the need for robust intervention strategies. Alphaviral arthritis can persist for months after the initial acute disease, and is mediated by cellular immune responses. A common strategy to limit inflammation and pathology is to dampen the overwhelming inflammatory responses by modulating proinflammatory cytokine pathways. Here, we investigate the contribution of interleukin-17 (IL-17), a cytokine involved in arthropathies such as rheumatoid arthritis, in the development RRV-induced arthritis and myositis. IL-17 was quantified in serum from RRV-infected patients, and mice were infected with RRV and joints and muscle tissues collected to analyse cellular infiltrates, tissue mRNA, cytokine expression, and joint and muscle histopathology. IL-17 expression was increased in musculoskeletal tissues and serum of RRV-infected mice and humans, respectively. IL-17-producing T cells and neutrophils contributed to the cellular infiltrate in the joint and muscle tissue during acute RRV disease in mice. Blockade of IL-17A/F using a monoclonal antibody (mAb) reduced disease severity in RRV-infected mice and led to decreased proinflammatory proteins, cellular infiltration in synovial tissues and cartilage damage, without affecting viral titers in inflamed tissues. IL-17A/F blockade triggered a shift in transcriptional profile of both leukocyte infiltrates and musculoskeletal stromal cells by downregulating proinflammatory genes. This study highlights a previously uncharacterized role for an effector cytokine in alphaviral pathology and points towards potential therapeutic benefit in targeting IL-17 to treat patients presenting with RRV-induced arthropathy.


Subject(s)
Arthritis, Rheumatoid/immunology , Immunity, Cellular , Inflammation/immunology , Interleukin-17/immunology , Myositis/immunology , Ross River virus/immunology , Alphavirus Infections/immunology , Alphavirus Infections/virology , Animals , Arthritis, Rheumatoid/virology , Chlorocebus aethiops , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Myositis/virology , Vero Cells , Viral Load
5.
Immunity ; 43(3): 554-65, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26297566

ABSTRACT

The dynamics of when and where CD4(+) T cells provide help for CD8(+) T cell priming and which dendritic cells (DCs) activate CD4(+) T cells in vivo after localized infection are poorly understood. By using a cutaneous herpes simplex virus infection model combined with intravital 2-photon imaging of the draining lymph node (LN) to concurrently visualize pathogen-specific CD4(+) and CD8(+) T cells, we found that early priming of CD4(+) T cells involved clustering with migratory skin DCs. CD8(+) T cells did not interact with migratory DCs and their activation was delayed, requiring later clustering interactions with LN-resident XCR1(+) DCs. CD4(+) T cells interacted with these late CD8(+) T cell clusters on resident XCR1(+) DCs. Together, these data reveal asynchronous T cell activation by distinct DC subsets and highlight the key role of XCR1(+) DCs as the central platform for cytotoxic T lymphocyte activation and the delivery of CD4(+) T cell help.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/immunology , Lymph Nodes/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Dendritic Cells/metabolism , Flow Cytometry , Fluorescent Dyes/chemistry , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Host-Pathogen Interactions/immunology , Lymph Nodes/cytology , Lymph Nodes/virology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Rhodamines/chemistry , Simplexvirus/immunology , Simplexvirus/physiology
6.
J Virol ; 96(17): e0099922, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000846

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Subject(s)
Alphavirus Infections , Arthritis , Myositis , Polysaccharides , Ross River virus , Skin , Alphavirus Infections/complications , Alphavirus Infections/immunology , Animals , Antiviral Agents/immunology , Arthritis/complications , Arthritis/immunology , Culicidae/virology , Dendritic Cells , Disease Models, Animal , Glycosylation , Humans , Mass Spectrometry , Mice , Monocytes , Myositis/complications , Myositis/immunology , Neutrophils , Polysaccharides/chemistry , Polysaccharides/immunology , Ross River virus/immunology , Skin/immunology , Skin/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
8.
Pak J Med Sci ; 39(3): 698-703, 2023.
Article in English | MEDLINE | ID: mdl-37250585

