Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1286750, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022679

RESUMO

The ability to expand and activate natural Killer (NK) cells ex vivo has dramatically changed the landscape in the development of novel adoptive cell therapies for treating cancer over the last decade. NK cells have become a key player for cancer immunotherapy due to their innate ability to kill malignant cells while not harming healthy cells, allowing their potential use as an "off-the-shelf" product. Furthermore, recent advancements in NK cell genetic engineering methods have enabled the efficient generation of chimeric antigen receptor (CAR)-expressing NK cells that can exert both CAR-dependent and antigen-independent killing. Clinically, CAR-NK cells have shown promising efficacy and safety for treating CD19-expressing hematologic malignancies. While the number of pre-clinical studies using CAR-NK cells continues to expand, it is evident that solid tumors pose a unique challenge to NK cell-based adoptive cell therapies. Major barriers for efficacy include low NK cell trafficking and infiltration into solid tumor sites, low persistence, and immunosuppression by the harsh solid tumor microenvironment (TME). In this review we discuss the barriers posed by the solid tumor that prevent immune cell trafficking and NK cell effector functions. We then discuss promising strategies to enhance NK cell infiltration into solid tumor sites and activation within the TME. This includes NK cell-intrinsic and -extrinsic mechanisms such as NK cell engineering to resist TME-mediated inhibition and use of tumor-targeted agents such as oncolytic viruses expressing chemoattracting and activating payloads. We then discuss opportunities and challenges for using combination therapies to extend NK cell therapies for the treatment of solid tumors.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Microambiente Tumoral , Células Matadoras Naturais , Imunoterapia Adotiva/métodos
2.
NPJ Vaccines ; 8(1): 25, 2023 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-36823425

RESUMO

Viral-vectored vaccines are highly amenable for respiratory mucosal delivery as a means of inducing much-needed mucosal immunity at the point of pathogen entry. Unfortunately, current monovalent viral-vectored tuberculosis (TB) vaccine candidates have failed to demonstrate satisfactory clinical protective efficacy. As such, there is a need to develop next-generation viral-vectored TB vaccine strategies which incorporate both vaccine antigen design and delivery route. In this study, we have developed a trivalent chimpanzee adenoviral-vectored vaccine to provide protective immunity against pulmonary TB through targeting antigens linked to the three different growth phases (acute/chronic/dormancy) of Mycobacterium tuberculosis (M.tb) by expressing an acute replication-associated antigen, Ag85A, a chronically expressed virulence-associated antigen, TB10.4, and a dormancy/resuscitation-associated antigen, RpfB. Single-dose respiratory mucosal immunization with our trivalent vaccine induced robust, sustained tissue-resident multifunctional CD4+ and CD8+ T-cell responses within the lung tissues and airways, which were further quantitatively and qualitatively improved following boosting of subcutaneously BCG-primed hosts. Prophylactic and therapeutic immunization with this multivalent trivalent vaccine in conventional BALB/c mice provided significant protection against not only actively replicating M.tb bacilli but also dormant, non-replicating persisters. Importantly, when used as a booster, it also provided marked protection in the highly susceptible C3HeB/FeJ mice, and a single respiratory mucosal inoculation was capable of significant protection in a humanized mouse model. Our findings indicate the great potential of this next-generation TB vaccine strategy and support its further clinical development for both prophylactic and therapeutic applications.

3.
Viruses ; 14(9)2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-36146734

RESUMO

In people living with HIV, Mycobacterium tuberculosis (Mtb) is the major cause of death. Due to the increased morbidity/mortality in co-infection, further research is urgently required. A limiting factor to research in HIV and HIV/Mtb co-infection is the lack of accessible in vivo models. Next-generation humanized mice expressing HLA transgenes report improved human immune reconstitution and functionality, which may better recapitulate human disease. This study compares well-established huNRG mice and next-generation HLA I/II-transgenic (huDRAG-A2) mice for immune reconstitution, disease course, and pathology in HIV and TB. HuDRAG-A2 mice have improved engraftment of key immune cell types involved in HIV and TB disease. Upon intravaginal HIV-1 infection, both models developed significant HIV target cell depletion in the blood and tissues. Upon intranasal Mtb infection, both models sustained high bacterial load within the lungs and tissue dissemination. Some huDRAG-A2 granulomas appeared more classically organized, characterized by focal central necrosis, multinucleated giant cells, and foamy macrophages surrounded by a halo of CD4+ T cells. HIV/Mtb co-infection in huNRG mice trended towards worsened TB pathology and showed potential for modeling co-infection. Both huNRG and huDRAG-A2 mice are viable options for investigating HIV and TB, but the huDRAG-A2 model may offer advantages.


Assuntos
Coinfecção , Infecções por HIV , Mycobacterium tuberculosis , Tuberculose , Animais , Linfócitos T CD4-Positivos , Modelos Animais de Doenças , Humanos , Camundongos
4.
Cell ; 185(5): 896-915.e19, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35180381

RESUMO

The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.


Assuntos
Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Imunidade nas Mucosas , Administração Intranasal , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , COVID-19/virologia , Vacinas contra COVID-19/imunologia , Citocinas/sangue , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vetores Genéticos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Testes de Neutralização , Nucleocapsídeo/genética , Nucleocapsídeo/imunologia , Nucleocapsídeo/metabolismo , Pan troglodytes , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Breast Cancer Res ; 19(1): 76, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28668076

RESUMO

BACKGROUND: Natural killer (NK) cells play a critical role in cancer immunosurveillance. Recent developments in NK cell ex-vivo expansion makes it possible to generate millions of activated NK cells from a small volume of peripheral blood. We tested the functionality of ex vivo expanded NK cells in vitro against breast cancer cell lines and in vivo using a xenograft mouse model. The study aim was to assess functionality and phenotype of expanded NK cells from breast cancer patients against breast cancer cell lines and autologous primary tumours. METHODS: We used a well-established NK cell co-culture system to expand NK cells ex vivo from healthy donors and breast cancer patients and examined their surface marker expression. Moreover, we tested the ability of expanded NK cells to lyse the triple negative breast cancer and HER2-positive breast cancer cell lines MDA-MB-231 and MDA-MB-453, respectively. We also tested their ability to prevent tumour growth in vivo using a xenograft mouse model. Finally, we tested the cytotoxicity of expanded NK cells against autologous and allogeneic primary breast cancer tumours in vitro. RESULTS: After 3 weeks of culture we observed over 1000-fold expansion of NK cells isolated from either breast cancer patients or healthy donors. We also showed that the phenotype of expanded NK cells is comparable between those from healthy donors and cancer patients. Moreover, our results confirm the ability of ex vivo expanded NK cells to lyse tumour cell lines in vitro. While the cell lines examined had differential sensitivity to NK cell killing we found this was correlated with level of major histocompatibility complex (MHC) class I expression. In our in vivo model, NK cells prevented tumour establishment and growth in immunocompromised mice. Finally, we showed that NK cells expanded from the peripheral blood of breast cancer patients show high cytotoxicity against allogeneic and autologous patient-derived tumour cells in vitro. CONCLUSION: NK cells from breast cancer patients can be expanded similarly to those from healthy donors, have a high cytotoxic ability against breast cancer cell lines and patient-derived tumour cells, and can be compatible with current cancer treatments to restore NK cell function in cancer patients.


Assuntos
Neoplasias da Mama/imunologia , Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Animais , Biomarcadores , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Técnicas de Cocultura , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Imunofenotipagem , Imunoterapia Adotiva , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Knockout , Receptor ErbB-2/metabolismo
6.
J Leukoc Biol ; 101(1): 285-295, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27493241

RESUMO

The crosstalk between NK cells and M1 macrophages has a vital role in the protection against infections and tumor development. However, macrophages in the tumor resemble an M2 phenotype, and, at present, their effect on NK cells is less clear. This study investigated whether tumor-associated macrophages (TAMs) have a role in altering NK cell function and phenotype using in vitro cocultures of murine NK cells with peritoneal or bone marrow-derived, M2-polarized macrophages or TAMs isolated from spontaneous mouse breast tumors. We report here that both peritoneal and bone marrow-derived M2 macrophages, as well as TAMs, substantially inhibit NK cell activation and concordant cytotoxicity against tumor cells. The mechanism for this inhibition was found to require contact between the respective cell types. Both M2 macrophages and TAMs are producers of the immunosuppressive cytokine TGF-ß. The inhibition of TGF-ß restored the cytotoxicity of NK cells in contact with M2 macrophages, implicating TGF-ß in the mechanism for NK cell inhibition. In addition to affecting NK cell function, TAMs also induced a CD27lowCD11bhigh-exhausted NK cell phenotype, which corresponds with the reduced activation and cytotoxicity observed. This study reveals a novel implication of TAMs in the tumor-associated inhibition of NK cell function by demonstrating their capacity to directly alter NK cell cytotoxicity and phenotype in a contact-dependent mechanism involving TGF-ß. These findings identify the interaction between NK cells and TAMs as a prospective therapeutic target to enhance NK cell effector function for effective NK cell cancer therapies.


Assuntos
Comunicação Celular , Polaridade Celular , Células Matadoras Naturais/patologia , Macrófagos Peritoneais/patologia , Neoplasias/patologia , Animais , Células da Medula Óssea/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Técnicas de Cocultura , Citotoxicidade Imunológica , Células Matadoras Naturais/metabolismo , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Fenótipo , Fator de Crescimento Transformador beta/metabolismo , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
7.
BMC Cancer ; 16: 555, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27469239

RESUMO

BACKGROUND: Lymph node (LN) status is the most important prognostic variable used to guide ER positive (+) breast cancer treatment. While a positive nodal status is traditionally associated with a poor prognosis, a subset of these patients respond well to treatment and achieve long-term survival. Several gene signatures have been established as a means of predicting outcome of breast cancer patients, but the development and indication for use of these assays varies. Here we compare the capacity of two approved gene signatures and a third novel signature to predict outcome in distinct LN negative (-) and LN+ populations. We also examine biological differences between tumours associated with LN- and LN+ disease. METHODS: Gene expression data from publically available data sets was used to compare the ability of Oncotype DX and Prosigna to predict Distant Metastasis Free Survival (DMFS) using an in silico platform. A novel gene signature (Ellen) was developed by including patients with both LN- and LN+ disease and using Prediction Analysis of Microarrays (PAM) software. Gene Set Enrichment Analysis (GSEA) was used to determine biological pathways associated with patient outcome in both LN- and LN+ tumors. RESULTS: The Oncotype DX gene signature, which only used LN- patients during development, significantly predicted outcome in LN- patients, but not LN+ patients. The Prosigna gene signature, which included both LN- and LN+ patients during development, predicted outcome in both LN- and LN+ patient groups. Ellen was also able to predict outcome in both LN- and LN+ patient groups. GSEA suggested that epigenetic modification may be related to poor outcome in LN- disease, whereas immune response may be related to good outcome in LN+ disease. CONCLUSIONS: We demonstrate the importance of incorporating lymph node status during the development of prognostic gene signatures. Ellen may be a useful tool to predict outcome of patients regardless of lymph node status, or for those with unknown lymph node status. Finally we present candidate biological processes, unique to LN- and LN+ disease, that may indicate risk of relapse.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Metástase Linfática/genética , Transcriptoma , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Feminino , Perfilação da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática/patologia , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Modelos de Riscos Proporcionais , Receptor ErbB-2/biossíntese , Receptores de Estrogênio/biossíntese , Tamoxifeno/uso terapêutico
8.
Cell Mol Immunol ; 13(5): 628-39, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26277898

RESUMO

Natural killer (NK) cells are innate immune cells with the ability to identify and eliminate transformed cells. However, within tumors, many studies have described NK cells as non-functional. The developmental stage of tumor-associated NK cells and how this may relate to functionality has not been explored. We examined the developmental state of NK cells from polyoma middle T antigen (pyMT) transgenic mouse (MMTV-pMT) breast tumors. In pyMT tumors, NK cells were immature as evidenced by their decreased expression of DX5 and their CD27(low)CD11b(low) phenotype. These immature NK cells also had increased expression of NKG2A and expressed low levels of NKp46, perforin, and granzyme B. In contrast, splenic NK cells isolated from the same mice maintained their maturity and their expression of activation markers. To delineate whether the tumor microenvironment directly alters NK cells, we adoptively transferred labeled NK cells and followed their activation status in both the spleen and the tumor. NK cells that arrived at the tumor had half the expression of NKp46 within three days of transfer in comparison to those which arrived at the spleen. In an effort to modify the tumor microenvironment and assess the plasticity of intratumoral NK cells, we treated pyMT tumors with IL-12 and anti-TGF-ß. After one week of treatment, the maturity of tumor-associated NK cells was increased; thus, indicating that these cells possess the ability to mature and become activated. A better understanding of how NK cells are modified by the tumor microenvironment will help to develop strategies aimed at bolstering immune responses against tumors.


Assuntos
Células Matadoras Naturais/patologia , Neoplasias Mamárias Animais/patologia , Microambiente Tumoral , Transferência Adotiva , Animais , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Antígenos Transformantes de Poliomavirus/metabolismo , Diferenciação Celular , Feminino , Granzimas/metabolismo , Interleucina-12/metabolismo , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Perforina/metabolismo , Fenótipo , Baço/patologia , Fator de Crescimento Transformador beta/metabolismo
9.
BMC Cancer ; 15: 293, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25879689

RESUMO

BACKGROUND: Natural Killer (NK) cells play an important role in tumor prevention, but once tumors form, the numbers as well as the cytotoxic functions of NK cells are reduced. IL-15 is a cytokine that increases and activates NK cells. Here we will examine the anti-tumor role of IL-15 in a spontaneous breast cancer model. METHODS: To achieve this, Polyoma Middle T (MT) mice that form spontaneous breast cancer were crossed with mice that either overexpress IL-15 (IL-15 transgenic (TG)) or mice that lack IL-15 (IL-15 knockout (KO)). We compared survival curves and tumor formation in IL-15 KO/MT, MT and IL-15 TG/MT groups. In addition, the phenotype, activation and contribution of NK cells and CD8 T cells to tumor formation were examined in each of these mouse strains via flow cytometry, ELISA, adoptive transfer and antibody depletion experiments. RESULTS: IL-15KO/MT tumors formed and progressed to endpoint more quickly than MT tumors. These tumors displayed little apoptosis and poor CD8 T cell infiltration. In contrast, IL-15 TG/MT mice had increased survival and the tumors displayed extensive cell death, high proportions of activated NK cells and a higher infiltration of CD8 T cells than MT tumors. CD8 T cells in IL-15 TG/MT tumors were capable of secreting IFNγ, possessed markers of memory, did not display an exhausted phenotype and were frequently NK1.1+. Long-term antibody depletion studies in IL-15 TG/MT mice revealed that NK1.1+, but not CD8 T cells, were critical for tumor destruction. Lastly, human NK cells, when exposed to a similar cytokine environment as that found in IL-15TG/MT tumors, were capable of killing human breast cancer cells. CONCLUSIONS: This study reveals that high levels of IL-15 can promote tumor destruction and reduce metastasis in breast cancer via effects on NK1.1+ cells. Our results suggest that strategies aimed at increasing NK cell activation may be effective against solid epithelial cancers.


Assuntos
Interleucina-15/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Mamárias Animais/metabolismo , Animais , Antígenos Ly/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-15/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Ativação Linfocitária , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo
10.
PLoS One ; 9(12): e114799, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25517731

RESUMO

Interleukin-15 (IL-15) is an immunomodulatory cytokine that affects body mass regulation independent of lymphocytes; however, the underlying mechanism(s) involved remains unknown. In an effort to investigate these mechanisms, we performed metabolic cage studies, assessed intestinal bacterial diversity and macronutrient absorption, and examined adipose mitochondrial activity in cultured adipocytes and in lean IL-15 transgenic (IL-15tg), overweight IL-15 deficient (IL-15-/-), and control C57Bl/6 (B6) mice. Here we show that differences in body weight are not the result of differential activity level, food intake, or respiratory exchange ratio. Although intestinal microbiota differences between obese and lean individuals are known to impact macronutrient absorption, differing gut bacteria profiles in these murine strains does not translate to differences in body weight in colonized germ free animals and macronutrient absorption. Due to its contribution to body weight variation, we examined mitochondrial factors and found that IL-15 treatment in cultured adipocytes resulted in increased mitochondrial membrane potential and decreased lipid deposition. Lastly, IL-15tg mice have significantly elevated mitochondrial activity and mass in adipose tissue compared to B6 and IL-15-/- mice. Altogether, these results suggest that IL-15 is involved in adipose tissue regulation and linked to altered mitochondrial function.


Assuntos
Tecido Adiposo/citologia , Interleucina-15/metabolismo , Mitocôndrias/metabolismo , Tamanho Mitocondrial , Células 3T3-L1 , Animais , Peso Corporal , Quimiocinas/biossíntese , Feminino , Regulação da Expressão Gênica , Humanos , Interleucina-15/deficiência , Interleucina-6/biossíntese , Intestinos/microbiologia , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Transgênicos , Microbiota , Sobrepeso/metabolismo , Sobrepeso/patologia
11.
J Immunol ; 193(12): 6184-91, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25355926

RESUMO

IL-15 is a cytokine that can affect many immune cells, including NK cells and CD8 T cells. In several tumor models, IL-15 delays primary tumor formation and can prevent or reduce metastasis. In this study, we have employed a model of breast cancer metastasis to examine the mechanism by which IL-15 affects metastasis. When breast tumor cells were injected i.v. into IL-15(-/-), C57BL/6, IL-15 transgenic (TG) and IL-15/IL-15Rα-treated C57BL/6 mice, there were high levels of metastasis in IL-15(-/-) mice and virtually no metastasis in IL-15 TG or IL-15-treated mice. In fact, IL-15(-/-) mice were 10 times more susceptible to metastasis, whereas IL-15 TG mice were at least 10 times more resistant to metastasis when compared with control C57BL/6 mice. Depletion of NK cells from IL-15 TG mice revealed that these cells were important for protection from metastasis. When NK cells were depleted from control C57BL/6 mice, these mice did not form as many metastatic foci as IL-15(-/-) mice, suggesting that other cell types may be contributing to metastasis in the absence of IL-15. We then examined the role of CD4 T cells and macrophages. In IL-15(-/-) mice, in vivo depletion of CD4 T cells decreased metastasis. The lack of IL-15 in IL-15(-/-) mice, and possibly the Th2-polarized CD4 T cells, was found to promote the formation of M2 macrophages that are thought to contribute to metastasis formation. This study reveals that whereas IL-15 effects on NK cells are important, it also has effects on other immune cells that contribute to metastasis.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linfócitos T CD4-Positivos/metabolismo , Expressão Gênica , Interleucina-15/genética , Células Matadoras Naturais/metabolismo , Macrófagos/metabolismo , Animais , Neoplasias da Mama/imunologia , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Interleucina-15/administração & dosagem , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Macrófagos/imunologia , Neoplasias Mamárias Experimentais , Melanoma Experimental , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica
12.
Antiviral Res ; 92(2): 346-55, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21945041

RESUMO

Fimbriae H protein (FimH) is a novel TLR4 ligand that has been shown to stimulate the innate immune system and elicits protective responses against bacterial and viral infections. Here, we evaluated the protective role of local delivery of FimH against influenza A infection in a mouse model. We show that intranasal delivery of FimH prior to lethal challenge with influenza A virus, resulted in decreased morbidity and mortality in wild-type, but not TLR4(-/-), mice. Importantly, FimH was able to reduce the early viral burden in the lung leading to minimal cell infiltration into the airway lumen and reduced pulmonary pathology following infection in wild type mice compared to TLR4(-/-) mice. Local delivery of FimH to C57BL/6, not TLR4(-/-), mice in a prophylactic manner increased the IL-12 and RANTES responses as well as neutrophil recruitment into the airway lumen. These effects correlate to the course of influenza infection. The FimH-mediated antiviral response against influenza virus appears to be partially dependent on alveolar macrophages. The antiviral effects are likely mediated by the innate mediators (TNF-α, IL-12 or RANTES) and/or by activation of a feedback inhibition loop to curtail the pulmonary inflammation possibly be the potential mechanisms involved in FimH-mediated protection. FimH thus holds promise to be a possible prophylactic mean of control against influenza viral infection.


Assuntos
Adesinas de Escherichia coli/administração & dosagem , Adesinas de Escherichia coli/imunologia , Proteínas de Fímbrias/administração & dosagem , Proteínas de Fímbrias/imunologia , Imunidade Inata/efeitos dos fármacos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Receptor 4 Toll-Like/administração & dosagem , Receptor 4 Toll-Like/imunologia , Administração Intranasal , Animais , Movimento Celular , Quimiocina CCL5/metabolismo , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Interleucina-12/metabolismo , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/patologia , Análise de Sobrevida , Fator de Necrose Tumoral alfa/metabolismo
13.
Expert Rev Clin Immunol ; 7(3): 367-82, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21595603

RESUMO

Current cancer immunotherapies have begun to target cell types involved in innate immunity, such as natural killer (NK) cells that recognize and kill tumor cells. Recent advances in the study of NK cell biology have generated interest in manipulating these cells to generate anti-tumor responses. A rise in the number of activated NK cells has been shown to prevent and treat cancer in many preclinical models and is a positive clinical factor in human tumors. This article will focus on recent research on the ability of IL-15 and Toll-like receptor ligands to stimulate NK cell activity against cancer. The potential of these therapies, both alone and in conjunction with traditional and other vaccine platforms, will be reviewed. The current status of these therapies in clinical trials will also be discussed. Targeting these cell types in the context of human cancers may be an essential factor in future cancer treatments.


Assuntos
Imunoterapia/tendências , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Humanos , Células Matadoras Naturais/citologia , Neoplasias/prevenção & controle
14.
Am J Reprod Immunol ; 63(4): 299-309, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20070285

RESUMO

PROBLEM: Previously we reported that ovariectomized (OVX) mice receiving estradiol (E) prior to immunization with an attenuated strain of HSV-2 (TK-HSV-2) were not protected. Lack of protection in the E group was because of the inability of TK-HSV-2 to penetrate the thick keratinized epithelium. In this study, we determined the outcome of immunization after the thickening of vaginal epithelium following E-treatment waned. OVX, C57BL/6 mice were given Progesterone (P), E or saline (S) for 3 days and immunized with IVAG TK-HSV-2. METHOD OF STUDY: To determine the time point at which E-treated mice could be successfully immunized, the mice were inoculated with TK-HSV-2 between days 1 and 7 (ED1-ED7) post-E-treatment and challenged with IVAG HSV-2 three weeks later. RESULTS: The level of infection post-immunization correlated with HSV-2-specific IgG antibody level, which correlated with sterile protection. No viral infection was observed in ED1-ED3 groups and no specific antibodies were detected, resulting in no protection. Moderate infection was seen in ED5 group, resulting in low antibody production and non-sterile protection in 87.5% of mice. High antibody titers and sterile protection were observed in all groups that experienced robust infection post-immunization. CONCLUSION: The results show that estradiol leads to limited viral replication and diminished mucosal IgG response, resulting in non-sterile immune protection against genital herpes infection.


Assuntos
Estradiol/fisiologia , Herpes Genital/imunologia , Herpesvirus Humano 2/imunologia , Imunoglobulina G/imunologia , Replicação Viral , Administração Intravaginal , Animais , Feminino , Herpes Genital/prevenção & controle , Herpes Genital/virologia , Imunização , Camundongos , Camundongos Endogâmicos C57BL , Mucosa/imunologia
15.
Biol Reprod ; 77(6): 1049-59, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17881767

RESUMO

Genital epithelial cells (ECs) are the first line of defense that sexually transmitted viruses encounter. The mechanism of viral pathogenesis in these cells is not well understood. Here, we show that a primary cell culture model from human reproductive tract tissues can be used as a novel ex vivo model in examining the interaction of herpes simplex virus, type 2 (HSV-2), with female genital mucosa. Confluent, polarized primary cultures of human endometrial and cervical ECs were established and shown to be free from any significant contamination of any other cell type. Both endometrial and cervical ECs were found to be highly susceptible to HSV-2 infection. The kinetic of infection was similar to in vivo infection, with the earliest viral shedding seen at 18 h postinfection. Primary EC monolayers could be infected both apically and basolaterally, but preferential viral shedding was seen on the apical side of cells. Following treatment of the monolayers with poly (I:C), an innate immune activator that acts via TLR3, viral shedding was reduced significantly, comparable to levels seen when an antiviral formulation, acyclovir, was used. Treatment of epithelial and stromal co-cultures with estradiol increased HSV-2 infection in endometrial ECs, but viral shedding decreased following treatment with progesterone. To the best of our knowledge, this is the first study that examines the interaction of primary human female genital ECs with HSV-2, using an ex vivo culture model. The results provide valuable information regarding the susceptibility of women's genital ECs to HSV-2 and the ability of innate immunity and hormones to modify this susceptibility.


Assuntos
Colo do Útero/virologia , Endométrio/virologia , Células Epiteliais/virologia , Herpes Genital/virologia , Herpesvirus Humano 2/fisiologia , Antivirais/farmacologia , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Herpesvirus Humano 2/efeitos dos fármacos , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Poli I-C/farmacologia , Progesterona/farmacologia , Células Estromais/virologia , Receptor 3 Toll-Like/agonistas
16.
Am J Reprod Immunol ; 57(5): 367-77, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17430501

RESUMO

PROBLEM: This study was undertaken to evaluate the susceptibility of upper and lower reproductive tract epithelial cells (ECs) to herpes simplex virus type 2 (HSV-2) infection and examine their cytokine secretion patterns prior to and following infection. METHOD OF STUDY: Primary EC cultures, grown from murine vaginal and uterine tissue, were inoculated with HSV-2. Viral shedding was measured in apical and basolateral compartments. Multi-analyte bead-based immunoassays run on Luminex, were used to analyse cytokine profiles. RESULTS: Both vaginal and uterine ECs became productively infected with HSV-2, ex-vivo. Uterine ECs displayed varying degrees of infection, dependent on transepithelial resistance of the monolayers. Co-culturing stromal cells did not significantly change levels of viral shedding from ECs. Uterine ECs and epithelial-stromal co-cultures constitutively secreted interleukin (IL)-1alpha, IL-6, mouse homologue of human IL-8 (KC) and monocyte chemotactic protein-1 (MCP-1), while vaginal epithelial-stromal co-cultures secreted granulocyte-macrophage colony stimulating factor (GM-CSF) and KC. Following exposure to HSV-2, IL-6 and MCP-1 levels decreased in uterine EC cultures. CONCLUSIONS: This data shows that ECs from the upper and lower reproductive tract have different cytokine secretion profiles and respond differentially to infection. HSV-2 may be able to suppress epithelial cytokine secretion as a strategy to evade host immune system.


Assuntos
Citocinas/metabolismo , Células Epiteliais/virologia , Herpes Genital/virologia , Herpesvirus Humano 2 , Útero/virologia , Vagina/virologia , Animais , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CXCL1 , Quimiocinas CXC/metabolismo , Técnicas de Cocultura , Células Epiteliais/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Herpes Genital/metabolismo , Humanos , Interleucina-1alfa/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Estromais/metabolismo , Células Estromais/virologia , Fatores de Tempo , Útero/metabolismo , Vagina/metabolismo , Eliminação de Partículas Virais
17.
J Virol ; 79(5): 3107-16, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15709030

RESUMO

We report here that sex hormones modulate susceptibility to a sexually transmitted viral agent, herpes simplex virus type 2 (HSV-2), in a mouse model. Ovariectomized mice were administered either saline (control), estradiol (E(2)), progesterone (P(4)), or a combination of both estradiol and progesterone (E+P) and infected intravaginally with HSV-2. With an inoculation dose of 10(5) PFU, the saline- and P(4)-treated mice were found to be highly susceptible to genital HSV-2 infection. Both groups had extensive pathology and high viral titers in vaginal secretions, and 100% of mice succumbed by day 4 postinfection. E(2)-treated mice were protected from HSV-2 infection at the same dose and did not display any vaginal pathology or viral shedding. There was a slow progression of genital pathology in the combination hormone-treated group, along with prolonged viral shedding; 80% of animals succumbed by day 13. With lower inoculation doses of 10(3) and 10(2) PFU, 50 and 100%, respectively, of the combination hormone-treated mice survived. Localization of HSV-2 infection showed extensive infection in the vaginal epithelium of P(4)- and saline-treated animals within 24 h of inoculation. E(2)-treated animals were clear of infection, while the E+P-treated group had focal infection at 24 h that had progressed extensively by day 3. Infection was accompanied by persistent inflammation and infiltration of neutrophils in the P(4)-treated group. An analysis of the genes in the vaginal tissue showed that inflammation in the P(4)-treated group correlated with local induction of chemokines and chemokine receptors that were absent in the E(2)-treated mice and in uninfected P(4)-treated mice. The results show that sex hormones regulate initiation of infection and immune responses to genital HSV-2 infection.


Assuntos
Estradiol/farmacologia , Herpes Genital/etiologia , Herpesvirus Humano 2/efeitos dos fármacos , Herpesvirus Humano 2/patogenicidade , Progesterona/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Herpes Genital/patologia , Herpes Genital/virologia , Herpesvirus Humano 2/isolamento & purificação , Inflamação/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Vagina/patologia , Vagina/virologia , Virulência/efeitos dos fármacos
18.
J Virol ; 79(5): 3117-26, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15709031

RESUMO

The present study was undertaken to examine the effect of the hormonal environment on immunization with an attenuated strain of herpes simplex virus type 2 (HSV-2 TK(-)) and subsequent protection against challenge. Ovariectomized mice were administered saline (S; control), estradiol (E(2)), progesterone (P(4)), or a combination of estradiol and progesterone (E+P) and immunized intravaginally (IVAG) with HSV-2 TK(-). Three weeks later, the immunized mice were challenged IVAG with wild-type HSV-2. Mice that were immunized following E treatment were not protected, whereas complete protection against the challenge was seen in mice from the S- and P(4)-treated groups. In the P(4)-treated group, 15% of mice developed chronic pathology following TK(-) immunization. Interestingly, about 40% of the E+P-treated mice were also protected. Upon examination of viral shedding in the vaginal secretions, it was clear that protection against challenge was dependent on the ability of the TK(-) virus to cause productive genital infection under different hormonal conditions. In the protected mice (the S and P groups and part of the E+P group), induced vagina-associated lymphoid tissues composed of CD11c(+) dendritic cells and CD3(+) and CD4(+) T cells were formed transiently in the vaginal lamina propria from day 2 to day 5 postchallenge. These aggregates were absent in the unprotected mice (the E group and part of the E+P group). Significant HSV-2-specific activation of lymphocytes was observed in the local draining lymph nodes of protected mice. This response was absent in the unprotected groups. High titers of gB-specific local immunoglobulin A (IgA) antibodies were present in the vaginal secretions of S- and P(4)-treated immunized mice following HSV-2 challenge. The S-treated group of mice also had high gB-specific IgG titers. These studies show that sex hormones modify the induction of protective immune responses following IVAG immunization.


Assuntos
Herpes Genital/imunologia , Herpes Genital/prevenção & controle , Tecido Linfoide/imunologia , Vagina/imunologia , Animais , Anticorpos Antivirais/metabolismo , Estradiol/farmacologia , Feminino , Herpes Genital/patologia , Herpes Genital/virologia , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/imunologia , Herpesvirus Humano 2/patogenicidade , Imunização , Imunoglobulina A Secretora/metabolismo , Imunoglobulina G/metabolismo , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Progesterona/farmacologia , Linfócitos T/imunologia , Vacinas Atenuadas/farmacologia , Vagina/patologia , Vagina/virologia , Vacinas Virais/farmacologia
19.
Oncogene ; 22(57): 9151-5, 2003 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-14668796

RESUMO

Activation of the epidermal growth factor receptor (EGFR) family is thought to play an important role in mammary tumorigenesis and metastasis. The potent transforming activity of the EGFR family is due to their ability to heterodimerize with each other in response to a number of mitogenic ligands. The formation of EGFR and ErbB-2 heterodimers has been recently implicated as an important factor in the induction of sporadic human breast cancers. To directly assess whether the catalytic activity of EGFR is required for ErbB-2 induction of mammary tumors, we have interbred transgenic mice expressing ErbB-2 oncogene under the transcriptional control of the mouse mammary tumor virus (MMTV) promoter/enhancer to a naturally occurring mouse mutant carrying a catalytically impaired EGFR (waved-2 mice). Although the female transgenic mice possessing mutant EGFR developed mammary tumors, the tumors occurred only after a delayed latency period, and were fewer in number. The impaired tumor phenotype was further correlated with debilitated phosphorylation of the Gab1 multisubstrate adapter. These observations provide evidence that efficient ErbB-2-induced mammary tumor progression requires EGFR-dependent activation of Gab1.


Assuntos
Receptores ErbB/fisiologia , Neoplasias Mamárias Animais/genética , Fosfoproteínas/metabolismo , Receptor ErbB-2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Cruzamentos Genéticos , Progressão da Doença , Elementos Facilitadores Genéticos , Receptores ErbB/deficiência , Receptores ErbB/genética , Feminino , Neoplasias Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Fosfoproteínas/genética , Fosforilação , Regiões Promotoras Genéticas , Transcrição Gênica , Células Tumorais Cultivadas
20.
J Virol ; 77(18): 9845-51, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941893

RESUMO

Depo-Provera (Depo) is a long-acting progestational formulation that is a popular form of contraception for women. In animal models of sexually transmitted diseases, it is used to facilitate infection. Here we report that treatment with Depo, in a mouse model of genital herpes simplex virus type 2 (HSV-2), altered immune responses depending on the length of time that animals were exposed to Depo prior to immunization. Mice immunized intravaginally (i.vag.) with an attenuated strain (TK(-)) of HSV-2 following longer (15 days) exposure to Depo (Depo 15 group) failed to show protection when challenged with wild-type HSV-2. In contrast, mice that were immunized shortly after Depo treatment (5 days; Depo 5 group) were fully protected and showed no genital pathology after HSV-2 challenge. High viral titers were detected in the vaginal washes of the Depo 15 group up to 6 days postchallenge. In contrast, no viral shedding was observed beyond day 3 postchallenge in the Depo 5 group. Following i.vag. TK(-) immunization, high levels of gamma interferon (IFN-gamma) were detected locally in vaginal washes of the Depo 5 group but not the Depo 15 group. After HSV-2 challenge, an early peak of IFN-gamma in the Depo 5 group coincided with clearance of the virus. In Depo 15 animals IFN-gamma was present throughout the 6 days postinfection. HSV-2-specific T-cell cytokine responses measured in the lymph node cells of Depo 5 TK(-)-immunized mice indicated a significantly higher Th1 response than that of Depo 15 TK(-)-immunized mice. The protection after HSV-2 challenge in the Depo 5 group correlated with increased local HSV-2 glycoprotein B (gB)-specific immunoglobulin G (IgG) and IgA responses seen in the vaginal secretions. The Depo 15 group had poor gB-specific antibody responses in the genital tract after HSV-2 challenge. These results indicate that longer exposure to Depo leads to poor innate and adaptive immune responses to HSV-2 that fail to protect mice from subsequent genital challenges.


Assuntos
Herpesvirus Humano 2/imunologia , Vacinas contra Herpesvirus/imunologia , Progesterona/toxicidade , Vagina/imunologia , Animais , Anticorpos Antivirais/análise , Feminino , Imunidade nas Mucosas , Imunização , Imunoglobulina A/análise , Imunoglobulina G/análise , Interferon gama/análise , Camundongos , Camundongos Endogâmicos C57BL , Infecções Sexualmente Transmissíveis/prevenção & controle , Linfócitos T/imunologia , Vacinas de Produtos Inativados/imunologia , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA