Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 10(5)2022 05.
Article in English | MEDLINE | ID: mdl-35577501

ABSTRACT

Immunotherapy with gene engineered CAR and TCR transgenic T-cells is a transformative treatment in cancer medicine. There is a rich pipeline with target antigens and sophisticated technologies that will enable establishing this novel treatment not only in rare hematological malignancies, but also in common solid tumors. The T2EVOLVE consortium is a public private partnership directed at accelerating the preclinical development of and increasing access to engineered T-cell immunotherapies for cancer patients. A key ambition in T2EVOLVE is to assess the currently available preclinical models for evaluating safety and efficacy of engineered T cell therapy and developing new models and test parameters with higher predictive value for clinical safety and efficacy in order to improve and accelerate the selection of lead T-cell products for clinical translation. Here, we review existing and emerging preclinical models that permit assessing CAR and TCR signaling and antigen binding, the access and function of engineered T-cells to primary and metastatic tumor ligands, as well as the impact of endogenous factors such as the host immune system and microbiome. Collectively, this review article presents a perspective on an accelerated translational development path that is based on innovative standardized preclinical test systems for CAR and TCR transgenic T-cell products.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy , Immunotherapy, Adoptive , Neoplasms/therapy , T-Lymphocytes
3.
Front Immunol ; 11: 554725, 2020.
Article in English | MEDLINE | ID: mdl-33072095

ABSTRACT

Systemic Lupus Erythematosus (SLE) is a chronic inflammatory autoimmune disease in which type I interferons (IFN) play a key role. The IFN response can be triggered when oxidized DNA engages the cytosolic DNA sensing platform cGAS-STING, but the repair mechanisms that modulate this process and govern disease progression are unclear. To gain insight into this biology, we interrogated the role of oxyguanine glycosylase 1 (OGG1), which repairs oxidized guanine 8-Oxo-2'-deoxyguanosine (8-OH-dG), in the pristane-induced mouse model of SLE. Ogg1-/- mice showed increased influx of Ly6Chi monocytes into the peritoneal cavity and enhanced IFN-driven gene expression in response to short-term exposure to pristane. Loss of Ogg1 was associated with increased auto-antibodies (anti-dsDNA and anti-RNP), higher total IgG, and expression of interferon stimulated genes (ISG) to longer exposure to pristane, accompanied by aggravated skin pathology such as hair loss, thicker epidermis, and increased deposition of IgG in skin lesions. Supporting a role for type I IFNs in this model, skin lesions of Ogg1-/- mice had significantly higher expression of type I IFN genes (Isg15, Irf9, and Ifnb). In keeping with loss of Ogg1 resulting in dysregulated IFN responses, enhanced basal and cGAMP-dependent Ifnb expression was observed in BMDMs from Ogg1-/- mice. Use of the STING inhibitor, H151, reduced both basal and cGAMP-driven increases, indicating that OGG1 regulates Ifnb expression through the cGAS-STING pathway. Finally, in support for a role for OGG1 in the pathology of cutaneous disease, reduced OGG1 expression in monocytes associated with skin involvement in SLE patients and the expression of OGG1 was significantly lower in lesional skin compared with non-lesional skin in patients with Discoid Lupus. Taken together, these data support an important role for OGG1 in protecting against IFN production and SLE skin disease.


Subject(s)
DNA Damage/immunology , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Systemic/immunology , Skin/immunology , Terpenes/adverse effects , Animals , DNA Glycosylases/deficiency , DNA Glycosylases/immunology , Disease Models, Animal , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lupus Erythematosus, Cutaneous/chemically induced , Lupus Erythematosus, Cutaneous/genetics , Lupus Erythematosus, Cutaneous/pathology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Oxidation-Reduction/drug effects , Skin/pathology , Terpenes/pharmacology
4.
Nature ; 585(7823): 96-101, 2020 09.
Article in English | MEDLINE | ID: mdl-32814898

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative disorders that overlap in their clinical presentation, pathology and genetic origin. Autoimmune disorders are also overrepresented in both ALS and FTD, but this remains an unexplained epidemiologic observation1-3. Expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial ALS and FTD (C9-ALS/FTD), and lead to both repeat-containing RNA and dipeptide accumulation, coupled with decreased C9orf72 protein expression in brain and peripheral blood cells4-6. Here we show in mice that loss of C9orf72 from myeloid cells alone is sufficient to recapitulate the age-dependent lymphoid hypertrophy and autoinflammation seen in animals with a complete knockout of C9orf72. Dendritic cells isolated from C9orf72-/- mice show marked early activation of the type I interferon response, and C9orf72-/- myeloid cells are selectively hyperresponsive to activators of the stimulator of interferon genes (STING) protein-a key regulator of the innate immune response to cytosolic DNA. Degradation of STING through the autolysosomal pathway is diminished in C9orf72-/- myeloid cells, and blocking STING suppresses hyperactive type I interferon responses in C9orf72-/- immune cells as well as splenomegaly and inflammation in C9orf72-/- mice. Moreover, mice lacking one or both copies of C9orf72 are more susceptible to experimental autoimmune encephalitis, mirroring the susceptibility to autoimmune diseases seen in people with C9-ALS/FTD. Finally, blood-derived macrophages, whole blood and brain tissue from patients with C9-ALS/FTD all show an elevated type I interferon signature compared with samples from people with sporadic ALS/FTD; this increased interferon response can be suppressed with a STING inhibitor. Collectively, our results suggest that patients with C9-ALS/FTD have an altered immunophenotype because their reduced levels of C9orf72 cannot suppress the inflammation mediated by the induction of type I interferons by STING.


Subject(s)
C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Inflammation/metabolism , Inflammation/prevention & control , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Aging/immunology , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein/deficiency , Dendritic Cells/cytology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Membrane Proteins/antagonists & inhibitors , Mice , Myeloid Cells/immunology , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology
5.
JACC Basic Transl Sci ; 5(6): 582-598, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32613145

ABSTRACT

In the Ldlr -/- mouse model of atherosclerosis, female Nlrp3 -/- bone marrow chimera and Nlrp3 -/- mice developed significantly smaller lesions in the aortic sinus and decreased lipid content in aorta en face, but a similar protection was not observed in males. Ovariectomized female mice lost protection from atherosclerosis in the setting of NLRP3 deficiency, whereas atherosclerosis showed a greater dependency on NLRP3 in castrated males. Thus, castration increased the dependency of atherosclerosis on the NLRP3 inflammasome, suggesting that testosterone may block inflammation in atherogenesis. Conversely, ovariectomy reduced the dependency on NLRP3 inflammasome components for atherogenesis, suggesting that estrogen may promote inflammasome-mediated atherosclerosis.

6.
Nat Commun ; 11(1): 1613, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32235862

ABSTRACT

In men, the incidence of melanoma rises rapidly after age 50, and nearly two thirds of melanoma deaths are male. The immune system is known to play a key role in controlling the growth and spread of malignancies, but whether age- and sex-dependent changes in immune cell function account for this effect remains unknown. Here, we show that in castrated male mice, neutrophil maturation and function are impaired, leading to elevated metastatic burden in two models of melanoma. Replacement of testosterone effectively normalized the tumor burden in castrated male mice. Further, the aberrant neutrophil phenotype was also observed in prostate cancer patients receiving androgen deprivation therapy, highlighting the evolutionary conservation and clinical relevance of the phenotype. Taken together, these results provide a better understanding of the role of androgen signaling in neutrophil function and the impact of this biology on immune control of malignancies.


Subject(s)
Androgen Antagonists/pharmacology , Neutrophils/immunology , Neutrophils/metabolism , Testosterone/metabolism , Androgen Antagonists/therapeutic use , Androgens , Animals , Antineoplastic Agents/pharmacology , Bone Marrow/pathology , Bone Marrow Transplantation , Disease Models, Animal , Female , Hormone Replacement Therapy/methods , Lung/pathology , Male , Melanoma/immunology , Melanoma/pathology , Melanoma/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Testosterone/immunology
8.
Arterioscler Thromb Vasc Biol ; 40(3): 802-818, 2020 03.
Article in English | MEDLINE | ID: mdl-31996019

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the leading cause of acute vasculitis and acquired heart disease in children in developed countries. Notably, KD is more prevalent in males than females. We previously established a key role for IL (interleukin)-1 signaling in KD pathogenesis, but whether this pathway underlies the sex-based difference in susceptibility is unknown. Approach and Results: The role of IL-1 signaling was investigated in the Lactobacillus casei cell wall extract-induced experimental mouse model of KD vasculitis. Five-week-old male and female mice were injected intraperitoneally with PBS, Lactobacillus caseicell wall extract, or a combination of Lactobacillus caseicell wall extract and the IL-1 receptor antagonist Anakinra. Aortitis, coronary arteritis inflammation score and abdominal aorta dilatation, and aneurysm development were assessed. mRNA-seq (messenger RNA sequencing) analysis was performed on abdominal aorta tissue. Publicly available human transcriptomics data from patients with KD was analyzed to identify sex differences and disease-associated genes. Male mice displayed enhanced aortitis and coronary arteritis as well as increased incidence and severity of abdominal aorta dilatation and aneurysm, recapitulating the increased incidence in males that is observed in human KD. Gene expression data from patients with KD and abdominal aorta tissue of Lactobacillus caseicell wall extract-injected mice showed enhanced Il1b expression and IL-1 signaling genes in males. Although the more severe IL-1ß-mediated disease phenotype observed in male mice was ameliorated by Anakinra treatment, the milder disease phenotype in female mice failed to respond. CONCLUSIONS: IL-1ß may play a central role in mediating sex-based differences in KD, with important implications for the use of anti-IL-1ß therapies to treat male and female patients with KD.


Subject(s)
Aorta, Abdominal/metabolism , Interleukin-1beta/metabolism , Mucocutaneous Lymph Node Syndrome/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Aorta, Abdominal/immunology , Case-Control Studies , Disease Models, Animal , Drug Resistance , Female , Health Status Disparities , Humans , Incidence , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/genetics , Lacticaseibacillus casei , Male , Mice, Inbred C57BL , Mucocutaneous Lymph Node Syndrome/drug therapy , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/microbiology , Risk Factors , Severity of Illness Index , Sex Factors , Signal Transduction
9.
Nat Commun ; 10(1): 3850, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31462642

ABSTRACT

Brain glioma treatment with checkpoint inhibitor antibodies to cytotoxic T-lymphocyte-associated antigen 4 (a-CTLA-4) and programmed cell death-1 (a-PD-1) was largely unsuccessful due to their inability to cross blood-brain barrier (BBB). Here we describe targeted nanoscale immunoconjugates (NICs) on natural biopolymer scaffold, poly(ß-L-malic acid), with covalently attached a-CTLA-4 or a-PD-1 for systemic delivery across the BBB and activation of local brain anti-tumor immune response. NIC treatment of mice bearing intracranial GL261 glioblastoma (GBM) results in an increase of CD8+ T cells, NK cells and macrophages with a decrease of regulatory T cells (Tregs) in the brain tumor area. Survival of GBM-bearing mice treated with NIC combination is significantly longer compared to animals treated with single checkpoint inhibitor-bearing NICs or free a-CTLA-4 and a-PD-1. Our study demonstrates trans-BBB delivery of tumor-targeted polymer-conjugated checkpoint inhibitors as an effective GBM treatment via activation of both systemic and local privileged brain tumor immune response.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunoconjugates/administration & dosage , Nanoconjugates/chemistry , Animals , Antineoplastic Agents, Immunological/pharmacokinetics , Biopolymers/chemistry , Blood-Brain Barrier/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Glioma/immunology , Glioma/pathology , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Malates/chemistry , Mice , Permeability , Physarum polycephalum/chemistry , Polymers/chemistry , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
10.
Immunity ; 49(5): 873-885.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30366765

ABSTRACT

Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses.


Subject(s)
Cell Differentiation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Atherosclerosis , Biomarkers , Caspase Activation and Recruitment Domain , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/mortality , Gene Expression , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/biosynthesis , Interleukin-1beta , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
11.
ACS Nano ; 9(5): 5594-608, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25906400

ABSTRACT

Differential diagnosis of brain magnetic resonance imaging (MRI) enhancement(s) remains a significant problem, which may be difficult to resolve without biopsy, which can be often dangerous or even impossible. Such MRI enhancement(s) can result from metastasis of primary tumors such as lung or breast, radiation necrosis, infections, or a new primary brain tumor (glioma, meningioma). Neurological symptoms are often the same on initial presentation. To develop a more precise noninvasive MRI diagnostic method, we have engineered a new class of poly(ß-l-malic acid) polymeric nanoimaging agents (NIAs). The NIAs carrying attached MRI tracer are able to pass through the blood-brain barrier (BBB) and specifically target cancer cells for efficient imaging. A qualitative/quantitative "MRI virtual biopsy" method is based on a nanoconjugate carrying MRI contrast agent gadolinium-DOTA and antibodies recognizing tumor-specific markers and extravasating through the BBB. In newly developed double tumor xenogeneic mouse models of brain metastasis this noninvasive method allowed differential diagnosis of HER2- and EGFR-expressing brain tumors. After MRI diagnosis, breast and lung cancer brain metastases were successfully treated with similar tumor-targeted nanoconjugates carrying molecular inhibitors of EGFR or HER2 instead of imaging contrast agent. The treatment resulted in a significant increase in animal survival and markedly reduced immunostaining for several cancer stem cell markers. Novel NIAs could be useful for brain diagnostic MRI in the clinic without currently performed brain biopsies. This technology shows promise for differential MRI diagnosis and treatment of brain metastases and other pathologies when biopsies are difficult to perform.


Subject(s)
Biopsy/methods , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Brain/pathology , Magnetic Resonance Imaging/methods , Nanoconjugates , Nanomedicine/methods , Animals , Base Sequence , Blood-Brain Barrier/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Diagnosis, Differential , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Nanoconjugates/chemistry , Neoplasm Metastasis , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Receptor, ErbB-2/metabolism , Survival Analysis
12.
J Gastrointest Oncol ; 6(2): 208-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25830040

ABSTRACT

The development of cancer is a multi-step process involving the gradual loss of regulation over the growth and functional capabilities of normal cells. Much research has been focused on the numerous cell intrinsic factors that govern this process; however, recent attention has turned to understanding the cell extrinsic factors in the tumor microenvironment that appear equally critical to the progression and treatment of cancer. One critical component of the tumor microenvironment is the immune system and it has become increasingly evident that the immune system plays an integral role in preventing and promoting the development of cancer. Understanding the immune cell types and pathways involved in this process has enabled the development of novel biomarkers for prognosis and accelerated the development of immune-based therapeutics, both of which have the potential to forever change the treatment paradigms for colorectal cancer (CRC). In this review, we discuss the impact of the immune system on the initiation, progression and treatment of cancer, specifically focusing on CRC.

13.
Adv Drug Deliv Rev ; 65(13-14): 1866-79, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24120656

ABSTRACT

Nanomedicine is an emerging form of therapy that focuses on alternative drug delivery and improvement of the treatment efficacy while reducing detrimental side effects to normal tissues. Cancer drug resistance is a complicated process that involves multiple mechanisms. Here we discuss the major forms of drug resistance and the new possibilities that nanomedicines offer to overcome these treatment obstacles. Novel nanomedicines that have a high ability for flexible, fast drug design and production based on tumor genetic profiles can be created making drug selection for personal patient treatment much more intensive and effective. This review aims to demonstrate the advantage of the young medical science field, nanomedicine, for overcoming cancer drug resistance. With the advanced design and alternative mechanisms of drug delivery known for different nanodrugs including liposomes, polymer conjugates, micelles, dendrimers, carbon-based, and metallic nanoparticles, overcoming various forms of multi-drug resistance looks promising and opens new horizons for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Nanomedicine/methods , Nanoparticles/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Clinical Trials as Topic , Drug Design , Humans , Nanomedicine/trends , Neoplasms/drug therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Tumor Microenvironment/drug effects
14.
J Drug Target ; 21(10): 956-967, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24032759

ABSTRACT

Engineered nanoparticles are widely used for delivery of drugs but frequently lack proof of safety for cancer patient's treatment. All-in-one covalent nanodrugs of the third generation have been synthesized based on a poly(ß-L-malic acid) (PMLA) platform, targeting human triple-negative breast cancer (TNBC). They significantly inhibited tumor growth in nude mice by blocking synthesis of epidermal growth factor receptor, and α4 and ß1 chains of laminin-411, the tumor vascular wall protein and angiogenesis marker. PMLA and nanodrug biocompatibility and toxicity at low and high dosages were evaluated in vitro and in vivo. The dual-action nanodrug and single-action precursor nanoconjugates were assessed under in vitro conditions and in vivo with multiple treatment regimens (6 and 12 treatments). The monitoring of TNBC treatment in vivo with different drugs included blood hematologic and immunologic analysis after multiple intravenous administrations. The present study demonstrates that the dual-action nanoconjugate is highly effective in preclinical TNBC treatment without side effects, supported by hematologic and immunologic assays data. PMLA-based nanodrugs of the Polycefin™ family passed multiple toxicity and efficacy tests in vitro and in vivo on preclinical level and may prove to be optimized and efficacious for the treatment of cancer patients in the future.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Malates/chemistry , Polymers/chemistry , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Carriers/chemistry , Drug Evaluation, Preclinical , ErbB Receptors/antagonists & inhibitors , Female , Humans , Laminin/metabolism , Mice , Mice, Nude , Nanoconjugates , Nanoparticles , Polyesters/chemistry , Rabbits , Triple Negative Breast Neoplasms/pathology
15.
Exp Toxicol Pathol ; 65(7-8): 1063-71, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23688656

ABSTRACT

Air pollution negatively impacts pulmonary, cardiovascular, and central nervous systems. Although its influence on brain cancer is unclear, toxic pollutants can cause blood-brain barrier disruption, enabling them to reach the brain and cause alterations leading to tumor development. By gene microarray analysis validated by quantitative RT-PCR and immunostaining we examined whether rat (n=104) inhalation exposure to air pollution particulate matter (PM) resulted in brain molecular changes similar to those associated with human brain tumors. Global brain gene expression was analyzed after exposure to PM (coarse, 2.5-10µm; fine, <2.5µm; or ultrafine, <0.15µm) and purified air for different times, short (0.5, 1, and 3 months) and chronic (10 months), for 5h per day, four days per week. Expression of select gene products was also studied in human brain (n=7) and in tumors (n=83). Arc/Arg3.1 and Rac1 genes, and their protein products were selected for further examination. Arc was elevated upon two-week to three-month exposure to coarse PM and declined after 10-month exposure. Rac1 was significantly elevated upon 10-month coarse PM exposure. On human brain tumor sections, Arc was expressed in benign meningiomas and low-grade gliomas but was much lower in high-grade tumors. Conversely, Rac1 was elevated in high-grade vs. low-grade gliomas. Arc is thus associated with early brain changes and low-grade tumors, whereas Rac1 is associated with long-term PM exposure and highly aggressive tumors. In summary, exposure to air PM leads to distinct changes in rodent brain gene expression similar to those observed in human brain tumors.


Subject(s)
Air Pollution/adverse effects , Brain Neoplasms/genetics , Cytoskeletal Proteins/genetics , Gene Expression Regulation/drug effects , Nerve Tissue Proteins/genetics , Particulate Matter/adverse effects , rac1 GTP-Binding Protein/genetics , Animals , Cytoskeletal Proteins/biosynthesis , Humans , Immunohistochemistry , Los Angeles , Male , Nerve Tissue Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred F344 , Real-Time Polymerase Chain Reaction , Transcriptome , rac1 GTP-Binding Protein/biosynthesis
16.
Cell Mol Immunol ; 9(6): 455-63, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23042535

ABSTRACT

Previously, we established a model in which physiologically adequate function of the autologous ß cells was recovered in non-obese diabetic (NOD) mice after the onset of hyperglycemia by rendering them hemopoietic chimera. These mice were termed antea-diabetic. In the current study, we addressed the role of T regulatory (Treg) cells in the mechanisms mediating the restoration of euglycemia in the antea-diabetic NOD model. The data generated in this study demonstrated that the numbers of Treg cells were decreased in unmanipulated NOD mice, with the most profound deficiency detected in the pancreatic lymph nodes (PLNs). The impaired retention of the Treg cells in the PLNs correlated with the locally compromised profile of the chemokines involved in their trafficking, with the most prominent decrease observed in SDF-1. The amelioration of autoimmunity and restoration of euglycemia observed in the antea-diabetic mice was associated with restoration of the Treg cell population in the PLNs. These data indicate that the function of the SDF-1/CXCR4 axis and the retention of Treg cells in the PLNs have a potential role in diabetogenesis and in the amelioration of autoimmunity and ß cell regeneration in the antea-diabetic model. We have demonstrated in the antea-diabetic mouse model that lifelong recovery of the ß cells has a strong correlation with normalization of the Treg cell population in the PLNs. This finding offers new opportunities for testing the immunomodulatory regimens that promote accumulation of Treg cells in the PLNs as a therapeutic approach for type 1 diabetes (T1D).


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Regeneration/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Count , Cell Movement/immunology , Chemokines/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Histocompatibility Antigens Class I/immunology , Immune Tolerance/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Mice , Mice, Inbred NOD
17.
PLoS One ; 7(2): e31070, 2012.
Article in English | MEDLINE | ID: mdl-22355336

ABSTRACT

Treatment options for triple negative breast cancer (TNBC) are generally limited to cytotoxic chemotherapy. Recently, anti-epidermal growth factor receptor (EGFR) therapy has been introduced for TNBC patients. We engineered a novel nanobioconjugate based on a poly(ß-L-malic acid) (PMLA) nanoplatform for TNBC treatment. The nanobioconjugate carries anti-tumor nucleosome-specific monoclonal antibody (mAb) 2C5 to target breast cancer cells, anti-mouse transferrin receptor (TfR) antibody for drug delivery through the host endothelial system, and Morpholino antisense oligonucleotide (AON) to inhibit EGFR synthesis. The nanobioconjugates variants were: (1) P (BioPolymer) with AON, 2C5 and anti-TfR for tumor endothelial and cancer cell targeting, and EGFR suppression (P/AON/2C5/TfR), and (2) P with AON and 2C5 (P/AON/2C5). Controls included (3) P with 2C5 but without AON (P/2C5), (4) PBS, and (5) P with PEG and leucine ester (LOEt) for endosomal escape (P/mPEG/LOEt). Drugs were injected intravenously to MDA-MB-468 TNBC bearing mice. Tissue accumulation of injected nanobioconjugates labeled with Alexa Fluor 680 was examined by Xenogen IVIS 200 (live imaging) and confocal microscopy of tissue sections. Levels of EGFR, phosphorylated and total Akt in tumor samples were detected by western blotting. In vitro western blot showed that the leading nanobioconjugate P/AON/2C5/TfR inhibited EGFR synthesis significantly better than naked AON. In vivo imaging revealed that 2C5 increased drug-tumor accumulation. Significant tumor growth inhibition was observed in mice treated with the lead nanobioconjugate (1) [P = 0.03 vs. controls; P<0.05 vs. nanobioconjugate variant (2)]. Lead nanobioconjugate (1) also showed stronger inhibition of EGFR expression and Akt phosphorylation than other treatments. Treatment of TNBC with the new nanobioconjugate results in tumor growth arrest by inhibiting EGFR and its downstream signaling intermediate, phosphorylated Akt. The nanobioconjugate represents a new generation of nanodrugs for treatment of TNBC.


Subject(s)
Biopolymers/therapeutic use , Breast Neoplasms/therapy , ErbB Receptors/antagonists & inhibitors , Malates/chemistry , Nanoparticles/therapeutic use , Oligonucleotides, Antisense/therapeutic use , Polymers/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Nude , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Front Biosci (Elite Ed) ; 3(2): 453-62, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196325

ABSTRACT

HIV-1-based vectors are widely used in gene therapy. In somatic cells, these vectors mainly integrate within genes. However, no distinct integration site preferences have been observed with regard to large chromosomal regions. The recent emergence of induced pluripotent stem (iPS) cells, similar to embryonic stem (ES) cells, has raised questions about where integration occurs in these cells. In this work we investigated the integration site preferences of HIV-1-based vectors in a pluripotent, ES-like cell line. We show that approximately 30% of the integrations occur in the vicinity of telomeres. We have analyzed integration sites in various somatic cells, as reported by us and other groups, and observed that this integration pattern is unique to the analyzed pluripotent cell line. We conclude that pluripotent cells may contain distinct cellular cofactors that participate in integration targeting and that are not present in somatic cells.


Subject(s)
Genetic Therapy/methods , HIV-1/metabolism , Pluripotent Stem Cells/physiology , Telomere/metabolism , Virus Integration/physiology , Cell Line, Tumor , Cloning, Molecular , DNA Primers/genetics , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genomic Library , Humans , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...