Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 18(6): e0276089, 2023.
Article in English | MEDLINE | ID: mdl-37347732

ABSTRACT

Diversity enriches the educational experience by improving intellectual engagement, self-motivation, citizenship, cultural engagement, and academic skills like critical thinking, problem-solving, and writing for students of all races. Faculty role models from similar backgrounds are essential for students from traditionally underrepresented groups as it sends a powerful message of support, belonging, and the confidence to pursue higher education. However, in the biomedical sciences, the percentage of historically underrepresented tenure-track faculty is far lower than that of their white colleagues. For this to change, a strong strategic plan and commitment from the university are imperative. This scoping review will assess the size and scope of available peer-reviewed research literature on diversity programs that aim to increase the recruitment and retention of biomedical sciences research faculty and are implemented and evaluated at American Universities. The information provided in this scoping review will help universities identify novel, successful diversity-based approaches for recruiting and retaining biomedical science faculty that might suit their own unique academic and geographic needs and be incorporated into their diversity initiatives and policies. The review follows the Population-Concept-Context methodology for Joanna Briggs Institution Scoping Reviews. Relevant peer-reviewed studies published in English between June 1, 2012, to June 1, 2022, will be identified from the following electronic databases; MEDLINE (PubMed), Scopus (Elsevier), EMBASE (Elsevier), CINAHL (EBSCO), and ERIC (EBSCO). The search strings using the key variables "biomedical research faculty," "recruitment/retention," "diversity/ minority/ underrepresented, and "mentoring" will be conducted using Boolean logic. Two independent reviewers will conduct all title and abstract screening, followed by a full article screening and data extraction. Due to the possible heterogeneity of the studies, we hope to use either a narrative analysis and/or descriptive figures/tables to depict the results.


Subject(s)
Faculty , Peer Review , Humans , United States , Universities , Educational Status , Students , Review Literature as Topic
2.
Mol Cell Oncol ; 9(1): 2136476, 2022.
Article in English | MEDLINE | ID: mdl-36313480

ABSTRACT

Prostate cancer (PC) is a commonly diagnosed malignancy in men and is associated with high mortality rates. Current treatments for PC include surgery, chemotherapy, and radiation therapy. However, recent advances in targeted delivery systems have yielded promising new approaches to PC treatment. As PC epithelial cells express high levels of prostate-specific membrane antigen (PSMA) on the cell surface, new drug conjugates focused on PSMA targeting have been developed. microRNAs (miRNAs) are small noncoding RNAs that regulate posttranscriptional gene expression in cells and show excellent possibilities for use in developing new therapeutics for PC. PSMA-targeted therapies based on a miRNA payload and that selectively target PC cells enhances therapeutic efficacy without eliciting damage to normal surrounding tissue. This review discusses the rationale for utilizing miRNAs to target PSMA, revealing their potential in therapeutic approaches to PC treatment. Different delivery systems for miRNAs and challenges to miRNA therapy are also explored.

3.
Front Cell Dev Biol ; 10: 895433, 2022.
Article in English | MEDLINE | ID: mdl-35898402

ABSTRACT

Zinc (Zn) is an essential trace element that plays a key role in several biological processes, including transcription, signaling, and catalysis. A subcellular network of transporters ensures adequate distribution of Zn to facilitate homeostasis. Among these are a family of importers, the Zrt/Irt-like proteins (ZIP), which consists of 14 members (ZIP1-ZIP14) that mobilize Zn from the extracellular domain and organelles into the cytosol. Expression of these transporters varies among tissues and during developmental stages, and their distribution at various cellular locations is essential for defining the net cellular Zn transport. Normally, the ion is bound to proteins or sequestered in organelles and vesicles. However, though research has focused on Zn internalization in mammalian cells, little is known about Zn mobilization within organelles, including within the nuclei under both normal and pathological conditions. Analyses from stomach and colon tissues isolated from mouse suggested that ZIP11 is the only ZIP transporter localized to the nucleus of mammalian cells, yet no clear cellular role has been attributed to this protein. We hypothesized that ZIP11 is essential to maintaining nuclear Zn homeostasis in mammalian cells. To test this, we utilized HeLa cells, as research in humans correlated elevated expression of ZIP11 with poor prognosis in cervical cancer patients. We stably knocked down ZIP11 in HeLa cancer cells and investigated the effect of Zn dysregulation in vitro. Our data show that ZIP11 knockdown (KD) reduced HeLa cells proliferation due to nuclear accumulation of Zn. RNA-seq analyses revealed that genes related to angiogenesis, apoptosis, mRNA metabolism, and signaling pathways are dysregulated. Although the KD cells undergoing nuclear Zn stress can activate the homeostasis response by MTF1 and MT1, the RNA-seq analyses showed that only ZIP14 (an importer expressed on the plasma membrane and endocytic vesicles) is mildly induced, which may explain the sensitivity to elevated levels of extracellular Zn. Consequently, ZIP11 KD HeLa cells have impaired migration, invasive properties and decreased mitochondrial potential. Furthermore, KD of ZIP11 delayed cell cycle progression and rendered an enhanced senescent state in HeLa cells, pointing to a novel mechanism whereby maintenance of nuclear Zn homeostasis is essential for cancer progression.

4.
Bioconjug Chem ; 32(11): 2386-2396, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34699177

ABSTRACT

The tunable nature of phosphoramidate linkers enables broad applicability as pH-triggered controlled-release platforms, particularly in the context of antibody- and small-molecule-drug conjugates (ADCs and SMDCs), where there remains a need for new linker technology. Herein, we explored in-depth the release of turn-on fluorogenic payloads from a homoserinyl-based phosphoramidate acid-cleavable linker. Kinetics of payload release from the scaffold was observed in buffers representing the pH conditions of systemic circulation, early and late endosomes, and lysosomes. It was found that payload release takes place in two key consecutive steps: (1) P-N bond hydrolysis and (2) spacer immolation. These two steps were found to follow pseudo-first-order kinetics and had opposite dependencies on pH. P-N bond hydrolysis increased with decreasing pH, while spacer immolation was most rapid at physiological pH. Despite the contrasting release kinetics of these two steps, maximal payload release was observed at the mildly acidic pH (5.0-5.5), while minimal payload release occurred at physiological pH. We integrated this phosphoramidate-payload linker system into a PSMA-targeted fluorescent turn-on probe to study the intracellular trafficking and release of a fluorescent payload in PSMA-expressing prostate cancer cells. Results showed excellent turn-on and accumulation of the coumarin payload in the late endosomal and lysosomal compartments of these cells. The release properties of this linker mark it as an attractive alternative in the modular design of ADCs and SMDCs, which demand selective intracellular payload release triggered by the pH changes that accompany intracellular trafficking.


Subject(s)
Prostate , Humans , Male
5.
BMC Syst Biol ; 12(1): 50, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29631571

ABSTRACT

BACKGROUND: Cell death as a result of ischemic injury triggers powerful mechanisms regulated by germline-encoded Pattern Recognition Receptors (PRRs) with shared specificity that recognize invading pathogens and endogenous ligands released from dying cells, and as such are essential to human health. Alternatively, dysregulation of these mechanisms contributes to extreme inflammation, deleterious tissue damage and impaired healing in various diseases. The Toll-like receptors (TLRs) are a prototypical family of PRRs that may be powerful anti-inflammatory targets if agents can be designed that antagonize their harmful effects while preserving host defense functions. This requires an understanding of the complex interactions and consequences of targeting the TLR-mediated pathways as well as technologies to analyze and interpret these, which will then allow the simulation of perturbations targeting specific pathway components, predict potential outcomes and identify safe and effective therapeutic targets. RESULTS: We constructed a multiscale mathematical model that spans the tissue and intracellular scales, and captures the consequences of targeting various regulatory components of injury-induced TLR4 signal transduction on potential pro-inflammatory or pro-healing outcomes. We applied known interactions to simulate how inactivation of specific regulatory nodes affects dynamics in the context of injury and to predict phenotypes of potential therapeutic interventions. We propose rules to link model behavior to qualitative estimates of pro-inflammatory signal activation, macrophage infiltration, production of reactive oxygen species and resolution. We tested the validity of the model by assessing its ability to reproduce published data not used in its construction. CONCLUSIONS: These studies will enable us to form a conceptual framework focusing on TLR4-mediated ischemic repair to assess potential molecular targets that can be utilized therapeutically to improve efficacy and safety in treating ischemic/inflammatory injury.


Subject(s)
Immunity, Innate , Ischemia/immunology , Models, Immunological , CD13 Antigens/metabolism , Cell Death/immunology , Cell Membrane/metabolism , Endosomes/metabolism , Humans , Ischemia/metabolism , Ischemia/pathology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism
6.
Immunology ; 142(4): 636-47, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24627994

ABSTRACT

CD13/Aminopeptidase N is a transmembrane metalloproteinase that is expressed in many tissues where it regulates various cellular functions. In inflammation, CD13 is expressed on myeloid cells, is up-regulated on endothelial cells at sites of inflammation and mediates monocyte/endothelial adhesion by homotypic interactions. In animal models the lack of CD13 alters the profiles of infiltrating inflammatory cells at sites of ischaemic injury. Here, we found that CD13 expression is enriched specifically on the pro-inflammatory subset of monocytes, suggesting that CD13 may regulate trafficking and function of specific subsets of immune cells. To further dissect the mechanisms regulating CD13-dependent trafficking we used the murine model of thioglycollate-induced sterile peritonitis. Peritoneal monocytes, macrophages and dendritic cells were significantly decreased in inflammatory exudates from global CD13(KO) animals when compared with wild-type controls. Furthermore, adoptive transfer of wild-type and CD13(KO) primary myeloid cells, or wild-type myeloid cells pre-treated with CD13-blocking antibodies into thioglycollate-challenged wild-type recipients demonstrated fewer CD13(KO) or treated cells in the lavage, suggesting that CD13 expression confers a competitive advantage in trafficking. Similarly, both wild-type and CD13(KO) cells were reduced in infiltrates in CD13(KO) recipients, confirming that both monocytic and endothelial CD13 contribute to trafficking. Finally, murine monocyte cell lines expressing mouse/human chimeric CD13 molecules demonstrated that the C-terminal domain of the protein mediates CD13 adhesion. Therefore, this work verifies that the altered inflammatory trafficking in CD13(KO) mice is the result of aberrant myeloid cell subset trafficking and further defines the molecular mechanisms underlying this regulation.


Subject(s)
CD13 Antigens/immunology , Cell Movement/immunology , Macrophages, Peritoneal/immunology , Monocytes/immunology , Animals , CD13 Antigens/genetics , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Humans , Macrophages, Peritoneal/cytology , Mice , Mice, Knockout , Monocytes/cytology , U937 Cells
7.
J Immunol ; 191(7): 3905-12, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23997214

ABSTRACT

CD13 is a large cell surface peptidase expressed on the monocytes and activated endothelial cells that is important for homing to and resolving the damaged tissue at sites of injury. We showed previously that cross-linking of human monocytic CD13 with activating Abs induces strong adhesion to endothelial cells in a tyrosine kinase- and microtubule-dependent manner. In the current study, we examined the molecular mechanisms underlying these observations in vitro and in vivo. We found that cross-linking of CD13 on U937 monocytic cells induced phosphorylation of a number of proteins, including Src, FAK, and ERK, and inhibition of these abrogated CD13-dependent adhesion. We found that CD13 itself was phosphorylated in a Src-dependent manner, which was an unexpected finding because its 7-aa cytoplasmic tail was assumed to be inert. Furthermore, CD13 was constitutively associated with the scaffolding protein IQGAP1, and CD13 cross-linking induced complex formation with the actin-binding protein α-actinin, linking membrane-bound CD13 to the cytoskeleton, further supporting CD13 as an inflammatory adhesion molecule. Mechanistically, mutation of the conserved CD13 cytoplasmic tyrosine to phenylalanine abrogated adhesion; Src, FAK, and ERK phosphorylation; and cytoskeletal alterations upon Ab cross-linking. Finally, CD13 was phosphorylated in isolated murine inflammatory peritoneal exudate cells, and adoptive transfer of monocytic cell lines engineered to express the mutant CD13 were severely impaired in their ability to migrate into the inflamed peritoneum, confirming that CD13 phosphorylation is relevant to inflammatory cell trafficking in vivo. Therefore, this study identifies CD13 as a novel, direct activator of intracellular signaling pathways in pathophysiological conditions.


Subject(s)
CD13 Antigens/metabolism , Cell Movement/immunology , Monocytes/immunology , Monocytes/metabolism , Animals , CD13 Antigens/genetics , Cell Adhesion/immunology , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Knockout , Phosphorylation , Plakins/metabolism , Protein Binding , Signal Transduction , src-Family Kinases/metabolism
8.
PLoS One ; 7(7): e41285, 2012.
Article in English | MEDLINE | ID: mdl-22815987

ABSTRACT

BACKGROUND: Aberrant growth of blood vessels in the eye forms the basis of many incapacitating diseases and currently the majority of patients respond to anti-angiogenic therapies based on blocking the principal angiogenic growth factor, vascular endothelial growth factor (VEGF). While highly successful, new therapeutic targets are critical for the increasing number of individuals susceptible to retina-related pathologies in our increasingly aging population. Prostate specific membrane antigen (PSMA) is a cell surface peptidase that is absent on normal tissue vasculature but is highly expressed on the neovasculature of most solid tumors, where we have previously shown to regulate angiogenic endothelial cell invasion. Because pathologic angiogenic responses are often triggered by distinct signals, we sought to determine if PSMA also contributes to the pathologic angiogenesis provoked by hypoxia of the retina, which underlies many debilitating retinopathies. METHODOLOGY/PRINCIPAL FINDINGS: Using a mouse model of oxygen-induced retinopathy, we found that while developmental angiogenesis is normal in PSMA null mice, hypoxic challenge resulted in decreased retinal vascular pathology when compared to wild type mice as assessed by avascular area and numbers of vascular tufts/glomeruli. The vessels formed in the PSMA null mice were more organized and highly perfused, suggesting a more 'normal' phenotype. Importantly, the decrease in angiogenesis was not due to an impaired hypoxic response as levels of pro-angiogenic factors are comparable; indicating that PSMA regulation of angiogenesis is independent of VEGF. Furthermore, both systemic and intravitreal administration of a PSMA inhibitor in wild type mice undergoing OIR mimicked the PSMA null phenotype resulting in improved retinal vasculature. CONCLUSIONS/SIGNIFICANCE: Our data indicate that PSMA plays a VEGF-independent role in retinal angiogenesis and that the lack of or inhibition of PSMA may represent a novel therapeutic strategy for treatment of angiogenesis-based ocular diseases.


Subject(s)
Antigens, Surface/biosynthesis , Gene Expression Regulation , Glutamate Carboxypeptidase II/biosynthesis , Retinal Neovascularization/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Membrane/metabolism , Hydrogen-Ion Concentration , Hypoxia , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Oxygen/metabolism , Peptide Hydrolases/chemistry , Perfusion , Phenotype , Retina/metabolism , Retina/pathology , Retinal Vein/pathology
9.
J Biol Chem ; 285(14): 10487-96, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20097759

ABSTRACT

Phogrin is a transmembrane protein expressed in cells with stimulus-coupled peptide hormone secretion, including pancreatic beta cells, in which it is localized to the membrane of insulin-containing dense-core vesicles. By sequence, phogrin is a member of the family of receptor-like protein-tyrosine phosphatases, but it contains substitutions in conserved catalytic sequences, and no significant enzymatic activity for phogrin has ever been reported. We report here that phogrin is able to dephosphorylate specific inositol phospholipids, including phosphatidylinositol (PI) 3-phosphate and PI 4,5-diphosphate but not PI 3,4,5-trisphosphate. The phosphatidylinositol phosphatase (PIPase) activity of phogrin was measurable but low when evaluated by the ability of a catalytic domain fusion protein to hydrolyze soluble short-chain phosphatidylinositol phospholipids. Unlike most PIPases, which are cytoplasmic proteins that associate with membranes, mature phogrin is a transmembrane protein. When the transmembrane form of phogrin was overexpressed in mammalian cells, it reduced plasma membrane phosphatidylinositol 4,5-disphosphate levels in a dose-dependent manner. When purified and assayed in vitro, the transmembrane form had a specific activity of 142 mol/min/mol, 75-fold more active than the catalytic domain fusion protein and comparable with the specific activities of the other PIPases. The PIPase activity of phogrin depended on the catalytic site cysteine and correlated with effects on glucose-stimulated insulin secretion. We propose that phogrin functions as a phosphatidylinositol phosphatase that contributes to maintaining subcellular differences in levels of PIP that are important for regulating stimulus-coupled exocytosis of insulin.


Subject(s)
Hypoglycemic Agents/metabolism , Insulin/metabolism , Insulinoma/metabolism , Pancreatic Neoplasms/metabolism , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Secretory Vesicles/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Exocytosis/physiology , Fluorescent Antibody Technique , Glucose/metabolism , Insulin Secretion , Insulinoma/genetics , Insulinoma/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Phosphotyrosine/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Receptor-Like Protein Tyrosine Phosphatases, Class 8/antagonists & inhibitors , Receptor-Like Protein Tyrosine Phosphatases, Class 8/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...