Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Front Immunol ; 12: 753849, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790197

RESUMEN

Background: CD14+ monocytes present antigens to adaptive immune cells via monocytic human leukocyte antigen receptor (mHLA-DR), which is described as an immunological synapse. Reduced levels of mHLA-DR can display an acquired immune defect, which is often found in sepsis and predisposes for secondary infections and fatal outcomes. Monocytic HLA-DR expression is reliably induced by interferon- γ (IFNγ) therapy. Case Report: We report a case of multidrug-resistant superinfected COVID-19 acute respiratory distress syndrome (ARDS) on extracorporeal membrane oxygenation (ECMO) support. The resistance profiles of the detected Klebsiella pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumannii and Citrobacter freundii isolates were equipped with resistance to all four antibiotic classes including carbapenems (4MRGN) and Cefiderocol in the case of K. pneumoniae. A causal therapeutic antibiotic strategy was not available. Therefore, we measured the immune status of the patient aiming to identify a potential acquired immune deficiency. Monocyte HLA-DR expression identified by FACS analysis revealed an expression level of 34% positive monocytes and suggested severe immunosuppression. We indicated IFNγ therapy, which resulted in a rapid increase in mHLA-DR expression (96%), rapid resolution of invasive bloodstream infection, and discharge from the hospital on day 70. Discussion: Superinfection is a dangerous complication of COVID-19 pneumonia, and sepsis-induced immunosuppression is a risk factor for it. Immunosuppression is expressed by a disturbed antigen presentation of monocytes to cells of the adaptive immune system. The case presented here is remarkable as no validated antibiotic regimen existed against the detected bacterial pathogens causing bloodstream infection and severe pneumonia in a patient suffering from COVID-19 ARDS. Possible restoration of the patient's own immunity by IFNγ was a plausible option to boost the patient's immune system, eliminate the identified 4MRGNs, and allow for lung recovery. This led to the conclusion that immune status monitoring is useful in complicated COVID-19-ARDS and that concomitant IFNγ therapy may support antibiotic strategies. Conclusion: After a compromised immune system has been detected by suppressed mHLA-DR levels, the immune system can be safely reactivated by IFNγ.


Asunto(s)
Bacterias/inmunología , COVID-19/inmunología , Resistencia a Múltiples Medicamentos/inmunología , Antígenos HLA/inmunología , Interferón gamma/inmunología , Monocitos/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Adulto , Humanos , Receptores de Interferón/inmunología , Receptor de Interferón gamma
2.
Front Immunol ; 11: 583305, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193396

RESUMEN

Novel therapeutics against the global threat of multidrug-resistant Neisseria gonorrhoeae are urgently needed. Gonococci possess several mechanisms to evade killing by human complement, including binding of factor H (FH), a key inhibitor of the alternative pathway. FH comprises 20 short consensus repeat (SCR) domains organized in a head-to-tail manner as a single chain. N. gonorrhoeae binds two regions in FH; domains 6 and 7 and domains 18 through 20. We designed a novel anti-infective immunotherapeutic molecule that fuses domains 18-20 of FH containing a D-to-G mutation in domain 19 at position 1119 (called FH*) with human IgG1 Fc. FH*/Fc retained binding to gonococci but did not lyse human erythrocytes. Expression of FH*/Fc in tobacco plants was undertaken as an alternative, economical production platform. FH*/Fc was expressed in high yields in tobacco plants (300-600 mg/kg biomass). The activities of plant- and CHO-cell produced FH*/Fc against gonococci were similar in vitro and in the mouse vaginal colonization model of gonorrhea. The addition of flexible linkers [e.g., (GGGGS)2 or (GGGGS)3] between FH* and Fc improved the bactericidal efficacy of FH*/Fc 2.7-fold. The linkers also improved PMN-mediated opsonophagocytosis about 11-fold. FH*/Fc with linker also effectively reduced the duration and burden of colonization of two gonococcal strains tested in mice. FH*/Fc lost efficacy: i) in C6-/- mice (no terminal complement) and ii) when Fc was mutated to abrogate complement activation, suggesting that an intact complement was necessary for FH*/Fc function in vivo. In summary, plant-produced FH*/Fc represent promising prophylactic or adjunctive immunotherapeutics against multidrug-resistant gonococci.


Asunto(s)
Resistencia a Múltiples Medicamentos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Neisseria gonorrhoeae/inmunología , Nicotiana/genética , Plantas Modificadas Genéticamente , Animales , Antibacterianos/farmacología , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Gonorrea , Humanos , Inmunoglobulina G , Inmunoterapia , Ratones , Plantas Modificadas Genéticamente/genética , Proteínas Recombinantes de Fusión/inmunología
3.
Drug Resist Updat ; 53: 100715, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32679188

RESUMEN

It is well established that multifactorial drug resistance hinders successful cancer treatment. Tumor cell interactions with the tumor microenvironment (TME) are crucial in epithelial-mesenchymal transition (EMT) and multidrug resistance (MDR). TME-induced factors secreted by cancer cells and cancer-associated fibroblasts (CAFs) create an inflammatory microenvironment by recruiting immune cells. CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) and inflammatory tumor associated macrophages (TAMs) are main immune cell types which further enhance chronic inflammation. Chronic inflammation nurtures tumor-initiating/cancer stem-like cells (CSCs), induces both EMT and MDR leading to tumor relapses. Pro-thrombotic microenvironment created by inflammatory cytokines and chemokines from TAMs, MDSCs and CAFs is also involved in EMT and MDR. MDSCs are the most common mediators of immunosuppression and are also involved in resistance to targeted therapies, e.g. BRAF inhibitors and oncolytic viruses-based therapies. Expansion of both cancer and stroma cells causes hypoxia by hypoxia-inducible transcription factors (e.g. HIF-1α) resulting in drug resistance. TME factors induce the expression of transcriptional EMT factors, MDR and metabolic adaptation of cancer cells. Promoters of several ATP-binding cassette (ABC) transporter genes contain binding sites for canonical EMT transcription factors, e.g. ZEB, TWIST and SNAIL. Changes in glycolysis, oxidative phosphorylation and autophagy during EMT also promote MDR. Conclusively, EMT signaling simultaneously increases MDR. Owing to the multifactorial nature of MDR, targeting one mechanism seems to be non-sufficient to overcome resistance. Targeting inflammatory processes by immune modulatory compounds such as mTOR inhibitors, demethylating agents, low-dosed histone deacetylase inhibitors may decrease MDR. Targeting EMT and metabolic adaptation by small molecular inhibitors might also reverse MDR. In this review, we summarize evidence for TME components as causative factors of EMT and anticancer drug resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Desmetilación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/inmunología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
4.
Nat Commun ; 11(1): 3418, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647286

RESUMEN

The emergence and spread of antiviral drug-resistant viruses have been a worldwide challenge and a great concern for patient care. We report A4 antibody specifically recognizing and binding to the mutant I223R/H275Y neuraminidase and prove the applicability of A4 antibody for direct detection of antiviral multidrug-resistant viruses in various sensing platforms, including naked-eye detection, surface-enhanced Raman scattering-based immunoassay, and lateral flow system. The development of the A4 antibody enables fast, simple, and reliable point-of-care assays of antiviral multidrug-resistant influenza viruses. In addition to current influenza virus infection testing methods that do not provide information on the antiviral drug-resistance of the virus, diagnostic tests for antiviral multidrug-resistant viruses will improve clinical judgment in the treatment of influenza virus infections, avoid the unnecessary prescription of ineffective drugs, and improve current therapies.


Asunto(s)
Anticuerpos Antivirales/inmunología , Resistencia a Múltiples Medicamentos/inmunología , Farmacorresistencia Viral/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Mutación/genética , Neuraminidasa/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/química , Afinidad de Anticuerpos/inmunología , Antígenos Virales/metabolismo , Líquidos Corporales/virología , Análisis Mutacional de ADN , Perros , Epítopos/química , Epítopos/inmunología , Humanos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/enzimología , Células de Riñón Canino Madin Darby , Simulación del Acoplamiento Molecular , Imagen Óptica , Unión Proteica , Espectrometría Raman
5.
Mil Med ; 185(Suppl 1): 637-643, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32074338

RESUMEN

INTRODUCTION: Infection frequently complicates the treatment of combat-related wounds, impairs healing, and leads to worse outcomes. To better manage wound infections, antimicrobial therapies that are effective against biofilm and designed for direct wound application are needed. The primary objective of this work was to evaluate a chitosan matrix for delivery of two engineered antimicrobial peptides, (ASP)-1 and ASP-2, to treat biofilm-associated bacteria. A secondary objective was to determine whether replacing the levorotatory (L) form amino acids in ASP-2 with dextrorotatory (D) form amino acids would impact peptide activity. MATERIALS AND METHODS: Chitosan gels loaded with antimicrobial peptides were evaluated for peptide release over 7 days and tested for efficacy against biofilms grown both in vitro on polymer mesh and ex vivo on porcine skin. RESULTS: When delivered via chitosan, 70% to 80% of peptides were released over 7 days. Gels eradicated biofilms of gram-positive and gram-negative, drug-resistant bacteria in vitro and ex vivo. Under the conditions tested, no meaningful differences in peptide activity between the L and D forms of ASP-2 were detected. CONCLUSIONS: Chitosan serves as an effective delivery platform for ASP-1 and ASP-2 to treat biofilm-embedded bacteria and warrants further development as a topical treatment.


Asunto(s)
Biopelículas/efectos de los fármacos , Quitosano/farmacocinética , Animales , Vendajes/normas , Vendajes/estadística & datos numéricos , Quitosano/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Múltiples Medicamentos/inmunología , Geles/uso terapéutico , Proteínas Citotóxicas Formadoras de Poros/farmacocinética , Proteínas Citotóxicas Formadoras de Poros/uso terapéutico , Porcinos , Cicatrización de Heridas/efectos de los fármacos
6.
Semin Liver Dis ; 40(1): 11-19, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31370067

RESUMEN

Severe alcoholic hepatitis (SAH) is a condition characterized by jaundice and liver failure that develops after heavy and prolonged alcohol consumption. Infection frequently complicates the natural history of the disease and is independently associated with mortality. Objective recognition and recording of infection are therefore essential in the evaluation of therapeutic interventions and for antibiotic stewardship. This review will evaluate infections that complicate SAH at admission and beyond. Factors that associate with the development of infection will be identified and clinical and laboratory techniques available to identify infection will be discussed. Common pathogens and frequently used antibiotics will be reviewed and recommendations will be made for the management of infection for SAH patients. New techniques to assess infection earlier and more precisely may improve diagnosis and treatment of this important driver of mortality in SAH.


Asunto(s)
Hepatitis Alcohólica/complicaciones , Antiinfecciosos/uso terapéutico , Programas de Optimización del Uso de los Antimicrobianos , Resistencia a Múltiples Medicamentos/inmunología , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/etiología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/etiología , Infecciones por Bacterias Grampositivas/microbiología , Hepatitis Alcohólica/microbiología , Humanos , Micosis/tratamiento farmacológico , Micosis/etiología , Micosis/microbiología
7.
Crit Rev Ther Drug Carrier Syst ; 37(5): 473-509, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33389848

RESUMEN

Multidrug resistance (MDR) remains a major obstacle to ensure effective chemotherapy in cancer patients. Several factors could be associated with cancer cells' drug resistance such as overexpression of P-glycoprotein (P-gp), cancer stem cells (CSCs), defect in apoptosis, mutation and alteration in DNA repair pathways, angiogenesis, autophagy, and modulation in metabolic enzymes. Until now, drug efflux by ABC transporters has been a univocal and well-established mechanism of chemotherapeutic associated drug resistance. To explore the mechanics involved in ABC transporter associated drug resistance, many crucial studies have been conducted from identification of drug binding sites to elucidation of their structure. Due to our continuous battle with drug resistance, several strategies have been employed to combat MDR, including P-gp modulators, siRNAs, antibodies, as well as peptides. Furthermore, various nanoparticle and different effective combination nanomedicine strategies also suggest some exciting results. Thus, to improve nanomedicine approaches to overcome MDR, in this evolutionary review, we have focused on fundamentals of possible strategies as well as the latest accomplishments to reverse MDR.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Nanomedicina Teranóstica/métodos , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Resistencia a Múltiples Medicamentos/genética , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Humanos , Ratones , Nanopartículas/química , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/mortalidad , ARN Interferente Pequeño/administración & dosificación , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
8.
ACS Appl Mater Interfaces ; 11(46): 42904-42916, 2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31657540

RESUMEN

Potentiating systemic immunity against breast cancer is in the most urgent demand as breast cancer is less sensitive to immune checkpoint blockade. Although phototherapy and some chemotherapy can trigger immunogenic cell death (ICD) for T cell-mediated antitumor immune response, their immunotherapy efficacy is severely restricted by insufficient phototherapeutic capability and severe multidrug resistance (MDR). Inspired by both the hypersensitivity to phototherapy and the key role of MDR for mitochondria, a rationally engineered immunity amplifier via mitochondria-targeted photochemotherapeutic nanoparticles was, for the first time, achieved to fight against low-immunogenic breast cancer without additional immune agents. The newly synthesized task-specific mitochondria-targeted IR780 derivative (T780) was integrated with chemotherapeutic doxorubicin (DOX) to form multifunctional nanoparticles via an assembling strategy along with bovine serum albumin (BSA) as a biomimetic corona (BSA@T780/DOX NPs). The in situ enhancement in both phototherapy and MDR reversal by targeting mitochondria with BSA@T780/DOX NPs boosted highly efficient ICD toward excellent antitumor immune response. The newly developed strategy not only eradicated the primary tumor but also eliminated the bilateral tumors efficiently, as well as preventing metastasis and postsurgical recurrence, demonstrating great interest for fighting against low-immunogenic breast cancer.


Asunto(s)
Materiales Biomiméticos , Doxorrubicina , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Inmunoterapia , Indoles , Neoplasias Mamarias Experimentales , Nanopartículas , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Bovinos , Línea Celular , Doxorrubicina/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Femenino , Indoles/química , Indoles/farmacología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Corona de Proteínas/química , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacología
9.
PLoS Pathog ; 15(8): e1007460, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31381597

RESUMEN

Candida auris is an emerging, multi-drug resistant, health care-associated fungal pathogen. Its predominant prevalence in hospitals and nursing homes indicates its ability to adhere to and colonize the skin, or persist in an environment outside the host-a trait unique from other Candida species. Besides being associated globally with life-threatening disseminated infections, C. auris also poses significant clinical challenges due to its ability to adhere to polymeric surfaces and form highly drug-resistant biofilms. Here, we performed bioinformatic studies to identify the presence of adhesin proteins in C. auris, with sequence as well as 3-D structural homologies to the major adhesin/invasin of C. albicans, Als3. Anti-Als3p antibodies generated by vaccinating mice with NDV-3A (a vaccine based on the N-terminus of Als3 protein formulated with alum) recognized C. auris in vitro, blocked its ability to form biofilms and enhanced macrophage-mediated killing of the fungus. Furthermore, NDV-3A vaccination induced significant levels of C. auris cross-reactive humoral and cellular immune responses, and protected immunosuppressed mice from lethal C. auris disseminated infection, compared to the control alum-vaccinated mice. The mechanism of protection is attributed to anti-Als3p antibodies and CD4+ T helper cells activating tissue macrophages. Finally, NDV-3A potentiated the protective efficacy of the antifungal drug micafungin, against C. auris candidemia. Identification of Als3-like adhesins in C. auris makes it a target for immunotherapeutic strategies using NDV-3A, a vaccine with known efficacy against other Candida species and safety as well as efficacy in clinical trials. Considering that C. auris can be resistant to almost all classes of antifungal drugs, such an approach has profound clinical relevance.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Linfocitos T CD4-Positivos/inmunología , Candida/inmunología , Candidiasis/prevención & control , Resistencia a Múltiples Medicamentos/inmunología , Proteínas Fúngicas/inmunología , Vacunas Fúngicas/administración & dosificación , Compuestos de Alumbre/química , Animales , Candidiasis/inmunología , Candidiasis/microbiología , Ratones , Ratones Endogámicos ICR , Vacunación
10.
Mem. Inst. Oswaldo Cruz ; 114: e190054, 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1012675

RESUMEN

BACKGROUND The central repetitive region (CRR) of the Plasmodium vivax circumsporozoite surface protein (CSP) is composed of a repetitive sequence that is characterised by three variants: VK210, VK247 and P. vivax-like. The most important challenge in the treatment of P. vivax infection is the possibility of differential response based on the parasite genotype. OBJECTIVES To characterise the CSP variants in P. vivax isolates from individuals residing in a malaria-endemic region in Brazil and to profile these variants based on sensitivity to chloroquine and mefloquine. METHODS The CSP variants were determined by sequencing and the sensitivity of the P. vivax isolates to chloroquine and mefloquine was determined by Deli-test. FINDINGS Although five different allele sizes were amplified, the sequencing results showed that all of the isolates belonged to the VK210 variant. However, we observed substantial genetic diversity in the CRR, resulting in the identification of 10 different VK210 subtypes. The frequency of isolates that were resistant to chloroquine and mefloquine was 11.8 and 23.8%, respectively. However, we did not observe any difference in the frequency of the resistant isolates belonging to the VK210 subtypes. MAIN CONCLUSION The VK210 variant is the most frequently observed in the studied region and there is significant genetic variability in the CRR of the P. vivax CSP. Moreover, the antimalarial drug sensitivity profiles of the isolates does not seem to be related to the VK210 subtypes.


Asunto(s)
Plasmodium vivax/efectos de los fármacos , Mefloquina/uso terapéutico , Cloroquina/uso terapéutico , Resistencia a Múltiples Medicamentos/inmunología , Brasil
11.
Biochem Biophys Res Commun ; 503(3): 1812-1818, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30060952

RESUMEN

Cancer immunoediting enriches NANOG expression in tumor cells, resulting in multi-drug resistance and stem-like phenotypes. We previously demonstrated that these NANOG-associated phenotypes are promoted through HDAC1 transcriptional upregulation. In this study, we identified that NANOG also contributes to the stabilization of HDAC1 protein through the AKT signaling pathway. NANOG-AKT axis leads to phosphor-dependent inactivation of CHFR, an E3 ligase for HDAC1 protein, and thereby inhibiting the ubiquitin-mediated degradation of HDAC1. Furthermore, AKT inhibition disrupts HDAC1 WT-mediated phenotypes but had no effect on the phenotypes mediated by HDAC1 FM, a mutant that is unable to interact with CHFR. Critically, we applied a catalytic dead mutant, HDAC1-H141A, to uncover that HDAC1 confers immune-resistance, drug-resistance and stem-like phenotype in tumor cells through its catalytic activity. Collectively, our results establish a firm molecular link in immune-edited tumor cells among NANOG, AKT, CHFR, and HDAC1, identifying HDAC1 as a molecular target in controlling NANOGHIGH immune-refractory cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Histona Desacetilasa 1/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias del Cuello Uterino/inmunología , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/inmunología , Femenino , Células HEK293 , Células HeLa , Histona Desacetilasa 1/genética , Humanos , Mutagénesis Sitio-Dirigida , Fenotipo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
12.
Clin Pharmacol Ther ; 103(1): 102-111, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29071710

RESUMEN

There has been an increasing interest in the association between human disease and altered gut microbiota, and therapeutics to modulate microbiota to treat disease. Healthy human gastrointestinal microbiota is highly diverse and rich, and harbors between 500 and 2,000 species. Diseases associated with dysbiotic microbiota include antibiotic-associated diarrhea, Clostridium difficile infection, multidrug-resistant organisms, inflammatory bowel disease, obesity, metabolic syndrome, diabetes mellitus, neuropsychiatric diseases, and systemic autoimmune diseases. Microbiota replacement therapies have shown immense promise in treatment of recurrent C. difficile infection and are being studied for other indications. Microbiota replacement therapies for indications other than C. difficile infection should be performed only in research settings. There is an immense need for standardized microbiota replacement therapies for C. difficile infection. Studies are needed to elucidate long-term safety and adverse events from these therapies.


Asunto(s)
Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/fisiología , Enfermedades del Sistema Inmune/terapia , Inflamación/terapia , Terapia Biológica/métodos , Infecciones por Clostridium/fisiopatología , Resistencia a Múltiples Medicamentos/inmunología , Disbiosis/terapia , Humanos , Enfermedades del Sistema Inmune/fisiopatología , Inflamación/inmunología , Resultado del Tratamiento
13.
Drug Resist Updat ; 32: 1-15, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-29145974

RESUMEN

Patients with breast tumors that do not express the estrogen receptor, the progesterone receptor, nor Her-2/neu are hence termed "triple negatives", and generally have a poor prognosis, with high rates of systemic recurrence and refractoriness to conventional therapy regardless of the choice of adjuvant treatment. Thus, more effective therapeutic options are sorely needed for triple-negative breast cancer (TNBC), which occurs in approximately 20% of diagnosed breast cancers. In recent years, exploiting intrinsic mechanisms of the host immune system to eradicate cancer cells has achieved impressive success, and the advances in immunotherapy have yielded potential new therapeutic strategies for the treatment of this devastating subtype of breast cancer. It is anticipated that the responses initiated by immunotherapeutic interventions will explicitly target and annihilate tumor cells, while at the same time spare normal cells. Various immunotherapeutic approaches have been already developed and tested, which include the blockade of immune checkpoints using neutralizing or blocking antibodies, induction of cytotoxic T lymphocytes (CTLs), adoptive cell transfer-based therapy, and modulation of the tumor microenvironment to enhance the activity of CTLs. One of the most important areas of breast cancer research today is understanding the immune features and profiles of TNBC and devising novel immune-modulatory strategies to tackling TNBC, a subtype of breast cancer notorious for its poor prognosis and its imperviousness to conventional treatments. On the optimal side, one can anticipate that novel, effective, and personalized immunotherapy for TNBC will soon achieve more success and impact clinical treatment of this disease which afflicts approximately 20% of patients with breast cancer. In the present review, we highlight the current progress and encouraging developments in cancer immunotherapy, with a goal to discuss the challenges and to provide future perspectives on how to exploit a variety of new immunotherapeutic approaches including checkpoint inhibitors and neoadjuvant immunotherapy for the treatment of patients with TNBC.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Recurrencia Local de Neoplasia/terapia , Neoplasias de la Mama Triple Negativas/terapia , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/inmunología , Ensayos Clínicos como Asunto , Terapia Combinada/métodos , Terapia Combinada/tendencias , Resistencia a Múltiples Medicamentos/genética , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Femenino , Inestabilidad Genómica , Humanos , Inmunoterapia/tendencias , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Terapia Neoadyuvante/métodos , Terapia Neoadyuvante/tendencias , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/inmunología
14.
PLoS Pathog ; 13(11): e1006696, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29112952

RESUMEN

Klebsiella pneumoniae is a significant cause of nosocomial pneumonia and an alarming pathogen owing to the recent isolation of multidrug resistant strains. Understanding of immune responses orchestrating K. pneumoniae clearance by the host is of utmost importance. Here we show that type I interferon (IFN) signaling protects against lung infection with K. pneumoniae by launching bacterial growth-controlling interactions between alveolar macrophages and natural killer (NK) cells. Type I IFNs are important but disparate and incompletely understood regulators of defense against bacterial infections. Type I IFN receptor 1 (Ifnar1)-deficient mice infected with K. pneumoniae failed to activate NK cell-derived IFN-γ production. IFN-γ was required for bactericidal action and the production of the NK cell response-amplifying IL-12 and CXCL10 by alveolar macrophages. Bacterial clearance and NK cell IFN-γ were rescued in Ifnar1-deficient hosts by Ifnar1-proficient NK cells. Consistently, type I IFN signaling in myeloid cells including alveolar macrophages, monocytes and neutrophils was dispensable for host defense and IFN-γ activation. The failure of Ifnar1-deficient hosts to initiate a defense-promoting crosstalk between alveolar macrophages and NK cell was circumvented by administration of exogenous IFN-γ which restored endogenous IFN-γ production and restricted bacterial growth. These data identify NK cell-intrinsic type I IFN signaling as essential driver of K. pneumoniae clearance, and reveal specific targets for future therapeutic exploitations.


Asunto(s)
Interferón Tipo I/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Klebsiella/inmunología , Macrófagos Alveolares/inmunología , Transducción de Señal/inmunología , Animales , Resistencia a Múltiples Medicamentos/inmunología , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cross-Talk/inmunología , Infecciones del Sistema Respiratorio/inmunología
15.
Cancer Res ; 77(18): 5039-5053, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28716899

RESUMEN

Cancer immunoediting drives the adaptation of tumor cells to host immune surveillance. Immunoediting driven by antigen (Ag)-specific T cells enriches NANOG expression in tumor cells, resulting in a stem-like phenotype and immune resistance. Here, we identify HDAC1 as a key mediator of the NANOG-associated phenotype. NANOG upregulated HDAC1 through promoter occupancy, thereby decreasing histone H3 acetylation on K14 and K27. NANOG-dependent, HDAC1-driven epigenetic silencing of cell-cycle inhibitors CDKN2D and CDKN1B induced stem-like features. Silencing of TRIM17 and NOXA induced immune and drug resistance in tumor cells by increasing antiapoptotic MCL1. Importantly, HDAC inhibition synergized with Ag-specific adoptive T-cell therapy to control immune refractory cancers. Our results reveal that NANOG influences the epigenetic state of tumor cells via HDAC1, and they encourage a rational application of epigenetic modulators and immunotherapy in treatment of NANOG+ refractory cancer types. Cancer Res; 77(18); 5039-53. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Resistencia a Múltiples Medicamentos/inmunología , Epigénesis Genética , Histona Desacetilasa 1/metabolismo , Proteína Homeótica Nanog/metabolismo , Células Madre Neoplásicas/patología , Neoplasias del Cuello Uterino/patología , Acetilación , Animales , Apoptosis , Biomarcadores de Tumor , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Proliferación Celular , Terapia Combinada , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/inmunología , Femenino , Histona Desacetilasa 1/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína Homeótica Nanog/genética , Estadificación de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Pronóstico , Activación Transcripcional , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Microb Drug Resist ; 23(2): 206-214, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27257915

RESUMEN

Since the implementation of routine PCV13 immunization in Japan, nonvaccine serotypes (NVTs) have been increasing among clinical isolates of Streptococcus pneumoniae. In this study, susceptibility to 18 antibiotics was tested for all the 231 isolates with NVTs, which were collected from children <16 years of age in northern Japan in 2014 (July-November). High resistance rates were observed for macrolides (>90.9%), tetracycline (91.3%), and clindamycin (75.3%), while penicillin (PEN) nonsusceptibility (PNSP; MIC ≥0.12 µg/ml) was detected in 42.9% of the pneumococci [39.4%; PEN-intermediate S. pneumoniae (PISP), 3.5%; PEN-resistant S. pneumoniae (PRSP)]. All serotype 15A isolates were PRSP (MIC, ≥2 µg/ml) or PISP, and PNSP was prevalent in also serotypes 23A (96.9%), 6C (41%), and 35B (33.3%). Overall, 42.0% of the isolates showed multidrug resistance (MDR). Sequence types (STs) determined for 20 PNSP isolates with NVTs were ST63 (15A), STs 242 or 5832 (6C), STs 338 or 5242 (23A), and ST558 (35B). All the PNSP isolates possessed tet(M), and erm(B) or mefA(A/E), and 70% of them were gPRSP having three altered genes pbp1a, pbp2x, and pbp2b. Among alterations in transpeptidase-coding region of penicillin-binding proteins (PBPs), two substitutions of T371S in the STMK motif and TSQF574-577NTGY in PBP1a were common to all PRSP isolates. The present study showed the spread of PNSP in NVTs 15A, 23A, 6C, and 35B, and the emergence of the MDR international clone Sweden15A-ST63 in northern Japan.


Asunto(s)
Resistencia a Múltiples Medicamentos/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/inmunología , Adolescente , Secuencia de Aminoácidos , Antibacterianos/inmunología , Humanos , Japón , Pruebas de Sensibilidad Microbiana/métodos , Proteínas de Unión a las Penicilinas/inmunología , Penicilinas/inmunología , Peptidil Transferasas/inmunología , Infecciones Neumocócicas/inmunología , Prevalencia , Alineación de Secuencia , Serogrupo , Serotipificación/métodos
17.
PLoS Pathog ; 11(12): e1005290, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26630657

RESUMEN

Neisseria gonorrhoeae deploys a novel immune evasion strategy wherein the lacto-N-neotetraose (LNnT) structure of lipooligosaccharide (LOS) is capped by the bacterial sialyltransferase, using host cytidine-5'-monophosphate (CMP)-activated forms of the nine-carbon nonulosonate (NulO) sugar N-acetyl-neuraminic acid (Neu5Ac), a sialic acid (Sia) abundant in humans. This allows evasion of complement-mediated killing by recruiting factor H (FH), an inhibitor of the alternative complement pathway, and by limiting classical pathway activation ("serum-resistance"). We utilized CMP salts of six additional natural or synthetic NulOs, Neu5Gc, Neu5Gc8Me, Neu5Ac9Ac, Neu5Ac9Az, legionaminic acid (Leg5Ac7Ac) and pseudaminic acid (Pse5Ac7Ac), to define structural requirements of Sia-mediated serum-resistance. While all NulOs except Pse5Ac7Ac were incorporated into the LNnT-LOS, only Neu5Gc incorporation yielded high-level serum-resistance and FH binding that was comparable to Neu5Ac, whereas Neu5Ac9Az and Leg5Ac7Ac incorporation left bacteria fully serum-sensitive and did not enhance FH binding. Neu5Ac9Ac and Neu5Gc8Me rendered bacteria resistant only to low serum concentrations. While serum-resistance mediated by Neu5Ac was associated with classical pathway inhibition (decreased IgG binding and C4 deposition), Leg5Ac7Ac and Neu5Ac9Az incorporation did not inhibit the classical pathway. Remarkably, CMP-Neu5Ac9Az and CMP-Leg5Ac7Ac each prevented serum-resistance despite a 100-fold molar excess of CMP-Neu5Ac in growth media. The concomitant presence of Leg5Ac7Ac and Neu5Ac on LOS resulted in uninhibited classical pathway activation. Surprisingly, despite near-maximal FH binding in this instance, the alternative pathway was not regulated and factor Bb remained associated with bacteria. Intravaginal administration of CMP-Leg5Ac7Ac to BALB/c mice infected with gonorrhea (including a multidrug-resistant isolate) reduced clearance times and infection burden. Bacteria recovered from CMP-Leg5Ac7Ac-treated mice were sensitive to human complement ex vivo, simulating in vitro findings. These data reveal critical roles for the Sia exocyclic side-chain in gonococcal serum-resistance. Such CMP-NulO analogs may provide a novel therapeutic strategy against the global threat of multidrug-resistant gonorrhea.


Asunto(s)
Citidina Monofosfato/análogos & derivados , Farmacorresistencia Microbiana/inmunología , Resistencia a Múltiples Medicamentos/inmunología , Gonorrea/inmunología , Ácidos Siálicos/farmacología , Animales , Western Blotting , Proteínas del Sistema Complemento/inmunología , Citidina Monofosfato/farmacología , Ácido N-Acetilneuramínico Citidina Monofosfato/análogos & derivados , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Gonorrea/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/metabolismo
18.
Scand J Gastroenterol ; 50(8): 1025-31, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25862144

RESUMEN

OBJECTIVE: Not all patients with autoimmune hepatitis (AIH) respond to standard medical therapy with corticosteroids and azathioprine. Such patients may develop end-stage liver disease with poor prognosis unless transplantation is considered. Alternatively, the introduction of new therapeutic strategies could potentially ameliorate deterioration of liver function. Patients in our tertiary center were selected for everolimus therapy when exhibiting nonresponse or intolerance to combinations of the standard and empirical drugs in use (e.g., mycophenolate mofetil, calcineurin inhibitors [CNIs]). We here report the efficacy of everolimus treatment of patients with AIH. MATERIALS AND METHODS: Seven patients (six female, mean age 47 years, range 22-62 years) in whom disease control could not be achieved with standard therapy or the alternative drugs in use were included. RESULTS: Treatment with everolimus induced a clear reduction of transaminases within 2 weeks. After 3-5 months three patients had normal alanine aminotransferase (ALT) levels (10-45 IU) and four patients had ALT levels below 55 IU compared to a three- to fivefold elevated level prior to everolimus treatment. Sustained remission after 1 year of treatment was observed in three patients; in another two patients ALT was 45-68 U/L. Four patients in remission after 3 years were rebiopsied. Two showed no histological progression, and in two the fibrosis had decreased. Side effects noted were myalgias and minor bacterial infections not leading to discontinuation of the drug. CONCLUSION: Our experience indicates that everolimus may be of value in selected patients with therapy-resistant AIH and comorbidity/side effects that excludes the use of CNIs.


Asunto(s)
Alanina Transaminasa/sangre , Resistencia a Múltiples Medicamentos/inmunología , Everolimus/uso terapéutico , Hepatitis Autoinmune/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Corticoesteroides/uso terapéutico , Adulto , Azatioprina/uso terapéutico , Everolimus/efectos adversos , Femenino , Humanos , Inmunosupresores/efectos adversos , Pruebas de Función Hepática , Masculino , Persona de Mediana Edad , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Inducción de Remisión , Centros de Atención Terciaria , Resultado del Tratamiento , Adulto Joven
19.
Curr Opin Pediatr ; 27(1): 124-31, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25565574

RESUMEN

PURPOSE OF REVIEW: The prevalence of multidrug-resistant (MDR) infections caused by Gram-negative rods (GNRs) such as Escherichia coli and Klebsiella pneumoniae is increasing in large part because of the production of extended-spectrum ß-lactamases (ESBLs) that confer resistance to third-generation cephalosporins such as ceftriaxone. This review examines the clinical implication of changes in the epidemiology, diagnostic tools, and reporting criteria for ESBL-GNRs. RECENT FINDINGS: Pediatric ESBL-GNR infections, once associated only with hospitals, are increasing in the community. The most common risk factor for ESBL-GNR infection is previous antibiotic use. Evidence also exists for prolonged fecal carriage after discharge from neonatal ICUs, leading to community transmission with close contact of known carriers. Controversial changes in the laboratory criteria for GNR cephalosporin resistance have also contributed to the increased numbers of reported MDR-GNR infections. Newer diagnostic tools will enable more rapid identification of MDR-GNR infections, leading to earlier implementation of appropriate therapy. SUMMARY: ESBL-GNR infections are no longer limited to ICUs and are now being identified in children presenting from the community. Appropriate antibiotic prescribing practices are critical to limit the spread of ESBL-GNRs, and pediatricians must prepare to identify and treat these challenging infections.


Asunto(s)
Infecciones Comunitarias Adquiridas/microbiología , Resistencia a Múltiples Medicamentos/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Klebsiella/microbiología , beta-Lactamasas/metabolismo , Antibacterianos , Niño , Preescolar , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Infecciones Comunitarias Adquiridas/epidemiología , Infección Hospitalaria , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/inmunología , Infecciones por Bacterias Gramnegativas/epidemiología , Humanos , Lactante , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/patogenicidad , Pruebas de Sensibilidad Microbiana , Prevalencia , Resistencia betalactámica/inmunología
20.
Cancer Lett ; 347(2): 159-66, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24657660

RESUMEN

Multi-drug resistance (MDR) has become the largest obstacle to the success of cancer chemotherapies. The mechanisms of MDR and the approaches to test MDR have been discovered, yet not fully understood. This review covers the in vivo and in vitro approaches for the detection of MDR in the laboratory and the mechanisms of MDR in cancers. This study also envisages the future developments toward the clinical and therapeutic applications of MDR in cancer treatment. Future therapeutics for cancer treatment will likely combine the existing therapies with drugs originated from MDR mechanisms such as anti-cancer stem cell drugs, anti-miRNA drugs or anti-epigenetic drugs. The challenges for the clinical detection of MDR will be to find new biomarkers and to determine new evaluation systems before the drug resistance emerges.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Autofagia , Resistencia a Múltiples Medicamentos/genética , Resistencia a Múltiples Medicamentos/inmunología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Epigénesis Genética , Humanos , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA