1
|
Bozzano F, Perrone C, Moretta L, De Maria A. NK Cell Precursors in Human Bone Marrow in Health and Inflammation. Front Immunol 2019; 10:2045. [PMID: 31555276 PMCID: PMC6724745 DOI: 10.3389/fimmu.2019.02045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/13/2019] [Indexed: 12/22/2022] Open
Abstract
NK cells are generated from hematopoietic stem cells (HSC) residing in the bone marrow (BM), similar to other blood cells. Development toward mature NK cells occurs largely outside the BM through travel of CD34+ and other progenitor intermediates toward secondary lymphoid organs. The BM harbors multipotent CD34+ common lymphoid progenitors (CLPs) that generate T, B, NK, and Dendritic Cells and are devoid of erythroid, myeloid, and megakaryocytic potential. Over recent years, there has been a quest for single-lineage progenitors predominantly with the objective of manipulation and intervention in mind, which has led to the identification of unipotent NK cell progenitors devoid of other lymphoid lineage potential. Research efforts for the study of lymphopoiesis have almost exclusively concentrated on healthy donor tissues and on repopulation/transplant models. This has led to the widely accepted assumption that lymphopoiesis during disease states reflects the findings of these models. However, compelling evidences in animal models show that inflammation plays a fundamental role in the regulation of HSC maturation and release in the BM niches through several mechanisms including modulation of the CXCL12-CXCR4 expression. Indeed, recent findings during systemic inflammation in patients provide evidence that a so-far overlooked CLP exists in the BM (Lin−CD34+DNAM-1brightCXCR4+) and that it overwhelmingly exits the BM during systemic inflammation. These “inflammatory” precursors have a developmental trajectory toward surprisingly functional NK and T cells as reviewed here and mirror the steady state maintenance of the NK cell pool by CD34+DNAM-1−CXCR4− precursors. Our understanding of NK cell precursor development may benefit from including a distinct “inflammatory” progenitor modeling of lymphoid precursors, allowing rapid deployment of specialized Lin−CD34+DNAM-1brightCXCR4+ -derived resources from the BM.
Collapse
Affiliation(s)
| | - Carola Perrone
- Centro di Eccellenza per la Ricerca Biomedica, Università di Genova, Genoa, Italy
| | | | - Andrea De Maria
- Centro di Eccellenza per la Ricerca Biomedica, Università di Genova, Genoa, Italy.,Clinica Malattie Infettive, Ospedale Policlinico S. Martino IRCCS, Genoa, Italy.,Dipartimento di Scienze Dell Salute, Università Degli Studi di Genova, Genoa, Italy
| |
Collapse
|
2
|
Golden-Mason L, McMahan RH, Kriss MS, Kilgore AL, Cheng L, Dran RJ, Wieland A, Rosen HR. Early and late changes in natural killer cells in response to ledipasvir/sofosbuvir treatment. Hepatol Commun 2018; 2:364-375. [PMID: 29619416 PMCID: PMC5880201 DOI: 10.1002/hep4.1166] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/17/2018] [Accepted: 02/10/2018] [Indexed: 01/01/2023] Open
Abstract
Chronic hepatitis C virus (HCV) infection is characterized by dysregulated natural killer (NK) cell responses. NKs play a critical role in achieving sustained responses to interferon (IFN)‐α‐based therapy. Rapid sustained HCV‐RNA clearance is now achieved with direct‐acting antivirals (DAAs). Studies of patients receiving first‐wave DAAs suggest NK functional restoration. Here, we investigate the effect of mainstream DAA treatment on NKs. We collected a prospective cohort of male HCV genotype 1‐infected patients treated with ledipasvir/sofosbuvir (n = 22). Peripheral blood was obtained at treatment start, week 2 (W2), W4, W8, and W12 of treatment and 12 weeks posttreatment. Flow cytometry was used to characterize NK responses to therapy. Mean baseline viral load was 1.75 million IU/mL. All subjects rapidly cleared virus and remained HCV RNA‐negative posttreatment. No change was seen in total NK levels; however, the frequency of immature NKs (clusters of differentiation [CD]56bright) decreased by W2 and was maintained throughout the study. Phenotypic changes were evident by W2/W4, coincident with rapid viral clearance. At W2, T‐cell immunoglobulin and mucin‐domain containing‐3 and CD161 were significantly increased, returning to pretreatment levels by W12. Some changes were not evident until late (W12 or posttreatment). Down‐regulation of several activation markers, including NKp30 and tumor necrosis factor–related apoptosis‐inducing ligand, was observed at W12 and sustained posttreatment. No difference was observed in IFN‐γ production or cytokine‐mediated killing of NK‐sensitive cell line K562 posttreatment compared to pretreatment. Conclusion: Our phenotype data suggest transient activation followed by dampening of NK cell activity to pretreatment levels. The NK response to ledipasvir/sofosbuvir is not universal in a homogeneous patient cohort. More studies are needed to elucidate the roles of NK cells in IFN‐free regimens, which will have implications for protection from re‐infection and fibrosis progression. (Hepatology Communications 2018;2:364‐375)
Collapse
Affiliation(s)
- Lucy Golden-Mason
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Rachel H McMahan
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Michael S Kriss
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Alexandra L Kilgore
- Department of Pediatrics, School of Medicine University of Colorado Denver Aurora CO.,Children's Hospital Colorado, Section of Pediatric Gastroenterology, Hepatology, and Nutrition Digestive Health Institute Aurora CO
| | - Linling Cheng
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Rachael J Dran
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Amanda Wieland
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO
| | - Hugo R Rosen
- Division of Gastroenterology and Hepatology University of Colorado Denver Aurora CO.,Eastern-Colorado Denver Veteran's Affairs Medical Center Denver CO
| |
Collapse
|
3
|
Ascierto ML, Idowu MO, Zhao Y, Khalak H, Payne KK, Wang XY, Dumur CI, Bedognetti D, Tomei S, Ascierto PA, Shanker A, Bear HD, Wang E, Marincola FM, De Maria A, Manjili MH. Molecular signatures mostly associated with NK cells are predictive of relapse free survival in breast cancer patients. J Transl Med 2013; 11:145. [PMID: 23758773 PMCID: PMC3694475 DOI: 10.1186/1479-5876-11-145] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/04/2013] [Indexed: 01/10/2023] Open
Abstract
Background Recent observations suggest that immune-mediated tissue destruction is dependent upon coordinate activation of immune genes expressed by cells of the innate and adaptive immune systems. Methods Here, we performed a retrospective pilot study to investigate whether the coordinate expression of molecular signature mostly associated with NK cells could be used to segregate breast cancer patients into relapse and relapse-free outcomes. Results By analyzing primary breast cancer specimens derived from patients who experienced either 58–116 months (~5-9 years) relapse-free survival or developed tumor relapse within 9–76 months (~1-6 years) we found that the expression of molecules involved in activating signaling of NK cells and in NK cells: target interaction is increased in patients with favorable prognosis. Conclusions The parameters identified in this study, together with the prognostic signature previously reported by our group, highlight the cooperation between the innate and adaptive immune components within the tumor microenvironment.
Collapse
|
4
|
Cell mediated innate responses of cattle and swine are diverse during foot-and-mouth disease virus (FMDV) infection: a unique landscape of innate immunity. Immunol Lett 2013; 152:135-43. [PMID: 23727070 PMCID: PMC7112845 DOI: 10.1016/j.imlet.2013.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/06/2013] [Accepted: 05/13/2013] [Indexed: 12/21/2022]
Abstract
Harnessing the innate immunity can protect domestic animals from viruses. Innate immune cells have potential capacity to afford protection against infection. Understanding the innate and adaptive immunity will aid rational vaccine design.
Pathogens in general and pathogenic viruses in particular have evolved a myriad of mechanisms to escape the immune response of mammalian species. Viruses that cause acute disease tend to bear characteristics that make them very contagious, as survival does not derive from chronicity of infection, but spread of disease throughout the herd. Foot-and-mouth disease virus (FMDV) is one of the most contagious viruses known. Upon infection of susceptible species, cloven-hoofed animals, the virus proliferates rapidly and causes a vesicular disease within 2–4 days. Disease symptoms resolve by 10 days to 2 weeks and in most cases, virus can no longer be detected. Periods of fever and viremia are usually brief, 1–3 days. In vivo control of virus infection and clearance of the virus during and following acute infection is of particular interest. The interaction of this virus with cells mediating the early, innate immune response has been analyzed in a number of recent studies. In most reports, the virus has a distinct inhibitory effect on the response of cells early in infection. Here we review these new data and discuss the dynamics of the interaction of virus with different cell types mediating the immune response to infection.
Collapse
|
5
|
The effect of alloferon on the enhancement of NK cell cytotoxicity against cancer via the up-regulation of perforin/granzyme B secretion. Immunobiology 2013; 218:1026-33. [PMID: 23434435 DOI: 10.1016/j.imbio.2012.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 12/28/2012] [Accepted: 12/29/2012] [Indexed: 01/09/2023]
Abstract
Alloferon is a novel immunomodulatory peptide originally isolated from infected insects. It has anti-viral and anti-tumor effects via the activation of NK cells. However, specific mechanisms leading to NK cell activation and anti-tumor responses yet to be clarified. In this study, we demonstrate that alloferon increases killing activity of NK cells to cancer cells via the up-regulation of the expression of NK-activating receptors, 2B4. In addition, the production of IFN-γ and TNF-α and granule exocytosis from NK cells against cancer cell were increased by alloferon. Lastly, the anti-tumor effect of alloferon was confirmed in vivo to demonstrate effective retardation of tumor growth in the human-to-mouse xenograft model. All taken together, these results suggest that alloferon has anti-tumor effects through up-regulation of NK-activating receptor 2B4 and the enhancement of granule exocytosis from NK cells.
Collapse
|
6
|
Mariee NG, Tuckerman E, Laird S, Li TC. The correlation of autoantibodies and uNK cells in women with reproductive failure. J Reprod Immunol 2012; 95:59-66. [PMID: 22884101 DOI: 10.1016/j.jri.2012.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 04/16/2012] [Accepted: 04/23/2012] [Indexed: 12/19/2022]
Abstract
There is conflicting evidence on the role of autoimmune disorders in reproductive failure, including recurrent miscarriage (RM) and recurrent implantation failure (RIF), after in vitro fertilisation (IVF). Several commonly studied autoimmune markers in women with reproductive failure include antiphospholipid antibodies (APAs), thyroid peroxidase antibodies (TPA) and uterine natural killer (uNK) cells. However, there have not been any studies that have examined the correlation of these markers in women with reproductive failure. To determine if women who tested positive for autoantibodies (APA and thyroid peroxidase antibodies) have significantly higher uNK cell numbers than women who tested negative for these antibodies, the percentage of stromal cells that stained positive for CD56 was identified by immunocytochemistry in endometrial biopsies from 42 women with unexplained RM (29 women tested negative for autoantibodies and 13 women tested positive for autoantibodies) and 40 women with unexplained RIF (30 women tested negative for autoantibodies and 10 women tested positive for autoantibodies). Biopsies were obtained on days LH+7 to LH+9. There was no significant difference in uNK cell numbers between women with unexplained RM who tested negative and those who tested positive for autoantibodies. Similarly, there was no significant difference in uNK cell numbers between women with unexplained RIF who tested negative and those who tested positive for autoantibodies. In women with reproductive failure the presence of autoantibodies does not appear to affect the numbers of uNK cells in the endometrium around the time of implantation.
Collapse
Affiliation(s)
- N G Mariee
- Academic Unit of Reproductive and Developmental Medicine, The University of Sheffield, Sheffield S10 2SF, UK.
| | | | | | | |
Collapse
|
7
|
El Costa H, Casemayou A, Aguerre-Girr M, Rabot M, Berrebi A, Parant O, Clouet-Delannoy M, Lombardelli L, Jabrane-Ferrat N, Rukavina D, Bensussan A, Piccinni MP, Le Bouteiller P, Tabiasco J. Critical and differential roles of NKp46- and NKp30-activating receptors expressed by uterine NK cells in early pregnancy. THE JOURNAL OF IMMUNOLOGY 2008; 181:3009-17. [PMID: 18713971 DOI: 10.4049/jimmunol.181.5.3009] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In early human pregnancy, uterine decidual NK cells (dNK) are abundant and considered as cytokine producers but poorly cytotoxic despite their cytolytic granule content, suggesting a negative control of this latter effector function. To investigate the basis of this control, we examined the relative contribution to the cytotoxic function of different activating receptors expressed by dNK. Using a multicolor flow cytometry analysis, we found that freshly isolated dNK exhibit a unique repertoire of activating and inhibitory receptors, identical among all the donors tested. We then demonstrated that in fresh dNK, mAb-specific engagement of NKp46-, and to a lesser extent NKG2C-, but not NKp30-activating receptors induced intracellular calcium mobilization, perforin polarization, granule exocytosis and efficient target cell lysis. NKp46-mediated cytotoxicity is coactivated by CD2 but dramatically blocked by NKG2A coengagement, indicating that the dNK cytotoxic potential could be tightly controlled in vivo. We finally found that in dNK, mAb-specific engagement of NKp30, but not NKp46, triggered the production of IFN-gamma, TNF-alpha, MIP-1alpha, MIP-1beta, and GM-CSF proinflammatory molecules. These data demonstrate a differential, controlled role of NKp46- and NKp30-activating receptors expressed by dNK that could be critical for the outcome of pregnancy and the killing of uterine cells infected by pathogens.
Collapse
Affiliation(s)
- Hicham El Costa
- Institut National de la Santé et de la Recherche Médicale; U563. France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Influenza virus infection augments NK cell inhibition through reorganization of major histocompatibility complex class I proteins. J Virol 2008; 82:8030-7. [PMID: 18524833 DOI: 10.1128/jvi.00870-08] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The killing by natural killer (NK) cells is regulated by inhibitory, costimulatory, and activating receptors. The inhibitory receptors recognize mainly major histocompatibility complex (MHC) class I molecules, while the activating NK receptors recognize stress-induced ligands and viral products. Thus, changes in the expression of the various inhibitory and activating ligands will determine whether target cells will be killed or protected. Here, we demonstrate that after influenza virus infection the binding of the two NK inhibitory receptors, KIR2DL1 and the LIR1, to the infected cells is specifically increased. The increased binding occurs shortly after the influenza virus infection, prior to the increased recognition of the infected cells by the NK activating receptor, NKp46. We also elucidate the mechanism responsible for this effect and demonstrate that, after influenza virus infection, MHC class I proteins redistribute on the cell surface and accumulate in the lipid raft microdomains. Such redistribution allows better recognition by the NK inhibitory receptors and consequently increases resistance to NK cell attack. In contrast, T-cell activity was not influenced by the redistribution of MHC class I proteins. Thus, we present here a novel mechanism, developed by the influenza virus, of inhibition of NK cell cytotoxicity, through the reorganization of MHC class I proteins on the cell surface.
Collapse
|
9
|
Gruda R, Achdout H, Stern-Ginossar N, Gazit R, Betser-Cohen G, Manaster I, Katz G, Gonen-Gross T, Tirosh B, Mandelboim O. Intracellular cysteine residues in the tail of MHC class I proteins are crucial for extracellular recognition by leukocyte Ig-like receptor 1. THE JOURNAL OF IMMUNOLOGY 2007; 179:3655-61. [PMID: 17785801 DOI: 10.4049/jimmunol.179.6.3655] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activity of NK cells is regulated by activating receptors that recognize mainly stress-induced ligands and by inhibitory receptors that recognize mostly MHC class I proteins on target cells. Comparing the cytoplasmic tail sequences of various MHC class I proteins revealed the presence of unique cysteine residues in some of the MHC class I molecules which are absent in others. To study the role of these unique cysteines, we performed site specific mutagenesis, generating MHC class I molecules lacking these cysteines, and demonstrated that their expression on the cell surface was impaired. Surprisingly, we demonstrated that these cysteines are crucial for the surface binding of the leukocyte Ig-like receptor 1 inhibitory receptor to the MHC class I proteins, but not for the binding of the KIR2DL1 inhibitory receptor. In addition, we demonstrated that the cysteine residues in the cytoplasmic tail of MHC class I proteins are crucial for their egress from the endoplasmic reticulum and for their palmitoylation, thus probably affecting their expression on the cell surface. Finally, we show that the cysteine residues are important for proper extracellular conformation. Thus, although the interaction between leukocyte Ig-like receptor 1 and MHC class I proteins is formed between two extracellular surfaces, the intracellular components of MHC class I proteins play a crucial role in this recognition.
Collapse
Affiliation(s)
- Raizy Gruda
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Ben-Eliyahu S, Page GG, Schleifer SJ. Stress, NK cells, and cancer: Still a promissory note. Brain Behav Immun 2007; 21:881-7. [PMID: 17662574 DOI: 10.1016/j.bbi.2007.06.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 06/18/2007] [Accepted: 06/19/2007] [Indexed: 12/19/2022] Open
Abstract
Although the last decades have provided ample evidence for deleterious effects of stress on immunity and on cancer development and suggested mediating mechanisms, no psychoneuroimmunology (PNI)-related intervention has become a standard of care in conventional cancer treatment. We believe the reasons for this include the unique nature of cancer evolvement and interactions with the immune system, and the many conceptual and technical obstacles to studying stress effects on immune activity and their implications for human resistance to malignancy. However, the numerous and diverse interactions between malignant tissue and immunocytes are now better understood, and suggestions can be made with respect to certain critical periods to be investigated in cancer-PNI research. Animal models of cancer progression are instrumental in suggesting neuroendocrine and immunological mediators of stress effects on specific aspects of cancer progression, especially with respect to the role of NK cell activity. The ultimate clinical relevance, however, must be tested in cancer patients. Recent animal studies suggest a role for the sympathetic nervous system in mediating biologically relevant stress effects on immunity and on tumor progression. Related interventions can now be tested in patients to support or refute the promise of such studies.
Collapse
Affiliation(s)
- Shamgar Ben-Eliyahu
- Neuroimmunology Research Unit, Department of Psychology, Tel Aviv University, Tel Aviv 69978, Israel.
| | | | | |
Collapse
|
11
|
Kalinski P, Mailliard RB, Giermasz A, Zeh HJ, Basse P, Bartlett DL, Kirkwood JM, Lotze MT, Herberman RB. Natural killer-dendritic cell cross-talk in cancer immunotherapy. Expert Opin Biol Ther 2006; 5:1303-15. [PMID: 16197336 DOI: 10.1517/14712598.5.10.1303] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cells and dendritic cells (DCs), two important components of the immune system, can exchange bidirectional activating signals in a positive feedback. Myeloid DCs, the cell type specialised in the presentation of antigen and initiation of antigen-specific immune responses, have recently been documented to be involved in supporting innate immunity, promoting the production of cytokines and cytotoxicity of NK cells, and enhancing their tumouricidal activity. Natural interferon-producing cells/plasmacytoid DCs (IPCs/PDCs) play an additional role in NK cell activation. Reciprocally, NK cells, traditionally considered to be major innate effector cells, have also recently been shown to play immunoregulatory 'helper' functions, being able to activate DCs and to enhance their ability to produce pro-inflammatory cytokines, and to stimulate T helper (Th) 1 and cytotoxic T lymphocyte (CTL) responses of tumour-specific CD4+ and CD8+ T cells. Activated NK cells induce the maturation of myeloid DCs into stable type-1 polarised DCs (DC1), characterised by up to a 100-fold enhanced ability to produce IL-12p70 in response to subsequent interaction with Th cells. In addition, the ability of NK cells to kill tumour cells may facilitate the generation of tumour-related antigenic material, further accelerating the induction of tumour-specific immunity. DC1, induced by NK cells or by NK cell-related soluble factors, are stable, resistant to tumour-related suppressive factors, and demonstrate a strongly enhanced ability to induce Th1 and CTL responses in human in vitro and mouse in vivo models. Compared with the standard mature DCs that are used in clinical trials at present, human NK cell-induced DC1s act as superior inducers of anticancer CTL responses during in vitro sensitisation. This provides a strong rationale for the combined use of NK cells and DCs in the immunotherapy of patients with cancer and patients with chronic infections that are resistant to standard forms of treatment. Stage I/II clinical trials that are being implemented at present should allow evaluation of the immunological and clinical efficacy of combined NK-DC therapy of melanoma and other cancers.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Aguilar P, Mathieu CP, Clerc G, Ethevenot G, Fajraoui M, Mattei S, Faure GC, Bene MC. Modulation of natural killer (NK) receptors on NK (CD3-/CD56+), T (CD3+/CD56-) and NKT-like (CD3+/CD56+) cells after heart transplantation. J Heart Lung Transplant 2006; 25:200-5. [PMID: 16446221 DOI: 10.1016/j.healun.2005.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Revised: 07/26/2005] [Accepted: 08/07/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND After undergoing heart transplantation and the subsequent compulsive immunosuppressive treatments, patients are at risk of rejection episodes, infectious complications or cancer development. Thus, it is probable that the various subsets of peripheral cytotoxic lymphocytes are modulated in such patients. This area of study can now be investigated by examining the numerous recently described natural killer (NK)-cell-related surface receptors. METHODS A prospective cohort of 60 heart transplant recipients and 60 controls was studied. The partitioning of lymphocyte subsets, especially NK (CD3-/CD56+), T (CD3+/CD56-) and NKT-like (CD3+/CD56+) cells, was compared in both groups using multi-parametric flow cytometry. Moreover, expression of a series of seven NK-related receptors was compared on the three subsets defined by CD56 expression. RESULTS A significant increase in NK-cell levels was observed in transplanted patients, as compared with controls, whereas T and NKT-like cells were in similar proportions in both groups. Two NK-related receptors showed significantly different levels of expression in heart transplant recipients: the cytotoxic effector, CD244, which was in a significantly increased proportion on T and NKT-like cells; and the activating receptor, CD161, which was expressed significantly less on NK and NKT-like cells, but more on T cells. CONCLUSIONS These findings indicate that cytotoxic NK-related cells, increased in proportion, also display increased levels of activity-associated markers in heart transplant recipients. Viral infection or the immunosuppressive regimen could be responsible for the modulation of regulatory receptors on NK and NKT-like cells in heart transplant recipients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/analysis
- Antigens, Surface/analysis
- CD3 Complex/analysis
- CD56 Antigen/analysis
- Case-Control Studies
- Female
- Graft Rejection/immunology
- Graft Rejection/pathology
- Heart Transplantation/immunology
- Heart Transplantation/pathology
- Heart Transplantation/physiology
- Humans
- Immunosuppressive Agents/pharmacology
- Immunosuppressive Agents/therapeutic use
- Infections/immunology
- Infections/pathology
- Killer Cells, Natural/chemistry
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Lectins, C-Type/analysis
- Lymphocyte Subsets/chemistry
- Lymphocyte Subsets/drug effects
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/pathology
- Male
- Middle Aged
- NK Cell Lectin-Like Receptor Subfamily B
- Prospective Studies
- Receptors, Immunologic/analysis
- Receptors, Immunologic/drug effects
- Receptors, Immunologic/physiology
- Receptors, Natural Killer Cell
- Signaling Lymphocytic Activation Molecule Family
- T-Lymphocytes/chemistry
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Patricia Aguilar
- Immunology Laboratory, Faculty of Medicine and CHU, BP184, Nancy, France.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Bottley G, Cook GP, Meade JL, Holt JR, Hoeben RC, Blair GE. Differential expression of LFA-3, Fas and MHC Class I on Ad5- and Ad12-transformed human cells and their susceptibility to lymphokine-activated killer (LAK) cells. Virology 2005; 338:297-308. [PMID: 15963548 DOI: 10.1016/j.virol.2005.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Revised: 02/25/2005] [Accepted: 05/12/2005] [Indexed: 11/19/2022]
Abstract
Adenovirus (Ad) E1A is a potent oncogene and has been shown to deregulate the expression of a large number of cellular genes leading to cellular transformation. Here we have analysed the expression of several immunomodulatory molecules on the surface of a set of human cell lines transformed with either Ad12 or Ad5. Human cells transformed with Ad12 demonstrated reduced expression of cell surface LFA-3, Fas and MHC class I when compared to Ad5-transformed cells. Furthermore, Ad12-transformed human cell lines demonstrated greater susceptibility to lysis by lymphokine-activated killer (LAK) cells, compared to Ad5-transformed human cell lines. In contrast, previous studies with rodent cells showed that both Ad5- and Ad12-transformed rat cells were susceptible to LAK cells. Thus, transformation of human cells with Ad5 or Ad12 results in differences in the expression of immunomodulatory molecules on the cell surface and differential recognition of these virus-transformed cells by immune effector cells.
Collapse
Affiliation(s)
- Graham Bottley
- Molecular Cell Biology Research Group and School of Biochemistry and Microbiology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | |
Collapse
|
14
|
Nelson GW, Martin MP, Gladman D, Wade J, Trowsdale J, Carrington M. Cutting edge: heterozygote advantage in autoimmune disease: hierarchy of protection/susceptibility conferred by HLA and killer Ig-like receptor combinations in psoriatic arthritis. THE JOURNAL OF IMMUNOLOGY 2004; 173:4273-6. [PMID: 15383555 DOI: 10.4049/jimmunol.173.7.4273] [Citation(s) in RCA: 204] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Functionally relevant combinations of HLA and killer Ig-like receptor (KIR) genotypes influence resistance to several diseases in humans. Analysis of genetic data from such studies is challenging because it involves multiple linked and unlinked loci that exert their influence in an epistatic manner. We previously reported that subjects with certain activating receptors were susceptible to developing psoriatic arthritis (PsA), an effect that was strongest when HLA ligands for corresponding homologous inhibitory receptors were missing. In this study, we present a novel model in which susceptibility to PsA is determined by the overall balance of activating and inhibitory composite KIR-HLA genotypes. This model fits our knowledge of clonal NK cell expression of KIR and regulation of NK cell activity better than does the previous model, as reflected in a robust trend for increasing susceptibility to PsA with more activating genotypes. These data emphasize the remarkable influence of KIR/HLA combinations on this disease.
Collapse
Affiliation(s)
- George W Nelson
- Basic Research Program, Science Applications International Corporation-Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | | | | | |
Collapse
|
15
|
Young RM, Zheng X, Holowka D, Baird B. Reconstitution of regulated phosphorylation of FcepsilonRI by a lipid raft-excluded protein-tyrosine phosphatase. J Biol Chem 2004; 280:1230-5. [PMID: 15537644 DOI: 10.1074/jbc.m408339200] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To examine the exquisite regulation of IgE-FcepsilonRI tyrosine phosphorylation by Lyn kinase that is stimulated by antigen-mediated cross-linking, we utilized co-expression of FcepsilonRI and Lyn in Chinese hamster ovary cells, which results in high basal levels of Lyn kinase activity and spontaneous phosphorylation of FcepsilonRI. We found that co-expression of a lipid raft-excluded transmembrane tyrosine phosphatase, PTPalpha, suppresses Lyn kinase activity and markedly reduces the level of spontaneous phosphorylation of FcepsilonRI, while facilitating its antigen-stimulated phosphorylation. Other tyrosine phosphatases, including SHP-1, CD45, and a lipid raft-preferring chimeric version of PTPalpha fail to reconstitute antigen-dependent FcepsilonRI phosphorylation. We concluded that both substrate specificity and submembrane location are critical to phosphatase-mediated regulation of Lyn kinase activity that supports activation of FcepsilonRI.
Collapse
Affiliation(s)
- Ryan M Young
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | | | | | | |
Collapse
|
16
|
Hultström AL, Bratt G, Cosma A, Erfle V, Wahren B, Carbone E. Autologous cytotoxicity of natural killer cells derived from HIV-infected patients. Immunol Lett 2004; 91:155-8. [PMID: 15019284 DOI: 10.1016/j.imlet.2003.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2003] [Accepted: 11/24/2003] [Indexed: 11/21/2022]
Abstract
NK cells recognize target cells with reduced expression of MHC class I molecules. Human immunodeficiency virus (HIV) infection decreases MHC class I on the cell membrane. The aim of this study was to directly evaluate the role and conditions of NK cell effects in HIV seropositive patients ex vivo. Autologous HIV-infected CD4+ T cells were exposed to NK cells recognition. We discovered an increased lysis of the target cells after infection with human immunodeficiency virus-1 (HIV-1). The expression of the HIV-1 nef gene or the combined expression of nef and tat confers NK susceptibility to autologous CD4+ targets. Downregulation of MHC class I but not HLA-C or CD4 correlated with increased recognition by NK cells. The specific recognition is correlated with downregulation of MHC class I molecules on the infected target cells.
Collapse
Affiliation(s)
- Anna Linda Hultström
- Department of Virology, Swedish Institute for Infectious Disease Control, Karolinska Institute, Solna
| | | | | | | | | | | |
Collapse
|
17
|
Dornand J, Lafont V, Oliaro J, Terraza A, Castaneda-Roldan E, Liautard JP. Impairment of intramacrophagic Brucella suis multiplication by human natural killer cells through a contact-dependent mechanism. Infect Immun 2004; 72:2303-11. [PMID: 15039355 PMCID: PMC375199 DOI: 10.1128/iai.72.4.2303-2311.2004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Brucella spp. are facultative intracellular bacteria that can establish themselves and cause chronic disease in humans and animals. NK cells play a key role in host defense. They are implicated in an early immune response to a variety of pathogens. However, it was shown that they do not control Brucella infection in mice. On the other hand, NK cell activity is impaired in patients with acute brucellosis, and recently it was demonstrated that human NK cells mediate the killing of intramacrophagic Mycobacterium tuberculosis in in vitro infection. Therefore, we have analyzed the behavior of Brucella suis infecting isolated human macrophages in the presence of syngeneic NK cells. We show that (i) NK cells impair the intramacrophagic development of B. suis, a phenomenon enhanced by NK cell activators, such as interleukin-2; (ii) NK cells cultured in the presence of infected macrophages are highly activated and secrete gamma interferon and tumor necrosis factor alpha; (iii) impairment of bacterial multiplication inside infected cells is marginally associated with the cytokines produced during the early phase of macrophage-NK cell cocultures; (iv) direct cell-to-cell contact is required for NK cells to mediate the inhibition of B. suis development; and (v) inhibition of B. suis development results from an induction of NK cell cytotoxicity against infected macrophages. Altogether, these findings show that NK cells could participate early in controlling the intramacrophagic development of B. suis in humans. It seems thus reasonable to hypothesize a role for NK cells in the control of human brucellosis. However, by impairing the activity of these cells in the acute phase of the illness, the pathogen should avoid this control.
Collapse
Affiliation(s)
- Jacques Dornand
- INSERM U431, Université de Montpellier 2, 34095 Montpellier, France.
| | | | | | | | | | | |
Collapse
|
18
|
Prud'homme GJ. Altering immune tolerance therapeutically: the power of negative thinking. J Leukoc Biol 2003; 75:586-99. [PMID: 14657212 DOI: 10.1189/jlb.0803394] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The etiology of most human autoimmune diseases remains largely unknown. However, investigators have identified several negative regulatory mechanisms acting at the level of innate and/or adaptive immunity. Mutations resulting in a deficiency of some key regulatory molecules are associated with systemic or organ-specific inflammatory disorders, which often have a prominent autoimmune component. Genetic studies have implicated the negative regulator cytotoxic T-lymphocyte antigen 4 (CTLA-4) and other regulatory molecules in human autoimmune diseases. In addition to CTLA-4, key inhibitory molecules include programmed death 1 and B and T lymphocyte attenuator. Transforming growth factor beta1 and interleukin-10 also play major anti-inflammatory and regulatory roles. Tumor cells and infectious agents use negative regulatory pathways to escape immunity. The therapeutic blockage of negative signaling (particularly of CTLA-4) increases immunity against tumor antigens but also induces or aggravates autoimmune diseases. It appears that under normal conditions, the immune system is under strong "negative influences" that prevent autoimmunity and that release of this suppression results in disease. Regulation involves communication between the immune system and nonlymphoid tissues, and the latter can deliver inhibitory or stimulatory signals. Recent studies reveal that the generation of negative signals by selective engagement of inhibitory molecules is feasible and is likely to be of therapeutic benefit in autoimmune diseases and allograft rejection.
Collapse
Affiliation(s)
- Gérald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, St Michael's Hospital, University of Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Achdout H, Arnon TI, Markel G, Gonen-Gross T, Katz G, Lieberman N, Gazit R, Joseph A, Kedar E, Mandelboim O. Enhanced recognition of human NK receptors after influenza virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:915-23. [PMID: 12847262 DOI: 10.4049/jimmunol.171.2.915] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The NK cell cytotoxic activity is regulated by both inhibitory and activating NK receptors. Thus, changes in the expression levels and in the affinity or avidity of those receptors will have a major effect on the killing of target cells. In this study, we demonstrate that the binding of NK-inhibitory receptors is enhanced after influenza virus infection. Surprisingly, however, no change in the level of class I MHC protein expression was observed on the surface of the infected cells. The increased binding was general, because it was observed in both the killer cell Ig-like receptor 2 domain long tail 1 and leukocyte Ig-like receptor-1. The increased binding was functional, was not dependent on the interaction with viral hemagglutinin-neuraminidase, was not dependent on the glycosylation site, and was not abolished after mutating the transmembrane or cytosolic portions of the class I MHC proteins. Confocal microscopy experiments showed increased binding of NK receptor-coated beads to infected cells expressing the appropriate class I MHC proteins. In addition, specific cell-free bead aggregates covered with class I MHC proteins were observed only in infected cells. We therefore suggest that the influenza virus use a novel mechanism for the inhibition of NK cell activity. This mechanism probably involves the generation of class I MHC complexes in infected cells that cause increased recognition of NK receptors.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- COS Cells
- Cell Line, Transformed
- Cells, Cultured
- Cytotoxicity, Immunologic/genetics
- HLA-C Antigens/genetics
- HLA-C Antigens/metabolism
- HLA-C Antigens/physiology
- Humans
- Influenza A virus/immunology
- Influenza A virus/metabolism
- Interleukin-2/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/virology
- Leukocyte Immunoglobulin-like Receptor B1
- Lymphocyte Activation/genetics
- Mice
- Microspheres
- Peptide Fragments/genetics
- Peptide Fragments/metabolism
- Protein Binding/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, KIR2DL1
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/metabolism
- Sendai virus/immunology
- Species Specificity
- Transfection
- Tumor Cells, Cultured
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Hagit Achdout
- The Lautenberg Center for General and Tumor Immunology, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Herren B, Burrows PD. B cell-restricted human mb-1 gene: expression, function, and lineage infidelity. Immunol Res 2003; 26:35-43. [PMID: 12403343 DOI: 10.1385/ir:26:1-3:035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The antigen receptor on B cells (B cell receptor [BCR]) consists of two noncovalently associated modules. Immunoglobulin genes created somatically during B cell development encode the antigen-specific component of the receptor. The Igalpha/beta heterodimer, encoded by the mb-1 and B29 genes, is necessary to escort the receptor complex to the plasma membrane. Following antigen engagement of the BCR, Igalpha/beta nucleates signal transduction and promotes endocytosis of bound antigen for intracellular degradation and presentation to helper T-cells. In this review, we outline the discovery of the mb-1 gene; summarize results from other laboratories on the function of Igalpha/beta in B cells; and conclude with our recent studies, which indicate that mb-1 is not a B-lineage-restricted gene as originally proposed.
Collapse
Affiliation(s)
- Bettie Herren
- Department of Microbiology, University of Alabama at Birmingham, 35294, USA
| | | |
Collapse
|
21
|
Le Bouteiller P, Barakonyi A, Giustiniani J, Lenfant F, Marie-Cardine A, Aguerre-Girr M, Rabot M, Hilgert I, Mami-Chouaib F, Tabiasco J, Boumsell L, Bensussan A. Engagement of CD160 receptor by HLA-C is a triggering mechanism used by circulating natural killer (NK) cells to mediate cytotoxicity. Proc Natl Acad Sci U S A 2002; 99:16963-8. [PMID: 12486241 PMCID: PMC139252 DOI: 10.1073/pnas.012681099] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Circulating human natural killer (NK) lymphocytes have been functionally defined by their ability to exert cytotoxic activity against MHC class I-negative target cell lines, including K562. Therefore, it was proposed that NK cells recognized the "missing self." We show here that the Ig-like CD160 receptor expressed by circulating CD56(dim+) NK cells or IL-2-deprived NK cell lines is mainly involved in their cytotoxic activity against K562 target cells. Further, we report that HLA-C molecules that are constitutively expressed by K562 trigger NK cell lysis through CD160 receptor engagement. In addition, we demonstrate, with recombinant soluble HLA-Cw3 and CD160 proteins, direct interaction of these molecules. We also find that CD158b inhibitory receptors partially interfere with CD160-mediated cytotoxicity, whereas CD94CD159a and CD85j have no effect on engagement with their respective ligands. Thus, CD160HLA-C interaction constitutes a unique pathway to trigger NK cell cytotoxic activity.
Collapse
Affiliation(s)
- Philippe Le Bouteiller
- Institut National de la Santé et de la Recherche Médicale U563, Hôpital Purpan, 31059 Toulouse Cedex 3, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Farrell HE, Davis-Poynter NJ, Andrews DM, Degli-Esposti MA. Function of CMV-encoded MHC class I homologues. Curr Top Microbiol Immunol 2002; 269:131-51. [PMID: 12224506 DOI: 10.1007/978-3-642-59421-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Homologues of MHC class I proteins have been identified in the genomes of human, murine and rat cytomegaloviruses (CMVs). Given the pivotal role of the MHC class I protein in cellular immunity, it has been postulated that the viral homologues subvert the normal antiviral immune response of the host, thus promoting virus replication and dissemination in an otherwise hostile environment. This review focuses on recent studies of the CMV MHC class I homologues at the molecular, cellular and whole animal level and presents current hypotheses for their roles in the CMV life cycle.
Collapse
Affiliation(s)
- H E Farrell
- Animal Health Trust, Virology Section, Kentford, Newmarket, Suffolk CB8 7UU, UK
| | | | | | | |
Collapse
|
23
|
Burgert HG, Ruzsics Z, Obermeier S, Hilgendorf A, Windheim M, Elsing A. Subversion of host defense mechanisms by adenoviruses. Curr Top Microbiol Immunol 2002; 269:273-318. [PMID: 12224514 DOI: 10.1007/978-3-642-59421-2_16] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adenoviruses (Ads) cause acute and persistent infections. Alike the much more complex herpesviruses, Ads encode numerous immunomodulatory functions. About a third of the viral genome is devoted to counteract both the innate and the adaptive antiviral immune response. Immediately upon infection, E1A blocks interferon-induced gene expression and the VA-RNA inhibits interferon-induced PKR activity. At the same time, E1A reprograms the cell for DNA synthesis and induces the intrinsic cellular apoptosis program that is interrupted by E1B/19K and E1B/55K proteins, the latter inhibits p53-mediated apoptosis. Most other viral stealth functions are encoded by a separate transcription units, E3. Several E3 products prevent death receptor-mediated apoptosis. E3/14.7K seems to interfere with the cytolytic and pro-inflammatory activities of TNF while E3/10.4K and 14.5K proteins remove Fas and TRAIL receptors from the cell surface by inducing their degradation in lysosomes. These and other functions that may afect granule-mediated cell death might drastically limit lysis by NK cells and cytotoxic T cells (CTL). Moreover, Ads interfere with recognition of infected cell by CTL. The paradigmatic E3/19K protein subverts antigen presentation by MHC class I molecules by inhibiting their transport to the cell surface. In concert, these viral countermeasures ensure prolonged survival in the infected host and, as a consequence, facilitate transmission. Elucidating the molecular mechanisms of Ad-mediated immune evasion has stimulated corresponding research on other viruses. This knowledge will also be instrumental for designing better vectors for gene therapy and vaccination, and may lead to a more rational treatment of life-threatening Ad infections, e.g. in transplantation patients.
Collapse
Affiliation(s)
- H G Burgert
- Max von Pettenkofer-Institut, Lehrstuhl Virologie, Genzentrum der Ludwig-Maximilians-Universität, Feodor-Lynen-Str. 25, 81377 München, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Farag SS, Fehniger TA, Ruggeri L, Velardi A, Caligiuri MA. Natural killer cell receptors: new biology and insights into the graft-versus-leukemia effect. Blood 2002; 100:1935-47. [PMID: 12200350 DOI: 10.1182/blood-2002-02-0350] [Citation(s) in RCA: 362] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells have held great promise for the immunotherapy of cancer for more than 3 decades. However, to date only modest clinical success has been achieved manipulating the NK cell compartment in patients with malignant disease. Progress in the field of NK cell receptors has revolutionized our concept of how NK cells selectively recognize and lyse tumor and virally infected cells while sparing normal cells. Major families of cell surface receptors that inhibit and activate NK cells to lyse target cells have been characterized, including killer cell immunoglobulinlike receptors (KIRs), C-type lectins, and natural cytotoxicity receptors (NCRs). Further, identification of NK receptor ligands and their expression on normal and transformed cells completes the information needed to begin development of rational clinical approaches to manipulating receptor/ligand interactions for clinical benefit. Indeed, clinical data suggest that mismatch of NK receptors and ligands during allogeneic bone marrow transplantation may be used to prevent leukemia relapse. Here, we review how NK cell receptors control natural cytotoxicity and novel approaches to manipulating NK receptor-ligand interactions for the potential benefit of patients with cancer.
Collapse
Affiliation(s)
- Sherif S Farag
- Department of Internal Medicine, Division of Hematology/Oncology, The Ohio State University, A433A Starling Loving Hall, 320 W Tenth Avenue, Columbus, OH 43210, USA.
| | | | | | | | | |
Collapse
|
25
|
Briard D, Brouty-Boyé D, Azzarone B, Jasmin C. Fibroblasts from human spleen regulate NK cell differentiation from blood CD34(+) progenitors via cell surface IL-15. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4326-32. [PMID: 11970974 DOI: 10.4049/jimmunol.168.9.4326] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Besides a structural role in tissue architecture, fibroblasts have been shown to regulate the proliferation and differentiation of other neighboring specialized cell types, but differently according to the anatomic site and pathologic status of their tissue of origin. In this study we report a novel regulatory function of human spleen-derived fibroblasts in the development of NK cells from adult resting blood progenitors. When CD34(+) cells were cocultured with spleen-derived fibroblasts in monolayers, nonadherent CD56(+)CD3(-) NK cells were predominantly produced after 2-3 wk of culture in the absence of exogenous cytokines. Most NK cells expressed class I-recognizing CD94 and NK p46, p44, and p30 receptors as well as perforin and granzyme lytic granules. Moreover, these cells demonstrated spontaneous killing activity. Cell surface immunophenotyping of spleen-derived fibroblasts revealed a low and consistent expression of IL-15, Flt3 ligand, and c-kit ligand. Additionally, low picogram amounts of the three cytokines were produced extracellularly. Neutralizing Abs to IL-15, but not the other two ligands, blocked NK cell development. Additionally, suppressing direct contacts of CD34(+) progenitors and fibroblasts by microporous membrane abrogated NK cell production. We conclude that stromal fibroblasts within the human spleen are involved via constitutive cell surface expression of bioactive IL-15 in the development of functional activated NK cells under physiologic conditions.
Collapse
Affiliation(s)
- Diane Briard
- Institut National de la Santé et de la Recherche Médical, Unité 268, Institut André Lwoff, Hôpital Paul-Brousse, Villejuif, France
| | | | | | | |
Collapse
|
26
|
Vankayalapati R, Wizel B, Weis SE, Safi H, Lakey DL, Mandelboim O, Samten B, Porgador A, Barnes PF. The NKp46 receptor contributes to NK cell lysis of mononuclear phagocytes infected with an intracellular bacterium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3451-7. [PMID: 11907104 DOI: 10.4049/jimmunol.168.7.3451] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We used human tuberculosis as a model to investigate the role of NK cytotoxic mechanisms in the immune response to intracellular infection. Freshly isolated NK cells and NK cell lines from healthy donors lysed Mycobacterium tuberculosis-infected monocytes to a greater extent than uninfected monocytes. Lysis of infected monocytes was associated with increased expression of mRNA for the NKp46 receptor, but not the NKp44 receptor. Antisera to NKp46 markedly inhibited lysis of infected monocytes. NK cell-mediated lysis was not due to reduced expression of MHC class I molecules on the surface of infected monocytes or to enhanced production of IL-18 or IFN-gamma. NK cell lytic activity against M. tuberculosis-infected monocytes and NKp46 mRNA expression were reduced in tuberculosis patients with ineffective immunity to M. tuberculosis compared with findings in healthy donors. These observations suggest that 1) the NKp46 receptor participates in NK cell-mediated lysis of cells infected with an intracellular pathogen, and 2) the reduced functional capacity of NK cells is associated with severe manifestations of infectious disease.
Collapse
MESH Headings
- Cell Line
- Cell Survival/immunology
- Cells, Cultured
- Coculture Techniques
- Cytokines/biosynthesis
- Cytokines/immunology
- Cytotoxicity, Immunologic/immunology
- Histocompatibility Antigens Class I/biosynthesis
- Humans
- Immune Sera/pharmacology
- Intracellular Fluid/immunology
- Intracellular Fluid/microbiology
- K562 Cells
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/microbiology
- Monocytes/immunology
- Monocytes/metabolism
- Monocytes/microbiology
- Mycobacterium tuberculosis/immunology
- Natural Cytotoxicity Triggering Receptor 1
- Natural Cytotoxicity Triggering Receptor 2
- Phagocytes/immunology
- Phagocytes/microbiology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/biosynthesis
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/physiology
- Tuberculosis/immunology
Collapse
Affiliation(s)
- Ramakrishna Vankayalapati
- Center for Pulmonary and Infectious Disease Control, University of Texas Health Center, Tyler, TX 75708, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Verastegui E, Morales R, Barrera JL, Müeller A, Guzman B, Meneses A, Alfaro G. Immunological approach in the evaluation of regional lymph nodes of patients with squamous cell carcinoma of the head and neck. Clin Immunol 2002; 102:37-47. [PMID: 11781066 DOI: 10.1006/clim.2001.5130] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In cancer, regional lymph node (LN) cells are one of the first components of the immune system to have contact with tumor cells or their products. Therefore, the phenotype and functional properties of hematopoietic cells present within the tumor-draining LN are important to understanding their role in the control of malignant cells. Based on the locoregional metastatic behavior of squamous cell carcinoma of the head and neck (SCCH&N) region, we analyzed tumor-draining lymph nodes from SCCH&N patients to obtain insights into regional tumor immunity. Using a three-color fluorescent labeling technique, surface antigen expression was visualized in mononuclear cells of lymph nodes that were obtained from head and neck cancer patients and compared to mononuclear cells of normal lymph nodes. Cell cycle analyses were performed using propidium iodide. Proliferation after phytohemagglutinin stimulation was measured by a sodium tetrazolium-based assay. LN histology was correlated with flow cytometric findings. Regional lymph nodes of head and neck cancer patients undergo morphologic and functional changes. Flow cytometry revealed a decrease in CD8(+) T cells and in some lymph nodes the presence of second or third populations of larger cells with distinct size and granularity that expressed both T (gammadelta/alphabeta) and different natural killer cell markers. Moreover, cell cycle analyses and proliferation assays showed a diminished response to mitogenic stimuli. These changes were found in both metastatic and hyperplastic lymph nodes from head and neck cancer patients; however, no alterations were found in control lymph nodes or peripheral blood mononuclear cells from noncancer patients. The immune alterations detected in lymphocytes present within the draining lymph nodes of head and neck cancer patients may improve our understanding of how tumor cells escape host immunosurveillance. However, this dysfunction in local draining lymph nodes may not be detected systemically.
Collapse
Affiliation(s)
- Emma Verastegui
- Department of Immunology, Division of Surgery, Instituto Nacional de Cancerologia, Avenida San Fernando 22, Tlalpan, 14000 México City, D.F, México.
| | | | | | | | | | | | | |
Collapse
|
28
|
Fassett MS, Davis DM, Valter MM, Cohen GB, Strominger JL. Signaling at the inhibitory natural killer cell immune synapse regulates lipid raft polarization but not class I MHC clustering. Proc Natl Acad Sci U S A 2001; 98:14547-52. [PMID: 11724921 PMCID: PMC64719 DOI: 10.1073/pnas.211563598] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2001] [Indexed: 12/23/2022] Open
Abstract
Natural killer (NK) cell cytotoxicity is determined by a balance of positive and negative signals. Negative signals are transmitted by NK inhibitory receptors (killer immunoglobulin-like receptors, KIR) at the site of membrane apposition between an NK cell and a target cell, where inhibitory receptors become clustered with class I MHC ligands in an organized structure known as an inhibitory NK immune synapse. Immune synapse formation in NK cells is poorly understood. Because signaling by NK inhibitory receptors could be involved in this process, the human NK tumor line YTS was transfected with signal-competent and signal-incompetent KIR2DL1. The latter were generated by truncating the KIR2DL1 cytoplasmic tail or by introducing mutations in the immunoreceptor tyrosine-based inhibition motifs. The KIR2DL1 mutants retained their ability to cluster class I MHC ligands on NK cell interaction with appropriate target cells. Therefore, receptor-ligand clustering at the inhibitory NK immune synapse occurs independently of KIR2DL1 signal transduction. However, parallel examination of NK cell membrane lipid rafts revealed that KIR2DL1 signaling is critical for blocking lipid raft polarization and NK cell cytotoxicity. Moreover, raft polarization was inhibited by reagents that disrupt microtubules and actin filaments, whereas synapse formation was not. Thus, NK lipid raft polarization and inhibitory NK immune synapse formation occur by fundamentally distinct mechanisms.
Collapse
Affiliation(s)
- M S Fassett
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | |
Collapse
|
29
|
Chuang SS, Kumaresan PR, Mathew PA. 2B4 (CD244)-mediated activation of cytotoxicity and IFN-gamma release in human NK cells involves distinct pathways. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6210-6. [PMID: 11714782 DOI: 10.4049/jimmunol.167.11.6210] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
2B4 (CD244), a member of the CD2 subset of the Ig superfamily receptors, is expressed on all human NK cells, a subpopulation of T cells, basophils and monocytes. 2B4 activates NK cell mediated cytotoxicity, induces secretion of IFN-gamma and matrix metalloproteinases, and NK cell invasiveness. Although there have been several molecules shown to interact with 2B4, the signaling mechanism of 2B4-mediated activation of NK cells is still unknown. In this study, we found cross-linking of 2B4 on YT cells, a human NK cell line, results in the increased DNA binding activity of activator protein-1 (AP-1), an important regulator of nuclear gene expression in leukocytes. We investigated the possible role of various signaling molecules that may be involved in the activation of lytic function of YT cells via 2B4. Treatment of YT cells with various specific inhibitors indicate that 2B4-stimulation of YT cells in spontaneous and Ab-dependent cytotoxicity is Ras/Raf dependent and involves multiple MAPK signaling pathways (ERK1/2 and p38). However, only inhibitors of transcription and p38 inhibited 2B4-mediated IFN-gamma release indicating distinct pathways are involved in cytotoxicity and cytokine release. In this study we also show that 2B4 constitutively associates with the linker for activation of T cells (LAT) and that 2B4 may mediate NK cell activation via a LAT-dependent signaling pathway. These results indicate that 2B4-mediated activation of NK cells involves complex interactions involving LAT, Ras, Raf, ERK and p38 and that cytolytic function and cytokine production may be regulated by distinct pathways.
Collapse
Affiliation(s)
- S S Chuang
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | | | | |
Collapse
|
30
|
Warren HS, Campbell AJ, Waldron JC, Lanier LL. Biphasic response of NK cells expressing both activating and inhibitory killer Ig-like receptors. Int Immunol 2001; 13:1043-52. [PMID: 11470774 DOI: 10.1093/intimm/13.8.1043] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
NK cells can co-express inhibitory and activating killer Ig-like receptors (KIR) recognizing the same HLA class I ligand. We present evidence from experiments with NK cells expressing both activating (KIR2DS2) and inhibitory (KIR2DL2 and KIR2DL3) receptors that the activating KIR can function without apparent interference from the inhibitory KIR. These studies used CD158b mAb that is equally reactive with KIR2DS2, KIR2DL2 and KIR2DL3. First, we show using plastic-immobilized CD158b mAb that the activating KIR2DS2 is stimulated, resulting in NK cell division and degranulation. Second, we show using soluble CD158b mAb and FcRII (+) P815 cells that high concentrations of CD158b mAb trigger the inhibitory KIR, whereas low concentrations stimulate the activating KIR2DS2 resulting in NK cell division and cytolysis. These results demonstrate that the activating KIR2DS2 can function on cells co-expressing the inhibitory KIR2DL2 and/or KIR2DL3, indicating the potential for independent function of activating KIR with natural ligand.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, CD/immunology
- Binding Sites, Antibody
- Cell Degranulation/immunology
- Cell Division/immunology
- Cell Line
- Cytotoxicity Tests, Immunologic/methods
- Humans
- Immunoglobulin G/immunology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/immunology
- Lymphocyte Subsets/immunology
- Lymphocyte Subsets/metabolism
- Mice
- Plastics/metabolism
- Receptors, IgG/biosynthesis
- Receptors, IgG/immunology
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/immunology
- Receptors, Immunologic/physiology
- Receptors, KIR
- Receptors, KIR2DL2
- Receptors, KIR2DL3
- Sarcoma, Experimental
- Solubility
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- H S Warren
- Cancer Research Unit, The Canberra Hospital, Canberra, ACT 2606, Australia
| | | | | | | |
Collapse
|
31
|
Lima M, Teixeira MA, Queirós ML, Leite M, Santos AH, Justiça B, Orfão A. Immunophenotypic characterization of normal blood CD56+lo versus CD56+hi NK-cell subsets and its impact on the understanding of their tissue distribution and functional properties. Blood Cells Mol Dis 2001; 27:731-43. [PMID: 11778657 DOI: 10.1006/bcmd.2001.0443] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present study we have compared the immunophenotypic characteristics of the CD56+lo and CD56+hi NK-cell subsets in a group of normal healthy adults. Our results show that CD56+hi NK-cells display greater light-scatter properties than CD56+lo NK-cells at the same time they have higher levels of CD25 and CD122 IL-2 chains, together with a higher reactivity for HLA-DR and CD45RO and lower levels of CD45RA, supporting that, as opposed to the majority of the CD56+lo population, CD56+hi NK-cells might correspond to a subset of activated circulating NK-lymphocytes. Higher expression of the CD2 and CD7 costimulatory molecules found for the CD56+hi NK-cells would support their greater ability to respond to various stimuli. In addition, CD56+hi NK-cells expressed higher levels of several adhesion molecules such as CD2, CD11c, CD44, CD56, and CD62L compared to CD56+lo NK-cells, supporting a particular ability of these cells to migrate from blood to tissues and/or a potential advantage to form conjugates with target cells. Interestingly, CD56+lo and CD56+hi NK-cells showed a different pattern of expression of killer receptors that might determine different activation requirements for each of these NK-cell subsets. For instance, absence or low levels of CD16 expression might explain the lower antibody-dependent cytotoxicity activity of CD56+hi NK-cells. On the other hand, the virtual absence of expression of the CD158a and NKB1 immunoglobulin-like and the greater reactivity for the CD94 lectin-like killer receptors on CD56+hi in comparison to CD56+lo NK-cells might determine different MHC-class I specificities for both NK-cell subsets, a possibility that deserves further studies to be confirmed.
Collapse
MESH Headings
- Adult
- Antigens, CD/analysis
- Antigens, Differentiation/analysis
- Antigens, Differentiation, T-Lymphocyte/analysis
- CD56 Antigen/analysis
- Cell Adhesion Molecules/analysis
- Cell Differentiation
- Female
- Humans
- Immunophenotyping
- Killer Cells, Natural/chemistry
- Killer Cells, Natural/classification
- Killer Cells, Natural/immunology
- Leukocyte Common Antigens/analysis
- Lymphocyte Activation
- Male
- Receptors, Immunologic/analysis
- Receptors, KIR
- Receptors, KIR2DL1
- Receptors, KIR3DL1
- Receptors, Lymphocyte Homing/analysis
Collapse
Affiliation(s)
- M Lima
- Service of Clinical Hematology, Unit of Cytometry, Hospital Geral de Santo António, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
32
|
Kung SK, Su RC, Shannon J, Miller RG. Characterization of four new monoclonal antibodies that recognize mouse natural killer activation receptors. Hybridoma (Larchmt) 2001; 20:91-101. [PMID: 11394535 DOI: 10.1089/02724570152057580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
With the aim of identifying natural killer (NK) activation receptors, we immunized BALB/c mice with (BALB/cxB6)F1 NK LAK cells and made B-cell hybridomas. These were screened for monoclonal antibody (MAb) reacting with an NK activation receptor by using an antibody-induced redirected lysis (AIRL) assay against FcR-bearing P815 targets. Four hybridomas, clones 1C10, 1F10, 2D10 and 4G4, were selected for further characterization. Protein G-purified MAbs from these clones activated both resting and IL-2 activated B6 or F1 NK cells in the AIRL assay. 1F10 MAb, but not the other three MAbs, could compete for the binding of anti-NK1.1 (PK136) MAb to F1 NK cells. The four MAbs were screened for their ability to bind to or activate NK cells from the mouse strains SJL/J, DBA/2, 129/J, C3H/J, and BALB.K. None showed activity except IC10, which could bind to and activate SJL/J NK cells. When members of the NKR-P1 family from both B6 mice (A, B, and C genes expressed) and SJL mice (only A and B genes expressed) were expressed in Jurkat cells and tested for their antibody reactivity, PK136 MAb was found to recognize B6 NKR-P1C and SJL/J NKR-P1B; IC10 MAb was found to recognize NKR-P1-A, -B and -C from B6, but not NKR-P1A or -B from SJL/J; and 1F10 MAb was found to react only with B6 NKR-P1C.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/pharmacology
- Antibody Specificity
- Antigens, Ly
- Antigens, Surface/immunology
- Binding, Competitive
- Cross Reactions
- Flow Cytometry
- Hybridomas
- Killer Cells, Lymphokine-Activated/chemistry
- Killer Cells, Lymphokine-Activated/drug effects
- Killer Cells, Lymphokine-Activated/immunology
- Killer Cells, Natural/chemistry
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lectins, C-Type
- Lymphocyte Activation/drug effects
- Mice
- NK Cell Lectin-Like Receptor Subfamily B
- Receptors, Immunologic/immunology
Collapse
Affiliation(s)
- S K Kung
- Department of Medical Biophysics, Ontario Cancer Institute, University of Toronto, Canada
| | | | | | | |
Collapse
|
33
|
Beatty G, Paterson Y. IFN-gamma-dependent inhibition of tumor angiogenesis by tumor-infiltrating CD4+ T cells requires tumor responsiveness to IFN-gamma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:2276-82. [PMID: 11160282 DOI: 10.4049/jimmunol.166.4.2276] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The importance of CD4(+) T cells in the induction of an optimal antitumor immune response has largely been attributed to their ability to provide costimulatory signals for the priming of MHC class I-restricted CD8(+) CTL. However, many reports have demonstrated a requirement for CD4(+) T cells in the effector phase of tumor rejection indicating a greater responsibility for CD4(+) T cells in controlling tumor outgrowth. We demonstrate here a critical role for CD4(+) T cells in restraining initial tumor development through the inhibition of tumor angiogenesis. Using a tumor variant that is unresponsive to IFN-gamma, we show that tumor responsiveness to IFN-gamma is necessary for IFN-gamma-dependent inhibition of tumor angiogenesis by CD4(+) T cells. These studies reveal a pivotal role for CD4(+) T cells in controlling early tumor development through inhibition of tumor angiogenesis.
Collapse
MESH Headings
- Angiogenesis Inhibitors/immunology
- Angiogenesis Inhibitors/metabolism
- Angiogenesis Inhibitors/physiology
- Animals
- Antibodies, Monoclonal/administration & dosage
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Division/immunology
- Female
- Injections, Intraperitoneal
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Neoplasm Transplantation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- G Beatty
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
34
|
Frumento G, Ottonello L, Bertolotto M, Franchello S, Melioli G, Dallegri F. Spontaneous apoptosis in neutrophils is associated with downregulation of HLA Class I and is prevented by ligation of Class I. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Guido Frumento
- Immunogenetics Laboratory and Flow Cytometry Unit, National Institute for Cancer Research, Genoa, Italy; and
- Immunogenetics Laboratory and Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Luciano Ottonello
- Immunogenetics Laboratory and Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Maria Bertolotto
- Immunogenetics Laboratory and Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Silvia Franchello
- Immunogenetics Laboratory and Flow Cytometry Unit, National Institute for Cancer Research, Genoa, Italy; and
- Immunogenetics Laboratory and Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Giovanni Melioli
- Immunogenetics Laboratory and Flow Cytometry Unit, National Institute for Cancer Research, Genoa, Italy; and
| | - Franco Dallegri
- Immunogenetics Laboratory and Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|