ABSTRACT

Objective: To determine that early needle-knife sphincterotomy does not increase post-ERCP pancreatitis in patients with difficult biliary cannulation as compared to standard cannulation. Method: This prospective single-centre cohort study was carried out at Pak Emirates Military Hospital from January 2021 to June 2021. Patients requiring ERCP were enrolled in the study (according to inclusion and exclusion criteria) and were subsequently allotted different groups according to the technique used for deep biliary cannulation. Qualitative data was analysed using frequencies and chi square statistics whereas, quantitative data was analysed using mean±SD and one way ANOVA test. Result: The cohort included 114 patients with 52.6% male patients and predominance of relatively younger age group (31-45 years). The most common indication for ERCP was choledocholithiasis (36%) with an overall technical success rate of 96%. Deep cannulation was achieved either through standard cannulation (56%), double guidewire and/or pancreatic stent assisted (10.5%), use of early Needle-Knife Sphincterotomy (19%), NKS as a last resort (3.5%) or Transpancreatic Stenting and/or combined sphincterotomy (6%). Pancreatitis as a complication occurred in 4(3.5%) patients, bleeding in 2(1.8%), on-table desaturation in 2(1.8%) and perforation in 1(0.9%) patient. The occurrence of pancreatitis was only related significantly to inadvertent PD cannulation through univariate and logistic regression analysis whereas, multiple cannulations (>5), gender, age, classification of papilla and the use of early NKS had no impact on pancreatitis or the occurrence of other complications. Conclusion: NKS is an effective and safe modality for deep biliary cannulation and achieving technical success where cannulation is deemed difficult and does not increase the risk of PEP if done by experienced endoscopists in high volume centres.

9.
Pak J Med Sci ; 39(5): 1232-1237, 2023.
Article in English | MEDLINE | ID: mdl-37680842

ABSTRACT

Objective: To compare the efficacy and safety of left lateral decubitus versus prone position during endoscopic retrograde cholangio-pancreaticography (ERCP). Methods: This prospective single-centre cohort study was carried out at Pak Emirates Military Hospital from January to June 2021. Patients requiring ERCP were subsequently allotted LL or PP group randomly (unequal randomization) except patients with recent abdominal surgery, in-dwelling catheters, raised intra-abdominal pressure, cervical spine abnormalities and limb contractures. Qualitative data was analysed using frequencies and chi square statistics whereas, quantitative data was analysed using mean±SD and student T or Mann Whitney U-test. Results: A total of 114 patients were enrolled according to the inclusion criteria with 62(54%) males and majority of the patients (42%) belonging to the age group 31-45 years. The most common ERCP indication was choledocholithiasis (36%). Technical success was achieved in 109(96%) patients with no statistically significant difference between the two groups. The total time of procedure, time for deep cannulation, time for acquiring therapeutic goal and ERCP complexity level were all similar between the two groups. The rate of inadvertent PD cannulation and PEP were relatively higher for the PP group but were statistically non-significant through univariate and logistic regression analyses and the only outcome measure that showed significance was multiple cannulations in the PP group. Conclusion: The study concludes that LL is non-inferior to PP and both positions have comparable outcomes with non-significant differences in terms of technical success rate, complications (specifically PEP), total procedure time, time required for deep cannulation and attainment of goal, ERCP complexity level and inadvertent PD cannulation.

