15 results on '"Nargess Hassanzadeh-Kiabi"'
Search Results
2. Blimp-1 Functions as a Molecular Switch to Prevent Inflammatory Activity in Foxp3+RORγt+ Regulatory T Cells
- Author
-
Chihiro Ogawa, Rashmi Bankoti, Truc Nguyen, Nargess Hassanzadeh-Kiabi, Samantha Nadeau, Rebecca A. Porritt, Michael Couse, Xuemo Fan, Deepti Dhall, Gerald Eberl, Caspar Ohnmacht, and Gislâine A. Martins
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Foxp3+ regulatory T cells (Treg) are essential modulators of immune responses, but the molecular mechanisms underlying their function are not fully understood. Here we show that the transcription factor Blimp-1 is a crucial regulator of the Foxp3+RORγt+ Treg subset. The intrinsic expression of Blimp-1 in these cells is required to prevent production of Th17-associated cytokines. Direct binding of Blimp-1 to the Il17 locus in Treg is associated with inhibitory histone modifications but unaltered binding of RORγt. In the absence of Blimp-1, the Il17 locus is activated, with increased occupancy of the co-activator p300 and abundant binding of the transcriptional regulator IRF4, which is required, along with RORγt, for IL-17 expression in the absence of Blimp-1. We also show that despite their sustained expression of Foxp3, Blimp-1−/− RORγt+IL-17-producing Treg lose suppressor function and can promote intestinal inflammation, indicating that repression of Th17-associated cytokines by Blimp-1 is a crucial requirement for RORγt+ Treg function. : Ogawa et al. demonstrate that the transcription factor Blimp-1 is required to prevent production of Th17-associated cytokines and inflammatory activity of microbiota-specific Foxp3+RORγt+ Treg. These findings uncover a critical role for Blimp-1 in Foxp3+ Treg function and shed light on the intricate mechanisms underlying Treg phenotypic stability. Keywords: Blimp-1, Prdm1, Foxp3, Treg, RORγt, IL-17, IRF4, transcription, Foxp3, intestinal
- Published
- 2018
- Full Text
- View/download PDF
3. Phagocytic Synapses
- Author
-
Helen S. Goodridge and Nargess Hassanzadeh-Kiabi
- Published
- 2023
- Full Text
- View/download PDF
4. 564 Potency-reduced and extended half-life IL12 heterodimeric Fc-fusions exhibit strong anti-tumor activity with potentially improved therapeutic index compared to native IL12 agents
- Author
-
Umesh Muchhal, Christine Bonzon, John R. Desjarlais, Ke Liu, Rajat Varma, Nicole Rodriguez, Connie Ardila, Matthew J. Bernett, Rumana Rashid, Seung Y. Chu, and Nargess Hassanzadeh-Kiabi
- Subjects
biology ,Chemistry ,Pharmacology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,In vitro ,Immune system ,MHC class I ,Interleukin 12 ,biology.protein ,Potency ,IL-2 receptor ,STAT4 ,CD8 - Abstract
Background Interleukin-12 (IL12) is a proinflammatory cytokine produced by activated antigen-presenting cells that induces differentiation of Th1 cells and increased proliferation and cytotoxicity of T and NK cells. Stimulation of these cells by IL12 leads to production of high levels of IFNγ. These immune-stimulating aspects of IL12 may help to establish an inflammatory tumor microenvironment critical for anti-tumor responses. Preclinical studies in mice revealed that native IL12 can dramatically shrink syngeneic tumors, however clinical studies in humans resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior pharmacokinetics, pharmacodynamics, and safety in non-human primates through reduction of receptor-mediated clearance. Applying similar principles to IL12, we created IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency to improve tolerability, slow receptor-mediated clearance, and extend half-life. Methods IL12 is a heterodimeric protein consisting of two subunits, so we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and the IL12p40 subunit to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity was assessed by engrafting MCF-7 cells into PBMC engrafted NSG MHC class I and II double-knockout mice and by measuring tumor volume, lymphocyte activation/proliferation, and IFNγ production over time. Results IL12-Fc were produced with good yield and purity. An IL12-Fc potency series was created, and variants had up to a 10,000-fold reduction in STAT4 signaling potency and IFNγ production in an MLR assay compared to native IL12-Fc. Anti-tumor activity in the huPBMC-MCF7 model was achieved with potency-reduced IL12-Fc as a single-agent and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells, as evidenced by upregulation of CD25. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum indicated up to 200-fold increases in IFNγ levels. Conclusions Combined, these data indicate that potency-reduced IL12-Fc retain strong anti-tumor activity, while potentially overcoming safety and tolerability issues related to small therapeutic index associated with recombinant native IL12 or IL12-Fc agents.
- Published
- 2020
- Full Text
- View/download PDF
5. 707 IL12 Fc-fusions engineered for reduced potency and extended half-life exhibit strong anti-tumor activity and improved therapeutic index compared to wild-type IL12 agents
- Author
-
Hanh Nho Nguyen, Umesh Muchhal, Katrina Bykova, Irene Leung, John R. Desjarlais, Rumana Rashid, Matthew J. Bernett, Ke Liu, Araz Eivazi, Christine Bonzon, Connie Ardila, Kendra N. Avery, Nicole Rodriguez, Norman J. Barlow, Nargess Hassanzadeh-Kiabi, Rajat Varma, and Michael Hackett
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,Chemistry ,Immunology ,Wild type ,Half-life ,Therapeutic index ,Oncology ,Interleukin 12 ,Molecular Medicine ,Immunology and Allergy ,Potency - Abstract
BackgroundInterleukin-12 (IL12) is a proinflammatory cytokine that induces differentiation of Th1 cells and increased cytotoxicity of T and NK cells. Stimulation by IL12 leads to production of IFNγ and an inflammatory tumor microenvironment critical for anti-tumor responses. Studies in mice revealed IL12 can dramatically shrink syngeneic tumors, however human clinical studies resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior therapeutic index (TI) in non-human primates (NHP) by reducing receptor-mediated clearance. Applying similar principles to IL12, we created IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency to improve TI.MethodsIL12 is a heterodimer of two subunits, so we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and IL12p40 to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity of human IL12-Fc was assessed in huPBMC-NSG-DKO and huCD34+ MCF7 xenograft models. Surrogate mouse potency-reduced IL12-Fc were evaluated in syngeneic tumor models. Tolerability and pharmacodynamic activity were assessed in NHP.ResultsAn IL12-Fc potency series was created, and variants had up to a 10,000-fold reduction in STAT4 signaling and IFNγ production in an MLR assay compared to wild-type IL12-Fc. Anti-tumor activity was achieved with potency-reduced IL12-Fc as single-agents and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum indicated up to 200-fold increases in IFNγ levels. Surrogate potency-reduced IL12-Fc had improved tolerability and greater selectivity of IFNγ production in tumors compared to spleen and less production of IL10 compared to wild-type IL12-Fc. In NHP, potency-reduced IL12-Fc had superior exposure with slower, more sustained accumulation of IFNγ and IP10, and a more gradual dose-dependent peak response, as well as more sustained margination of T and NK cells compared to wild-type IL12-Fc.ConclusionsPotency-reduced IL12-Fc retain strong anti-tumor activity, while potentially overcoming safety and tolerability issues related to narrow TI associated with wild-type IL12 or IL12-Fc agents.
- Published
- 2021
- Full Text
- View/download PDF
6. Abstract 1743: IL12 heterodimeric Fc-fusions engineered for reduced potency exhibit strong anti-tumor activity and improved therapeutic index compared to native IL12 agents
- Author
-
Rumana Rashid, Nargess Hassanzadeh-Kiabi, Irene Leung, Norm Barlow, Nicole Rodriguez, Araz Eivazi, Michael Hackett, Duc-Hanh T. Nguyen, Connie Ardila, Rajat Varma, Matthew J. Bernett, Seung Y. Chu, Christine Bonzon, John R. Desjarlais, Ke Liu, Umesh Muchhal, and Katrina Bykova
- Subjects
Antitumor activity ,Cancer Research ,Therapeutic index ,Oncology ,Chemistry ,Interleukin 12 ,Potency ,Pharmacology - Abstract
Interleukin-12 (IL12) is a proinflammatory cytokine produced by activated antigen-presenting cells that induces differentiation of Th1 cells and increased proliferation and cytotoxicity of T and NK cells. Stimulation of these cells by IL12 leads to production of high levels of IFNγ. These immune-stimulating aspects of IL12 may help to establish an inflammatory tumor microenvironment critical for anti-tumor responses. Preclinical studies in mice revealed that native IL12 can dramatically shrink syngeneic tumors, however clinical studies in humans resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior pharmacokinetics, pharmacodynamics, and safety in non-human primates through reduction of receptor-mediated clearance. Applying similar principles to IL12, we created IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency to improve tolerability, slow receptor-mediated clearance, and extend half-life. IL12 is a heterodimeric protein consisting of two subunits, so we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and the IL12p40 subunit to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity of human IL12-Fc were assessed in a human PBMC engrafted mouse MCF7 tumor model. Surrogate mouse IL12-Fc were evaluated in additional murine tumor models. Tolerability and pharmacodynamic activity were assessed in non-human primates. IL12-Fc were produced with good yield and purity. An IL12-Fc potency series was created, and variants had up to a 10,000-fold reduction in STAT4 signaling potency and IFNγ production in an MLR assay compared to native IL12-Fc. Anti-tumor activity was achieved with potency-reduced IL12-Fc as a single-agent and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum indicated up to 200-fold increases in IFNγ levels. Combined, these data indicate that potency-reduced IL12-Fc retain strong anti-tumor activity, while potentially overcoming safety and tolerability issues related to narrow therapeutic index associated with recombinant native IL12 or IL12-Fc agents. Citation Format: Matthew J. Bernett, Ke Liu, Christine Bonzon, Rumana Rashid, Nicole Rodriguez, Nargess Hassanzadeh-Kiabi, Connie Ardila, Katrina Bykova, Michael Hackett, Norm Barlow, Irene Leung, Duc-Hanh Nguyen, Araz Eivazi, Seung Y. Chu, Rajat Varma, Umesh S. Muchhal, John R. Desjarlais. IL12 heterodimeric Fc-fusions engineered for reduced potency exhibit strong anti-tumor activity and improved therapeutic index compared to native IL12 agents [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1743.
- Published
- 2021
- Full Text
- View/download PDF
7. Abstract 1831: Affinity tuned XmAb®2+1 GPC3 x CD3 bispecific antibodies demonstrate selective activity in liver cancer models
- Author
-
Umesh Muchhal, Seung Y. Chu, Juan Diaz, Rumana Rashid, John R. Desjarlais, Nargess Hassanzadeh-Kiabi, Katrina Bykova, Alex Nisthal, Kendra N. Avery, and Gregory L. Moore
- Subjects
Cancer Research ,Bispecific antibody ,Oncology ,biology ,Chemistry ,CD3 ,biology.protein ,Cancer research ,medicine ,Liver cancer ,medicine.disease - Abstract
Bispecific T cell engagers simultaneously bind CD3 on T cells and tumor-associated antigens to promote T cell-mediated killing of tumor cells. These agents provide synthetic immunity by expanding, activating, and redirecting T cells against a target of interest. While targeting lineage-restricted antigens such as CD19 or CD20 have found clinical success in hematopoietic cancers, targeting solid tumors requires high expressing tumor associated antigens (TAAs) with minimal normal tissue expression. Glypican 3 (GPC3), a lipid-anchored cell surface protein, is over-expressed in the majority of hepatocellular carcinoma (HCC) and a smaller subset of lung squamous cell carcinoma. It is considered an ideal target because its expression is not detected in adult tissues, and its function can promote tumor growth through the Wnt/beta-catenin pathway. Although GPC3 demonstrates good differential expression, bispecific T cell engagers are powerful immunomodulatory agents and careful tuning can improve the therapeutic window on all targets. Building upon the XmAb® heterodimeric Fc platform, we generated bispecific antibodies in an XmAb 2+1 Fab2-scFv-Fc format that are bivalent for GPC3 and monovalent for CD3. Reducing the affinity of GPC3 encouraged avid binding and strong killing activity only on high GPC3 expressing cell lines while minimizing reactivity on low expressing cell lines. We found modulating the CD3 affinity, either directly through mutation or indirectly by the molecular format, contributes to the selectivity of the bispecific antibodies. To ensure biologically valid antigen densities were being considered, IHC was conducted on paraffin embedded arrays of tumor tissue, normal tissue, and cell lines. Upon matching the staining intensity between the three sample types, we identified cell lines with corresponding GPC3 antigen density that could serve as proxies of tumor and normal tissue. In vitro T cell-dependent cellular cytotoxicity (TDCC) assays on the relevant cell lines confirmed selective potency on high versus low expressing cells. Citation Format: Alex Nisthal, Nargess Hassanzadeh-Kiabi, Kendra N. Avery, Rumana Rashid, Juan E. Diaz, Umesh S. Muchhal, Seung Y. Chu, Gregory L. Moore, Katrina Bykova, John R. Desjarlais. Affinity tuned XmAb®2+1 GPC3 x CD3 bispecific antibodies demonstrate selective activity in liver cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1831.
- Published
- 2021
- Full Text
- View/download PDF
8. Autocrine Type I IFN Signaling in Dendritic Cells Stimulated with Fungal β-Glucans or Lipopolysaccharide Promotes CD8 T Cell Activation
- Author
-
Helen S. Goodridge, Gislâine A. Martins, Ivy Dang, David M. Underhill, Alberto Yáñez, and Nargess Hassanzadeh-Kiabi
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Granzyme B production ,beta-Glucans ,Blotting, Western ,Immunology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,Fungal Proteins ,Mice ,03 medical and health sciences ,0302 clinical medicine ,MHC class I ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Autocrine signalling ,Mice, Knockout ,CD86 ,CD40 ,biology ,CD44 ,Dendritic Cells ,Flow Cytometry ,Coculture Techniques ,Cell biology ,Mice, Inbred C57BL ,Autocrine Communication ,030104 developmental biology ,Granzyme ,Interferon Type I ,biology.protein ,Signal Transduction ,030215 immunology - Abstract
Type I IFNs are key mediators of immune defense against viruses and bacteria. Type I IFNs were also previously implicated in protection against fungal infection, but their roles in antifungal immunity have not been thoroughly investigated. A recent study demonstrated that bacterial and fungal β-glucans stimulate IFN-β production by dendritic cells (DCs) following detection by the Dectin-1 receptor, but the effects of β-glucan–induced type I IFNs have not been defined. We investigated whether type I IFNs regulate CD8 T cell activation by fungal β-glucan particle–stimulated DCs. We demonstrate that β-glucan–stimulated DCs induce CD8 T cell proliferation, activation marker (CD44 and CD69) expression, and production of IFN-γ, IL-2, and granzyme B. Moreover, we show that type I IFNs support robust CD8 T cell activation (proliferation and IFN-γ and granzyme B production) by β-glucan–stimulated DCs in vitro and in vivo due to autocrine effects on the DCs. Specifically, type I IFNs promote Ag presentation on MHC I molecules, CD86 and CD40 expression, and the production of IL-12 p70, IL-2, IL-6, and TNF-α by β-glucan–stimulated DCs. We also demonstrate a role for autocrine type I IFN signaling in bacterial LPS-induced DC maturation, although, in the context of LPS stimulation, this mechanism is not so critical for CD8 T cell activation (promotes IFN-γ production but not proliferation or granzyme B production). This study provides insight into the mechanisms underlying CD8 T cell activation during infection, which may be useful in the rational design of vaccines directed against pathogens and tumors.
- Published
- 2017
- Full Text
- View/download PDF
9. Abstract 5549: Potency-reduced IL12 heterodimeric Fc-fusions exhibit strong anti-tumor activity
- Author
-
Matthew J. Bernett, Umesh Muchhal, Nargess Hassanzadeh-Kiabi, Seung Y. Chu, Connie Ardila, John R. Desjarlais, Ke Liu, Nicole Rodriguez, Christine Bonzon, Rajat Varma, and Rumana Rashid
- Subjects
Cancer Research ,Immune system ,Oncology ,In vivo ,Chemistry ,Interleukin 12 ,Potency ,IL-2 receptor ,Molecular biology ,STAT4 ,CD8 ,In vitro - Abstract
Interleukin-12 (IL12) is a proinflammatory cytokine produced by activated antigen-presenting cells that induces differentiation of Th1 cells and increased proliferation and cytotoxicity of T and NK cells. Stimulation of these cells by IL12 leads to production of high levels of IFNγ. These immune-stimulating aspects of IL12 are promising for cancer treatment and may help to convert immunologically suppressed “cold” tumors into inflamed “hot” tumors. Preclinical studies in mice revealed that IL12 can have a dramatic effect on shrinking syngeneic tumors, however clinical studies in humans have resulted in severe toxicity and a small therapeutic window, limiting response rates. Prior work at Xencor demonstrated that reduced-potency IL15/IL15Rα-Fc fusion proteins exhibited superior pharmacokinetics, pharmacodynamics, and safety in non-human primates through reduction of receptor-mediated clearance. Applying similar principles to IL12, we created various IL12 heterodimeric Fc-fusions (IL12-Fc) with reduced potency in order to improve tolerability, slow receptor-mediated clearance, and prolong half-life. IL12 is a heterodimeric protein consisting of two subunits, and thus we engineered IL12-Fc fusions by fusing the IL12p35 subunit to one side of a heterodimeric (and inactive) Fc domain, and the IL12p40 subunit to the other side. These Fc-fusions were tuned for optimal activity by introducing amino acid substitutions at putative receptor-interface positions and screening for reductions of in vitro potency. In vitro activity was assessed on normal human PBMCs by measuring signaling in a STAT4 phosphorylation assay and IFNγ production in a mixed-lymphocyte reaction (MLR). In vivo anti-tumor activity was assessed by engrafting pp65-MCF-7 cells into human PBMC engrafted NSG MHC class I and II double-knockout mice and by measuring tumor volume, lymphocyte activation/proliferation, and IFNγ production over time. IL12-Fc were produced with good yield and purity. An IL12-Fc potency series was created, and weaker variants had up to a 10,000-fold reduction in STAT4 signaling potency and IFNγ production in an MLR assay compared to a wild-type IL12-Fc. In vivo anti-tumor activity in the huPBMC-pp65-MCF7 model was achieved with reduced-potency IL12-Fc as a single-agent and in combination with anti-PD1, with weaker variants maintaining anti-tumor activity at higher dose levels. Analysis of peripheral lymphocytes indicated increased numbers of T and NK cells as well as activation of CD8+ T cells, as evidenced by upregulation of CD25. Increased expression of immune checkpoints including PD1 was also observed. Analysis of serum cytokine indicated up to 200-fold increases in IFNγ levels. Combined, these data indicate that reduced-potency IL12-Fc retain strong anti-tumor activity, with potential for improvement of therapeutic index. Citation Format: Rajat Varma, Ke Liu, Christine Bonzon, Rumana Rashid, Nicole Rodriguez, Nargess Hassanzadeh-Kiabi, Connie Ardila, Seung Y. Chu, Umesh S. Muchhal, John R. Desjarlais, Matthew J. Bernett. Potency-reduced IL12 heterodimeric Fc-fusions exhibit strong anti-tumor activity [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5549.
- Published
- 2020
- Full Text
- View/download PDF
10. IRF8 acts in lineage-committed rather than oligopotent progenitors to control neutrophil vs monocyte production
- Author
-
Alberto Yáñez, Madelena Y. Ng, Nargess Hassanzadeh-Kiabi, and Helen S. Goodridge
- Subjects
Neutrophils ,Cellular differentiation ,Immunology ,Apoptosis ,Granulocyte ,Biology ,Biochemistry ,Monocytes ,Mice ,medicine ,Animals ,Humans ,Cell Lineage ,Granulocyte Precursor Cells ,Progenitor cell ,Cells, Cultured ,Cell Proliferation ,Mice, Knockout ,Macrophages ,Monocyte ,Cell Differentiation ,Cell Biology ,Hematology ,Dendritic cell ,Flow Cytometry ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Monocyte differentiation ,Interferon Regulatory Factors ,Myelopoiesis ,IRF8 ,Granulocytes - Abstract
Interferon regulatory factor 8 (IRF8) is a key regulator of myelopoiesis in mice and humans. IRF8-deficient mice exhibit increased neutrophil numbers but defective monocyte and dendritic cell (DC) production. It has therefore been hypothesized that IRF8 regulates granulocyte vs monocyte/DC lineage commitment by oligopotent progenitors. Alternatively, IRF8 could control the differentiation of lineage-committed progenitors. In this study, we defined the role of IRF8 in lineage commitment and neutrophil vs monocyte differentiation using a novel sorting strategy that for the first time allows us to separate oligopotent granulocyte-monocyte progenitors (GMPs) and their lineage-committed progeny: granulocyte progenitors (GPs) and monocyte progenitors (MPs). We show that IRF8 is highly expressed by both GPs and MPs, but not GMPs, and is not required for GP or MP production by GMPs. In fact, IRF8-deficient mice have more GPs and MPs. This is not due to IRF8-mediated suppression of GP and MP production by GMPs, but rather to selective effects in GPs and MPs. We identify roles for IRF8 in regulating progenitor survival and differentiation and preventing leukemic cell accumulation. Thus, IRF8 does not regulate granulocytic vs monocytic fate in GMPs, but instead acts downstream of lineage commitment to selectively control neutrophil and monocyte production.
- Published
- 2015
- Full Text
- View/download PDF
11. Detection of a TLR2 agonist by hematopoietic stem and progenitor cells impacts the function of the macrophages they produce
- Author
-
Helen S. Goodridge, George Y. Liu, Nargess Hassanzadeh-Kiabi, David M. Underhill, Alberto Yáñez, Madelena Y. Ng, Javier Megías, M. Luisa Gil, and Aparna Subramanian
- Subjects
Myeloid ,medicine.medical_treatment ,Cellular differentiation ,Immunology ,Biology ,Cell biology ,Haematopoiesis ,Cytokine ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Macrophage ,Myelopoiesis ,Stem cell ,Progenitor cell - Abstract
Several groups have shown that detection of microbial components by TLRs on hematopoietic stem and progenitor cells (HSPCs) instructs myeloid cell generation, raising interest in the possibility of targeting TLRs on HSPCs to boost myelopoiesis. However, although "TLR-derived" cells exhibit myeloid cell characteristics (phagocytosis, cytokine production, antigen presentation), it is not clear whether they are functionally equivalent to macrophages derived in the absence of TLR activation. Our in vitro and in vivo studies show that macrophages derived from mouse and human HSPC subsets (including stem cells) exposed to a TLR2 agonist prior to or during macrophage differentiation produce lower levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) and reactive oxygen species. This is in contrast to prior exposure of differentiated macrophages to the TLR2 agonist ("tolerance"), which suppresses inflammatory cytokine production, but elevates reactive oxygen species. Soluble factors produced following exposure of HSPCs to a TLR2 agonist can also act in a paracrine manner to influence the function of macrophages derived from unexposed HSPCs. Our data demonstrate that macrophage function can be influenced by TLR signaling in the HSPCs from which they are derived, and that this may impact the clinical utility of targeting TLRs on HSPCs to boost myelopoiesis.
- Published
- 2013
- Full Text
- View/download PDF
12. Blimp-1 Functions as a Molecular Switch to Prevent Inflammatory Activity in Foxp3+RORγt+ Regulatory T Cells
- Author
-
Rashmi Bankoti, Deepti Dhall, Nargess Hassanzadeh-Kiabi, Rebecca A. Porritt, Gislâine A. Martins, Truc Nguyen, Samantha Nadeau, Michael Couse, Chihiro Ogawa, Caspar Ohnmacht, Xuemo Fan, and Gerald Eberl
- Subjects
0301 basic medicine ,Regulator ,chemical and pharmacologic phenomena ,Biology ,T-Lymphocytes, Regulatory ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,RAR-related orphan receptor gamma ,PRDM1 ,Transcriptional regulation ,Animals ,lcsh:QH301-705.5 ,Transcription factor ,Inflammation ,Interleukin-17 ,FOXP3 ,hemic and immune systems ,Forkhead Transcription Factors ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Colitis ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Blimp-1 ,Foxp3 ,Il-17 ,Irf4 ,Prdm1 ,Rorγt ,Treg ,Intestinal ,Transcription ,lcsh:Biology (General) ,Interferon Regulatory Factors ,Female ,Interleukin 17 ,Positive Regulatory Domain I-Binding Factor 1 ,IRF4 - Abstract
SUMMARY Foxp3+ regulatory T cells (Treg) are essential modulators of immune responses, but the molecular mechanisms underlying their function are not fully understood. Here we show that the transcription factor Blimp-1 is a crucial regulator of the Foxp3+RORγt+ Treg subset. The intrinsic expression of Blimp-1 in these cells is required to prevent production of Th17-associated cytokines. Direct binding of Blimp-1 to the Il17 locus in Treg is associated with inhibitory histone modifications but unaltered binding of RORgt. In the absence of Blimp-1, the Il17 locus is activated, with increased occupancy of the co-activator p300 and abundant binding of the transcriptional regulator IRF4, which is required, along with RORγt, for IL-17 expression in the absence of Blimp-1. We also show that despite their sustained expression of Foxp3, Blimp-1−/− RORγt+IL-17-producing Treg lose suppressor function and can promote intestinal inflammation, indicating that repression of Th17-associated cytokines by Blimp-1 is a crucial requirement for RORγt+ Treg function., Graphical Abstract, In Brief Ogawa et al. demonstrate that the transcription factor Blimp-1 is required to prevent production of Th17-associated cytokines and inflammatory activity of microbiota-specific Foxp3+RORγt+ Treg. These findings uncover a critical role for Blimp-1 in Foxp3+Treg function and shed light on the intricate mechanisms underlying Treg phenotypic stability.
- Published
- 2018
13. 175 ENHANCING DENDRITIC CELL VACCINES WITH LIGANDS FOR DECTIN-1 AND TOLL-LIKE RECEPTOR-9
- Author
-
Nargess Hassanzadeh-Kiabi, Hyung J. Kim, Yanping Wang, Helen S. Goodridge, and David M. Underhill
- Subjects
chemistry.chemical_classification ,business.industry ,Urology ,TLR9 ,Dendritic cell ,Lymphocyte proliferation ,Tumor antigen ,Immune system ,chemistry ,Cancer research ,Medicine ,Cytotoxic T cell ,business ,CD8 ,Glucan - Abstract
INTRODUCTION AND OBJECTIVES: Cancer vaccines have the potential to target tumors while sparing normal tissue. Dectin-1 receptor plays an important role in activating a cellular immune response against fungal infection, providing a rational for evaluating Dectin-1 ligand as an adjuvant for cancer vaccines. Activation of toll-like receptors (TLRs), particularly TLR-3, 4 and 9, has been reported to produce robust antitumor immune responses. Therefore, Dectin stimulation was evaluated alone or in combination with TLR stimulate. METHODS: To assess CD8 T cell responses, B16 lysatepulsed DCs were co-cultured with lymphocytes from pmel-1 mice. Pmel-1 transgenic mice express CD8 T cells with specificity for a Db-restricted epitope from the melanoma tumor antigen gp10025-33. The effect of stimulating DCs with ligands for Dectin-1, TLR, or both was assessed by monitoring pmel-1 lymphocyte proliferation and activation. Antitumor activity was assessed in vivo using syngeneic mouse tumor models for melanoma and renal cell carcinoma. RESULTS: Treating DCs with Dectin-1 ligand ( -glucan) alone did not increase CD8 T cell proliferation or IFNproduction. Furthermore, there was no significant effect on CD8 T cells when DCs were treated with -glucan combined with TLR4 ligand (lipopolysaccharide) or TLR3 ligand (poly I:C). However, the combination of -glucan and TLR9 ligand (unmethylated CpG oligonucleotides, CpG) increased CD8 lymphocyte proliferation significantly more than CpG or -glucan alone. In fact, the increase in CD8 proliferation was greater than with -glucan plus all 3 TLR ligands. The combination of -glucan and CpG was significantly more effective than -glucan alone in stimulating IFNproduction. Based on these results, the combination of -glucan and CpG was evaluated for enhancing DC vaccines in vivo. Bone marrow derived DCs were pulsed with tumor lysate and stimulated with -glucan, CpG or both, prior to injection in mice. Following two DC vaccine treatments, mice were subcutaneously challenged with B16 tumors. A tumor prevention model was used to unequivocally demonstrate that antitumor effects were immune mediated with no possibility of direct cytotoxic effect by -glucan or CpG. None of the mice treated with -glucan plus CpG grew tumor while all control mice rapidly grew tumor. These findings were replicated using the in vivo RENCA model for renal cell carcinoma. CONCLUSIONS: Preclinical data suggests that tumor lysatepulsed DC vaccines can be enhanced by stimulation of Dectin-1 and TLR-9.
- Published
- 2013
- Full Text
- View/download PDF
14. Detection of a TLR2 agonist by hematopoietic stem and progenitor cells impacts the function of the macrophages they produce
- Author
-
Alberto, Yáñez, Nargess, Hassanzadeh-Kiabi, Madelena Y, Ng, Javier, Megías, Aparna, Subramanian, George Y, Liu, David M, Underhill, M Luisa, Gil, and Helen S, Goodridge
- Subjects
Mice, Knockout ,Myelopoiesis ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,Macrophages ,Interleukin-1beta ,Cell Differentiation ,Hematopoietic Stem Cells ,Toll-Like Receptor 2 ,Article ,Mice, Inbred C57BL ,Lipopeptides ,Mice ,Phagocytosis ,Animals ,Myeloid Cells ,Reactive Oxygen Species ,Cells, Cultured ,Signal Transduction - Abstract
Several groups have shown that detection of microbial components by Toll-like receptors (TLRs) on hematopoietic stem and progenitor cells (HSPCs) instructs myeloid cell generation, raising interest in the possibility of targeting TLRs on HSPCs to boost myelopoiesis. However, although “TLR-derived” cells exhibit myeloid cell characteristics (phagocytosis, cytokine production, antigen presentation), it isn’t clear whether they are functionally equivalent to macrophages derived in the absence of TLR activation. Our in vitro and in vivo studies show that macrophages derived from mouse and human HSPC subsets (including stem cells) exposed to a TLR2 agonist prior to or during macrophage differentiation produce lower levels of inflammatory cytokines (TNF-α, IL-6 and IL-1β) and reactive oxygen species (ROS). This is in contrast to prior exposure of differentiated macrophages to the TLR2 agonist (“tolerance”), which suppresses inflammatory cytokine production, but elevates ROS. Soluble factors produced following exposure of HSPCs to a TLR2 agonist can also act in a paracrine manner to influence the function of macrophages derived from unexposed HSPCs. Our data demonstrate that macrophage function can be influenced by TLR signaling in the HSPCs from which they are derived, and that this may impact the clinical utility of targeting TLRs on HSPCs to boost myelopoiesis.
- Published
- 2013
15. The first trimester human placenta is a site for terminal maturation of primitive erythroid cells
- Author
-
Hanna K. A. Mikkola, Angela Chen, Nargess Hassanzadeh-Kiabi, Sacha L. Prashad, Eija Hämäläinen, Mattias Magnusson, Boriana Atanassova, Andy Huang, and Ben Van Handel
- Subjects
Placenta ,Immunology ,Enucleation ,Biology ,Biochemistry ,Red Cells, Iron, and Erythropoiesis ,Fetus ,Erythroid Cells ,Fetal membrane ,Pregnancy ,hemic and lymphatic diseases ,medicine ,Humans ,Erythropoiesis ,Macrophages ,hemic and immune systems ,Cell Biology ,Hematology ,Cell biology ,Haematopoiesis ,Pregnancy Trimester, First ,medicine.anatomical_structure ,embryonic structures ,Chorionic villi ,Fetoplacental Circulation ,Female ,Chorionic Villi ,circulatory and respiratory physiology - Abstract
Embryonic hematopoiesis starts via the generation of primitive red blood cells (RBCs) that satisfy the embryo's immediate oxygen needs. Although primitive RBCs were thought to retain their nuclei, recent studies have shown that primitive RBCs in mice enucleate in the fetal liver. It has been unknown whether human primitive RBCs enucleate, and what hematopoietic site might support this process. Our data indicate that the terminal maturation and enucleation of human primitive RBCs occurs in first trimester placental villi. Extravascular ζ-globin+ primitive erythroid cells were found in placental villi between 5-7 weeks of development, at which time the frequency of enucleated RBCs was higher in the villous stroma than in circulation. RBC enucleation was further evidenced by the presence of primitive reticulocytes and pyrenocytes (ejected RBC nuclei) in the placenta. Extravascular RBCs were found to associate with placental macrophages, which contained ingested nuclei. Clonogenic macrophage progenitors of fetal origin were present in the chorionic plate of the placenta before the onset of fetoplacental circulation, after which macrophages had migrated to the villi. These findings indicate that placental macrophages may assist the enucleation process of primitive RBCs in placental villi, implying an unexpectedly broad role for the placenta in embryonic hematopoiesis.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.