The detection of ADAM8 protein on cells of the human immune system and the demonstration of its expression on peripheral blood B cells, dendritic cells and monocyte subsets

Immunobiology ◽  
2007 ◽  
Vol 212 (1) ◽  
pp. 29-38 ◽  
Author(s):  
J RICHENS ◽  
L FAIRCLOUGH ◽  
A GHAEMMAGHAMI ◽  
J MAHDAVI ◽  
F SHAKIB ◽  
...  
Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2781-2781 ◽  
Author(s):  
Fumihiko Ishikawa ◽  
Masaki Yasukawa ◽  
Bonnie Lyons ◽  
Shuro Yoshida ◽  
Leonard D. Shultz ◽  
...  

Abstract (Purpose) We aimed to develop a new model for studying the development of a human hematopoietic and immune systems in vivo. (Methods) In order to establish a new model of xenogeneic transplantation, we establish an immune-compromised strain, NOD.Cg-PrkdcscidIL2rgtmlWjl/Sz (NOD/SCID/IL2rg-null) mice by backcrossing a complete null mutation of the IL2 receptor common gamma chain (IL2rg) onto the NOD/SCID background. 1 x 105 human CB-derived lineage antigen negative (Lin−) CD34+ cells were intravenously transplanted into newborn NOD/SCID/IL2rg-null mice following 100cGy irradiation. At 3 months post-transplantation, the engraftment of human cells was evaluated by flow cytometric analysis, immunostaining, and functional assays for production of human immunoglobulin and T-cell cytotoxicity against allogeneic cells. (Results) NOD/SCID/IL2rg-null mice showed extremely low activity of NK cells along with the complete lack of mature B cells and T cells. During post-natal development of the NOD/SCID/IL2rg-null mice, a human hematopoietic system was developed following injection of human CB-derived Lin-CD34+ cells. In BM of the recipient mice, human glycophorin A+ erythroid cells were present at 9.5 +/− 6.2% (n=5), and human CD41+ megakaryocytes were present at 1.64 +/− 0.42% (n=5). Human CB-derived Lin−CD34+ cells generated multi-lineage leukocytes, CD33+ myeloid cells, CD19+ B cells, and CD3+ T cells. The engraftment level of human CD45+ cells in peripheral blood was significantly higher (68.9 +/− 11.6%, n=5) in NOD/SCID/IL2rg-null mice than that in NOD/SCID/b2mnull mice (12.4 +/− 5.9%, n=4). Mature erythrocytes and platelets were identified in peripheral blood. The xenogeneic environment supported the systemic development of a human immune system, containing each stage of B cells and T cells in primary and secondary lymphoid tissues. CD34+CD19+ pro-B cells, CD10+CD19+ B cells, and CD19+CD20hi mature B cells were identified in the BM and spleen. Immature CD4+CD8+ double positive T cells were the major cell populations in the thymus, while spleen contained abundant single positive T cells at 1.39 +/− 0.61 (n=5) CD4/CD8 ratio, suggesting that human CB stem/progenitor-derived T cells underwent the maturation and proliferation similarly as identified in human body. Transplanted human stem cells reconstituted mucosal immunity in intestinal tracts as evidenced by human IgA+ B cells and CD3+ T cells. Adaptive human immune system cooperatively functioned in xenogeneic environment to produce antigen-specific human IgM and IgG antibodies, when engrafted mice were immunized with ovalbumin. Furthermore, human CD4+ T cells as well as CD8+ T cells generated in the xenogeneic host exerted cytotoxicity against allogeneic target cells. (Conclusion) The neonatal NOD/SCID/IL2rg-null model will facilitate studying post-natal development of the human hematopoietic and immune systems and for studying of human immune surveillance in vivo against exogenous antigens.


2021 ◽  
Vol 118 (16) ◽  
pp. e2021570118
Author(s):  
Thiago Alves da Costa ◽  
Jacob N. Peterson ◽  
Julie Lang ◽  
Jeremy Shulman ◽  
Xiayuan Liang ◽  
...  

Central B cell tolerance, the process restricting the development of many newly generated autoreactive B cells, has been intensely investigated in mouse cells while studies in humans have been hampered by the inability to phenotypically distinguish autoreactive and nonautoreactive immature B cell clones and the difficulty in accessing fresh human bone marrow samples. Using a human immune system mouse model in which all human Igκ+ B cells undergo central tolerance, we discovered that human autoreactive immature B cells exhibit a distinctive phenotype that includes lower activation of ERK and differential expression of CD69, CD81, CXCR4, and other glycoproteins. Human B cells exhibiting these characteristics were observed in fresh human bone marrow tissue biopsy specimens, although differences in marker expression were smaller than in the humanized mouse model. Furthermore, the expression of these markers was slightly altered in autoreactive B cells of humanized mice engrafted with some human immune systems genetically predisposed to autoimmunity. Finally, by treating mice and human immune system mice with a pharmacologic antagonist, we show that signaling by CXCR4 is necessary to prevent both human and mouse autoreactive B cell clones from egressing the bone marrow, indicating that CXCR4 functionally contributes to central B cell tolerance.


2021 ◽  
Vol 12 ◽  
Author(s):  
Isabelle Serr ◽  
Maria Kral ◽  
Martin G. Scherm ◽  
Carolin Daniel

Immunodeficient mice engrafted with a functional human immune system [Human immune system (HIS) mice] have paved the way to major advances for personalized medicine and translation of immune-based therapies. One prerequisite for advancing personalized medicine is modeling the immune system of individuals or disease groups in a preclinical setting. HIS mice engrafted with peripheral blood mononuclear cells have provided fundamental insights in underlying mechanisms guiding immune activation vs. regulation in several diseases including cancer. However, the development of Graft-vs.-host disease restrains relevant long-term studies in HIS mice. Alternatively, engraftment with hematopoietic stem cells (HSCs) enables mimicking different disease stages, however, low frequencies of HSCs in peripheral blood of adults impede engraftment efficacy. One possibility to overcome those limitations is the use of patient-derived induced pluripotent stem cells (iPSCs) reprogrammed into HSCs, a challenging process which has recently seen major advances. Personalized HIS mice bridge research in mice and human diseases thereby facilitating the translation of immunomodulatory therapies. Regulatory T cells (Tregs) are important mediators of immune suppression and thereby contribute to tumor immune evasion, which has made them a central target for cancer immunotherapies. Importantly, studying Tregs in the human immune system in vivo in HIS mice will help to determine requirements for efficient Treg-targeting. In this review article, we discuss advances on personalized HIS models using reprogrammed iPSCs and review the use of HIS mice to study requirements for efficient targeting of human Tregs for personalized cancer immunotherapies.


2019 ◽  
Vol 2019 ◽  
pp. 1-16
Author(s):  
Liangliang Jiang ◽  
Tao Jiang ◽  
Jianhua Luo ◽  
Yanliang Kang ◽  
Yue Tong ◽  
...  

Single B cell antibody technology is a method for isolating antigen-specific B cells from human peripheral blood and obtaining antibody genes in developing antibody drugs. However, owing to immune tolerance to autoantigen, human autoantigen-specific B cells are difficult to acquire by conventional single B cell technology. In this study, we constructed a nitrated T-cell epitope named NitraTh by incorporating p-nitrophenylalanine into a universal T helper epitope. NitraTh had enhanced ability to activate CD4+ T cells and can be recognized by CD4+ T cells with different HLA class II haplotypes. This NitraTh can also break immune tolerance to autoantigens, such as human epidermal growth factor receptor 2 (HER2) and cannabinoid receptor 1, and induce strong specific IgM+ B cell responses in vitro. HER2-NitraTh vaccine can also stimulate the generation of HER2-specific IgG+ B cells in human immune system mice, which was established by cotransplanting lymphocytes and autologous dendritic cells in immunodeficient mice. We obtained 30 fully human IgG antibody genes by sorting single B cells from the human immune system mice immunized with HER2-NitraTh vaccine. The analysis of antibody genes showed that sorted B cells underwent the extensive somatic mutation of the antibody genes. We randomly selected eight genes for cloning, six of which expressed antibodies that can bind to HER2. Hence, we provided a convenient and effective method in acquiring fully human antibody genes against self-proteins, which can be used in developing therapeutic antibody drugs.


2015 ◽  
Vol 37 (4) ◽  
pp. 1355-1368 ◽  
Author(s):  
Zhen Gong ◽  
Hanzi Xu ◽  
Yiping Su ◽  
Wangfei Wu ◽  
Lin Hao ◽  
...  