10.
J Immunol ; 205(7): 1842-1856, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32839238

ABSTRACT

Follicular dendritic cells and macrophages have been strongly implicated in presentation of native Ag to B cells. This property has also occasionally been attributed to conventional dendritic cells (cDC) but is generally masked by their essential role in T cell priming. cDC can be divided into two main subsets, cDC1 and cDC2, with recent evidence suggesting that cDC2 are primarily responsible for initiating B cell and T follicular helper responses. This conclusion is, however, at odds with evidence that targeting Ag to Clec9A (DNGR1), expressed by cDC1, induces strong humoral responses. In this study, we reveal that murine cDC1 interact extensively with B cells at the border of B cell follicles and, when Ag is targeted to Clec9A, can display native Ag for B cell activation. This leads to efficient induction of humoral immunity. Our findings indicate that surface display of native Ag on cDC with access to both T and B cells is key to efficient humoral vaccination.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/metabolism , Receptors, Immunologic/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Antigen Presentation , Autoantigens/immunology , Autoantigens/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Immunity, Humoral , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Vaccination
11.
Cell Immunol ; 350: 103913, 2020 04.
Article in English | MEDLINE | ID: mdl-30992120

ABSTRACT

Intravital imaging of cutaneous immune responses has revealed intricate links between the skin's structural properties, the immune cells that reside therein, and the carefully orchestrated migratory dynamics that enable rapid sensing and subsequent elimination of skin pathogens. In particular, the development of 2-photon intravital microscopy (2P-IVM), which enables the excitation of fluorescent molecules within deep tissue with minimal light scattering and tissue damage, has proven an invaluable tool in the characterization of different cell subset's roles in skin infection. The ability to visualize cells, tissue structures, pathogens and track migratory dynamics at designated times following infection, or during inflammatory responses has been crucial in defining how immune responses in the skin are coordinated, either locally or in concert with circulating immune cells. Skin pathogens affect millions of people worldwide, and skin infections leading to cutaneous pathology have a considerable impact on the quality of life and longevity of people affected. In contrast, pathogens that infect the skin to later cause systemic illness, such as malaria parasites and a variety of arthropod-borne viruses, or infection in distant anatomical sites are a significant cause of morbidity and mortality worldwide. Here, we review recent advances and seminal studies that employed intravital imaging to characterize key immune response mechanisms in the context of viral, bacterial and parasitic skin infections, and provide insights on skin pathogens of global significance that would benefit from such investigative approaches.


Subject(s)
Intravital Microscopy/methods , Skin Diseases, Infectious/diagnostic imaging , Skin Diseases, Infectious/immunology , Animals , Dermis/diagnostic imaging , Dermis/immunology , Epidermis/diagnostic imaging , Epidermis/immunology , Humans , Keratinocytes/immunology , Keratinocytes/metabolism , Mice , Microscopy, Fluorescence, Multiphoton/methods , Quality of Life , Skin/diagnostic imaging , Skin/immunology , Skin Diseases, Infectious/microbiology
12.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142130

ABSTRACT

Resolution of virus infections depends on the priming of virus-specific CD8+ T cells by dendritic cells (DC). While this process requires major histocompatibility complex (MHC) class I-restricted antigen presentation by DC, the relative contribution to CD8+ T cell priming by infected DC is less clear. We have addressed this question in the context of a peripheral infection with herpes simplex virus 1 (HSV). Assessing the endogenous, polyclonal HSV-specific CD8+ T cell response, we found that effective in vivo T cell priming depended on the presence of DC subsets specialized in cross-presentation, while Langerhans cells and plasmacytoid DC were dispensable. Utilizing a novel mouse model that allows for the in vivo elimination of infected DC, we also demonstrated in vivo that this requirement for cross-presenting DC was not related to their infection but instead reflected their capacity to cross-present HSV-derived antigen. Taking the results together, this study shows that infected DC are not required for effective CD8+ T cell priming during a peripheral virus infection.IMPORTANCE The ability of some DC to present viral antigen to CD8+ T cells without being infected is thought to enable the host to induce killer T cells even when viruses evade or kill infected DC. However, direct experimental in vivo proof for this notion has remained elusive. The work described in this study characterizes the role that different DC play in the induction of virus-specific killer T cell responses and, critically, introduces a novel mouse model that allows for the selective elimination of infected DC in vivo Our finding that HSV-specific CD8+ T cells can be fully primed in the absence of DC infection shows that cross-presentation by DC is indeed sufficient for effective CD8+ T cell priming during a peripheral virus infection.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Herpes Simplex/immunology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Flow Cytometry , Herpesvirus 1, Human , Mice , Mice, Inbred C57BL
13.
J Immunol ; 199(7): 2451-2459, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28855310

ABSTRACT

Infection or inflammation of the skin recruits effector CD8+ T cells that enter the epidermis and form populations of long-lived tissue-resident memory T (TRM) cells. These skin TRM cells migrate within the constrained epidermal environment by extending multiple dynamic dendritic projections and squeezing between keratinocytes to survey the tissue for pathogens. In this study, we examined the signals required for this distinctive mode of T cell migration by inhibiting key cytoskeletal components and performing intravital two-photon microscopy to visualize TRM cell behavior. We found that TRM cell motility and dendrite formation required an intact actomyosin cytoskeleton and the Rho-associated coiled-coil containing kinases. We also identified an essential role for microtubules for maintaining skin TRM cell shape and cellular integrity. We reveal a role for pertussis toxin-sensitive signaling for TRM cell dendritic morphology and migration that is independent of CXCR3 or CXCR6, or the skin-selective chemokine receptors CCR10 and CCR8. However, we found that CXCR6 and CCR10 expression by CD8+ T cells was required for the optimal formation of memory T cell populations, in particular TRM cell populations in the skin.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Cell Movement , Epidermis/immunology , Immunologic Memory , Receptors, Chemokine/metabolism , Skin/immunology , Actomyosin/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Epidermal Cells , Intravital Microscopy/methods , Mice , Microtubules/metabolism , Pertussis Toxin/metabolism , Receptors, CCR10/genetics , Receptors, CCR10/metabolism , Receptors, CCR8/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR6 , Receptors, Chemokine/genetics , Signal Transduction , Skin/anatomy & histology , Skin/cytology , rho-Associated Kinases/metabolism
14.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24025841

ABSTRACT

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/microbiology , Signal Transduction , Virulence Factors/metabolism , Animals , Caspase 8/metabolism , Cell Death , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/physiology , Enteropathogenic Escherichia coli/pathogenicity , Enzyme Activation , Escherichia coli Infections/pathology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , N-Acetylglucosaminyltransferases/metabolism , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/metabolism , fas Receptor/deficiency , fas Receptor/metabolism
15.
Clin Sci (Lond) ; 131(14): 1713-1721, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28667069

ABSTRACT

Since its discovery in 2001, human metapneumovirus (hMPV) has been identified as an important cause of respiratory tract infection in young children, second only to the closely related respiratory syncytial virus (RSV). Clinical evidence suggests that hMPV is associated with acute exacerbations of asthma in both children and adults, and may play a role in initiating asthma development in children. Animal models have demonstrated that airway hyperresponsiveness (AHR) and inflammation are triggered following hMPV infection, and hMPV is able to persist in vivo by inhibiting innate immune responses and causing aberrant adaptive responses. In this review, we discuss the prevalence of hMPV infection in pediatric and adult populations and its potential role in asthma exacerbation. We also review recent advances made in animal models to determine immune responses following hMPV infection, and compare to what is known about RSV.


Subject(s)
Asthma/virology , Metapneumovirus , Paramyxoviridae Infections/complications , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus, Human , Acute Disease , Animals , Disease Models, Animal , Humans , Immunity, Innate , Paramyxoviridae Infections/immunology , Respiratory Syncytial Virus Infections/immunology
16.
J Immunol ; 195(3): 1006-14, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26101322

