Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cell Metab ; 35(12): 2119-2135.e5, 2023 12 05.
Article in English | MEDLINE | ID: mdl-37913768

ABSTRACT

The rising pancreatic cancer incidence due to obesity and type 2 diabetes is closely tied to hyperinsulinemia, an independent cancer risk factor. Previous studies demonstrated reducing insulin production suppressed pancreatic intraepithelial neoplasia (PanIN) pre-cancerous lesions in Kras-mutant mice. However, the pathophysiological and molecular mechanisms remained unknown, and in particular it was unclear whether hyperinsulinemia affected PanIN precursor cells directly or indirectly. Here, we demonstrate that insulin receptors (Insr) in KrasG12D-expressing pancreatic acinar cells are dispensable for glucose homeostasis but necessary for hyperinsulinemia-driven PanIN formation in the context of diet-induced hyperinsulinemia and obesity. Mechanistically, this was attributed to amplified digestive enzyme protein translation, triggering of local inflammation, and PanIN metaplasia in vivo. In vitro, insulin dose-dependently increased acinar-to-ductal metaplasia formation in a trypsin- and Insr-dependent manner. Collectively, our data shed light on the mechanisms connecting obesity-driven hyperinsulinemia and pancreatic cancer development.


Subject(s)
Carcinoma in Situ , Diabetes Mellitus, Type 2 , Hyperinsulinism , Insulins , Pancreatic Neoplasms , Mice , Animals , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, Insulin/metabolism , Diabetes Mellitus, Type 2/metabolism , Pancreatic Neoplasms/metabolism , Acinar Cells/metabolism , Acinar Cells/pathology , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Inflammation/metabolism , Hyperinsulinism/complications , Metaplasia/metabolism , Metaplasia/pathology , Obesity/metabolism , Insulins/metabolism
2.
J Ayurveda Integr Med ; 14(6): 100806, 2023.
Article in English | MEDLINE | ID: mdl-37857033

ABSTRACT

BACKGROUND: Prediabetes is an intermediate state of hyperglycemia, which acts as a precursor to Diabetes mellitus if left untreated. Nisha (Curcuma longa) and Amalaki (Emblica officinalis) combination has been advocated as drugs of choice to treat the early manifestations of Diabetes mellitus. OBJECTIVE: This prospective, randomized, single-blind, placebo-controlled, comparative study was planned to assess the efficacy and safety of Nisha-Amalaki capsules in preventing progression to Diabetes mellitus in prediabetic patients when administered for 6 months. METHODS: The study was conducted on prediabetic participants randomized to receive either Nisha-Amalaki (500 mg) or placebo one capsule twice a day for six months. The effect of study medications on IDRS (Indian Diabetes Risk Score), BMI (Body Mass Index), blood sugar, serum insulin, HOMA-IR (Homeostasis Model Assessment-Estimated Insulin Resistance), HbA1c (glycated hemoglobin), oxidative markers, Ayurvedic symptoms and Quality of Life (QoL) scores was assessed at regular intervals. RESULTS: 58 of the 62 participants enrolled completed the study. Significant fall in IDRS score [p < 0.001], BMI [p < 0.001], fasting, and 2 h post-OGTT sugar, insulin, HbA1c, HOMA-IR, and oxidative stress markers [p < 0.001] was observed in patients receiving Nisha-Amalaki at 6 months. Ayurvedic symptoms and QoL scores also improved at 6 months in the treatment group. CONCLUSION: Treatment with Nisha-Amalaki capsules improved all study parameters including insulin sensitivity at 6 months as compared to placebo in prediabetic patients. Thus Nisha-Amalaki should be considered as prophylactic therapy in prediabetics to delay progression to diabetes.

4.
Sci Data ; 10(1): 70, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36737439

ABSTRACT

We introduce Cháksu-a retinal fundus image database for the evaluation of computer-assisted glaucoma prescreening techniques. The database contains 1345 color fundus images acquired using three brands of commercially available fundus cameras. Each image is provided with the outlines for the optic disc (OD) and optic cup (OC) using smooth closed contours and a decision of normal versus glaucomatous by five expert ophthalmologists. In addition, segmentation ground-truths of the OD and OC are provided by fusing the expert annotations using the mean, median, majority, and Simultaneous Truth and Performance Level Estimation (STAPLE) algorithm. The performance indices show that the ground-truth agreement with the experts is the best with STAPLE algorithm, followed by majority, median, and mean. The vertical, horizontal, and area cup-to-disc ratios are provided based on the expert annotations. Image-wise glaucoma decisions are also provided based on majority voting among the experts. Cháksu is the largest Indian-ethnicity-specific fundus image database with expert annotations and would aid in the development of artificial intelligence based glaucoma diagnostics.


Subject(s)
Glaucoma , Optic Disk , Humans , Algorithms , Artificial Intelligence , Fundus Oculi , Glaucoma/diagnostic imaging , Optic Disk/diagnostic imaging
5.
Cell Mol Gastroenterol Hepatol ; 15(5): 1219-1246, 2023.
Article in English | MEDLINE | ID: mdl-36758798

ABSTRACT

BACKGROUND & AIMS: Loss of AT-rich interactive domain-containing protein 1A (ARID1A) fosters acinar-to-ductal metaplasia (ADM) and pancreatic carcinogenesis by down-regulating transcription programs controlling acinar cell identity. However, how ARID1A reacts to metaplasia-triggering environmental cues remains elusive. Here, we aimed to elucidate the role of ARID1A in controlling ductal pancreatic gene signatures and deciphering hierarchical signaling cues determining ARID1A-dependent chromatin regulation during acinar cell reprogramming. METHODS: Acinar cell explants with differential ARID1A status were subjected to genome-wide expression analyses. The impact of epidermal growth factor receptor (EGFR) signaling, NFATc1 activity, and ARID1A status on acinar reprogramming processes were characterized by ex vivo ADM assays and transgenic mouse models. EGFR-dependent ARID1A chromatin binding was studied by chromatin immunoprecipitation sequencing analysis and cellular fractionation. RESULTS: EGFR signaling interferes with ARID1A-dependent transcription by inducing genome-wide ARID1A displacement, thereby phenocopying ARID1A loss-of-function mutations and inducing a shift toward ADM permissive ductal transcription programs. Moreover, we show that EGFR signaling is required to push ARID1A-deficient acinar cells toward a metaplastic phenotype. Mechanistically, we identified the transcription factor nuclear factor of activated T cells 1 (NFATc1) as the central regulatory hub mediating both EGFR signaling-induced genomic ARID1A displacement and the induction of ADM-promoting gene signatures in the absence of ARID1A. Consequently, pharmacologic inhibition of NFATc1 or its depletion in transgenic mice not only preserves genome-wide ARID1A occupancy, but also attenuates acinar metaplasia led by ARID1A loss. CONCLUSIONS: Our data describe an intimate relationship between environmental signaling and chromatin remodeling in orchestrating cell fate decisions in the pancreas, and illustrate how ARID1A loss influences transcriptional regulation in acinar cell reprogramming.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Acinar Cells/metabolism , Chromatin , Pancreatic Neoplasms/genetics , Carcinoma, Pancreatic Ductal/genetics , Cellular Reprogramming , Transcription Factors/genetics , ErbB Receptors/genetics , Mice, Transgenic , Metaplasia , DNA-Binding Proteins/genetics , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism
6.
Cancers (Basel) ; 14(14)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35884510

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDAC) represents a lethal malignancy with a consistently poor outcome. Besides mutations in PDAC driver genes, the aggressive tumor biology of the disease and its remarkable therapy resistance are predominantly installed by potentially reversible epigenetic dysregulation. However, epigenetic regulators act in a context-dependent manner with opposing implication on tumor progression, thus critically determining the therapeutic efficacy of epigenetic targeting. Herein, we aimed at exploring the molecular prerequisites and underlying mechanisms of oncogenic Enhancer of Zeste Homolog 2 (EZH2) activity in PDAC progression. Preclinical studies in EZH2 proficient and deficient transgenic and orthotopic in vivo PDAC models and transcriptome analysis identified the TP53 status as a pivotal context-defining molecular cue determining oncogenic EZH2 activity in PDAC. Importantly, the induction of pro-apoptotic gene signatures and processes as well as a favorable PDAC prognosis upon EZH2 depletion were restricted to p53 wildtype (wt) PDAC subtypes. Mechanistically, we illustrate that EZH2 blockade de-represses CDKN2A transcription for the subsequent posttranslational stabilization of p53wt expression and function. Together, our findings suggest an intact CDKN2A-p53wt axis as a prerequisite for the anti-tumorigenic consequences of EZH2 depletion and emphasize the significance of molecular stratification for the successful implementation of epigenetic targeting in PDAC.

7.
Visc Med ; 38(1): 4-10, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35295896

ABSTRACT

Background: Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease that has no effective early detection method or treatment to date. Summary: The normal cell type that initiates PDAC, or its cellular origin, is still unknown. To investigate the contribution of distinct normal epithelial cell types to PDAC tumorigenesis, genetically engineered mouse models were used to show that both acinar and ductal cells are capable of giving rise to PDAC. These studies indicated that genetic mutations and pancreatic injury interact differently with each cellular origin to affect their predilection and process for forming PDAC. In this review, we summarize recent findings using various genetically engineered mouse models in the identification and characterization of the PDAC cell of origin. We also discuss potential implications for cellular origin on tumor development, PDAC transcriptional subtype, and disease prognosis of patients. Key Message: Although it is clear that both ductal and acinar cells have the potential to form PDAC, whether cellular origin can indeed influence patient prognosis and whether knowledge of cellular origin will aid in the diagnosis or treatment of patients in the future will need further study.

8.
Aging (Albany NY) ; 14(3): 1068-1086, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35158337

ABSTRACT

Radiation therapy is a commonly used treatment modality for cancer. Although effective in providing local tumor control, radiation causes oxidative stress, inflammation, immunomodulatory and mitogenic cytokine production, extracellular matrix production, and premature senescence in lung parenchyma. The senescence associated secretory phenotype (SASP) can promote inflammation and stimulate alterations in the surrounding tissue. Therefore, we hypothesized that radiation-induced senescent parenchymal cells in irradiated lung would enhance tumor growth. Using a murine syngeneic tumor model of melanoma and non-small cell lung cancer lung metastasis, we demonstrate that radiation causes a significant increase in markers of premature senescence in lung parenchyma within 4 to 8 weeks. Further, injection of B16F0 (melanoma) or Lewis Lung carcinoma (epidermoid lung cancer) cells at these time points after radiation results in an increase in the number and size of pulmonary tumor nodules relative to unirradiated mice. Treatment of irradiated mice with a senolytic agent (ABT-737) or agents that prevent senescence (rapamycin, INK-128) was sufficient to reduce radiation-induced lung parenchymal senescence and to mitigate radiation-enhanced tumor growth. These agents abrogated radiation-induced expression of 12-Lipoxygenase (12-LOX), a molecule implicated in several deleterious effects of senescence. Deficiency of 12-LOX prevented radiation-enhanced tumor growth. Together, these data demonstrate the pro-tumorigenic role of radiation-induced senescence, introduces the dual TORC inhibitor INK-128 as an effective agent for prevention of radiation-induced normal tissue senescence, and identifies senescence-associated 12-LOX activity as an important component of the pro-tumorigenic irradiated tissue microenvironment. These studies suggest that combining senotherapeutic agents with radiotherapy may decrease post-therapy tumor growth.


Subject(s)
Carcinoma, Lewis Lung , Lung Neoplasms , Melanoma, Experimental , Animals , Arachidonate 12-Lipoxygenase/pharmacology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Cell Growth Processes , Cellular Senescence , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Tumor Microenvironment
9.
Cells ; 10(12)2021 12 08.
Article in English | MEDLINE | ID: mdl-34943970

ABSTRACT

BACKGROUND: The Nuclear Factor of Activated T-cells 1 (NFATc1) transcription factor and the methyltransferase Enhancer of Zeste Homolog 2 (EZH2) significantly contribute to the aggressive phenotype of pancreatic ductal adenocarcinoma (PDAC). Herein, we aimed at dissecting the mechanistic background of their interplay in PDAC progression. METHODS: NFATc1 and EZH2 mRNA and protein expression and complex formation were determined in transgenic PDAC models and human PDAC specimens. NFATc1 binding on the Ezh2 gene and the consequences of perturbed NFATc1 expression on Ezh2 transcription were explored by Chromatin Immunoprecipitation (ChIP) and upon transgenic or siRNA-mediated interference with NFATc1 expression, respectively. Integrative analyses of RNA- and ChIP-seq data was performed to explore NFATc1-/EZH2-dependent gene signatures. RESULTS: NFATc1 targets the Ezh2 gene for transcriptional activation and biochemically interacts with the methyltransferase in murine and human PDAC. Surprisingly, our genome-wide binding and expression analyses do not link the protein complex to joint gene regulation. In contrast, our findings provide evidence for chromatin-independent functions of the NFATc1:EZH2 complex and reveal posttranslational EZH2 phosphorylation at serine 21 as a prerequisite for robust complex formation. CONCLUSION: Our findings disclose a previously unknown NFATc1-EZH2 axis operational in the pancreas and provide mechanistic insights into the conditions fostering NFATc1:EZH2 complex formation in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , NFATC Transcription Factors/genetics , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/genetics , Chromatin/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Mice , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Small Interfering/genetics , Trans-Activators/genetics
10.
Cancers (Basel) ; 13(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34944784

ABSTRACT

To improve the treatment of pancreatic ductal adenocarcinoma (PDAC), a promising strategy consists of personalized chemotherapy based on gene expression profiles. Investigating a panel of PDAC-derived human cell lines, we found that their sensitivities towards cisplatin fall in two distinct classes. The platinum-sensitive class is characterized by the expression of GATA6, miRNA-200a, and miRNA-200b, which might be developable as predictive biomarkers. In the case of resistant PDAC cells, we identified a synergism of cisplatin with HSP90 inhibitors. Mechanistic explanations of this synergy include the degradation of Fanconi anemia pathway factors upon HSP90 inhibition. Treatment with the drug combination resulted in increased DNA damage and chromosome fragmentation, as we have reported previously for ovarian cancer cells. On top of this, HSP90 inhibition also enhanced the accumulation of DNA-bound platinum. We next investigated an orthotopic syngeneic animal model consisting of tumors arising from KPC cells (LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre, C57/BL6 genetic background). Here again, when treating established tumors, the combination of cisplatin with the HSP90 inhibitor onalespib was highly effective and almost completely prevented further tumor growth. We propose that the combination of platinum drugs and HSP90 inhibitors might be worth testing in the clinics for the treatment of cisplatin-resistant PDACs.

11.
Anesth Pain Med ; 11(3): e117197, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34540647

ABSTRACT

Adjuvant drugs for peripheral nerve blocks are a promising solution to acute postoperative pain and the transition to chronic pain treatment. Peripheral nerve blocks (PNB) are used in the brachial plexus, lumbar plexus, femoral nerve, sciatic nerve, and many other anatomic locations for site-specific pain relief. However, the duration of action of a PNB is limited without an adjuvant drug. The use of non-opioid adjuvant drugs for single-shot peripheral nerve blocks (sPNB), such as alpha-2 agonists, dexamethasone, midazolam, and non-steroidal anti-inflammatory drugs, can extend the duration of local anesthetics and reduce the dose-dependent adverse effects of local anesthetics. Tramadol is a weak opioid that acts as a central analgesic. It can block voltage-dependent sodium and potassium channels, cause serotonin release, and inhibit norepinephrine reuptake and can also be used as an adjuvant in PNBs. However, tramadol's effectiveness and safety as an adjuvant to local anesthetic for PNB are inconsistent. The effects of the adjuvants on neurotoxicity must be further evaluated with further studies to delineate the safety in their use in PNB. Further research needs to be done. However, the use of adjuvants in PNB can be a way to help control postoperative pain.

12.
Curr Pain Headache Rep ; 24(11): 73, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33098008

ABSTRACT

PURPOSE OF REVIEW: Multimodal pain management is the most effective way to treat postsurgical pain. However, the use of opioids for acute pain management has unfortunately been a significant contributor to the current opioid epidemic. The use of opioids should be limited and only considered a "rescue" pain medication after other modalities of pain management have been utilized. RECENT FINDINGS: It may be difficult to curtail the use of opioids in the treatment of chronic pain; however, in the postsurgical setting, there is compelling evidence that an opioid-centric analgesic approach is not necessary for good patient outcomes and healthcare cost benefits. Opioid-related adverse effects are the leading cause of preventable harm in the hospital setting. After the realization in recent years of the many harmful effects of opioids, alternative regimens including the use of multimodal analgesia have become a standard practice in acute pain management. Exparel, a long-lasting liposomal bupivacaine local anesthetic agent, has many significant benefits in the management of postoperative pain. Overall, the literature suggests that Exparel may be a significant component for postoperative multimodal pain control owing to its efficacy and long duration of action.


Subject(s)
Anesthetics, Local/administration & dosage , Bupivacaine/administration & dosage , Pain Management/methods , Pain, Postoperative/drug therapy , Delayed-Action Preparations/administration & dosage , Humans , Liposomes
13.
Cancer Res ; 80(21): 4620-4632, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32907838

ABSTRACT

Recent studies have thoroughly described genome-wide expression patterns defining molecular subtypes of pancreatic ductal adenocarcinoma (PDAC), with different prognostic and predictive implications. Although the reversible nature of key regulatory transcription circuits defining the two extreme PDAC subtype lineages "classical" and "basal-like" suggests that subtype states are not permanently encoded but underlie a certain degree of plasticity, pharmacologically actionable drivers of PDAC subtype identity remain elusive. Here, we characterized the mechanistic and functional implications of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) in controlling PDAC plasticity, dedifferentiation, and molecular subtype identity. Utilization of transgenic PDAC models and human PDAC samples linked EZH2 activity to PDAC dedifferentiation and tumor progression. Combined RNA- and chromatin immunoprecipitation sequencing studies identified EZH2 as a pivotal suppressor of differentiation programs in PDAC and revealed EZH2-dependent transcriptional repression of the classical subtype defining transcription factor Gata6 as a mechanistic basis for EZH2-dependent PDAC progression. Importantly, genetic or pharmacologic depletion of EZH2 sufficiently increased GATA6 expression, thus inducing a gene signature shift in favor of a less aggressive and more therapy-susceptible, classical PDAC subtype state. Consistently, abrogation of GATA6 expression in EZH2-deficient PDAC cells counteracted the acquisition of classical gene signatures and rescued their invasive capacities, suggesting that GATA6 derepression is critical to overcome PDAC progression in the context of EZH2 inhibition. Together, our findings link the EZH2-GATA6 axis to PDAC subtype identity and uncover EZH2 inhibition as an appealing strategy to induce subtype-switching in favor of a less aggressive PDAC phenotype. SIGNIFICANCE: This study highlights the role of EZH2 in PDAC progression and molecular subtype identity and suggests EZH2 inhibition as a strategy to recalibrate GATA6 expression in favor of a less aggressive disease. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/21/4620/F1.large.jpg.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , GATA6 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic/physiology , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Disease Progression , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/metabolism
14.
Hum Vaccin Immunother ; 16(8): 1937-1944, 2020 08 02.
Article in English | MEDLINE | ID: mdl-31977268

ABSTRACT

Two vaccines, 23-valent pneumococcal polysaccharide vaccine (PPSV23) and 13-valent pneumococcal conjugate vaccine (PCV13), are widely available for the prevention of pneumococcal disease in adults. However, it is unclear how cost-effective these pneumococcal vaccine choices are in the Hong Kong healthcare environment. We aimed to assess the cost-effectiveness of a sequential administration of PCV13 followed by PPSV23 compared to a single dose of PPSV23 vaccination for pneumococcal disease control in Hong Kong adults aged ≥65 years and individuals aged 20-64 years with immunocompromising and chronic conditions. A previously developed deterministic cohort sequential model was applied to compare the outcomes of two vaccination strategies from a societal perspective. Population-specific model input, including incidence, mortality, case-fatality, risk group distribution, vaccination costs, disease management, and productivity loss, was estimated from a Hong Kong-wide electronic medical database. Costs were valued in US$ in 2017. Vaccination strategies with an incremental cost-effectiveness ratio (ICER, defined as incremental cost per QALY saved) less than one local GDP per capita ($46,193 in 2017) were defined as highly cost-effective. Deterministic sensitivity analyses (SA) were conducted. Compared with single-dose PPSV23, sequential vaccination of PCV13 followed by PPSV23 was cost-saving for adults aged ≥20 years. In the deterministic SA, the base-case results were robust for tested parameter uncertainties. Future vaccination policies should consider the cost-effectiveness of a sequential vaccination strategy as a measure to reduce the vaccine-preventable pneumococcal disease burden in Hong Kong.


Subject(s)
Pneumococcal Infections , Pneumonia, Pneumococcal , Adult , Cost-Benefit Analysis , Hong Kong/epidemiology , Humans , Pneumococcal Infections/epidemiology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines , Vaccination , Vaccines, Conjugate
15.
Iran J Pathol ; 14(4): 317-321, 2019.
Article in English | MEDLINE | ID: mdl-31754362

ABSTRACT

BACKGROUND & OBJECTIVE: Colorectal cancer is the third most prevalent malignancy with high mortality rate, necessitating markers that predict survival and guide the treatment. Previous studies have examined the immunohistochemical expression of Bcl-2, an apoptotic marker, in colorectal carcinoma, but results have been contradictory. To evaluate the histopathological features of colorectal carcinoma, immunohistochemical expression of Bcl-2 must be analyzed to find out statistical association of Bcl-2 expression with certain prognostic factors histopathologic type, grade and TNM staging. METHODS: This prospective study was conducted on the colectomy specimens of colorectal carcinoma, over a period of two years. The tumor morphology and Bcl-2 status were evaluated by immunohistochemistry in each case. RESULTS: The study included 58 cases, with mean patient age of 47.07 years and male: female ratio of 1.89:1. Bcl-2 positivity was seen in 32.7% of the cases. Weak, moderate, and strong expression of Bcl-2 was seen in 12.1%, 12.1%, and 8.5% of cases respectively. Even though early stages of colorectal carcinoma showed greater frequency of Bcl-2 expression than advanced stages (36.3% versus 28%), however this association was not statistically significant. CONCLUSION: Lack of statistically significant correlation between Bcl-2 immuno-histochemical expression and prognostic parameters like tumor grade and stage, suggests that Bcl-2 immunoexpression may not be a significant prognostic marker in colorectal carcinoma.

16.
Radiat Res ; 192(4): 367-379, 2019 08.
Article in English | MEDLINE | ID: mdl-31373871

ABSTRACT

Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive complication of therapeutic irradiation of the thorax. It has been suggested that senescence of type II pneumocytes (AECIIs), an alveolar stem cell, plays a role in the development of RIPF through loss of replicative reserve and via senescent AECII-driven release of proinflammatory and profibrotic cytokines. Within this context, we hypothesized that arachidonate 12-lipoxygenase (12-LOX) is a critical mediator of AECII senescence and RIPF. Treatment of wild-type AECIIs with 12S-hydroxyeicosateraenoic acid (12S-HETE), a downstream product of 12-LOX, was sufficient to induce senescence in a NADPH oxidase 4 (NOX4)-dependent manner. Mice deficient in 12-LOX exhibited reduced AECII senescence, pulmonary collagen accumulation and accumulation of alternatively activated (M2) macrophages after thoracic irradiation (5 × 6 Gy) compared to wild-type mice. Conditioned media from irradiated or 12S-HETE-treated primary pneumocytes contained elevated levels of IL-4 and IL-13 compared to untreated pneumocytes. Primary macrophages treated with conditioned media from irradiated AECII demonstrated preferential M2 type polarization when AECIIs were derived from wild-type mice compared to 12-LOX-deficient mice. Together, these data identified 12-LOX as a critical component of RIPF and a therapeutic target for radiation-induced lung injury.


Subject(s)
Alveolar Epithelial Cells/pathology , Arachidonate 12-Lipoxygenase/metabolism , Cellular Senescence/radiation effects , Macrophages/radiation effects , Radiation Pneumonitis/enzymology , Alveolar Epithelial Cells/radiation effects , Animals , Arachidonate 12-Lipoxygenase/genetics , Female , Gene Expression Regulation, Enzymologic/radiation effects , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Radiation Pneumonitis/genetics , Radiation Pneumonitis/immunology , Radiation Pneumonitis/pathology
17.
Curr Pain Headache Rep ; 23(9): 67, 2019 Jul 29.
Article in English | MEDLINE | ID: mdl-31359193

ABSTRACT

PURPOSE OF REVIEW: Anticoagulant use among patients is prevalent and increasing. It is important for anesthesiologists to be aware of patients on anticoagulants while performing regional anesthesia. RECENT FINDINGS: In recent years, the FDA has approved many new anticoagulants. With new drugs coming to the market, new side effect profiles should be considered when treating patients, especially when using regional anesthesia. Both ASRA and European agencies have laid out recommendations regarding anticoagulant use and neuraxial/regional techniques. Regarding newer anticoagulants, the guidelines for discontinuation prior to neuraxial injection are based on pharmacokinetics, including half-life duration for each drug. While each clinical scenario requires an individualized approach, general guidelines can serve as a starting point to help with anesthetic planning and potentially improve patient safety in this evolving field.


Subject(s)
Anesthesia, Conduction/methods , Anticoagulants/administration & dosage , Evidence-Based Medicine/methods , Administration, Oral , Anesthesia, Conduction/adverse effects , Anticoagulants/adverse effects , Evidence-Based Medicine/trends , Hemorrhage/chemically induced , Hemorrhage/diagnosis , Hemorrhage/prevention & control , Humans , Risk Factors
18.
Cells ; 8(6)2019 06 09.
Article in English | MEDLINE | ID: mdl-31181844

ABSTRACT

Background: Pancreatic ductal adenocarcinoma (PDAC) is highly resistant to standard chemo- and radiotherapy. Recently, a new class of non-platinum-based halogenated molecules (called FMD compounds) was discovered that selectively kills cancer cells. Here, we investigate the potential of 1,2-Diamino-4,5-dibromobenzene (2Br-DAB) in combination with standard chemotherapy and radiotherapy in murine and human PDAC. Methods: Cell viability and colony formation was performed in human (Panc1, BxPC3, PaTu8988t, MiaPaCa) and three murine LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre (KPC) pancreatic cancer cell lines. In vivo, preclinical experiments were conducted in LSL-KrasG12D/+;p48-Cre (KC) and KPC mice using 2Br-DAB (7 mg/kg, i.p.), +/- radiation (10 × 1.8 Gy), gemcitabine (100 mg/kg, i.p.), or a combination. Tumor growth and therapeutic response were assessed by high-resolution ultrasound and immunohistochemistry. Results: 2Br-DAB significantly reduced cell viability in human and murine pancreatic cancer cell lines in a dose-dependent manner. In particular, colony formation in human Panc1 cells was significantly decreased upon 25 µM 2Br-DAB + radiation treatment compared with vehicle control (p = 0.03). In vivo, 2Br-DAB reduced tumor frequency in KC mice. In the KPC model, 2Br-DAB or gemcitabine monotherapy had comparable therapeutic effects. Furthermore, the combination of gemcitabine and 2Br-DAB or 2Br-DAB and 18 Gy irradiation showed additional antineoplastic effects. Conclusions: 2Br-DAB is effective in killing pancreatic cancer cells in vitro. 2Br-DAB was not toxic in vivo, and additional antineoplastic effects were observed in combination with irradiation.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzene Derivatives/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Benzene Derivatives/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/radiotherapy , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Cisplatin/therapeutic use , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Gamma Rays , Genetic Engineering , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy
19.
Ann Pediatr Cardiol ; 12(2): 135-137, 2019.
Article in English | MEDLINE | ID: mdl-31143039

ABSTRACT

We report two cases of agenesis of the right pulmonary veins (PVs) associated with sequestration of the right lung with systemic to pulmonary collateral. Both the children were referred for evaluation of recurrent lower respiratory tract infections. Transthoracic echocardiographic evaluation showed reversal of flow in the right pulmonary artery (PA) and absent right PVs. Cardiac catheterization confirmed the diagnosis of agenesis of the right PVs. Transcatheter closure of collaterals was performed in view of significant shunt from systemic to PA. Both the patients are asymptomatic on follow-up.

20.
EBioMedicine ; 40: 394-405, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30709769

ABSTRACT

BACKGROUND: Cytosolic 5'-nucleotidase 1A (NT5C1A) dephosphorylates non-cyclic nucleoside monophosphates to produce nucleosides and inorganic phosphates. Here, we investigate NT5C1A expression in pancreatic ductal adenocarcinoma (PDAC) and its impact on gemcitabine metabolism and therapeutic efficacy. METHODS: NT5C1A expression was determined by semiquantitative immunohistochemistry using tissue microarrays. Gemcitabine metabolites and response were assessed in several human and murine PDAC cell lines using crystal violet assays, Western blot, viability assays, and liquid chromatography tandem mass-spectrometry (LC-MS/MS). FINDINGS: NT5C1A was strongly expressed in tumor cells of a large subgroup of resected PDAC patients in two independent patient cohorts (44-56% score 2 and 8-26% score 3, n = 414). In contrast, NT5C1A was expressed at very low levels in the tumor stroma, and neither stromal nor tumoral expression was a prognostic marker for postoperative survival. In vitro, NT5C1A overexpression increased gemcitabine resistance by reducing apoptosis levels and significantly decreased intracellular amounts of cytotoxic dFdCTP in +NT5C1A tumor cells. Co-culture experiments with conditioned media from +NT5C1A PSCs improved gemcitabine efficacy in tumor cells. In vivo, therapeutic efficacy of gemcitabine was significantly decreased and serum levels of the inactive gemcitabine metabolite dFdU significantly increased in mice bearing NT5C1A overexpressing tumors. INTERPRETATION: NT5C1A is robustly expressed in tumor cells of resected PDAC patients. Moreover, NT5C1A mediates gemcitabine resistance by decreasing the amount of intracellular dFdCTP, leading to reduced tumor cell apoptosis and larger pancreatic tumors in mice. Further studies should clarify the role of NT5C1A as novel predictor for gemcitabine treatment response in patients with PDAC.


Subject(s)
5'-Nucleotidase/genetics , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Expression , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Animals , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Models, Biological , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Prognosis , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...