Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Nat Commun ; 14(1): 5346, 2023 09 02.
Article in English | MEDLINE | ID: mdl-37660083

ABSTRACT

Chimeric antigen receptor (CAR) T cells have transformed the treatment landscape for hematological malignancies. However, CAR T cells are less efficient against solid tumors, largely due to poor infiltration resulting from the immunosuppressive nature of the tumor microenvironment (TME). Here, we assessed the efficacy of Lewis Y antigen (LeY)-specific CAR T cells in patient-derived xenograft (PDX) models of prostate cancer. In vitro, LeY CAR T cells directly killed organoids derived from androgen receptor (AR)-positive or AR-null PDXs. In vivo, although LeY CAR T cells alone did not reduce tumor growth, a single prior dose of carboplatin reduced tumor burden. Carboplatin had a pro-inflammatory effect on the TME that facilitated early and durable CAR T cell infiltration, including an altered cancer-associated fibroblast phenotype, enhanced extracellular matrix degradation and re-oriented M1 macrophage differentiation. In a PDX less sensitive to carboplatin, CAR T cell infiltration was dampened; however, a reduction in tumor burden was still observed with increased T cell activation. These findings indicate that carboplatin improves the efficacy of CAR T cell treatment, with the extent of the response dependent on changes induced within the TME.


Subject(s)
Cancer-Associated Fibroblasts , Prostatic Neoplasms , Male , Animals , Humans , Carboplatin/pharmacology , Carboplatin/therapeutic use , Tumor Microenvironment , T-Lymphocytes , Prostatic Neoplasms/drug therapy , Disease Models, Animal
2.
Ann Oncol ; 32(11): 1366-1380, 2021 11.
Article in English | MEDLINE | ID: mdl-34375680

ABSTRACT

Chimeric antigen receptor (CAR)-T-cell therapy is a promising approach for the treatment of childhood cancers, particularly high-risk tumors that fail to respond to standard therapies. CAR-T cells have been highly successful in treating some types of hematological malignancies. However, CAR-T cells targeting solid cancers have had limited success so far for multiple reasons, including their poor long-term persistence and proliferation. Evidence is emerging to show that maintaining CAR-T cells in an early, less-differentiated state in vitro results in superior persistence, proliferation, and antitumor effects in vivo. Children are ideal candidates for receiving less-differentiated CAR-T cells, because their peripheral T-cell pool primarily comprises naïve cells that could readily be harvested in large numbers to generate early-phenotype CAR-T cells. Although several studies have reported different approaches to successfully generate early CAR-T cells, there are only a few clinical trials testing these in adult patients. No trials are currently testing early CAR-T cells in children. Here, we summarize the different strategies used to maintain CAR-T cells in an early phenotypic stage and present evidence suggesting that this approach may be particularly relevant to treating childhood cancers.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Phenotype , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
3.
J Neuroimmunol ; 327: 1-9, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30685070

ABSTRACT

Type 1 Diabetes (T1D) is an autoimmune disease in which insulin producing beta cells of the pancreas are selectively destroyed. Glial Fibrillary Acidic Protein (GFAP) expressed in peri-islet Schwann cells (pSCs) and in the ductal cells of the pancreas is one of the candidate autoantigens for T1D. Immune responses to GFAP expressing cell types precede the islet autoimmunity in Non-Obese Diabetic (NOD) mice. By removing MHC class I from GFAP expressing cell types, we tested the role of autoantigens presented by these cell types in the development of invasive insulitis. Our findings indicate that antigens expressed by pancreatic ductal cells are important in the development of invasive insulitis in NOD mice.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Islets of Langerhans/immunology , Animals , Islets of Langerhans/pathology , Mice , Mice, Inbred NOD , Mice, Knockout
4.
Sci Rep ; 8(1): 11158, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30042403

ABSTRACT

A prospective study explored the heterogeneous nature of metastatic melanoma using Multiplex immunohistochemistry (IHC) and flow cytometry (FACS). Multiplex IHC data quantitated immune subset number present intra-tumoral (IT) vs the tumor stroma, plus distance of immune subsets from the tumor margin (TM). In addition, mIHC showed a close association between the presence of IT CD8+ T cells and PDL1 expression in melanoma, which was more prevalent on macrophages than on melanoma cells. In contrast, FACS provided more detailed information regarding the T cell subset differentiation, their activation status and expression of immune checkpoint molecules. Interestingly, mIHC detected significantly higher Treg numbers than FACS and showed preferential CD4+ T cell distribution in the tumor stroma. Based on the mIHC and FACS data, we provide a model which defines metastatic melanoma immune context into four categories using the presence or absence of PDL1+ melanoma cells and/or macrophages, and their location within the tumor or on the periphery, combined with the presence or absence of IT CD8+ T cells. This model interprets melanoma immune context as a spectrum of tumor escape from immune control, and provides a snapshot upon which interpretation of checkpoint blockade inhibitor (CBI) therapy responses can be built.


Subject(s)
Immunohistochemistry/methods , Melanoma/immunology , Melanoma/pathology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Humans , Ipilimumab/immunology , Ipilimumab/therapeutic use , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating , Macrophages/metabolism , Melanoma/drug therapy , Metastasectomy , Middle Aged , Prospective Studies , Statistics, Nonparametric , T-Lymphocytes, Regulatory/immunology , Tumor Escape
5.
Proc Natl Acad Sci U S A ; 115(9): E2068-E2076, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440406

ABSTRACT

Chimeric antigen receptor T (CAR-T) cells are effective serial killers with a faster off-rate from dying tumor cells than CAR-T cells binding target cells through their T cell receptor (TCR). Here we explored the functional consequences of CAR-mediated signaling using a dual-specific CAR-T cell, where the same cell was triggered via TCR (tcrCTL) or CAR (carCTL). The carCTL immune synapse lacked distinct LFA-1 adhesion rings and was less reliant on LFA to form stable conjugates with target cells. carCTL receptors associated with the synapse were found to be disrupted and formed a convoluted multifocal pattern of Lck microclusters. Both proximal and distal receptor signaling pathways were induced more rapidly and subsequently decreased more rapidly in carCTL than in tcrCTL. The functional consequence of this rapid signaling in carCTL cells included faster lytic granule recruitment to the immune synapse, correlating with faster detachment of the CTL from the target cell. This study provides a mechanism for how CAR-T cells can debulk large tumor burden quickly and may contribute to further refinement of CAR design for enhancing the quality of signaling and programming of the T cell.


Subject(s)
Immunological Synapses/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD3 Complex , Cell Adhesion , Cell Death , Cell Line, Tumor , Computational Biology , Cytokines/metabolism , Dyneins/chemistry , Ligands , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Microtubules/metabolism , Signal Transduction
6.
Drugs Aging ; 33(2): 151-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26833349

ABSTRACT

BACKGROUND: Sleep complaints and the consumption of medications for sleep are common in older adults. Falls are also a significant concern for older adults and sedative use has been identified as a risk factor for falls. Sleep quality is a potential confounder in studies evaluating the relationship between sleep medication use and falls. However, very few studies have assessed the combined impact of sleep medication use and sleep quality on the risk of falls. OBJECTIVE: The objective of this study was to evaluate the association between sleep medication use, poor sleep quality, and falls in community-dwelling older adults. METHODS: This was a multicenter, 6-month prospective cohort study conducted in senior housings settings in central Virginia, USA. Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI) and a medication review was conducted. Data regarding falls were collected over 6 months by use of a diary. Logistic regression modeling was used to examine the effects of poor sleep quality, sleep medication use, and both, on the risk of falls. RESULTS: Among 113 independently living older adults (mean age ± standard deviation 81.1 ± 8.6), 46.9 % fell at least once during a 6-month period; 62.8 % (n = 71) had poor sleep quality, and 44.2 % (n = 50) used medications or treatments to aid sleep. Compared with participants reporting good sleep quality and no sleep medication use, those who reported poor sleep quality and sleep medication use had an increased risk of falls after adjusting for covariates (odds ratio 3.23, 95 % confidence interval 1.05-9.91). The group with good sleep quality and sleep medication use, as well as the group with poor sleep quality and no sleep medication use had no significantly greater risk for falls compared with the group with good sleep quality and no sleep medication use. CONCLUSION: A combined effect of sleep quality and sleep medication use on the risk of falls suggests that medication effectiveness may be an important factor to consider in understanding the risk of falls associated with sedative medications.


Subject(s)
Accidental Falls/prevention & control , Hypnotics and Sedatives , Sleep Wake Disorders , Accidental Falls/statistics & numerical data , Aged , Aged, 80 and over , Female , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Independent Living/statistics & numerical data , Logistic Models , Male , Odds Ratio , Prospective Studies , Risk Assessment , Risk Factors , Self Report , Sleep , Sleep Wake Disorders/complications , Sleep Wake Disorders/drug therapy , Socioeconomic Factors , United States
7.
Bioelectromagnetics ; 33(4): 298-308, 2012 May.
Article in English | MEDLINE | ID: mdl-21953117

ABSTRACT

Microalgal biotechnology could generate substantial amounts of biofuels with minimal environmental impact if the economics can be improved by increasing the rate of biomass production. Chlorella kessleri was grown in a small-scale raceway pond and in flask cultures with the entire volume, 1% (v/v) at any instant, periodically exposed to static magnetic fields to demonstrate increased biomass production and investigate physiological changes, respectively. The growth rate in flasks was maximal at a field strength of 10 mT, increasing from 0.39 ± 0.06 per day for the control to 0.88 ± 0.06 per day. In the raceway pond the 10 mT field increased the growth rate from 0.24 ± 0.03 to 0.45 ± 0.05 per day, final biomass from 0.88 ± 0.11 to 1.56 ± 0.18 g/L per day, and maximum biomass production from 0.11 ± 0.02 to 0.38 ± 0.04 g/L per day. Increased pigment, protein, Ca, and Zn content made the biomass produced with magnetic stimulation nutritionally superior. An increase in oxidative stress was measured indirectly as a decrease in antioxidant capacity from 26 ± 2 to 17 ± 1 µmol antioxidant/g biomass. Net photosynthetic capacity (NPC) and respiratory rate were increased by factors of 2.1 and 3.1, respectively. Loss of NPC enhancement after the removal of magnetic field fit a first-order model well (R(2) = 0.99) with a half-life of 3.3 days. Transmission electron microscopy showed enlarged chloroplasts and decreased thylakoid order with 10 mT treatment. By increasing daily biomass production about fourfold, 10 mT magnetic field exposure could make algal oil cost competitive with other biodiesel feedstocks.


Subject(s)
Chlorella/growth & development , Chlorella/metabolism , Magnetic Fields , Microalgae/growth & development , Microalgae/metabolism , Photosynthesis , Biomass , Chlorella/ultrastructure , Microalgae/ultrastructure
8.
Environ Sci Technol ; 45(20): 9061-8, 2011 10 15.
Article in English | MEDLINE | ID: mdl-21879741

ABSTRACT

Ultramafic mine tailings from the Diavik Diamond Mine, Canada and the Mount Keith Nickel Mine, Western Australia are valuable feedstocks for sequestering CO2 via mineral carbonation. In microcosm experiments, tailings were leached using various dilute acids to produce subsaline solutions at circumneutral pH that were inoculated with a phototrophic consortium that is able to induce carbonate precipitation. Geochemical modeling of the experimental solutions indicates that up to 2.5% and 16.7% of the annual emissions for Diavik and Mount Keith mines, respectively, could be sequestered as carbonate minerals and phototrophic biomass. CO2 sequestration rates are mainly limited by cation availability and the uptake of CO2. Abundant carbonate mineral precipitation occurred when heterotrophic oxidation of acetate acted as an alternative pathway for CO2 delivery. These experiments highlight the importance of heterotrophy in producing sufficient DIC concentrations while phototrophy causes alkalinization of waters and produces biomass (fatty acids = 7.6 wt.%), a potential feedstock for biofuel production. Tailings storage facilities could be redesigned to promote CO2 sequestration by directing leachate waters from tailings piles into specially designed ponds where carbonate precipitation would be mediated by both chemical and biological processes, thereby storing carbon in stable carbonate minerals and potentially valuable biomass.


Subject(s)
Heterotrophic Processes/physiology , Minerals/metabolism , Phototrophic Processes/physiology , Biodegradation, Environmental , Biomass , Carbon Dioxide/metabolism , Mining , Water Microbiology
9.
Gene Ther ; 17(9): 1105-16, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20428216

ABSTRACT

The anti-tumor efficacy of adoptively transferred T cells requires their in vivo persistence and memory polarization. It is unknown if human chimeric antigen receptor (CAR)-expressing T cells can also undergo memory polarization. We examined the functional status of CAR CD8(+) T cells, re-directed to Lewis Y antigen (LeY-T), throughout a period of ex vivo expansion. Immediately before culture CD8(+) T cells comprised a mixture of phenotypes including naive (CD45RA(+)/CCR7(+)/CD27(+)/CD28(+)/perforin-), central memory (CM, CD45RA(-)/CCR7(lo)/CD27(+)/CD28(+)/perforin(lo)), effector memory (EM, CD45RA(-)/CCR7(-)/CD27(+)/CD28(+)/perforin(mod)) and effector (Eff, CD45RA(+)/CCR7(-)/CD27(-)/CD28(-)/perforin(hi)) cells. After transduction and expansion culture of peripheral blood mononuclear cells from normal donors or multiple myeloma patients, CD8(+) LeY-T cells polarized to EM- and CM-like phenotype. CD8(+) LeY-T cells differed from starting CD8(+) CM and EM T cells in that CD27, but not CD28, was downregulated. In addition, CD8(+) LeY-T cells expressed high levels of perforin, similar to starting CD8(+) Eff. CD8(+) LeY-T cells also showed hallmarks of both memory and Eff function, underwent homeostatic proliferation in response to interleukin (IL)-15, and showed interferon (IFN)-γ production and cytotoxicity in response to Le-Y antigen on OVCAR-3 (human ovarian adenocarcinoma) cells. This study confirms CD8(+) LeY-T cells have a CM- and EM-like phenotype and heterogeneous function consistent with potential to persist in vivo after adoptive transfer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Immunologic Memory , Receptors, Antigen/genetics , CD28 Antigens/immunology , Cell Proliferation , Humans , Interferon-gamma/metabolism , Leukocyte Common Antigens/immunology , Phenotype , Receptors, Antigen/immunology , Receptors, Antigen/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
10.
Gene Ther ; 17(5): 678-86, 2010 May.
Article in English | MEDLINE | ID: mdl-20200563

ABSTRACT

We have evaluated the carbohydrate antigen Lewis(Y) (Le(Y)) as a potential target for T-cell immunotherapy of hematological neoplasias. Analysis of 81 primary bone marrow samples revealed moderate Le(Y) expression on plasma cells of myeloma patients and myeloblasts of patients with acute myeloid leukemia (AML) (52 and 46% of cases, respectively). We developed a retroviral vector construct encoding a chimeric T-cell receptor that recognizes the Le(Y) antigen in a major histocompatibility complex-independent manner and delivers co-stimulatory signals to achieve T-cell activation. We have shown efficient transduction of peripheral blood-derived T cells with this construct, resulting in antigen-restricted interferon-gamma secretion and cell lysis of Le(Y)-expressing tumor cells. In vivo activity of gene-modified T cells was demonstrated in the delayed growth of myeloma xenografts in NOD/SCID mice, which prolonged survival. Therefore, targeting Le(Y)-positive malignant cells with T cells expressing a chimeric receptor recognizing Le(Y) was effective both in vitro and in a myeloma mouse model. Consequently, we plan to use T cells manufactured under Good Manufacturing Practice conditions in a phase I immunotherapy study for patients with Le(Y)-positive myeloma or AML.


Subject(s)
Genetic Therapy/methods , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/therapy , Lewis Blood Group Antigens/immunology , Multiple Myeloma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/transplantation , Animals , Female , Genetic Vectors , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Antigen, T-Cell/immunology , Retroviridae/genetics , T-Lymphocytes/immunology , Transduction, Genetic
11.
Tissue Antigens ; 74(4): 277-89, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19775368

ABSTRACT

Adoptive cellular immunotherapy involving transfer of tumor-reactive T cells has shown some notable antitumor responses in a minority of cancer patients. In particular, transfer of tumor-infiltrating lymphocytes has resulted in long-term objective responses in patients with advanced melanoma. However, the inability to isolate sufficient numbers of tumor-specific T cells from most malignancies has restricted the broad utility of this approach. An emerging approach to circumvent this limitation involves the genetic modification of effector cells with T cell receptor (TCR) transgenes or chimeric single-chain variable fragment (scFv) receptors that can specifically redirect T cells to tumor. There has been much progress in the design of TCR and scFv receptors to enhance the antigen-specific activation of effector cells and their trafficking and persistence in vivo. Considerable effort has been directed toward improving the safety of this approach and reducing the immunogenicity of the receptor. This review discusses the latest developments in the field of adoptive immunotherapy using genetically modified immune cells that have been transduced with either TCR or scFv receptor transgenes and used in preclinical and clinical settings as anticancer agents.


Subject(s)
Genetic Engineering , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Humans , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology
12.
Comp Med ; 59(1): 27-36, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19295052

ABSTRACT

Murine noroviruses are a recently discovered group of viruses found within mouse research colonies in many animal facilities worldwide. In this study, we used 2 novel mouse norovirus (MNV) wildtype isolates to examine the kinetics of transmission and tissue distribution in breeding units of NOD.CB17-Prkdc(scid)/J and backcrossed NOD.CB17-Prkdc(scid)/J x NOD/ShiLtJ (N1) mice. Viral shedding in feces and dissemination to tissues of infected offspring mice were monitored by RT-PCR over a 6-wk period postpartum. Histologic sections of tissues from mice exposed to MNV were examined for lesions and their sera monitored for the presence of antibodies to MNV. Viruses shed in feces of parental and offspring mice were compared for sequence homology of the Orf2 gene. Studies showed that the wildtype viruses MNV5 and MNV6 behaved differently in terms of the kinetics of transmission and distribution to tissues of offspring mice. For MNV5, virus transmission from parents to offspring was not seen before 3 wk after birth, and neither isolate was transmitted between cages of infected and control mice. Susceptibility to infection was statistically different between the 2 mouse strains used in the study. Both immunodeficient NOD.CB17-Prkdc(scid)/J mice and NOD. CB17-Prkdc(scid)/J x NOD/ShiLtJ offspring capable of mounting an immune response shed virus in their feces throughout the 6-wk study period, but no gross or histologic lesions were present in infected tissues. Progeny viruses isolated from the feces of infected offspring showed numerous mutations in the Orf2 gene for MNV5 but not MNV6. These results confirm previous studies demonstrating that the biology of MNV in mice varies substantially with each virus isolate and mouse strain infected.


Subject(s)
Caliciviridae Infections/veterinary , Infectious Disease Transmission, Vertical/veterinary , Norovirus/physiology , Rodent Diseases/virology , Animals , Antibodies, Viral/analysis , Caliciviridae Infections/diagnosis , Caliciviridae Infections/transmission , Feces/virology , Female , Immunocompromised Host , Male , Mice , Mice, SCID , Norovirus/isolation & purification , Norovirus/pathogenicity , Pregnancy , RNA, Viral/analysis , Rodent Diseases/blood , Rodent Diseases/transmission , Serologic Tests/veterinary , Species Specificity , Time Factors , Virus Shedding/physiology
13.
Gene Ther ; 15(14): 1056-66, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18369322

ABSTRACT

There is considerable concern regarding the transforming potential of retroviral vectors currently used for gene therapy, with evidence that retroviral integration can lead to leukemia in recipients of gene-modified stem cells. However, it is not clear whether retroviral-mediated transduction of T cells can lead to malignancy. We transduced mouse T cells with a Moloney murine retroviral gene construct and transferred them into congenic mice, which were preconditioned to enhance the engraftment of transferred T cells. Recipients were then observed long-term for evidence of cancer. Transferred T cells persisted in mice throughout life at levels up to 17% with gene copy numbers up to 5.89 x 10(5) per million splenocytes. Mice receiving gene-modified T cells developed tumors at a similar rate as control mice that did not receive T cells, and tumors in both groups of mice were of a similar range of histologies. Hematological malignancies comprised approximately 60% of cancers, and the remaining cancers consisted largely of carcinomas. Importantly, the incidence of lymphomas was similar in both groups of mice, and no lymphomas were found to be of donor T-cell origin. This study indicates that the use of retroviral vectors to transduce T cells does not lead to malignant transformation.


Subject(s)
Adoptive Transfer , Genetic Therapy/adverse effects , Genetic Vectors/administration & dosage , Moloney murine leukemia virus/physiology , T-Lymphocytes/virology , Animals , Cell Transformation, Viral , Leukemia/virology , Lymphoma/virology , Mice , Mice, SCID , Moloney murine leukemia virus/genetics , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/transplantation , Time , Transduction, Genetic/methods , Transgenes
14.
Cancer Gene Ther ; 14(9): 773-80, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17599092

ABSTRACT

Dendritic cells (DC) perform an important role in the initiation of the immune response through the local secretion of inflammatory mediators within diseased tissue in response to Toll-like receptor (TLR) ligation. However, DC vaccine strategies fail to make use of this capability against cancer. To harness the TLR response capability of DC against cancer, we tested a series of recombinant genes for their ability to redirect DC function specifically against a tumor-associated antigen. Each gene encoded a cell surface chimeric protein made up of extracellular single-chain immunoglobulin anti-erbB2 linked to an intracellular TLR-signaling component composed of either myeloid differentiation factor 88, interleukin-1 receptor-associated kinase-1 (IRAK-1) or the cytoplasmic domain of TLR4. Each gene was expressed in the DC line, JAWS II, to a similar degree following retroviral transduction. However, only the chimera containing IRAK-1 was able to mediate interleukin-12 and tumor necrosis factor-alpha secretion. Since TLR engagement can also activate DC and enhance their ability to stimulate T cells, we ligated the chimeric anti-erbB2-IRAK-1 receptor and determined the effect on the stimulation of T cells. We found that JAWS II cells triggered through chimeric anti-erbB2-IRAK-1 displayed an enhanced ability to stimulate ovalbumin-specific OT-II CD4(+) T cells. This first description of the generation of tumor-reactive DC may lead to the development of new cell-based vaccines that can act at both the tumor site to induce danger and at the lymph node to stimulate a specific T-cell response.


Subject(s)
Dendritic Cells/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Toll-Like Receptors/metabolism , Animals , Cell Line, Tumor , Dendritic Cells/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Lymphocyte Activation , Mice
15.
Cancer Gene Ther ; 14(5): 499-508, 2007 May.
Article in English | MEDLINE | ID: mdl-17332777

ABSTRACT

Activation and expansion of T cells are important in disease resolution, but tumors do not usually satisfy these immune requirements. Therefore, we employed a novel strategy whereby dual-specific T cells were generated that could respond to both tumor and influenza virus, reasoning that immunization with influenza virus would activate and expand tumor-specific cells, and inhibit tumor growth. Dual-specific T cells were generated by gene modification of influenza virus-specific mouse T cells with a chimeric gene-encoding reactivity against the erbB2 tumor-associated antigen. Dual-specific T cells were demonstrated to respond against both tumor and influenza in vitro, and expanded in vitro in response to influenza to a much greater degree than in response to tumor cells. Following adoptive transfer and immunization of tumor-bearing mice with influenza virus, dual-specific T cells expanded greatly in numbers in the peritoneal cavity and spleen. This resulted in a significant increase in time of survival of mice. However, tumors were not eradicated, which may have been due to the observed poor penetration of tumor by T cells. This is the first demonstration that the potent immunogenic nature of an infectious agent can be utilized to directly impact on T-cell expansion and activity against tumor in vivo.


Subject(s)
Immunotherapy, Adoptive/methods , Mammary Neoplasms, Animal/therapy , Orthomyxoviridae/immunology , Receptor, ErbB-2/antagonists & inhibitors , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , Mice , Mice, Inbred Strains , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology
16.
Oncogene ; 26(16): 2340-52, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17016432

ABSTRACT

Progression of colorectal cancer (CRC) involves spatial and temporal occurrences of epithelial-mesenchymal transition (EMT), whereby tumour cells acquire a more invasive and metastatic phenotype. Subsequently, the disseminated mesenchymal tumour cells must undergo a reverse transition (mesenchymal-epithelial transition, MET) at the site of metastases, as most metastases recapitulate the pathology of their corresponding primary tumours. Importantly, initiation of tumour growth at the secondary site is the rate-limiting step in metastasis. However, investigation of this dynamic reversible EMT and MET that underpins CRC morphogenesis has been hindered by a lack of suitable in vitro models. To this end, we have established a unique in vitro model of CRC morphogenesis, which we term LIM1863-Mph (morphogenetic). LIM1863-Mph cells spontaneously undergo cyclic transitions between two-dimensional monolayer (migratory, mesenchymal) and three-dimensional sphere (carcinoid, epithelial) states. Using RNAi, we demonstrate that FZD7 is necessary for MET of the monolayer cells as loss of FZD7 results in the persistence of a mesenchymal state (increased SNAI2/decreased E-cadherin). Moreover, FZD7 is also required for migration of the LIM1863-Mph monolayer cells. During development, FZD7 orchestrates either migratory or epithelialization events depending on the context. Our findings strongly implicate similar functional diversity for FZD7 during CRC morphogenesis.


Subject(s)
Carcinoid Tumor/pathology , Colorectal Neoplasms/pathology , Frizzled Receptors/physiology , Receptors, G-Protein-Coupled/physiology , Carcinoid Tumor/ultrastructure , Cell Cycle , Cell Differentiation , Cell Division , Cell Line, Tumor , Colorectal Neoplasms/ultrastructure , Epithelial Cells/cytology , Frizzled Receptors/deficiency , Frizzled Receptors/genetics , Humans , Mesoderm/cytology , RNA Interference , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , beta Catenin/physiology
17.
Diabetes ; 54(5): 1603-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15855353

ABSTRACT

The diabetogenic major histocompatibility complex (MHC) (H2(g7)) of NOD mice comprises contributions from several class II loci collectively designated as Idd1. Introduction of the H2(gx) haplotype from the related but diabetes-resistant cataract Shionogi (CTS) strain demonstrated an additional MHC-linked locus designated Idd16. The NOD-related alloxan resistant (ALR)/Lt strain is also characterized by the H2(gx) haplotype, which does not differ from H2(g7) from the class I H2-K(d) gene distally through the class II and into the class III region. Polymorphisms distal to the heat shock protein 70 locus (Hspa1b) include a rare H2-D(dx) rather than the H2(g7) encoded D(b) allele. Two differential-length NOD.ALR-H2(gx) congenic stocks (D.R1 and D.R2), both containing H2-D(dx), significantly suppressed diabetogenesis. This protection was lost when ALR alleles between the class III region and H2-D were removed in a shorter interval congenic (D.R3). Because no differences were observed in the ALR-derived interval extending 0.41 mB proximal to H2-K in any of these congenic stocks, a component of what was originally designated "Idd16" was sited to an interval shorter than 7.33 mB, distinguishing D.R2 from D.R3. Evidence supporting the candidacy of the ALR/CTS-shared H2-D(dx) MHC class I variant present in both diabetes-resistant stocks, but not the susceptible stock, is discussed.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , Major Histocompatibility Complex , Aging , Animals , Biomarkers , Diabetes Mellitus, Type 1/epidemiology , Female , Incidence , Male , Mice , Mice, Inbred NOD , Sex Characteristics
18.
Cell Death Differ ; 11(9): 1028-37, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15131592

ABSTRACT

P-glycoprotein (P-gp) can induce multidrug resistance (MDR) through the ATP-dependent efflux of chemotherapeutic agents. We have previously shown that P-gp can inhibit nondrug apoptotic stimuli by suppressing the activation of caspases. To determine if this additional activity is functionally linked to ATP hydrolysis, we expressed wild-type and ATPase-mutant P-gp and showed that cells expressing mutant P-gp could not efflux chemotherapeutic drugs but remained relatively resistant to apoptosis. CEM lymphoma cells expressing mutant P-gp treated with vincristine showed a decrease in the fraction of cells with apoptotic morphology, cytochrome c release from the mitochondria and suppression of caspase activation, yet still accumulated in mitosis and showed a loss of clonogenic potential. The loss of clonogenicity in vincristine-treated cells expressing mutant P-gp was associated with accumulation of cells in mitosis and the presence of multinucleated cells consistent with mitotic catastrophe. The antiapoptotic effect of mutant P-gp was not affected by antibodies that inhibit the efflux function of the protein. These data are consistent with a dual activity model for P-gp-induced MDR involving both ATPase-dependent drug efflux and ATPase-independent inhibition of apoptosis. The structure-function analyses described herein provide novel insight into the mechanisms of action of P-gp in mediating MDR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenosine Triphosphate/metabolism , Caspases/metabolism , Adenosine Triphosphatases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Survival , Cytochromes c/metabolism , DNA Mutational Analysis , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Enzyme Activation , Green Fluorescent Proteins/metabolism , Humans , Hydrolysis , Hydroxamic Acids/pharmacology , Idarubicin/pharmacology , Lymphoma/drug therapy , Mitosis , Mutation , Retroviridae/genetics , Structure-Activity Relationship , Time Factors , Vincristine/pharmacology
19.
J Biol Chem ; 279(11): 10593-605, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14676205

ABSTRACT

Here we identified two novel proteins denoted EH domain protein 2 (EHD2) and EHD2-binding protein 1 (EHBP1) that link clathrin-mediated endocytosis to the actin cytoskeleton. EHD2 contains an N-terminal P-loop and a C-terminal EH domain that interacts with NPF repeats in EHBP1. Disruption of EHD2 or EHBP1 function by small interfering RNA-mediated gene silencing inhibits endocytosis of transferrin into EEA1-positive endosomes as well as GLUT4 endocytosis into cultured adipocytes. EHD2 localizes with cortical actin filaments, whereas EHBP1 contains a putative actin-binding calponin homology domain. High expression of EHD2 or EHBP1 in intact cells mediates extensive actin reorganization. Thus EHD2 appears to connect endocytosis to the actin cytoskeleton through interactions of its N-terminal domain with membranes and its C-terminal EH domain with the novel EHBP1 protein.


Subject(s)
Actins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cytoskeleton/metabolism , Endocytosis , 3T3-L1 Cells , Actins/chemistry , Adipocytes/metabolism , Amino Acid Sequence , Animals , Biological Transport , Blotting, Northern , Blotting, Western , COS Cells , Carrier Proteins/metabolism , Cell Membrane/metabolism , DNA, Complementary/metabolism , Endosomes/metabolism , Gene Silencing , Glucose/metabolism , Green Fluorescent Proteins , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Mice , Microscopy, Electron , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats , Rhodamines/chemistry , Time Factors , Tissue Distribution , Transfection , Transferrin/chemistry , Transferrin/metabolism
20.
Nature ; 420(6917): 821-4, 2002.
Article in English | MEDLINE | ID: mdl-12490950

ABSTRACT

Insulin stimulates glucose uptake in muscle and adipocytes by signalling the translocation of GLUT4 glucose transporters from intracellular membranes to the cell surface. The translocation of GLUT4 may involve signalling pathways that are both independent of and dependent on phosphatidylinositol-3-OH kinase (PI(3)K). This translocation also requires the actin cytoskeleton, and the rapid movement of GLUT4 along linear tracks may be mediated by molecular motors. Here we report that the unconventional myosin Myo1c is present in GLUT4-containing vesicles purified from 3T3-L1 adipocytes. Myo1c, which contains a motor domain, three IQ motifs and a carboxy-terminal cargo domain, is highly expressed in primary and cultured adipocytes. Insulin enhances the localization of Myo1c with GLUT4 in cortical tubulovesicular structures associated with actin filaments, and this colocalization is insensitive to wortmannin. Insulin-stimulated translocation of GLUT4 to the adipocyte plasma membrane is augmented by the expression of wild-type Myo1c and inhibited by a dominant-negative cargo domain of Myo1c. A decrease in the expression of endogenous Myo1c mediated by small interfering RNAs inhibits insulin-stimulated uptake of 2-deoxyglucose. Thus, myosin Myo1c functions in a PI(3)K-independent insulin signalling pathway that controls the movement of intracellular GLUT4-containing vesicles to the plasma membrane.


Subject(s)
Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Myosins/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/ultrastructure , Amino Acid Motifs , Animals , Biological Transport/drug effects , Cell Differentiation , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Glucose/metabolism , Glucose Transporter Type 4 , Mice , Myosin Type I , Myosins/chemistry , Myosins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...