Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Scand J Immunol ; 95(3): e13132, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34936119

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. For unresectable HCC, transarterial radioembolization (TARE) with Yttrium-90 is a widely used treatment. The aim of this study was to investigate whether monocytic myeloid-derived suppressor cells (M-MDSC) and CD39+ T cells can be non-invasive predictive biomarkers of radiological response and prognosis in patients with HCC treated with TARE. This study was conducted on 39 patients with HCC who were treated with TARE between August 2018 and December 2019 and the control group consisted of 23 healthy volunteers. CD4+, CD8+, CD39+ T cells, Natural killer (NK) cells, myeloid cells (MC) and M-MDSC parameters are examined in the course of TARE treatment with student t test and Kaplan-Meier method. There were statistically significant differences in M-MDSC, CD39+ T cells and MC values between healthy controls and HCC patients. A statistically significant difference was found in M-MDSC and CD4+ T cells values in the HCC patient group who responded to the treatment compared to those who did not. Survival analysis found that patients with lower frequencies (under 3.81%) of M-MDSC showed more prominent differences of overall survival (OS) compared to patients with all high groups. We found that M-MDSC in the peripheral blood might be a useful non-invasive biomarker to predict OS. We have shown for the first time that M-MDSC is correlated with treatment response in HCC patients treated with TARE. Additionally, we have found that the percentage of CD39+ T cells is high in HCC patients and these cells are positively correlated with M-MDSC.


Subject(s)
Carcinoma, Hepatocellular/immunology , HLA-DR Antigens/immunology , Lipopolysaccharide Receptors/immunology , Liver Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/therapy , Case-Control Studies , Embolization, Therapeutic/methods , Female , Humans , Liver Neoplasms/therapy , Male , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/radiation effects , Myeloid-Derived Suppressor Cells/radiation effects , Prognosis , Prospective Studies , Survival Analysis , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Yttrium Radioisotopes/therapeutic use
2.
Proc Natl Acad Sci U S A ; 116(47): 23714-23723, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31712430

ABSTRACT

Tumor-associated myeloid cells (TAMCs) are key drivers of immunosuppression in the tumor microenvironment, which profoundly impedes the clinical response to immune-dependent and conventional therapeutic modalities. As a hallmark of glioblastoma (GBM), TAMCs are massively recruited to reach up to 50% of the brain tumor mass. Therefore, they have recently been recognized as an appealing therapeutic target to blunt immunosuppression in GBM with the hope of maximizing the clinical outcome of antitumor therapies. Here we report a nano-immunotherapy approach capable of actively targeting TAMCs in vivo. As we found that programmed death-ligand 1 (PD-L1) is highly expressed on glioma-associated TAMCs, we rationally designed a lipid nanoparticle (LNP) formulation surface-functionalized with an anti-PD-L1 therapeutic antibody (αPD-L1). We demonstrated that this system (αPD-L1-LNP) enabled effective and specific delivery of therapeutic payload to TAMCs. Specifically, encapsulation of dinaciclib, a cyclin-dependent kinase inhibitor, into PD-L1-targeted LNPs led to a robust depletion of TAMCs and an attenuation of their immunosuppressive functions. Importantly, the delivery efficiency of PD-L1-targeted LNPs was robustly enhanced in the context of radiation therapy (RT) owing to the RT-induced up-regulation of PD-L1 on glioma-infiltrating TAMCs. Accordingly, RT combined with our nano-immunotherapy led to dramatically extended survival of mice in 2 syngeneic glioma models, GL261 and CT2A. The high targeting efficiency of αPD-L1-LNP to human TAMCs from GBM patients further validated the clinical relevance. Thus, this study establishes a therapeutic approach with immense potential to improve the clinical response in the treatment of GBM and warrants a rapid translation into clinical practice.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Myeloid Cells/pathology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cyclic N-Oxides , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Indolizines , Mice , Myeloid Cells/drug effects , Myeloid Cells/radiation effects , Nanoparticles , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/therapeutic use , Tumor Microenvironment , Xenograft Model Antitumor Assays
3.
J Neuroinflammation ; 17(1): 279, 2020 Sep 20.
Article in English | MEDLINE | ID: mdl-32951604

ABSTRACT

BACKGROUND: Microglia, the primary resident myeloid cells of the brain, play critical roles in immune defense by maintaining tissue homeostasis and responding to injury or disease. However, microglial activation and dysfunction has been implicated in a number of central nervous system (CNS) disorders, thus developing tools to manipulate and replace these myeloid cells in the CNS is of therapeutic interest. METHODS: Using whole body irradiation, bone marrow transplant, and colony-stimulating factor 1 receptor inhibition, we achieve long-term and brain-wide (~ 80%) engraftment and colonization of peripheral bone marrow-derived myeloid cells (i.e., monocytes) in the brain parenchyma and evaluated the long-term effects of their colonization in the CNS. RESULTS: Here, we identify a monocyte signature that includes an upregulation in Ccr1, Ms4a6b, Ms4a6c, Ms4a7, Apobec1, Lyz2, Mrc1, Tmem221, Tlr8, Lilrb4a, Msr1, Nnt, and Wdfy1 and a downregulation of Siglech, Slc2a5, and Ccl21a/b. We demonstrate that irradiation and long-term (~ 6 months) engraftment of the CNS by monocytes induces brain region-dependent alterations in transcription profiles, astrocytes, neuronal structures, including synaptic components, and cognition. Although our results show that microglial replacement with peripherally derived myeloid cells is feasible and that irradiation-induced changes can be reversed by the replacement of microglia with monocytes in the hippocampus, we also observe that brain-wide engraftment of peripheral myeloid cells (relying on irradiation) can result in cognitive and synaptic deficits. CONCLUSIONS: These findings provide insight into better understanding the role and complexity of myeloid cells in the brain, including their regulation of other CNS cells and functional outcomes.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Transplantation/methods , Brain/cytology , Brain/immunology , Myeloid Cells/immunology , Animals , Bone Marrow/immunology , Bone Marrow/radiation effects , Brain/radiation effects , Central Nervous System/cytology , Central Nervous System/immunology , Central Nervous System/radiation effects , Male , Mice , Mice, Inbred C57BL , Monocytes/physiology , Monocytes/radiation effects , Myeloid Cells/radiation effects , Transcription, Genetic/physiology , Transcription, Genetic/radiation effects
4.
J Pathol ; 247(5): 606-614, 2019 04.
Article in English | MEDLINE | ID: mdl-30632153

ABSTRACT

Historically, our understanding of the cytotoxicity of radiation has centred on tumour cell-autonomous mechanisms of cell death. Here, tumour cell death occurs when a threshold number of radiation-induced non-reparable double-stranded DNA breaks is exceeded. However, in recent years, the importance of immune mechanisms of cell death has been increasingly recognised, as well as the impact of radiotherapy on non-malignant cellular components of the tumour microenvironment. Conserved antiviral pathways that detect foreign nucleic acid in the cytosol and drive downstream interferon (IFN) responses via the cyclic guanosine monophosphate-adenosine monophosphate synthase/stimulator of IFN genes (cGAS/STING) pathway are key components of the immune response to radiation-induced DNA damage. In preclinical models, acute induction of a type 1 IFN response is important for both direct and abscopal tumour responses to radiation. Inhibitors of the DNA damage response show promise in augmenting this inflammatory IFN response. However, a substantial proportion of tumours show chronic IFN signalling prior to radiotherapy, which paradoxically drives immunosuppression. This chronic IFN signalling leads to treatment resistance, and heterotypic interactions between stromal fibroblasts and tumour cells contribute to an aggressive tumour phenotype. The effect of radiotherapy on myeloid cell populations, particularly tumour-associated macrophages, has an additional impact on the immune tumour microenvironment. It is not yet clear how the above preclinical findings translate into a human context. Human tumours show greater intratumoural genomic heterogeneity and more variable levels of chromosomal instability than experimental murine models. High-quality translational studies of immunological changes occurring during radiotherapy that incorporate intrinsic tumour biology will enable a better understanding of the immunological consequences of radiation-induced DNA damage in patients. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
DNA Damage/radiation effects , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cancer-Associated Fibroblasts/immunology , Chromosomal Instability/genetics , Chromosomal Instability/immunology , Combined Modality Therapy , DNA Damage/immunology , Disease Models, Animal , Humans , Immune Tolerance/immunology , Immunologic Factors/therapeutic use , Interferon Type I/biosynthesis , Interferon Type I/radiation effects , Mice , Myeloid Cells/immunology , Myeloid Cells/radiation effects , Neoplasms/immunology , Neoplasms/radiotherapy , Radiation Dosage , Signal Transduction/immunology
5.
Int J Cancer ; 143(5): 1017-1028, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29417588

ABSTRACT

Cervical cancer is the fourth most commonly diagnosed cancer and the fourth leading cause of cancer death in women worldwide. Approximately half of cervical cancer patients present with locally advanced disease, for which surgery is not an option. These cases are nonetheless potentially curable with radiotherapy and cisplatin chemotherapy. Unfortunately, some tumours are resistant to treatment, and lymph node and distant recurrences are major problems in patients with advanced disease at diagnosis. New targeted treatments that can overcome treatment resistance and reduce metastases are urgently needed. The CXCL12/CXCR4 chemokine pathway is ubiquitously expressed in many normal tissues and cancers, including cervical cancer. Emerging evidence indicates that it plays a central role in cervical cancer pathogenesis, malignant progression, the development of metastases and radiation treatment response. Pre-clinical studies of standard-of-care fractionated radiotherapy and concurrent weekly cisplatin plus the CXCR4 inhibitor Plerixafor (AMD3100) in patient-derived orthotopic cervical cancer xenografts have shown improved primary tumour response and reduced lymph node metastases with no increase in early or late side effects. These studies have pointed the way forward to future clinical trials of radiotherapy/cisplatin plus Plerixafor or other newly emerging CXCL12 or CXCR4 inhibitors in women with cervical cancer.


Subject(s)
Chemokine CXCL12/antagonists & inhibitors , Myeloid Cells/pathology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy , Antineoplastic Agents/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Myeloid Cells/drug effects , Myeloid Cells/radiation effects , Radiotherapy , Uterine Cervical Neoplasms/pathology
6.
Blood Cells Mol Dis ; 63: 1-8, 2017 03.
Article in English | MEDLINE | ID: mdl-27888688

ABSTRACT

An oral therapeutic which reduces duration of cytopenias and is active following accidental radiation exposures is an unmet need in radiation countermeasures. Alpha methylhydrocinnamate (ST7) prolongs STAT-5 phosphorylation, reduces growth-factor dependency of multi-lineage cell lines, and stimulates erythropoiesis. Here, ST7 and its isomers were studied for their effects on myeloid progenitors and hematopoietic stem cells (HSCs) following radiation, in nonhuman primates, and murine irradiation models. Addition of ST7 or ST7-S increased CFU-GM production by 1.7-fold (p<0.001), reduced neutrophil apoptosis comparable to G-CSF, and enhanced HSC survival post-radiation by 2-fold, (p=0.028). ST7 and ST7-S administered in normal baboons increased ANC and platelet counts by 50-400%. In sub-lethally-irradiated mice, ANC nadir remained >200/mm3 and neutropenia recovered in 6days with ST7 treatment and 18days in controls (p<0.05). In lethally-irradiated mice, marrow pathology at 15days was hypocellular (10% cellularity) in controls, but normal (55-75% cellularity) with complete neutrophil maturation with ST7-S treatment. Following lethal irradiation, ST7, given orally for 4days, reduced mortality, with 30% survival in ST7-animals vs 8% in controls, (p<0.05). Collectively, the studies indicate that ST7 and ST7-S enhance myeloid recovery post-radiation and merit further evaluation to accelerate hematologic recovery in conditions of radiation-related and other marrow hypoplasias.


Subject(s)
Myeloid Cells/drug effects , Neutrophils/drug effects , Phenylpropionates/therapeutic use , Recovery of Function/drug effects , Whole-Body Irradiation/adverse effects , Animals , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Mice , Myeloid Cells/radiation effects , Neutrophils/radiation effects , Papio , Phenylpropionates/pharmacology , Radiation Exposure/adverse effects , Survival Rate , Whole-Body Irradiation/mortality
7.
J Neuroinflammation ; 13: 30, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26842770

ABSTRACT

BACKGROUND: Cranial radiotherapy is used to treat tumors of the central nervous system (CNS), as well as non-neoplastic conditions such as arterio-venous malformations; however, its use is limited by the tolerance of adjacent normal CNS tissue, which can lead to devastating long-term sequelae for patients. Despite decades of research, the underlying mechanisms by which radiation induces CNS tissue injury remain unclear. Neuroinflammation and immune cell infiltration are a recognized component of the CNS radiation response; however, the extent and mechanisms by which bone marrow-derived (BMD) immune cells participate in late radiation injury is unknown. Thus, we set out to better characterize the response and tested the hypothesis that C-C chemokine receptor type 2 (CCR2) signaling was required for myeloid cell recruitment following brain irradiation. METHODS: We used young adult C57BL/6 male bone marrow chimeric mice created with donor mice that constitutively express enhanced green fluorescent protein (eGFP). The head was shielded to avoid brain radiation exposure during chimera construction. Radiation dose and time response studies were conducted in wild-type chimeras, and additional experiments were performed with chimeras created using donor marrow from CCR2 deficient, eGFP-expressing mice. Infiltrating eGFP+ cells were identified and quantified using immunofluorescent microscopy. RESULTS: Brain irradiation resulted in a dose- and time-dependent infiltration of BMD immune cells (predominately myeloid) that began at 1 month and persisted until 6 months following ≥15 Gy brain irradiation. Infiltration was limited to areas that were directly exposed to radiation. CCR2 signaling loss resulted in decreased numbers of infiltrating cells at 6 months that appeared to be restricted to cells also expressing major histocompatibility complex class II molecules. CONCLUSIONS: The potential roles played by infiltrating immune cells are of current importance due to increasing interest in immunotherapeutic approaches for cancer treatment and a growing clinical interest in survivorship and quality of life issues. Our findings demonstrate that injury from brain radiation facilitates a dose- and time-dependent recruitment of BMD cells that persists for at least 6 months and, in the case of myeloid cells, is dependent on CCR2 signaling.


Subject(s)
Brain Injuries/etiology , Brain Injuries/pathology , Myeloid Cells/radiation effects , Radiation Injuries/complications , Receptors, CCR2/metabolism , Signal Transduction/radiation effects , Animals , Bone Marrow Transplantation , Calcium-Binding Proteins/metabolism , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Gene Expression Regulation/radiation effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Myosin Heavy Chains/metabolism , Neutrophil Infiltration/radiation effects , Radiation Chimera/physiology , Receptors, CCR2/genetics , Time Factors
8.
Carcinogenesis ; 35(6): 1310-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24469308

ABSTRACT

In human tumors, and in mouse models, cyclooxygenase-2 (COX-2) levels are frequently correlated with tumor development/burden. In addition to intrinsic tumor cell expression, COX-2 is often present in fibroblasts, myofibroblasts and endothelial cells of the tumor microenvironment, and in infiltrating immune cells. Intrinsic cancer cell COX-2 expression is postulated as only one of many sources for prostanoids required for tumor promotion/progression. Although both COX-2 inhibition and global Cox-2 gene deletion ameliorate ultraviolet B (UVB)-induced SKH-1 mouse skin tumorigenesis, neither manipulation can elucidate the cell type(s) in which COX-2 expression is required for tumorigenesis; both eliminate COX-2 activity in all cells. To address this question, we created Cox-2(flox/flox) mice, in which the Cox-2 gene can be eliminated in a cell-type-specific fashion by targeted Cre recombinase expression. Cox-2 deletion in skin epithelial cells of SKH-1 Cox-2(flox/flox);K14Cre(+) mice resulted, following UVB irradiation, in reduced skin hyperplasia and increased apoptosis. Targeted epithelial cell Cox-2 deletion also resulted in reduced tumor incidence, frequency, size and proliferation rate, altered tumor cell differentiation and reduced tumor vascularization. Moreover, Cox-2(flox/flox);K14Cre(+) papillomas did not progress to squamous cell carcinomas. In contrast, Cox-2 deletion in SKH-1 Cox-2(flox/flox); LysMCre(+) myeloid cells had no effect on UVB tumor induction. We conclude that (i) intrinsic epithelial COX-2 activity plays a major role in UVB-induced skin cancer, (ii) macrophage/myeloid COX-2 plays no role in UVB-induced skin cancer and (iii) either there may be another COX-2-dependent prostanoid source(s) that drives UVB skin tumor induction or there may exist a COX-2-independent pathway(s) to UVB-induced skin cancer.


Subject(s)
Cyclooxygenase 2/metabolism , Skin Neoplasms/etiology , Skin Neoplasms/metabolism , Ultraviolet Rays/adverse effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Cell Proliferation/radiation effects , Cyclooxygenase 2/genetics , DNA Damage/radiation effects , Disease Models, Animal , Epidermis/metabolism , Epidermis/pathology , Epidermis/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Gene Deletion , Gene Expression , Gene Targeting , Homozygote , Humans , Hyperplasia/genetics , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Cells/radiation effects , Neovascularization, Pathologic/genetics , Organ Specificity/genetics , Skin Neoplasms/pathology
9.
Clin Dev Immunol ; 2013: 281958, 2013.
Article in English | MEDLINE | ID: mdl-23653658

ABSTRACT

Radiation therapy is showing potential as a partner for immunotherapies in preclinical cancer models and early clinical studies. As has been discussed elsewhere, radiation provides debulking, antigen and adjuvant release, and inflammatory targeting of effector cells to the treatment site, thereby assisting multiple critical checkpoints in antitumor adaptive immunity. Adaptive immunity is terminated by inflammatory resolution, an active process which ensures that inflammatory damage is repaired and tissue function is restored. We discuss how radiation therapy similarly triggers inflammation followed by repair, the consequences to adaptive immune responses in the treatment site, and how the myeloid response to radiation may impact immunotherapies designed to improve control of residual cancer cells.


Subject(s)
Inflammation/radiotherapy , Myeloid Cells/radiation effects , Neoplasms/radiotherapy , Adaptive Immunity/radiation effects , Animals , Antigens, Neoplasm/immunology , Clinical Trials as Topic , Disease Models, Animal , Humans , Immunomodulation , Immunotherapy , Myeloid Cells/immunology , Neoplasms/immunology , Wound Healing/radiation effects
10.
Proc Natl Acad Sci U S A ; 107(18): 8363-8, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20404138

ABSTRACT

Despite recent advances in radiotherapy, loco-regional failures are still the leading cause of death in many cancer patients. We have previously reported that bone marrow-derived CD11b(+) myeloid cells are recruited to tumors grown in irradiated tissues, thereby restoring the vasculature and tumor growth. In this study, we examined whether neutralizing CD11b monoclonal antibodies could inhibit the recruitment of myeloid cells into irradiated tumors and inhibit their regrowth. We observed a significant enhancement of antitumor response to radiation in squamous cell carcinoma xenografts in mice when CD11b antibodies are administered systemically. Histological examination of tumors revealed that CD11b antibodies reduced infiltration of myeloid cells expressing S100A8 and matrix metalloproteinase-9. CD11b antibodies further inhibited bone marrow-derived cell adhesion and transmigration to C166 endothelial cell monolayers and chemotactic stimuli, respectively, to levels comparable to those from CD11b knockout or CD18 hypomorphic mice. Given the clinical availability of humanized CD18 antibodies, we tested two murine tumor models in CD18 hypomorphic or CD11b knockout mice and found that tumors were more sensitive to irradiation when grown in CD18 hypomorphic mice but not in CD11b knockout mice. When CD18 hypomorphism was partially rescued by reconstitution with the wild-type bone marrow, the resistance of the tumors to irradiation was restored. Our study thus supports the rationale of using clinically available Mac-1 (CD11b/CD18) antibodies as an adjuvant therapy to radiotherapy.


Subject(s)
CD11b Antigen/immunology , CD18 Antigens/immunology , Carcinoma, Squamous Cell/immunology , Cell Movement , Macrophage-1 Antigen/immunology , Myeloid Cells/immunology , Animals , Antibodies/immunology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/radiotherapy , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Knockout , Mice, Nude , Myeloid Cells/cytology , Myeloid Cells/radiation effects , Recurrence , Xenograft Model Antitumor Assays
11.
Methods Cell Biol ; 174: 17-30, 2023.
Article in English | MEDLINE | ID: mdl-36710048

ABSTRACT

The use of ionizing radiation (IR) is a cornerstone for the treatment of cancer and radiotherapy (RT) is used in roughly 50% of cancer patients. It is now well established that RT exerts widespread effects on the tumor stroma, including the immune environment. Together with its deeply characterized effects on the lymphoid compartment, RT also deeply affects the myeloid cell compartment. Fluorescence-activated flow cytometry is one of the most widely used technologies in immunology, allowing the multiparametric analysis of cells on a cell-by-cell basis. Here, we provide a detailed flow cytometry protocol to analyze the myeloid cell populations of human papillomavirus (HPV)-positive TC1/Luc tumors engrafted in the oral mucosa of immunocompetent mice, and to evaluate their modulations in response to RT. The same method, with slight modifications, can be used to study the tumor myeloid cells from a variety of other mouse tumors.


Subject(s)
Myeloid Cells , Neoplasms , Animals , Humans , Mice , Neoplasms/radiotherapy , Radiation, Ionizing , Myeloid Cells/radiation effects , Flow Cytometry
12.
Glia ; 60(5): 833-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22362506

ABSTRACT

Previous studies have shown that following whole-body irradiation bone marrow (BM)-derived cells can migrate into the central nervous system, including the retina, to give rise to microglia-like cells. The detailed mechanism, however, remains elusive. We show in this study that a single-dose whole-body γ-ray irradiation (8 Gy) induced subclinical damage (i.e., DNA damage) in the neuronal retina, which is accompanied by a low-grade chronic inflammation, para-inflammation, characterized by upregulated expression of chemokines (CCL2, CXCL12, and CX3CL1) and complement components (C4 and CFH), and microglial activation. The upregulation of chemokines CCL2 and CXCL12 and complement C4 lasted for more than 160 days, whereas the expression of CX3CL1 and CFH was upregulated for 2 weeks. Both resident microglia and BM-derived phagocytes displayed mild activation in the neuronal retina following irradiation. When BM cells from CX3CR1(gfp/+) mice or CX3CR1(gfp/gfp) mice were transplanted to wild-type C57BL/6 mice, more than 90% of resident CD11b(+) cells were replaced by donor-derived GFP(+) cells after 6 months. However, when transplanting CX3CR1(gfp/+) BM cells into CCL2-deficient mice, only 20% of retinal CD11b(+) cells were replaced by donor-derived cells at 6 month. Our results suggest that the neuronal retina suffers from a chronic stress following whole-body irradiation, and a para-inflammatory response is initiated, presumably to rectify the insults and maintain homeostasis. The recruitment of BM-derived myeloid cells is a part of the para-inflammatory response and is CCL2 but not CX3CL1 dependent.


Subject(s)
Bone Marrow Cells/metabolism , Chemokine CCL2/physiology , Inflammation Mediators/physiology , Myeloid Cells/metabolism , Retina/metabolism , Whole-Body Irradiation/adverse effects , Animals , Bone Marrow Cells/pathology , Bone Marrow Cells/radiation effects , Chemokine CCL2/radiation effects , Chemokine CXCL1/physiology , Inflammation Mediators/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/pathology , Myeloid Cells/radiation effects , Retina/pathology , Retina/radiation effects
13.
Br J Haematol ; 155(2): 167-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21810091

ABSTRACT

NUP98-HOXA9 [t(7;11) (p15;p15)] is associated with inferior prognosis in de novo and treatment-related acute myeloid leukaemia (AML) and contributes to blast crisis in chronic myeloid leukaemia (CML). We have engineered an inducible transgenic zebrafish harbouring human NUP98-HOXA9 under the zebrafish spi1(pu.1) promoter. NUP98-HOXA9 perturbed zebrafish embryonic haematopoiesis, with upregulated spi1 expression at the expense of gata1a. Markers associated with more differentiated myeloid cells, lcp1, lyz, and mpx were also elevated, but to a lesser extent than spi1, suggesting differentiation of early myeloid progenitors may be impaired by NUP98-HOXA9. Following irradiation, NUP98-HOXA9-expressing embryos showed increased numbers of cells in G2-M transition compared to controls and absence of a normal apoptotic response, which may result from an upregulation of bcl2. These data suggest NUP98-HOXA9-induced oncogenesis may result from a combination of defects in haematopoiesis and an aberrant response to DNA damage. Importantly, 23% of adult NUP98-HOXA9-transgenic fish developed a myeloproliferative neoplasm (MPN) at 19-23 months of age. In summary, we have identified an embryonic haematopoietic phenotype in a transgenic zebrafish line that subsequently develops MPN. This tool provides a unique opportunity for high-throughput in vivo chemical modifier screens to identify novel therapeutic agents in high risk AML.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Leukemia, Experimental/genetics , Myeloid Cells/pathology , Myeloproliferative Disorders/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Animals, Genetically Modified , Apoptosis , Cell Cycle , Cell Lineage , DNA Damage , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Genes, Reporter , Hematopoiesis/genetics , Homeodomain Proteins/physiology , Humans , Leukemia, Experimental/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Myeloid Cells/radiation effects , Myeloproliferative Disorders/pathology , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/physiology , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/physiology , Trans-Activators/genetics , Transgenes , Zebrafish/embryology , Zebrafish Proteins/physiology
14.
Viruses ; 12(8)2020 08 13.
Article in English | MEDLINE | ID: mdl-32823598

ABSTRACT

HIV-1 infects 39.5 million people worldwide, and cART is effective in preventing viral spread by reducing HIV-1 plasma viral loads to undetectable levels. However, viral reservoirs persist by mechanisms, including the inhibition of autophagy by HIV-1 proteins (i.e., Nef and Tat). HIV-1 reservoirs can be targeted by the "shock and kill" strategy, which utilizes latency-reversing agents (LRAs) to activate latent proviruses and immunotarget the virus-producing cells. Yet, limitations include reduced LRA permeability across anatomical barriers and immune hyper-activation. Ionizing radiation (IR) induces effective viral activation across anatomical barriers. Like other LRAs, IR may cause inflammation and modulate the secretion of extracellular vesicles (EVs). We and others have shown that cells may secrete cytokines and viral proteins in EVs and, therefore, LRAs may contribute to inflammatory EVs. In the present study, we mitigated the effects of IR-induced inflammatory EVs (i.e., TNF-α), through the use of mTOR inhibitors (mTORi; Rapamycin and INK128). Further, mTORi were found to enhance the selective killing of HIV-1-infected myeloid and T-cell reservoirs at the exclusion of uninfected cells, potentially via inhibition of viral transcription/translation and induction of autophagy. Collectively, the proposed regimen using cART, IR, and mTORi presents a novel approach allowing for the targeting of viral reservoirs, prevention of immune hyper-activation, and selectively killing latently infected HIV-1 cells.


Subject(s)
Cytokines/immunology , Extracellular Vesicles/immunology , HIV-1/radiation effects , Radiation, Ionizing , TOR Serine-Threonine Kinases/antagonists & inhibitors , Virus Latency/drug effects , Antiviral Agents/pharmacology , Autophagy/drug effects , Benzoxazoles/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/radiation effects , CD4-Positive T-Lymphocytes/virology , Extracellular Vesicles/virology , Female , HIV-1/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Male , Myeloid Cells/drug effects , Myeloid Cells/radiation effects , Myeloid Cells/virology , Pyrimidines/pharmacology , Sirolimus/pharmacology , U937 Cells , Virus Activation/radiation effects
15.
Radiat Res ; 170(3): 307-15, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18763856

ABSTRACT

We investigated the mechanism underlying the radioadaptive response that rescues mice from hematopoietic failure. C57BL/6 mice were irradiated with low-dose acute X rays (0.5 Gy) for priming 2 weeks prior to a high-dose (6 Gy) challenge irradiation. Bone marrow cells, erythrocytes and platelets in low-dose-preirradiated mice showed earlier recovery after the challenge irradiation than those in mice subjected only to the challenge irradiation. This suggests that hematopoiesis is enhanced after a challenge irradiation in preirradiated mice. The rapid recovery of bone marrow cells after the challenge irradiation was consistent with the proliferation of hematopoietic progenitors expressing the cell surface markers Lin-, Sca-1- and c-Kit+ in low-dose-preirradiated mice. A subpopulation of myeloid (Mac-1+/Gr-1+) cells, which were descendants of Lin-, Sca-1- and c-Kit+ cells, rapidly recovered in the bone marrow of low-dose-preirradiated mice, whereas the number of B-lymphoid (CD19+/B220+) cells did not show a statistically significant increase. Plasma cytokine profiles were analyzed using antibody arrays, and results indicated that the concentrations of several growth factors for myelopoiesis after the challenge irradiation were considerably increased by low-dose preirradiation. The rapid recovery of erythrocytes and platelets but not leukocytes was observed in the peripheral blood of preirradiated mice, suggesting that low-dose preirradiation triggered the differentiation to myelopoiesis. Thus the adaptive response induced by low-dose preirradiation in terms of the recovery kinetics of the number of hematopoietic cells may be due to the rapid recovery of the number of myeloid cells after high-dose irradiation.


Subject(s)
Adaptation, Physiological/physiology , Cytokines/metabolism , Myeloid Cells/physiology , Myeloid Cells/radiation effects , Recovery of Function/radiation effects , Whole-Body Irradiation/methods , Adaptation, Physiological/radiation effects , Animals , Female , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Radiation Tolerance
16.
Radiat Res ; 170(1): 15-22, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18582153

ABSTRACT

Irradiation of mice at doses of 1-1.5 Gy induced a predominant regeneration of the B-cell lineage but suppressed the regeneration of the myeloid lineage. The mechanisms underlying such reciprocal regulation of regeneration and the relationship between the two lineages remain unclear. Because the predominant regeneration of the B-cell lineage observed is considered to depend on the stromal cell function, and because the impairment of such stromal function may nullify such reciprocal responses, mouse models of senescent stromal cell impairment (SCI) and the less senescent stage of SCI (non-SCI) were compared to elucidate the mechanisms underlying the reciprocal regulation of both lineages after radiation exposure. In non-SCI mice irradiated with 1 Gy, the numbers of B-lymphocyte progenitor (CFU-preB) and granulocyte-macrophage progenitor (CFU-GM) cells in the bone marrow decreased rapidly during the first 24 h. Then the number of CFU-preB cells in the bone marrow promptly recovered from the nadir and exceeded the pretreatment level, whereas that of CFU-GM cells remained lower than the pretreatment level. The expression of genes encoding positive regulators of the B-lymphoid lineage [interleukin (IL)10, Flt3 ligand and IL7] was up-regulated; in contrast, expression of the positive regulators of the myeloid lineage [granulocyte macrophage colony-stimulating factor (GM-CSF) and stem cell factor (SCF)] was down-regulated. In SCI mice irradiated with 1 Gy, the oscillatory changes in the numbers of femoral CFU-preB and CFU-GM cells and in the expression levels of cytokine genes were less marked than those in the non-SCI mice. These results thus imply that the reciprocal regeneration depends on the up-regulation of IL10, Flt3 ligand and IL7 expression and the down-regulation of GM-CSF and SCF expression in the bone marrow, possibly depending on the hematopoietic microenvironment.


Subject(s)
B-Lymphocytes/radiation effects , Cell Lineage/radiation effects , Cytokines/metabolism , Interleukins/immunology , Membrane Proteins/metabolism , Myeloid Cells/radiation effects , Whole-Body Irradiation , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blood Cell Count , Bone Marrow/radiation effects , Cell Differentiation/immunology , Cell Differentiation/radiation effects , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/radiation effects , Male , Mice , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism
17.
J Leukoc Biol ; 81(3): 632-41, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17158608

ABSTRACT

Although granulocyte transfusion is a logical, therapeutic option for neutropenic patients with refractory infections, significant technical barriers have prevented its widespread use. A novel phagocyte transfusion strategy has been developed based on activation of a human myeloid cell line HL-60. To further define the potential for HL-60 cells to recapitulate white cell transfusions, a shortened duration of activation was evaluated, facile quality control markers were defined, and the impact of low-dose irradiation on cell function was determined. Three days of activation resulted in increased cell viability and in vitro candidacidal capacity but with slightly higher cell replication compared with 7 days of activation. Cell viability and several flow cytometric measurements were accurate, quality control markers for HL-60 activation. In combination with activation, low-dose irradiation abrogated replication while sparing the candidacidal effects of the HL-60 cells. Infusion of irradiated, activated HL-60 cells improved survival of neutropenic, candidemic mice significantly. In summary, activated, irradiated HL-60 cells are microbicidal, have virtually no replicative capacity, and are safe and effective at protecting neutropenic mice against an otherwise 100% fatal candidal infection. With continued development, this strategy to recapitulate neutrophil functions has the potential to serve as an effective alternative to granulocyte transfusions.


Subject(s)
Candidiasis/immunology , Leukocyte Transfusion , Myeloid Cells/immunology , Myeloid Cells/transplantation , Neutropenia/therapy , Animals , Candida albicans/immunology , Candidiasis/therapy , Cell Survival/immunology , Disease Models, Animal , Flow Cytometry , HL-60 Cells , Humans , Leukocyte Transfusion/methods , Male , Mice , Mice, Inbred BALB C , Myeloid Cells/radiation effects , Neutropenia/immunology , Transplantation, Heterologous
18.
PLoS One ; 12(7): e0181577, 2017.
Article in English | MEDLINE | ID: mdl-28732024

ABSTRACT

Our ability to use ionizing radiation as an energy source, as a therapeutic agent, and, unfortunately, as a weapon, has evolved tremendously over the past 120 years, yet our tool box to handle the consequences of accidental and unwanted radiation exposure remains very limited. We have identified a novel group of small molecule compounds with a 4-nitrophenylsulfonamide (NPS) backbone in common that dramatically decrease mortality from the hematopoietic acute radiation syndrome (hARS). The group emerged from an in vitro high throughput screen (HTS) for inhibitors of radiation-induced apoptosis. The lead compound also mitigates against death after local abdominal irradiation and after local thoracic irradiation (LTI) in models of subacute radiation pneumonitis and late radiation fibrosis. Mitigation of hARS is through activation of radiation-induced CD11b+Ly6G+Ly6C+ immature myeloid cells. This is consistent with the notion that myeloerythroid-restricted progenitors protect against WBI-induced lethality and extends the possible involvement of the myeloid lineage in radiation effects. The lead compound was active if given to mice before or after WBI and had some anti-tumor action, suggesting that these compounds may find broader applications to cancer radiation therapy.


Subject(s)
Acute Radiation Syndrome/drug therapy , Piperazines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cells, Cultured , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/radiation effects
19.
Transplantation ; 82(3): 332-9, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16906030

ABSTRACT

BACKGROUND: We previously described a reduced-intensity hematopoietic cell transplantation (HCT) regimen in dog leukocyte antigen (DLA)-haploidentical littermate recipients consisting of 450 cGy total body irradiation (TBI) and anti-CD44 monoclonal antibody (mAb) S5 before and mycophenolate mofetil (MMF)/cyclosporine (CSP) after HCT. METHODS: We tested a nonmyeloablative regimen of mAb S5 and 200 cGy TBI with postgrafting MMF/CSP in 44 DLA-haploidentical recipients using eight different regimens. Ten dogs also received escalating doses of donor lymphocyte infusion (DLI) alone or with pentostatin to convert to complete donor chimerism. RESULTS: All dogs achieved initial engraftment between one to two weeks after HCT with peripheral blood mononuclear cell (PBMC) donor chimerism ranging from 2% to 98% (median 37%) on day +35. Twenty-five (57%) dogs rejected their donor grafts at a median of seven (range; 1-19) weeks after HCT. Low levels of PBMC donor chimerism at three weeks predicted graft rejection. DLI neither facilitated conversion to full donor chimerism after HCT nor prevented rejection. Higher total nucleated cells, CD4+, CD8+, and CD14+ cell subset numbers in the PBMC graft were associated with stable full donor engraftment. Dogs given higher cell subset doses of infused PBMC achieved longer duration of mixed chimerism. CONCLUSIONS: Nonmyeloablative conditioning with 200 cGy TBI and anti-CD44 mAb was sufficient for initial uniform engraftment across DLA haplotype-mismatched barriers. However, sustained donor engraftment was seen in only 43% of recipients. Graft composition and donor-dominant chimerism early after HCT may be the most important factors for sustained donor engraftment.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Hematopoietic Stem Cell Transplantation , Hyaluronan Receptors/immunology , Immunotherapy , Leukocytes/immunology , Stem Cells/immunology , Animals , Cell Count , Chimera/immunology , Dogs , Dose-Response Relationship, Drug , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft vs Host Disease/immunology , Haploidy , Immunosuppressive Agents/pharmacology , Leukocytes/radiation effects , Models, Animal , Myeloid Cells/cytology , Myeloid Cells/radiation effects , Stem Cells/drug effects , Time Factors , Transplantation Conditioning , Transplantation, Homologous/immunology
20.
Tissue Eng Part C Methods ; 22(5): 509-15, 2016 05.
Article in English | MEDLINE | ID: mdl-26993746

ABSTRACT

Studies on hematopoiesis currently rely on animal models because in vitro culture methods do not accurately recapitulate complex bone marrow physiology. We recently described a bone marrow-on-a-chip microfluidic device that enables the culture of living hematopoietic bone marrow and mimics radiation toxicity in vitro. In the present study, we used this microdevice to demonstrate continuous blood cell production in vitro and model bone marrow responses to potential radiation countermeasure drugs. The device maintained mouse hematopoietic stem and progenitor cells in normal proportions for at least 2 weeks in culture. Increases in the number of leukocytes and red blood cells into the microfluidic circulation also could be detected over time, and addition of erythropoietin induced a significant increase in erythrocyte production. Exposure of the bone marrow chip to gamma radiation resulted in reduction of leukocyte production, and treatment of the chips with two potential therapeutics, granulocyte-colony stimulating factor or bactericidal/permeability-increasing protein (BPI), induced significant increases in the number of hematopoietic stem cells and myeloid cells in the fluidic outflow. In contrast, BPI was not found to have any effect when analyzed using static marrow cultures, even though it has been previously shown to accelerate recovery from radiation-induced toxicity in vivo. These findings demonstrate the potential value of the bone marrow-on-a-chip for modeling blood cell production, monitoring responses to hematopoiesis-modulating drugs, and testing radiation countermeasures in vitro.


Subject(s)
Bone Marrow/pathology , Gamma Rays/adverse effects , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Microfluidic Analytical Techniques/instrumentation , Models, Biological , Myeloid Cells/cytology , Animals , Antimicrobial Cationic Peptides/administration & dosage , Blood Proteins/administration & dosage , Bone Marrow/radiation effects , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/radiation effects , Mice , Mice, Inbred C57BL , Myeloid Cells/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL