Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Intensive Care Med ; : 8850666241245703, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38613376

RESUMEN

Aconite poisoning refers to toxicity resulting from plants belonging to the Aconitum genus, which comprises over 350 different species of perennial flowering plants that grow in temperate mountainous areas of the northern hemisphere (North America, Europe, Asia). These plants contain a group of toxins known as aconite alkaloids, which encompass numerous closely related toxic compounds. Conventional teaching from toxicology textbooks has broadly classified these alkaloids based on their mechanism of action, often simplifying them as substances that prevent sodium channel inactivation. However, this is an oversimplified and sometimes inaccurate description, as some aconite alkaloids can act as sodium channel blockers. Aconite alkaloids have a long history of use as poisonous substances and have been historically employed for hunting, assassinations, traditional medicine, and self-inflicted harm. Toxicity can occur due to the consumption of traditional medicines derived from aconitum plants or the ingestion of aconite plants and their derivatives. The clinical manifestations of aconite poisoning may encompass gastrointestinal symptoms, sensory alterations, seizures, and life-threatening dysrhythmias that may not respond to standard treatments. Treatment is primarily supportive however evaluation and management of these patients should be personalized and carried out in collaboration with a toxicologist.

2.
Am J Emerg Med ; 77: 158-163, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38150986

RESUMEN

PURPOSE: The preferred vasopressor in post-cardiac arrest shock has not been established with robust clinical outcomes data. Our goal was to perform a systematic review and meta-analysis comparing rates of in-hospital mortality, refractory shock, and hemodynamic parameters in post-cardiac arrest patients who received either norepinephrine or epinephrine as primary vasopressor support. METHODS: We conducted a search of PubMed, Cochrane Library, and CINAHL from 2000 to 2022. Included studies were prospective, retrospective, or published abstracts comparing norepinephrine and epinephrine in adults with post-cardiac arrest shock or with cardiogenic shock and extractable post-cardiac arrest data. The primary outcome of interest was in-hospital mortality. Other outcomes included incidence of arrhythmias or refractory shock. RESULTS: The database search returned 2646 studies. Two studies involving 853 participants were included in the systematic review. The proposed meta-analysis was deferred due to low yield. Crude incidence of in-hospital mortality was numerically higher in the epinephrine group compared with norepinephrine in both studies, but only statistically significant in one. Risk of bias was moderate to severe for in-hospital mortality. Additional outcomes were reported differently between studies, minimizing direct comparison. CONCLUSION: The vasopressor with the best mortality and hemodynamic outcomes in post-cardiac arrest shock remains unclear. Randomized studies are crucial to remedy this.


Asunto(s)
Paro Cardíaco , Choque , Adulto , Humanos , Norepinefrina/uso terapéutico , Choque Cardiogénico/etiología , Estudios Prospectivos , Estudios Retrospectivos , Epinefrina/uso terapéutico , Vasoconstrictores/uso terapéutico , Paro Cardíaco/tratamiento farmacológico , Paro Cardíaco/complicaciones , Choque/tratamiento farmacológico , Choque/complicaciones , Hemodinámica
3.
Cancer Sci ; 114(12): 4499-4510, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37776054

RESUMEN

The Accum™ technology was initially designed to enhance the bioaccumulation of a given molecule in target cells. It does so by triggering endosomal membrane damages allowing endocytosed products to enter the cytosol, escaping the harsh environmental cues of the endosomal lumen. In an attempt to minimize manufacturing hurdles associated with Accum™ conjugation, we tested whether free Accum™ admixed with antigens could lead to outcomes similar to those obtained with conjugated products. Surprisingly, unconjugated Accum™ was found to promote cell death in vitro, an observation further confirmed on various murine tumor cell lines (EL4, CT-26, B16, and 4 T1). At the molecular level, unconjugated Accum™ triggers the production of reactive oxygen species and elicits immunogenic cell death while retaining its innate ability to cause endosomal damages. When administered as a monotherapy to animals with pre-established EL4 T-cell lymphoma, Accum™ controlled tumor growth in a dose-dependent manner, and its therapeutic effect relies on CD4 and CD8 T cells. Although unconjugated Accum™ synergizes with various immune checkpoint inhibitors (anti-CTLA4, anti-PD-1, or anti-CD47) at controlling tumor growth, its therapeutic potency could not be further enhanced when combined with all three tested immune checkpoint inhibitors at once due to its dependency on a specific dosing regimen. In sum, we report in this study an unprecedented new function for unconjugated Accum™ as a novel anticancer molecule. These results could pave the path for a new line of investigation aimed at exploring the pro-killing properties of additional Accum™ variants as a mean to develop second-generation anticancer therapeutics.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Linfoma de Células T , Animales , Ratones , Linfocitos T CD8-positivos , Línea Celular Tumoral
4.
Front Immunol ; 14: 1099459, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969187

RESUMEN

Introduction: Adipocytes in the tumour microenvironment are highly dynamic cells that have an established role in tumour progression, but their impact on anti-cancer therapy resistance is becoming increasingly difficult to overlook. Methods: We investigated the role of adipose tissue and adipocytes in response to oncolytic virus (OV) therapy in adipose-rich tumours such as breast and ovarian neoplasms. Results: We show that secreted products in adipocyte-conditioned medium significantly impairs productive virus infection and OV-driven cell death. This effect was not due to the direct neutralization of virions or inhibition of OV entry into host cells. Instead, further investigation of adipocyte secreted factors demonstrated that adipocyte-mediated OV resistance is primarily a lipid-driven phenomenon. When lipid moieties are depleted from the adipocyte-conditioned medium, cancer cells are re-sensitized to OV-mediated destruction. We further demonstrated that blocking fatty acid uptake by cancer cells, in a combinatorial strategy with virotherapy, has clinical translational potential to overcome adipocyte-mediated OV resistance. Discussion: Our findings indicate that while adipocyte secreted factors can impede OV infection, the impairment of OV treatment efficacy can be overcome by modulating lipid flux in the tumour milieu.


Asunto(s)
Viroterapia Oncolítica , Virus Oncolíticos , Neoplasias Ováricas , Humanos , Femenino , Microambiente Tumoral , Medios de Cultivo Condicionados , Virus Oncolíticos/fisiología , Neoplasias Ováricas/terapia , Lípidos
5.
Front Immunol ; 14: 1098344, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36860852

RESUMEN

Introduction: Triple negative breast cancer (TNBC) is the most aggressive and hard-to-treat subtype of breast cancer, affecting 10-20% of all women diagnosed with breast cancer. Surgery, chemotherapy and hormone/Her2 targeted therapies are the cornerstones of treatment for breast cancer, but women with TNBC do not benefit from these treatments. Although the prognosis is dismal, immunotherapies hold significant promise in TNBC, even in wide spread disease because TNBC is infiltrated with more immune cells. This preclinical study is proposing to optimize an oncolytic virus-infected cell vaccine (ICV) based on a prime-boost vaccination strategy to address this unmet clinical need. Methods: We used various classes of immunomodulators to improve the immunogenicity of whole tumor cells in the prime vaccine, followed by their infection with oncolytic Vesicular Stomatitis Virus (VSVd51) to deliver the boost vaccine. For in vivo studies, we compared the efficacy of a homologous prime-boost vaccination regimen to a heterologous strategy by treating 4T1 tumor bearing BALB/c mice and further by conducting re-challenge studies to evaluate immune memory responses in surviving mice. Due to the aggressive nature of 4T1 tumor spread (akin to stage IV TNBC in human patients), we also compared early surgical resection of primary tumors versus later surgical resection combined with vaccination. Results: In vitro results demonstrated that immunogenic cell death (ICD) markers and pro-inflammatory cytokines were released at the highest levels following treatment of mouse 4T1 TNBC cells with oxaliplatin chemotherapy and influenza vaccine. These ICD inducers also contributed towards higher dendritic cell recruitment and activation. With the top ICD inducers at hand, we observed that treatment of TNBC-bearing mice with the influenza virus-modified prime vaccine followed by VSVd51 infected boost vaccine resulted in the best survival. Furthermore, higher frequencies of both effector and central memory T cells along with a complete absence of recurrent tumors were observed in re-challenged mice. Importantly, early surgical resection combined with prime-boost vaccination led to improved overall survival in mice. Conclusion: Taken together, this novel cancer vaccination strategy following early surgical resection could be a promising therapeutic avenue for TNBC patients.


Asunto(s)
Vacunas contra la Influenza , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Animales , Ratones , Neoplasias de la Mama Triple Negativas/terapia , Recurrencia Local de Neoplasia , Vacunación , Oncogenes , Inmunoterapia
6.
Cell Death Discov ; 9(1): 45, 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36746928

RESUMEN

Gemcitabine is a first-line treatment agent for pancreatic ductal adenocarcinoma (PDAC). Contributing to its cytotoxicity, this chemotherapeutic agent is primarily a DNA replication inhibitor that also induces DNA damage. However, its therapeutic effects are limited owing to chemoresistance. Evidence in the literature points to a role for autophagy in restricting the efficacy of gemcitabine. Autophagy is a catabolic process in which intracellular components are delivered to degradative organelles lysosomes. Interfering with this process sensitizes PDAC cells to gemcitabine. It is consequently inferred that autophagy and lysosomal function need to be tightly regulated to maintain homeostasis and provide resistance to environmental stress, such as those imposed by chemotherapeutic drugs. However, the mechanism(s) through which gemcitabine promotes autophagy remains elusive, and the impact of gemcitabine on lysosomal function remains largely unexplored. Therefore, we applied complementary approaches to define the mechanisms triggered by gemcitabine that support autophagy and lysosome function. We found that gemcitabine elicited ERK-dependent autophagy in PDAC cells, but did not stimulate ERK activity or autophagy in non-tumoral human pancreatic epithelial cells. Gemcitabine also promoted transcription factor EB (TFEB)-dependent lysosomal function in PDAC cells. Indeed, treating PDAC cells with gemcitabine caused expansion of the lysosomal network, as revealed by Lysosome associated membrane protein-1 (LAMP1) and LysoTracker staining. More specific approaches have shown that gemcitabine promotes the activity of cathepsin B (CTSB), a cysteine protease playing an active role in lysosomal degradation. We showed that lysosomal function induced by gemcitabine depends on TFEB, the master regulator of autophagy and lysosomal biogenesis. Interfering with TFEB function considerably limited the clonogenic growth of PDAC cells and hindered the capacity of TFEB-depleted PDAC cells to develop orthotopic tumors.

7.
Mol Ther Oncolytics ; 24: 507-521, 2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35229029

RESUMEN

A significant proportion of non-muscle invasive bladder cancer cases will progress to muscle invasive disease. Transurethral resection followed by Bacillus Calmette Guerin immunotherapy can reduce this risk, while cystectomy prior to muscle invasion provides the best option for survival. Currently, there are no effective treatments for Bacillus Calmette Guerin refractory disease. A novel oncolytic vesicular stomatitis virus containing the human GM-CSF transgene (VSVd51-hGM-CSF) was rescued and tested as a potential bladder-sparing therapy for aggressive bladder cancer. The existing variant expressing mouse GM-CSF was also used. Measurement of gene expression and protein level alterations of canonical immunogenic cell death associated events on mouse and human bladder cancer cell lines and spheroids showed enhanced release of danger signals and immunogenic factors following infection with VSVd51-m/hGM-CSF. Intravesical instillation of VSVd51-mGM-CSF into MB49 bladder cancer bearing C57Bl/6 mice demonstrated enhanced activation of peripheral and bladder infiltrating effector immune cells, along with improved survival and reduced tumor volume. Importantly, virus-mediated anti-tumor immunity was recapitulated in bladder cancer patient-derived organoids. These results suggest that VSVd51-hGM-CSF is a promising viro/immunotherapy that could benefit bladder cancer patients.

8.
Skin Pharmacol Physiol ; 34(4): 214-228, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33910205

RESUMEN

INTRODUCTION: The continuous availability of open micropores is crucial for a successful microneedle (MN) drug delivery strategy. However, micropore lifetime depends on intrinsic skin functional and anatomical characteristics, which vary significantly at different anatomical sites. OBJECTIVE: This pilot study explored if differences exist in micropore closure timeframes at 3 anatomical sites - upper arm, volar forearm, and abdomen. METHODS: Healthy subjects (n = 35) self-identifying as Asian (n = 9), Bi-/multiracial (n = 2), Black (n = 9), Latino (n = 6), and White (n = 9) completed the study. The upper arm, volar forearm, and abdomen were treated with MNs; skin impedance and transepidermal water loss (TEWL) were measured at baseline and post-MN to confirm micropore formation. Impedance was measured for 3 days to evaluate micropore lifetime. Measurements of L*, which quantifies the skin lightness/darkness, were made using a tristimulus colorimeter. Micropore lifetime was determined by comparing baseline and post-MN impedance measurements, and micropore closure half-life was predicted using mathematical modeling. RESULTS: Post-MN increase in TEWL and decrease in impedance were significant (p < 0.05), confirming successful micropore formation at all anatomical sites. When data were analyzed according to subject self-identified racial/ethnic groups, the mean micropore closure time at the abdomen (63.09 ± 13.13 h) was longer than the upper arm (60.34 ± 14.69 h) and volar forearm (58.29 ± 16.76 h). The predicted micropore closure half-life at anatomical sites was the abdomen (25.86 ± 14.96 h) ≈ upper arm (23.69 ± 13.67 h) > volar forearm (20.2 ± 11.99 h). Differences were not statistically significant between groups. Objective categorization by L* showed that the darker skin may be associated with longer micropore closure time at the abdomen site. CONCLUSIONS: Our results suggest that anatomical site of application may not be a source of significant variability in micropore closure time. These findings may help reduce the number of physiological parameters that need to be explicitly considered when developing drug products to support MN-assisted drug delivery strategies.


