Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Commun Biol ; 7(1): 507, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678127

ABSTRACT

Our nervous system contains billions of neurons that form precise connections with each other through interactions between cell surface proteins. In Drosophila, the Dpr and DIP immunoglobulin protein subfamilies form homophilic or heterophilic interactions to instruct synaptic connectivity, synaptic growth, and cell survival. However, the upstream regulatory mechanisms of Dprs and DIPs are not clear. On the other hand, while transcription factors have been implicated in target recognition, their downstream cell surface proteins remain mostly unknown. We conduct an F1 dominant modifier genetic screen to identify regulators of Dprs and DIPs. We identify huckebein (hkb), a transcription factor previously implicated in target recognition of the dorsal Is motor neuron. We show that hkb genetically interacts with DIP-α and loss of hkb leads to complete removal of DIP-α expression specifically in dorsal Is motor neurons. We then confirm that this specificity is through the dorsal Is motor neuron specific transcription factor, even-skipped (eve), which acts downstream of hkb. Analysis of the genetic interaction between hkb and eve reveals that they act in the same pathway to regulate dorsal Is motor neuron connectivity. Our study provides insight into the transcriptional regulation of DIP-α and suggests that distinct regulatory mechanisms exist for the same CSP in different neurons.


Subject(s)
Drosophila Proteins , Transcription Factors , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Neuromuscular Junction/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
2.
bioRxiv ; 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37905128

ABSTRACT

Our nervous system contains billions of neurons that form precise connections with each other through interactions between cell surface proteins (CSPs). In Drosophila, the Dpr and DIP immunoglobulin protein subfamilies form homophilic or heterophilic interactions to instruct synaptic connectivity, synaptic growth and cell survival. However, the upstream regulation and downstream signaling mechanisms of Dprs and DIPs are not clear. In the Drosophila larval neuromuscular system, DIP-α is expressed in the dorsal and ventral type-Is motor neurons (MNs). We conducted an F1 dominant modifier genetic screen to identify regulators of Dprs and DIPs. We found that the transcription factor, huckebein (hkb), genetically interacts with DIP-α and is important for target recognition specifically in the dorsal Is MN, but not the ventral Is MN. Loss of hkb led to complete removal of DIP-α expression. We then confirmed that this specificity is through the dorsal Is MN specific transcription factor, even-skipped (eve), which acts downstream of hkb. Genetic interaction between hkb and eve revealed that they act in the same pathway to regulate dorsal Is MN connectivity. Our study provides insight into the transcriptional regulation of DIP-α and suggests that distinct regulatory mechanisms exist for the same CSP in different neurons.

3.
J Hematol Oncol ; 15(1): 80, 2022 06 11.
Article in English | MEDLINE | ID: mdl-35690784

ABSTRACT

Despite recent advances in cancer therapeutics, glioblastoma (GBM) remains one of the most difficult cancers to treat in both the primary and recurrent settings. GBM presents a unique therapeutic challenge given the immune-privileged environment of the brain and the aggressive nature of the disease. Furthermore, it can change phenotypes throughout the course of disease-switching between mesenchymal, neural, and classic gene signatures, each with specific markers and mechanisms of resistance. Recent advancements in the field of immunotherapy-which utilizes strategies to reenergize or alter the immune system to target cancer-have shown striking results in patients with many types of malignancy. Immune checkpoint inhibitors, adoptive cellular therapy, cellular and peptide vaccines, and other technologies provide clinicians with a vast array of tools to design highly individualized treatment and potential for combination strategies. There are currently over 80 active clinical trials evaluating immunotherapies for GBM, often in combination with standard secondary treatment options including re-resection and anti-angiogenic agents, such as bevacizumab. This review will provide a clinically focused overview of the immune environment present in GBM, which is frequently immunosuppressive and characterized by M2 macrophages, T cell exhaustion, enhanced transforming growth factor-ß signaling, and others. We will also outline existing immunotherapeutic strategies, with a special focus on immune checkpoint inhibitors, chimeric antigen receptor therapy, and dendritic cell vaccines. Finally, we will summarize key discoveries in the field and discuss currently active clinical trials, including combination strategies, burgeoning technology like nucleic acid and nanoparticle therapy, and novel anticancer vaccines. This review aims to provide the most updated summary of the field of immunotherapy for GBM and offer both historical perspective and future directions to help inform clinical practice.


Subject(s)
Brain Neoplasms , Glioblastoma , Physicians , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Immune Checkpoint Inhibitors , Immunologic Factors , Immunotherapy/methods , T-Lymphocytes
5.
Sci Transl Med ; 12(525)2020 01 08.
Article in English | MEDLINE | ID: mdl-31915300

ABSTRACT

Cancer-associated thrombocytosis and high concentrations of circulating transforming growth factor-ß1 (TGF-ß1) are frequently observed in patients with progressive cancers. Using genetic and pharmacological approaches, we show a direct link between thrombin catalytic activity and release of mature TGF-ß1 from platelets. We found that thrombin cleaves glycoprotein A repetitions predominant (GARP), a cell surface docking receptor for latent TGF-ß1 (LTGF-ß1) on platelets, resulting in liberation of active TGF-ß1 from the GARP-LTGF-ß1 complex. Furthermore, systemic inhibition of thrombin obliterates TGF-ß1 maturation in platelet releasate and rewires the tumor microenvironment toward favorable antitumor immunity, which translates into efficient cancer control either alone or in combination with programmed cell death 1-based immune checkpoint blockade therapy. Last, we demonstrate that soluble GARP and GARP-LTGF-ß1 complex are present in the circulation of patients with cancer. Together, our data reveal a mechanism of cancer immune evasion that involves thrombin-mediated GARP cleavage and the subsequent TGF-ß1 release from platelets. We propose that blockade of GARP cleavage is a valuable therapeutic strategy to overcome cancer's resistance to immunotherapy.


Subject(s)
Blood Platelets/metabolism , Immune Evasion , Latent TGF-beta Binding Proteins/metabolism , Membrane Proteins/metabolism , Proteolysis , Thrombin/metabolism , Animals , Carcinogenesis/drug effects , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Progression , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Evasion/drug effects , Latent TGF-beta Binding Proteins/blood , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Protein Binding/drug effects , Proteolysis/drug effects , Tumor Microenvironment/drug effects
6.
Exp Hematol Oncol ; 8: 27, 2019.
Article in English | MEDLINE | ID: mdl-31709128

ABSTRACT

The use of chimeric antigen receptor (CAR) T cell technology as a therapeutic strategy for the treatment blood-born human cancers has delivered outstanding clinical efficacy. However, this treatment modality can also be associated with serious adverse events in the form of cytokine release syndrome. While several avenues are being pursued to limit the off-target effects, it is critically important that any intervention strategy has minimal consequences on long term efficacy. A recent study published in Science Translational Medicine by Dr. Hudecek's group proved that dasatinib, a tyrosine kinase inhibitor, can serve as an on/off switch for CD19-CAR-T cells in preclinical models by limiting toxicities while maintaining therapeutic efficacy. In this editorial, we discuss the recent strategies for generating safer CAR-T cells, and also important questions surrounding the use of dasatinib for emergency intervention of CAR-T cell mediated cytokine release syndrome.

7.
Cancer Res ; 79(6): 1178-1190, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30674536

ABSTRACT

Activated regulatory T (Treg) cells express the surface receptor glycoprotein-A repetitions predominant (GARP), which binds and activates latent TGFß. How GARP modulates Treg function in inflammation and cancer remains unclear. Here we demonstrate that loss of GARP in Treg cells leads to spontaneous inflammation with highly activated CD4+ and CD8+ T cells and development of enteritis. Treg cells lacking GARP were unable to suppress pathogenic T-cell responses in multiple models of inflammation, including T-cell transfer colitis. GARP-/- Treg cells were significantly reduced in the gut and exhibited a reduction in CD103 expression, a colon-specific migratory marker. In the colitis-associated colon cancer model, GARP on Treg cells dampened immune surveillance, and mice with GARP-/- Treg cells exhibited improved antitumor immunity. Thus, GARP empowers the functionality of Treg cells and their tissue-specific accumulation, highlighting the importance of cell surface TGFß in Treg function and GARP as a potential therapeutic target for colorectal cancer therapy.Significance: These findings uncover functions of membrane-bound TGFß and GARP that tune the activity of Treg cells, highlighting a potential treatment strategy in autoimmune diseases and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Immune Tolerance/immunology , Inflammation/immunology , Membrane Proteins/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
8.
JCI Insight ; 3(7)2018 04 05.
Article in English | MEDLINE | ID: mdl-29618665

ABSTRACT

GARP, a cell surface docking receptor for binding and activating latent TGF-ß, is highly expressed by platelets and activated Tregs. While GARP is implicated in immune invasion in cancer, the roles of the GARP-TGF-ß axis in systemic autoimmune diseases are unknown. Although B cells do not express GARP at baseline, we found that the GARP-TGF-ß complex is induced on activated human and mouse B cells by ligands for multiple TLRs, including TLR4, TLR7, and TLR9. GARP overexpression on B cells inhibited their proliferation, induced IgA class-switching, and dampened T cell-independent antibody production. In contrast, B cell-specific deletion of GARP-encoding gene Lrrc32 in mice led to development of systemic autoimmune diseases spontaneously as well as worsening of pristane-induced lupus-like disease. Canonical TGF-ß signaling more readily upregulates GARP in Peyer patch B cells than in splenic B cells. Furthermore, we demonstrated that B cells are required for the induction of oral tolerance of T cell-dependent antigens via GARP. Our studies reveal for the first time to our knowledge that cell surface GARP-TGF-ß is an important checkpoint for regulating B cell peripheral tolerance, highlighting a mechanism of autoimmune disease pathogenesis.


Subject(s)
B-Lymphocytes/immunology , Immune Tolerance/immunology , Membrane Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adjuvants, Immunologic/administration & dosage , Animals , Autoimmune Diseases/immunology , B-Lymphocytes/metabolism , Bone Marrow Transplantation , Cells, Cultured , Female , Gene Knock-In Techniques , Healthy Volunteers , Humans , Immune Tolerance/drug effects , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Transgenic , Primary Cell Culture , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Transplantation Chimera
9.
J Hematol Oncol ; 11(1): 24, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458436

ABSTRACT

GARP (glycoprotein-A repetitions predominant) is a type I transmembrane cell surface docking receptor for latent transforming growth factor-ß (TGF-ß) that is abundantly expressed on regulatory T lymphocytes and platelets. GARP regulates the availability of membrane-bound latent TGF-ß and modulates its activation. For this reason, GARP expression on immune and non-immune cells is involved in maintaining peripheral tolerance. It plays an important role in preventing inflammatory diseases such as allergy and graft versus host disease (GvHD). GARP is also frequently hijacked by cancer cells to promote oncogenesis. This review summarizes the most important features of GARP biology described to date including gene regulation, protein expression and mechanism in activating latent TGF-ß, and the function of GARP in regulatory T cell biology and peripheral tolerance, as well as GARP's increasingly recognized roles in platelet-mediated cancer immune evasion. The promise for GARP-targeted strategy as a novel immunotherapy of cancer is also highlighted.


Subject(s)
Inflammation/immunology , Membrane Proteins/immunology , Neoplasms/immunology , Transforming Growth Factor beta/immunology , Animals , Blood Platelets/immunology , Blood Platelets/pathology , Gene Expression Regulation, Neoplastic , Humans , Immune Tolerance , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Membrane Proteins/analysis , Membrane Proteins/genetics , Neoplasms/complications , Neoplasms/genetics , Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/analysis
10.
J Nurs Adm ; 47(11): 545-550, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29045355

ABSTRACT

OBJECTIVE: The aim of this study is to determine if the pattern of monthly medical expense can be used to identify individuals at risk of dying, thus supporting providers in proactively engaging in advanced care planning discussions. BACKGROUND: Identifying the right time to discuss end of life can be difficult. Improved predictive capacity has made it possible for nurse leaders to use large data sets to guide clinical decision making. METHODS: We examined the patterns of monthly medical expense of Medicare beneficiaries with life-limiting illness during the last 24 months of life using analysis of variance, t tests, and stepwise hierarchical linear modeling. RESULTS: In the final year of life, monthly medical expense increases rapidly for all disease groupings and forms distinct patterns of change. CONCLUSION: Type of condition can be used to classify decedents into distinctly different cost trajectories. Conditions including chronic disease, system failure, or cancer may be used to identify patients who may benefit from supportive care.


Subject(s)
Advance Care Planning/standards , Centers for Medicare and Medicaid Services, U.S./economics , Chronic Disease/economics , Hospice Care/economics , Terminally Ill/statistics & numerical data , Advance Care Planning/organization & administration , Aged , Centers for Medicare and Medicaid Services, U.S./statistics & numerical data , Chronic Disease/classification , Chronic Disease/mortality , Communication , Costs and Cost Analysis , Electronic Health Records/standards , Electronic Health Records/statistics & numerical data , Hospice Care/statistics & numerical data , Humans , Meaningful Use/standards , Meaningful Use/statistics & numerical data , Physician-Patient Relations , Prognosis , Retrospective Studies , Risk Assessment/methods , United States/epidemiology , Unnecessary Procedures/economics , Unnecessary Procedures/statistics & numerical data
11.
Nat Struct Mol Biol ; 24(10): 834-839, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28869608

ABSTRACT

The unfolded protein response (UPR) in the endoplasmic reticulum (ER) is a highly conserved protein-quality-control mechanism critical for cells to make survival-or-death decisions under ER-stress conditions. However, how UPR sensors are activated remains unclear. Here, we report that ER luminal protein canopy homolog 2 (CNPY2) is released from grp78 upon ER stress. Free CNPY2 then engages protein kinase R-like ER kinase (PERK) to induce expression of the transcription factor C/EBP homologous protein (CHOP), thereby initiating the UPR. Indeed, deletion of CNPY2 blocked the PERK-CHOP pathway and protected mice from UPR-induced liver damage and steatosis. Additionally, CNPY2 is transcriptionally upregulated by CHOP in a forward-feed loop to further enhance UPR signaling. These findings demonstrate the critical roles of CNPY2 in ER stress and suggest that CNPY2 is a potential new therapeutic target for UPR-related diseases such as metabolic disorders, inflammation and cancer.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Transcription Factor CHOP/metabolism , Unfolded Protein Response , eIF-2 Kinase/metabolism , Animals , Cell Line , Endoplasmic Reticulum Chaperone BiP , Gene Deletion , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout
12.
J Biol Chem ; 292(44): 18091-18097, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28912269

ABSTRACT

Glycoprotein A repetitions predominant (GARP) (encoded by the Lrrc32 gene) plays important roles in cell-surface docking and activation of TGFß. However, GARP's role in organ development in mammalian systems is unclear. To determine the function of GARP in vivo, we generated a GARP KO mouse model. Unexpectedly, the GARP KO mice died within 24 h after birth and exhibited defective palatogenesis without apparent abnormalities in other major organs. Furthermore, we observed decreased apoptosis and SMAD2 phosphorylation in the medial edge epithelial cells of the palatal shelf of GARP KO embryos at embryonic day 14.5 (E14.5), indicating a defect in the TGFß signaling pathway in the GARP-null developing palates. Of note, the failure to develop the secondary palate and concurrent reduction of SMAD phosphorylation without other defects in GARP KO mice phenocopied TGFß3 KO mice, although GARP has not been suggested previously to interact with TGFß3. We found that GARP and TGFß3 co-localize in medial edge epithelial cells at E14.5. In vitro studies confirmed that GARP and TGFß3 directly interact and that GARP is indispensable for the surface expression of membrane-associated latent TGFß3. Our findings indicate that GARP is essential for normal morphogenesis of the palate and demonstrate that GARP plays a crucial role in regulating TGFß3 signaling during embryogenesis. In conclusion, we have uncovered a novel function of GARP in positively regulating TGFß3 activation and function.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Organogenesis , Palate/metabolism , Protein Processing, Post-Translational , Smad2 Protein/metabolism , Transforming Growth Factor beta3/agonists , Animals , Animals, Newborn , Apoptosis , Cleft Palate/embryology , Cleft Palate/metabolism , Cleft Palate/pathology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Gene Knock-In Techniques , HEK293 Cells , Heterozygote , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice, Knockout , Palate/abnormalities , Palate/embryology , Palate/pathology , Phosphorylation , Pregnancy , Protein Multimerization , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta3/chemistry , Transforming Growth Factor beta3/genetics , Transforming Growth Factor beta3/metabolism
13.
Sci Immunol ; 2(11)2017 May 05.
Article in English | MEDLINE | ID: mdl-28763790

ABSTRACT

Cancer-associated thrombocytosis has long been linked to poor clinical outcome, but the underlying mechanism is enigmatic. We hypothesized that platelets promote malignancy and resistance to therapy by dampening host immunity. We show that genetic targeting of platelets enhances adoptive T cell therapy of cancer. An unbiased biochemical and structural biology approach established transforming growth factor ß (TGFß) and lactate as major platelet-derived soluble factors to obliterate CD4+ and CD8+ T cell functions. Moreover, we found that platelets are the dominant source of functional TGFß systemically as well as in the tumor microenvironment through constitutive expression of the TGFß-docking receptor glycoprotein A repetitions predominant (GARP) rather than secretion of TGFß per se. Platelet-specific deletion of the GARP-encoding gene Lrrc32 blunted TGFß activity at the tumor site and potentiated protective immunity against both melanoma and colon cancer. Last, this study shows that T cell therapy of cancer can be substantially improved by concurrent treatment with readily available antiplatelet agents. We conclude that platelets constrain T cell immunity through a GARP-TGFß axis and suggest a combination of immunotherapy and platelet inhibitors as a therapeutic strategy against cancer.

14.
Proc Natl Acad Sci U S A ; 114(28): E5549-E5558, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652336

ABSTRACT

Neutral sphingomyelinase 2 (nSMase2, product of the SMPD3 gene) is a key enzyme for ceramide generation that is involved in regulating cellular stress responses and exosome-mediated intercellular communication. nSMase2 is activated by diverse stimuli, including the anionic phospholipid phosphatidylserine. Phosphatidylserine binds to an integral-membrane N-terminal domain (NTD); however, how the NTD activates the C-terminal catalytic domain is unclear. Here, we identify the complete catalytic domain of nSMase2, which was misannotated because of a large insertion. We find the soluble catalytic domain interacts directly with the membrane-associated NTD, which serves as both a membrane anchor and an allosteric activator. The juxtamembrane region, which links the NTD and the catalytic domain, is necessary and sufficient for activation. Furthermore, we provide a mechanistic basis for this phenomenon using the crystal structure of the human nSMase2 catalytic domain determined at 1.85-Å resolution. The structure reveals a DNase-I-type fold with a hydrophobic track leading to the active site that is blocked by an evolutionarily conserved motif which we term the "DK switch." Structural analysis of nSMase2 and the extended N-SMase family shows that the DK switch can adopt different conformations to reposition a universally conserved Asp (D) residue involved in catalysis. Mutation of this Asp residue in nSMase2 disrupts catalysis, allosteric activation, stimulation by phosphatidylserine, and pharmacological inhibition by the lipid-competitive inhibitor GW4869. Taken together, these results demonstrate that the DK switch regulates ceramide generation by nSMase2 and is governed by an allosteric interdomain interaction at the membrane interface.


Subject(s)
Allosteric Site , Ceramides/biosynthesis , Sphingomyelin Phosphodiesterase/chemistry , Aniline Compounds/chemistry , Benzylidene Compounds/chemistry , Catalytic Domain , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Lipids/chemistry , MCF-7 Cells , Protein Binding , Protein Folding , Saccharomyces cerevisiae , Signal Transduction
15.
Cancer Res ; 76(24): 7106-7117, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27913437

ABSTRACT

GARP encoded by the Lrrc32 gene is the cell surface docking receptor for latent TGFß, which is expressed naturally by platelets and regulatory T cells (Treg). Although Lrrc32 is amplified frequently in breast cancer, the expression and relevant functions of GARP in cancer have not been explored. Here, we report that GARP exerts oncogenic effects, promoting immune tolerance by enriching and activating latent TGFß in the tumor microenvironment. We found that human breast, lung, and colon cancers expressed GARP aberrantly. In genetic studies in normal mammary gland epithelial and carcinoma cells, GARP expression increased TGFß bioactivity and promoted malignant transformation in immunodeficient mice. In breast carcinoma-bearing mice that were immunocompetent, GARP overexpression promoted Foxp3+ Treg activity, which in turn contributed to enhancing cancer progression and metastasis. Notably, administration of a GARP-specific mAb limited metastasis in an orthotopic model of human breast cancer. Overall, these results define the oncogenic effects of the GARP-TGFß axis in the tumor microenvironment and suggest mechanisms that might be exploited for diagnostic and therapeutic purposes. Cancer Res; 76(24); 7106-17. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Carcinogenesis , Membrane Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Escape/physiology , Animals , Blotting, Western , Female , Heterografts , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Polymerase Chain Reaction , Tissue Array Analysis
16.
FASEB J ; 30(12): 4159-4171, 2016 12.
Article in English | MEDLINE | ID: mdl-27609772

ABSTRACT

Alterations in sphingolipid metabolism, especially ceramide and sphingosine 1-phosphate, have been linked to colon cancer, suggesting that enzymes of sphingolipid metabolism may emerge as novel regulators and targets in colon cancer. Neutral ceramidase (nCDase), a key enzyme in sphingolipid metabolism that hydrolyzes ceramide into sphingosine, is highly expressed in the intestine; however, its role in colon cancer has not been defined. Here we show that molecular and pharmacological inhibition of nCDase in colon cancer cells increases ceramide, and this is accompanied by decreased cell survival and increased apoptosis and autophagy, with minimal effects on noncancerous cells. Inhibition of nCDase resulted in loss of ß-catenin and inhibition of ERK, components of pathways relevant for colon cancer development. Furthermore, inhibition of nCDase in a xenograft model delayed tumor growth and increased ceramide while decreasing proliferation. It is noteworthy that mice lacking nCDase treated with azoxymethane were protected from tumor formation. Taken together, these studies show that nCDase is pivotal for regulating initiation and development of colon cancer, and these data suggest that this enzyme is a suitable and novel target for colon cancer therapy.-García-Barros, M., Coant, N., Kawamori, T., Wada, M., Snider, A. J., Truman, J.-P., Wu, B. X., Furuya, H., Clarke, C. J., Bialkowska, A. B., Ghaleb, A., Yang, V. W., Obeid, L. M., Hannun, Y. A. Role of neutral ceramidase in colon cancer.


Subject(s)
Ceramides/metabolism , Colonic Neoplasms/enzymology , Lipid Metabolism/physiology , Neutral Ceramidase/metabolism , Animals , Colon/metabolism , Humans , Male , Mice, Knockout , Sphingolipids/metabolism , beta Catenin/metabolism
17.
Invest Ophthalmol Vis Sci ; 57(10): 4476-84, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27571014

ABSTRACT

PURPOSE: Acid sphingomyelinase (ASMase) catalyzes the hydrolysis of sphingomyelin to ceramide and mediates multiple responses involved in inflammatory and apoptotic signaling. However, the role ASMase plays in ischemic retinal injury has not been investigated. The purpose of this study was to investigate how reduced ASMase expression impacts retinal ischemic injury. METHODS: Changes in ceramide levels and ASMase activity were determined by high performance liquid chromatography-tandem mass spectrometry analysis and ASMase activity. Retinal function and morphology were assessed by electroretinography (ERG) and morphometric analyses. Levels of TNF-α were determined by ELISA. Activation of p38 MAP kinase was assessed by Western blot analysis. RESULTS: In wild-type mice, ischemia produced a significant increase in retinal ASMase activity and ceramide levels. These increases were associated with functional deficits as measured by ERG analysis and significant structural degeneration in most retinal layers. In ASMase+/- mice, retinal ischemia did not significantly alter ASMase activity, and the rise in ceramide levels were significantly reduced compared to levels in retinas from wild-type mice. In ASMase+/- mice, functional and morphometric analyses of ischemic eyes revealed significantly less retinal degeneration than in injured retinas from wild-type mice. The ischemia-induced increase in retinal TNF-α levels was suppressed by the administration of the ASMase inhibitor desipramine, or by reducing ASMase expression. CONCLUSIONS: Our results demonstrate that reducing ASMase expression provides partial protection from ischemic injury. Hence, the production of ceramide and subsequent mediators plays a role in the development of ischemic retinal injury. Modulating ASMase may present new opportunities for adjunctive therapies when treating retinal ischemic disorders.


Subject(s)
Desipramine/pharmacology , Reperfusion Injury/prevention & control , Retina/drug effects , Retinal Degeneration/prevention & control , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Ceramides/metabolism , Chromatography, High Pressure Liquid , Disease Models, Animal , Electroretinography , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/complications , Reperfusion Injury/metabolism , Retina/diagnostic imaging , Retina/physiopathology , Retinal Degeneration/diagnosis , Retinal Degeneration/etiology , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Curr Top Med Chem ; 16(25): 2765-78, 2016.
Article in English | MEDLINE | ID: mdl-27072698

ABSTRACT

As an endoplasmic reticulum heat shock protein (HSP) 90 paralogue, glycoprotein (gp) 96 possesses immunological properties by chaperoning antigenic peptides for activation of T cells. Genetic studies in the last decade have unveiled that gp96 is also an essential master chaperone for multiple receptors and secreting proteins including Toll-like receptors (TLRs), integrins, the Wnt coreceptor, Low Density Lipoprotein Receptor-Related Protein 6 (LRP6), the latent TGFß docking receptor, Glycoprotein A Repetitions Predominant (GARP), Glycoprotein (GP) Ib and insulin-like growth factors (IGF). Clinically, elevated expression of gp96 in a variety of cancers correlates with the advanced stage and poor survival of cancer patients. Recent preclinical studies have also uncovered that gp96 expression is closely linked to cancer progression in multiple myeloma, hepatocellular carcinoma, breast cancer and inflammation-associated colon cancer. Thus, gp96 is an attractive therapeutic target for cancer treatment. The chaperone function of gp96 depends on its ATPase domain, which is structurally distinct from other HSP90 members, and thus favors the design of highly selective gp96-targeted inhibitors against cancer. We herein discuss the strategically important oncogenic clients of gp96 and their underlying biology. The roles of cell-intrinsic gp96 in T cell biology are also discussed, in part because it offers another opportunity of cancer therapy by manipulating levels of gp96 in T cells to enhance host immune defense.


Subject(s)
Membrane Glycoproteins/physiology , Oncogenes , Humans , Neoplasms/genetics , Neoplasms/physiopathology , Neoplasms/therapy , T-Lymphocytes/metabolism
19.
Adv Cancer Res ; 129: 165-90, 2016.
Article in English | MEDLINE | ID: mdl-26916005

ABSTRACT

As an endoplasmic reticulum heat-shock protein 90 (HSP90) paralog, GRP94 (glucose-regulated protein 94)/gp96 (hereafter referred to as GRP94) has been shown to be an essential master chaperone for multiple receptors including Toll-like receptors, Wnt coreceptors, and integrins. Clinically, expression of GRP94 correlates with advanced stage and poor survival in a variety of cancers. Recent preclinical studies have also revealed that GRP94 expression is closely linked to cancer growth and metastasis in melanoma, ovarian cancer, multiple myeloma, lung cancer, and inflammation-associated colon cancer. Thus, GRP94 is an attractive therapeutic target in a number of malignancies. The chaperone function of GRP94 depends on its ATPase domain, which is structurally distinct from HSP90, allowing design of highly selective GRP94-targeted inhibitors. In this chapter, we discuss the biology and structure-function relationship of GRP94. We also summarize the immunological roles of GRP94 based on the studies documented over the last two decades, as these pertain to tumorigenesis and cancer progression. Finally, the structure-based rationale for the design of selective small-molecule inhibitors of GRP94 and their potential application in the treatment of cancer are highlighted.


Subject(s)
Antineoplastic Agents/chemistry , Drug Design , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/immunology , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/immunology , Neoplasms/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenesis/immunology , Endoplasmic Reticulum/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Integrins/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Membrane Glycoproteins/chemistry , Mice , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Protein Structure, Tertiary , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptors/metabolism
20.
J Healthc Qual ; 38(1): 3-16, 2016.
Article in English | MEDLINE | ID: mdl-26730804

ABSTRACT

Hospitalized adult Medicaid recipients with chronic disease are at risk for rehospitalization within 90 days of discharge, but most research has focused on the Medicare population. The purpose of this study is to examine the impact of population-based care management intensity on inpatient readmissions in Medicaid adults with pre-existing chronic disease. Retrospective analyses of 2,868 index hospital admissions from 2012 New York State Medicaid Data Warehouse claims compared 90-day post-discharge utilization in populations with and without transitional care management interventions. High intensity managed care organization interventions were associated with higher outpatient and lower emergency department post-discharge utilization than low intensity fee-for-service management. However, readmission rates were higher for the managed care cases. Shorter time to readmission was associated with managed care, diagnoses that include heart and kidney failure, shorter length of stay for index hospitalization, and male sex; with no relationship to age. This unexpected result flags the need to re-evaluate readmission as a quality indicator in the complex Medicaid population. Quality improvement efforts should focus on care continuity during transitions and consider population-specific factors that influence readmission. Optimum post-discharge utilization in the Medicaid population requires a balance between outpatient, emergency and inpatient services to improve access and continuity.


Subject(s)
Chronic Disease/therapy , Hospitalization/statistics & numerical data , Medicaid/statistics & numerical data , Patient Discharge/statistics & numerical data , Patient Readmission/statistics & numerical data , Quality Improvement/organization & administration , Risk Management/methods , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Inpatients/statistics & numerical data , Male , Medicare/statistics & numerical data , Middle Aged , New York , Retrospective Studies , Sex Factors , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...