Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Artif Organs ; 48(7): 723-733, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38385713

RESUMO

BACKGROUND: The SARS-CoV-2 pandemic has spurred an unparalleled scientific endeavor to elucidate the virus' structure, infection mechanisms, and pathogenesis. Two-dimensional culture systems have been instrumental in shedding light on numerous aspects of COVID-19. However, these in vitro systems lack the physiological complexity to comprehend the infection process and explore treatment options. Three-dimensional (3D) models have been proposed to fill the gap between 2D cultures and in vivo studies. Specifically, spheroids, composed of lung cell types, have been suggested for studying SARS-CoV-2 infection and serving as a drug screening platform. METHODS: 3D lung spheroids were prepared by coculturing human alveolar or bronchial epithelial cells with human lung stromal cells. The morphology, size, and ultrastructure of spheroids before and after SARS-CoV-2 infection were analyzed using optical and electron microscopy. Immunohistochemistry was used to detect spike protein and, thus, the virus presence in the spheroids. Multiplex analysis elucidated the cytokine release after virus infection. RESULTS: The spheroids were stable and kept their size and morphology after SARS-CoV-2 infection despite the presence of multivesicular bodies, endoplasmic reticulum rearrangement, tubular compartment-enclosed vesicles, and the accumulation of viral particles. The spheroid responded to the infection releasing IL-6 and IL-8 cytokines. CONCLUSION: This study demonstrates that coculture spheroids of epithelial and stromal cells can serve as a cost-effective infection model for the SARS-CoV-2 virus. We suggest using this 3D spheroid as a drug screening platform to explore new treatments related to the cytokines released during virus infection, especially for long COVID treatment.


Assuntos
COVID-19 , Avaliação Pré-Clínica de Medicamentos , Pulmão , SARS-CoV-2 , Esferoides Celulares , Humanos , Esferoides Celulares/virologia , COVID-19/virologia , SARS-CoV-2/fisiologia , Pulmão/virologia , Pulmão/patologia , Tratamento Farmacológico da COVID-19 , Antivirais/farmacologia , Antivirais/uso terapêutico , Técnicas de Cocultura , Citocinas/metabolismo , Análise Custo-Benefício , Células Epiteliais/virologia
2.
Nature ; 607(7918): 345-350, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35768512

RESUMO

Enteric viruses like norovirus, rotavirus and astrovirus have long been accepted as spreading in the population through fecal-oral transmission: viruses are shed into feces from one host and enter the oral cavity of another, bypassing salivary glands (SGs) and reaching the intestines to replicate, be shed in feces and repeat the transmission cycle1. Yet there are viruses (for example, rabies) that infect the SGs2,3, making the oral cavity one site of replication and saliva one conduit of transmission. Here we report that enteric viruses productively and persistently infect SGs, reaching titres comparable to those in the intestines. We demonstrate that enteric viruses get released into the saliva, identifying a second route of viral transmission. This is particularly significant for infected infants, whose saliva directly transmits enteric viruses to their mothers' mammary glands through backflow during suckling. This sidesteps the conventional gut-mammary axis route4 and leads to a rapid surge in maternal milk secretory IgA antibodies5,6. Lastly, we show that SG-derived spheroids7 and cell lines8 can replicate and propagate enteric viruses, generating a scalable and manageable system of production. Collectively, our research uncovers a new transmission route for enteric viruses with implications for therapeutics, diagnostics and importantly sanitation measures to prevent spread through saliva.


Assuntos
Saliva , Glândulas Salivares , Viroses , Vírus , Astroviridae , Aleitamento Materno , Células Cultivadas , Fezes/virologia , Feminino , Humanos , Imunoglobulina A/imunologia , Lactente , Norovirus , Rotavirus , Saliva/virologia , Glândulas Salivares/virologia , Esferoides Celulares/virologia , Viroses/transmissão , Viroses/virologia , Vírus/crescimento & desenvolvimento
3.
J Am Soc Nephrol ; 32(9): 2242-2254, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34112705

RESUMO

BACKGROUND: Although coronavirus disease 2019 (COVID-19) causes significan t morbidity, mainly from pulmonary involvement, extrapulmonary symptoms are also major componen ts of the disease. Kidney disease, usually presenting as AKI, is particularly severe among patients with COVID-19. It is unknown, however, whether such injury results from direct kidney infection with COVID-19's causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), or from indirect mechanisms. METHODS: Using ex vivo cell models, we sought to analyze SARS-CoV-2 interactions with kidney tubular cells and assess direct tubular injury. These models comprised primary human kidney epithelial cells (derived from nephrectomies) and grown as either proliferating monolayers or quiescent three-dimensional kidney spheroids. RESULTS: We demonstrated that viral entry molecules and high baseline levels of type 1 IFN-related molecules were present in monolayers and kidney spheroids. Although both models support viral infection and replication, they did not exhibit a cytopathic effect and cell death, outcomes that were strongly present in SARS-CoV-2-infected controls (African green monkey kidney clone E6 [Vero E6] cultures). A comparison of monolayer and spheroid cultures demonstrated higher infectivity and replication of SARS-CoV-2 in actively proliferating monolayers, although the spheroid cultures exhibited high er levels of ACE2. Monolayers exhibited elevation of some tubular injury molecules-including molecules related to fibrosis (COL1A1 and STAT6) and dedifferentiation (SNAI2)-and a loss of cell identity, evident by reduction in megalin (LRP2). The three-dimensional spheroids were less prone to such injury. CONCLUSIONS: SARS-CoV-2 can infect kidney cells without a cytopathic effect. AKI-induced cellular proliferation may potentially intensify infectivity and tubular damage by SARS-CoV-2, suggesting that early intervention in AKI is warranted to help minimize kidney infection.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/virologia , COVID-19/complicações , SARS-CoV-2/patogenicidade , Esferoides Celulares/virologia , Animais , Células Cultivadas , Chlorocebus aethiops , Estudos de Coortes , Efeito Citopatogênico Viral , Células Epiteliais/patologia , Células Epiteliais/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Interferon Tipo I/metabolismo , Rim/imunologia , Rim/patologia , Rim/virologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Pandemias , Receptores Virais/metabolismo , Estudos Retrospectivos , SARS-CoV-2/fisiologia , Esferoides Celulares/patologia , Células Vero , Replicação Viral
4.
J Mol Med (Berl) ; 99(3): 425-438, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33484281

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumorigenic virus and the etiological agent of an endothelial tumor (Kaposi's sarcoma) and two B cell proliferative diseases (primary effusion lymphoma and multicentric Castleman's disease). While in patients with late stage of Kaposi's sarcoma the majority of spindle cells are KSHV-infected, viral copies are rapidly lost in vitro, both upon culture of tumor-derived cells or from newly infected endothelial cells. We addressed this discrepancy by investigating a KSHV-infected endothelial cell line in various culture conditions and in tumors of xenografted mice. We show that, in contrast to two-dimensional endothelial cell cultures, KSHV genomes are maintained under 3D cell culture conditions and in vivo. Additionally, an increased rate of newly infected cells was detected in 3D cell culture. Furthermore, we show that the PI3K/Akt/mTOR and ATM/γH2AX pathways are modulated and support an improved KSHV persistence in 3D cell culture. These mechanisms may contribute to the persistence of KSHV in tumor tissue in vivo and provide a novel target for KS specific therapeutic interventions. KEY MESSAGES: In vivo maintenance of episomal KSHV can be mimicked in 3D spheroid cultures 3D maintenance of KSHV is associated with an increased de novo infection frequency PI3K/Akt/mTOR and ATM/ γH2AX pathways contribute to viral maintenance.


Assuntos
Técnicas de Cultura de Células em Três Dimensões , Células Endoteliais/virologia , Herpesvirus Humano 8/fisiologia , Cultura de Vírus/métodos , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Transformada , Doxiciclina/farmacologia , Células Endoteliais/citologia , Genoma Viral , Xenoenxertos , Histonas/fisiologia , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Plasmídeos , Proteínas Proto-Oncogênicas c-akt/fisiologia , Sarcoma de Kaposi/virologia , Transdução de Sinais/fisiologia , Esferoides Celulares/transplante , Esferoides Celulares/virologia , Serina-Treonina Quinases TOR/fisiologia , Latência Viral , Liberação de Vírus , Replicação Viral
5.
Artif Organs ; 45(6): 548-558, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33264436

RESUMO

The new coronavirus (2019-nCoV) or the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was officially declared by the World Health Organization (WHO) as a pandemic in March 2020. To date, there are no specific antiviral drugs proven to be effective in treating SARS-CoV-2, requiring joint efforts from different research fronts to discover the best route of treatment. The first decisions in drug discovery are based on 2D cell culture using high-throughput screening. In this context, spheroids and organoids emerge as a reliable alternative. Both are scaffold-free 3D engineered constructs that recapitulate key cellular and molecular events of tissue physiology. Different studies have already shown their advantages as a model for different infectious diseases, including SARS-CoV-2 and for drug screening. The use of these 3D engineered tissues as an in vitro model can fill the gap between 2D cell culture and in vivo preclinical assays (animal models) as they could recapitulate the entire viral life cycle. The main objective of this review is to understand spheroid and organoid biology, highlighting their advantages and disadvantages, and how these scaffold-free engineered tissues can contribute to a better comprehension of viral infection by SARS-CoV-2 and to the development of in vitro high-throughput models for drug screening.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Organoides/fisiologia , Esferoides Celulares/fisiologia , Engenharia Tecidual/métodos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Humanos , Organoides/virologia , SARS-CoV-2 , Esferoides Celulares/virologia , Alicerces Teciduais
6.
EMBO Mol Med ; 12(8): e12697, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32473600

RESUMO

Baricitinib is an oral Janus kinase (JAK)1/JAK2 inhibitor approved for the treatment of rheumatoid arthritis (RA) that was independently predicted, using artificial intelligence (AI) algorithms, to be useful for COVID-19 infection via proposed anti-cytokine effects and as an inhibitor of host cell viral propagation. We evaluated the in vitro pharmacology of baricitinib across relevant leukocyte subpopulations coupled to its in vivo pharmacokinetics and showed it inhibited signaling of cytokines implicated in COVID-19 infection. We validated the AI-predicted biochemical inhibitory effects of baricitinib on human numb-associated kinase (hNAK) members measuring nanomolar affinities for AAK1, BIKE, and GAK. Inhibition of NAKs led to reduced viral infectivity with baricitinib using human primary liver spheroids. These effects occurred at exposure levels seen clinically. In a case series of patients with bilateral COVID-19 pneumonia, baricitinib treatment was associated with clinical and radiologic recovery, a rapid decline in SARS-CoV-2 viral load, inflammatory markers, and IL-6 levels. Collectively, these data support further evaluation of the anti-cytokine and anti-viral activity of baricitinib and support its assessment in randomized trials in hospitalized COVID-19 patients.


Assuntos
Antivirais/farmacologia , Inteligência Artificial , Azetidinas/farmacologia , Betacoronavirus , Infecções por Coronavirus/tratamento farmacológico , Pandemias , Pneumonia Viral/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Sulfonamidas/farmacologia , Adulto , Idoso , Antivirais/farmacocinética , Antivirais/uso terapêutico , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , COVID-19 , Citocinas/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Leucócitos/efeitos dos fármacos , Fígado , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Purinas , Pirazóis , SARS-CoV-2 , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/virologia , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapêutico , Tratamento Farmacológico da COVID-19
7.
Adv Biosyst ; 4(2): e1900143, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32293137

RESUMO

Replicable oncolytic viruses (OVs) induce tumor cell lysis and release viral progeny. The released progeny virions and cell debris can spread within surrounding tumor cells or blood vessels. These released molecules may also induce bystander damage in additional tumor cells through spreading within surrounding tumor cells or blood vessels. However, this effect has not been clearly demonstrated due to the difficulty of direct observation. Here, the bystander infection of OVs by vessel delivery and selective infection in 3D multicellular tumoroids (MCTs) in an in vitro microphysiological system (MPS) with integrated medium flow is demonstrated. This study uses replicable vesicular stomatitis virus (VSV)-green fluorescence protein (GFP) to identify the location of infection in 3D MCTs. Using this MPS, the oncoselective infection by VSV-GFP and the spreading by delivery of OVs through flow via block-to-block linkage of the primary infected MPS with uninfected 3D MCTs in an integrated MPS is observed. This MPS enables real-time monitoring and various analysis for the bystander infection of OVs. It is expected that the 3D in vitro MPS can be suitable to investigate the oncoselective spreading and bystander infection of OVs.


Assuntos
Técnicas Citológicas , Modelos Biológicos , Terapia Viral Oncolítica/efeitos adversos , Vírus Oncolíticos , Células A549 , Células Cultivadas , Técnicas Citológicas/instrumentação , Técnicas Citológicas/métodos , Desenho de Equipamento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Infecções por Rhabdoviridae/virologia , Esferoides Celulares/virologia , Células Tumorais Cultivadas/virologia , Vesiculovirus/genética
8.
J BUON ; 24(4): 1700-1705, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31646828

RESUMO

PURPOSE: Nasopharyngeal carcinoma (NPC) is one of the common types of cancer that originate from the nasopharyngeal region. Recurrence and early metastasis represent major problems associated with NPC mortality. These are mainly caused by various molecular changes that take place during the conversion of normal stem cells into treatment-resistant stem cells. The aim of our study was to investigate the proliferative behavior of cancer stem cells in different stages of NPC and to identify the functional roles of SPLUNC1 and MLL3 associated with cancer stem cells. METHODS: We successfully developed a NPC mouse model using C666-1 cells. Immunohistochemistry and Western blotting were used to analyze the expression of SOX2, SPLUNC1 and MLL3. RESULTS: Null BALB/c mice developed initial and aggressive stages of NPC in 3 and 10 weeks, respectively. Histological results showed that the proliferative ability of cells increased as the tumor progressed to the next level. The SOX2 protein showed a peculiar pattern of upregulation in aggressive NPC when compared with control tissues and initial NPC. Remarkably, our study found that SPLUNC1 and MLL3 expression showed upregulation in initial NPC, which indicates their role in the tumor resistance mechanism even if their expression was downregulated in aggressive NPC. CONCLUSIONS: Our results conclude that SPLUNC1 and MLL3 expression control the resistance mechanism of cancer stem cells in initial NPC, but their downregulation in aggressive stages contributes to developing resistance in nasopharyngeal cancer stem cells.


Assuntos
Proteínas de Ligação a DNA/genética , Glicoproteínas/genética , Carcinoma Nasofaríngeo/genética , Fosfoproteínas/genética , Fatores de Transcrição SOXB1/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Herpesvirus Humano 4/patogenicidade , Humanos , Camundongos , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/virologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/virologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/virologia , Esferoides Celulares/metabolismo , Esferoides Celulares/virologia
9.
J Virol Methods ; 265: 99-104, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30582939

RESUMO

The way viruses interact with cultured cells and their surrounding environment is still a matter of debate. From a technical point of view, 2D cell cultures only partially exhibit the morpho-molecular pattern required for viral tropism, not reflecting the complexity of the microenvironment in vivo. Therefore, 3D cell cultures are envisioned as an alternative approach to study viral replication possibly closer to in vivo conditions than 2D, representing the link between traditional cell culture and in vivo models. The use of cellular spheroids is proving to be useful to optimize and overcome constraints related to conventional in vitro systems for viral isolation. In order to create an advanced 3D in vitro isolation system, we compared the classic 2D shell vial system with the spheroid culture method based on the adhesion inhibition technique with pHema. In this study, we evaluated which of the most common viral cell lines used in our laboratory (A-549, 293 T, CaCo2, KB, HUH-7, VERO, and MRC-5) (Fig. 1) could be grown as 3D cultures and all proved to be able to grow as spheroids. Subsequently, we compared the sensitivity and efficiency of isolation of three viral species of medical interest (Adenovirus, CMV, HSV-1) in 2D and 3D cell cultures obtained from the respective susceptible cells. Our results indicate earlier and more sensitive virus isolation than in traditional 2D shell vial system for all three viruses tested, thus confirming how the establishment of 3D culture systems in the virological field is crucial to the improvement and evolution of more accurate and faster virus isolation protocols.


Assuntos
Adenoviridae/crescimento & desenvolvimento , Citomegalovirus/crescimento & desenvolvimento , Herpesvirus Humano 1/crescimento & desenvolvimento , Esferoides Celulares/virologia , Cultura de Vírus/métodos , Replicação Viral , Adenoviridae/isolamento & purificação , Animais , Linhagem Celular , Citomegalovirus/isolamento & purificação , Herpesvirus Humano 1/isolamento & purificação , Humanos
10.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30045986

RESUMO

Herpes simplex virus 1 (HSV-1)-mediated oncolytic therapy is an emerging cancer treatment modality with potential effectiveness against a variety of malignancies. To better understand the interaction of HSV-1 with neoplastic cells, we inoculated three-dimensional (3D) cultures of human uveal melanoma cells with HSV-1. 3D melanoma cultures were established by placing tumor cells on the surface of a Matrigel matrix, which was followed by the growth of tumor cells on the matrix surface and invasion of the Matrigel matrix by some tumor cells to form multicellular tumor spheroids within the matrix. When established 3D melanoma cultures were inoculated with HSV-1 by placing virus on the surface of cultures, virus infection caused extensive death of melanoma cells growing on the surface of the 3D matrix and significantly decreased the number of tumor cell spheroids within the matrix. However, HSV-1 infection did not lead to a complete destruction of tumor cells in the 3D cultures during a 17-day observation period and, surprisingly, HSV-1 infection promoted the growth of some melanoma cells within the matrix as determined by the significantly increased size of residual viable multicellular tumor spheroids in virus-inoculated 3D cultures at 17 days after virus inoculation. Acyclovir treatment inhibited HSV-1-induced tumor cell killing but did not block the virus infection-induced increase in spheroid size. These findings suggest that although HSV-1 oncolytic virotherapy may cause extensive tumor cell killing, it may also be associated with the unintended promotion of the growth of some tumor cells.IMPORTANCE Cancer cells are exposed to HSV-1 during oncolytic virotherapy with the intention of killing tumor cells. Our observations reported here suggest that potential dangers of HSV-1 oncolytic therapy include promotion of growth of some tumor cells. Furthermore, our findings raise the possibility that HSV-1 infection of neoplastic cells during natural infections or vaccinations may promote the growth of tumors. Our study indicates that HSV-1 infection of 3D tumor cell cultures provides an experimental platform in which mechanisms of HSV-1-mediated promotion of tumor cell growth can be effectively studied.


Assuntos
Herpes Simples/complicações , Herpesvirus Humano 1/patogenicidade , Melanoma/patologia , Terapia Viral Oncolítica , Esferoides Celulares/patologia , Neoplasias Uveais/patologia , Replicação Viral , Proliferação de Células , Herpes Simples/virologia , Humanos , Melanoma/terapia , Melanoma/virologia , Esferoides Celulares/virologia , Células Tumorais Cultivadas , Neoplasias Uveais/terapia , Neoplasias Uveais/virologia
11.
Nat Commun ; 9(1): 1485, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29662124

RESUMO

Cohesin is a multi-subunit nuclear protein complex that coordinates sister chromatid separation during cell division. Highly frequent somatic mutations in genes encoding core cohesin subunits have been reported in multiple cancer types. Here, using a genome-wide CRISPR-Cas9 screening approach to identify host dependency factors and novel innate immune regulators of rotavirus (RV) infection, we demonstrate that the loss of STAG2, an important component of the cohesin complex, confers resistance to RV replication in cell culture and human intestinal enteroids. Mechanistically, STAG2 deficiency results in spontaneous genomic DNA damage and robust interferon (IFN) expression via the cGAS-STING cytosolic DNA-sensing pathway. The resultant activation of JAK-STAT signaling and IFN-stimulated gene (ISG) expression broadly protects against virus infections, including RVs. Our work highlights a previously undocumented role of the cohesin complex in regulating IFN homeostasis and identifies new therapeutic avenues for manipulating the innate immunity.


Assuntos
Antígenos Nucleares/imunologia , Proteínas de Ciclo Celular/imunologia , Proteínas Cromossômicas não Histona/imunologia , Interações Hospedeiro-Patógeno , Proteínas de Membrana/imunologia , Nucleotidiltransferases/imunologia , Rotavirus/imunologia , Esferoides Celulares/imunologia , Antígenos Nucleares/genética , Sistemas CRISPR-Cas , Células CACO-2 , Proteínas de Ciclo Celular/genética , Núcleo Celular/imunologia , Núcleo Celular/virologia , Proteínas Cromossômicas não Histona/genética , Dano ao DNA , Deleção de Genes , Edição de Genes , Regulação da Expressão Gênica , Genoma Humano , Células HEK293 , Células HT29 , Células HeLa , Humanos , Interferons/genética , Interferons/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Janus Quinases/genética , Janus Quinases/imunologia , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , Rotavirus/crescimento & desenvolvimento , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Esferoides Celulares/virologia , Coesinas
12.
J Virol ; 92(11)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29593030

RESUMO

Human papillomavirus (HPV) infection of the genital tract is common; however, only about 10 to 15% of infections persist, and approximately 10 to 15% of these persistent infections result in cancer. Basal epidermal stem cells are the presumed target cells for HPV infection, providing a reservoir of latently infected cells that persist over time and initiate lesions. However, it is not known whether stem cell density has any influence on transformation of human keratinocytes by HPV. We explored the relationship between stem cell properties of normal human keratinocytes and their susceptibility to transformation by HPV16 DNA. Normal human keratinocyte isolates (NHKc) derived from different donors were cultured in three-dimensional anchorage-free suspension to assess their spheroid-forming ability. NHKc spheroids were then plated back into plastic monolayer culture and transfected with full-length HPV16 DNA, which we have previously shown to integrate into the host cell genome upon transfection. Spheroid-derived NHKc (SD-NHKc) and fluorescence-activated cell sorting-purified populations of basal stem-like keratinocytes, expressing low levels of epidermal growth factor receptor and high levels of integrin alpha 6 (EGFRlo/ITGα6hi), responded to transfection with HPV16 DNA with more vigorous proliferation, greater immortalization efficiency, and faster progression to differentiation resistance than autologous mass-cultured cells. Conversely, cells committed to terminal differentiation (EGFRhi/ITGα6lo) grew slowly after transfection with HPV16 and failed to generate immortalized or DR clones. HPV16 DNA induced stem cell properties in mass-cultured NHKc. We conclude that HPV16 preferentially immortalizes basal keratinocytes with stem cell properties and that these cells readily achieve a differentiation-resistant phenotype upon immortalization by HPV16.IMPORTANCE This paper explores the relationship between the stem cell properties of normal human epidermal cells in culture and these cells' susceptibility to transformation by HPV16 DNA, the HPV type present in about 50% of cervical cancers. We report variable susceptibilities to HPV16-mediated transformation among different keratinocyte isolates derived from neonatal foreskin. Our findings provide strong experimental evidence that HPV16 preferentially transforms basal keratinocytes with stem cell properties. Insights gained from these studies increase our understanding of the host cell-specific factors influencing individual susceptibility to HPV-driven transformation and the contributing factors leading to preneoplastic and neoplastic progression of HPV-positive lesions.


Assuntos
Transformação Celular Viral/genética , Papillomavirus Humano 16/genética , Queratinócitos/virologia , Células-Tronco/virologia , Linhagem Celular Transformada , Proliferação de Células/genética , DNA Viral/genética , Receptores ErbB/metabolismo , Feminino , Prepúcio do Pênis/citologia , Humanos , Cadeias alfa de Integrinas/metabolismo , Queratinócitos/citologia , Masculino , Esferoides Celulares/virologia , Células-Tronco/citologia , Transfecção , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia
13.
Int J Mol Med ; 40(2): 400-410, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28627604

RESUMO

The hepatitis B virus X (HBx) protein is an important factor in hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC). The C-terminal region of HBx plays a major role in the replication of HBV. Notably, HBx promotes the expansion and tumourigenesis of hepatic progenitor cells (HPCs) in mice. However, it remains unclear as to whether the C-terminal region of HBx is required for the stimulation fo the proliferation of mouse foetal HPCs (FHPCs). In our study, we used EpCAM+, CD133+ and CD49f+ FHPCs, which are bipotential clonogenic cells. These FHPCs transformed into mature hepatocytes and cholangiocytes when cultured under conditions that facilitate differentiation. Compared with the FHPCs grown as monolayers, spherical cell proliferation occurred more rapidly. Furthermore, spherically cultured FHPCs can grow in semi-solid agar and tend to maintain the morphology and characteristics of stem cells compared with growth in rat tail collagen. Notably, we also demonstrate that the C-terminus of HBx stimulates the proliferation of FHPCs, but is not required for the formation of spheroids, similar to hepatic cancer stem cells. These findings enhance our understanding of the HBx-induced tumourigenicity of FHPCs and may aid in the treatment of HCC.


Assuntos
Proliferação de Células , Vírus da Hepatite B/fisiologia , Hepatócitos/virologia , Células-Tronco/virologia , Transativadores/metabolismo , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virologia , Diferenciação Celular , Células Cultivadas , Hepatite B/metabolismo , Hepatite B/virologia , Vírus da Hepatite B/química , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/virologia , Domínios Proteicos , Esferoides Celulares/citologia , Esferoides Celulares/metabolismo , Esferoides Celulares/virologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Transativadores/química , Proteínas Virais Reguladoras e Acessórias
14.
BMC Cancer ; 16: 495, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27431799

RESUMO

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated epithelial malignancy that exhibits distinct geographical and ethnic prevalence. Although the contemporary therapeutic approach of radio-/chemotherapy provides excellent results for patients with early-stage disease, it is far from satisfactory for those with disease remission and distant metastasis. Promising therapeutic strategies for advanced and relapsed NPC are still lacking. We recently identified and characterized a cancer stem-like cell (CSC) subpopulation in NPC that appeared to play an important role in tumor progression. Microarray analysis revealed downregulation of several stemness-inhibiting miRNAs in these CSC cells. Among these miRNAs, miR-96 and miR-183 showed the highest fold change and were selected to elucidate their role in repressing NPC CSC properties. METHODS: MiR-96 and miR-183 expression in NPC CSCs was detected by qRT-PCR. Transient and stable transfection was performed in EBV-positive NPC C666-1 cells to examine the effects of ectopic expression of miR-96 and miR-183 on repressing cell growth and CSC properties. Anchorage-dependent (colony formation) and anchorage-independent (tumor sphere formation) growths of these miR-96 and miR-183 expressing cells were determined. Expression of multiple CSC markers and related molecules were accessed by flow cytometry and Western blotting. The tumorigenicity of the stable miR-96- and miR-183-transfected NPC cells was examined in an in vivo nude mice model. RESULTS: Downregulation of miR-96 and miR-183 was confirmed in NPC spheroids. Using transient or stable transfection, we showed that ectopic expression of miR-96 and miR-183 suppressed cell growth and tumor sphere formation in NPC. Reduced NICD3 and NICD4 in miR-96- and miR-183-expressing NPC cells suggests the involvement of the NOTCH signaling pathway in their tumor suppressive function. Finally, we showed that the tumorigenicity of cells stably expressing miR-183 was significantly inhibited in the in vivo nude mice model. CONCLUSIONS: miR-183 is a tumor-suppressive miRNA in EBV-associated NPC. Its abilities to suppress CSC properties in vitro and effectively reduce tumor growth in vivo shed light on its role as a potential therapeutic target.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Neoplasias Nasofaríngeas/genética , Células-Tronco Neoplásicas/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Feminino , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virologia , Células-Tronco Neoplásicas/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptor Notch3/genética , Receptor Notch3/metabolismo , Receptor Notch4 , Receptores Notch/genética , Receptores Notch/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esferoides Celulares/metabolismo , Esferoides Celulares/virologia , Transplante Heterólogo/métodos
15.
IEEE Trans Nanobioscience ; 14(8): 946-51, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26661633

RESUMO

Glioblastoma (GBM) is the most aggressive brain tumor, with 12-15 months median survival time despite current treatment efforts. Among the alternative treatment approaches that have gained acceptance over the last decade is the use of replication-competent oncolytic adenoviruses, which are promising due to their relatively low toxicity and tumor-specific targeting. Three-dimensional (3D) tumor models can mimic the physiological microenvironment of GBM tumors and provide valuable information about the interaction between tumor cells and adenoviruses. Therefore, robust in vitro 3D tumor models are critical to investigate the mechanisms underlying tumor progression and explore the cytotoxicity effect of the adenovirus on tumor cells. In this study, we used a hydrogel microwell platform to generate in vitro 3D GBM spheroids and studied their interactions with the Delta-24-RGD adenovirus. The results showed that the cultured 3D spheroids were successfully infected by the Delta-24-RGD. A significant cell lysis was observed. Cell viability was decreased approximately 37%, 54% and 65% with 10, 50, and 100 MOIs, respectively. The infection of the Delta-24-RGD was found more effective on 3D spheroids when compared to 2D monolayer cell culture. These results implicate that our hydrogel microwell platform could provide a promising 3D model to investigate the oncolytic potential of the viruses in vitro.


Assuntos
Adenoviridae/genética , Glioblastoma , Modelos Biológicos , Esferoides Celulares/fisiologia , Microambiente Tumoral/fisiologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Técnicas de Transferência de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Oligopeptídeos/genética , Polietilenoglicóis/química , Esferoides Celulares/virologia , Células Tumorais Cultivadas
16.
Cancer Gene Ther ; 21(6): 256-60, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24874841

RESUMO

The relationship between ligand-receptor affinity and antitumor potency of an oncolytic virus was investigated using a panel of six HER2/neu (HER2)-targeted measles viruses (MVs) displaying single-chain antibodies (scFv) that bind to the same epitope on HER2, but with affinities ranging from 10(-6) to 10(-11) M. All viruses were able to infect SKOV3ip.1 human ovarian cancer cells in vitro, but only the high-affinity MV (Kd≥10(-8) M) induced cytopathic effects of syncytia formation in the cell monolayers. In contrast, all six viruses were therapeutically active in vivo against orthotopic human ovarian SKOV3ip.1 tumor xenografts in athymic mice compared with saline-treated controls. The oncolytic activities of MV displaying the high-affinity scFv (Kd=10(-9), 10(-10), 10(-11) M) were not significantly superior to MV displaying scFv with Kd of 10(-8) M or less. Results from this study suggest that increasing the receptor affinity of the attachment protein of an oncolytic MV has minimal impact on its in vivo efficacy against a tumor that expresses the targeted receptor.


Assuntos
Vírus do Sarampo/imunologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Anticorpos de Cadeia Única/metabolismo , Animais , Modelos Animais de Doenças , Epitopos/metabolismo , Feminino , Humanos , Injeções Intraperitoneais , Camundongos Nus , Vírus Oncolíticos/patogenicidade , Neoplasias Ovarianas/virologia , Receptor ErbB-2/metabolismo , Esferoides Celulares/virologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Pharm ; 11(7): 2106-14, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24761996

RESUMO

Developing effective new drugs against hepatitis C (HCV) virus has been challenging due to the lack of appropriate small animal and in vitro models recapitulating the entire life cycle of the virus. Current in vitro models fail to recapitulate the complexity of human liver physiology. Here we present a method to study HCV infection and replication on spheroid cultures of Huh 7.5 cells and primary human hepatocytes. Spheroid cultures are constructed using a galactosylated cellulosic sponge with homogeneous macroporosity, enabling the formation and maintenance of uniformly sized spheroids. This facilitates easy handling of the tissue-engineered constructs and overcomes limitations inherent of traditional spheroid cultures. Spheroids formed in the galactosylated cellulosic sponge show enhanced hepatic functions in Huh 7.5 cells and maintain liver-specific functions of primary human hepatocytes for 2 weeks in culture. Establishment of apical and basolateral polarity along with the expression and localization of all HCV specific entry proteins allow for a 9-fold increase in viral entry in spheroid cultures over conventional monolayer cultures. Huh 7.5 cells cultured in the galactosylated cellulosic sponge also support replication of the HCV clone, JFH (Japanese fulminant hepatitis)-1 at higher levels than in monolayer cultures. The advantages of our system in maintaining liver-specific functions and allowing HCV infection together with its ease of handling make it suitable for the study of HCV biology in basic research and pharmaceutical R&D.


Assuntos
Técnicas de Cultura de Células/métodos , Hepacivirus/genética , Hepatite C/virologia , Hepatócitos/virologia , Esferoides Celulares/virologia , Engenharia Tecidual/métodos , Replicação Viral/genética , Materiais Biocompatíveis/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Celulose/metabolismo , Galactose/metabolismo , Hepatite C/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/virologia , Esferoides Celulares/metabolismo
18.
Gene Ther ; 21(6): 609-17, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24739522

RESUMO

In the canonical pathway, infection of cells by the wild-type mammalian orthoreovirus Type 3 Dearing (T3D) is dependent on the interaction of the viral spike protein σ1 with the high-affinity cellular receptor junction adhesion molecule-A (JAM-A). We previously demonstrated that the human glioblastoma cell line U-118 MG does not express JAM-A and resists reovirus T3D infection in standard cell culture conditions (SCCC). Heterologous JAM-A expression sensitises U-118 MG cells to reovirus T3D. Here we studied reovirus infection in U-118 MG cells grown in spheroid cultures with the premise that cells in such cultures resemble cells in tumours more than those grown under standard adherent cell culture conditions on a plastic surface. Although the U-118 MG cells in spheroids do not express JAM-A, they are susceptible to reovirus T3D infection. We show that this can be attributed to factors secreted by cells in the spheroids. The concentration of active extracellular proteases cathepsin B and L in the medium of spheroid cultures was increased 19- and 24-fold, respectively, as compared with SCCC. These enzymes can convert the reovirus particles into a form that can infect the U-118 MG cells independent of JAM-A. Taken together, these data demonstrate that infection of tumour cells by wild-type reovirus T3D is not strictly dependent on the expression of JAM-A on the cell surface.


Assuntos
Moléculas de Adesão Celular/metabolismo , Glioblastoma/patologia , Glioblastoma/virologia , Orthoreovirus Mamífero 3/patogenicidade , Receptores de Superfície Celular/metabolismo , Esferoides Celulares/virologia , Catepsina B/metabolismo , Catepsina L/metabolismo , Humanos
19.
Oncol Rep ; 29(3): 1108-14, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23292314

RESUMO

Accumulating evidence suggests that breast cancer originates from cancer stem cells (CSCs), which comprise a small percentage of the overall tumor but are highly tumorigenic and pluripotent with unlimited proliferation potential. Furthermore, CSCs are highly resistant to conventional treatment, which may explain certain difficulties in treating cancer with current therapy options. In this study, the third generation oncolytic herpes simplex virus (oHSV) vector G47∆ effectively killed different subtypes of breast cancer cells, with more than 98% of the tumor cells killed by Day 5. Moreover, G47∆ targeted equally non-cancer stem cells (NCSCs) and CSCs which showed resistance to paclitaxel. We demonstrated that G47∆ effectively replicated and spread among CSCs. G47∆ also impaired the self-renewal ability of CSCs, as the viable cells were unable to form secondary tumor spheres. We also showed that G47∆ was able to induce the regression of tumor xenografts in BALB/c nude mice and demonstrated the ability of G47∆ to synergize with paclitaxel by killing both NCSCs and CSCs, suggesting that oHSV may be an effective treatment modality for patients with breast cancer.


Assuntos
Neoplasias da Mama/terapia , Células-Tronco Neoplásicas/fisiologia , Vírus Oncolíticos/fisiologia , Paclitaxel/farmacologia , Simplexvirus/fisiologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/virologia , Esferoides Celulares/fisiologia , Esferoides Celulares/virologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cancer Gene Ther ; 18(12): 897-905, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21904396

RESUMO

Accumulating evidence supports the concept that cancer stem cells (CSCs) are responsible for tumor initiation and maintenance. They are also considered as an attractive target for advanced cancer therapy. Using a sphere culture method that favors the growth of self-renewal cells, we have isolated sphere-forming cells (SFCs) from cervical cancer cell lines HeLa and SiHa. HeLa-SFCs were resistant to multiple chemotherapeutic drugs and were more tumorigenic, as evidenced by the growth of tumors following injection of immunodeficient mice with 1 × 10(4) cells, compared with 1 × 10(6) parental HeLa cells required to grow tumors of similar size in the same time frame. These cells showed an expression pattern of CD44(high)/CD24(low) that resembles the CSC surface biomarker of breast cancer. We further demonstrated that HeLa-SFCs expressed a higher level (6.9-fold) of the human papillomavirus oncogene E6, compared with that of parental HeLa cells. Gene silencing of E6 with a lentiviral-short-hairpin RNA (shRNA) profoundly inhibited HeLa-SFC sphere formation and cell growth. The inhibition of cell growth was even greater than that for sphere formation after E6 silence, suggesting that the loss of self-renewing ability may be more important. We then measured the expression of self-renewal genes, transformation growth factor-beta (TGF-ß) and leukemia-inhibitory factor (LIF), in shRNA-transduced HeLa-SFCs and found that expression of all three TGF-ß isoforms was significantly downregulated while LIF remained unchanged. Expression of the Ras gene (a downstream component of TGF-ß) was also markedly decreased, suggesting that the growth-inhibitory effect could be via the TGF-ß pathway. The above data indicate RNA interference-based therapy may offer a new approach for CSC-targeted cancer therapy.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Terapia Genética/métodos , Células-Tronco Neoplásicas/virologia , Proteínas Oncogênicas Virais/antagonistas & inibidores , RNA Interferente Pequeno/genética , Esferoides Celulares/virologia , Neoplasias do Colo do Útero/terapia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Células HeLa , Humanos , Fator Inibidor de Leucemia/metabolismo , Camundongos , Camundongos Nus , Proteínas Oncogênicas Virais/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...