Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Proc Natl Acad Sci U S A ; 116(25): 12442-12451, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31147469

ABSTRACT

Tumor-associated macrophages contribute to tumor progression and therapeutic resistance in breast cancer. Within the tumor microenvironment, tumor-derived factors activate pathways that modulate macrophage function. Using in vitro and in vivo models, we find that tumor-derived factors induce activation of the Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway in macrophages. We also demonstrate that loss of STAT3 in myeloid cells leads to enhanced mammary tumorigenesis. Further studies show that macrophages contribute to resistance of mammary tumors to the JAK/STAT inhibitor ruxolitinib in vivo and that ruxolitinib-treated macrophages produce soluble factors that promote resistance of tumor cells to JAK inhibition in vitro. Finally, we demonstrate that STAT3 deletion and JAK/STAT inhibition in macrophages increases expression of the protumorigenic factor cyclooxygenase-2 (COX-2), and that COX-2 inhibition enhances responsiveness of tumors to ruxolitinib. These findings define a mechanism through which macrophages promote therapeutic resistance and highlight the importance of understanding the impact of targeted therapies on the tumor microenvironment.


Subject(s)
Carcinogenesis , Janus Kinase Inhibitors/pharmacology , Macrophages/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Macrophages/enzymology , Mice , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Tumor Microenvironment
2.
Undersea Hyperb Med ; 49(3): 329-331, 2022.
Article in English | MEDLINE | ID: mdl-36001565

ABSTRACT

Carbon monoxide (CO) inhalation is a common method of suicide. The combination of formic acid with sulfuric acid creates carbon monoxide. This novel method is described in readily accessible internet-based resources. We present the case of a 35-year-old woman who developed CO toxicity by using this method. It is important for hyperbaric medicine physicians to be aware of this source of CO toxicity.


Subject(s)
Carbon Monoxide Poisoning , Hyperbaric Oxygenation , Physicians , Administration, Inhalation , Adult , Carbon Monoxide/toxicity , Carbon Monoxide Poisoning/therapy , Female , Humans , Suicide, Attempted
3.
Breast Cancer Res ; 23(1): 104, 2021 11 07.
Article in English | MEDLINE | ID: mdl-34743736

ABSTRACT

BACKGROUND: In breast cancer, complex interactions between tumor cells and cells within the surrounding stroma, such as macrophages, are critical for tumor growth, progression, and therapeutic response. Recent studies have highlighted the complex nature and heterogeneous populations of macrophages associated with both tumor-promoting and tumor-inhibiting phenotypes. Defining the pathways that drive macrophage function is important for understanding their complex phenotypes within the tumor microenvironment. Signal transducer and activator of transcription (STAT) transcription factors, such as STAT5, are key regulators of immune cell function. The studies described here investigate the functional contributions of STAT5 to tumor-associated macrophage function in breast cancer. METHODS: Initial studies were performed using a panel of human breast cancer and mouse mammary tumor cell lines to determine the ability of tumor cell-derived factors to induce STAT5 activation in macrophages. Further studies used these models to identify soluble factors that activate STAT5 in macrophages. To delineate STAT5-specific contributions to macrophage function, a conditional model of myeloid STAT5 deletion was used for in vitro, RNA-sequencing, and in vivo studies. The effects of STAT5 deletion in macrophages on tumor cell migration and metastasis were evaluated using in vitro co-culture migration assays and an in vivo tumor cell-macrophage co-injection model. RESULTS: We demonstrate here that STAT5 is robustly activated in macrophages by tumor cell-derived factors and that GM-CSF is a key cytokine stimulating this pathway. The analysis of RNA-seq studies reveals that STAT5 promotes expression of immune stimulatory genes in macrophages and that loss of STAT5 in macrophages results in increased expression of tissue remodeling factors. Finally, we demonstrate that loss of STAT5 in macrophages promotes tumor cell migration in vitro and mammary tumor metastasis in vivo. CONCLUSIONS: Breast cancer cells produce soluble factors, such as GM-CSF, that activate the STAT5 pathway in macrophages and drive expression of inflammatory factors. STAT5 deletion in myeloid cells enhances metastasis, suggesting that STAT5 activation in tumor-associated macrophages protects against tumor progression. Understanding mechanisms that drive macrophage function in the tumor microenvironment will ultimately lead to new approaches that suppress tumor-promoting functions while enhancing their anti-tumor functions.


Subject(s)
Breast Neoplasms/metabolism , STAT5 Transcription Factor/metabolism , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism , Adaptive Immunity/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Neoplasm Metastasis/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/immunology
4.
Acc Chem Res ; 51(3): 583-590, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29498267

ABSTRACT

Batteries are dynamic devices composed of multiple components that operate far from equilibrium and may operate under extreme stress and varying loads. Studies of isolated battery components are valuable to the fundamental understanding of the physical processes occurring within each constituent element. When the components are integrated into a full device and operated under realistic conditions, it can be difficult to decouple the physical processes that occur across multiple interfaces and multiple length scales. Thus, the physical processes studied in isolated components may change in a full battery setup or may be irrelevant to performance. Simulation studies on many length scales play a key role in the analysis of experiments and in the elucidation of the relevant physical processes impacting performance. In this Account, we aim to highlight the use of modeling on multiple length scales to identify rate limiting phenomena in lithium-ion batteries. To illustrate the utility of modeling, we examine lithium-ion batteries with nanostructured magnetite, Fe3O4, as the positive electrode active material against a solid Li0 negative electrode. Due to continuous operation away from equilibrium, batteries exhibit highly nonideal behavior, and a model that aims to reproduce behavior under realistic operating conditions must be able to capture the physics occurring on the length scales relevant to the performance of the system. It is our experience that limiting behavior in lithium-ion batteries can be observed on the atomic scale and up through the electrode scale and thus, predictive models must be capable of integrating and communicating physics across multiple length scales. Magnetite is studied as an electrode material for lithium-ion batteries, but it is found to suffer from slow solid-state transport of lithium, slow reaction kinetics, and poor cycling. Magnetite (Fe3O4) is a material capable of undergoing multiple electron transfers (MET), and can accept up to eight lithium per formula unit (Li8Fe3O4). Magnetite, (Fe8a3+)[Fe3+Fe2+]16dO4,32e2-, has a close-packed inverse spinel structure and undergoes intercalation, structural rearrangement, and conversion reactions upon full lithiation. (1) To overcome solid-state transport resistances, magnetite can be nanostructured to decrease Li+ diffusion lengths, and this has been shown to increase capacity. Additionally, unique architectures incorporating both carbon and Fe3O4 have shown to alleviate transport and cycling issues in the material. (2) Here, we solely address traditional composite electrodes, in which Fe3O4 is synthesized as nanoparticles and combined with additives to fabricate the electrode. In the case of nanoparticulate magnetite, it has been found that the electrode fabrication process results in the formation of micrometer-sized agglomerates of the Fe3O4 nanoparticles, introducing a secondary structural motif. The agglomerates may form in one or more fabrication processes, and their elimination may not be straightforward or warranted. Here, we highlight the impact of these secondary formations on the performance of the Fe3O4 lithium-ion battery. We illustrate how simulations can be used to design experiments, prioritize research efforts, and predict performance.

5.
Dev Biol ; 428(1): 232-244, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28606561

ABSTRACT

Macrophages are required for proper mammary gland development and maintaining tissue homeostasis. However, the mechanisms by which macrophages regulate this process remain unclear. Here, we identify STAT5 as an important regulator of macrophage function in the developing mammary gland. Analysis of mammary glands from mice with STAT5-deficient macrophages demonstrates delayed ductal elongation, enhanced ductal branching and increased epithelial proliferation. Further analysis reveals that STAT5 deletion in macrophages leads to enhanced expression of proliferative factors such as Cyp19a1/aromatase and IL-6. Mechanistic studies demonstrate that STAT5 binds directly to the Cyp19a1 promoter in macrophages to suppress gene expression and that loss of STAT5 results in enhanced stromal expression of aromatase. Finally, we demonstrate that STAT5 deletion in macrophages cooperates with oncogenic initiation in mammary epithelium to accelerate the formation of estrogen receptor (ER)-positive hyperplasias. These studies establish the importance of STAT5 in macrophages during ductal morphogenesis in the mammary gland and demonstrate that altering STAT5 function in macrophages can affect the development of tissue-specific disease.


Subject(s)
Macrophage Activation/genetics , Macrophages/cytology , Mammary Glands, Animal/embryology , Morphogenesis/genetics , Organogenesis/genetics , STAT5 Transcription Factor/genetics , Animals , Aromatase/biosynthesis , Aromatase/genetics , Cell Line , Cell Proliferation/genetics , Enzyme Activation/genetics , Epithelial Cells/cytology , Female , Gene Expression Regulation/genetics , Interleukin-6/biosynthesis , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Estrogen/metabolism
7.
Mediators Inflamm ; 2016: 4549676, 2016.
Article in English | MEDLINE | ID: mdl-26884646

ABSTRACT

Macrophages are critical mediators of inflammation and important regulators of developmental processes. As a key phagocytic cell type, macrophages evolved as part of the innate immune system to engulf and process cell debris and pathogens. Macrophages produce factors that act directly on their microenvironment and also bridge innate immune responses to the adaptive immune system. Resident macrophages are important for acting as sensors for tissue damage and maintaining tissue homeostasis. It is now well-established that macrophages are an integral component of the breast tumor microenvironment, where they contribute to tumor growth and progression, likely through many of the mechanisms that are utilized during normal wound healing responses. Because macrophages contribute to normal mammary gland development and breast cancer growth and progression, this review will discuss both resident mammary gland macrophages and tumor-associated macrophages with an emphasis on describing how macrophages interact with their surrounding environment during normal development and in the context of cancer.


Subject(s)
Breast Neoplasms/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mammary Glands, Human/metabolism , Animals , Breast Neoplasms/immunology , Female , Humans , Inflammation/immunology , Macrophages/immunology , Mammary Glands, Human/immunology
8.
Mol Cancer ; 14: 138, 2015 Jul 29.
Article in English | MEDLINE | ID: mdl-26215578

ABSTRACT

BACKGROUND: The epidermal growth factor (EGF) family of ligands has been implicated in promoting breast cancer initiation, growth and progression. The contributions of EGF family ligands and their receptors to breast cancer are complex, and the specific mechanisms through which different ligands regulate breast tumor initiation and growth are not well-defined. These studies focus on the EGF family member epiregulin (EREG) as a mediator of early stage breast tumorigenesis. METHODS: EREG expression levels were assessed in both cell lines and human samples of ductal carcinoma in situ (DCIS) using quantitative RT-PCR, ELISA and immunohistochemistry. Gene knock-down approaches using shRNA-based strategies were used to determine the requirement of EREG for growth of MCF10DCIS cells in vivo, and for identifying mechanisms through which EREG promotes tumor cell survival. Experiments were performed using a combination of two-dimensional culture, three-dimensional culture and tumor growth in vivo. RESULTS: In comparison with other EGF family members, EREG was induced in MCF10DCIS cells compared with MCF10A and MCF10AT cells and its expression was partially regulated by fibroblast growth factor receptor (FGFR) activity. Reduced EREG expression in MCF10DCIS cells led to decreased tumor growth in vivo, which was associated with reduced cell survival. Furthermore, treatment of MCF10A cells with exogenous EREG enhanced cell survival both in three-dimensional culture and in response to chemotherapeutic agents. Examination of EREG-induced signaling pathways demonstrated that EREG promoted survival of MCF10A cells through regulating expression of matrix metalloproteinase-1 (MMP-1). To determine the relevance of these findings in human tumors, samples of DCIS were analyzed for EREG and MMP-1 expression. EREG was induced in DCIS lesions compared to normal breast epithelium, and EREG and MMP-1 were correlated in a subset of DCIS samples. CONCLUSIONS: Together, these studies lead to identification of a novel pathway involving EREG and MMP-1 that contributes to the formation of early stage breast cancer. Understanding these complex pathways could ultimately lead to the development of novel biomarkers of neoplastic progression and/or new therapeutic strategies for patients with early stage cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Epiregulin/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Breast Neoplasms/pathology , Carcinogenesis/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Cell Line, Tumor , Epiregulin/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 1/genetics , Neoplasm Staging , Receptor, Fibroblast Growth Factor, Type 1/genetics
9.
Popul Health Manag ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38682441

ABSTRACT

The objective was to identify medical conditions associated with 30-day readmission, determine patient characteristics for which outpatient follow-up is most associated with reduced readmission, and evaluate how readmission risk changes with time to outpatient follow-up within a mobile integrated health-community paramedicine (MIH-CP) program. This retrospective observational study used data from 1,118 adult patient enrollments in a MIH-CP program operating in Baltimore, Maryland, from May 14, 2018, to December 21, 2021. Bivariate analysis identified chronic disease exacerbations associated with higher 30-day readmission risk. Kaplan-Meier curves and Cox proportional hazard regressions were used to measure how hazard of readmission changes with outpatient follow-up and how that association may vary with other factors. Receiver operating characteristic analysis was used to evaluate how well time to follow-up could predict 30-day readmission. Timely outpatient follow-up was associated with a significant reduction in hazard of readmission for patients aged 50 and younger and for patients with fewer than 5 social determinants of health needs identified. No significant association between readmission and specific chronic disease exacerbations was observed. An optimal follow-up time frame to reduce readmissions could not be identified. Timely outpatient follow-up may be effective for reducing readmissions in younger patients and patients who are less socially complex. Programs and policies aiming to reduce 30-day readmissions may have more success by expanding efforts to include these patients.

10.
Nat Commun ; 15(1): 363, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191471

ABSTRACT

In the complex tumor microenvironment (TME), mesenchymal cells are key players, yet their specific roles in prostate cancer (PCa) progression remain to be fully deciphered. This study employs single-cell RNA sequencing to delineate molecular changes in tumor stroma that influence PCa progression and metastasis. Analyzing mesenchymal cells from four genetically engineered mouse models (GEMMs) and correlating these findings with human tumors, we identify eight stromal cell populations with distinct transcriptional identities consistent across both species. Notably, stromal signatures in advanced mouse disease reflect those in human bone metastases, highlighting periostin's role in invasion and differentiation. From these insights, we derive a gene signature that predicts metastatic progression in localized disease beyond traditional Gleason scores. Our results illuminate the critical influence of stromal dynamics on PCa progression, suggesting new prognostic tools and therapeutic targets.


Subject(s)
Mesenchymal Stem Cells , Prostatic Neoplasms , Humans , Male , Animals , Mice , Prostatic Neoplasms/genetics , Prostate , Stromal Cells , Cell Differentiation , Tumor Microenvironment/genetics
11.
JBJS Case Connect ; 13(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36763710

ABSTRACT

CASE: Phosphodiesterase type-5 enzyme (PDE5) inhibitors are well tolerated and used to treat erectile dysfunction. We report a case of prosthetic knee effusions associated with PDE5 inhibitor use in a 65-year-old man after total knee arthroplasty (TKA). PDE5 inhibitor treatment was stopped, and the patient had no further episodes of painful effusions. CONCLUSION: This report describes a previously unknown adverse effect of PDE5 inhibitor use in a prosthetic joint after TKA. We hope to encourage physicians managing patients after joint replacement to be aware of the association between PDE5 inhibitor use and recurrent joint effusions to improve postoperative outcomes.


Subject(s)
Arthroplasty, Replacement, Knee , Erectile Dysfunction , Male , Humans , Aged , Phosphodiesterase 5 Inhibitors/adverse effects , Arthroplasty, Replacement, Knee/adverse effects , Erectile Dysfunction/chemically induced , Erectile Dysfunction/drug therapy , Phosphoric Diester Hydrolases/therapeutic use
12.
J Hand Surg Eur Vol ; 48(2_suppl): 42S-50S, 2023 09.
Article in English | MEDLINE | ID: mdl-37704027

ABSTRACT

Metacarpal fractures are common and can be functionally disabling. The majority are managed non-operatively. When surgical intervention is indicated, various methods of fixation are available with the utility of each being based on injury pattern, patient function and surgeon preference. Early mobilization, especially in case of open reduction and internal fixation, is a critical component of treatment to prevent stiffness and restore function. When possible, a fixation construct that can withstand the applied forces of early postoperative motion is chosen. We provide an updated description for diagnosis, treatment options and operative fixation for metacarpal fractures.


Subject(s)
Fractures, Bone , Hand Injuries , Metacarpal Bones , Humans , Metacarpal Bones/surgery , Fractures, Bone/surgery , Hand Injuries/surgery , Fracture Fixation, Internal , Open Fracture Reduction
13.
Arthroplast Today ; 21: 101139, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37151404

ABSTRACT

Background: The relationship between elevated body mass index (BMI) and adverse outcomes in joint arthroplasty is well established in the literature. This paper aims to challenge the conventional thought of excluding patients from a total knee or hip replacement based on BMI alone. Instead, we propose using the metabolic syndrome (MetS) and its defining components to better identify patients at high risk for intraoperative and postoperative complications. Methods: Patients who underwent primary, elective total knee and total hip arthroplasty were identified in the 2015-2020 American College of Surgeons National Surgical Quality Improvement Program database. Several defining components of MetS, such as hypertension, diabetes, and obesity, were compared to a metabolically healthy cohort. Postoperative outcomes assessed included mortality, length of hospital stay, 30-day surgical and medical complications, and discharge. Results: The outcomes of 529,737 patients from the American College of Surgeons National Surgical Quality Improvement Program who underwent total knee and total hip arthroplasty were assessed. MetS is associated with increased complications and increased mortality. Both hypertension and diabetes are associated with increased complications but have no impact on mortality. Interestingly, while obesity was associated with increased complications, there was a significant decrease in mortality. Conclusions: Our results show that the impact of MetS is more than the sum of its constitutive parts. Additionally, obese patients experience a protective effect, with lower mortality than their nonobese counterparts. This study supports moving away from strict BMI cutoffs alone for someone to be eligible for an arthroplasty surgery and offers more granular data for risk stratification and patient selection.

14.
bioRxiv ; 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-37034687

ABSTRACT

Alterations in tumor stroma influence prostate cancer progression and metastatic potential. However, the molecular underpinnings of this stromal-epithelial crosstalk are largely unknown. Here, we compare mesenchymal cells from four genetically engineered mouse models (GEMMs) of prostate cancer representing different stages of the disease to their wild-type (WT) counterparts by single-cell RNA sequencing (scRNA-seq) and, ultimately, to human tumors with comparable genotypes. We identified 8 transcriptionally and functionally distinct stromal populations responsible for common and GEMM-specific transcriptional programs. We show that stromal responses are conserved in mouse models and human prostate cancers with the same genomic alterations. We noted striking similarities between the transcriptional profiles of the stroma of murine models of advanced disease and those of of human prostate cancer bone metastases. These profiles were then used to build a robust gene signature that can predict metastatic progression in prostate cancer patients with localized disease and is also associated with progression-free survival independent of Gleason score. Taken together, this offers new evidence that stromal microenvironment mediates prostate cancer progression, further identifying tissue-based biomarkers and potential therapeutic targets of aggressive and metastatic disease.

15.
Cancer Res Commun ; 3(8): 1447-1459, 2023 08.
Article in English | MEDLINE | ID: mdl-37546702

ABSTRACT

Although recent efforts have led to the development of highly effective androgen receptor (AR)-directed therapies for the treatment of advanced prostate cancer, a significant subset of patients will progress with resistant disease including AR-negative tumors that display neuroendocrine features [neuroendocrine prostate cancer (NEPC)]. On the basis of RNA sequencing (RNA-seq) data from a clinical cohort of tissue from benign prostate, locally advanced prostate cancer, metastatic castration-resistant prostate cancer and NEPC, we developed a multi-step bioinformatics pipeline to identify NEPC-specific, overexpressed gene transcripts that encode cell surface proteins. This included the identification of known NEPC surface protein CEACAM5 as well as other potentially targetable proteins (e.g., HMMR and CESLR3). We further showed that cadherin EGF LAG seven-pass G-type receptor 3 (CELSR3) knockdown results in reduced NEPC tumor cell proliferation and migration in vitro. We provide in vivo data including laser capture microdissection followed by RNA-seq data supporting a causal role of CELSR3 in the development and/or maintenance of the phenotype associated with NEPC. Finally, we provide initial data that suggests CELSR3 is a target for T-cell redirection therapeutics. Further work is now needed to fully evaluate the utility of targeting CELSR3 with T-cell redirection or other similar therapeutics as a potential new strategy for patients with NEPC. Significance: The development of effective treatment for patients with NEPC remains an unmet clinical need. We have identified specific surface proteins, including CELSR3, that may serve as novel biomarkers or therapeutic targets for NEPC.


Subject(s)
Neuroendocrine Tumors , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/genetics , Neuroendocrine Tumors/genetics , Prostate/metabolism , Cell Membrane/metabolism , Cadherins/genetics
16.
Cureus ; 14(2): e21813, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35261833

ABSTRACT

An 81-year-old male patient who underwent a Medacta GMK sphere kinematically aligned (KA) total knee arthroplasty (TKA) for end-stage knee osteoarthritis presented with a dislocated medial pivot (MP) tibia polyethylene (PE) insert on routine six-week postoperative x-rays. The patient presented asymptomatic with a normal range of motion. Dissociation of a fixed-bearing (FB) PE implant is an uncommon complication after TKA. There are only a few cases reported in the literature. We report for the first time a case of non-traumatic dissociation of MP PE from the tibial baseplate in a KA TKA in an asymptomatic patient but identified on routine postoperative radiographs.

17.
Resuscitation ; 181: 140-146, 2022 12.
Article in English | MEDLINE | ID: mdl-36410605

ABSTRACT

INTRODUCTION: Transcutaneous cardiac pacing (TCP) is a lifesaving procedure for patients with certain types of unstable bradycardia. We aimed to assess the difference in the pacing thresholds between the anteroposterior (AP) and anterolateral (AL) pacer pad positions. The second aim was to characterize the severity of chest wall muscle contractions during TCP. METHODS: In this prospective crossover trial, we enrolled patients presenting to the electrophysiology laboratory for elective cardioversion. After successful cardioversion, sedated participants were sequentially paced in both positions. The study procedure concluded after successful capture or inability to achieve capture by 140 mA (the pacer's maximum output) in both positions. Pacing thresholds were compared between positions, using a student's paired t-test, assigning a value of 141 mA to any trials with non-capture. RESULTS: Forty-one patients were screened; 20 were enrolled in the study. Seven participants were excluded from the paired analysis (three were prevented from pacing in the second position at the anesthesiologist's discretion, and 4 did not capture in either position). The study population consisted of 14 men and 6 women with a median age of 65 years. The mean pacing threshold was 33 mA lower (P = 0.001, 95% CI 20-45) in the AP (93 mA) versus the AL (126 mA) position. The median contraction severity score was 3 in the AL position versus 4 in the AP position (P = 0.005). CONCLUSIONS: Placing pacer pads in the AP position requires less energy to capture. Major resuscitation guidelines may favor the AP position for TCP. CLINICALTRIALS: gov Identifier: NCT03898050 https://clinicaltrials.gov/ct2/show/NCT03898050.


Subject(s)
Bradycardia , Cardiac Pacing, Artificial , Aged , Female , Humans , Male , Bradycardia/therapy , Cardiac Pacing, Artificial/methods , Electric Countershock , Heart , Prospective Studies , Cross-Over Studies
18.
Elife ; 112022 12 13.
Article in English | MEDLINE | ID: mdl-36511483

ABSTRACT

Advanced prostate malignancies are a leading cause of cancer-related deaths in men, in large part due to our incomplete understanding of cellular drivers of disease progression. We investigate prostate cancer cell dynamics at single-cell resolution from disease onset to the development of androgen independence in an in vivo murine model. We observe an expansion of a castration-resistant intermediate luminal cell type that correlates with treatment resistance and poor prognosis in human patients. Moreover, transformed epithelial cells and associated fibroblasts create a microenvironment conducive to pro-tumorigenic immune infiltration, which is partially androgen responsive. Androgen-independent prostate cancer leads to significant diversification of intermediate luminal cell populations characterized by a range of androgen signaling activity, which is inversely correlated with proliferation and mRNA translation. Accordingly, distinct epithelial populations are exquisitely sensitive to translation inhibition, which leads to epithelial cell death, loss of pro-tumorigenic signaling, and decreased tumor heterogeneity. Our findings reveal a complex tumor environment largely dominated by castration-resistant luminal cells and immunosuppressive infiltrates.


Subject(s)
Androgens , Prostatic Neoplasms , Male , Humans , Mice , Animals , Prostate/metabolism , Prostatic Neoplasms/pathology , Orchiectomy , Population Dynamics , Receptors, Androgen/metabolism , Disease Progression , Tumor Microenvironment
19.
Adv Mater ; 34(2): e2100096, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34676924

ABSTRACT

Following treatment with androgen receptor (AR) pathway inhibitors, ≈20% of prostate cancer patients progress by shedding their AR-dependence. These tumors undergo epigenetic reprogramming turning castration-resistant prostate cancer adenocarcinoma (CRPC-Adeno) into neuroendocrine prostate cancer (CRPC-NEPC). No targeted therapies are available for CRPC-NEPCs, and there are minimal organoid models to discover new therapeutic targets against these aggressive tumors. Here, using a combination of patient tumor proteomics, RNA sequencing, spatial-omics, and a synthetic hydrogel-based organoid, putative extracellular matrix (ECM) cues that regulate the phenotypic, transcriptomic, and epigenetic underpinnings of CRPC-NEPCs are defined. Short-term culture in tumor-expressed ECM differentially regulated DNA methylation and mobilized genes in CRPC-NEPCs. The ECM type distinctly regulates the response to small-molecule inhibitors of epigenetic targets and Dopamine Receptor D2 (DRD2), the latter being an understudied target in neuroendocrine tumors. In vivo patient-derived xenograft in immunocompromised mice showed strong anti-tumor response when treated with a DRD2 inhibitor. Finally, we demonstrate that therapeutic response in CRPC-NEPCs under drug-resistant ECM conditions can be overcome by first cellular reprogramming with epigenetic inhibitors, followed by DRD2 treatment. The synthetic organoids suggest the regulatory role of ECM in therapeutic response to targeted therapies in CRPC-NEPCs and enable the discovery of therapies to overcome resistance.


Subject(s)
Organoids , Prostatic Neoplasms, Castration-Resistant , Androgen Receptor Antagonists/pharmacology , Androgen Receptor Antagonists/therapeutic use , Animals , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Extracellular Matrix/metabolism , Humans , Hydrogels/pharmacology , Hydrogels/therapeutic use , Male , Mice , Organoids/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/therapeutic use
20.
JBJS Case Connect ; 11(4)2021 10 20.
Article in English | MEDLINE | ID: mdl-34669655

ABSTRACT

CASE: We report a case of bilateral capitate osteonecrosis in a patient who has a history of acute lymphocytic leukemia treated with systemic steroids and other chemotherapeutic agents. After exhausting conservative treatment, the patient underwent surgical management with a right-sided 4-corner arthrodesis and left-sided vascular pedicle graft, providing pain relief and improved function. CONCLUSION: In patients with a history of hematologic malignancy, clinicians should consider osteonecrosis of the capitate as a cause of wrist pain. Salvage procedures and vascularized grafts can provide pain relief in the presence of both early and late capitate osteonecrosis or collapse.


Subject(s)
Capitate Bone , Osteonecrosis , Arthralgia , Arthrodesis , Capitate Bone/diagnostic imaging , Capitate Bone/surgery , Humans , Osteonecrosis/diagnostic imaging , Osteonecrosis/surgery , Upper Extremity
SELECTION OF CITATIONS
SEARCH DETAIL