Asunto(s)
Abdomen/fisiología , Brazo/fisiología , Microinyecciones/métodos , Absorción Cutánea/fisiología , Adolescente , Adulto , Impedancia Eléctrica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Grupos Raciales , Fenómenos Fisiológicos de la Piel , Adulto Joven
9.
Sci Rep ; 10(1): 18963, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33144596

RESUMEN

Microneedles (MNs) allow transdermal delivery of skin-impermeable drugs by creating transient epidermal micropores, and micropore lifetime directly affects drug diffusion timeframes. Healthy subjects (n = 111) completed the study, self-identifying as Asian (n = 32), Bi-/multi-racial (n = 10), Black (n = 22), White (n = 23), Latino (n = 23), and Native American/Hawaiian (n = 1). L* was measured with tristimulus colorimetry to objectively describe skin lightness/darkness. MNs were applied to the upper arm; impedance and transepidermal water loss (TEWL) were measured at baseline and post-MN to confirm micropore formation. Impedance was repeated for 4 days to determine micropore lifetime. Post-MN changes in TEWL and impedance were significant in all groups (p < 0.05), confirming micropore formation regardless of skin type. Micropore lifetime was significantly longer in Blacks (66.5 ± 19.5 h) versus Asians (44.1 ± 14.0 h), Bi-/multi-racial (48.0 ± 16.0 h), and Whites (50.2 ± 2.6 h). Latinos (61.1 ± 16.1 h) had significantly longer micropore closure time versus Asians (44.1 ± 14.0 h). When categorizing data according to L*, micropore lifetime was significantly longer in darker skin. We report for the first time that micropore lifetime differences are present in human subjects of different ethnic/racial backgrounds, with longer micropore lifetime in skin of color. These results also suggest that objectively measured skin color is a better predictor of micropore lifetime than self-identified race/ethnicity.


Asunto(s)
Microinyecciones/métodos , Pigmentación de la Piel/fisiología , Piel/metabolismo , Administración Cutánea , Colorimetría , Espectroscopía Dieléctrica , Sistemas de Liberación de Medicamentos , Humanos
10.
EJNMMI Res ; 10(1): 69, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32592121

RESUMEN

BACKGROUND: Nuclear medicine is on the constant search of precision radiopharmaceutical approaches to improve patient management. Although discordant expression of the estrogen receptor (ER) and the human epidermal growth factor receptor 2 (HER2) in breast cancer is a known dilemma for appropriate patient management, traditional tumor sampling is often difficult or impractical. While 2-deoxy-2[18F]fluoro-D-glucose (18F-FDG)-positron emission tomography (PET) is an option to detect subclinical metastases, it does not provide phenotype information. Radiolabeled antibodies are able to specifically target expressed cell surface receptors. However, their long circulating half-lives (days) require labeling with long-lived isotopes, such as 89Zr, in order to allow sufficient time for tracer clearance from the blood compartment and to accumulate adequately in target tumors and, thus, generate high-quality PET images. The aim of this study was to develop a dual-tracer PET imaging approach consisting of a fast-clearing small molecule and a slow-clearing antibody. This approach was evaluated in a model consisting of mice harboring separate breast cancer xenografts with either an ER+/HER2- or ER-/HER2+ phenotype, comparable to human metastatic disease with intertumor heterogeneity. Lastly, the aim of our study was to determine the feasibility of specifically identifying these two important phenotypes in an acceptable time window. METHODS: Female nude mice were subcutaneously implanted on opposite shoulders with the ER+/HER2- and ER-/HER2+ MCF-7 and JIMT-1 tumor cell lines, respectively. A second model was developed consisting of mice implanted orthotopically with either MCF-7 or JIMT-1 cells. Pharmacokinetic analysis, serial PET imaging, and biodistribution were first performed for [89Zr]Zr-DFO-trastuzumab (89Zr-T) up to 8 days post-injection (p.i.) in JIMT-1 bearing mice. Region-of-interest (ROI) and biodistribution-derived uptake (% injected-activity/gram of tissue [%IA/g]) values and tumor-to-background ratios were obtained. Results were compared in order to validate ROI and identify early time points that provided high contrast tumor images. For the dual-tracer approach, cohorts of tumor-bearing mice were then subjected to sequential tracer PET imaging. On day 1, mice were administered 4-fluoro-11ß-methoxy-16α-[18F]-fluoroestradiol (4FMFES) which targets ER and imaged 45 min p.i. This was immediately followed by the injection of 89Zr-T. Mice were then imaged on day 3 or day 7. ROI analysis was performed, and uptake was calculated in tumors and selected healthy organs for all radiotracers. Quality of tumor targeting for all tracers was evaluated by tumor contrast visualization, tumor and normal tissue uptake, and tumor-to-background ratios. RESULTS: 89Zr-T provided sufficiently high tumor and low background uptake values that furnished high contrast tumor images by 48 h p.i. For the dual-tracer approach, 4FMFES provided tumor uptake values that were significantly increased in MCF-7 tumors. When 89Zr-T-PET was combined with 18F-4FMFES-PET, the entire dual-tracer sequential-imaging procedure provided specific high-quality contrast images of ER+/HER2- MCF-7 and ER-/HER2+ JIMT-1 tumors for 4FMFES and 89Zr-T, respectively, as short as 72 h from start to finish. CONCLUSIONS: This protocol can provide high contrast images of tumors expressing ER or HER2 within 3 days from injection of 4FMFES to final scan of 89Zr-T and, hence, provides a basis for future dual-tracer combinations that include antibodies.

12.
BMC Cancer ; 19(1): 823, 2019 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-31429730

RESUMEN

BACKGROUND: Natural killer (NK) cell dysfunction following cancer surgery has been shown to promote metastases. Recent studies demonstrate an emerging role for lipids in the modulation of NK cell innate responses. However, the mechanisms involved in lipid modulation of NK cell postoperative anti-tumor function are unknown. This current study will determine whether the lipid accumulation via scavenger receptors on NK cells is responsible for the increase in postoperative metastasis. METHODS: Lipid content in mouse and human NK cells was evaluated by flow cytometry. NK cell scavenger receptor (SR) expression was measured by microarray analysis, validated by qRT-PCR and flow cytometry. NK cell ex vivo and in vivo tumor killing was measured by chromium-release and adoptive transfer assays, respectively. The mediating role of surgery-expanded granulocytic myeloid derived suppressor cells (gMDSC) in SR induction on NK cells was evaluated using co-culture assays. RESULTS: NK cells in surgery-treated mice demonstrated increased lipid accumulation, which occurred via up-regulation of MSR1, CD36 and CD68. NK cells with high lipid content had diminished ability to lyse tumor targets ex vivo. Adoptive transfer of lipid-laden NK cells into NK cell-deficient mice were unable to protect against a lung tumor challenge. Granulocytic MDSC from surgery-treated mice increased SR expression on NK cells. Colorectal cancer surgical patients showed increased NK cell lipid content, higher CD36 expression, decreased granzyme B and perforin production in addition to reduced cytotoxicity in the postoperative period. CONCLUSIONS: Postoperative lipid accumulation promotes the formation of metastases by impairing NK cell function in both preclinical surgical models and human surgical colorectal cancer patient samples. Understanding and targeting the mechanisms underlying lipid accumulation in innate immune NK cells can improve prognosis in cancer surgical patients.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Citotoxicidad Inmunológica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ácidos Palmíticos/metabolismo , Traslado Adoptivo , Animales , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos CD36/genética , Neoplasias Colorrectales/cirugía , Modelos Animales de Enfermedad , Femenino , Granzimas/metabolismo , Humanos , Células K562 , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Perforina/metabolismo , Periodo Posoperatorio , Receptores Depuradores/genética , Receptores Depuradores de Clase A/genética
13.
Viruses ; 11(5)2019 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-31083491

RESUMEN

Oncolytic viruses (OVs) are a form of immunotherapy that release tumor antigens in the context of highly immunogenic viral signals following tumor-targeted infection and destruction. Emerging preclinical and clinical evidence suggests that this in situ vaccine effect is critical for successful viro-immunotherapy. In this review, we discuss the application of OV as an infected cell vaccine (ICV) as one method of enhancing the potency and breadth of anti-tumoral immunity. We focus on understanding and manipulating the critical role of natural killer (NK) cells and their interactions with other immune cells to promote a clinical outcome. With a synergistic tumor killing and immune activating mechanism, ICVs represent a valuable new addition to the cancer fighting toolbox with the potential to treat malignant disease.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Humanos , Inmunoterapia , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/inmunología
14.
mBio ; 10(1)2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30755505

RESUMEN

The large genome of human cytomegalovirus (HCMV) is transcribed by RNA polymerase II (Pol II). However, it is not known how closely this betaherpesvirus follows host transcriptional paradigms. We applied PRO-Seq and PRO-Cap methods to profile and quantify transcription initiation and productive elongation across the host and virus genomes in late infection. A major similarity between host transcription and viral transcription is that treatment of cells with the P-TEFb inhibitor flavopiridol preempts virtually all productive elongation, which otherwise covers most of the HCMV genome. The deep, nucleotide resolution identification of transcription start sites (TSSs) enabled an extensive analysis of core promoter elements. An important difference between host and viral transcription is that initiation is much more pervasive on the HCMV genome. The sequence preferences in the initiator region around the TSS and the utilization of upstream T/A-rich elements are different. Upstream TATA positions the TSS and boosts initiation in both the host and the virus, but upstream TATT has a significant stimulatory impact only on the viral template. The major immediate early (MIE) promoter remained active during late infection and was accompanied by transcription of both strands of the MIE enhancer from promoters within the enhancer. Surprisingly, we found that the long noncoding RNA4.9 is intimately associated with the viral origin of replication (oriLyt) and was transcribed to a higher level than any other viral or host promoter. Finally, our results significantly contribute to the idea that late in infection, transcription takes place on viral genomes that are not highly chromatinized.IMPORTANCE Human cytomegalovirus infects more than half of humans, persists silently in virtually all tissues, and produces life-threatening disease in immunocompromised individuals. HCMV is also the most common infectious cause of birth defects and the leading nongenetic cause of sensorineural hearing loss in the United States. Because there is no vaccine and current drugs have problems with potency, toxicity, and antiviral drug resistance, alternative treatment strategies that target different points of viral control are needed. Our current study contributes to this goal by applying newly developed methods to examine transcription of the HCMV and host genomes at nucleotide resolution in an attempt to find targetable differences between the two. After a thorough analysis of productive elongation and of core promoter element usage, we found that some mechanisms of regulating transcription are shared between the host and HCMV but that others are distinctly different. This suggests that HCMV transcription may be a legitimate target for future antiviral therapies and this might translate to other herpesviruses.


Asunto(s)
Citomegalovirus/genética , Genoma Humano , Genoma Viral , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética , Células Cultivadas , Inhibidores Enzimáticos/metabolismo , Flavonoides/metabolismo , Humanos , Piperidinas/metabolismo
15.
J Immunother Cancer ; 6(1): 86, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30176921

RESUMEN

BACKGROUND: Cancer surgery is necessary and life-saving. However, the majority of patients develop postoperative recurrence and metastasis, which are the main causes of cancer-related deaths. The postoperative stress response encompasses a broad set of physiological changes that have evolved to safeguard the host following major tissue trauma. These stress responses, however, intersect with cellular mediators and signaling pathways that contribute to cancer proliferation. MAIN: Previous descriptive and emerging mechanistic studies suggest that the surgery-induced prometastatic effect is linked to impairment of both innate and adaptive immunity. Existing studies that combine surgery and immunotherapies have revealed that this combination strategy is not straightforward and patients have experienced both therapeutic benefit and drawbacks. This review will specifically assess the immunological pathways that are disrupted by oncologic surgical stress and provide suggestions for rationally combining cancer surgery with immunotherapies to improve immune and treatment outcomes. SHORT CONCLUSION: Given the prevalence of surgery as frontline therapy for solid cancers, the emerging data on postoperative immunosuppression and the rapid development of immunotherapy for oncologic treatment, we believe that future targeted studies of perioperative immunotherapy are warranted.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Atención Perioperativa , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Manejo de la Enfermedad , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/cirugía , Atención Perioperativa/efectos adversos , Atención Perioperativa/métodos , Nivel de Atención , Resultado del Tratamiento
16.
Nucleic Acids Res ; 45(19): 11088-11105, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28977633

RESUMEN

Oxidative stress has pervasive effects on cells but how they respond transcriptionally upon the initial insult is incompletely understood. We developed a nuclear walk-on assay that semi-globally quantifies nascent transcripts in promoter-proximal paused RNA polymerase II (Pol II). Using this assay in conjunction with ChIP-Seq, in vitro transcription, and a chromatin retention assay, we show that within a minute, hydrogen peroxide causes accumulation of Pol II near promoters and enhancers that can best be explained by a rapid decrease in termination. Some of the accumulated polymerases slowly move or 'creep' downstream. This second effect is correlated with and probably results from loss of NELF association and function. Notably, both effects were independent of DNA damage and ADP-ribosylation. Our results demonstrate the unexpected speed at which a global transcriptional response can occur. The findings provide strong support for the residence time of paused Pol II elongation complexes being much shorter than estimated from previous studies.


Asunto(s)
Genoma Humano/genética , Estrés Oxidativo , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Interferencia de ARN , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
17.
Proc Natl Acad Sci U S A ; 114(18): 4781-4786, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28428342

RESUMEN

The cell wall of Gram-positive bacteria contains abundant surface-exposed carbohydrate molecules that are highly conserved within and often across species. The potential therapeutic usefulness of high-affinity antibodies to cell wall carbohydrates is unquestioned, however obtaining such antibodies is challenging due to the poor overall immunogenicity of these bacterial targets. Autolysins and phage lysins are peptidoglycan hydrolases, enzymes that have evolved over a billion years to degrade bacterial cell wall. Such wall hydrolases are modular enzymes, composed of discrete domains for high-affinity binding to cell wall carbohydrates and cleavage activity. In this study, we demonstrate that binding domains from autolysins and lysins can be fused to the Fc region of human IgG, creating a fully functional homodimer (or "lysibody") with high-affinity binding and specificity for carbohydrate determinants on the bacterial surface. Furthermore, we demonstrate that this process is reproducible with three different binding domains specific to methicillin-resistant Staphylococcus aureus (MRSA). Cell-bound lysibodies induced the fixation of complement on the bacterial surface, promoted phagocytosis by macrophages and neutrophils, and protected mice from MRSA infection in two model systems. The lysibody approach could be used to target a range of difficult-to-treat pathogenic bacteria, given that cell wall hydrolases are ubiquitous in nature.


Asunto(s)
Pared Celular/metabolismo , Fragmentos Fc de Inmunoglobulinas/farmacología , N-Acetil Muramoil-L-Alanina Amidasa/farmacología , Fagocitosis/efectos de los fármacos , Polisacáridos Bacterianos/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Staphylococcus aureus/metabolismo , Células HL-60 , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , N-Acetil Muramoil-L-Alanina Amidasa/genética , Polisacáridos Bacterianos/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética
18.
Infect Immun ; 84(7): 2051-2058, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27113359

RESUMEN

The Treponema denticola FhbB protein contributes to immune evasion by binding factor H (FH). Cleavage of FH by the T. denticola protease, dentilisin, may contribute to the local immune dysregulation that is characteristic of periodontal disease (PD). Although three FhbB phyletic types have been defined (FhbB1, FhbB2, and FhbB3), the in vivo expression patterns and antigenic heterogeneity of FhbB have not been assessed. Here, we demonstrate that FhbB is a dominant early antigen that elicits FhbB type-specific antibody (Ab) responses. Using the murine skin abscess model, we demonstrate that the presence or absence of FhbB or dentilisin significantly influences Ab responses to infection and skin abscess formation. Competitive binding analyses revealed that α-FhbB Ab can compete with FH for binding to T. denticola and block dentilisin-mediated FH cleavage. Lastly, we demonstrate that dentilisin cleavage sites reside within critical functional domains of FH, including the complement regulatory domain formed by CCPs 1 to 4. Analysis of the FH cleavage products revealed that they lack cofactor activity. The data presented here provide insight into the in vivo significance of dentilisin, FhbB and its antigenic diversity, and the potential impact of FH cleavage on the regulation of complement activation.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Péptido Hidrolasas/metabolismo , Treponema denticola/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Especificidad de Anticuerpos/inmunología , Antígenos Bacterianos/metabolismo , Unión Competitiva , Factor H de Complemento/química , Ratones , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Dominios y Motivos de Interacción de Proteínas , Proteolisis
19.
Breast Cancer Res ; 17: 47, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25880415

RESUMEN

INTRODUCTION: Focal adhesion kinase (FAK) controls cell growth and survival downstream of integrin-matrix receptors. Upon adhesion loss or FAK inhibition, FAK can translocate to the nucleus. The nucleolus is a non-membrane nuclear structure that regulates ribosome biogenesis and cell proliferation. Nucleostemin (NS), a nucleolar-localized protein, modulates cell cycle progression, stemness, and three-dimensional tumor spheroid formation. The signaling pathways that regulate NS levels in tumors remain undefined. METHODS: Human breast carcinoma cells were evaluated for growth in culture (adherent and anchorage-independent spheroid) and as orthotopic tumors. FAK signaling was evaluated by pharmacological FAK inhibitor addition (PF-271, IC50~0.1 µM) and by small hairpin RNA (shRNA) knockdown followed by re-expression of FAK wildtype (WT) or a kinase-dead (KD, K454R) FAK point mutant. Immunoblotting was used to evaluate FAK, NS, nucleolar phosphoprotein B23, and nucleolin levels. Total and phosphospecific antibody imunoblotting were used to detect changes in FAK, Akt kinase (Akt also known as protein kinase B), and 4E-binding protein 1 (4E-BP1) phosphorylation, a translation repressor protein and target of the mammalian target of rapamycin (mTOR) complex. Immunohistochemical, co-immunoprecipitation, and cellular fractionation analyses were used to evaluate FAK association with nucleoli. RESULTS: Pharmacological (0.1 µM PF-271) or genetic inhibition of FAK activity prevents MDA-MB-231 and 4T1L breast carcinoma growth as spheroids and as orthotopic tumors. FAK inhibition triggers proteasome-mediated decreased NS levels but no changes in other nucleolar proteins such as B23 (nucleophosmin) or nucleolin. Active FAK was associated with purified nucleoli of anchorage-independent cells and present within nucleoli of human invasive ductal carcinoma tumor samples. FAK co-immunoprecipitated with B23 that binds NS and a complex between FAK, NS, Akt, and mTOR was detected. Constitutively-active Akt kinase promoted tumor spheroid growth, stabilized NS levels, and promoted pS65 4E-BP1 phosphorylation in the presence of inhibited FAK. Rapamycin lowered NS levels and inhibited pS65 4E-BP1 phosphorylation in cells with activated Akt-mTOR signaling. CONCLUSIONS: FAK signaling occurs in the nucleolus, active FAK protects NS, and Akt-mTOR pathway regulates NS protein stability needed for breast carcinoma spheroid and tumor growth.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas Nucleares/metabolismo , Animales , Neoplasias de la Mama/genética , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Activación Enzimática , Femenino , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Ratones , Nucleofosmina , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Sirolimus/farmacología , Esferoides Celulares , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
20.
Mol Cancer Ther ; 13(8): 2050-61, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899686

RESUMEN

Ovarian cancer ascites fluid contains matrix proteins that can impact tumor growth via integrin receptor binding. In human ovarian tumor tissue arrays, we find that activation of the cytoplasmic focal adhesion (FAK) tyrosine kinase parallels increased tumor stage, ß5 integrin, and osteopontin matrix staining. Elevated osteopontin, ß5 integrin, and FAK mRNA levels are associated with decreased serous ovarian cancer patient survival. FAK remains active within ovarian cancer cells grown as spheroids, and anchorage-independent growth analyses of seven ovarian carcinoma cell lines identified sensitive (HEY, OVCAR8) and resistant (SKOV3-IP, OVCAR10) cells to 0.1 µmol/L FAK inhibitor (VS-4718, formerly PND-1186) treatment. VS-4718 promoted HEY and OVCAR8 G0-G1 cell-cycle arrest followed by cell death, whereas growth of SKOV3-IP and OVCAR10 cells was resistant to 1.0 µmol/L VS-4718. In HEY cells, genetic or pharmacological FAK inhibition prevented tumor growth in mice with corresponding reductions in ß5 integrin and osteopontin expression. ß5 knockdown reduced HEY cell growth in soft agar, tumor growth in mice, and both FAK Y397 phosphorylation and osteopontin expression in spheroids. FAK inhibitor-resistant (SKOV3-IP, OVCAR10) cells exhibited anchorage-independent Akt S473 phosphorylation, and expression of membrane-targeted and active Akt in sensitive cells (HEY, OVCAR8) increased growth but did not create a FAK inhibitor-resistant phenotype. These results link osteopontin, ß5 integrin, and FAK in promoting ovarian tumor progression. ß5 integrin expression may serve as a biomarker for serous ovarian carcinoma cells that possess active FAK signaling.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Cadenas beta de Integrinas/metabolismo , Neoplasias Quísticas, Mucinosas y Serosas/metabolismo , Neoplasias Ováricas/metabolismo , Aminopiridinas/farmacología , Animales , Antineoplásicos/farmacología , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Quísticas, Mucinosas y Serosas/mortalidad , Osteopontina/metabolismo , Neoplasias Ováricas/mortalidad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...