Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Clin Immunol ; 260: 109915, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38286172

RESUMEN

The increased risk for acquiring secondary illnesses in people living with HIV (PLWH) has been associated with immune dysfunction. We have previously found that circulating monocytes from PLWH display a trained phenotype. Here, we evaluated the metabolic profile of these cells and found increased mitochondrial respiration and glycolysis of monocyte-derived macrophages (MDMs) from PLWH. We additionally found that cART shifted the energy metabolism of MDMs from controls toward increased utilization of mitochondrial respiration. Importantly, both downregulation of IKAROS expression and inhibition of the mTOR pathway reversed the metabolic profile of MDMs from PLWH and cART-treated control-MDMs. Altogether, this study reveals a very specific metabolic adaptation of MDMs from PLWH, which involves an IKAROS/mTOR-dependent increase of mitochondrial respiration and glycolysis. We propose that this metabolic adaptation decreases the ability of these cells to respond to environmental cues by "locking" PLWH monocytes in a pro-inflammatory and activated phenotype.


Asunto(s)
Infecciones por VIH , Humanos , Macrófagos , Monocitos , Fenotipo , Serina-Treonina Quinasas TOR/metabolismo
2.
J Hematol Oncol ; 16(1): 48, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37143124

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of several human cancers, including Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL), which preferentially arise in immunocompromised patients while lack of effective therapeutic options. Oncoproteins Myc and hypoxia-inducible factor-1α (HIF1α) have been found closely related to KSHV infection, replication and oncogenesis. However, the strategies of dual targeting these two oncoproteins have never been developed and tested for treatments of KSHV-related malignancies. In the current study, we report that treatment of echinomycin dramatically regresses cell growth both in vitro-cultured KSHV + tumor cells and in vivo KS or PEL xenograft mice models, through simultaneously inhibiting Myc and HIF1α expression. Echinomycin treatment also induces viral lytic gene expression whereas not increasing infectious virions production from KSHV + tumor cells. Our comparative transcriptomic analysis has identified a bunch of new Echinomycin-regulated, Myc- and HIF1α-related genes contributed to KSHV pathogenesis, including KDM4B and Tau, which are required for the survival of KSHV + tumor cells with functional validation. These data together reveal that dual targeting Myc and HIF1α such as using Echinomycin may represent a new and promising option for treatments of these virus-associated malignancies.


Asunto(s)
Equinomicina , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Animales , Ratones , Herpesvirus Humano 8/genética , Equinomicina/farmacología , Equinomicina/uso terapéutico , Latencia del Virus/genética , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/metabolismo , Ciclo Celular , Histona Demetilasas con Dominio de Jumonji
3.
Viruses ; 15(4)2023 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-37112991

RESUMEN

Although Kaposi's sarcoma-associated herpesvirus (KSHV) has been reported to cause several human cancers including Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL), the mechanisms of KSHV-induced tumorigenesis, especially virus-host interaction network, are still not completely understood, which therefore hinders the development of effective therapies. Histamine, together with its receptors, plays an important role in various allergic diseases by regulating different inflammation and immune responses. Our previous data showed that antagonists targeting histamine receptors effectively repressed KSHV lytic replication. In the current study, we determined that histamine treatment increased cell proliferation and anchorage-independent growth abilities of KSHV-infected cells. Furthermore, histamine treatment affected the expression of some inflammatory factors from KSHV-infected cells. For clinical relevance, several histamine receptors were highly expressed in AIDS-KS tissues when compared to normal skin tissues. We determined that histamine treatment promoted KSHV-infected lymphoma progression in immunocompromised mice models. Therefore, besides viral replication, our data indicate that the histamine and related signaling are also involved in other functions of KSHV pathogenesis and oncogenesis.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Animales , Ratones , Herpesvirus Humano 8/fisiología , Histamina , Receptores Histamínicos/uso terapéutico , Carcinogénesis , Transformación Celular Neoplásica
4.
Front Cell Infect Microbiol ; 12: 1049624, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36457850

RESUMEN

Objective: Kaposi's Sarcoma-associated Herpesvirus (KSHV) is the etiologic agent of several human cancers, including Kaposi's sarcoma (KS) and Primary effusion lymphoma (PEL), which are usually seen in immunocompromised patients while lack of effective therapeutic options. Interleukin1 (IL1) family is a major mediator for inflammation response and has functional role in both innate and adaptive immunity. In contrast to the well-studied IL1 molecules, the activation and functional role of IL1 receptor/co-receptor and other related ligands, such as the IL1 receptor accessory protein (IL1RAP), in KSHV pathogenesis and tumorigenesis remain almost unknown. Methods: In the current study, a series of KSHV negative and positive primary or tumor cells, as well as AIDS-KS tumor samples from cohort HIV+ patients were used to compare and determine the activation status of IL1 signaling molecules, and their functional roles in KSHV pathogenesis. Results: We reported the high activation of multiple IL1 signaling molecules, including IL1, IL36, IL1R1, IL1RAP and IRAKs, during KSHV latent and lytic stages, as well as in clinical samples from patients with KSHV-related malignancies. Directly targeting these molecules especially IL1R1 and IL1RAP significantly impaired the survival and growth of KSHV+ tumor cells, as well as their colony formation on 3-D culture. Conclusion: Our data indicate the importance of IL1 signaling molecules in KSHV pathogenesis and tumorigenesis, which may represent attractive therapeutic targets against these virus-associated diseases.


Asunto(s)
Herpesvirus Humano 8 , Humanos , Carcinogénesis , Transformación Celular Neoplásica , Inmunidad Adaptativa , Receptores de Interleucina-1
5.
Front Microbiol ; 13: 882520, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35516440

RESUMEN

Kaposi's Sarcoma (KS) caused by Kaposi's sarcoma-associated herpesvirus (KSHV) continues to be the most common AIDS-associated tumor. Involvement of the oral cavity represents one of the most common clinical manifestations of this tumor. Numerous types of cancer are associated with the alterations of in components of the microbiome. However, little is known about how KSHV coinfection affects the oral microbiome in HIV+ patients, especially in a "pre-cancer" niche. Using 16S rRNA pyrosequencing, we found that oral shedding of KSHV correlated with altered oral microbiome signatures in HIV+ patients, including a reduction in the microbiota diversity, changing the relative composition of specific phyla and species, and regulating microbial functions. Furthermore, we found that Streptococcus sp., one of the most increased species in the oral cavity of HIV+/KSHV+ patients, induced KSHV lytic reactivation in primary oral cells. Together, these data indicate that oral shedding of KSHV may manipulate the oral microbiome to promote viral pathogenesis and tumorigenesis especially in immunocompromised patients.

6.
Sci Rep ; 12(1): 3384, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35232976

RESUMEN

Glioblastomas are the most aggressive brain tumors for which therapeutic options are limited. Current therapies against glioblastoma include surgical resection, followed by radiotherapy plus concomitant treatment and maintenance with temozolomide (TMZ), however, these standard therapies are often ineffective, and average survival time for glioblastoma patients is between 12 and 18 months. We have previously reported a strong anti-glioblastoma activity of several metabolic compounds, which were synthetized based compounds, which were synthetized based on the chemical structure of a common lipid-lowering drug, fenofibrate, and share a general molecular skeleton of benzoylphenoxyacetamide (BPA). Extensive computational analyses of phenol and naphthol moieties added to the BPA skeleton were performed in this study with the objective of selecting new BPA variants for subsequent compound preparation and anti-glioblastoma testing. Initially, 81 structural variations were considered and their physical properties such as solubility (logS), blood-brain partitioning (logBB), and probability of entering the CNS calculated by the Central Nervous System-Multiparameter Optimization (MPO-CNS) algorithm were evaluated. From this initial list, 18 compounds were further evaluated for anti-glioblastoma activity in vitro. Nine compounds demonstrated desirable glioblastoma cell toxicity in cell culture, and two of them, HR51, and HR59 demonstrated significantly improved capability of crossing the model blood-brain-barrier (BBB) composed of endothelial cells, astrocytes and pericytes.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Antineoplásicos Alquilantes/farmacología , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/patología , Células Endoteliales/metabolismo , Glioblastoma/patología , Humanos , Temozolomida/farmacología
7.
Front Immunol ; 12: 785905, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34917094

RESUMEN

Persons living with HIV (PLWH) are at higher risk of developing secondary illnesses than their uninfected counterparts, suggestive of a dysfunctional immune system in these individuals. Upon exposure to pathogens, monocytes undergo epigenetic remodeling that results in either a trained or a tolerant phenotype, characterized by hyper-responsiveness or hypo-responsiveness to secondary stimuli, respectively. We utilized CD14+ monocytes from virally suppressed PLWH and healthy controls for in vitro analysis following polarization of these cells toward a pro-inflammatory monocyte-derived macrophage (MDM) phenotype. We found that in PLWH-derived MDMs, pro-inflammatory signals (TNFA, IL6, IL1B, miR-155-5p, and IDO1) dominate over negative feedback signals (NCOR2, GSN, MSC, BIN1, and miR-146a-5p), favoring an abnormally trained phenotype. The mechanism of this reduction in negative feedback involves the attenuated expression of IKZF1, a transcription factor required for de novo synthesis of RELA during LPS-induced inflammatory responses. Furthermore, restoring IKZF1 expression in PLWH-MDMs partially reinstated expression of negative regulators of inflammation and lowered the expression of pro-inflammatory cytokines. Overall, this mechanism may provide a link between dysfunctional immune responses and susceptibility to co-morbidities in PLWH with low or undetectable viral load.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Infecciones por VIH/inmunología , Factor de Transcripción Ikaros/metabolismo , Macrófagos/inmunología , Factor de Transcripción ReIA/metabolismo , Fármacos Anti-VIH/administración & dosificación , Estudios de Casos y Controles , Citocinas/metabolismo , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica/inmunología , VIH/inmunología , VIH/aislamiento & purificación , Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Voluntarios Sanos , Humanos , Inflamación/sangre , Inflamación/inmunología , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Factor de Transcripción ReIA/genética , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
8.
Commun Biol ; 4(1): 682, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083759

RESUMEN

An outbreak of the novel coronavirus SARS-CoV-2, the causative agent of Coronavirus Disease-2019 (COVID-19), a respiratory disease, has infected almost one hundred million people since the end of 2019, killed over two million, and caused worldwide social and economic disruption. Because the mechanisms of SARS-CoV-2 infection of host cells and its pathogenesis remain largely unclear, there are currently no antiviral drugs with proven efficacy. Besides severe respiratory and systematic symptoms, several comorbidities increase risk of fatal disease outcome. Therefore, it is required to investigate the impacts of COVID-19 on pre-existing diseases of patients, such as cancer and other infectious diseases. In the current study, we report that SARS-CoV-2 encoded proteins and some currently used anti-COVID-19 drugs are able to induce lytic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV), one of major human oncogenic viruses, through manipulation of intracellular signaling pathways. Our data indicate that those KSHV + patients especially in endemic areas exposure to COVID-19 or undergoing the treatment may have increased risks to develop virus-associated cancers, even after they have fully recovered from COVID-19.


Asunto(s)
Antivirales/farmacología , COVID-19/complicaciones , Herpesvirus Humano 8/fisiología , SARS-CoV-2/fisiología , Sarcoma de Kaposi/etiología , Activación Viral , Azitromicina/farmacología , Benzamidinas/farmacología , Línea Celular , Guanidinas/farmacología , Infecciones por Herpesviridae/inducido químicamente , Infecciones por Herpesviridae/etiología , Herpesvirus Humano 8/efectos de los fármacos , Humanos , Virus Oncogénicos/efectos de los fármacos , Virus Oncogénicos/fisiología , SARS-CoV-2/efectos de los fármacos , Sarcoma de Kaposi/inducido químicamente , Proteínas Virales/metabolismo , Activación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
9.
Front Cell Infect Microbiol ; 10: 587929, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33194830

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) represents the etiological agent for several human malignancies, including Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease (MCD), which are mostly seen in immunocompromised patients. In fact, KSHV has developed many strategies to hijack host immune response, including the regulation of inflammatory cytokine production. Interleukin-1 (IL-1) family represents a major mediator for inflammation and plays an important role in both innate and adaptive immunity. Furthermore, a broadening list of diseases has revealed the pathologic role of IL-1 mediated inflammation. In the current mini-review, we have summarized recent findings about how this oncogenic virus is able to manipulate the activities of IL-1 signaling pathway to facilitate disease progression. We also discuss the therapeutic potential of IL-1 blockade against KSHV-related diseases and several unsolved questions in this interesting field.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Interleucina-1 , Transducción de Señal
10.
J Med Virol ; 92(12): 3862-3867, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32436999

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) infection causes several human cancers, including Kaposi's sarcoma (KS), one of the most common AIDS-associated tumors. The involvement of the oral cavity represents one common clinical manifestation of AIDS-KS individuals with periodontal diseases and an oral carriage of a variety of pathogenic bacteria, including Porphyromonas gingivalis. In the current study, we report the clinical relevance of P. gingivalis and KSHV coinfection in the oral cavity of a cohort of HIV+ patients. Furthermore, we found that P. gingivalis conditioned medium or derived lipopolysaccharide effectively induced KSHV lytic reactivation from infected oral cells. This reactivation requires TLR4 as well as the activities of p38 and Jun N-terminal kinase- mitogen-activated protein kinase signaling pathways. Our findings reveal the mechanisms through which coinfected periodontal pathogens potentially promote oncogenic virus pathogenesis in the unique niche of immunocompromised patients.

11.
Drug Discov Today Dis Models ; 32(Pt A): 5-11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33692833

RESUMEN

Neurocognitive disorders associated with HIV-1 infection affect more than half of persons living with HIV (PLWH) under retroviral therapy. Understanding the molecular mechanisms and the complex cellular network communication underlying neurological dysfunction is critical for the development of an effective therapy. As with other neurological disorders, challenges to studying HIV infection of the brain include limited access to clinical samples and proper reproducibility of the complexity of brain networks in cellular and animal models. This review focuses on cellular models used to investigate various aspects of neurological dysfunction associated with HIV infection.

12.
J Vis Exp ; (154)2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31885371

RESUMEN

Human immunodeficiency virus (HIV) remains a major health concern despite the introduction of combined antiretroviral therapy (cART) in the mid-1990s. While antiretroviral therapy efficiently lowers systemic viral load and restores normal CD4+ T cell counts, it does not reconstitute a completely functional immune system. A dysfunctional immune system in HIV-infected individuals undergoing cART may be characterized by immune activation, early aging of immune cells, or persistent inflammation. These conditions, along with comorbid factors associated with HIV infection, add complexity to the disease, which cannot be easily reproduced in cellular and animal models. To investigate the molecular events underlying immune dysfunction in these patients, a system to culture and manipulate human primary monocytes in vitro is presented here. Specifically, the protocol allows for the culture and transfection of primary CD14+ monocytes obtained from HIV-infected individuals undergoing cART as well as from HIV-negative controls. The method involves isolation, culture, and transfection of monocytes and monocyte-derived macrophages. While commercially available kits and reagents are employed, the protocol provides important tips and optimized conditions for successful adherence and transfection of monocytes with miRNA mimics and inhibitors as well as with siRNAs.


Asunto(s)
Separación Celular/métodos , Monocitos/citología , Transfección , Animales , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Regulación hacia Abajo , Humanos , Activación de Macrófagos , Macrófagos/citología , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo , ARN Interferente Pequeño/metabolismo
13.
Virology ; 536: 16-19, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31394407

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) causes several cancers such as Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). PD-1/PD-Ls immune checkpoint molecules play important roles in cancer cell immune escape. The expression of PD-1/PD-Ls and their regulation by oncogenic viruses, in particular KSHV, remain largely undefined. Here we demonstrate strong PD-1/PD-L1/PD-L2 expression in KS tissues from a cohort of HIV + patients. We found that induction of KSHV lytic reactivation significantly upregulates PD-L1 expression on infected tumor cells, potentially through several major cellular signaling pathways and IL-1ß, which may represent a novel mechanism for virus-associated tumor cell immune escape.


Asunto(s)
Antígeno B7-H1/genética , Infecciones por VIH/genética , Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno/genética , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/genética , Sarcoma de Kaposi/genética , Adulto , Antígeno B7-H1/inmunología , Línea Celular Tumoral , Estudios de Cohortes , Coinfección , Regulación de la Expresión Génica , Células HEK293 , VIH/inmunología , VIH/patogenicidad , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Herpesvirus Humano 8/inmunología , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Masculino , Persona de Mediana Edad , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/virología , Transducción de Señal , Escape del Tumor/genética , Activación Viral , Latencia del Virus
14.
Viruses ; 8(2)2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26907327

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 viral microRNAs (miRNAs) that are expressed during latency. Research into KSHV miRNA function has suffered from a lack of genetic systems to study viral miRNA mutations in the context of the viral genome. We used the Escherichia coli Red recombination system together with a new bacmid background, BAC16, to create mutants for all known KSHV miRNAs. The specific miRNA deletions or mutations and the integrity of the bacmids have been strictly quality controlled using PCR, restriction digestion, and sequencing. In addition, stable viral producer cell lines based on iSLK cells have been created for wildtype KSHV, for 12 individual miRNA knock-out mutants (ΔmiR-K12-1 through -12), and for mutants deleted for 10 of 12 (ΔmiR-cluster) or all 12 miRNAs (ΔmiR-all). NGS, in combination with SureSelect technology, was employed to sequence the entire latent genome within all producer cell lines. qPCR assays were used to verify the expression of the remaining viral miRNAs in a subset of mutants. Induction of the lytic cycle leads to efficient production of progeny viruses that have been used to infect endothelial cells. Wt BAC16 and miR mutant iSLK producer cell lines are now available to the research community.


Asunto(s)
Herpesvirus Humano 8/genética , MicroARNs/genética , ARN Viral/genética , Sarcoma de Kaposi/virología , Eliminación de Secuencia , Herpesvirus Humano 8/metabolismo , Humanos , MicroARNs/metabolismo , ARN Viral/metabolismo
15.
Oncotarget ; 7(9): 10459-71, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26859574

RESUMEN

Kaposi's Sarcoma-associated Herpesvirus (KSHV) is the etiologic agent of several malignancies, including Kaposi's Sarcoma (KS), which preferentially arise in immunocompromised patients such as HIV+ subpopulation and lack effective therapeutic options. Heme oxygenase-1 (HO-1) has been reported as an important regulator of endothelial cell cycle control, proliferation and angiogenesis. HO-1 has also been found to be highly expressed in KSHV-infected endothelial cells and oral AIDS-KS lesions. We previously demonstrate that the multifunctional glycoprotein CD147 is required for KSHV/LANA-induced endothelial cell invasiveness. During the identification of CD147 controlled downstream genes by microarray analysis, we found that the expression of HO-1 is significantly elevated in both CD147-overexpressing and KSHV-infected HUVEC cells when compared to control cells. In the current study, we further identify the regulation of HO-1 expression and mediated cellular functions by both CD147 and KSHV-encoded LANA proteins. Targeting HO-1 by either RNAi or the chemical inhibitor, SnPP, effectively induces cell death of KSHV-infected endothelial cells (the major cellular components of KS) through DNA damage and necrosis process. By using a KS-like nude mouse model, we found that SnPP treatment significantly suppressed KSHV-induced tumorigenesis in vivo. Taken together, our data demonstrate the important role of HO-1 in the pathogenesis and tumorigenesis of KSHV-infected endothelial cells, the underlying regulatory mechanisms for HO-1 expression and targeting HO-1 may represent a promising therapeutic strategy against KSHV-related malignancies.


Asunto(s)
Basigina/metabolismo , Hemo-Oxigenasa 1/metabolismo , Herpesvirus Humano 8/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Sarcoma de Kaposi/patología , Animales , Antígenos Virales/metabolismo , Apoptosis , Carcinogénesis/efectos de los fármacos , Proliferación Celular , Daño del ADN/efectos de los fármacos , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Metaloporfirinas/farmacología , Ratones , Ratones Desnudos , Necrosis/inducido químicamente , Proteínas Nucleares/metabolismo , Protoporfirinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Sarcoma de Kaposi/virología
16.
Oncotarget ; 7(4): 3806-18, 2016 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-26675551

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of several human cancers, including Kaposi's sarcoma (KS), which preferentially arise in immunocompromised patients and lack effective therapeutic options. We have previously shown that KSHV or viral protein LANA up-regulates the glycoprotein CD147, thereby inducing primary endothelial cell invasiveness. In the current study, we identify the global network controlled by CD147 in KSHV-infected endothelial cells using Illumina microarray analysis. Among downstream genes, two specific metalloproteases, ADAMTS1 and 9, are strongly expressed in AIDS-KS tissues and contribute to KSHV-infected endothelial cell invasiveness through up-regulation of IL-6 and VEGF. By using a KS-like nude mouse model, we found that targeting CD147 and downstream ADAMTSs significantly suppressed KSHV-induced tumorigenesis in vivo. Taken together, targeting CD147 and associated proteins may represent a promising therapeutic strategy against these KSHV-related malignancies.


Asunto(s)
Proteínas ADAM/metabolismo , Basigina/metabolismo , Transformación Celular Viral , Endotelio Vascular/patología , Herpesvirus Humano 8/patogenicidad , Sarcoma de Kaposi/patología , Proteínas ADAM/genética , Proteína ADAMTS1 , Animales , Apoptosis , Basigina/genética , Western Blotting , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Endotelio Vascular/virología , Perfilación de la Expresión Génica , Humanos , Ratones , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Blood ; 126(26): 2821-31, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26531163

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) is a principal causative agent of primary effusion lymphoma (PEL) with a poor prognosis in immunocompromised patients. However, it still lacks effective treatment which urgently requires the identification of novel therapeutic targets for PEL. Here, we report that the hepatocyte growth factor (HGF)/c-MET pathway is highly activated by KSHV in vitro and in vivo. The selective c-MET inhibitor, PF-2341066, can induce PEL apoptosis through cell cycle arrest and DNA damage, and suppress tumor progression in a xenograft murine model. By using microarray analysis, we identify many novel genes that are potentially controlled by HGF/c-MET within PEL cells. One of the downstream candidates, ribonucleoside-diphosphate reductase subunit M2 (RRM2), also displays the promising therapeutic value for PEL treatment. Our findings provide the framework for development of HGF/c-MET-focused therapy and implementation of clinical trials for PEL patients.


Asunto(s)
Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Linfoma de Efusión Primaria/patología , Piperidinas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Adulto , Animales , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ensayo Cometa , Crizotinib , Daño del ADN/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Infecciones por Herpesviridae/complicaciones , Herpesvirus Humano 8 , Humanos , Immunoblotting , Huésped Inmunocomprometido , Linfoma de Efusión Primaria/inmunología , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirazoles , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
18.
Cancer Lett ; 362(2): 158-66, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25837851

RESUMEN

Kaposi sarcoma-associated herpesvirus (KSHV) is the etiologic agent for Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL), malignancies arising primarily in immunocompromised patients particularly AIDS-patients, which still lack effective therapy. Hyaluronan (HA) is a large glucuronic acid and has been found closely related to multiple functions in cancer cells, although its role in viral oncogenesis remains largely unknown. Here we provide first evidence that KSHV de novo infection induces HA production from primary endothelial cells through upregulation of HA synthase gene 1 (Has1) and a multifunctional glycoprotein, CD147. Further data demonstrate that KSHV-induced HA production requires viral latent protein, LANA (in particular functional domain A) and MAPK/ERK signaling activities. In functions, HA production is necessary for KSHV/LANA-induced primary endothelial cell invasion, a hallmark feature for KS development. For clinical relevance, our data indicate that the KSHV+ group has higher levels of HA and Has1 activities in its plasma than the KSHV- group of cohort HIV-infected patients. Together, our findings provide innovative insights into the mechanisms of oncogenic virus activation of HA production and its role in virus-associated malignancy pathogenesis, which may help to develop novel therapeutic strategies by targeting HA and related signaling.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/metabolismo , Síndrome de Inmunodeficiencia Adquirida/virología , Herpesvirus Humano 8/metabolismo , Herpesvirus Humano 8/patogenicidad , Ácido Hialurónico/biosíntesis , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Adulto , Anciano , Antígenos Virales/metabolismo , Basigina/metabolismo , Femenino , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , VIH/metabolismo , VIH/patogenicidad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Hialuronano Sintasas , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/patología , Regulación hacia Arriba , Adulto Joven
19.
Viruses ; 6(10): 4005-23, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25341664

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) microRNAs are encoded in the latency-associated region. Knockdown of KSHV miR-K12-3 and miR-K12-11 increased expression of lytic genes in BC-3 cells, and increased virus production from latently infected BCBL-1 cells. Furthermore, iSLK cells infected with miR-K12-3 and miR-K12-11 deletion mutant viruses displayed increased spontaneous reactivation and were more sensitive to inducers of reactivation than cells infected with wild type KSHV. Predicted binding sites for miR-K12-3 and miR-K12-11 were found in the 3'UTRs of the cellular transcription factors MYB, Ets-1, and C/EBPα, which activate RTA, the KSHV replication and transcription activator. Targeting of MYB by miR-K12-11 was confirmed by cloning the MYB 3'UTR downstream from the luciferase reporter. Knockdown of miR­K12-11 resulted in increased levels of MYB transcript, and knockdown of miR-K12-3 increased both C/EBPα and Ets-1 transcripts. Thus, miR-K12-11 and miR-K12-3 contribute to maintenance of latency by decreasing RTA expression indirectly, presumably via down-regulation of MYB, C/EBPα and Ets-1, and possibly other host transcription factors.


Asunto(s)
Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , MicroARNs/genética , Proteínas Virales/metabolismo , Línea Celular , Regulación hacia Abajo , Células Endoteliales/virología , Técnicas de Silenciamiento del Gen , Herpesvirus Humano 8/inmunología , Herpesvirus Humano 8/metabolismo , Humanos , MicroARNs/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Receptores Virales/fisiología , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Virales/genética , Internalización del Virus , Latencia del Virus
20.
PLoS One ; 9(7): e102314, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25010828

RESUMEN

Phosphorylation of sphingosine by sphingosine kinases (SphK1 and SphK2) generates sphingosine-1-phosphate (S1P), a bioactive sphingolipid which promotes cancer cell survival and tumor progression in vivo. We have recently reported that targeting SphK2 induces apoptosis for human primary effusion lymphoma (PEL) cell lines infected by the Kaposi's sarcoma-associated herpesvirus (KSHV), and this occurs in part through inhibition of canonical NF-κB activation. In contrast, pharmacologic inhibition of SphK2 has minimal impact for uninfected B-cell lines or circulating human B cells from healthy donors. Therefore, we designed additional studies employing primary human endothelial cells to explore mechanisms responsible for the selective death observed for KSHV-infected cells during SphK2 targeting. Using RNA interference and a clinically relevant pharmacologic approach, we have found that targeting SphK2 induces apoptosis selectively for KSHV-infected endothelial cells through induction of viral lytic gene expression. Moreover, this effect occurs through repression of KSHV-microRNAs regulating viral latency and signal transduction, including miR-K12-1 which targets IκBα to facilitate activation of NF-κB, and ectopic expression of miR-K12-1 restores NF-κB activation and viability for KSHV-infected endothelial cells during SphK2 inhibition. These data illuminate a novel survival mechanism and potential therapeutic target for KSHV-infected endothelial cells: SphK2-associated maintenance of viral latency.


Asunto(s)
Herpesvirus Humano 8/genética , MicroARNs/biosíntesis , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Latencia del Virus/genética , Apoptosis/genética , Células Endoteliales/metabolismo , Células Endoteliales/virología , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Cultivo Primario de Células , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...