Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Thorac Dis ; 16(4): 2528-2538, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38738248

ABSTRACT

Background: The mortality rate of coronary artery disease ranks first in developed countries, and coronary revascularization therapy is an important cornerstone of its treatment. The postoperative pulmonary complications (PPCs) in patients receiving one-stop hybrid coronary revascularization (HCR) aggravate the dysfunction of multiple organs such as the heart and lungs, therefore increasing mortality. However, the risk factors are still unclear. The objective of this study was to explore the risk factors of PPCs after HCR surgery. Methods: In this study, the perioperative data of 311 patients undergoing HCR surgery were reviewed. All patients were divided into two groups according to whether the PPCs occurred. The baseline information and surgery-related indicators in preoperative laboratory examination, intraoperative fluid management, and anesthesia management were compared between the two groups. Results: Advanced age [odds ratio (OR): 1.065, 95% confidence interval (CI): 1.030-1.101, P<0.001], high body mass index (BMI; OR: 1.113, 95% CI: 1.011-1.225, P=0.02), history of percutaneous coronary intervention (PCI) surgery (OR: 2.831, 95% CI: 1.388-5.775, P=0.004), one-lung volume ventilation (OR: 3.804, 95% CI: 1.923-7.526, P<0.001), inhalation of high concentration oxygen (OR: 3.666, 95% CI: 1.719-7.815, P=0.001), the application of positive end-expiratory pressure (PEEP; OR: 2.567, 95% CI: 1.338-4.926, P=0.005), and long one-lung ventilation time (OR: 1.015, 95% CI: 1.006-1.023, P=0.001) may be risk factors for postoperative PPCs in patients undergoing one-stop coronary revascularization surgery. Using the above seven factors to jointly predict the risk of PPCs in patients undergoing one-stop coronary revascularization surgery, the receiver operating characteristic (ROC) curve showed an area under the curve (AUC) =0.873, 95% CI: 0.835-0.911, sensitivity: 84.81%, and specificity: 75.82%; the predictive model was shown to be effective. Conclusions: Patients undergoing HCR surgery with advanced age, high BMI, a history of PCI surgery, one-lung volume ventilation, inhalation of high concentration oxygen, use of PEEP, and prolonged single lung ventilation are more prone to PPCs.

2.
Adv Sci (Weinh) ; 11(18): e2307233, 2024 May.
Article in English | MEDLINE | ID: mdl-38487926

ABSTRACT

The gut microbiome has emerged as a potential target for the treatment of cardiovascular disease. Ischemia/reperfusion (I/R) after myocardial infarction is a serious complication and whether certain gut bacteria can serve as a treatment option remains unclear. Lactobacillus reuteri (L. reuteri) is a well-studied probiotic that can colonize mammals including humans with known cholesterol-lowering properties and anti-inflammatory effects. Here, the prophylactic cardioprotective effects of L. reuteri or its metabolite γ-aminobutyric acid (GABA) against acute ischemic cardiac injury caused by I/R surgery are demonstrated. The prophylactic gavage of L. reuteri or GABA confers cardioprotection mainly by suppressing cardiac inflammation upon I/R. Mechanistically, GABA gavage results in a decreased number of proinflammatory macrophages in I/R hearts and GABA gavage no longer confers any cardioprotection in I/R hearts upon the clearance of macrophages. In vitro studies with LPS-stimulated bone marrow-derived macrophages (BMDM) further reveal that GABA inhibits the polarization of macrophages toward the proinflammatory M1 phenotype by inhibiting lysosomal leakage and NLRP3 inflammasome activation. Together, this study demonstrates that the prophylactic oral administration of L. reuteri or its metabolite GABA attenuates macrophage-mediated cardiac inflammation and therefore alleviates cardiac dysfunction after I/R, thus providing a new prophylactic strategy to mitigate acute ischemic cardiac injury.


Subject(s)
Disease Models, Animal , Limosilactobacillus reuteri , Mice, Inbred C57BL , Probiotics , gamma-Aminobutyric Acid , Animals , Limosilactobacillus reuteri/metabolism , Mice , gamma-Aminobutyric Acid/metabolism , Probiotics/administration & dosage , Probiotics/therapeutic use , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Macrophages/metabolism , Gastrointestinal Microbiome , Myocardial Ischemia/metabolism , Myocardial Ischemia/prevention & control
3.
BMC Anesthesiol ; 24(1): 76, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38408904

ABSTRACT

BACGROUND: Off-pump coronary artery bypass graft (OPCABG) has a high incidence of postoperative systemic inflammation response syndrome (SIRS), and perioperative endothelial glycocalyx layer (EGL) disruption can be one of the predisposing factors. We hypothesized that EGL shedding happened earlier in OPCABG which can influence on postoperative SIRS, and sevoflurane might preserve EGL better than propofol. METHODS: We randomly allocated 50 patients undergoing OPCABG to receive either sevoflurane-sufentanil or propofol-sufentanil anesthesia. Plasma syndecan-1, heparan sulfate (HS), atrial natriuretic peptide (ANP), IL-6, and cardiac troponin I (cTnI) were measured. Blood samples were collected at 6 timepoints: induction (T1), before grafting (T2), after grafting(T3), surgery done (T4), postoperative day1 (POD1,T5) and POD2 (T6). SIRS criteria and sequential organ failure assessment (SOFA) score were examined. RESULTS: There were neither differences of syndecan-1, HS, IL-6 nor of SIRS criteria or SOFA score between the sevoflurane and propofol groups. All patients were pooled as a single group for further statistical analyses, plasma syndecan-1 (P < 0.001) and IL-6 (P < 0.001) increased significantly as a function of time; syndecan-1 increasing correlated significantly with the duration of coronary graft anastomosis (r = 0.329, P = 0.026). Syndecan-1(T3) correlated significantly with ANP(T3) (r = 0.0.354, P = 0.016) and IL-6 (T5) (r = 0.570, P < 0.001). The maximum value of IL-6 correlated significantly with SIRS (r = 0.378, P = 0.010), the maximum value of SOFA score (r = 0.399, P = 0.006) and ICU days (r = 0.306, P = 0.039). The maximum value of SOFA score correlated significantly with the occurrence of SIRS (r = 0.568, P < 0.001) and ICU days (r = 0.338, P = 0.022). CONCLUSIONS: OPCABG intraoperative early EGL shedding caused of grafts anastomosis greatly affected postoperative SIRS and SOFA score, sevoflurane did not clinically preserve EGL better. TRIAL REGISTRATION: ChiCTR-IOR-17012535. Registered on 01/09/2017.


Subject(s)
Glycocalyx , Propofol , Humans , Syndecan-1 , Propofol/pharmacology , Sevoflurane , Sufentanil , Interleukin-6 , Inflammation , Systemic Inflammatory Response Syndrome
4.
Sci Adv ; 9(47): eadg8876, 2023 11 24.
Article in English | MEDLINE | ID: mdl-38000020

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive soft tissue sarcomas with limited treatment options, and new effective therapeutic strategies are desperately needed. We observe antiproliferative potency of genetic depletion of PTPN11 or pharmacological inhibition using the SHP2 inhibitor (SHP2i) TNO155. Our studies into the signaling response to SHP2i reveal that resistance to TNO155 is partially mediated by reduced RB function, and we therefore test the addition of a CDK4/6 inhibitor (CDK4/6i) to enhance RB activity and improve TNO155 efficacy. In combination, TNO155 attenuates the adaptive response to CDK4/6i, potentiates its antiproliferative effects, and converges on enhancement of RB activity, with greater suppression of cell cycle and inhibitor-of-apoptosis proteins, leading to deeper and more durable antitumor activity in in vitro and in vivo patient-derived models of MPNST, relative to either single agent. Overall, our study provides timely evidence to support the clinical advancement of this combination strategy in patients with MPNST and other tumors driven by loss of NF1.


Subject(s)
Neurofibrosarcoma , Humans , Signal Transduction , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics
5.
bioRxiv ; 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36778419

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNST) are highly aggressive soft tissue sarcomas with limited treatment options, and novel effective therapeutic strategies are desperately needed. We observe anti-proliferative efficacy of genetic depletion or pharmacological inhibition using the clinically available SHP2 inhibitor (SHP2i) TNO155. Our studies into the signaling response to SHP2i reveal that resistance to TNO155 is partially mediated by reduced RB function, and we therefore test the addition of a CDK4/6 inhibitor (CDK4/6i) to enhance RB activity and improve TNO155 efficacy. In combination, TNO155 attenuates the adaptive response to CDK4/6i, potentiates its anti-proliferative effects, and converges on enhancement of RB activity, with greater suppression of cell cycle and inhibitor-of-apoptosis proteins, leading to deeper and more durable anti-tumor activity in in vitro and in vivo patient-derived models of MPNST, relative to either single agent. Overall, our study provides timely evidence to support the clinical advancement of this combination strategy in patients with MPNST and other tumors driven by loss of NF1.

7.
Oncogene ; 41(21): 2973-2983, 2022 05.
Article in English | MEDLINE | ID: mdl-35459782

ABSTRACT

Activating RAS mutations are found in a subset of fusion-negative rhabdomyosarcoma (RMS), and therapeutic strategies to directly target RAS in these tumors have been investigated, without clinical success to date. A potential strategy to inhibit oncogenic RAS activity is the disruption of RAS prenylation, an obligate step for RAS membrane localization and effector pathway signaling, through inhibition of farnesyltransferase (FTase). Of the major RAS family members, HRAS is uniquely dependent on FTase for prenylation, whereas NRAS and KRAS can utilize geranylgeranyl transferase as a bypass prenylation mechanism. Tumors driven by oncogenic HRAS may therefore be uniquely sensitive to FTase inhibition. To investigate the mutation-specific effects of FTase inhibition in RMS we utilized tipifarnib, a potent and selective FTase inhibitor, in in vitro and in vivo models of RMS genomically characterized for RAS mutation status. Tipifarnib reduced HRAS processing, and plasma membrane localization leading to decreased GTP-bound HRAS and decreased signaling through RAS effector pathways. In HRAS-mutant cell lines, tipifarnib reduced two-dimensional and three-dimensional cell growth, and in vivo treatment with tipifarnib resulted in tumor growth inhibition exclusively in HRAS-mutant RMS xenografts. Our data suggest that small molecule inhibition of FTase is active in HRAS-driven RMS and may represent an effective therapeutic strategy for a genomically-defined subset of patients with RMS.


Subject(s)
Rhabdomyosarcoma, Embryonal , Rhabdomyosarcoma , Farnesyltranstransferase/genetics , Genes, ras , Humans , Prenylation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/genetics
8.
Clin Cancer Res ; 27(22): 6197-6208, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34433654

ABSTRACT

PURPOSE: Selective RAF-targeted therapy is effective in some patients with BRAFV600E-mutated glioma, though emergent and adaptive resistance occurs through ill-defined mechanisms. EXPERIMENTAL DESIGN: Paired pre-/post- RAF inhibitor (RAFi)-treated glioma samples (N = 15) were obtained and queried for treatment-emergent genomic alterations using DNA and RNA sequencing (RNA-seq). Functional validation of putative resistance mechanisms was performed using established and patient-derived BRAFV600E-mutant glioma cell lines. RESULTS: Analysis of 15 tissue sample pairs identified 13 alterations conferring putative resistance were identified among nine paired samples (including mutations involving ERRFI1, BAP1, ANKHD1, and MAP2K1). We performed functional validation of mechanisms of resistance, including loss of NF1, PTEN, or CBL, in BRAFV600E-mutant glioma lines, and demonstrate they are capable of conferring resistance in vitro. Knockdown of CBL resulted in increased EGFR expression and phosphorylation, a possible mechanism for maintaining ERK signaling within the cell. Combination therapy with a MEKi or EGFR inhibitor was able to overcome resistance to BRAFi, in NF1 knockdown and CBL knockdown, respectively. Restoration of wild-type PTEN in B76 cells (PTEN-/-) restored sensitivity to BRAFi. We identified and validated CRAF upregulation as a mechanism of resistance in one resistant sample. RNA-seq analysis identified two emergent expression patterns in resistant samples, consistent with expression patterns of known glioma subtypes. CONCLUSIONS: Resistance mechanisms to BRAFi in glioma are varied and may predict effective precision combinations of targeted therapy, highlighting the importance of a personalized approach.


Subject(s)
Glioma , Proto-Oncogene Proteins B-raf , Glioma/drug therapy , Glioma/genetics , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins , Signal Transduction , Tumor Suppressor Proteins , Ubiquitin Thiolesterase
9.
Cancer Res ; 81(3): 747-762, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33203698

ABSTRACT

Malignant peripheral nerve sheath tumors often arise in patients with neurofibromatosis type 1 and are among the most treatment-refractory types of sarcoma. Overall survival in patients with relapsed disease remains poor, and thus novel therapeutic approaches are needed. NF1 is essential for negative regulation of RAS activity and is altered in about 90% of malignant peripheral nerve sheath tumors (MPNST). A complex interplay of upstream signaling and parallel RAS-driven pathways characterizes NF1-driven tumorigenesis, and inhibiting more than one RAS effector pathway is therefore necessary. To devise potential combination therapeutic strategies, we identified actionable alterations in signaling that underlie adaptive and acquired resistance to MEK inhibitor (MEKi). Using a series of proteomic, biochemical, and genetic approaches in an in vitro model of MEKi resistance provided a rationale for combination therapies. HGF/MET signaling was elevated in the MEKi-resistant model. HGF overexpression conferred resistance to MEKi in parental cells. Depletion of HGF or MET restored sensitivity of MEKi-resistant cells to MEKi. Finally, a combination of MEK and MET inhibition demonstrated activity in models of MPNST and may therefore be effective in patients with MPNST harboring genetic alterations in NF1. SIGNIFICANCE: This study demonstrates that MEKi plus MET inhibitor may delay or prevent a novel mechanism of acquired MEKi resistance, with clinical implications for MPNST patients harboring NF1 alterations.


Subject(s)
Drug Resistance, Neoplasm , Neoplasm Recurrence, Local/enzymology , Nerve Sheath Neoplasms/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation , Female , Hepatocyte Growth Factor/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Nerve Sheath Neoplasms/drug therapy , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/mortality , Neurofibromatosis 1/complications , Neurofibromatosis 1/metabolism , Neurofibromin 1/deficiency , Neurofibromin 1/genetics , Protein Kinase Inhibitors/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proteomics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pyridones/pharmacology , Pyrimidinones/pharmacology , Random Allocation , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , ras Proteins/antagonists & inhibitors , ras Proteins/metabolism
10.
J Cardiothorac Vasc Anesth ; 35(7): 2201-2211, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32736999

ABSTRACT

Despite advances in the therapy for pulmonary hypertension over the past decades, the prognosis of pregnant patients with pulmonary hypertension remains poor, with high maternal mortality. This poses a particular challenge for the mother and her medical team. In the present review, the authors have updated the classification and definition of pulmonary hypertension, summarized the current knowledge with regard to perioperative management and anesthesia considerations for these patients, and stressed the importance of a "pregnancy heart team" to improve long-term outcomes of pregnant women with pulmonary hypertension.


Subject(s)
Anesthesia , Hypertension, Pulmonary , Pregnancy Complications, Cardiovascular , Cesarean Section , Female , Humans , Hypertension, Pulmonary/complications , Pregnancy , Pregnant Women
11.
Neurooncol Adv ; 2(1): vdaa138, 2020.
Article in English | MEDLINE | ID: mdl-33235998

ABSTRACT

BACKGROUND: RAS effector signaling pathways such as PI3K/mTOR and ERK are frequently dysregulated in glioblastoma. While small molecule targeted therapies against these pathways have appeared promising in preclinical studies, they have been disappointing in clinical trials due to toxicity and de novo and adaptive resistance. To identify predictors of glioblastoma sensitivity to dual pathway inhibition with mTORC1/2 and MEK inhibitors, we tested these agents, alone and in combination, in a cohort of genomically characterized glioblastoma cell lines. METHODS: Seven genomically characterized, patient-derived glioblastoma neurosphere cell lines were evaluated for their sensitivity to the dual mTORC1/2 kinase inhibitor sapanisertib (MLN0128, TAK-228) alone or in combination with the MEK1/2 inhibitor trametinib (GSK1120212), using assessment of proliferation and evaluation of the downstream signaling consequences of these inhibitors. RESULTS: Sapanisertib inhibited cell growth in neurosphere lines, but induced apoptosis only in a subset of lines, and did not completely inhibit downstream mTOR signaling via ribosomal protein S6 (RPS6). Growth sensitivity to MEK inhibitor monotherapy was observed in a subset of lines defined by loss of NF1, was predicted by an ERK-dependent expression signature, and was associated with effective phospho-RPS6 inhibition. In these lines, combined MEK/mTOR treatment further inhibited growth and induced apoptosis. Combined MEK and mTOR inhibition also led to modest antiproliferative effects in lines with intact NF1 and insensitivity to MEK inhibitor monotherapy. CONCLUSIONS: These data demonstrate that combined MEK/mTOR inhibition is synergistic in glioblastoma cell lines and may be more potent in NF1-deficient glioblastoma.

12.
Cancer Res ; 80(23): 5367-5379, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33032988

ABSTRACT

Loss of the RAS GTPase-activating protein (RAS-GAP) NF1 drives aberrant activation of RAS/MEK/ERK signaling and other effector pathways in the majority of malignant peripheral nerve sheath tumors (MPNST). These dysregulated pathways represent potential targets for therapeutic intervention. However, studies of novel single agents including MEK inhibitors (MEKi) have demonstrated limited efficacy both preclinically and clinically, with little advancement in overall patient survival. By interrogation of kinome activity through an unbiased screen and targeted evaluation of the signaling response to MEK inhibition, we have identified global activation of upstream receptor tyrosine kinases (RTK) that converges on activation of RAS as a mechanism to limit sensitivity to MEK inhibition. As no direct inhibitors of pan-RAS were available, an inhibitor of the protein tyrosine phosphatase SHP2, a critical mediator of RAS signal transduction downstream of multiple RTK, represented an alternate strategy. The combination of MEKi plus SHP099 was superior to MEKi alone in models of NF1-MPNST, including those with acquired resistance to MEKi. Our findings have immediate translational implications and may inform future clinical trials for patients with MPNST harboring alterations in NF1. SIGNIFICANCE: Combined inhibition of MEK and SHP2 is effective in models of NF1-MPNST, both those naïve to and those resistant to MEKi, as well as in the MPNST precursor lesion plexiform neurofibroma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Nerve Sheath Neoplasms/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Humans , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Sheath Neoplasms/metabolism , Neurofibroma/drug therapy , Neurofibroma/pathology , Neurofibromin 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Pyridones/administration & dosage , Pyridones/pharmacology , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacology , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/metabolism
13.
J Int Med Res ; 48(8): 300060520945895, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32812467

ABSTRACT

Cardiac tamponade is a rare complication that occurs during hemihepatectomy. This particular complication has a high degree of mortality and morbidity. A 51-year-old woman was admitted to our hospital for surgical treatment of a malignant liver tumor. During surgery, she developed sudden hemodynamic instability and signs suggesting cardiac tamponade, which was confirmed via transthoracic echocardiogram. Cardiac compression and creation of a pericardial window resulted in immediate hemodynamic improvement. At completion of surgery, a repeated transthoracic echocardiogram showed no pericardial effusion. Early ultrasound-assisted diagnosis and treatment of cardiac tamponade are crucial. Although cardiac tamponade rarely occurs during hemihepatectomy, medics should be aware of this possibility to ensure prompt diagnosis. Our findings strongly support the use of early cardiac compression in cardiac arrest during surgery with echocardiography for prompt and accurate diagnosis of cardiac tamponade. Additionally, our findings will hopefully make anesthesiologists aware of the need to maintain a high index of suspicion for cardiac tamponade with sudden hypotension and a large reduction in differential pressure, and encourage early use of echocardiography and timely cardiac compression.


Subject(s)
Cardiac Tamponade , Pericardial Effusion , Cardiac Tamponade/diagnostic imaging , Cardiac Tamponade/etiology , Echocardiography , Female , Heart , Humans , Middle Aged , Pericardial Effusion/diagnostic imaging , Pericardial Effusion/etiology , Ultrasonography
14.
Genes (Basel) ; 11(6)2020 06 24.
Article in English | MEDLINE | ID: mdl-32599735

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNST) are rare, aggressive soft tissue sarcomas that occur with significantly increased incidence in people with the neuro-genetic syndrome neurofibromatosis type I (NF1). These complex karyotype sarcomas are often difficult to resect completely due to the involvement of neurovascular bundles, and are relatively chemotherapy- and radiation-insensitive. The lifetime risk of developing MPNST in the NF1 population has led to great efforts to characterize the genetic changes that drive the development of these tumors and identify mutations that may be used for diagnostic or therapeutic purposes. Advancements in genetic sequencing and genomic technologies have greatly enhanced researchers' abilities to broadly and deeply investigate aberrations in human MPNST genomes. Here, we review genetic sequencing efforts in human MPNST samples over the past three decades. Particularly for NF1-associated MPNST, these overall sequencing efforts have converged on a set of four common genetic changes that occur in most MPNST, including mutations in neurofibromin 1 (NF1), CDKN2A, TP53, and members of the polycomb repressor complex 2 (PRC2). However, broader genomic studies have also identified recurrent but less prevalent genetic variants in human MPNST that also contribute to the molecular landscape of MPNST and may inform further research. Future studies to further define the molecular landscape of human MPNST should focus on collaborative efforts across multiple institutions in order to maximize information gathered from large numbers of well-annotated MPNST patient samples, both in the NF1 and the sporadic MPNST populations.


Subject(s)
Evolution, Molecular , Neoplasm Proteins/genetics , Neurofibrosarcoma/genetics , Transcriptome/genetics , Chromosome Aberrations , Cyclin-Dependent Kinase Inhibitor p16/genetics , Genomics , Humans , Mutation/genetics , Neurofibromin 1/genetics , Neurofibrosarcoma/pathology , Proteomics , Tumor Suppressor Protein p53/genetics
15.
Sci Data ; 7(1): 184, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561749

ABSTRACT

Nerve sheath tumors occur as a heterogeneous group of neoplasms in patients with neurofibromatosis type 1 (NF1). The malignant form represents the most common cause of death in people with NF1, and even when benign, these tumors can result in significant disfigurement, neurologic dysfunction, and a range of profound symptoms. Lack of human tissue across the peripheral nerve tumors common in NF1 has been a major limitation in the development of new therapies. To address this unmet need, we have created an annotated collection of patient tumor samples, patient-derived cell lines, and patient-derived xenografts, and carried out high-throughput genomic and transcriptomic characterization to serve as a resource for further biologic and preclinical therapeutic studies. In this work, we release genomic and transcriptomic datasets comprised of 55 tumor samples derived from 23 individuals, complete with clinical annotation. All data are publicly available through the NF Data Portal and at http://synapse.org/jhubiobank.


Subject(s)
Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/pathology , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Cell Line, Tumor , Genomics , High-Throughput Nucleotide Sequencing , Humans , Transcriptome
17.
Clin Hemorheol Microcirc ; 75(2): 135-142, 2020.
Article in English | MEDLINE | ID: mdl-31903986

ABSTRACT

BACKGROUND: Damage-associated molecular patterns (DAMPs) generated by major surgery can induce global inflammation response and may degrade the vascular endothelial glycocalyx layer (EGL); in turn, the resulting EGL fragments can act as DAMPs, in a destructive positive feedback loop, to promote exacerbation of inflammation. Ulinastatin (UTI) may attenuate EGL shedding by inhibiting serine proteases and hyaluronidase. OBJECTIVE: This trail evaluates whether EGL shedding elicited by Traditional Whipple Procedure (TWP) could be decreased by using UTI. METHODS: We divided 60 patients undergoing TWP into a control group and a UTI group (n = 30 for both). Blood samples were collected before (T0), near the end (T1), and 1 hour after (T2) surgery. Levels of syndecan-1, ICAM-1, VCAM-1, IL-6, C-reactive protein, thrombomodulin, Hbg and serum albumin were measured and plasma albumin leakage was estimated. RESULTS: IL-6 levels significantly elevated at T1 and T2 in the control group compared with T0, but not the UTI group. Syndecan-1 levels significantly elevated at T1 and T2 in the control group but only T2 in the UTI group compared with T0. CONCLUSIONS: We found global inflammation reaction and EGL degradation during TWP. Perioperative UTI treatment can attenuate this EGL shedding and might alleviate plasma albumin leakage.


Subject(s)
Glycocalyx/drug effects , Glycoproteins/therapeutic use , Pancreatic Neoplasms/surgery , Perioperative Care/methods , Trypsin Inhibitors/therapeutic use , Female , Glycoproteins/pharmacology , Humans , Male , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Trypsin Inhibitors/pharmacology
18.
Life Sci ; 247: 116942, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-31715185

ABSTRACT

AIMS: The cardioprotective effects of preconditioning against ischemia-reperfusion (I/R) injury depend on the structural integrity of membrane caveolae and signaling through G protein-coupled receptors (GPCRs). However, the mechanisms underlying opioid preconditioning are not fully understood. Here, we examined whether caveolins transmitted opioid-GPCR signals to the mitochondria to mediate cardioprotection. MAIN METHODS: Mice were treated with pertussis toxin (PTX) or saline. Thirty-six hours later, mice from each group were randomly assigned to receive the δ-opioid receptor agonist SNC-121 or saline intraperitoneally 15 min before in vivo I/R. Infarct sizes in each group were compared, and immunoblot analysis was used to detect caveolin expression. The structures of caveolae and mitochondria were determined by electron microscopy (EM). The opening degree of the mitochondrial permeability transition pore (mPTP) was assessed by colorimetry, and mitochondrial respiratory function was assessed by Oxygraph-2k. KEY FINDINGS: Treatment with an opioid receptor agonist reduced the myocardial infarct size after I/R injury, increased caveolin expression, decreased mitochondrial mPTP opening, and improved mitochondrial respiratory function. EM analysis revealed that opioids induced caveolae formation in myocytes and tended to promote translocation to mitochondria. However, these protective effects were blocked by PTX. SIGNIFICANCE: Opioid-induced preconditioning depended on Gi signaling, which promoted caveolin translocation to mitochondria, supported their functional integrity, and enhanced cardiac stress adaption. Verification of this pathway will establish new targets for opioid agents in the field of cardiac protection.


Subject(s)
Benzamides/pharmacology , Cardiotonic Agents/pharmacology , Caveolins/metabolism , Mitochondria, Heart/metabolism , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/metabolism , Animals , Caveolae/metabolism , Caveolae/ultrastructure , Male , Mice , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/ultrastructure , Mitochondrial Permeability Transition Pore , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Receptors, G-Protein-Coupled/metabolism
19.
PLoS One ; 14(6): e0217399, 2019.
Article in English | MEDLINE | ID: mdl-31158244

ABSTRACT

The clinical significance of BRAF alterations in well-differentiated (WD) metastatic pancreatic neuroendocrine tumor (panNET) is unknown, but BRAF-mutated panNET could represent a subset characterized by an identifiable and clinically actionable driver. Following the identification of two patients with WD metastatic panNET whose tumors harbored BRAF mutations, we queried the MSK-IMPACT series of 80 patients with WD metastatic panNET for additional mutations in BRAF, and in other genes involved in RAS/ RTK/ PI3K signaling pathways. BRAF mutations were identified in six samples (7.5%): two tumors harbored V600E mutations, one tumor each expressed K601E, T599K, and T310I mutations, and one tumor expressed both G596D and E451K BRAF. Few additional actionable driver alterations were identified. To determine the ERK activating capability of four BRAF mutations not previously characterized, mutant constructs were tested in model systems. Biochemical characterization of BRAF mutations revealed both high and low activity mutants. Engineered cells expressing BRAF K601E and V600E were used for in vitro drug testing of RAF and MEK inhibitors currently in clinical use. BRAF K601E demonstrated reduced sensitivity to dabrafenib compared to BRAF V600E, but the combination of RAF plus MEK inhibition was effective in cells expressing this mutation. Herein, we describe the clinical course of a patient with BRAF K601E and a patient with BRAF V600E WD metastatic panNET, and the identification of four mutations in BRAF not previously characterized. The combined clinical and biochemical data support a potential role for RAF and MEK inhibitors, or a combination of these, in a selected panNET population.


Subject(s)
Imidazoles/pharmacology , MAP Kinase Kinase Kinases , MAP Kinase Signaling System/drug effects , Mutation, Missense , Neuroendocrine Tumors , Oximes/pharmacology , Pancreatic Neoplasms , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf , Amino Acid Substitution , Animals , Cell Line, Tumor , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , NIH 3T3 Cells , Neoplasm Metastasis , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/enzymology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism
20.
Cancer Discov ; 8(9): 1130-1141, 2018 09.
Article in English | MEDLINE | ID: mdl-29880583

ABSTRACT

BRAFV600E hyperactivates ERK and signals as a RAF inhibitor-sensitive monomer. Although RAF inhibitors can produce impressive clinical responses in patients with mutant BRAF tumors, the mechanisms of resistance to these drugs are incompletely characterized. Here, we report a complete response followed by clinical progression in a patient with a BRAFV600E-mutant brain tumor treated with dabrafenib. Whole-exome sequencing revealed a secondary BRAFL514V mutation at progression that was not present in the pretreatment tumor. Expressing BRAFV600E/L514V induces ERK signaling, promotes RAF dimer formation, and is sufficient to confer resistance to dabrafenib. Newer RAF dimer inhibitors and an ERK inhibitor are effective against BRAFL514V-mediated resistance. Collectively, our results validate a novel biochemical mechanism of RAF inhibitor resistance mediated by a secondary mutation, emphasizing that, like driver mutations in cancer, the spectrum of mutations that drive resistance to targeted therapy are heterogeneous and perhaps emerge with a lineage-specific prevalence.Significance: In contrast to receptor tyrosine kinases, in which secondary mutations are often responsible for acquired resistance, second-site mutations in BRAF have not been validated in clinically acquired resistance to RAF inhibitors. We demonstrate a secondary mutation in BRAF (V600E/L514V) following progression on dabrafenib and confirm functionally that this mutation is responsible for resistance. Cancer Discov; 8(9); 1130-41. ©2018 AACR.See related commentary by Romano and Kwong, p. 1064This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
Brain Neoplasms/genetics , Drug Resistance, Neoplasm , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Brain Neoplasms/drug therapy , Disease Progression , Humans , Imidazoles/therapeutic use , Male , Oximes/therapeutic use , Protein Multimerization/drug effects , Proto-Oncogene Proteins B-raf/chemistry , Exome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...