Background/Aims: The aim of this study was to develop a novel model by transplanting human bladder cancer xenografts into humanized immunodeficient mice (SCID). Methods: The animals first underwent sublethal irradiation and then were subjected to simultaneous transplantation of human lymphocytes (5 × 107 cells/mouse i.p.) and human bladder cancer cells (3 × 106 cells/mouse s.c.). Results: The xenografts developed in all 12 mice that had received bladder cancer BIU-87 cells, and the tumor specimens were evaluated histologically. All 6 model mice expressed human CD3 mRNA and/or protein in the peripheral blood, spleens and xenografts. The mean proportion of human CD3+ cells was 19% with a level of human IgG 532.4µ/ml in the peripheral blood at Week 6 after transplant inoculation. The re-constructed human immune system in these mice was confirmed to be functional by individual in vitro testing of their proliferative, secretory and cytotoxic responses. Conclusion: The successful engraftment of the human bladder cancer xenografts and the establishment of the human immune system in our in vivo model described here may provide a useful tool for the development of novel therapeutic strategies targeting at bladder cancer.


2019 ◽  
Vol 221 (2) ◽  
pp. 201-213 ◽  
Author(s):  
Jordana G A Coelho-Dos-Reis ◽  
Ryota Funakoshi ◽  
Jing Huang ◽  
Felipe Valença Pereira ◽  
Sho Iketani ◽  
...  

Abstract Background For the purpose of studying functional human dendritic cells (DCs) in a humanized mouse model that mimics the human immune system (HIS), a model referred to as HIS mice was established. Methods Human immune system mice were made by engrafting NOD/SCID/IL2Rgammanull (NSG) mice with human hematopoietic stem cells (HSCs) following the transduction of genes encoding human cytokines and human leukocyte antigen (HLA)-A2.1 by adeno-associated virus serotype 9 (AAV9) vectors. Results Our results indicate that human DC subsets, such as CD141+CD11c+ and CD1c+CD11c+ myeloid DCs, distribute throughout several organs in HIS mice including blood, bone marrow, spleen, and draining lymph nodes. The CD141+CD11c+ and CD1c+CD11c+ human DCs isolated from HIS mice immunized with adenoviruses expressing malaria/human immunodeficiency virus (HIV) epitopes were able to induce the proliferation of malaria/HIV epitopes-specific human CD8+ T cells in vitro. Upregulation of CD1c was also observed in human CD141+ DCs 1 day after immunization with the adenovirus-based vaccines. Conclusions Establishment of such a humanized mouse model that mounts functional human DCs enables preclinical assessment of the immunogenicity of human vaccines in vivo.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A598-A598
Author(s):  
Dhwani Haria ◽  
Jayamary Divya Ravichandar ◽  
Jill Desnoyer ◽  
Sabina Lau ◽  
Jina Lee ◽  
...  

BackgroundDespite the unprecedented clinical success of immune checkpoint therapy (ICI) in many cancers there is still a large unmet need since many patients have an inadequate response. Fecal Matter Transplant from anti-PD-1 responding subjects into non-responders improves response in many subjects, providing a strong rationale that the gut microbiome has a critical role in modulating effects of ICI therapies. However, the causative mechanisms and host targets that mediate these beneficial therapeutic effects are still poorly understood. The objective of the present study was to demonstrate that peptides derived from the microbiome of ICI responders, have the potential to bring a similar therapeutic benefit to ICI non-responders via direct immunomodulatory effects through a host target.MethodsUsing Second Genome’s proprietary algorithms, we derived a microbial signature that was enriched in responding subjects across multiple cohorts. The peptides identified from these strains were screened in a classical drug discovery pipeline to discover peptides that modulate the human immune system by secreting proinflammatory cytokines and chemokines such as CXCL10 and TNF-α by human dendritic cells (DCs), and mediate anti-tumor immunity and improve response to anti-PD-1 therapy in mouse tumor models.ResultsWe found several peptides which bind primary human cells, exert immune function, and exert anti-tumor effects. Particularly, peptide SG-3-00802 showed improved anti-tumor responses in combination with anti-PD-1 in the anti-PD-1 insensitive RENCA model. Treatment with SG-3-00802, alone or in combination with anti-PD-1 significantly improved overall survival with many animals showing complete tumor regression. Surviving animals with fully regressed tumors rejected newly implanted tumors when rechallenged with RENCA cells, indicating that combination treatment of SG-3-00802 + anti-PD-1 generates long lasting anti-tumor memory responses.ConclusionsMicrobiome interacts with the human immune system to impact anti-tumor immunity. Microbiome-derived peptides identified by Second Genome’s discovery platform sg4sight modulate innate immune cells e.g. dendritic cells to promote anti-tumor immunity. SG-3-00802 peptide has the greatest potential to impact subjects who are potentially going to be non-responders to anti-PD-1 or have derived inadequate response to ICI therapies.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4899-4899
Author(s):  
Tomomi Nagashima ◽  
Akihiko Yokohama ◽  
Kie Nagai ◽  
Rumi Ino ◽  
Noriyuki Takahashi ◽  
...  

Abstract Introduction:Erythropoietin (EPO) is hematopoietic factors participating in red blood cell production by accelerating cell proliferation and inhibition of apoptosis of erythroblast. It is reported that EPO has pleiotropic effects including anti-apoptotic action for some cells, antioxidant action, vascularization action, and promoting cell division in addition to stimulation of erythropoiesis as well, whereas there are conflicting results of small cohorts as to its effect on blood immune cells. We analyzed peripheral white blood cell subsets in patients who received one bolus administration of recombinant human erythropoietin (rHuEPO) to examine the effect of EPO on human immune system. Materials and Methods:One hundred twenty eight autologous blood donors (male/female 64/64) in Gunma University Hospital were enrolled in this study after written informed consent. All the patients had no infections or inflammation. Fifty six patients were administered rHuEPO (Epoetin alpha or Epoetin beta, 24,000 IU) once after blood donation because of low hemoglobin concentration (EPO group) and 72 were not administered (non-EPO group). Peripheral blood samples were obtained at the time of the first phlebotomy and after 7 days from the same patient. We measured the number of WBC, lymphocytes, myeloid dendritic cells (mDC), plasmacytoid dendritic cells (pDC), CD4+ T cells, CD8+ T cells, Natural killer (NK) cells, B cells, monocytes, and neutrophils of peripheral blood before and after rHuEPO administration by flow cytometry. Also we compared EPO group with non-EPO group. Paired and unpaired Student's t-test were used to compare absolute counts and percentages of each cell, P values < 0.05 were considered significant. This study was approved by the research ethics committee of our hospital. Results:In EPO group, absolute number and percentage of lymphocyte in WBC decreased significantly after rHuEPO administration from 1754.9 ± 535.4/µl to 1627.2 ± 467.7/µl, (p<0.01) and from 30.9 ± 8.00% to 28.2 ± 6.77% (p<0.01), respectively. The numbers of whole WBC, mDC, pDC, monocyte and neutrophil did not show significant changes. In regards to lymphocyte subset, absolute number of CD8+ T cell significantly decreased from 358.9 ± 257.0/µl to 311.5 ± 210.9/µl (p=0.04) and that of NK cell from 290.6 ± 157.6/µl to 257.4 ± 141.8/µl (p<0.01), but their percentages did not change. B cells significantly decreased in absolute number from 258.3 ± 154.2 /µl to 221.6 ± 129.2 /µl (p<0.01) and also in percentages from 14.8 ± 6.92% to 13.6 ± 6.07% (p<0.01). Regarding B cell subsets, absolute number of Naïve B cells significantly decreased from 171.3 ± 93.5 /µl to 153.0 ± 84.2 /µl (p<0.01); moreover other B cell subsets, such as transitional B cells, memory B cells and marginal zone B cells, also showed a trend of decrease, so that the percentage of Naïve B cell did not change in total B cells. These suggested that whole B cell decreased, not a specific subset of B cell. In non-EPO group, there was no significant change in any type of immune cells. When compared the rate of change between before and after rHuEPO treatment, B cell number significant decreased in EPO group compared to non-EPO group (0.87 vs 0.98, p<0.01). Multiple regression analysis revealed rHuEPO administration significantly influenced B cell number (p=0.015); however age, sex, disease status and the type of rHuEPO preparations (epoetin alfa or beta) did not influence. Conclusion:These findings suggested that just one administration of rHuEPO influenced human immune system, especially via reduction of B cell in peripheral blood. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document