ABSTRACT

Targeting Ags to dendritic cell (DC) surface receptors can induce a variety of responses depending on the DC type targeted, the receptor targeted, and the adjuvant used. Clec9A (DNGR-1), which is expressed by CD8(+) DCs, has been shown to bind F-actin exposed on damaged cells. Targeting Ag to this receptor in mice and nonhuman primates induces strong humoral immunity even in the absence of adjuvant, a process seen for a few select DC receptors. In contrast with other receptors, however, targeting Clec9A induces long-lived, affinity-matured Ab responses that are associated with efficient CD4(+) T cell responses shown to possess properties of follicular Th cells (TFH). In this article, we provide definitive evidence that Clec9A targeting promotes the development of TFH by showing that responding CD4 T cells express CXCR5, PD1, the TFH transcription factor Bcl6, and the cytokine IL-21, and that these cells localize to germinal centers. Furthermore, we extend studies from the model Ag OVA to the viral Ag glycoprotein D of HSV-1 and examine the capacity of primed TFH to form functional memory. We show that targeting glycoprotein D to Clec9A even in the absence of adjuvant induced long-lived memory CXCR5(+) PD1(hi) CD4(+) T cells that proliferated extensively upon secondary challenge and rapidly developed into effector TFH. This was associated with enhanced germinal center B cell responses and accelerated Ab production. Our study indicates that targeting Ags to Clec9A in the absence of adjuvant routinely generates TFH responses that form long-lived memory capable of robust secondary TFH responses.


Subject(s)
Dendritic Cells/immunology , Immunologic Memory/immunology , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antigens/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , DNA-Binding Proteins/biosynthesis , Germinal Center/cytology , Germinal Center/immunology , Interleukin-21 Receptor alpha Subunit/genetics , Interleukins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Programmed Cell Death 1 Receptor/biosynthesis , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR5/biosynthesis , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/transplantation , Viral Envelope Proteins/immunology
17.
Nature ; 477(7363): 216-9, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841802

ABSTRACT

Infections localized to peripheral tissues such as the skin result in the priming of T-cell responses that act to control pathogens. Activated T cells undergo migrational imprinting within the draining lymph nodes, resulting in memory T cells that provide local and systemic protection. Combinations of migrating and resident memory T cells have been implicated in long-term peripheral immunity, especially at the surfaces that form pathogen entry points into the body. However, T-cell immunity consists of separate CD4(+) helper T cells and CD8(+) killer T cells, with distinct effector and memory programming requirements. Whether these subsets also differ in their ability to form a migrating pool involved in peripheral immunosurveillance or a separate resident population responsible for local infection control has not been explored. Here, using mice, we show key differences in the migration and tissue localization of memory CD4(+) and CD8(+) T cells following infection of the skin by herpes simplex virus. On resolution of infection, the skin contained two distinct virus-specific memory subsets; a slow-moving population of sequestered CD8(+) T cells that were resident in the epidermis and confined largely to the original site of infection, and a dynamic population of CD4(+) T cells that trafficked rapidly through the dermis as part of a wider recirculation pattern. Unique homing-molecule expression by recirculating CD4(+) T effector-memory cells mirrored their preferential skin-migratory capacity. Overall, these results identify a complexity in memory T-cell migration, illuminating previously unappreciated differences between the CD4(+) and CD8(+) subsets.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Immunologic Memory , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , E-Selectin/metabolism , Genomic Imprinting , Herpes Simplex/immunology , Herpes Simplex/virology , Immunologic Surveillance/immunology , Ligands , Mice , P-Selectin/metabolism , Simplexvirus/immunology , Skin/cytology , Skin/immunology , Skin/virology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
18.
Proc Natl Acad Sci U S A ; 111(14): 5307-12, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706879

ABSTRACT

Barrier tissues such as the skin contain various populations of immune cells that contribute to protection from infections. These include recently identified tissue-resident memory T cells (TRM). In the skin, these memory CD8(+) T cells reside in the epidermis after being recruited to this site by infection or inflammation. In this study, we demonstrate prolonged persistence of epidermal TRM preferentially at the site of prior infection despite sustained migration. Computational simulation of TRM migration within the skin over long periods revealed that the slow rate of random migration effectively constrains these memory cells within the region of skin in which they form. Notably, formation of TRM involved a concomitant local reduction in dendritic epidermal γδ T-cell numbers in the epidermis, indicating that these populations persist in mutual exclusion and may compete for local survival signals. Accordingly, we show that expression of the aryl hydrocarbon receptor, a transcription factor important for dendritic epidermal γδ T-cell maintenance in skin, also contributes to the persistence of skin TRM. Together, these data suggest that skin tissue-resident memory T cells persist within a tightly regulated epidermal T-cell niche.


Subject(s)
Immunologic Memory , Skin/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Cell Movement , Flow Cytometry , Mice , Mice, Inbred C57BL , Microscopy/methods
19.
J Virol ; 89(3): 1564-78, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25410867

ABSTRACT

UNLABELLED: Human respiratory syncytial virus (RSV) is a major cause of morbidity and severe lower respiratory tract disease in the elderly and very young, with some infants developing bronchiolitis, recurrent wheezing, and asthma following infection. Previous studies in humans and animal models have shown that vaccination with formalin-inactivated RSV (FI-RSV) leads to prominent airway eosinophilic inflammation following RSV challenge; however, the roles of pulmonary eosinophilia in the antiviral response and in disease pathogenesis are inadequately understood. In vivo studies in mice with eotaxin and/or interleukin 5 (IL-5) deficiency showed that FI-RSV vaccination did not lead to enhanced pulmonary disease, where following challenge there were reduced pulmonary eosinophilia, inflammation, Th2-type cytokine responses, and altered chemokine (TARC and CCL17) responses. In contrast to wild-type mice, RSV was recovered at high titers from the lungs of eotaxin- and/or IL-5-deficient mice. Adoptive transfer of eosinophils to FI-RSV-immunized eotaxin- and IL-5-deficient (double-deficient) mice challenged with RSV was associated with potent viral clearance that was mediated at least partly through nitric oxide. These studies show that pulmonary eosinophilia has dual outcomes: one linked to RSV-induced airway inflammation and pulmonary pathology and one with innate features that contribute to a reduction in the viral load. IMPORTANCE: This study is critical to understanding the mechanisms attributable to RSV vaccine-enhanced disease. This study addresses the hypothesis that IL-5 and eotaxin are critical in pulmonary eosinophil response related to FI-RSV vaccine-enhanced disease. The findings suggest that in addition to mediating tissue pathology, eosinophils within a Th2 environment also have antiviral activity.


Subject(s)
Eosinophils/immunology , Lung/immunology , Lung/pathology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Female , Lung/virology , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Vaccines, Inactivated/immunology , Viral Load
20.
Heliyon ; 10(14): e34283, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39114011

ABSTRACT

Sickle cell disease (SCD) is a hereditary blood condition characterized by abnormal hemoglobin, leading to chronic hemolysis and vaso-occlusive complications. Caregivers of children with SCD often experience significant distress, akin to post-traumatic stress disorder (PTSD). This study aimed to measure the degree of trauma and post-traumatic growth among parents (caregivers) of children with SCD in the Kingdom of Saudi Arabia. A total of 294 primary caregivers were recruited for this study, through direct phone calls and online outreach using contact information obtained from their primary treating physician in Maternity and Children Hospitals and the Hereditary Blood Diseases Center in Al-Ahsa. Inclusion criteria required caregivers not to be receiving professional mental health care and to have a child with SCD below the age of 18. Results indicate that caregiver gender significantly affected IESR scores, with mothers reporting higher scores than fathers. Family income had a significant effect on IESR as well. In terms of education level, higher-educated caregivers were less likely to experience severe trauma. Significant differences emerged between online and phone interview participants, with online respondents reporting higher post-traumatic growth and higher trauma levels. This study represents a crucial step in understanding the challenges faced by caregivers of children with SCD in Al-Ahsa, Saudi Arabia. However, the study has limitations, including a substantial portion of the sample being from a single clinic and a cross-sectional design.

SELECTION OF CITATIONS
SEARCH DETAIL