Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Commun Biol ; 6(1): 760, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37479733

ABSTRACT

Brain metastases is the most common intracranial tumor and account for approximately 20% of all systematic cancer cases. It is a leading cause of death in advanced-stage cancer, resulting in a five-year overall survival rate below 10%. Therefore, there is a critical need to identify effective biomarkers that can support frequent surveillance and promote efficient drug guidance in brain metastasis. Recently, the remarkable breakthroughs in single-cell RNA-sequencing (scRNA-seq) technology have advanced our insights into the tumor microenvironment (TME) at single-cell resolution, which offers the potential to unravel the metastasis-related cellular crosstalk and provides the potential for improving therapeutic effects mediated by multifaceted cellular interactions within TME. In this study, we have applied scRNA-seq and profiled 10,896 cells collected from five brain tumor tissue samples originating from breast and lung cancers. Our analysis reveals the presence of various intratumoral components, including tumor cells, fibroblasts, myeloid cells, stromal cells expressing neural stem cell markers, as well as minor populations of oligodendrocytes and T cells. Interestingly, distinct cellular compositions are observed across different samples, indicating the influence of diverse cellular interactions on the infiltration patterns within the TME. Importantly, we identify tumor-associated fibroblasts in both our in-house dataset and external scRNA-seq datasets. These fibroblasts exhibit high expression of type I collagen genes, dominate cell-cell interactions within the TME via the type I collagen signaling axis, and facilitate the remodeling of the TME to a collagen-I-rich extracellular matrix similar to the original TME at primary sites. Additionally, we observe M1 activation in native microglial cells and infiltrated macrophages, which may contribute to a proinflammatory TME and the upregulation of collagen type I expression in fibroblasts. Furthermore, tumor cell-specific receptors exhibit a significant association with patient survival in both brain metastasis and native glioblastoma cases. Taken together, our comprehensive analyses identify type I collagen-secreting tumor-associated fibroblasts as key mediators in metastatic brain tumors and uncover tumor receptors that are potentially associated with patient survival. These discoveries provide potential biomarkers for effective therapeutic targets and intervention strategies.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Collagen Type I , Brain , Fibroblasts , Tumor Microenvironment
2.
Front Oncol ; 11: 734959, 2021.
Article in English | MEDLINE | ID: mdl-34956864

ABSTRACT

BACKGROUND: Triggering receptor expressed on myeloid cells (TREM)-1 is a key mediator of innate immunity previously associated with the severity of inflammatory disorders, and more recently, the inferior survival of lung and liver cancer patients. Here, we investigated the prognostic impact and immunological correlates of TREM1 expression in breast tumors. METHODS: Breast tumor microarray and RNAseq expression profiles (n=4,364 tumors) were analyzed for associations between gene expression, tumor immune subtypes, distant metastasis-free survival (DMFS) and clinical response to neoadjuvant chemotherapy (NAC). Single-cell (sc)RNAseq was performed using the 10X Genomics platform. Statistical associations were assessed by logistic regression, Cox regression, Kaplan-Meier analysis, Spearman correlation, Student's t-test and Chi-square test. RESULTS: In pre-treatment biopsies, TREM1 and known TREM-1 inducible cytokines (IL1B, IL8) were discovered by a statistical ranking procedure as top genes for which high expression was associated with reduced response to NAC, but only in the context of immunologically "hot" tumors otherwise associated with a high NAC response rate. In surgical specimens, TREM1 expression varied among tumor molecular subtypes, with highest expression in the more aggressive subtypes (Basal-like, HER2-E). High TREM1 significantly and reproducibly associated with inferior distant metastasis-free survival (DMFS), independent of conventional prognostic markers. Notably, the association between high TREM1 and inferior DMFS was most prominent in the subset of immunogenic tumors that exhibited the immunologically hot phenotype and otherwise associated with superior DMFS. Further observations from bulk and single-cell RNAseq analyses indicated that TREM1 expression was significantly enriched in polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and M2-like macrophages, and correlated with downstream transcriptional targets of TREM-1 (IL8, IL-1B, IL6, MCP-1, SPP1, IL1RN, INHBA) which have been previously associated with pro-tumorigenic and immunosuppressive functions. CONCLUSIONS: Together, these findings indicate that increased TREM1 expression is prognostic of inferior breast cancer outcomes and may contribute to myeloid-mediated breast cancer progression and immune suppression.

3.
Front Immunol ; 11: 57, 2020.
Article in English | MEDLINE | ID: mdl-32117236

ABSTRACT

Background: Understanding how tumors subvert immune destruction is essential to the development of cancer immunotherapies. New evidence suggests that tumors limit anti-tumor immunity by exploiting transcriptional programs that regulate intratumoral trafficking and accumulation of effector cells. Here, we investigated the gene expression profiles that distinguish immunologically "cold" and "hot" tumors across diverse tumor types. Methods: RNAseq profiles of tumors (n = 8,920) representing 23 solid tumor types were analyzed using immune gene signatures that quantify CD8+ T cell abundance. Genes and pathways associated with a low CD8+ T cell infiltration profile (CD8-Low) were identified by correlation, differential expression, and statistical ranking methods. Gene subsets were evaluated in immunotherapy treatment cohorts and functionally characterized in cell lines and mouse tumor models. Results: Among different cancer types, we observed highly significant overlap of genes enriched in CD8-Low tumors, which included known immunomodulatory genes (e.g., BMP7, CMTM4, KDM5B, RCOR2) and exhibited significant associations with Wnt signaling, neurogenesis, cell-cell junctions, lipid biosynthesis, epidermal development, and cancer-testis antigens. Analysis of mutually exclusive gene clusters demonstrated that different transcriptional programs may converge on the T cell-cold phenotype as well as predict for response and survival of patients to Nivo treatment. Furthermore, we confirmed that a top-ranking candidate belonging to the TGF-ß superfamily, BMP7, negatively regulates CD8+ T cell abundance in immunocompetent murine tumor models, with and without anti-PD-L1 treatment. Conclusions: This study presents the first evidence that solid tumors of diverse anatomical origin acquire conserved transcriptional alterations that may be operative in the T cell-cold state. Our findings demonstrate the potential clinical utility of CD8-Low tumor-associated genes for predicting patient immunotherapy outcomes and point to novel mechanisms with potential for broad therapeutic exploitation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Transcriptome/immunology , Animals , Bone Morphogenetic Protein 7 , Cell Line , Co-Repressor Proteins/metabolism , Computational Biology , Female , Gene Regulatory Networks , Humans , Immunologic Factors , Immunotherapy , Mice , Mice, Inbred BALB C , Prognosis
4.
Cancer Med ; 8(6): 3072-3085, 2019 06.
Article in English | MEDLINE | ID: mdl-31033233

ABSTRACT

Tumor-infiltrating myeloid cells are the most abundant leukocyte population within tumors. Molecular cues from the tumor microenvironment promote the differentiation of immature myeloid cells toward an immunosuppressive phenotype. However, the in situ dynamics of the transcriptional reprogramming underlying this process are poorly understood. Therefore, we applied single cell RNA-seq (scRNA-seq) to computationally investigate the cellular composition and transcriptional dynamics of tumor and adjacent normal tissues from 4 early-stage non-small cell lung cancer (NSCLC) patients. Our scRNA-seq analyses identified 11 485 cells that varied in identity and gene expression traits between normal and tumor tissues. Among these, myeloid cell populations exhibited the most diverse changes between tumor and normal tissues, consistent with tumor-mediated reprogramming. Through trajectory analysis, we identified a differentiation path from CD14+ monocytes to M2 macrophages (monocyte-to-M2). This differentiation path was reproducible across patients, accompanied by increased expression of genes (eg, MRC1/CD206, MSR1/CD204, PPARG, TREM2) with significantly enriched functions (Oxidative phosphorylation and P53 pathway) and decreased expression of genes (eg, CXCL2, IL1B) with significantly enriched functions (TNF-α signaling via NF-κB and inflammatory response). Our analysis further identified a co-regulatory network implicating upstream transcription factors (JUN, NFKBIA) in monocyte-to-M2 differentiation, and activated ligand-receptor interactions (eg, SFTPA1-TLR2, ICAM1-ITGAM) suggesting intratumoral mechanisms whereby epithelial cells stimulate monocyte-to-M2 differentiation. Overall, our study identified the prevalent monocyte-to-M2 differentiation in NSCLC, accompanied by an intricate transcriptional reprogramming mediated by specific transcriptional activators and intercellular crosstalk involving ligand-receptor interactions.


Subject(s)
Cell Plasticity/genetics , Myeloid Cells/metabolism , RNA-Seq/methods , Humans , Signal Transduction , Tumor Microenvironment
5.
Sci Rep ; 5: 12981, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26257336

ABSTRACT

Tumor proliferative capacity is a major biological correlate of breast tumor metastatic potential. In this paper, we developed a systems approach to investigate associations among gene expression patterns, representative protein-protein interactions, and the potential for clinical metastases, to uncover novel survival-related subnetwork signatures as a function of tumor proliferative potential. Based on the statistical associations between gene expression patterns and patient outcomes, we identified three groups of survival prognostic subnetwork signatures (SPNs) corresponding to three proliferation levels. We discovered 8 SPNs in the high proliferation group, 8 SPNs in the intermediate proliferation group, and 6 SPNs in the low proliferation group. We observed little overlap of SPNs between the three proliferation groups. The enrichment analysis revealed that most SPNs were enriched in distinct signaling pathways and biological processes. The SPNs were validated on other cohorts of patients, and delivered high accuracy in the classification of metastatic vs non-metastatic breast tumors. Our findings indicate that certain biological networks underlying breast cancer metastasis differ in a proliferation-dependent manner. These networks, in combination, may form the basis of highly accurate prognostic classification models and may have clinical utility in guiding therapeutic options for patients.


Subject(s)
Algorithms , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Databases, Factual , Female , Humans , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Prognosis , Protein Interaction Maps , Transcriptome
6.
Mol Pharmacol ; 87(5): 792-802, 2015 May.
Article in English | MEDLINE | ID: mdl-25680754

ABSTRACT

The human α2C-adrenergic receptor (α2C-AR) is localized intracellularly at physiologic temperature. Decreasing the environmental temperature strongly stimulates the receptor transport to the cell surface. In contrast, rat and mouse α2C-AR plasma membrane levels are less sensitive to decrease in temperature, whereas the opossum α2C-AR cell surface levels are not changed in these conditions. Structural analysis demonstrated that human α2C-AR has a high number of arginine residues in the third intracellular loop and in the C-terminus, organized as putative RXR motifs. Although these motifs do not affect the receptor subcellular localization at 37°C, deletion of the arginine clusters significantly enhanced receptor plasma membrane levels at reduced temperature. We found that this exaggerated transport of the human receptor is mediated by two functional arginine clusters, one in the third intracellular loop and one in the C-terminus. This effect is mediated by interactions with COPI vesicles, but not by 14-3-3 proteins. In rat α2C-AR, the arginine cluster from the third intracellular loop is shifted to the left due to three missing residues. Reinsertion of these residues in the rat α2C-AR restored the same temperature sensitivity as in the human receptor. Proteomic and coimmunoprecipitation experiments identified pontin as a molecule having stronger interactions with human α2C-AR compared with rat α2C-AR. Inhibition of pontin activity enhanced human receptor plasma membrane levels and signaling at 37°C. Our results demonstrate that human α2C-AR has a unique temperature-sensitive traffic pattern within the G protein-coupled receptor class due to interactions with different molecular chaperones, mediated in part by strict spatial localization of specific arginine residues.


Subject(s)
Protein Transport/physiology , Receptors, Adrenergic, alpha-2/metabolism , Animals , Arginine/metabolism , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , HEK293 Cells , Humans , Proteomics/methods , Rats , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Temperature
7.
Int J Mol Sci ; 14(2): 3595-620, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23434660

ABSTRACT

Extracellular-signal regulated kinase (ERK) signaling is required for a multitude of physiological and patho-physiological processes. However, the identities of the proteins that ERK phosphorylates to elicit these responses are incompletely known. Using an affinity purification methodology of general utility, here we identify cytoplasmic dynein intermediate chain 2 (DYNC1I-2, IC-2) as a novel substrate for ERK following epidermal growth factor receptor stimulation of fibroblasts. IC-2 is a subunit of cytoplasmic dynein, a minus-end directed motor protein necessary for transport of diverse cargos along microtubules. Emerging data support the hypothesis that post-translational modification regulates dynein but the signaling mechanisms used are currently unknown. We find that ERK phosphorylates IC-2 on a novel, highly conserved Serine residue proximal to the binding site for the p150Glued subunit of the cargo adapter dynactin. Surprisingly, neither constitutive phosphorylation nor a phosphomimetic substitution of this Serine influences binding of p150Glued to IC-2. These data suggest that ERK phosphorylation of IC-2 regulates dynein function through mechanisms other than its interaction with dynactin.

8.
J Neurosci ; 32(44): 15495-510, 2012 Oct 31.
Article in English | MEDLINE | ID: mdl-23115187

ABSTRACT

The retrograde transport of Trk-containing endosomes from the axon to the cell body by cytoplasmic dynein is necessary for axonal and neuronal survival. We investigated the recruitment of dynein to signaling endosomes in rat embryonic neurons and PC12 cells. We identified a novel phosphoserine on the dynein intermediate chains (ICs), and we observed a time-dependent neurotrophin-stimulated increase in intermediate chain phosphorylation on this site in both cell types. Pharmacological studies, overexpression of constitutively active MAP kinase kinase, and an in vitro assay with recombinant proteins demonstrated that the intermediate chains are phosphorylated by the MAP kinase ERK1/2, extracellular signal-regulated kinase, a major downstream effector of Trk. Live cell imaging with fluorescently tagged IC mutants demonstrated that the dephosphomimic mutants had significantly reduced colocalization with Trk and Rab7, but not a mitochondrial marker. The phosphorylated intermediate chains were enriched on immunoaffinity-purified Trk-containing organelles. Inhibition of ERK reduced the amount of phospho-IC and the total amount of dynein that copurified with the signaling endosomes. In addition, inhibition of ERK1/2 reduced the motility of Rab7- and TrkB-containing endosomes and the extent of their colocalization with dynein in axons. NGF-dependent survival of sympathetic neurons was significantly reduced by the overexpression of the dephosphomimic mutant IC-1B-S80A, but not WT IC-1B, further demonstrating the functional significance of phosphorylation on this site. These results demonstrate that neurotrophin binding to Trk initiates the recruitment of cytoplasmic dynein to signaling endosomes through ERK1/2 phosphorylation of intermediate chains for their subsequent retrograde transport in axons.


Subject(s)
Axonal Transport/physiology , Cytoplasm/physiology , Dyneins/physiology , Endosomes/physiology , MAP Kinase Signaling System/physiology , Receptor, trkA/physiology , Animals , Blotting, Western , Cell Membrane/metabolism , Cell Membrane/physiology , Cell Survival/physiology , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , MAP Kinase Signaling System/genetics , Nerve Growth Factor/physiology , Nerve Growth Factors/pharmacology , Neurons/physiology , Organelles/physiology , PC12 Cells , Phosphorylation , Plasmids/genetics , RNA, Small Interfering/genetics , Rats , Signal Transduction/physiology , Transfection
9.
Exp Biol Med (Maywood) ; 237(1): 38-49, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22185919

ABSTRACT

The interferon-stimulated gene 15 (ISG15) pathway is highly elevated in breast cancer; however, very little is known about how the ISG15 pathway contributes to breast tumorigenesis. In the current study, using the gene disruption approach, we demonstrate that both ISG15 and UbcH8 (ISG15-specific conjugating enzyme) disrupt F-actin architecture and formation of focal adhesions in ZR-75-1 breast cancer cells. In addition, ISG15 and UbcH8 promote breast cancer cell migration. We also demonstrate that ISG15 inhibits ubiquitin/26S proteasome-mediated turnover of proteins implicated in tumor cell motility, invasion and metastasis. Together, our results suggest that the aberrant activation of the ISG15 pathway confers a motile phenotype to breast cancer cells by disrupting cell architecture and stabilizing proteins involved in cell motility, invasion and metastasis. Because the cellular architecture is conserved and the ISG15 pathway is constitutively activated in tumor cells of different lineages, it is reasonable to assume that our observations in breast cancer must hold true for many other tumors.


Subject(s)
Breast Neoplasms/metabolism , Cytokines/metabolism , Cytoskeleton/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitins/metabolism , Actins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cytokines/genetics , Cytoskeleton/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Interferons , Neoplasm Invasiveness , Neoplasm Metastasis , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/genetics
10.
PLoS One ; 6(6): e20710, 2011.
Article in English | MEDLINE | ID: mdl-21677780

ABSTRACT

RACK1 proteins belong to the eukaryote WD40-repeat protein family and function as spatial regulators of multiple cellular events, including signaling pathways, the cell cycle and translation. For this latter role, structural and genetic studies indicate that RACK1 associates with the ribosome through two conserved positively charged amino acids in its first WD40 domain. Unlike RACK1s, including Trypanosoma brucei RACK1 (TbRACK1), only one of these two positively-charged residues is conserved in the first WD40 domain of the Leishmania major RACK1 ortholog, LACK. We compared virulence-attenuated LACK single copy (LACK/-) L. major, with L. major expressing either two LACK copies (LACK/LACK), or one copy each of LACK and TbRACK1 (LACK/TbRACK1), to evaluate the function of these structurally distinct RACK1 orthologs with respect to translation, viability at host temperatures and pathogenesis. Our results indicate that although the ribosome-binding residues are not fully conserved in LACK, both LACK and TbRACK1 co-sedimented with monosomes and polysomes in LACK/LACK and LACK/TbRACK1 L. major, respectively. LACK/LACK and LACK/TbRACK1 strains differed in their sensitivity to translation inhibitors implying that minor sequence differences between the RACK1 proteins can alter their functional properties. While biochemically distinguishable, both LACK/LACK and LACK/TbRACK1 lines were more tolerant of elevated temperatures, resistant to translation inhibitors, and displayed robust pathogenesis in vivo, contrasting to LACK/- parasites.


Subject(s)
Antigens, Protozoan/metabolism , Leishmania major/physiology , Leishmania major/pathogenicity , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Cell Cycle/physiology , Female , Leishmania major/cytology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Polyribosomes/metabolism , Protein Biosynthesis , Protozoan Proteins/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Sequence Alignment , Temperature , Transcription, Genetic , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
11.
Mol Cell Biol ; 30(13): 3233-48, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439493

ABSTRACT

Cell migration is critical for normal development and for pathological processes including cancer cell metastasis. Dynamic remodeling of focal adhesions and the actin cytoskeleton are crucial determinants of cell motility. The Rho family and the mitogen-activated protein kinase (MAPK) module consisting of MEK-extracellular signal-regulated kinase (ERK) are important regulators of these processes, but mechanisms for the integration of these signals during spreading and motility are incompletely understood. Here we show that ERK activity is required for fibronectin-stimulated Rho-GTP loading, Rho-kinase function, and the maturation of focal adhesions in spreading cells. We identify p190A RhoGAP as a major target for ERK signaling in adhesion assembly and identify roles for ERK phosphorylation of the C terminus in p190A localization and activity. These observations reveal a novel role for ERK signaling in adhesion assembly in addition to its established role in adhesion disassembly.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions/metabolism , Repressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Butadienes/metabolism , Cell Adhesion/physiology , Cell Line , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Fibronectins/metabolism , Humans , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Data , Nitriles/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Sequence Alignment , Vinculin/metabolism , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics
12.
Cell Commun Signal ; 7: 26, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19930650

ABSTRACT

ERK signaling regulates focal adhesion disassembly during cell movement, and increased ERK signaling frequently contributes to enhanced motility of human tumor cells. We previously found that the ERK scaffold MEK Partner 1 (MP1) is required for focal adhesion disassembly in fibroblasts. Here we test the hypothesis that MP1-dependent ERK signaling regulates motility of DU145 prostate cancer cells. We find that MP1 is required for motility on fibronectin, but not for motility stimulated by serum or EGF. Surprisingly, MP1 appears not to function through its known binding partners MEK1 or PAK1, suggesting the existence of a novel pathway by which MP1 can regulate motility on fibronectin. MP1 may function by regulating the stability or expression of paxillin, a key regulator of motility.

13.
Cell Signal ; 19(8): 1621-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553668

ABSTRACT

Cell migration is critical for many physiological processes and is often misregulated in developmental disorders and pathological conditions including cancer and neurodegeneration. MAPK signaling and the Rho family of proteins are known regulators of cell migration that exert their influence on cellular cytoskeleton during cell adhesion and migration. Here we review data supporting the view that localized ERK signaling mediated through recently identified scaffold proteins may regulate cell migration.


Subject(s)
Cytoskeleton/physiology , Mitogen-Activated Protein Kinases/physiology , Signal Transduction , Actins/metabolism , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Humans , Models, Biological , rho GTP-Binding Proteins/physiology
14.
Am J Physiol Renal Physiol ; 292(5): F1501-12, 2007 May.
Article in English | MEDLINE | ID: mdl-17287198

ABSTRACT

Following a blood meal, the mosquito Aedes aegypti will have acquired an enormous sodium load that must be rapidly excreted to restore ion homeostasis. It is a process that demands robust sodium and fluid transport capabilities. Even though the identities of the components involved in this ion transport across the mosquito Malpighian tubule epithelia have not been completely determined, electrophysiological studies suggest the contribution of a Na(+)/H(+) exchanger extruding cations into the lumen driven secondarily by the proton gradient created by the V-type H(+)-ATPase in the tubules' apical membrane. We have identified the putative exchanger and designated it AeNHE8. Immunolocalization studies demonstrated that AeNHE8 is expressed in the apical membranes of Malpighian tubules, gastric caecae, and rectum. When heterologously expressed in salt-sensitive yeast cells lacking Na(+) extrusion and Na(+)/H(+) exchange proteins, AeNHE8 rescues the salt-sensitive phenotype and restores the cells' ability to grow in high NaCl media. Furthermore, heterologous expression of AeNHE8 in NHE-deficient fibroblast cells results in an amiloride-sensitive (22)Na(+) uptake. To determine the exchanger's kinetic properties, we reconstituted membranes from yeast cells expressing the protein into lipid proteoliposomes and assayed for cation-dependent H(+) exchange by fluorimetric methods. Our results indicate that AeNHE8 mediates saturable exchange of Na(+) and K(+) for H(+). We propose that AeNHE8 may be coupled to the inward H(+) gradient across the Malpighian tubules and plays a role in the extrusion of excess sodium and potassium while maintaining steady intracellular pH in the principal cells.


Subject(s)
Aedes/metabolism , Amiloride/pharmacology , Malpighian Tubules/metabolism , Potassium/metabolism , Proteolipids/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Hydrogen Exchangers/physiology , Sodium/metabolism , Aedes/growth & development , Amino Acid Sequence , Animals , Biological Transport/drug effects , Catalysis , Cell Line , Gene Transfer Techniques , Larva , Molecular Sequence Data , Phospholipids/metabolism , Saccharomyces cerevisiae , Sodium-Hydrogen Exchangers/drug effects , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/pharmacology
15.
J Exp Biol ; 209(Pt 18): 3529-44, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16943493

ABSTRACT

Transport across insect epithelia is thought to depend on the activity of a vacuolar-type proton ATPase (V-ATPase) that energizes ion transport through a secondary proton/cation exchanger. Although several of the subunits of the V-ATPase have been cloned, the molecular identity of the exchanger has not been elucidated. Here, we present the identification of sodium/proton exchanger isoform 3 (NHE3) from yellow fever mosquito, Aedes aegypti (AeNHE3). AeNHE3 localizes to the basal plasma membrane of Malpighian tubule, midgut and the ion-transporting sector of gastric caeca. Midgut expression of NHE3 shows a different pattern of enrichment between larval and adult stages, implicating it in the maintenance of regional pH in the midgut during the life cycle. In all tissues examined, NHE3 predominantly localizes to the basal membrane. In addition the limited expression in intracellular vesicles in the median Malpighian tubules may reflect a potential functional versatility of NHE3 in a tissue-specific manner. The localization of V-ATPase and NHE3, and exclusion of Na+/K+-ATPase from the distal ion-transporting sector of caeca, indicate that the role of NHE3 in ion and pH regulation is intricately associated with functions of V-ATPase. The AeNHE3 complements yeast mutants deficient in yeast NHEs, NHA1 and NHX1. To further examine the functional property of AeNHE3, we expressed it in NHE-deficient fibroblast cells. AeNHE3 expressing cells were capable of recovering intracellular pH following an acid load. The recovery was independent of the large cytoplasmic region of AeNHE3, implying this domain to be dispensable for NHE3 ion transport function. 22Na+ uptake studies indicated that AeNHE3 is relatively insensitive to amiloride and EIPA and is capable of Na+ transport in the absence of the cytoplasmic tail. Thus, the core domain containing the transmembrane regions of NHE3 is sufficient for pH recovery and ion transport. The present data facilitate refinement of the prevailing models of insect epithelial transport by incorporating basal amiloride-insensitive NHE3 as a critical mediator of transepithelial ion and fluid transport and likely in the maintenance of intracellular pH.


Subject(s)
Aedes/metabolism , Insect Proteins/physiology , Insect Vectors/metabolism , Sodium-Hydrogen Exchangers/physiology , Aedes/anatomy & histology , Aedes/genetics , Amiloride/analogs & derivatives , Amiloride/pharmacology , Amino Acid Sequence , Animals , Animals, Genetically Modified/metabolism , Cell Line , Cricetinae , Cricetulus , Genetic Complementation Test , Hydrogen-Ion Concentration , Immunohistochemistry , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Vectors/genetics , Ion Transport/drug effects , Ion Transport/genetics , Ion Transport/physiology , Larva/anatomy & histology , Larva/genetics , Larva/metabolism , Malpighian Tubules/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/genetics
16.
J Exp Biol ; 206(Pt 21): 3857-68, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14506221

ABSTRACT

Membrane transport in insect epithelia appears to be energized through proton-motive force generated by the vacuolar type proton ATPase (V-ATPase). However, secondary transport mechanisms that are coupled to V-ATPase activity have not been fully elucidated. Following a blood meal, the female mosquito regulates fluid and ion homeostasis through a series of characteristic behaviors that require brain-derived factors to regulate ion secretion. Despite the knowledge on the behaviors of the mosquito, little is known of the targets of several factors that have been implicated in cellular changes following a blood meal. This review discusses current models of membrane transport in insects and specific data on mosquito ion regulation together with the molecular aspects of membrane transport systems that are potentially linked to V-ATPase activity, which collectively determine the functioning of mosquito midgut and Malpighian tubules. Ion transport mechanisms will be discussed from a comparative physiology perspective to gain appreciation of the exquisite mechanisms of mosquito ion regulation.


Subject(s)
Culicidae/genetics , Malpighian Tubules/physiology , Models, Biological , Phylogeny , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Animals , Culicidae/physiology , Ion Transport , Malpighian Tubules/metabolism , Protein Conformation , Sodium-Potassium-Chloride Symporters/genetics , Vacuolar Proton-Translocating ATPases/metabolism
17.
J Biol Chem ; 277(52): 50355-64, 2002 Dec 27.
Article in English | MEDLINE | ID: mdl-12393871

ABSTRACT

The cisternal maturation model proposes that secretory proteins transit the Golgi in cisternae that mature by the continuous retrograde transport of Golgi enzymes in vesicles. We have tested the hypothesis that de novo generation of transport intermediates containing medial, trans, and trans Golgi network (TGN) enzymes is reconstituted in vitro. Our analysis shows that the majority of transport is mediated by a steady state of transport intermediate production and consumption by Golgi cisternae, with only a minor contribution of pre-existing transport intermediates. Transport in the medial and trans regions of the stack involved intermediates containing Golgi enzymes, apparently moving in a retrograde direction. In contrast, transport between the trans Golgi and TGN was exclusively mediated by intermediates containing secretory protein, as expected for anterograde transport. These intermediates may be physiologically relevant, because only these two specific types of intermediates can be detected in cell homogenates. By analogy to the coatomer (COPI)-independent transport of Golgi enzymes to the endoplasmic reticulum, the steady-state production of intra-Golgi transport intermediates was not impaired by inhibition of COPI vesicle formation. These data suggest a model for COPI-independent intra-Golgi transport by cisternal maturation with a shift in mechanism to anterograde transport at the trans Golgi and TGN boundary.


Subject(s)
ADP-Ribosylation Factors/metabolism , Coatomer Protein/metabolism , Golgi Apparatus/metabolism , Protein Transport , trans-Golgi Network/metabolism , Animals , CHO Cells , Cattle , Cricetinae , Kinetics
18.
J Exp Biol ; 205(Pt 7): 911-26, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11916988

ABSTRACT

The hexameric ATPase, N-ethylmaleimide-sensitive factor (NSF) is implicated in the release of neurotransmitters and in mediating fusion between intracellular membranes. Due to the conservation of proteins in constitutive and regulated membrane fusion reactions, NSF and its downstream targets have been predicted also to participate in fusion reactions underlying endocrine function, but there is little experimental evidence to support such a role for NSF in insect neuroendocrine secretion. Here we have characterized the NSF orthologue (MsNSF) from the endocrine model for development Manduca sexta. MsNSF is developmentally regulated in endocrine organs of the protocerebral complex. Enrichment of MsNSF in corpora cardiaca (CC) and not in corpora allata (CA) indicates that it might play a preferential role in releasing hormones produced in CC. Endocrine/paracrine cells of the enteric system in M. sexta exhibit selective MsNSF enrichment. Together the data point to a more selective participation of MsNSF in development of M. sexta by its involvement in a subset of factors, whereas other as-yet-unidentified homolog(s) might regulate secretion from CA and a large set of endocrine/paracrine cells. We further characterized the in vivo role of MsNSF by heterologous expression. In contrast to vertebrate NSF, MsNSF is functional in yeast membrane fusion in vivo. MsNSF rectifies defects in SEC18 (yeast NSF homologue) at nearly all discernible steps where Sec18p has been implicated in the biosynthetic route. This underscores the utility of our approach to delineate functional roles for proteins from systems that are not currently amenable to in vitro reconstitution.


Subject(s)
Carrier Proteins/physiology , Glycoproteins , Manduca/metabolism , Membrane Transport Proteins/metabolism , Neurosecretory Systems/metabolism , Saccharomyces cerevisiae Proteins , Vesicular Transport Proteins , Adenosine Triphosphatases/metabolism , Animals , Brain/metabolism , Carboxypeptidases/metabolism , Carrier Proteins/genetics , Cathepsin A , Corpora Allata/physiology , Enteric Nervous System/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation, Developmental , Genetic Complementation Test , Golgi Apparatus/metabolism , Heat-Shock Proteins/metabolism , Immunohistochemistry , Manduca/genetics , Manduca/growth & development , Membrane Fusion/physiology , Mutation , N-Ethylmaleimide-Sensitive Proteins , Neurosecretory Systems/cytology , Neurotransmitter Agents/metabolism , Optic Lobe, Nonmammalian/metabolism , Temperature , Vacuoles/metabolism , Yeasts/genetics
19.
J Biol Chem ; 277(16): 13863-72, 2002 Apr 19.
Article in English | MEDLINE | ID: mdl-11836242

ABSTRACT

Lipid rafts are characterized by their insolubility in nonionic detergents such as Triton X-100 at 4 degrees C. They have been studied in mammals, where they play critical roles in protein sorting and signal transduction. To understand the potential role of lipid rafts in lepidopteran insects, we isolated and analyzed the protein and lipid components of these lipid raft microdomains from the midgut epithelial membrane of Heliothis virescens and Manduca sexta. Like their mammalian counterparts, H. virescens and M. sexta lipid rafts are enriched in cholesterol, sphingolipids, and glycosylphosphatidylinositol-anchored proteins. In H. virescens and M. sexta, pretreatment of membranes with the cholesterol-depleting reagent saponin and methyl-beta-cyclodextrin differentially disrupted the formation of lipid rafts, indicating an important role for cholesterol in lepidopteran lipid rafts structure. We showed that several putative Bacillus thuringiensis Cry1A receptors, including the 120- and 170-kDa aminopeptidases from H. virescens and the 120-kDa aminopeptidase from M. sexta, were preferentially partitioned into lipid rafts. Additionally, the leucine aminopeptidase activity was enriched approximately 2-3-fold in these rafts compared with brush border membrane vesicles. We also demonstrated that Cry1A toxins were associated with lipid rafts, and that lipid raft integrity was essential for in vitro Cry1Ab pore forming activity. Our study strongly suggests that these microdomains might be involved in Cry1A toxin aggregation and pore formation.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins , Endotoxins/metabolism , Membrane Microdomains/chemistry , beta-Cyclodextrins , Animals , Bacillus thuringiensis Toxins , Biotinylation , Blotting, Western , Cholesterol/metabolism , Cyclodextrins/chemistry , Detergents/pharmacology , Electrophoresis, Polyacrylamide Gel , Hemolysin Proteins , Immunoblotting , Insecta , Leucine/chemistry , Lipid Metabolism , Lipids/chemistry , Manduca , Membrane Potentials , Octoxynol/pharmacology , Phosphatidylinositol Diacylglycerol-Lyase , Protein Binding , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Time Factors , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL