251 results on '"Meruelo D"'
Search Results
52. Genetic control of radiation leukemia virus-induced tumorigenesis II. Influence of Srlv-1, a locus not linked to H-2.
- Author
-
Meruelo, D, Lieberman, M, Deak, B, and McDevitt, H O
- Abstract
A single locus, tentatively denoted Srlv-1 (susceptibility to radiation leukemia virus [RadLV]-1), confers dominant susceptibility to RadLV-induced leukemogenesis. Srlv-1 is not linked to H-2, and appears to be distinct from Fv-1 and Fv-2. Preliminary data suggest that Srlv-1 affected virus proliferation. A striking feature of this system is that Srlv-1 overrides the protection afforded by the H2D-associated dominant resistance to RadLV-induced neoplasia.
- Published
- 1977
- Full Text
- View/download PDF
53. Genetics of susceptibility for radiation-induced leukemia. Mapping of genes involved to chromosomes 1, 2, and 4, and implications for a viral etiology in the disease.
- Author
-
Meruelo, D, Offer, M, and Flieger, N
- Abstract
Susceptibility to radiation-induced leukemia in (A/J x B10)F2 mice is encoded for by genes in chromosomes 1, 2, and 4. The loci involved in chromosomes 1 and 4 are close to or similar to xenotropic virus inducibility locus on chromosome 1 and a locus-affecting expression of xenotropic MuLV envelope-related cell surface antigens. Radiation-induced leukemia-1 (Ril-1) on chromosome 2 plays an overriding influence in susceptibility to the disease. This locus might encode ecotropic viral-associated genetic information or might contain cellular sequences with oncogenic potential. These findings are of interest in view of the importance of recombinant viruses to leukemogenesis. Furthermore, it is intriguing that Ril-1 is located in a chromosomal site rich in thymus differentiation-specific loci. An explanation for tissue-specific activation of endogenous viruses is that activation of the virus in question is dependent on differentiation-specific steps.
- Published
- 1981
- Full Text
- View/download PDF
54. A role for elevated H-2 antigen expression in resistance to neoplasia caused by radiation-induced leukemia virus. Enhancement of effective tumor surveillance by killer lymphocytes.
- Author
-
Meruelo, D
- Abstract
Resistance to neoplasia caused by radiation-induced leukemia virus (RadLV) is mediated by gene(s) in the H-2D region of the major histocompatibility complex. The previous observation that rapid increases in cellular synthesis and cell-surface expression of H-2 antigens are detectable immediately after virus inoculation has suggested that altered expression of H-2 antigens may play a significant role in the mechanism(s) of host defense to virus infection. This concept is supported by the following observations. First, cell-mediated immunity against RadLV transformed or infected cells can be detected with ease when H-2-positive target cells are used in the cell-mediated lympholysis (CML) assay. (Although RadLV transformed cells obtained from overtly leukemic animals and maintained in tissue culture are H-2 negative, these cells can regain their H-2 phenotype by in vivo passage in normal animals. The H-2-negative cells are poor targets in a CML assay.) Second, resistant mice develop greater numbers of effectors when infected with RadLV than do susceptible mice. Third, injection of normal (uninfected) thymocytes into syngeneic recipients of resistant or susceptible H-2 type does not stimulate a CML response. However, injection of RadLV infected thymocytes from resistant mice produces a vigorous CMI response, and such thymocytes elicit the strongest response at a time when both H-2 and viral antigen expression is elevated. By contrast, injection of infected thymocytes from susceptible mice, which express viral antigens, but low levels of H-2 antigens, does not stimulate a CML reaction. These findings may explain the easier induction of leukemia found by many investigators when virus is inoculated into neonatal mice and the preferential thymus tropism of some oncogenic type-C RNA virus. Cells expressing very low levels of H-2, such as thymocytes, may serve as permissive targets for virus infection because they lack an important component (H-2 antigens) of the dual or altered recognition signal required to trigger a defensive host immune response.
- Published
- 1979
- Full Text
- View/download PDF
55. Increased synthesis and expression of H-2 antigens on thymocytes as a result of radiation leukemia virus infection: a possible mechanism for H-2 linked control of virus-induced neoplasia.
- Author
-
Meruelo, D, Nimelstein, S H, Jones, P P, Lieberman, M, and McDevitt, H O
- Abstract
Previous studies from this laboratory have mapped resistance and/or susceptibility to radiation-induced leukemia virus (RadLV)-induced neoplasia to the H-2D region. H-2 linked effects on virus replication can be detected subsequent to the initial virus infection, and clear-cut differences in numbers of virus infected thymus cells can be detected as early as 5 wk after RadLV inoculation. Rapid increases in cellular synthesis and cell surface expression of H-2 antigens are detectable immediately after virus inoculation. These changes have been studied by immunofluorescence, absorption, cell surface iodination followed by sodium dodecyl-sulfate-polyacrylamide gel electrophoresis, and two dimensional gel electrophoretic analysis of internally labeled lymphocyte proteins. Expression of H-2K molecules is significantly increased in cells of susceptible and resistant animals. However, significant increases in expression of H-2D antigens occurs only on thymus cells from resistant strains (H-2Dd). Transformed cells of resistant and susceptible H-2 haplotypes adapted to tissue culture lack detectable H-2 antigens as determined by serological absorption studies. It is argued that altered expression of H-2 antigens plays a very significant role in the mechanism of host defense to virus infection.
- Published
- 1978
- Full Text
- View/download PDF
56. Expression of a single major histocompatibility complex locus controls the immune complex locus controls the immune response to poly-L-(tyrosine, glutamic acid)-poly-DL-alanine--poly-L-lysine
- Author
-
Deak, BD, Meruelo, D, and McDevitt, HO
- Abstract
Genetic control of the immune response linked to the major histocompatibility (H-2) complex in the mouse has been described for synthetic polypeptide antigens and for low doses of native proteins. The phenomenon is well documented(1,2). Extensive screening of intra-H-2 crossover-derived recombinant strains has localized H-2-linked immune response (Ir) genes to the I-immune response region of the H-2 complex (3). For most antigens, Ir genes are autosomal, dominant, and they segregate as single loci. It is not known whether these crossover-defined loci respresent single genes with multiple alleles or clusters of tightly linked genes (4). In 1972, Stimpfling and Durham (5) postulated that two interacting loci within the H-2 complex were required for the response to the alloantigen, H-2.2 (6), and, in 1975, Dorf et. al. (7) observed a responder phenotype in a recombinant derived from two strains which were nonresponders to the synthetic linear terpolymer, L-glutamic acid, L-lysine, L-phenylaline (GLPhe). Analysis of additional recombinants and complementation tests with F(1) hybrids clearly demonstrated that genes in two intra-I-region loci controlled the immune response to GLPhe. Subsequently, requirement for genes mapping in two intra-I-region loci were reported for porcine LDH(B)(8), the alloantigen Thy-1.1 (9), and for the synthetic terpolymers L-glutamic acid, L-lysine, L-tyrosine and L-glutamic acid, L-lysine, L- leucine (6,10). Demonstration that responses to both synthetic polypeptide and native protein antigens can be controlled by genes in two distinct I-region loci prompted speculation that the phenotypic expression of two I-region genes is a general phenomenon which may provide the key for understanding the mechanism of Ir gene function and cellular collaboration in the immune response. Benacerraf and Dorf (10) have shown that Ir gene complementation is often more effective in the cis than in the trans configuration. This concept is further supported by the data reported for GLPhe (10-12) which indicate that both of the complementing genes must be expressed in each of the cell types participating in the interaction. Failure to detect complementation for the majority of antigens under H-2-linked Ir-gene control might be attributed to the limited number of available intra-I- region recombinant strains.
- Published
- 1978
- Full Text
- View/download PDF
57. Murine leukemia virus sequences are encoded in the murine major histocompatibility complex.
- Author
-
Meruelo, D, Kornreich, R, Rossomando, A, Pampeno, C, Mellor, A L, Weiss, E H, Flavell, R A, and Pellicer, A
- Abstract
The studies reported here localize murine leukemia viral sequences to the TL region of the major histocompatibility complex, H-2. We examined a battery of 38 cosmids, isolated from two large genomic libraries constructed from C57BL/10 spleen DNA, that define 25 class I gene sequences. The viral probes used hybridized with only four cosmids, containing overlapping mouse sequences, that define four class I gene-related sequences in a region of 90 kilobases of DNA. The data show that two distinct viral envelope sequences are contained in the cluster. One of these sequences is situated with its 3' end next to the 3' end of a class I sequence. The other sequence, which does not contain the entire viral envelope, is proximal to the 3' end of a different class I sequence. Hybridization of the viral probes with the H-2 cosmid clones does not appear to be due to homology between viral and H-2 sequences. Rather, the viral sequences detected appear to be linked to or inserted amid class I genes. These findings may be significant in understanding molecular mechanisms involved in the generation of H-2 class I gene diversity.
- Published
- 1984
- Full Text
- View/download PDF
58. Studies of the mechanisms of action of the antiretroviral agents hypericin and pseudohypericin.
- Author
-
Lavie, G, Valentine, F, Levin, B, Mazur, Y, Gallo, G, Lavie, D, Weiner, D, and Meruelo, D
- Abstract
Administration of the aromatic polycyclic dione compounds hypericin or pseudohypericin to experimental animals provides protection from disease induced by retroviruses that give rise to acute, as well as slowly progressive, diseases. For example, survival from Friend virus-induced leukemia is significantly prolonged by both compounds, with hypericin showing the greater potency. Viremia induced by LP-BM5 murine immunodeficiency virus is markedly suppressed after infrequent dosage of either substance. These compounds affect the retroviral infection and replication cycle at least at two different points: (i) Assembly or processing of intact virions from infected cells was shown to be affected by hypericin. Electron microscopy of hypericin-treated, virus-producing cells revealed the production of particles containing immature or abnormally assembled cores, suggesting the compounds may interfere with processing of gag-encoded precursor polyproteins. The released virions contain no detectable activity of reverse transcriptase. (ii) Hypericin and pseudohypericin also directly inactivate mature and properly assembled retroviruses as determined by assays for reverse transcriptase and infectivity. Accumulating data from our laboratories suggest that these compounds inhibit retroviruses by unconventional mechanisms and that the potential therapeutic value of hypericin and pseudohypericin should be explored in diseases such as AIDS.
- Published
- 1989
- Full Text
- View/download PDF
59. A new lymphocyte cell surface antigen, Ly-22.2, controlled by a locus on chromosome 4 and a second unlinked locus.
- Author
-
Meruelo, D, primary, Offer, M, additional, and Flieger, N, additional
- Published
- 1983
- Full Text
- View/download PDF
60. Definition of a new T lymphocyte cell surface antigen, Ly 11.2.
- Author
-
Meruelo, D, primary, Paolino, A, additional, Flieger, N, additional, and Offer, M, additional
- Published
- 1980
- Full Text
- View/download PDF
61. Functional properties of Ly 11.2 lymphocytes: a role for these cells in leukemia?
- Author
-
Meruelo, D, primary, Paolino, A, additional, Flieger, N, additional, Dworkin, J, additional, Offer, M, additional, Hirayama, N, additional, and Ovary, Z, additional
- Published
- 1980
- Full Text
- View/download PDF
62. RADIATION LEUKEMIA VIRUS AND ITS EFFECT ON H-2 GENE EXPRESSION
- Author
-
Brown, G. D., primary and Meruelo, D., additional
- Published
- 1989
- Full Text
- View/download PDF
63. A TCR Cell Recognizing a Novel TL-encoded Gene Product
- Author
-
Houlden, B.A., primary, Matis, L.A., additional, Cron, R.Q., additional, Widacki, S.M., additional, Brown, G.D., additional, Pampeno, C., additional, Meruelo, D., additional, and Bluestone, J.A., additional
- Published
- 1989
- Full Text
- View/download PDF
64. Hypericin as an activator of infectious viruses in blood components
- Author
-
Lavie, G., Mazur, Y., Lavie, D., Prince, A.M., Pascual, D., Liebes, L., Levin, B., and Meruelo, D.
- Subjects
Hypericin -- Health aspects ,Retroviruses -- Drug therapy ,HIV infection -- Drug therapy - Abstract
Lavie, G.; Mazur, Y.; Lavie, D.; Prince, A.M.; Pascual, D.; Liebes, L.; Levin, B.; Meruelo, D. "Hypericin as an Inactivator of Infectious Viruses in Blood Components." Transfusion, May 1995;35(5):392-400. According [...]
- Published
- 1995
65. Sindbis viral vector induced apoptosis requires translational inhibition and signaling through Mcl-1 and Bak
- Author
-
Meruelo Daniel and Venticinque Lisa
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Sindbis viral vectors are able to efficiently target and kill tumor cells in vivo, as shown using pancreatic and ovarian cancer models. Infection results in apoptosis both in vitro and in vivo. Sindbis vector uptake is mediated by the LAMR, which is upregulated on a number of different tumor types, thus conferring specificity of the vector to a wide range of cancers. In this study we elucidate the mechanism of apoptosis in two tumor cell lines, MOSEC, derived from the ovarian epithelium and Pan02, derived from a pancreatic adenocarcinoma. A comprehensive understanding of the mechanism of apoptosis would facilitate the design of more effective vectors for cancer therapy. Results The initial phase of Sindbis vector induced apoptosis in MOSEC and Pan02 models reconfirms that viral infection is sensed by PKR due to double-stranded RNA intermediates associated with genomic replication. PKR activation results in translation inhibition through eIF2α phosphorylation and initiation of the stress response. Our studies indicate that the roles of two proteins, Mcl-1 and JNK, intimately link Sindbis induced translational arrest and cellular stress. Translational arrest inhibits the synthesis of anti-apoptotic Bcl-2 protein, Mcl-1. JNK activation triggers the release of Bad from 14-3-3, which ultimately results in apoptosis. These signals from translational arrest and cellular stress are propagated to the mitochondria where Bad and Bik bind to Bcl-xl and Mcl-1 respectively. Formation of these heterodimers displaces Bak, which results in caspase 9 cleavage and signaling through the mitochondrial pathway of apoptosis. Conclusion The host cell response to Sindbis is triggered through PKR activation. Our studies demonstrate that PKR activation and subsequent translational arrest is linked to both cellular stress and apoptosis. We have also found the linkage point between translational arrest and apoptosis to be Mcl-1, a protein whose constant translation is required for inhibition of apoptosis. With this information vectors can be designed, which express or repress proteins implicated in this study, to enhance their therapeutic potential.
- Published
- 2010
- Full Text
- View/download PDF
66. In vivo stimulation of H-2D d expression following RadLV infection of thymocytes: increased transcription and DNA-binding activity to sequences 5' of the D d gene
- Author
-
Brown, G.D., Nobunaga, T., Morris, D.R., and Meruelo, D.
- Published
- 1991
- Full Text
- View/download PDF
67. ChemInform Abstract: The Chemical and Biological Properties of Hypericin - A Compound with a Broad Spectrum of Biological Activities.
- Author
-
LAVIE, G., MAZUR, Y., LAVIE, D., and MERUELO, D.
- Published
- 1995
- Full Text
- View/download PDF
68. Extraribosomal Functions Associated with the C Terminus of the 37/67 kDa Laminin Receptor are Required for Maintaining Cell Viability
- Author
-
Meruelo, D
- Published
- 2011
69. Structure-Guided Identification of a Laminin Binding Site on the Laminin Receptor Precursor
- Author
-
Meruelo, D
- Published
- 2011
- Full Text
- View/download PDF
70. Crystal Structure of the Human Laminin Receptor Precursor
- Author
-
Meruelo, D
- Published
- 2008
- Full Text
- View/download PDF
71. Role for elevated H-2 antigen expression in resistance to neoplasia caused by radiation-induced leukemia virus. Enhancement of effective tumor surveillance by killer lymphocytes
- Author
-
Meruelo, D
- Published
- 1979
- Full Text
- View/download PDF
72. Sindbis Virus Vaccine Platform: A Promising Oncolytic Virus-Mediated Approach for Ovarian Cancer Treatment.
- Author
-
Pampeno C, Opp S, Hurtado A, and Meruelo D
- Subjects
- Humans, Female, Sindbis Virus, Neoplasm Recurrence, Local therapy, Immunotherapy methods, Oncolytic Viruses, Oncolytic Virotherapy methods, Ovarian Neoplasms pathology, Vaccines
- Abstract
This review article provides a comprehensive overview of a novel Sindbis virus vaccine platform as potential immunotherapy for ovarian cancer patients. Ovarian cancer is the most lethal of all gynecological malignancies. The majority of high-grade serous ovarian cancer (HGSOC) patients are diagnosed with advanced disease. Current treatment options are very aggressive and limited, resulting in tumor recurrences and 50-60% patient mortality within 5 years. The unique properties of armed oncolytic Sindbis virus vectors (SV) in vivo have garnered significant interest in recent years to potently target and treat ovarian cancer. We discuss the molecular biology of Sindbis virus, its mechanisms of action against ovarian cancer cells, preclinical in vivo studies, and future perspectives. The potential of Sindbis virus-based therapies for ovarian cancer treatment holds great promise and warrants further investigation. Investigations using other oncolytic viruses in preclinical studies and clinical trials are also presented.
- Published
- 2024
- Full Text
- View/download PDF
73. Channeling the Natural Properties of Sindbis Alphavirus for Targeted Tumor Therapy.
- Author
-
Pampeno C, Hurtado A, Opp S, and Meruelo D
- Subjects
- Humans, Genetic Vectors genetics, Genetic Therapy methods, Alphavirus genetics, Neoplasms therapy, Encephalitis Virus, Venezuelan Equine
- Abstract
Sindbis alphavirus vectors offer a promising platform for cancer therapy, serving as valuable models for alphavirus-based treatment. This review emphasizes key studies that support the targeted delivery of Sindbis vectors to tumor cells, highlighting their effectiveness in expressing tumor-associated antigens and immunomodulating proteins. Among the various alphavirus vectors developed for cancer therapy, Sindbis-vector-based imaging studies have been particularly extensive. Imaging modalities that enable the in vivo localization of Sindbis vectors within lymph nodes and tumors are discussed. The correlation between laminin receptor expression, tumorigenesis, and Sindbis virus infection is examined. Additionally, we present alternative entry receptors for Sindbis and related alphaviruses, such as Semliki Forest virus and Venezuelan equine encephalitis virus. The review also discusses cancer treatments that are based on the alphavirus vector expression of anti-tumor agents, including tumor-associated antigens, cytokines, checkpoint inhibitors, and costimulatory immune molecules.
- Published
- 2023
- Full Text
- View/download PDF
74. Potent and Targeted Sindbis Virus Platform for Immunotherapy of Ovarian Cancer.
- Author
-
Opp S, Hurtado A, Pampeno C, Lin Z, and Meruelo D
- Subjects
- Humans, Female, Animals, Mice, Interleukin-12, Antibodies, Immunotherapy, Tumor Microenvironment, Sindbis Virus physiology, Ovarian Neoplasms metabolism
- Abstract
Our laboratory has been developing a Sindbis viral (SV) vector platform for treatments of ovarian and other types of cancers. In this study we show that SV.IL-12 combined with an agonistic OX40 antibody can eliminate ovarian cancer in a Mouse Ovarian Surface Epithelial Cell Line (MOSEC) model and further prevent tumors in mice rechallenged with tumor cells after approximately 5 months. Treatment efficacy is shown to be dependent upon T-cells that are transcriptionally and metabolically reprogramed. An influx of immune cells to the tumor microenvironment occurs. Combination of sequences encoding both IL-12 and anti-OX40 into a single SV vector, SV.IgGOX40.IL-12, facilitates the local delivery of immunoregulatory agents to tumors enhancing the anti-tumor response. We promote SV.IgGOX40.IL-12 as a safe and effective therapy for multiple types of cancer.
- Published
- 2022
- Full Text
- View/download PDF
75. Combination of a Sindbis-SARS-CoV-2 Spike Vaccine and αOX40 Antibody Elicits Protective Immunity Against SARS-CoV-2 Induced Disease and Potentiates Long-Term SARS-CoV-2-Specific Humoral and T-Cell Immunity.
- Author
-
Scaglione A, Opp S, Hurtado A, Lin Z, Pampeno C, Noval MG, Thannickal SA, Stapleford KA, and Meruelo D
- Subjects
- Angiotensin-Converting Enzyme 2 metabolism, Animals, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, COVID-19 immunology, Cricetinae, Female, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Sindbis Virus genetics, T-Lymphocytes immunology, Vaccination, Antigens, Differentiation immunology, COVID-19 prevention & control, COVID-19 Vaccines immunology, SARS-CoV-2 immunology, Sindbis Virus immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 is a major global public threat. Currently, a worldwide effort has been mounted to generate billions of effective SARS-CoV-2 vaccine doses to immunize the world's population at record speeds. However, there is still a demand for alternative effective vaccines that rapidly confer long-term protection and rely upon cost-effective, easily scaled-up manufacturing. Here, we present a Sindbis alphavirus vector (SV), transiently expressing the SARS-CoV-2 spike protein (SV.Spike), combined with the OX40 immunostimulatory antibody (αOX40) as a novel, highly effective vaccine approach. We show that SV.Spike plus αOX40 elicits long-lasting neutralizing antibodies and a vigorous T-cell response in mice. Protein binding, immunohistochemical, and cellular infection assays all show that vaccinated mice sera inhibits spike functions. Immunophenotyping, RNA Seq transcriptome profiles, and metabolic analysis indicate a reprogramming of T cells in vaccinated mice. Activated T cells were found to mobilize to lung tissue. Most importantly, SV.Spike plus αOX40 provided robust immune protection against infection with authentic coronavirus in transgenic mice expressing the human ACE2 receptor (hACE2-Tg). Finally, our immunization strategy induced strong effector memory response, potentiating protective immunity against re-exposure to SARS-CoV-2 spike protein. Our results show the potential of a new Sindbis virus-based vaccine platform to counteract waning immune response, which can be used as a new candidate to combat SARS-CoV-2. Given the T-cell responses elicited, our vaccine is likely to be effective against variants that are proving challenging, as well as serve as a platform to develop a broader spectrum pancoronavirus vaccine. Similarly, the vaccine approach is likely to be applicable to other pathogens., Competing Interests: All authors are employed by NYU Langone School of Medicine and have no employment relationship or consultancy agreement with Cynvec a biotechnology company that support some studies under a Research and Licensing agreement with NYU. AS, SO, AH, ZL, CP, and DM are inventors on one or several issued patents and/or patent applications held by NYU that cover Sindbis treatment of neoplasia and COVID19. As part of the Research and Licensing agreement authors who are inventors on patents are entitled to a portion of NYU Langone’s royalties received, should Sindbis vectors be approved by the FDA for the therapeutic or vaccination use., (Copyright © 2021 Scaglione, Opp, Hurtado, Lin, Pampeno, Noval, Thannickal, Stapleford and Meruelo.)
- Published
- 2021
- Full Text
- View/download PDF
76. Sindbis Virus with Anti-OX40 Overcomes the Immunosuppressive Tumor Microenvironment of Low-Immunogenic Tumors.
- Author
-
Scherwitzl I, Opp S, Hurtado AM, Pampeno C, Loomis C, Kannan K, Yu M, and Meruelo D
- Abstract
Despite remarkable responses to cancer immunotherapy in a subset of patients, many patients remain resistant to therapies. It is now clear that elevated levels of tumor-infiltrating T cells as well as a systemic anti-tumor immune response are requirements for successful immunotherapies. However, the tumor microenvironment imposes an additional resistance mechanism to immunotherapy. We have developed a practical and improved strategy for cancer immunotherapy using an oncolytic virus and anti-OX40. This strategy takes advantage of a preexisting T cell immune repertoire in vivo , removing the need to know about present tumor antigens. We have shown in this study that the replication-deficient oncolytic Sindbis virus vector expressing interleukin-12 (IL-12) (SV.IL12) activates immune-mediated tumor killing by inducing OX40 expression on CD4 T cells, allowing the full potential of the agonistic anti-OX40 antibody. The combination of SV.IL12 with anti-OX40 markedly changes the transcriptome signature and metabolic program of T cells, driving the development of highly activated terminally differentiated effector T cells. These metabolically reprogrammed T cells demonstrate enhanced tumor infiltration capacity as well as anti-tumor activity capable of overcoming the repressive tumor microenvironment. Our findings identify SV.IL12 in combination with anti-OX40 to be a novel and potent therapeutic strategy that can cure multiple types of low-immunogenic solid tumors., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
77. Molecular and metabolic pathways mediating curative treatment of a non-Hodgkin B cell lymphoma by Sindbis viral vectors and anti-4-1BB monoclonal antibody.
- Author
-
Yu M, Scherwitzl I, Opp S, Tsirigos A, and Meruelo D
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Cell Line, Tumor, Combined Modality Therapy, Cytokines genetics, Gene Expression Regulation, Neoplastic, Humans, Interferon-gamma metabolism, Lymphocytes, Tumor-Infiltrating drug effects, Lymphoma, Non-Hodgkin genetics, Lymphoma, Non-Hodgkin immunology, Mice, Mice, Inbred BALB C, Neoplasm Recurrence, Local, Oncolytic Virotherapy, Signal Transduction drug effects, Sindbis Virus genetics, Xenograft Model Antitumor Assays, 4-1BB Ligand agonists, Antibodies, Monoclonal administration & dosage, Gene Expression Profiling methods, Lymphoma, Non-Hodgkin therapy, Sindbis Virus physiology
- Abstract
Background: Limitations to current therapies for treating non-Hodgkin B cell lymphoma include relapse, toxicity and high cost. Thus, there remains a need for novel therapies. Oncolytic viral (OV) therapy has become a promising cancer immunotherapy because of its potential effectiveness, specificity and long-lasting immunity. We describe and characterize a novel cancer immunotherapy combining Sindbis virus (SV) vectors and the agonistic monoclonal antibody (mAb) to the T cell costimulatory receptor, 4-1BB (CD137)., Methods: A20 lymphoma was transfected with luciferase and tumor cells were inoculated to BALB/c mice. Tumor growth was monitored by IVIS imaging. Tumor bearing mice were treated with Sindbis virus, α4-1BB Ab or SV plus α4-1BB Ab. On day 7 after treatment, splenocytes were harvested and surface markers, cytokines, and transcription factors were measured by flow cytometry or Elispot. Splenic T cells were isolated and RNA transcriptome analysis was performed. Tumor cured mice were rechallenged with tumor for testing immunological memory., Results: SV vectors in combination with α4-1BB monoclonal antibody (mAb) completely eradicated a B-cell lymphoma in a preclinical mouse model, a result that could not be achieved with either treatment alone. Tumor elimination involves a synergistic effect of the combination that significantly boosts T cell cytotoxicity, IFNγ production, T cell proliferation, migration, and glycolysis. In addition, all mice that survived after treatment developed long lasting antitumor immunity, as shown by the rejection of A20 tumor rechallenge. We identified the molecular pathways, including upregulated cytokines, chemokines and metabolic pathways in T cells that are triggered by the combined therapy and help to achieve a highly effective anti-tumor response., Conclusions: Our study provides a novel, alternative method for B cell lymphoma treatment and describes a rationale to help translate SV vectors plus agonistic mAb into clinical applications.
- Published
- 2019
- Full Text
- View/download PDF
78. Systemically Administered Sindbis Virus in Combination with Immune Checkpoint Blockade Induces Curative Anti-tumor Immunity.
- Author
-
Scherwitzl I, Hurtado A, Pierce CM, Vogt S, Pampeno C, and Meruelo D
- Abstract
Oncolytic viruses represent a promising form of cancer immunotherapy. We investigated the potential of Sindbis virus (SV) for the treatment of solid tumors expressing the human cancer testis antigen NYESO-1. NYESO-1 is an immunogenic antigen frequently expressed in numerous cancers, such as ovarian cancer. We show that SV expressing the tumor-associated antigen NYESO-1 (SV-NYESO1) acts as an immunostimulatory agent, inducing systemic and rapid lymphocyte activation, leading to a pro-inflammatory environment. SV-NYESO1 treatment combined with anti-programmed death 1 (anti-PD-1) markedly augmented the anti-tumor immunity in mice over the course of treatment, resulting in an avid systemic and intratumoral immune response. This response involved reduced presence of granulocytic myeloid-derived suppressor cells in tumors and an increase in the activation of splenic and tumor-infiltrating T cells. Combined therapy also induced enhanced cytotoxic activity of T cells against NYESO-1-expressing tumors. These results were in line with an observed inverse correlation between T cell activation and tumor growth. Finally, we show that combined therapy resulted in complete clearance of NYESO-1-expressing tumors in vivo and led to long-term protection against recurrences. These findings provide a rationale for clinical studies of SV-NYESO1 combined with immune checkpoint blockade anti-PD-1 to be used in the treatment of NYESO-1-expressing tumors.
- Published
- 2018
- Full Text
- View/download PDF
79. Looking into laminin receptor: critical discussion regarding the non-integrin 37/67-kDa laminin receptor/RPSA protein.
- Author
-
DiGiacomo V and Meruelo D
- Subjects
- Humans, Laminin genetics, Receptors, Laminin genetics, Ribosomal Proteins genetics, Gene Expression Regulation physiology, Laminin metabolism, Receptors, Laminin metabolism, Ribosomal Proteins metabolism
- Abstract
The 37/67-kDa laminin receptor (LAMR/RPSA) was originally identified as a 67-kDa binding protein for laminin, an extracellular matrix glycoprotein that provides cellular adhesion to the basement membrane. LAMR has evolutionary origins, however, as a 37-kDa RPS2 family ribosomal component. Expressed in all domains of life, RPS2 proteins have been shown to have remarkably diverse physiological roles that vary across species. Contributing to laminin binding, ribosome biogenesis, cytoskeletal organization, and nuclear functions, this protein governs critical cellular processes including growth, survival, migration, protein synthesis, development, and differentiation. Unsurprisingly given its purview, LAMR has been associated with metastatic cancer, neurodegenerative disease and developmental abnormalities. Functioning in a receptor capacity, this protein also confers susceptibility to bacterial and viral infection. LAMR is clearly a molecule of consequence in human disease, directly mediating pathological events that make it a prime target for therapeutic interventions. Despite decades of research, there are still a large number of open questions regarding the cellular biology of LAMR, the nature of its ability to bind laminin, the function of its intrinsically disordered C-terminal region and its conversion from 37 to 67 kDa. This review attempts to convey an in-depth description of the complexity surrounding this multifaceted protein across functional, structural and pathological aspects., (© 2015 Cambridge Philosophical Society.)
- Published
- 2016
- Full Text
- View/download PDF
80. The Transition of the 37-Kda Laminin Receptor (Rpsa) to Higher Molecular Weight Species: Sumoylation or Artifact?
- Author
-
Digiacomo V, Gando IA, Venticinque L, Hurtado A, and Meruelo D
- Subjects
- Animals, Gene Knockdown Techniques, HeLa Cells, Humans, Immunoprecipitation, Mice, Molecular Weight, NIH 3T3 Cells, Receptors, Laminin genetics, Ribosomal Proteins genetics, Sumoylation, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Receptors, Laminin chemistry, Receptors, Laminin metabolism, Ribosomal Proteins chemistry, Ribosomal Proteins metabolism
- Abstract
The 37-kDa laminin receptor (37LRP or RPSA) is a remarkable, multifaceted protein that functions in processes ranging from matrix adhesion to ribosome biogenesis. Its ability to engage extracellular laminin is further thought to contribute to cellular migration and invasion. Most commonly associated with metastatic cancer, RPSA is also increasingly found to be important in other pathologies, including microbial infection, neurodegenerative disease and developmental malformations. Importantly, it is thought to have higher molecular weight forms, including a 67-kDa species (67LR), the expression of which is linked to strong laminin binding and metastatic behavior. The composition of these larger forms has remained elusive and controversial. Homo- and heterodimerization have been proposed as events capable of building the larger species from the monomeric 37-kDa precursor, but solid evidence is lacking. Here, we present data suggesting that higher molecular weight species require SUMOylation to form. We also comment on the difficulty of isolating larger RPSA species for unambiguous identification and demonstrate that cell lines stably expressing tagged RPSA for long periods of time fail to produce tagged higher molecular weight RPSA. It is possible that higher molecular weight species like 67LR are not derived from RPSA.
- Published
- 2015
- Full Text
- View/download PDF
81. TBLR1 as an androgen receptor (AR) coactivator selectively activates AR target genes to inhibit prostate cancer growth.
- Author
-
Daniels G, Li Y, Gellert LL, Zhou A, Melamed J, Wu X, Zhang X, Zhang D, Meruelo D, Logan SK, Basch R, and Lee P
- Subjects
- Adenocarcinoma genetics, Animals, Cell Growth Processes genetics, Cell Line, Tumor, Chromatin Immunoprecipitation, Humans, Male, Mice, Nude, Nuclear Proteins genetics, Prostatic Neoplasms genetics, RNA, Neoplasm chemistry, RNA, Neoplasm genetics, Receptors, Androgen genetics, Receptors, Cytoplasmic and Nuclear genetics, Repressor Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Tissue Array Analysis, Transcription, Genetic, Adenocarcinoma metabolism, Nuclear Proteins metabolism, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Repressor Proteins metabolism
- Abstract
Androgen receptor (AR), a steroid hormone receptor, is critical for prostate cancer growth. However, activation of AR by androgens can also lead to growth suppression and differentiation. Transcriptional cofactors play an important role in this switch between proliferative and anti-proliferative AR target gene programs. Transducin β-like-related protein 1 (TBLR1), a core component of the nuclear receptor corepressor complex, shows both corepressor and coactivator activities on nuclear receptors, but little is known about its effects on AR and prostate cancer. We characterized TBLR1 as a coactivator of AR in prostate cancer cells and determined that the activation is dependent on both phosphorylation and 19S proteosome. We showed that TBLR1 physically interacts with AR and directly occupies the androgen-response elements of the affected AR target genes in an androgen-dependent manner. TBLR1 is primarily localized in the nucleus in benign prostate cells and nuclear expression is significantly reduced in prostate cancer cells in culture. Similarly, in human tumor samples, the expression of TBLR1 in the nucleus is significantly reduced in the malignant glands compared with the surrounding benign prostatic glands (P<0.005). Stable ectopic expression of nuclear TBLR1 leads to androgen-dependent growth suppression of prostate cancer cells in vitro and in vivo by selective activation of androgen-regulated genes associated with differentiation (e.g. KRT18) and growth suppression (e.g. NKX3-1), but not cell proliferation of the prostate cancer. Understanding the molecular switches involved in the transition from AR-dependent growth promotion to AR-dependent growth suppression will lead to more successful treatments for prostate cancer.
- Published
- 2014
- Full Text
- View/download PDF
82. Interaction of human laminin receptor with Sup35, the [PSI⁺] prion-forming protein from S. cerevisiae: a yeast model for studies of LamR interactions with amyloidogenic proteins.
- Author
-
Pampeno C, Derkatch IL, and Meruelo D
- Subjects
- Centrifugation, Fluorescent Antibody Technique, Green Fluorescent Proteins metabolism, Humans, Immunoprecipitation, Protein Binding, Recombinant Fusion Proteins metabolism, Amyloidogenic Proteins metabolism, Models, Biological, Peptide Termination Factors metabolism, Receptors, Laminin metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism
- Abstract
The laminin receptor (LamR) is a cell surface receptor for extracellular matrix laminin, whereas the same protein within the cell interacts with ribosomes, nuclear proteins and cytoskeletal fibers. LamR has been shown to be a receptor for several bacteria and viruses. Furthermore, LamR interacts with both cellular and infectious forms of the prion protein, PrP(C) and PrP(Sc). Indeed, LamR is a receptor for PrP(C). Whether LamR interacts with PrP(Sc) exclusively in a capacity of the PrP receptor, or LamR specifically recognizes prion determinants of PrP(Sc), is unclear. In order to explore whether LamR has a propensity to interact with prions and amyloids, we examined LamR interaction with the yeast prion-forming protein, Sup35. Sup35 is a translation termination factor with no homology or functional relationship to PrP. Plasmids expressing LamR or LamR fused with the green fluorescent protein (GFP) were transformed into yeast strain variants differing by the presence or absence of the prion conformation of Sup35, respectively [PSI⁺] and [psi⁻]. Analyses by immunoprecipitation, centrifugal fractionation and fluorescent microscopy reveal interaction between LamR and Sup35 in [PSI⁺] strains. The presence of [PSI⁺] promotes LamR co-precipitation with Sup35 as well as LamR aggregation. In [PSI⁺] cells, LamR tagged with GFP or mCherry forms bright fluorescent aggregates that co-localize with visible [PSI⁺] foci. The yeast prion model will facilitate studying the interaction of LamR with amyloidogenic prions in a safe and easily manipulated system that may lead to a better understanding and treatment of amyloid diseases.
- Published
- 2014
- Full Text
- View/download PDF
83. Sindbis viral vectors transiently deliver tumor-associated antigens to lymph nodes and elicit diversified antitumor CD8+ T-cell immunity.
- Author
-
Granot T, Yamanashi Y, and Meruelo D
- Subjects
- Animals, Cricetinae, Cytotoxicity, Immunologic, Disease Models, Animal, Epitopes, T-Lymphocyte immunology, Female, Gene Expression, Gene Transfer Techniques, Genes, Reporter, Genetic Vectors administration & dosage, Genetic Vectors immunology, Immunologic Memory, Lymph Nodes metabolism, Lymphocyte Activation, Mice, Neoplasms pathology, Neoplasms therapy, Oncolytic Viruses genetics, Oncolytic Viruses immunology, Sindbis Virus immunology, Tumor Burden genetics, Tumor Burden immunology, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Genetic Vectors genetics, Lymph Nodes immunology, Neoplasms genetics, Neoplasms immunology, Sindbis Virus genetics
- Abstract
Tumors are theoretically capable of eliciting an antitumor immune response, but are often poorly immunogenic. Oncolytic viruses (OVs) have recently emerged as a promising strategy for the immunogenic delivery of tumor-associated antigens (TAAs) to cancer patients. However, safe and effective OV/TAA therapies have not yet been established. We have previously demonstrated that vectors based on Sindbis virus (SV) can inhibit tumor growth and activate the innate immune system in mice. Here, we demonstrate that SV vectors carrying a TAA generate a dramatically enhanced therapeutic effect in mice bearing subcutaneous, intraperitoneal, and lung cancers. Notably, SV/TAA efficacy was not dependent on tumor cell targeting, but was characterized by the transient expression of TAAs in lymph nodes draining the injection site. Early T-cell activation at this site was followed by a robust influx of NKG2D expressing antigen-specific cytotoxic CD8+ T cells into the tumor site, subsequently leading to the generation of long-lasting memory T cells which conferred protection against rechallenge with TAA-positive as well as TAA-negative tumor cells. By combining in vivo imaging, flow cytometry, cytotoxicity/cytokine assays, and tetramer analysis, we investigated the relationship between these events and propose a model for CD8+ T-cell activation during SV/TAA therapy.
- Published
- 2014
- Full Text
- View/download PDF
84. Tumor-specific targeting with modified Sindbis viral vectors: evaluation with optical imaging and positron emission tomography in vivo.
- Author
-
Stelter L, Tseng JC, Torosjan A, Levin B, Longo VA, Pillarsetty N, Zanzonico P, Meruelo D, and Larson SM
- Subjects
- Animals, Cell Line, Female, Fluorescent Dyes, Genetic Vectors genetics, Genetic Vectors metabolism, Humans, Mice, Mice, SCID, Receptors, Laminin genetics, Receptors, Laminin metabolism, Transfection, Drug Delivery Systems methods, Molecular Imaging methods, Optical Imaging methods, Positron-Emission Tomography methods, Sindbis Virus genetics
- Abstract
Purpose: Sindbis virus (SINV) infect tumor cells specifically and systemically throughout the body. Sindbis vectors are capable of expressing high levels of transduced suicide genes and thus efficiently produce enzymes for prodrug conversion in infected tumor cells. The ability to monitor suicide gene expression levels and viral load in patients, after administration of the vectors, would significantly enhance this tumor-specific therapeutic option., Procedures: The tumor specificity of SINV is mediated by the 67-kDa laminin receptor (LR). We probed different cancer cell lines for their LR expression and, to determine the specific role of LR-expression in the infection cycle, used different molecular imaging strategies, such as bioluminescence, fluorescence molecular tomography, and positron emission tomography, to evaluate SINV-mediated infection in vitro and in vivo., Results: All cancer cell lines showed a marked expression of LR. The infection rates of the SINV particles, however, differed significantly among the cell lines., Conclusion: We used novel molecular imaging techniques to visualize vector delivery to different neoplatic cells. SINV infection rates proofed to be not solely dependent on cellular LR expression. Further studies need to evaluate the herein discussed ways of cellular infection and viral replication.
- Published
- 2013
- Full Text
- View/download PDF
85. Comprehensive proteomic analysis of nonintegrin laminin receptor interacting proteins.
- Author
-
Venticinque L and Meruelo D
- Subjects
- Animals, Chaperonin Containing TCP-1 chemistry, Chaperonin Containing TCP-1 isolation & purification, Chromatography, Affinity, Cytoskeletal Proteins chemistry, Cytoskeletal Proteins isolation & purification, Hexosyltransferases, Histones chemistry, Histones isolation & purification, Humans, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins isolation & purification, Mice, NIH 3T3 Cells, Proteasome Endopeptidase Complex chemistry, Proteasome Endopeptidase Complex isolation & purification, Protein Binding, Proteome isolation & purification, Proteomics, RNA-Binding Proteins chemistry, RNA-Binding Proteins isolation & purification, Receptors, Laminin chemistry, Ribosomal Proteins chemistry, Ribosomal Proteins isolation & purification, Transcription Factors chemistry, Transcription Factors isolation & purification, Proteome chemistry
- Abstract
Human nonintegrin laminin receptor is a multifunctional protein acting as an integral component of the ribosome and a cell surface receptor for laminin-1. The laminin receptor is overexpressed in several human cancers and is also the cell surface receptor for several viruses and pathogenic prion proteins, making it a pathologically significant protein. This study focused on the proteomic characterization of laminin receptor interacting proteins from Mus musculus. The use of affinity chromatography with immobilized recombinant laminin receptor coupled with mass spectrometry analysis identified 45 proteins with high confidence. Following validation through coimmunoprecipitation, the proteins were classified based on predicted function into ribosomal, RNA processing, signal transduction/metabolism, protein processing, cytoskeleton/cell anchorage, DNA/chromatin, and unknown functions. A significant portion of the identified proteins is related to functions or localizations previously described for laminin receptor. This work represents a comprehensive proteomic approach to studying laminin receptor and provides an essential stepping stone to a better mechanistic understanding of this protein's diverse functions.
- Published
- 2012
- Full Text
- View/download PDF
86. ATM kinase is activated by sindbis viral vector infection.
- Author
-
Pampeno C, Hurtado A, and Meruelo D
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Line, Histones metabolism, Mice, Minichromosome Maintenance Complex Component 3, Nuclear Proteins metabolism, Phosphorylation, Time Factors, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Host-Pathogen Interactions, Protein Serine-Threonine Kinases metabolism, Sindbis Virus pathogenicity, Tumor Suppressor Proteins metabolism
- Abstract
Sindbis virus is a prototypic member of the Alphavirus genus, Togaviridae family. Sindbis replication results in cellular cytotoxicity, a feature that has been exploited by our laboratory for treatment of in vivo tumors. Understanding the interactions between Sindbis vectors and the host cell can lead to better virus production and increased efficacy of gene therapy vectors. Here we present studies investigating a possible cellular response to genotoxic effects of Sindbis vector infection. The Ataxia Telangiectasia Mutated (ATM) kinase, a sentinel against genomic and cellular stress, was activated by Sindbis vector infection at 3h post infection. ATM substrates, Mcm3 and the γH2AX histone, were subsequently phosphorylated, however, substrates involved with checkpoint arrest of DNA replication, p53, Chk1 and Chk2, were not differentially phosphorylated compared with uninfected cells. The ATM response suggests nuclear pertubation, resulting from cessation of host protein synthesis, as an early event in Sindbis vector infection., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
87. Interactions between laminin receptor and the cytoskeleton during translation and cell motility.
- Author
-
Venticinque L, Jamieson KV, and Meruelo D
- Subjects
- Actins metabolism, Humans, Multiprotein Complexes, Protein Binding, Receptors, Laminin physiology, Tubulin metabolism, Cell Movement, Cytoskeleton metabolism, Protein Biosynthesis, Receptors, Laminin metabolism
- Abstract
Human laminin receptor acts as both a component of the 40S ribosomal subunit to mediate cellular translation and as a cell surface receptor that interacts with components of the extracellular matrix. Due to its role as the cell surface receptor for several viruses and its overexpression in several types of cancer, laminin receptor is a pathologically significant protein. Previous studies have determined that ribosomes are associated with components of the cytoskeleton, however the specific ribosomal component(s) responsible has not been determined. Our studies show that laminin receptor binds directly to tubulin. Through the use of siRNA and cytoskeletal inhibitors we demonstrate that laminin receptor acts as a tethering protein, holding the ribosome to tubulin, which is integral to cellular translation. Our studies also show that laminin receptor is capable of binding directly to actin. Through the use of siRNA and cytoskeletal inhibitors we have shown that this laminin receptor-actin interaction is critical for cell migration. These data indicate that interactions between laminin receptor and the cytoskeleton are vital in mediating two processes that are intimately linked to cancer, cellular translation and migration.
- Published
- 2011
- Full Text
- View/download PDF
88. Structure-guided identification of a laminin binding site on the laminin receptor precursor.
- Author
-
Jamieson KV, Hubbard SR, and Meruelo D
- Subjects
- Amino Acid Sequence, Amino Acid Substitution genetics, Archaeal Proteins chemistry, Archaeoglobus fulgidus chemistry, Binding Sites, Conserved Sequence, Crystallography, X-Ray, Humans, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Protein Structure, Tertiary, Laminin metabolism, Receptors, Laminin chemistry, Receptors, Laminin metabolism
- Abstract
The 37/ 67-kDa human laminin receptor (LamR) is a cell surface receptor for laminin, prion protein, and a variety of viruses. Because of its wide range of ligands, LamR plays a role in numerous pathologies. LamR overexpression correlates with a highly invasive cell phenotype and increased metastatic ability, mediated by interactions between LamR and laminin. In addition, the specific targeting of LamR with small interfering RNAs, blocking antibodies, and Sindbis viral vectors confers anti-tumor effects. We adopted a structure-based approach to map a laminin binding site on human LamR by comparing the sequences and crystal structures of LamR and Archaeoglobus fulgidus S2p, a non-laminin-binding ortholog. Here, we identify a laminin binding site on LamR, comprising residues Phe32, Glu35, and Arg155, which are conserved among mammalian species. Mutation of these residues results in a significant loss of laminin binding. Further, recombinant wild-type LamR is able to act as a soluble decoy to inhibit cellular migration towards laminin. Mutation of this laminin binding site results in loss of migration inhibition, which demonstrates the physiological role of Phe32, Glu35, and Arg155 for laminin binding activity. Mapping of the LamR binding site should contribute to the development of therapeutics that inhibit LamR interactions with laminin and may aid in the prevention of tumor growth and metastasis., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
89. Activation of cytotoxic and regulatory functions of NK cells by Sindbis viral vectors.
- Author
-
Granot T, Venticinque L, Tseng JC, and Meruelo D
- Subjects
- Animals, Cell Count, Cell Line, Tumor, Cell Transformation, Neoplastic immunology, Cricetinae, Cytotoxicity, Immunologic genetics, Female, Genes, MHC Class II genetics, Genetic Vectors immunology, Humans, Interferon-gamma metabolism, Interleukin-12 genetics, Killer Cells, Natural cytology, Killer Cells, Natural metabolism, Killer Cells, Natural transplantation, Lac Operon genetics, Macrophages, Peritoneal immunology, Macrophages, Peritoneal metabolism, Mice, Peritoneum immunology, Peritoneum metabolism, Sindbis Virus physiology, Up-Regulation immunology, Cytotoxicity, Immunologic immunology, Genetic Vectors genetics, Killer Cells, Natural immunology, Oncolytic Virotherapy, Sindbis Virus genetics, Sindbis Virus immunology
- Abstract
Oncolytic viruses (OVs) represent a relatively novel anti-cancer modality. Like other new cancer treatments, effective OV therapy will likely require combination with conventional treatments. In order to design combinatorial treatments that work well together, a greater scrutiny of the mechanisms behind the individual treatments is needed. Sindbis virus (SV) based vectors have previously been shown to target and kill tumors in xenograft, syngeneic, and spontaneous mouse models. However, the effect of SV treatment on the immune system has not yet been studied. Here we used a variety of methods, including FACS analysis, cytotoxicity assays, cell depletion, imaging of tumor growth, cytokine blockade, and survival experiments, to study how SV therapy affects Natural Killer (NK) cell function in SCID mice bearing human ovarian carcinoma tumors. Surprisingly, we found that SV anti-cancer efficacy is largely NK cell-dependent. Furthermore, the enhanced therapeutic effect previously observed from Sin/IL12 vectors, which carry the gene for interleukin 12, is also NK cell dependent, but works through a separate IFNγ-dependent mechanism, which also induces the activation of peritoneal macrophages. These results demonstrate the multimodular nature of SV therapy, and open up new possibilities for potential synergistic or additive combinatorial therapies with other treatments.
- Published
- 2011
- Full Text
- View/download PDF
90. Sindbis viral vector induced apoptosis requires translational inhibition and signaling through Mcl-1 and Bak.
- Author
-
Venticinque L and Meruelo D
- Subjects
- Alphavirus Infections enzymology, Alphavirus Infections pathology, Alphavirus Infections virology, Animals, Cell Line, Tumor, Cytoplasmic Granules drug effects, Cytoplasmic Granules enzymology, Cytoplasmic Granules virology, Enzyme Activation drug effects, JNK Mitogen-Activated Protein Kinases metabolism, Mice, Mice, Inbred C57BL, Models, Biological, Myeloid Cell Leukemia Sequence 1 Protein, RNA, Double-Stranded metabolism, Stress, Physiological drug effects, eIF-2 Kinase metabolism, Apoptosis drug effects, Genetic Vectors pharmacology, Protein Biosynthesis drug effects, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction drug effects, Sindbis Virus genetics, bcl-2 Homologous Antagonist-Killer Protein metabolism
- Abstract
Background: Sindbis viral vectors are able to efficiently target and kill tumor cells in vivo, as shown using pancreatic and ovarian cancer models. Infection results in apoptosis both in vitro and in vivo. Sindbis vector uptake is mediated by the LAMR, which is upregulated on a number of different tumor types, thus conferring specificity of the vector to a wide range of cancers. In this study we elucidate the mechanism of apoptosis in two tumor cell lines, MOSEC, derived from the ovarian epithelium and Pan02, derived from a pancreatic adenocarcinoma. A comprehensive understanding of the mechanism of apoptosis would facilitate the design of more effective vectors for cancer therapy., Results: The initial phase of Sindbis vector induced apoptosis in MOSEC and Pan02 models reconfirms that viral infection is sensed by PKR due to double-stranded RNA intermediates associated with genomic replication. PKR activation results in translation inhibition through eIF2alpha phosphorylation and initiation of the stress response. Our studies indicate that the roles of two proteins, Mcl-1 and JNK, intimately link Sindbis induced translational arrest and cellular stress. Translational arrest inhibits the synthesis of anti-apoptotic Bcl-2 protein, Mcl-1. JNK activation triggers the release of Bad from 14-3-3, which ultimately results in apoptosis. These signals from translational arrest and cellular stress are propagated to the mitochondria where Bad and Bik bind to Bcl-xl and Mcl-1 respectively. Formation of these heterodimers displaces Bak, which results in caspase 9 cleavage and signaling through the mitochondrial pathway of apoptosis., Conclusion: The host cell response to Sindbis is triggered through PKR activation. Our studies demonstrate that PKR activation and subsequent translational arrest is linked to both cellular stress and apoptosis. We have also found the linkage point between translational arrest and apoptosis to be Mcl-1, a protein whose constant translation is required for inhibition of apoptosis. With this information vectors can be designed, which express or repress proteins implicated in this study, to enhance their therapeutic potential.
- Published
- 2010
- Full Text
- View/download PDF
91. Multiple functions of the 37/67-kd laminin receptor make it a suitable target for novel cancer gene therapy.
- Author
-
Scheiman J, Tseng JC, Zheng Y, and Meruelo D
- Subjects
- Animals, Blotting, Western, Cell Cycle genetics, Cell Cycle physiology, Cell Line, Cell Line, Tumor, Cell Proliferation, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinases metabolism, Female, Humans, Mice, Mice, SCID, Neoplasms metabolism, RNA, Small Interfering, Receptors, Laminin genetics, Ribosome Subunits, Small, Eukaryotic genetics, Ribosome Subunits, Small, Eukaryotic metabolism, Genetic Therapy methods, Neoplasms therapy, Receptors, Laminin physiology
- Abstract
The 37/67-kd laminin receptor, LAMR, is a multifunctional protein that associates with the 40S ribosomal subunit and also localizes to the cell membrane to interact with the extracellular matrix. LAMR is overexpressed in many types of cancer, playing important roles in tumor-cell migration and invasion. Here, we show that LAMR is also vital for tumor-cell proliferation, survival, and protein translation. Small-interfering RNA (siRNA)-mediated reduction in expression of LAMR leads to G1 phase cell-cycle arrest in vitro by altering cyclins A2/B1, cyclin-dependent kinases (CDKs) 1/2, Survivin, and p21 expression levels. In vivo, reduction in LAMR expression results in inhibition of HT1080 cells to develop tumors. We also found that LAMR's ribosomal functions are critical for translation as reduction in LAMR expression leads to a dramatic decrease in newly synthesized proteins. Further, cells with lower expression of LAMR have fewer 40S subunits and 80S monosomes, causing an increase in free 60S ribosomal subunits. These results indicate that LAMR is able to regulate tumor development in many ways; further enhancing its potential as a target for gene therapy. To test this, we developed a novel Sindbis/Lenti pseudotype vector carrying short-hairpin RNA (shRNA) designed against lamr. This pseudotype vector effectively reduces LAMR expression and specifically targets tumors in vivo. Treatment of tumor-bearing severe combine immunodeficient (SCID) mice with this pseudotype vector significantly inhibits tumor growth. Thus, we show that LAMR can be used as a target in novel therapy for tumor reduction and elimination.
- Published
- 2010
- Full Text
- View/download PDF
92. CCR7 signalling as an essential regulator of CNS infiltration in T-cell leukaemia.
- Author
-
Buonamici S, Trimarchi T, Ruocco MG, Reavie L, Cathelin S, Mar BG, Klinakis A, Lukyanov Y, Tseng JC, Sen F, Gehrie E, Li M, Newcomb E, Zavadil J, Meruelo D, Lipp M, Ibrahim S, Efstratiadis A, Zagzag D, Bromberg JS, Dustin ML, and Aifantis I
- Subjects
- Animals, Cell Adhesion, Cell Line, Tumor, Chemokine CCL19 deficiency, Chemokine CCL19 metabolism, Chemokine CCL21 metabolism, Humans, Mice, Mice, Inbred C57BL, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Receptors, CCR7 deficiency, Central Nervous System metabolism, Central Nervous System pathology, Leukemia, T-Cell metabolism, Leukemia, T-Cell pathology, Receptors, CCR7 metabolism, Signal Transduction
- Abstract
T-cell acute lymphoblastic leukaemia (T-ALL) is a blood malignancy afflicting mainly children and adolescents. T-ALL patients present at diagnosis with increased white cell counts and hepatosplenomegaly, and are at an increased risk of central nervous system (CNS) relapse. For that reason, T-ALL patients usually receive cranial irradiation in addition to intensified intrathecal chemotherapy. The marked increase in survival is thought to be worth the considerable side-effects associated with this therapy. Such complications include secondary tumours, neurocognitive deficits, endocrine disorders and growth impairment. Little is known about the mechanism of leukaemic cell infiltration of the CNS, despite its clinical importance. Here we show, using T-ALL animal modelling and gene-expression profiling, that the chemokine receptor CCR7 (ref. 5) is the essential adhesion signal required for the targeting of leukaemic T-cells into the CNS. Ccr7 gene expression is controlled by the activity of the T-ALL oncogene Notch1 and is expressed in human tumours carrying Notch1-activating mutations. Silencing of either CCR7 or its chemokine ligand CCL19 (ref. 6) in an animal model of T-ALL specifically inhibits CNS infiltration. Furthermore, murine CNS-targeting by human T-ALL cells depends on their ability to express CCR7. These studies identify a single chemokine-receptor interaction as a CNS 'entry' signal, and open the way for future pharmacological targeting. Targeted inhibition of CNS involvement in T-ALL could potentially decrease the intensity of CNS-targeted therapy, thus reducing its associated short- and long-term complications.
- Published
- 2009
- Full Text
- View/download PDF
93. Crystal structure of the human laminin receptor precursor.
- Author
-
Jamieson KV, Wu J, Hubbard SR, and Meruelo D
- Subjects
- Amino Acid Sequence, Animals, Caenorhabditis elegans, Crystallography, X-Ray, Humans, Ligands, Models, Molecular, Molecular Conformation, Molecular Sequence Data, Protein Binding, Receptors, Cell Surface chemistry, Saccharomyces cerevisiae, Sequence Homology, Amino Acid, Protein Precursors chemistry, Receptors, Laminin chemistry
- Abstract
The human laminin receptor (LamR) interacts with many ligands, including laminin, prions, Sindbis virus, and the polyphenol (-)-epigallocatechin-3-gallate (EGCG), and has been implicated in a number of diseases. LamR is overexpressed on tumor cells, and targeting LamR elicits anti-cancer effects. Here, we report the crystal structure of human LamR, which provides insights into its function and should facilitate the design of novel therapeutics targeting LamR.
- Published
- 2008
- Full Text
- View/download PDF
94. Tumor-specific in vivo transfection with HSV-1 thymidine kinase gene using a Sindbis viral vector as a basis for prodrug ganciclovir activation and PET.
- Author
-
Tseng JC, Zanzonico PB, Levin B, Finn R, Larson SM, and Meruelo D
- Subjects
- Animals, Cricetinae, Female, Kidney metabolism, Mice, Mice, SCID, Prodrugs chemistry, Transfection, Ganciclovir pharmacology, Gene Transfer Techniques, Herpesvirus 1, Human genetics, Positron-Emission Tomography methods, Sindbis Virus genetics, Thymidine Kinase genetics
- Abstract
Unlabelled: One type of gene therapy of tumors, gene-directed enzyme-prodrug therapy (GDEPT), holds considerable promise, although practical considerations limit its clinical applicability. These include the lack of acceptable noninvasive methods that are adaptable to humans for selective tumor targeting of the therapeutic genetic material. Sindbis virus is an oncolytic, alpha-virus that selectively targets tumors through the 67-kDa laminin receptor (LAMR). In this report we describe a novel approach that permits tumor-selective tumor targeting and quantitative in vivo monitoring using PET of a commonly applied GDEPT, based on herpes simplex virus thymidine kinase type 1 (HSVtk) and ganciclovir (GCV)., Methods: Sindbis/tk vectors were harvested from the supernatant of in vitro cultures of a packaging cell produced by electroporation of both replicon RNA (SinRep5/tk) and helper RNA (DH-BB) into baby hamster kidney (BHK) cells. The therapeutic effect of GCV was determined by incubation of transfected tumor cells with increasing concentrations of GCV. BHK tumors growing as xenografts in severe combined immunodeficiency disease (SCID) mice were transfected by parenteral administration of the vector. Imaging was performed using small-animal PET at 2 h after injection of 18F fluoro-ethyl-arabinosyluridine (18F-FEAU) and 24 h after the final parenteral injection of Sindbis/tk viral vector., Results: The vector efficiently expresses the HSVtk enzyme in infected tumor cells, both in vitro and in vivo. High levels of HSVtk expression ensure sufficient prodrug GCV conversion and activation for bystander effects that kill the surrounding untransduced tumor cells. Tumor localization of intravenously administered 18F-FEAU after 2 and 3 parenteral vector treatments of Sindbis/tk demonstrated uptake of 1.7 and 3.1 %ID/g (percentage injected dose per gram), respectively., Conclusion: The vector efficiently targets the HSVtk enzyme gene into Sindbis-infected tumor cells. High levels of HSVtk expression ensure sufficient prodrug GCV conversion and activation for bystander effects that killed many surrounding untransduced tumor cells. In addition, the HSVtk activities in tumors can be noninvasively monitored using PET after systemic Sindbis/tk treatments as a basis for determining the levels and tissue distribution of vector, noninvasively in living animals, and for optimizing in vivo transfection rates of tumor.
- Published
- 2006
95. Identification of amino acids of Sindbis virus E2 protein involved in targeting tumor metastases in vivo.
- Author
-
Hurtado A, Tseng JC, Boivin C, Levin B, Yee H, Pampeno C, and Meruelo D
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Mice, Mice, SCID, Mutation, Neoplasm Metastasis pathology, Protein Transport, Sequence Analysis, DNA, Transfection, Viral Envelope Proteins genetics, Viral Envelope Proteins therapeutic use, Genetic Vectors, Neoplasm Metastasis therapy, Plasmids genetics, Sindbis Virus genetics, Viral Envelope Proteins chemistry
- Abstract
Previous studies conducted in our laboratory with Sindbis viral vectors in animal models demonstrated excellent in vivo targeting of tumor cells and significant reduction of metastatic implant size. To explore the influence of Sindbis strain on these factors, we constructed new plasmids from the wild-type Ar-339 Sindbis virus strain and compared their sequences. We found differences in the replicase and envelope proteins between JT, HRSP, and Ar-339 sequences. We made chimeras combining both strains and studied their efficiency in SCID mice bearing tumor xenograft using IVIS in vivo imaging techniques. We found that JT envelope proteins targeted tumors more efficiently than those of Ar-339, while the Ar-339 replicase showed increased efficacy in tumor reduction. To determine which residues are responsible for tumor targeting, we made mutants of Ar-339 E2 envelope protein and tested them by IVIS imaging in ES-2 tumor-bearing and tumor-free mice. The change of only one amino acid from E70 to K70 in Ar-339 E2 suppressed the ability to target metastatic tumor implants in mice. A K70 and V251 double E2 mutant did not reverse the loss of targeting capability. Only the mutant with JT E2 and Ar-339 helper targeted tumor, though with less intensity.
- Published
- 2005
- Full Text
- View/download PDF
96. Anti-angiogenic activities of hypericin in vivo: potential for ophthalmologic applications.
- Author
-
Lavie G, Mandel M, Hazan S, Barliya T, Blank M, Grunbaum A, Meruelo D, and Solomon A
- Subjects
- Angiogenesis Inducing Agents, Angiogenesis Inhibitors therapeutic use, Animals, Anthracenes, Cornea blood supply, Corneal Neovascularization chemically induced, Disease Models, Animal, Endothelium, Vascular drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Fibroblast Growth Factor 2, Genes, Tumor Suppressor, Humans, Iris blood supply, Matrix Metalloproteinases, Membrane-Associated, Metalloendopeptidases antagonists & inhibitors, Nuclear Proteins, Perylene pharmacology, Perylene therapeutic use, Phosphorylation drug effects, Rats, Rats, Wistar, Retinal Neovascularization chemically induced, Retinal Neovascularization prevention & control, Vascular Endothelial Growth Factor A physiology, Angiogenesis Inhibitors pharmacology, Corneal Neovascularization prevention & control, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Perylene analogs & derivatives
- Abstract
Hypericin, a perihydroxylated dianthraquinone is shown here to be a highly potent inhibitor of angiogenesis in several ocular models examined in rat eyes. Extensive angiogenesis induced in the cornea and iris by intra-ocular administration of FGF-2 was effectively inhibited by a minimum of four dose regimens of hypericin (2 mg/kg) administered via the intraperitoneal route at 48 h intervals. Maximal inhibition was achieved when animal treatment with hypericin was initiated 48 h prior to inoculation of FGF-2. The molecular basis for the hypericin-mediated inhibition of angiogenesis in the anterior eye compartment appears to involve several sites in the cascade leading to angiogenesis. We show that the activating phosphorylation of extracellular signal-regulated MAP kinases (ERK1/2) is inhibited by hypericin in human retinal pigment epithelial cells and in EA.hy926 cells, an endothelial hybridoma expressing endothelial cell properties. ERK1/2 activity is required for the transactivation of hypoxia-inducible factor 1 alpha (HIF-1alpha) and in VEGF-induced blood vessel sprouting. MT1-MMP activity in human microvascular endothelial cells was also inhibited. The findings identify hypericin as a potentially useful agent in the treatment of ophthalmic neovascularization pathogeneses.
- Published
- 2005
- Full Text
- View/download PDF
97. Using sindbis viral vectors for specific detection and suppression of advanced ovarian cancer in animal models.
- Author
-
Tseng JC, Hurtado A, Yee H, Levin B, Boivin C, Benet M, Blank SV, Pellicer A, and Meruelo D
- Subjects
- Animals, Female, Genes, Reporter genetics, Genetic Vectors genetics, Genetic Vectors physiology, Humans, Immunohistochemistry, Luminescent Measurements, Mice, Mice, Inbred C57BL, Mice, SCID, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms therapy, Peritoneal Neoplasms secondary, Peritoneal Neoplasms virology, RNA, Small Interfering genetics, Receptors, Laminin biosynthesis, Receptors, Laminin genetics, Receptors, Laminin metabolism, Sindbis Virus genetics, Sindbis Virus pathogenicity, Xenograft Model Antitumor Assays, Ovarian Neoplasms virology, Sindbis Virus physiology
- Abstract
We studied the therapeutic value of Sindbis vectors for advanced metastatic ovarian cancer by using two highly reproducible and clinically accurate mouse models: a SCID xenograft model, established by i.p. inoculation of human ES-2 ovarian cancer cells, and a syngenic C57BL/6 model, established by i.p. inoculation of mouse MOSEC ovarian cancer cells. We demonstrate through imaging, histologic, and molecular data that Sindbis vectors systemically and specifically infect/detect and kill metastasized tumors in the peritoneal cavity, leading to significant suppression of the carcinomatosis in both animal models. Use of two different bioluminescent genetic markers for the IVIS Imaging System permitted demonstration, for the first time, of an excellent correlation between vector delivery and metastatic locations in vivo. Sindbis vector infection and growth suppression of murine MOSEC tumor cells indicate that Sindbis tumor specificity is not attributable to a species difference between human tumor and mouse normal cells. Sindbis virus is known to infect mammalian cells using the Mr 67,000 laminin receptor. Immunohistochemical staining of tumor cells indicates that laminin receptor is elevated in tumor versus normal cells. Down-regulated expression of laminin receptor with small interfering RNA significantly reduces the infectivity of Sindbis vectors. Tumor overexpression of the laminin receptor may explain the specificity and efficacy that Sindbis vectors demonstrate for tumor cells in vivo. We show that incorporation of antitumor cytokine genes such as interleukin-12 and interleukin-15 genes enhances the efficacy of the vector. These results suggest that Sindbis viral vectors may be promising agents for both specific detection and growth suppression of metastatic ovarian cancer.
- Published
- 2004
- Full Text
- View/download PDF
98. Antimetastatic activity of the photodynamic agent hypericin in the dark.
- Author
-
Blank M, Lavie G, Mandel M, Hazan S, Orenstein A, Meruelo D, and Keisari Y
- Subjects
- Animals, Anthracenes, Breast Neoplasms veterinary, Carcinoma, Squamous Cell veterinary, Disease Models, Animal, Female, Humans, Light, Lung Neoplasms veterinary, Male, Mice, Mice, Inbred BALB C, Signal Transduction, Survival Analysis, Tissue Distribution, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Breast Neoplasms pathology, Carcinoma, Squamous Cell pathology, Lung Neoplasms drug therapy, Lung Neoplasms secondary, Perylene analogs & derivatives, Perylene pharmacokinetics, Perylene pharmacology
- Abstract
A unique property of the photodynamic signal transduction inhibitor hypericin (HY) is high functionality in the dark, which has been shown to result in portfolio of anticancer activities both in vitro and in vivo. Here we show that treatment with HY significantly reduces growth rate of metastases in 2 murine models: breast adenocarcinoma (DA3) and squamous cell carcinoma (SQ2). Focus on metastases was achieved by resection of primary tumors at stages in which micrometastases exist in lungs. Long-term animal survival in DA3 tumor-excised groups increased from 15.6% in controls to 34.5% following supplementary treatment with HY. In mice bearing SQ2 tumor metastases, therapy with HY increased animal survival from 17.7% in controls to 46.1%. Using Laser-induced fluorescence and multipixel spectral image analyses, we demonstrate that HY has a high tendency to accumulate in primary and metastatic tumors; HY content in lungs bearing metastases was approximately 2-fold higher than in the lungs of healthy animals. The tendency of HY to preferentially concentrate in lung metastases, combined with its potent antiproliferative activities, may render HY as a useful supplementary modality in the treatment of metastatic cancer irrespective of photoactivation., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
99. Systemic gene therapy by Sindbis vectors: A potentially safe and effective targeted therapy for identifying and killing tumor cells in vivo.
- Author
-
Meruelo D
- Abstract
Extract: A major obstacle to the development of gene therapy for cancer has been the inability to specifically and systemically deliver gene therapy vectors throughout the body to primary and/or metastasized tumor cells. Although intratumor injections of gene therapy vectors have sometimes been possible, no viral vector has been available that could be administered systemically and would selectively and efficiently target tumors without infection of normal tissues. Furthermore, even when locally injected, many viral vectors end up at high concentrations in the liver, because many cells of the body have low receptor numbers for some of the vectors in current use, whereas the liver has high numbers of such receptors. A number of ingenious approaches have been tried but none so far have fully resolved the problem. For example, tumor-specific promoters have been incorporated into the vectors so that gene expression and/or replication can occur in tumor cells but not normal cells. Unfortunately, only a small fraction of these vectors are typically taken up by the target tumor and expressed. In such cases, tumor cell death is generally insufficient to eradicate or significantly slow tumor growth.
- Published
- 2004
100. Systemic tumor targeting and killing by Sindbis viral vectors.
- Author
-
Tseng JC, Levin B, Hurtado A, Yee H, Perez de Castro I, Jimenez M, Shamamian P, Jin R, Novick RP, Pellicer A, and Meruelo D
- Subjects
- Animals, Cell Line, Female, Genetic Vectors, Immunohistochemistry, Luciferases metabolism, Mice, Mice, Inbred C57BL, Mice, SCID, Neoplasm Metastasis, Neoplasm Transplantation, Time Factors, Genetic Therapy methods, Neoplasms therapy, Sindbis Virus genetics
- Abstract
Successful cancer gene therapy requires a vector that systemically and specifically targets tumor cells throughout the body. Although several vectors have been developed to express cytotoxic genes via tumor-specific promoters or to selectively replicate in tumor cells, most are taken up and expressed by just a few targeted tumor cells. By contrast, we show here that blood-borne Sindbis viral vectors systemically and specifically infect tumor cells. A single intraperitoneal treatment allows the vectors to target most tumor cells, as demonstrated by immunohistochemistry, without infecting normal cells. Further, Sindbis infection is sufficient to induce complete tumor regression. We demonstrate systemic vector targeting of tumors growing subcutaneously, intrapancreatically, intraperitoneally and in the lungs. The vectors can also target syngeneic and spontaneous tumors in immune-competent mice. We document the anti-tumor specificity of a vector that systemically targets and eradicates tumor cells throughout the body without adverse effects.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.