Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 607(7917): 185-190, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35732735

RESUMO

Translation initiation defines the identity and quantity of a synthesized protein. The process is dysregulated in many human diseases1,2. A key commitment step is when the ribosomal subunits join at a translation start site on a messenger RNA to form a functional ribosome. Here, we combined single-molecule spectroscopy and structural methods using an in vitro reconstituted system to examine how the human ribosomal subunits join. Single-molecule fluorescence revealed when the universally conserved eukaryotic initiation factors eIF1A and eIF5B associate with and depart from initiation complexes. Guided by single-molecule dynamics, we visualized initiation complexes that contained both eIF1A and eIF5B using single-particle cryo-electron microscopy. The resulting structure revealed how eukaryote-specific contacts between the two proteins remodel the initiation complex to orient the initiator aminoacyl-tRNA in a conformation compatible with ribosomal subunit joining. Collectively, our findings provide a quantitative and architectural framework for the molecular choreography orchestrated by eIF1A and eIF5B during translation initiation in humans.


Assuntos
Fator de Iniciação 1 em Eucariotos , Fatores de Iniciação em Eucariotos , RNA de Transferência de Metionina , Subunidades Ribossômicas , Microscopia Crioeletrônica , Fator de Iniciação 1 em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/genética , Humanos , RNA de Transferência de Metionina/genética , RNA de Transferência de Metionina/metabolismo , Subunidades Ribossômicas/química , Subunidades Ribossômicas/metabolismo , Imagem Individual de Molécula
2.
J Biol Chem ; 300(5): 107242, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38569933

RESUMO

Messenger RNA (mRNA) recruitment to the 40S ribosomal subunit is mediated by eukaryotic initiation factor 4F (eIF4F). This complex includes three subunits: eIF4E (m7G cap-binding protein), eIF4A (DEAD-box helicase), and eIF4G. Mammalian eIF4G is a scaffold that coordinates the activities of eIF4E and eIF4A and provides a bridge to connect the mRNA and 40S ribosomal subunit through its interaction with eIF3. While the roles of many eIF4G binding domains are relatively clear, the precise function of RNA binding by eIF4G remains to be elucidated. In this work, we used an eIF4G-dependent translation assay to reveal that the RNA binding domain (eIF4G-RBD; amino acids 682-720) stimulates translation. This stimulating activity is observed when eIF4G is independently tethered to an internal region of the mRNA, suggesting that the eIF4G-RBD promotes translation by a mechanism that is independent of the m7G cap and mRNA tethering. Using a kinetic helicase assay, we show that the eIF4G-RBD has a minimal effect on eIF4A helicase activity, demonstrating that the eIF4G-RBD is not required to coordinate eIF4F-dependent duplex unwinding. Unexpectedly, native gel electrophoresis and fluorescence polarization assays reveal a previously unidentified direct interaction between eIF4G and the 40S subunit. Using binding assays, our data show that this 40S subunit interaction is separate from the previously characterized interaction between eIF4G and eIF3. Thus, our work reveals how eIF4F can bind to the 40S subunit using eIF3-dependent and eIF3-independent binding domains to promote translation initiation.


Assuntos
Fator de Iniciação 4E em Eucariotos , Biossíntese de Proteínas , Subunidades Ribossômicas Menores de Eucariotos , Humanos , Fator de Iniciação 3 em Eucariotos/química , Fator de Iniciação 3 em Eucariotos/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Ligação Proteica , Domínios Proteicos , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Fator de Iniciação 4E em Eucariotos/metabolismo
3.
J Biol Chem ; 298(10): 102368, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35963437

RESUMO

During translation initiation, the underlying mechanism by which the eukaryotic initiation factor (eIF) 4E, eIF4A, and eIF4G components of eIF4F coordinate their binding activities to regulate eIF4F binding to mRNA is poorly defined. Here, we used fluorescence anisotropy to generate thermodynamic and kinetic frameworks for the interaction of uncapped RNA with human eIF4F. We demonstrate that eIF4E binding to an autoinhibitory domain in eIF4G generates a high-affinity binding conformation of the eIF4F complex for RNA. In addition, we show that the nucleotide-bound state of the eIF4A component further regulates uncapped RNA binding by eIF4F, with a four-fold decrease in the equilibrium dissociation constant observed in the presence versus the absence of ATP. Monitoring uncapped RNA dissociation in real time reveals that ATP reduces the dissociation rate constant of RNA for eIF4F by ∼4-orders of magnitude. Thus, release of ATP from eIF4A places eIF4F in a dynamic state that has very fast association and dissociation rates from RNA. Monitoring the kinetic framework for eIF4A binding to eIF4G revealed two different rate constants that likely reflect two conformational states of the eIF4F complex. Furthermore, we determined that the eIF4G autoinhibitory domain promotes a more stable, less dynamic, eIF4A-binding state, which is overcome by eIF4E binding. Overall, our data support a model whereby eIF4E binding to eIF4G/4A stabilizes a high-affinity RNA-binding state of eIF4F and enables eIF4A to adopt a more dynamic interaction with eIF4G. This dynamic conformation may contribute to the ability of eIF4F to rapidly bind and release mRNA during scanning.


Assuntos
Fator de Iniciação 4A em Eucariotos , Fator de Iniciação 4E em Eucariotos , Humanos , Trifosfato de Adenosina/metabolismo , Fator de Iniciação 4A em Eucariotos/química , Fator de Iniciação 4E em Eucariotos/química , Fator de Iniciação 4F em Eucariotos/química , Fator de Iniciação Eucariótico 4G/química , Nucleotídeos/química , Ligação Proteica , RNA Mensageiro/metabolismo
4.
Blood ; 125(24): 3778-88, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25904246

RESUMO

Allogeneic hematopoietic cell transplant (allo-HCT) can be curative for certain hematologic malignancies, but the risk of graft-versus-host disease (GVHD) is a major limitation for wider application. Ideally, strategies to improve allo-HCT would involve suppression of T lymphocytes that drive GVHD while sparing those that mediate graft-versus-malignancy (GVM). Recently, using a xenograft model, we serendipitously discovered that myxoma virus (MYXV) prevented GVHD while permitting GVM. In this study, we show that MYXV binds to resting, primary human T lymphocytes but will only proceed into active virus infection after the T cells receive activation signals. MYXV-infected T lymphocytes exhibited impaired proliferation after activation with reduced expression of interferon-γ, interleukin-2 (IL-2), and soluble IL-2Rα, but did not affect expression of IL-4 and IL-10. MYXV suppressed T-cell proliferation in 2 patterns (full vs partial) depending on the donor. In terms of GVM, we show that MYXV-infected activated human T lymphocytes effectively deliver live oncolytic virus to human multiple myeloma cells, thus augmenting GVM by transfer of active oncolytic virus to residual cancer cells. Given this dual capacity of reducing GVHD plus increasing the antineoplastic effectiveness of GVM, ex vivo virotherapy with MYXV may be a promising clinical adjunct to allo-HCT regimens.


Assuntos
Mieloma Múltiplo/terapia , Myxoma virus/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Linfócitos T/imunologia , Linfócitos T/virologia , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/terapia , Transplante de Células-Tronco Hematopoéticas , Humanos , Ativação Linfocitária , Infecções por Poxviridae/imunologia , Linfócitos T/citologia , Infecções Tumorais por Vírus/imunologia
5.
Cytotherapy ; 18(3): 465-80, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26857235

RESUMO

BACKGROUND: Relapsing disease is a major challenge after hematopoietic cell transplantation for hematological malignancies. Myxoma virus (MYXV) is an oncolytic virus that can target and eliminate contaminating cancer cells from auto-transplant grafts. The aims of this study were to examine the impact of MYXV on normal hematopoietic stem and progenitor cells and define the optimal treatment conditions for ex vivo virotherapy. METHODS: Bone marrow (BM) and mobilized peripheral blood stem cells (mPBSCs) from patients with hematologic malignancies were treated with MYXV at various time, temperature and incubation media conditions. Treated BM cells from healthy normal donors were evaluated using flow cytometry for MYXV infection, long-term culture-initiating cell (LTC-IC) assay and colony-forming cell (CFC) assay. RESULTS: MYXV initiated infection in up to 45% of antigen-presenting monocytes, B cells and natural killer cells; however, these infections were uniformly aborted in >95% of all cells. Fresh graft sources showed higher levels of MYXV infection initiation than cryopreserved specimens, but in all cases less than 10% of CD34(+) cells could be infected after ex vivo MYXV treatment. MYXV did not impair LTC-IC colony numbers compared with mock treatment. CFC colony types and numbers were also not impaired by MYXV treatment. MYXV incubation time, temperature or culture media did not significantly change the percentage of infected cells, LTC-IC colony formation or CFC colony formation. CONCLUSIONS: Human hematopoietic cells are non-permissive for MYXV. Human hematopoietic stem and progenitor cells were not infected and thus unaffected by MYXV ex vivo treatment.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Neoplasias Hematológicas/patologia , Células-Tronco Hematopoéticas/citologia , Myxoma virus/fisiologia , Terapia Viral Oncolítica/métodos , Adulto , Antígenos CD34/metabolismo , Autoenxertos/normas , Medula Óssea/patologia , Células da Medula Óssea/patologia , Células Cultivadas , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/normas , Células-Tronco Hematopoéticas/fisiologia , Humanos , Masculino , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/prevenção & controle , Condicionamento Pré-Transplante/métodos
6.
J Biol Chem ; 288(46): 32932-40, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24092755

RESUMO

Recruitment of mRNA to the 40S ribosomal subunit requires the coordinated interaction of a large number of translation initiation factors. In mammals, the direct interaction between eukaryotic initiation factor 4G (eIF4G) and eIF3 is thought to act as the molecular bridge between the mRNA cap-binding complex and the 40S subunit. A discrete ∼90 amino acid domain in eIF4G is responsible for binding to eIF3, but the identity of the eIF3 subunit(s) involved is less clear. The eIF3e subunit has been shown to directly bind eIF4G, but the potential role of other eIF3 subunits in stabilizing this interaction has not been investigated. It is also not clear if the eIF4A helicase plays a role in stabilizing the interaction between eIF4G and eIF3. Here, we have used a fluorescence anisotropy assay to demonstrate that eIF4G binds to eIF3 independently of eIF4A binding to the middle region of eIF4G. By using a site-specific cross-linking approach, we unexpectedly show that the eIF4G-binding surface in eIF3 is comprised of the -c, -d and -e subunits. Screening multiple cross-linker positions reveals that eIF4G contains two distinct eIF3-binding subdomains within the previously identified eIF3-binding domain. Finally, by employing an eIF4G-dependent translation assay, we establish that both of these subdomains are required for efficient mRNA recruitment to the ribosome and stimulate translation. Our study reveals unexpected complexity to the eIF3-eIF4G interaction that provides new insight into the regulation of mRNA recruitment to the human ribosome.


Assuntos
Fator de Iniciação 3 em Eucariotos/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Iniciação Traducional da Cadeia Peptídica/fisiologia , RNA Mensageiro/metabolismo , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Fator de Iniciação 3 em Eucariotos/química , Fator de Iniciação Eucariótico 4G/química , Células HeLa , Humanos , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/química , Subunidades Ribossômicas Menores de Eucariotos/química
7.
Proc Natl Acad Sci U S A ; 108(51): 20473-8, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22135459

RESUMO

Protein fate in higher eukaryotes is controlled by three complexes that share conserved architectural elements: the proteasome, COP9 signalosome, and eukaryotic translation initiation factor 3 (eIF3). Here we reconstitute the 13-subunit human eIF3 in Escherichia coli, revealing its structural core to be the eight subunits with conserved orthologues in the proteasome lid complex and COP9 signalosome. This structural core in eIF3 binds to the small (40S) ribosomal subunit, to translation initiation factors involved in mRNA cap-dependent initiation, and to the hepatitis C viral (HCV) internal ribosome entry site (IRES) RNA. Addition of the remaining eIF3 subunits enables reconstituted eIF3 to assemble intact initiation complexes with the HCV IRES. Negative-stain EM reconstructions of reconstituted eIF3 further reveal how the approximately 400 kDa molecular mass structural core organizes the highly flexible 800 kDa molecular mass eIF3 complex, and mediates translation initiation.


Assuntos
Fator de Iniciação 3 em Eucariotos/química , Complexo do Signalossomo COP9 , DNA Complementar/metabolismo , Escherichia coli/metabolismo , Células HeLa , Hepacivirus/genética , Hepacivirus/metabolismo , Humanos , Microscopia Eletrônica/métodos , Modelos Moleculares , Conformação Molecular , Complexos Multiproteicos/química , Peptídeo Hidrolases/química , Ligação Proteica , Biossíntese de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Ribossomos/química
8.
Int J Mol Sci ; 15(7): 11523-38, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24979134

RESUMO

Eukaryotic translation initiation factors are the principal molecular effectors regulating the process converting nucleic acid to functional protein. Commonly referred to as eIFs (eukaryotic initiation factors), this suite of proteins is comprised of at least 25 individual subunits that function in a coordinated, regulated, manner during mRNA translation. Multiple facets of eIF regulation have yet to be elucidated; however, many of the necessary protein factors are phosphorylated. Herein, we have isolated, identified and quantified phosphosites from eIF2, eIF3, and eIF4G generated from log phase grown HeLa cell lysates. Our investigation is the first study to globally quantify eIF phosphosites and illustrates differences in abundance of phosphorylation between the residues of each factor. Thus, identification of those phosphosites that exhibit either high or low levels of phosphorylation under log phase growing conditions may aid researchers to concentrate their investigative efforts to specific phosphosites that potentially harbor important regulatory mechanisms germane to mRNA translation.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , Processamento de Proteína Pós-Traducional , Células HeLa , Humanos , Fosforilação
9.
Am Surg ; 90(9): 2249-2252, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38871348

RESUMO

BACKGROUND: The rise of value-based purchasing has led to decreased compensation for hospital-acquired conditions, including surgical site infections (SSI). This study aims to assess the risk factors for SSI in children and teenagers undergoing gastrointestinal surgery across US hospitals. METHODS: The 2018-2020 Nationwide Readmissions Database was queried for patients undergoing gastrointestinal surgery under the age of 18. The primary outcome was SSI during index admission or readmission within a year. Comparison groups were elective, trauma, and emergent surgery based on anatomic location and urgency. Univariable comparison used chi-squared tests for relevant variables. Confounders were addressed through multivariable logistic regression with significant variables from univariable analysis. RESULTS: 113 108 total patients met the study criteria. The SSI rate during admission or readmission was 2.9% (n = 3254). Infections during admission and readmission were 1.4% (n = 1560) and 1.5% (n = 1694), respectively. The most common site was organ space (48.6%, n = 1657). Increased infection risk was associated with trauma (OR 1.80 [1.51-2.16] P < .001), emergency surgery (OR 1.31 [1.17-1.47] P < .001), large bowel surgery (OR 2.78 [2.26-3.43] P < .001), and those with three or more comorbidities (OR 2.03 [1.69-2.45] P < .001). Investor-owned hospitals (OR .65 [.56-.76] P < .001) and highest quartile income (OR .80 [.73-.88] P < .001) were associated with decreased infection risk. CONCLUSIONS: Pediatric patients undergoing gastrointestinal surgery face an elevated risk of SSI, especially in trauma and emergency surgeries, particularly with multiple comorbidities. Meanwhile, a reduced risk is observed in high-income and investor-owned hospital settings. Hospitals and surgeons caring for high risk patients should advocate for risk adjustment in value-based payment systems.


Assuntos
Comorbidade , Procedimentos Cirúrgicos do Sistema Digestório , Readmissão do Paciente , Fatores Socioeconômicos , Infecção da Ferida Cirúrgica , Humanos , Infecção da Ferida Cirúrgica/epidemiologia , Criança , Adolescente , Masculino , Feminino , Pré-Escolar , Fatores de Risco , Procedimentos Cirúrgicos do Sistema Digestório/efeitos adversos , Readmissão do Paciente/estatística & dados numéricos , Estados Unidos/epidemiologia , Lactente , Estudos Retrospectivos
10.
bioRxiv ; 2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37808713

RESUMO

Messenger RNA (mRNA) recruitment to the 40S ribosomal subunit is mediated by eukaryotic initiation factor 4F (eIF4F). This complex includes 3 subunits: eIF4E (m 7 G cap binding protein), eIF4A (DEAD box helicase), and eIF4G. Mammalian eIF4G is a scaffold that coordinates the activities of eIF4E and eIF4A and provides a bridge to connect the mRNA and 40S ribosomal subunit through its interaction with eIF3. While the roles of many eIF4G binding domains are relatively clear, the precise function of RNA binding by eIF4G remains to be elucidated. In this work, we used an eIF4G-dependent translation assay to reveal that the RNA binding domain (eIF4G-RBD; amino acids 682-720) stimulates translation. This stimulating activity is observed when eIF4G is independently tethered to an internal region of the mRNA, suggesting that the eIF4G-RBD promotes translation by a mechanism that is independent of the m 7 G cap and mRNA tethering. Using a kinetic helicase assay, we show that the eIF4G-RBD has a minimal effect on eIF4A helicase activity, demonstrating that the eIF4G-RBD is not required to coordinate eIF4F-dependent duplex unwinding. Unexpectedly, native gel electrophoresis and fluorescence polarization assays reveal a previously unidentified direct interaction between eIF4G and the 40S subunit. Using binding assays, our data show that this 40S subunit interaction is separate from the previously characterized interaction between eIF4G and eIF3. Thus, our work reveals how eIF4F can bind to the 40S subunit using eIF3-dependent and eIF3-independent binding domains to promote translation initiation.

11.
J Clin Invest ; 133(15)2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37526080

RESUMO

Entry of antigen-specific T cells into human tumors is critical for immunotherapy, but the underlying mechanisms are poorly understood. Here, we combined high-dimensional spatial analyses with in vitro and in vivo modeling to study the mechanisms underlying immune infiltration in human multiple myeloma (MM) and its precursor monoclonal gammopathy of undetermined significance (MGUS). Clustered tumor growth was a feature of MM but not MGUS biopsies, and this growth pattern was reproduced in humanized mouse models. MM biopsies exhibited intralesional as well as spatial heterogeneity, with coexistence of T cell-rich and T cell-sparse regions and the presence of areas of T cell exclusion. In vitro studies demonstrated that T cell entry into MM clusters was regulated by agonistic signals and CD2-CD58 interactions. Upon adoptive transfer, antigen-specific T cells localized to the tumor site but required in situ DC-mediated antigen presentation for tumor entry. C-type lectin domain family 9 member A-positive (CLEC9A+) DCs appeared to mark portals of entry for gradients of T cell infiltration in MM biopsies, and their proximity to T cell factor 1-positive (TCF1+) T cells correlated with disease state and risk status. These data illustrate a role for tumor-associated DCs and in situ activation in promoting the infiltration of antigen-specific T cells in MM and provide insights into spatial alterations in tumor/immune cells with malignant evolution.


Assuntos
Mieloma Múltiplo , Lesões Pré-Cancerosas , Animais , Camundongos , Humanos , Mieloma Múltiplo/patologia , Linfócitos T , Lesões Pré-Cancerosas/patologia , Imunoterapia/métodos , Apresentação de Antígeno , Células Dendríticas
12.
Oncotarget ; 13: 490-504, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251496

RESUMO

Multiple myeloma (MM) is a hematological malignancy of plasma cells that remains incurable despite significant progress with myeloablative regimens and autologous stem cell transplantation for eligible patients and, more recently with T cell redirected immunotherapy. Recently, we reported that ex vivo virotherapy with oncolytic myxoma virus (MYXV) improved MM-free survival in an autologous-transplant Balb/c mouse model. Here, we tested the Vk*MYC transplantable C57BL/6 mouse MM model that more closely recapitulates human disease. In vitro, the murine bortezomib-resistant Vk12598 cell line is fully susceptible to MYXV infection. In vivo results demonstrate: (i) autologous bone marrow (BM) leukocytes armed ex vivo with MYXV exhibit moderate therapeutic effects against MM cells pre-seeded into recipient mice; (ii) Cyclophosphamide in combination with BM/MYXV delays the onset of myeloma in mice seeded with Vk12598 cells; (iii) BM/MYXV synergizes with the Smac-mimetics LCL161 and with immune checkpoint inhibitor α-PD-1 to control the progression of established MM in vivo, resulting in significant improvement of survival rates and decreased of tumor burden; (iv) Survivor mice from (ii) and (iii), when re-challenged with fresh Vk12598 cells, developed acquired anti-MM immunity. These results highlight the utility of autologous BM grafts armed ex vivo with oncolytic MYXV alone or in combination with chemotherapy/immunotherapy to treat drug-resistant MM in vivo.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Mieloma Múltiplo , Myxoma virus , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Medula Óssea , Bortezomib/farmacologia , Ciclofosfamida , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Inibidores de Checkpoint Imunológico , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica/métodos , Receptor de Morte Celular Programada 1 , Transplante Autólogo
13.
J Mol Evol ; 73(3-4): 134-52, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22009226

RESUMO

Proteins belonging to the Progestin and AdipoQ Receptor (PAQR) superfamily of membrane bound receptors are ubiquitously found in fungi. Nearly, all fungi possess two evolutionarily distinct paralogs of PAQR protein, which we have called the PQRA and PQRB subtypes. In the model fungus Saccharomyces cerevisiae, these subtypes are represented by the Izh2p and Izh3p proteins, respectively. S. cerevisiae also possesses two additional PQRA-type receptors called Izh1p and Izh4p that are restricted to other species within the "Saccharomyces complex". Izh2p has been the subject of several recent investigations and is of particular interest because it regulates fungal growth in response to proteins produced by plants and, as such, represents a new paradigm for interspecies communication. We demonstrate that IZH2 and IZH3 gene dosage affects resistance to polyene antifungal drugs. Moreover, we provide additional evidence that Izh2p and Izh3p negatively regulate fungal filamentation. These data suggest that agonists of these receptors might make antifungal therapeutics, either by inhibiting fungal development or by sensitizing fungi to the toxic effects of current antifungal therapies. This is particularly relevant for pathogenic fungi such as Candida glabrata that are closely related to S. cerevisiae and contain the same complement of PAQR receptors.


Assuntos
Hifas/metabolismo , Proteínas de Membrana/genética , Filogenia , Receptores de Superfície Celular/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Antifúngicos/farmacologia , Duplicação Cromossômica , Cromossomos Fúngicos/genética , Sequência Conservada , Farmacorresistência Fúngica , Evolução Molecular , Dosagem de Genes , Genoma Fúngico , Interações Hidrofóbicas e Hidrofílicas , Hifas/crescimento & desenvolvimento , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Nistatina/farmacologia , Fenótipo , Polienos/farmacologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos
14.
Methods Mol Biol ; 2225: 163-177, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33108662

RESUMO

Viruses engineered to express fluorescent proteins can be used with live-cell imaging techniques to monitor the progression of infection in real time. Here we describe a set of methods to track infection spreading from one cell population to another as well as to visualize transfer of virions between cells. This approach is extended to multiplexing with physiological readouts of cell death, which can be correlated with single-cell resolution to viral infection.


Assuntos
Rastreamento de Células/métodos , Engenharia Genética/métodos , Myxoma virus/genética , Imagem Óptica/métodos , Proteínas Virais/genética , Vírion/genética , Células A549 , Apoptose/genética , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células Jurkat , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Myxoma virus/crescimento & desenvolvimento , Myxoma virus/metabolismo , Imagem Óptica/estatística & dados numéricos , Proteínas Virais/metabolismo , Vírion/crescimento & desenvolvimento , Vírion/metabolismo , Replicação Viral/genética , Proteína Vermelha Fluorescente
15.
Mol Ther Oncolytics ; 22: 539-554, 2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34553039

RESUMO

Solid cancers that metastasize to the lungs represent a major therapeutic challenge. Current treatment paradigms for lung metastases consist of radiation therapy, chemotherapies, and surgical resection, but there is no single treatment or combination that is effective for all tumor types. To address this, oncolytic myxoma virus (MYXV) engineered to express human tumor necrosis factor (vMyx-hTNF) was tested after systemic administration in an immunocompetent mouse K7M2-Luc lung metastatic osteosarcoma model. Virus therapy efficacy against pre-seeded lung metastases was assessed after systemic infusion of either naked virus or ex vivo-loaded autologous bone marrow leukocytes or peripheral blood mononuclear cells (PBMCs). Results of this study showed that the PBMC pre-loaded strategy was the most effective at reducing tumor burden and increasing median survival time, but sequential intravenous multi-dosing with naked virus was comparably effective to a single infusion of PBMC-loaded virus. PBMC-loaded vMyx-hTNF also potentially synergized very effectively with immune checkpoint inhibitors anti-PD-1, anti-PD-L1, and anti-cytotoxic T lymphocyte associated protein 4 (CTLA-4). Finally, in addition to the pro-immune stimulation caused by unarmed MYXV, the TNF transgene of vMyx-hTNF further induced the unique expression of numerous additional cytokines associated with the innate and adaptive immune responses in this model. We conclude that systemic ex vivo virotherapy with TNF-α-armed MYXV represents a new potential strategy against lung metastatic cancers like osteosarcoma and can potentially act synergistically with established checkpoint immunotherapies.

16.
Methods Mol Biol ; 2058: 95-110, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31486033

RESUMO

Myxoma virus (MYXV) is a member of the Poxviridae family and the genus Leporipoxvirus. In nature MYXV tropism is restricted to lagomorphs, and is specifically pathogenic only for European rabbits (Oryctolagus cuniculus), in which this virus causes the lethal systemic disease called myxomatosis. Importantly, although MYXV cannot cause any disease pathology in humans, mice, or any other domestic animals other than rabbit, this virus can productively infect and kill a variety of human and murine cancer cells, from either primary sources or cultured cancer cell lines. Therefore, in the last decade, MYXV has emerged as a novel oncolytic virus against hematologic malignancies and various solid cancers. One novel aspect of MYXV virotherapy is a new systemic virus delivery strategy to cancer sites in the recipient, by which adsorption of the virus to isolated leukocytes is conducted prior to reinfusion of the virus-infected cells back into the recipient, via a procedure called ex vivo virotherapy (EVV, or simply EV2). The EV2 delivery strategy thus exploits the inherent migratory properties of leukocytes to ferry MYXV to tissue sites bearing cancer cells that are accessible to leukocyte chemotaxis. Here, we describe EV2 procedures with MYXV to systemically deliver the virus to sites of disseminated and/or metastatic cancer in situ via infected leukocytes derived from either bone marrow or peripheral blood.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Myxoma virus/genética , Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Animais , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Imunofenotipagem , Leucócitos Mononucleares/metabolismo , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/etiologia , Neoplasias/patologia , Terapia Viral Oncolítica/métodos , Transdução Genética , Carga Tumoral
17.
Mol Ther Oncolytics ; 18: 335-350, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32775618

RESUMO

Oncolytic viruses can target neoplasms, triggering oncolytic and immune effects. Their delivery to melanoma lesions remains challenging. Bone-marrow-derived mesenchymal stem cells (MSCs) were shown to be permissive for oncolytic myxoma virus (MYXV), allowing its transfer to melanoma cells, leading to their killing. Involvement of progeny virus was demonstrated in the transfer from MSCs to co-cultured melanoma cells. The inhibitory effect of virus on melanoma foci formation in murine lungs was revealed using melanoma cells previously co-cultured with MYXV-infected MSCs. Virus accumulation and persistence in lungs of lesion-bearing mice were shown following intravenous administration of MSC-shielded MYXV construct encoding luciferase. Therapy of experimentally induced lung melanoma in mice with interleukin (IL)-15-carrying MYXV construct delivered by MSCs led to marked regression of lesions and could increase survival. Elevated natural killer (NK) cell percentages in blood indicated robust innate responses against unshielded virus only. Lung infiltration by NK cells was followed by inflow of CD8+ T lymphocytes into melanoma lesions. Elevated expression of genes involved in adaptive immune response following oncolytic treatment was confirmed using RT-qPCR. No adverse pathological effects related to MSC-mediated oncolytic therapy with MYXV were observed. MSCs allow for safe and efficient ferrying of therapeutic MYXV to pulmonary melanoma foci triggering immune effects.

18.
Mol Ther Oncolytics ; 18: 171-188, 2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32695875

RESUMO

Multiple myeloma (MM) is a hematological malignancy of monoclonal plasma cells that remains incurable. Standard treatments for MM include myeloablative regimens and autologous cell transplantation for eligible patients. A major challenge of these treatments is the relapse of the disease due to residual MM in niches that become refractory to treatments. Therefore, novel therapies are needed in order to eliminate minimal residual disease (MRD). Recently, our laboratory reported that virotherapy with oncolytic myxoma virus (MYXV) improved MM-free survival in an allogeneic transplant mouse model. In this study, we demonstrate the capacity of donor autologous murine leukocytes, pre-armed with MYXV, to eliminate MRD in a BALB/c MM model. We report that MYXV-armed bone marrow (BM) carrier leukocytes are therapeutically superior to MYXV-armed peripheral blood mononuclear cells (PBMCs) or free virus. Importantly, when cured survivor mice were re-challenged with fresh myeloma cells, they developed immunity to the same MM that had comprised MRD. In vivo imaging demonstrated that autologous carrier cells armed with MYXV were very efficient at delivery of MYXV into the recipient tumor microenvironment. Finally, we demonstrate that treatment with MYXV activates the secretion of pro-immune molecules from the tumor bed. These results highlight the utility of exploiting autologous leukocytes to enhance tumor delivery of MYXV to treat MRD in vivo.

19.
Biochemistry ; 48(24): 5504-6, 2009 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-19453184

RESUMO

The progestin and AdipoQ receptor (PAQR) family of proteins comprises three distinct structural classes, each with seemingly different agonist specificities. For example, Class I receptors, like the human adiponectin receptors (AdipoR1 and AdipoR2), sense proteins with a particular three-dimensional fold, while Class II receptors are nonclassical membrane receptors for the steroid hormone progesterone. Using a previously developed heterologous expression system to study PAQR receptor activity, we demonstrate that human PAQRs from all three classes are antagonized by both 1(S),2(R)-d-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol, a ceramidase inhibitor, and TNFalpha, a homologue of adiponectin that functions antagonistically to both adiponectin and progesterone in human cells.


Assuntos
Ceramidases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Miristatos/farmacologia , Propanolaminas/farmacologia , Receptores de Adiponectina/antagonistas & inibidores , Receptores de Progesterona/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Ceramidases/metabolismo , Humanos , Modelos Biológicos , Receptores de Adiponectina/metabolismo , Receptores de Progesterona/metabolismo
20.
Mol Pharmacol ; 75(4): 866-75, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19066337

RESUMO

The Izh2p protein from Saccharomyces cerevisiae belongs to the newly characterized progestin and adipoQ receptor (PAQR) superfamily of receptors whose mechanism of signal transduction is still unknown. Izh2p functions as a receptor for the plant PR-5 defensin osmotin and has pleiotropic effects on cellular biochemistry. One example of this pleiotropy is the Izh2p-dependent repression of FET3, a gene involved in iron-uptake. Although the physiological purpose of FET3 repression by Izh2p is a matter of speculation, it provides a reporter with which to probe the mechanism of signal transduction by this novel class of receptor. Receptors in the PAQR family share sequence similarity with enzymes involved in ceramide metabolism, which led to the hypothesis that sphingolipids are involved in Izh2p-dependent signaling. In this study, we demonstrate that drugs affecting sphingolipid metabolism, such as d-erythro-MAPP and myriocin, inhibit the effect of Izh2p on FET3. We also show that Izh2p causes an increase in steady-state levels of sphingoid base. Moreover, we show that Izh2p-independent increases in sphingoid bases recapitulate the effect of Izh2p on FET3. Finally, our data indicate that the Pkh1p and Pkh2p sphingoid base-sensing kinases are essential components of the Izh2p-dependent signaling pathway. In conclusion, our data indicate that Izh2p produces sphingoid bases and that these bioactive lipids probably function as the second messenger responsible for the effect of Izh2p on FET3.


Assuntos
Proteínas de Saccharomyces cerevisiae/fisiologia , Sistemas do Segundo Mensageiro/fisiologia , Esfingolipídeos/fisiologia , Ceramidas/biossíntese , Ceramidas/química , Ceramidas/fisiologia , Ceruloplasmina/antagonistas & inibidores , Ceruloplasmina/genética , Ceruloplasmina/fisiologia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Modelos Genéticos , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sistemas do Segundo Mensageiro/genética , Esfingolipídeos/química , Esfingolipídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa