Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Am J Cancer Res ; 14(4): 1545-1560, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38726264

RESUMO

Breast cancer represents the leading cancer type and leading cause of cancer-related death among women in the world. Triple-negative breast cancer (TNBC) is a subset of breast cancer with the poorest prognosis and still lacking of effective therapeutic options. We recently screened a natural product library and identified 3 new hit compounds with selective and prominent anti-TNBC activities on different subtype of TNBC cell lines. Interestingly, all of these 3 hit compounds belong to "cytoskeletal drugs" that target tubulin and microtubule function. Our data also showed that these hit compounds showed consistently effective on TNBC cells which are resistant to those currently used antimicrotubule agents such as Paclitaxel. RNA-Sequencing analyses revealed the anti-TNBC mechanisms of these hit compounds and identified a subset of new cellular factors commonly affected by hit compounds in different subtypes of TNBC cells. Among them, we demonstrated AHCYL1 and SPG21 as new microtubule-associated proteins, which were required for TNBC cell survival with clinical implication through tissue array analysis. Our studies provide new insights into the mechanisms of TNBC pathogenesis and offer promising therapeutic directions for this aggressive breast cancer.

2.
Int J Mol Sci ; 25(2)2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38255987

RESUMO

The complex interactions between cancer cells and their surrounding microenvironment are fundamental in determining tumor progression, response to therapy, and, ultimately, patient prognosis [...].


Assuntos
Neoplasias da Mama , Humanos , Feminino , Microambiente Tumoral
3.
J Cancer Educ ; 39(2): 111-117, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37957501

RESUMO

Arkansas has a high cancer burden, and a pressing need exists for more medical students to pursue oncology as a career. The Partnership in Cancer Research (PCAR) program provides a summer research experience at the University of Arkansas for Medical Sciences for 12 medical students who have completed their first year of medical training. A majority of participants spend time pursuing cancer research in basic science, clinical, or community-based research. Students report on their research progress in an interactive "Live from the Lab!" series and assemble a final poster presentation describing their findings. Other activities include participation in a moderated, cancer-patient support group online, lecture series on cancer topics, medical simulations, palliative care clinic visit, "Death Over Dinner" event, and an entrepreneurship competition. Students completed surveys over PCAR's first 2 years in operation to evaluate all aspects of the program. Surveys reveal that students enthusiastically embraced the program in its entirety. This was especially true of the medical simulations which received the highest evaluations. Most significantly, surveys revealed that the program increased cancer knowledge and participant confidence to perform cancer research.


Assuntos
Neoplasias , Estudantes de Medicina , Humanos , Currículo , Pesquisa , Oncologia/educação , Neoplasias/terapia , Avaliação de Programas e Projetos de Saúde
4.
J Hematol Oncol ; 16(1): 48, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-37143124

RESUMO

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.


Assuntos
Equinomicina , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Animais , Camundongos , Herpesvirus Humano 8/genética , Equinomicina/farmacologia , Equinomicina/uso terapêutico , Latência Viral/genética , Sarcoma de Kaposi/tratamento farmacológico , Sarcoma de Kaposi/metabolismo , Ciclo Celular , Histona Desmetilases com o Domínio Jumonji
5.
Viruses ; 15(4)2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-37112991

RESUMO

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.


Assuntos
Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Animais , Camundongos , Herpesvirus Humano 8/fisiologia , Histamina , Receptores Histamínicos/uso terapêutico , Carcinogênese , Transformação Celular Neoplásica
6.
Int J Mol Sci ; 24(3)2023 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-36768830

RESUMO

The anti-CSPG4 monoclonal antibodies (mAbs) have shown anti-tumor activity and therapeutic potential for treating breast cancer. In addition, CSPG4 is a dominant tumor-associated antigen that is also involved in normal-tissue development in humans. Therefore, the potential for off-tumor activity remains a serious concern when targeting CSPG4 therapeutically. Previous work suggested that glycans contribute to the binding of specific anti-CSPG4 antibodies to tumor cells, but the specificity and importance of this contribution are unknown. In this study, the reactivity of anti-CSPG4 mAbs was characterized with a peptide mimetic of carbohydrate antigens expressed in breast cancer. ELISA, flow cytometry, and microarray assays were used to screen mAbs for their ability to bind to carbohydrate-mimicking peptides (CMPs), cancer cells, and glycans. The mAb VT68.2 displayed a distinctly strong binding to a CMP (P10s) and bound to triple-negative breast cancer cells. In addition, VT68.2 showed a higher affinity for N-linked glycans that contain terminal fucose and fucosylated lactosamines. The functional assays demonstrated that VT68.2 inhibited cancer cell migration. These results define the glycoform reactivity of an anti-CSPG4 antibody and may lead to the development of less toxic therapeutic approaches that target tumor-specific glyco-peptides.


Assuntos
Proteoglicanas , Neoplasias de Mama Triplo Negativas , Humanos , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Anticorpos Monoclonais/farmacologia , Antígenos Glicosídicos Associados a Tumores , Proteínas de Membrana
7.
Cancer Genomics Proteomics ; 20(1): 1-8, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36581341

RESUMO

BACKGROUND/AIM: Prostate cancer (PCa) is one of the most common types of cancer in men. Prostate-specific antigen (PSA) is currently the only biomarker used to screen for the risk of developing PCa. Because PSA tests may show false positives, identifying novel PCa-specific biomarkers would improve prediction and diagnosis at an early stage. Previously, we identified a number of genes/microRNAs (miRNAs) in prostate tissue as potential biomarkers of chronic prostatitis in a rat model of chemical-induced prostatitis. The current study aimed to evaluate their potential for use as translational, diagnostic markers in humans. MATERIALS AND METHODS: We performed quantitative polymerase chain reaction analysis using pathologically clear (normal) or confirmed PCa tissue samples from the same patients (N=18 per group). RESULTS: Levels (relative fold changes) of bone morphogenetic protein 7 (BMP7) transcripts were significantly lower in PCa tissues, compared with clear tissues, in a paired t-test (p=0.0075). Although neural cell adhesion molecule 1 (NCAM1) transcripts tended to be altered in PCa tissues, statistically insignificant differences were observed (p=0.0521). No statistically significant differences were observed for the other genes/miRNAs analyzed in PCa tissues due to a high degree of individual variance in expression. CONCLUSION: Similar to the results previously observed in rats, changes in the levels of BMP7 and NCAM1 transcripts were evident in human PCa tissues, suggesting that these genes may serve as potential diagnostic biomarkers during the early stages of PCa. Further studies are needed to determine the potential use of these molecules as biomarkers.


Assuntos
MicroRNAs , Neoplasias da Próstata , Prostatite , Masculino , Humanos , Animais , Ratos , Próstata/metabolismo , Antígeno Prostático Específico/análise , Antígeno Prostático Específico/metabolismo , Prostatite/diagnóstico , Prostatite/metabolismo , Pesquisa Translacional Biomédica , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo
8.
Hepatology ; 77(6): 1943-1957, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36052732

RESUMO

BACKGROUND: Morreton virus (MORV) is an oncolytic Vesiculovirus , genetically distinct from vesicular stomatitis virus (VSV). AIM: To report that MORV induced potent cytopathic effects (CPEs) in cholangiocarcinoma (CCA) and hepatocellular carcinoma (HCC) in vitro models. APPROACH AND RESULTS: In preliminary safety analyses, high intranasal doses (up to 10 10 50% tissue culture infectious dose [TCID 50 ]) of MORV were not associated with significant adverse effects in immune competent, non-tumor-bearing mice. MORV was shown to be efficacious in a Hep3B hepatocellular cancer xenograft model but not in a CCA xenograft HuCCT1 model. In an immune competent, syngeneic murine CCA model, single intratumoral treatments with MORV (1 × 10 7 TCID 50 ) triggered a robust antitumor immune response leading to substantial tumor regression and disease control at a dose 10-fold lower than VSV (1 × 10 8 TCID 50 ). MORV led to increased CD8 + cytotoxic T cells without compensatory increases in tumor-associated macrophages and granulocytic or monocytic myeloid-derived suppressor cells. CONCLUSIONS: Our findings indicate that wild-type MORV is safe and can induce potent tumor regression via immune-mediated and immune-independent mechanisms in HCC and CCA animal models without dose limiting adverse events. These data warrant further development and clinical translation of MORV as an oncolytic virotherapy platform.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Terapia Viral Oncolítica , Camundongos , Humanos , Animais , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/patologia , Vesiculovirus , Modelos Animais de Doenças , Linhagem Celular Tumoral
9.
Front Oncol ; 12: 1042250, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36457491

RESUMO

It has long been known that oncolytic viruses wield their therapeutic capability by priming an inflammatory state within the tumor and activating the tumor immune microenvironment, resulting in a multifaceted antitumor immune response. Vaccine-derived viruses, such as measles and mumps, have demonstrated promising potential for treating human cancer in animal models and clinical trials. However, the extensive cost of manufacturing current oncolytic viral products makes them far out of reach for most patients. Here by analyzing the impact of intratumoral (IT) administrations of the trivalent live attenuated measles, mumps, and rubella viruses (MMR) vaccine, we unveil the cellular and molecular basis of MMR-induced anti-cancer activity. Strikingly, we found that IT delivery of low doses of MMR correlates with tumor control and improved survival in murine hepatocellular cancer and colorectal cancer models via increased tumor infiltration of CD8+ granzyme B+ T-cells and decreased macrophages. Moreover, our data indicate that MMR activates key cellular effectors of the host's innate and adaptive antitumor immunity, culminating in an immunologically coordinated cancer cell death. These findings warrant further work on the potential for MMR to be repurposed as safe and cost-effective cancer immunotherapy to impact cancer patients globally.

10.
Front Cell Infect Microbiol ; 12: 1049624, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36457850

RESUMO

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.


Assuntos
Herpesvirus Humano 8 , Humanos , Carcinogênese , Transformação Celular Neoplásica , Imunidade Adaptativa , Receptores de Interleucina-1
11.
Front Microbiol ; 13: 882520, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35516440

RESUMO

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.

12.
Antimicrob Agents Chemother ; 66(3): e0239521, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35041508

RESUMO

Recently, remdesivir and molnupiravir were approved for treating COVID-19 caused by SARS-CoV-2 infection. However, little is known about the impact of these drugs on other viruses preexisted in COVID-19 patients. Here we report that remdesivir but not molnupiravir induced lytic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV), two major oncogenic herpesviruses. Remdesivir induced mature virion production from latently infected cells. Mechanistic studies showed that remdesivir induced KSHV and EBV reactivation by regulating several intracellular signaling pathways.


Assuntos
Tratamento Farmacológico da COVID-19 , Infecções por Vírus Epstein-Barr , Herpesvirus Humano 8 , Monofosfato de Adenosina/análogos & derivados , Alanina/análogos & derivados , Herpesvirus Humano 4/fisiologia , Humanos , SARS-CoV-2 , Transdução de Sinais , Ativação Viral
13.
Commun Biol ; 4(1): 682, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083759

RESUMO

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.


Assuntos
Antivirais/farmacologia , COVID-19/complicações , Herpesvirus Humano 8/fisiologia , SARS-CoV-2/fisiologia , Sarcoma de Kaposi/etiologia , Ativação Viral , Azitromicina/farmacologia , Benzamidinas/farmacologia , Linhagem Celular , Guanidinas/farmacologia , Infecções por Herpesviridae/induzido quimicamente , Infecções por Herpesviridae/etiologia , Herpesvirus Humano 8/efeitos dos fármacos , Humanos , Vírus Oncogênicos/efeitos dos fármacos , Vírus Oncogênicos/fisiologia , SARS-CoV-2/efeitos dos fármacos , Sarcoma de Kaposi/induzido quimicamente , Proteínas Virais/metabolismo , Ativação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
14.
J Cell Mol Med ; 25(6): 2795-2805, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33539648

RESUMO

Although the Eukaryotic Translation Initiation Factor 4 Gamma 1 (EIF4G1) has been found overexpressed in a variety of cancers, its role in non-small cell lung cancers (NSCLC) pathogenesis especially in immunoregulatory functions, its clinical relevance and therapeutic potential remain largely unknown. By using cancer patients tissue assays, the results indicate that EIF4G1 expressional levels are much higher in NSCLC tissues than in adjacent or normal lung tissues, which are also associated with NSCLC patient survival. By using an RNA-Sequencing based pipeline, the data show that EIF4G1 has a significant association with immune checkpoint molecules such as PD-1/PD-L1 in NSCLC. EIF4G1 small-molecule inhibitors effectively repress NSCLC growth in cell culture and xenograft animal models. Protein array results identify the signature of proteins controlled by EIF4G1 in NSCLC cells, in which new candidates such as MUC1 and NRG1 are required for NSCLC survival and tumorigenesis with clinical relevance. Taken together, these results have for the first time demonstrated the immunoregulatory functions, clinical relevance and therapeutic potential of the EIF4G1 network in NSCLC, which may represent a promising and novel target to improve lung cancer treatment.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Fator de Iniciação Eucariótico 4G/genética , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Neoplasias Pulmonares/genética , Animais , Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Progressão da Doença , Fator de Iniciação Eucariótico 4G/metabolismo , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/metabolismo , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Med Virol ; 93(6): 4028-4032, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32648938

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) causes several human cancers, including Kaposi's sarcoma (KS) and primary effusion lymphoma, which are mostly seen in immunocompromised patients, such as human immunodefeciency virus (HIV)+ individuals. Tuberculosis (TB), caused by the bacterial pathogen Mycobacterium tuberculosis (Mtb), remains one of the deadliest infectious diseases in the world. The risk of developing TB is dramatically higher in people living with HIV than among those without HIV infection. Case reports link cutaneous or pulmonary KS in HIV+ patients with mycobacterial co-infections, however, impacts of Mtb infection or its products on KSHV-infected cells are not known. We report here that ESAT-6, a secreted Mtb virulence factor, induces viral reactivation from KSHV-infected cells. KSHV-infected pulmonary endothelial cells were resistant to ESAT-6 induced inhibition of cell growth. Our data demonstrate that Mtb virulence factors influence the biology of KSHV-infected cells, highlighting the need to study the interactions between these two pathogens commonly found in people living with HIV.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Herpesvirus Humano 8/fisiologia , Mycobacterium tuberculosis/genética , Sarcoma de Kaposi/virologia , Ativação Viral , Antígenos de Bactérias/farmacologia , Proteínas de Bactérias/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/microbiologia , Células Endoteliais/virologia , Regulação Viral da Expressão Gênica , Humanos , Pulmão/citologia , Mycobacterium tuberculosis/patogenicidade , Fatores de Virulência , Replicação Viral
16.
bioRxiv ; 2020 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-33024968

RESUMO

An outbreak of the novel coronavirus SARS-CoV-2, the causative agent of Coronavirus Disease-2019 (COVID-19), a respiratory disease, has infected over 34,000,000 people since the end of 2019, killed over 1,000,000, and caused worldwide social and economic disruption. Due to the mechanisms of SARS-CoV-2 infection to host cells and its pathogenesis remain largely unclear, there are currently no antiviral drugs with proven efficacy nor are there vaccines for its prevention. Besides severe respiratory and systematic symptoms, several comorbidities may also 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 have reported that SARS-CoV-2 encoded proteins and some anti-COVID-19 drugs currently used 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.

17.
Blood ; 136(19): 2175-2187, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-32518949

RESUMO

Primary effusion lymphoma (PEL) is an aggressive malignancy with poor prognosis even under chemotherapy. Kaposi sarcoma-associated herpesvirus (KSHV), one of the human oncogenic viruses, is the principal causative agent. Currently, there is no specific treatment for PEL; therefore, developing new therapies is of great importance. Sphingolipid metabolism plays an important role in determining the fate of tumor cells. Our previous studies have demonstrated that there is a correlation between sphingolipid metabolism and KSHV+ tumor cell survival. To further develop sphingolipid metabolism-targeted therapy, after screening a series of newly synthesized ceramide analogs, here, we have identified compounds with effective anti-PEL activity. These compounds induce significant PEL apoptosis, cell-cycle arrest, and intracellular ceramide production through regulation of ceramide synthesizing or ceramide metabolizing enzymes and dramatically suppress tumor progression without visible toxicity in vivo. These new compounds also increase viral lytic gene expression in PEL cells. Our comparative transcriptomic analysis revealed their mechanisms of action for inducing PEL cell death and identified a subset of novel cellular genes, including AURKA and CDCA3, controlled by sphingolipid metabolism, and required for PEL survival with functional validation. These data provide the framework for the development of promising sphingolipid-based therapies against this virus-associated malignancy.


Assuntos
Aurora Quinase A/metabolismo , Ceramidas/farmacologia , Herpesvirus Humano 8/patogenicidade , Linfoma de Efusão Primária/tratamento farmacológico , Sarcoma de Kaposi/complicações , Esfingolipídeos/farmacologia , Animais , Apoptose , Aurora Quinase A/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular , Ceramidas/química , Feminino , Perfilação da Expressão Gênica , Humanos , Linfoma de Efusão Primária/etiologia , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Sarcoma de Kaposi/virologia , Células Tumorais Cultivadas , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Med Virol ; 92(12): 3862-3867, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32436999

RESUMO

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.

19.
J Infect Dis ; 221(8): 1331-1341, 2020 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-31111897

RESUMO

Collectively, viruses are the principal cause of cancers arising in patients with immune dysfunction, including human immunodeficiency virus (HIV)-positive patients. Kaposi sarcoma (KS) etiologically linked to Kaposi sarcoma-associated herpesvirus (KSHV) continues to be the most common AIDS-associated tumor. The involvement of the oral cavity represents one of the most common clinical manifestations of this tumor. HIV infection incurs an increased risk among individuals with periodontal diseases and oral carriage of a variety of pathogenic bacteria. However, whether interactions involving periodontal bacteria and oncogenic viruses in the local environment facilitate replication or maintenance of these viruses in the oral cavity of HIV-positive patients remain largely unknown. We previously showed that pathogen-associated molecular patterns (PAMPs) from specific periodontal bacteria promoted KSHV entry into oral cells and subsequent establishment of latency. In the current study, we demonstrate that Staphylococcus aureus, one of common pathogens causing infection in HIV-positive patients, and its PAMPs can effectively induce KSHV lytic reactivation from infected oral cells, through the Toll-like receptor reactive oxygen species and cyclin D1-Dicer-viral microRNA axis. This investigation provides further clinical evidence about the relevance of coinfection due to these 2 pathogens in the oral cavities of a cohort HIV-positive patients and reveals novel mechanisms through which these coinfecting pathogens potentially promote virus-associated cancer development in the unique niche of immunocompromised patients.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Boca/microbiologia , Boca/virologia , Vírus Oncogênicos/fisiologia , Ativação Viral/fisiologia , Adulto , Idoso , Linhagem Celular , Feminino , Infecções por HIV/microbiologia , Infecções por HIV/virologia , Infecções por Herpesviridae/microbiologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Vírus Oncogênicos/genética , Sarcoma de Kaposi/virologia , Staphylococcus aureus/patogenicidade , Latência Viral/fisiologia , Replicação Viral/genética , Adulto Jovem
20.
PLoS Pathog ; 15(12): e1008156, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790497

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) causes several human cancers, such as Kaposi's sarcoma (KS) and primary effusion lymphoma (PEL). Current treatment options for KSHV infection and virus associated diseases are sometimes ineffective, therefore, more effectively antiviral agents are urgently needed. As a herpesvirus, lytic replication is critical for KSHV pathogenesis and oncogenesis. In this study, we have established a high-throughput screening assay by using an inducible KSHV+ cell-line, iSLK.219. After screening a compound library that consisted of 1280 Food and Drug Administration (FDA)-approved drugs, 15 hit compounds that effectively inhibited KSHV virion production were identified, most of which have never been reported with anti-KSHV activities. Interestingly, 3 of these drugs target histamine receptors or signaling. Our data further confirmed that antagonists targeting different histamine receptors (HxRs) displayed excellent inhibitory effects on KSHV lytic replication from induced iSLK.219 or BCBL-1 cells. In contrast, histamine and specific agonists of HxRs promoted viral lytic replication from induced iSLK.219 or KSHV-infected primary cells. Mechanistic studies indicated that downstream MAPK and PI3K/Akt signaling pathways were required for histamine/receptors mediated promotion of KSHV lytic replication. Direct knockdown of HxRs in iSLK.219 cells effectively blocked viral lytic gene expression during induction. Using samples from a cohort of HIV+ patients, we found that the KSHV+ group has much higher levels of histamine in their plasma and saliva than the KSHV- group. Taken together, our data have identified new anti-KSHV agents and provided novel insights into the molecular bases of host factors that contribute to lytic replication and reactivation of this oncogenic herpesvirus.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 8/efeitos dos fármacos , Histamina/metabolismo , Sarcoma de Kaposi/virologia , Ativação Viral/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Herpesvirus Humano 8/fisiologia , Ensaios de Triagem em Larga Escala , Humanos , Receptores Histamínicos/metabolismo , Transdução de Sinais/fisiologia , Ativação Viral/fisiologia , Latência Viral/efeitos dos fármacos , Latência Viral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA