Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Cancer Biol ; 86(Pt 3): 971-980, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34033895

RESUMEN

Anti-PD-1 and oncolytic viruses (OVs) have non-overlapping anti-tumor mechanisms, since each agent works at different steps of the cancer-immunity cycle. Evidence suggests that OVs improve therapeutic responses to anti-PD-1 therapy by reversing immunosuppressive factors, increasing the number and diversity of infiltrating lymphocytes, and promoting PD-L1 expression in both injected and non-injected tumors. Many studies in preclinical models suggest that the timing of anti-PD-1 administration influences the therapeutic success of the combination therapy (anti-PD-1 + OV). Therefore, determining the appropriate sequencing of agents is of critical importance to designing a rationale OV-based combinational clinical trial. Currently, the combination of anti-PD-1 and OVs are being delivered using various schedules, and we have classified the timing of administration of anti-PD-1 and OVs into five categories: (i) anti-PD-1 lead-in → OV; (ii) concurrent administration; (iii) OV lead-in → anti-PD-1; (iv) concurrent therapy lead-in → anti-PD-1; and (v) OV lead-in → concurrent therapy. Based on the reported preclinical and clinical literature, the most promising treatment strategy to date is hypothesized to be OV lead-in → concurrent therapy. In the OV lead-in → concurrent therapy approach, initial OV treatment results in T cell priming and infiltration into tumors and an immunologically hot tumor microenvironment (TME), which can be counterbalanced by engagement of PD-L1 to PD-1 receptor on immune cells, leading to T cell exhaustion. Therefore, after initial OV therapy, concurrent use of both OV and anti-PD-1 is critical through which OV maintains T cell priming and an immunologically hot TME, whereas PD-1 blockade helps to overcome PD-L1/PD-1-mediated T cell exhaustion. It is important to note that the hypothetical conclusion drawn in this review is based on thorough literature review on current understanding of OV + anti-PD-1 combination therapies and rhythm of treatment-induced cancer-immunity cycle. A variety of confounding factors such as tumor types, OV types, presence or absence of cytokine transgenes carried by an OV, timing of treatment initiation, varying dosages and treatment frequencies/duration of OV and anti-PD-1, etc. may affect the validity of our conclusion that will need to be further examined by future research (such as side-by-side comparative studies using all five treatment schedules in a given tumor model).


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Viroterapia Oncolítica/métodos , Antígeno B7-H1/genética , Virus Oncolíticos/genética , Microambiente Tumoral , Neoplasias/terapia
2.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29793956

RESUMEN

Oncolytic viruses, including herpes simplex viruses (HSVs), are a new class of cancer therapeutic engineered to infect and kill cancer cells while sparing normal tissue. To ensure that oncolytic HSV (oHSV) is safe in the brain, all oHSVs in clinical trial for glioma lack the γ34.5 genes responsible for neurovirulence. However, loss of γ34.5 attenuates growth in cancer cells. Glioblastoma (GBM) is a lethal brain tumor that is heterogeneous and contains a subpopulation of cancer stem cells, termed GBM stem-like cells (GSCs), that likely promote tumor progression and recurrence. GSCs and matched serum-cultured GBM cells (ScGCs), representative of bulk or differentiated tumor cells, were isolated from the same patient tumor specimens. ScGCs are permissive to replication and cell killing by oHSV with deletion of the γ34.5 genes (γ34.5- oHSV), while patient-matched GSCs were not, implying an underlying biological difference between stem and bulk cancer cells. GSCs specifically restrict the synthesis of HSV-1 true late (TL) proteins, without affecting viral DNA replication or transcription of TL genes. A global shutoff of cellular protein synthesis also occurs late after γ34.5- oHSV infection of GSCs but does not affect the synthesis of early and leaky late viral proteins. Levels of phosphorylated eIF2α and eIF4E do not correlate with cell permissivity. Expression of Us11 in GSCs rescues replication of γ34.5- oHSV. The difference in degrees of permissivity between GSCs and ScGCs to γ34.5- oHSV illustrates a selective translational regulatory pathway in GSCs that may be operative in other stem-like cells and has implications for creating oHSVs.IMPORTANCE Herpes simplex virus (HSV) can be genetically engineered to endow cancer-selective replication and oncolytic activity. γ34.5, a key neurovirulence gene, has been deleted in all oncolytic HSVs in clinical trial for glioma. Glioblastoma stem-like cells (GSCs) are a subpopulation of tumor cells thought to drive tumor heterogeneity and therapeutic resistance. GSCs are nonpermissive for γ34.5- HSV, while non-stem-like cancer cells from the same patient tumors are permissive. GSCs restrict true late protein synthesis, despite normal viral DNA replication and transcription of all kinetic classes. This is specific for true late translation as early and leaky late transcripts are translated late in infection, notwithstanding shutoff of cellular protein synthesis. Expression of Us11 in GSCs rescues the replication of γ34.5- HSV. We have identified a cell type-specific innate response to HSV-1 that limits oncolytic activity in glioblastoma.


Asunto(s)
Neoplasias Encefálicas/virología , Eliminación de Gen , Glioblastoma/virología , Células Madre Neoplásicas/virología , Simplexvirus/fisiología , Proteínas Virales/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Chlorocebus aethiops , Glioblastoma/metabolismo , Glioblastoma/terapia , Herpes Simple/genética , Células Madre Neoplásicas/metabolismo , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Proteínas de Unión al ARN/metabolismo , Simplexvirus/genética , Células Vero , Proteínas Virales/metabolismo , Replicación Viral
3.
Mol Ther ; 24(8): 1435-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27357626

RESUMEN

Adoptive T-cell transfer is a promising treatment approach for metastatic cancer, but efficacy in solid tumors has only been achieved with toxic pre- and postconditioning regimens. Thus, adoptive T-cell therapies would benefit from complementary modalities that enable their full potential without excessive toxicity. We aimed to improve the efficacy and safety of adoptive T-cell transfer by using adenoviral vectors for direct delivery of immunomodulatory murine cytokines into B16.OVA melanoma tumors with concomitant T-cell receptor transgenic OT-I T-cell transfer. Armed adenoviruses expressed high local and low systemic levels of cytokine when injected into B16.OVA tumors, suggesting safety of virus-mediated cytokine delivery. Antitumor efficacy was significantly enhanced with adenoviruses coding for murine interleukin-2 (mIL-2) and tumor necrosis factor-α (mTNFα) when compared with T-cell transfer alone or viruses alone. Further improvement in efficacy was achieved with a triple combination of mIL-2, mTNFα, and OT-I T-cells. Mechanistic studies suggest that mIL-2 has an important role in activating T-cells at the tumor, while mTNFα induces chemokine expression. Furthermore, adenovirus treatments enhanced tumor-infiltration of OT-I T-cells as demonstrated by SPECT/CT imaging of (111)In-labeled cells. Our results suggest the utility of cytokine-coding adenoviruses for improving the efficacy of adoptive T-cell therapies.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Inmunoterapia Adoptiva , Interleucina-2/genética , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Factor de Necrosis Tumoral alfa/genética , Animales , Antígeno B7-H1/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Modelos Animales de Enfermedad , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Huésped Inmunocomprometido , Inyecciones Intralesiones , Interleucina-2/metabolismo , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma Experimental/diagnóstico , Melanoma Experimental/terapia , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Neurooncol ; 121(1): 91-100, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25213669

RESUMEN

Anti-angiogenic therapy is a promising therapeutic strategy for the highly vascular and malignant brain tumor, glioblastoma (GBM), although current clinical trials have failed to demonstrate an extension in overall survival. The small molecule tyrosine kinase inhibitor axitinib that targets vascular endothelial growth factor receptor, potently inhibits angiogenesis and has single-agent clinical activity in non-small cell lung, thyroid, and advanced renal cell cancer. Here we show that axitinib exerts direct cytotoxic activity against a number of patient-derived GBM stem cell (GSCs) and an endothelial cell line, and inhibits endothelial tube formation in vitro. Axitinib treatment of mice bearing hypervascular intracranial tumors generated from human U87 glioma cells, MGG4 GSCs and mouse 005 GSCs significantly extended survival that was associated with decreases in tumor-associated vascularity. We thus show for the first time the anti-angiogenic effect and survival prolongation provided by systemic single agent treatment with axitinib in preclinical orthotopic GBM models including clinically relevant GSC models. These results support further investigation of axitinib as an anti-angiogenic agent for GBM.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Imidazoles/farmacología , Indazoles/farmacología , Animales , Apoptosis/efectos de los fármacos , Axitinib , Neoplasias Encefálicas/fisiopatología , Línea Celular Tumoral , Femenino , Glioblastoma/fisiopatología , Humanos , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/fisiología , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Distribución Aleatoria , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Análisis de Supervivencia
5.
Drugs Future ; 40(11): 739-749, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26855472

RESUMEN

Malignant gliomas are the most common type of primary malignant brain tumor with no effective treatments. Current conventional therapies (surgical resection, radiation therapy, temozolomide (TMZ), and bevacizumab administration) typically fail to eradicate the tumors resulting in the recurrence of treatment-resistant tumors. Therefore, novel approaches are needed to improve therapeutic outcomes. Oncolytic viruses (OVs) are excellent candidates as a more effective therapeutic strategy for aggressive cancers like malignant gliomas since OVs have a natural preference or have been genetically engineered to selectively replicate in and kill cancer cells. OVs have been used in numerous preclinical studies in malignant glioma, and a large number of clinical trials using OVs have been completed or are underway that have demonstrated safety, as well as provided indications of effective antiglioma activity. In this review, we will focus on those OVs that have been used in clinical trials for the treatment of malignant gliomas (herpes simplex virus, adenovirus, parvovirus, reovirus, poliovirus, Newcastle disease virus, measles virus, and retrovirus) and OVs examined preclinically (vesicular stomatitis virus and myxoma virus), and describe how these agents are being used.

6.
Stem Cell Rev Rep ; 20(1): 3-24, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37861969

RESUMEN

Tumorigenic Cancer Stem Cells (CSCs), often called tumor-initiating cells (TICs), represent a unique subset of cells within the tumor milieu. They stand apart from the bulk of tumor cells due to their exceptional self-renewal, metastatic, and differentiation capabilities. Despite significant progress in classifying CSCs, these cells remain notably resilient to conventional radiotherapy and chemotherapy, contributing to cancer recurrence. In this review, our objective is to explore novel avenues of research that delve into the distinctive characteristics of CSCs within their surrounding tumor microenvironment (TME). We will start with an overview of the defining features of CSCs and then delve into their intricate interactions with cells from the lymphoid lineage, namely T cells, B cells, and natural killer (NK) cells. Furthermore, we will discuss their dynamic interplay with myeloid lineage cells, including macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs). Moreover, we will illuminate the crosstalk between CSCs and cells of mesenchymal origin, specifically fibroblasts, adipocytes, and endothelial cells. Subsequently, we will underscore the pivotal role of CSCs within the context of the tumor-associated extracellular matrix (ECM). Finally, we will highlight pre-clinical and clinical studies that target CSCs within the intricate landscape of the TME, including CAR-T therapy, oncolytic viruses, and CSC-vaccines, with the ultimate goal of uncovering novel avenues for CSC-based cancer immunotherapy.


Asunto(s)
Células Endoteliales , Recurrencia Local de Neoplasia , Humanos , Recurrencia Local de Neoplasia/patología , Linfocitos T , Inmunoterapia , Células Madre Neoplásicas/patología , Microambiente Tumoral
7.
J Immunother Cancer ; 12(5)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38821716

RESUMEN

Cytokines are small proteins that regulate the growth and functional activity of immune cells, and several have been approved for cancer therapy. Oncolytic viruses are agents that mediate antitumor activity by directly killing tumor cells and inducing immune responses. Talimogene laherparepvec is an oncolytic herpes simplex virus type 1 (oHSV), approved for the treatment of recurrent melanoma, and the virus encodes the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). A significant advantage of oncolytic viruses is the ability to deliver therapeutic payloads to the tumor site that can help drive antitumor immunity. While cytokines are especially interesting as payloads, the optimal cytokine(s) used in oncolytic viruses remains controversial. In this review, we highlight preliminary data with several cytokines and chemokines, including GM-CSF, interleukin 12, FMS-like tyrosine kinase 3 ligand, tumor necrosis factor α, interleukin 2, interleukin 15, interleukin 18, chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 5, chemokine (C-X-C motif) ligand 4, or their combinations, and show how these payloads can further enhance the antitumor immunity of oHSV. A better understanding of cytokine delivery by oHSV can help improve clinical benefit from oncolytic virus immunotherapy in patients with cancer.


Asunto(s)
Citocinas , Inmunoterapia , Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Virus Oncolíticos/genética , Citocinas/metabolismo , Inmunoterapia/métodos , Neoplasias/terapia , Neoplasias/inmunología , Animales , Simplexvirus/inmunología , Simplexvirus/genética , Herpesvirus Humano 1/inmunología
8.
J Immunother Cancer ; 12(4)2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599661

RESUMEN

BACKGROUND: Glioblastoma (GBM), a highly immunosuppressive and often fatal primary brain tumor, lacks effective treatment options. GBMs contain a subpopulation of GBM stem-like cells (GSCs) that play a central role in tumor initiation, progression, and treatment resistance. Oncolytic viruses, especially oncolytic herpes simplex virus (oHSV), replicate selectively in cancer cells and trigger antitumor immunity-a phenomenon termed the "in situ vaccine" effect. Although talimogene laherparepvec (T-VEC), an oHSV armed with granulocyte macrophage-colony stimulating factor (GM-CSF), is Food and Drug Administration (FDA)-approved for melanoma, its use in patients with GBM has not been reported. Interleukin 2 (IL-2) is another established immunotherapy that stimulates T cell growth and orchestrates antitumor responses. IL-2 is FDA-approved for melanoma and renal cell carcinoma but has not been widely evaluated in GBM, and IL-2 treatment is limited by its short half-life, minimal tumor accumulation, and significant systemic toxicity. We hypothesize that local intratumoral expression of IL-2 by an oHSV would avoid the systemic IL-2-related therapeutic drawbacks while simultaneously producing beneficial antitumor immunity. METHODS: We developed G47Δ-mIL2 (an oHSV expressing IL-2) using the flip-flop HSV BAC system to deliver IL-2 locally within the tumor microenvironment (TME). We then tested its efficacy in orthotopic mouse GBM models (005 GSC, CT-2A, and GL261) and evaluated immune profiles in the treated tumors and spleens by flow cytometry and immunohistochemistry. RESULTS: G47Δ-mIL2 significantly prolonged median survival without any observable systemic IL-2-related toxicity in the 005 and CT-2A models but not in the GL261 model due to the non-permissive nature of GL261 cells to HSV infection. The therapeutic activity of G47Δ-mIL2 in the 005 GBM model was associated with increased intratumoral infiltration of CD8+ T cells, critically dependent on the release of IL-2 within the TME, and CD4+ T cells as their depletion completely abrogated therapeutic efficacy. The use of anti-PD-1 immune checkpoint blockade did not improve the therapeutic outcome of G47Δ-mIL2. CONCLUSIONS: Our findings illustrate that G47Δ-mIL2 is efficacious, stimulates antitumor immunity against orthotopic GBM, and may also target GSC. OHSV expressing IL-2 may represent an agent that merits further exploration in patients with GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Herpes Simple , Viroterapia Oncolítica , Animales , Humanos , Ratones , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos , Glioblastoma/patología , Herpesvirus Humano 2 , Interleucina-2/uso terapéutico , Melanoma/terapia , Microambiente Tumoral , Estados Unidos
9.
J Drug Target ; 31(5): 1-15, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37026714

RESUMEN

Pulmonary arterial hypertension (PAH) is a rare pulmonary vascular disorder, wherein mean systemic arterial pressure (mPAP) becomes abnormally high because of aberrant changes in various proliferative and inflammatory signalling pathways of pulmonary arterial cells. Currently used anti-PAH drugs chiefly target the vasodilatory and vasoconstrictive pathways. However, an imbalance between bone morphogenetic protein receptor type II (BMPRII) and transforming growth factor beta (TGF-ß) pathways is also implicated in PAH predisposition and pathogenesis. Compared to currently used PAH drugs, various biologics have shown promise as PAH therapeutics that elicit their therapeutic actions akin to endogenous proteins. Biologics that have thus far been explored as PAH therapeutics include monoclonal antibodies, recombinant proteins, engineered cells, and nucleic acids. Because of their similarity with naturally occurring proteins and high binding affinity, biologics are more potent and effective and produce fewer side effects when compared with small molecule drugs. However, biologics also suffer from the limitations of producing immunogenic adverse effects. This review describes various emerging and promising biologics targeting the proliferative/apoptotic and vasodilatory pathways involved in PAH pathogenesis. Here, we have discussed sotatercept, a TGF-ß ligand trap, which is reported to reverse vascular remodelling and reduce PVR with an improved 6-minute walk distance (6-MWDT). We also elaborated on other biologics including BMP9 ligand and anti-gremlin1 antibody, anti-OPG antibody, and getagozumab monoclonal antibody and cell-based therapies. Overall, recent literature suggests that biologics hold excellent promise as a safe and effective alternative to currently used PAH therapeutics.


Asunto(s)
Productos Biológicos , Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/patología , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Ligandos , Factor de Crecimiento Transformador beta/metabolismo
10.
Expert Opin Drug Deliv ; 20(3): 335-348, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36720629

RESUMEN

INTRODUCTION: Drugs delivered via the lungs are predominantly used to treat various respiratory disorders, including asthma, chronic obstructive pulmonary diseases, respiratory tract infections and lung cancers, and pulmonary vascular diseases such as pulmonary hypertension. To treat respiratory diseases, targeted, modified or controlled release inhalation formulations are desirable for improved patient compliance and superior therapeutic outcome. AREAS COVERED: This review summarizes the important factors that have an impact on the inhalable modified release formulation approaches with a focus toward various formulation strategies, including dissolution rate-controlled systems, drug complexes, site-specific delivery, drug-polymer conjugates, and drug-polymer matrix systems, lipid matrix particles, nanosystems, and formulations that can bypass clearance via mucociliary system and alveolar macrophages. EXPERT OPINION: Inhaled modified release formulations can potentially reduce dosing frequency by extending drug's residence time in the lungs. However, inhalable modified or controlled release drug delivery systems remain unexplored and underdeveloped from the commercialization perspective. This review paper addresses the current state-of-the-art of inhaled controlled release formulations, elaborates on the avenues for developing newer technologies for formulating various drugs with tailored release profiles after inhalational delivery and explains the challenges associated with translational feasibility of modified release inhalable formulations.


Asunto(s)
Asma , Enfermedades Respiratorias , Humanos , Preparaciones de Acción Retardada , Sistemas de Liberación de Medicamentos , Pulmón , Asma/tratamiento farmacológico , Administración por Inhalación , Enfermedades Respiratorias/tratamiento farmacológico
11.
Hum Gene Ther ; 34(17-18): 878-895, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37578106

RESUMEN

Interleukin 2 (IL-2) plays a crucial role in T cell growth and survival, enhancing the cytotoxic activity of natural killer and cytotoxic T cells and thus functioning as a versatile master proinflammatory anticancer cytokine. The FDA has approved IL-2 cytokine therapy for the treatment of metastatic melanoma and metastatic renal cell carcinoma. However, IL-2 therapy has significant constraints, including a short serum half-life, low tumor accumulation, and life-threatening toxicities associated with high doses. Oncolytic viruses (OVs) offer a promising option for cancer immunotherapy, selectively targeting and destroying cancer cells while sparing healthy cells. Numerous studies have demonstrated the successful delivery of IL-2 to the tumor microenvironment without compromising safety using OVs such as vaccinia, Sendai, parvo, Newcastle disease, tanapox, and adenoviruses. Additionally, by engineering OVs to coexpress IL-2 with other anticancer transgenes, the immune properties of IL-2 can be further enhanced. Preclinical and clinical studies have shown promising antitumor effects of IL-2-expressing viral vectors, either alone or in combination with other anticancer therapies. This review summarizes the therapeutic potential of IL-2-expressing viral vectors and their antitumor mechanisms of action.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Humanos , Interleucina-2/genética , Interleucina-2/uso terapéutico , Citocinas , Virus Oncolíticos/genética , Inmunoterapia , Microambiente Tumoral
12.
J Gen Virol ; 93(Pt 7): 1548-1555, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492916

RESUMEN

Porcine circovirus 2 (PCV2) is the causative agent of porcine circovirus-associated diseases in pigs. Previously, it was demonstrated that mAbs 16G12, 38C1, 63H3 and 94H8 directed against the PCV2 capsid protein recognize PCV2 strains Stoon-1010 (PCV2a), 48285 (PCV2b), 1121 (PCV2a), 1147 (PCV2b) and II9F (PCV2b), but only neutralize Stoon-1010 and 48285. This points to the existence of two distinct PCV2 neutralization phenotypes: phenotype α (mAb recognition with neutralization; Stoon-1010 and 48285) and phenotype ß (mAb recognition without neutralization; 1121, 1147 and II9F). In the present study, amino acids that are important in determining the neutralization phenotype were identified in the capsid. Mutation of T at position 190 to A in strain 48285 (phenotype α) resulted in a capsid resembling that of strain 1147 (phenotype ß) and caused a loss of neutralization (switch from α to ß). Mutations of P at position 151 to T and A at position 190 to T in strain II9F (phenotype ß) resulted in a capsid resembling that of strain 48285 (phenotype α) and gave a gain of neutralization (switch from ß to α). Mutations of T at position 131 to P and of E at position 191 to R in Stoon-1010 (phenotype α) changed the capsid into that of 1121 (phenotype ß) and reduced neutralization (switch from α to ß). This study demonstrated that single amino acid changes in the capsid result in a phenotypic switch from α to ß or ß to α.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Circovirus/genética , Circovirus/inmunología , Mutación Missense , Sustitución de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Pruebas de Neutralización
13.
Virus Genes ; 44(2): 258-61, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22116682

RESUMEN

Porcine circovirus 2 (PCV2) is a small circular single-stranded DNA virus that causes postweaning multisystemic wasting syndrome in pigs. We obtained an infectious clone, derived from PCV2 strain 1147 (size 1,767 bp), with a 60 bp insertion in the cap-rep intergenic region at the 3' end of the rep. In vitro propagation and sequencing of virus showed mixed sequence for the cap-rep intergenic region. Cloning of this region of virus from the passages 2 and 20 showed deletions of various size of the 60 bp insertion. Viruses from the passage 20 showed deletions of the 60 bp insertion and had extra (+1 to +5) or less (-1 or -2) bp in the intergenic region as compared to 1,147. These findings suggest that insertion of a 60-bp DNA sequence at the 3' end of the rep is unstable in vitro. This finding can have implications for the genetic engineering of PCV2.


Asunto(s)
Circovirus/crecimiento & desarrollo , Circovirus/genética , Inestabilidad Genómica , Mutagénesis Insercional , Animales , ADN Viral/química , ADN Viral/genética , Ingeniería Genética/métodos , Análisis de Secuencia de ADN , Eliminación de Secuencia , Pase Seriado , Porcinos , Virología/métodos
15.
Biosensors (Basel) ; 12(4)2022 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-35448246

RESUMEN

3D printing (3DP) can serve not only as an excellent platform for producing solid dosage forms tailored to individualized dosing regimens but can also be used as a tool for creating a suitable 3D model for drug screening, sensing, testing and organ-on-chip applications. Several new technologies have been developed to convert the conventional dosing regimen into personalized medicine for the past decade. With the approval of Spritam, the first pharmaceutical formulation produced by 3DP technology, this technology has caught the attention of pharmaceutical researchers worldwide. Consistent efforts are being made to improvise the process and mitigate other shortcomings such as restricted excipient choice, time constraints, industrial production constraints, and overall cost. The objective of this review is to provide an overview of the 3DP process, its types, types of material used, and the pros and cons of each technique in the application of not only creating solid dosage forms but also producing a 3D model for sensing, testing, and screening of the substances. The application of producing a model for the biosensing and screening of drugs besides the creation of the drug itself, offers a complete loop of application for 3DP in pharmaceutics.


Asunto(s)
Medicina de Precisión , Impresión Tridimensional , Composición de Medicamentos , Evaluación Preclínica de Medicamentos
16.
Micromachines (Basel) ; 13(8)2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-36014279

RESUMEN

We present a robust, low-cost fabrication method for implementation in multilayer soft photolithography to create a PDMS microfluidic chip with features possessing multiple height levels. This fabrication method requires neither a cleanroom facility nor an expensive UV exposure machine. The central part of the method stays on the alignment of numerous PDMS slabs on a wafer-scale instead of applying an alignment for a photomask positioned right above a prior exposure layer using a sophisticated mask aligner. We used a manual XYZR stage attached to a vacuum tweezer to manipulate the top PDMS slab. The bottom PDMS slab sat on a rotational stage to conveniently align with the top part. The movement of the two slabs was observed by a monocular scope with a coaxial light source. As an illustration of the potential of this system for fast and low-cost multilayer microfluidic device production, we demonstrate the microfabrication of a 3D microfluidic chaotic mixer. A discussion on another alternative method for the fabrication of multiple height levels is also presented, namely the micromilling approach.

17.
Biosensors (Basel) ; 12(8)2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36004984

RESUMEN

This paper shows both experimental and in-depth theoretical studies (including simulations and analytical solutions) on a microfluidic platform to optimize its design and use for 3D multicellular co-culture applications, e.g., creating a tissue-on-chip model for investigating diseases such as pulmonary arterial hypertension (PAH). A tissue microfluidic chip usually has more than two channels to seed cells and supply media. These channels are often separated by barriers made of micro-posts. The optimization for the structures of these micro-posts and their spacing distances is not considered previously, especially for the aspects of rapid and cost-efficient fabrication toward scaling up and commercialization. Our experimental and theoretical (COMSOL simulations and analytical solutions) results showed the followings: (i) The cell seeding was performed successfully for this platform when the pressure drops across the two posts were significantly larger than those across the channel width. The circular posts can be used in the position of hexagonal or other shapes. (ii) In this work, circular posts are fabricated and used for the first time. They offer an excellent barrier effect, i.e., prevent the liquid and gel from migrating from one channel to another. (iii) As for rapid and cost-efficient production, our computer-aided manufacturing (CAM) simulation confirms that circular-post fabrication is much easier and more rapid than hexagonal posts when utilizing micro-machining techniques, e.g., micro-milling for creating the master mold, i.e., the shim for polymer injection molding. The findings open up a possibility for rapid, cost-efficient, large-scale fabrication of the tissue chips using micro-milling instead of expensive clean-room (soft) lithography techniques, hence enhancing the production of biochips via thermoplastic polymer injection molding and realizing commercialization.


Asunto(s)
Dispositivos Laboratorio en un Chip , Microfluídica , Técnicas de Cocultivo , Microfluídica/métodos , Polímeros/química , Impresión/métodos
18.
Micromachines (Basel) ; 13(9)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36144106

RESUMEN

Pulmonary arterial hypertension (PAH) is a rare pulmonary vascular disease that affects people of all ethnic origins and age groups including newborns. In PAH, pulmonary arteries and arterioles undergo a series of pathological changes including remodeling of the entire pulmonary vasculatures and extracellular matrices, mis-localized growth of pulmonary arterial cells, and development of glomeruloid-like lesions called plexiform lesions. Traditionally, various animal and cellular models have been used to understand PAH pathophysiology, investigate sex-disparity in PAH and monitor therapeutic efficacy of PAH medications. However, traditional models can only partially capture various pathological features of PAH, and they are not adaptable to combinatorial study design for deciphering intricately intertwined complex cellular processes implicated in PAH pathogenesis. While many microfluidic chip-based models are currently available for major diseases, no such disease-on-a-device model is available for PAH, an under investigated disease. In the absence of any chip-based models of PAH, we recently proposed a five-channel polydimethylsiloxane (PDMS)-based microfluidic device that can emulate major pathological features of PAH. However, our proposed model can make a bigger impact on the PAH field only when the larger scientific community engaged in PAH research can fabricate the device and develop the model in their laboratory settings. With this goal in mind, in this study, we have described the detailed methodologies for fabrication and development of the PAH chip model including a thorough explanation of scientific principles for various steps for chip fabrication, a detailed list of reagents, tools and equipment along with their source and catalogue numbers, description of laboratory setup, and cautionary notes. Finally, we explained the methodologies for on-chip cell seeding and application of this model for studying PAH pathophysiology. We believe investigators with little or no training in microfluidic chip fabrication can fabricate this eminently novel PAH-on-a-chip model. As such, this study will have a far-reaching impact on understanding PAH pathophysiology, unravelling the biological mystery associated with sexual dimorphism in PAH, and developing PAH therapy based on patient sex and age.

19.
Microb Pathog ; 51(3): 194-202, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21511026

RESUMEN

Reproductive failure due to porcine reproductive and respiratory syndrome virus (PRRSV) is characterized by late-term abortions, early farrowing and an increase of dead and mummified fetuses and weak-born piglets. The mechanism of PRRSV-induced reproductive failure is poorly understood. Human pregnancies, complicated by some pathogens leading to reproductive disorders exhibit increased apoptosis in the fetal membranes. Because PRRSV-target cells are present in endometrium/fetal placentas from healthy sows and PRRSV-infected macrophages in other organs die by apoptosis, we hypothesized that PRRSV can replicate and induce apoptosis in the fetal implantation sites at the last stage of gestation. In the present study, identification, localization and quantification of the PRRSV-positive and apoptotic cells were performed in the fetal implantation sites. Three dams were inoculated intranasally with 10(5) TCID(50) PRRSV 07V063 at 90 days of gestation and sampled at 10 days post-inoculation. Two non-inoculated dams that were euthanized at 100 days of gestation served as control animals. Inoculation of the dams resulted in a viremia that lasted until the end of the study. Transplacental PRRSV spread was detected in all inoculated dams. Using immunofluorescence staining, single PRRSV-positive cells were found in the endometrial connective tissues adjacent to both PRRSV-positive and PRRSV-negative fetuses. In the fetal placental mesenchyme of the PRRSV-positive fetuses, infected cells were more abundant and spread focally. Double staining showed that all PRRSV-positive cells in the fetal implantation sites were positive for sialoadhesin and CD163. Apoptotic cells (TUNEL+) were detected in endometrium and fetal placentas of both non- and PRRSV-inoculated dams. The number of apoptotic cells was significantly higher in PRRSV-positive endometrium/fetal placentas. PRRSV caused apoptosis in infected cells since 20-61% of PRRSV-positive cells were apoptotic and in surrounding cells since 43-91% of the apoptotic cells were virus-negative. The main conclusion obtained from the present study is that PRRSV replicates in the fetal implantation sites and causes apoptosis in infected macrophages and surrounding cells at the last stage of gestation. The possible mode of PRRSV replication in the fetal implantation sites and the events that might contribute to the reproductive disorders are discussed.


Asunto(s)
Apoptosis , Síndrome Respiratorio y de la Reproducción Porcina/patología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Animales , Antígenos CD/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Endometrio/patología , Endometrio/virología , Femenino , Intercambio Materno-Fetal , Glicoproteínas de Membrana/análisis , Placenta/patología , Placenta/virología , Embarazo , Receptores de Superficie Celular/análisis , Receptores Inmunológicos/análisis , Lectina 1 Similar a Ig de Unión al Ácido Siálico , Porcinos
20.
BMC Vet Res ; 7: 64, 2011 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-22018436

RESUMEN

BACKGROUND: Porcine circovirus type 1 (PCV1) has been described as a non-cytopathic contaminant of the PK-15 cell line. Several experimental infections with PCV1 failed to reproduce disease in pigs. Therefore, PCV1 is generally accepted as non-pathogenic to pigs. To our knowledge, nothing is known about the outcome of PCV1 infections in porcine foetuses. This was examined in the present study. RESULTS: Nine foetuses from three sows were inoculated at 55 days of gestation: three with 10(4.3) TCID(50) of the PCV1 cell culture strain ATCC-CCL33, three with 10(4.3) TCID(50) of the PCV1 field strain 3384 and three with cell culture medium (mock-inoculated). At 21 days post-inoculation, all 6 PCV1-inoculated and all 3 mock-inoculated foetuses had a normal external appearance. Microscopic lesions characterized by severe haemorrhages were observed in the lungs of two foetuses inoculated with CCL33. High PCV1 titres (up to 10(4.7) TCID(50)/g tissue) were found in the lungs of the CCL33-inoculated foetuses. All other organs of the CCL33-inoculated foetuses and all the organs of the 3384-inoculated foetuses were negative (< 10(1.7) TCID(50)/g tissue) by virus titration. PCV1-positive cells (up to 121 cells/10 mm(2) in CCL33-inoculated foetuses and up to 13 cells/10 mm(2) in 3384-inoculated foetuses) were found in the heart, lungs, spleen, liver, thymus and tonsils. PCR and DNA sequencing of Rep recovered CCL33 or 3384 sequences from CCL33- or 3384-inoculated foetuses, respectively. CONCLUSIONS: From this study, it can be concluded that cell culture PCV1 can replicate efficiently and produce pathology in the lungs of porcine foetuses inoculated at 55 days of foetal life.


Asunto(s)
Infecciones por Circoviridae/veterinaria , Circovirus , Enfermedades Fetales/veterinaria , Enfermedades de los Porcinos/virología , Animales , Infecciones por Circoviridae/virología , Circovirus/genética , Femenino , Enfermedades Fetales/virología , Técnica del Anticuerpo Fluorescente/veterinaria , Genes Virales/genética , Pulmón/virología , Embarazo , Porcinos/virología , Enfermedades de los Porcinos/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA