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










Publication year range
1.
Nat Commun ; 11(1): 4964, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009394

ABSTRACT

Thrombosis leads to platelet activation and subsequent degradation; therefore, replenishment of platelets from hematopoietic stem/progenitor cells (HSPCs) is needed to maintain the physiological level of circulating platelets. Platelet-derived microparticles (PMPs) are protein- and RNA-containing vesicles released from activated platelets. We hypothesized that factors carried by PMPs might influence the production of platelets from HSPCs, in a positive feedback fashion. Here we show that, during mouse acute liver injury, the density of megakaryocyte in the bone marrow increases following an increase in circulating PMPs, but without thrombopoietin (TPO) upregulation. In vitro, PMPs are internalized by HSPCs and drive them toward a megakaryocytic fate. Mechanistically, miR-1915-3p, a miRNA highly enriched in PMPs, is transported to target cells and suppresses the expression levels of Rho GTPase family member B, thereby inducing megakaryopoiesis. In addition, direct injection of PMPs into irradiated mice increases the number of megakaryocytes and platelets without affecting TPO levels. In conclusion, our data reveal that PMPs have a role in promoting megakaryocytic differentiation and platelet production.


Subject(s)
Blood Platelets/metabolism , Cell Differentiation , Cell-Derived Microparticles/metabolism , Megakaryocytes/cytology , MicroRNAs/metabolism , Animals , Base Sequence , Cell Line , Endocytosis , Gene Expression Profiling , Humans , Liver/injuries , Liver/pathology , Male , Megakaryocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Polyploidy , rhoB GTP-Binding Protein/metabolism
2.
Antib Ther ; 3(2): 146-154, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32671329

ABSTRACT

Although there are only two bispecific antibody (bsAb) drugs in the market, around 100 bsAb drug candidates are in clinical development. BsAbs have gained fast growing investment and attractions from the biopharmaceutical industry and academia in recent years. Antibody Engineering & Therapeutics 2019 (AET 2019) was held in San Diego, USA, from December 9th to 13th, 2019. This year's AET certainly reflected the trend. In this report, we selected eleven presentations from AET 2019 to highlight bsAbs' design and their potentials in cancer therapy. These presentations have discussed emerging strategies to improve bispecific antibody drugs in efficacy, safety, and production. As compared to CAR-Ts, some T cell-redirecting bsAbs may potentially achieve comparable efficacies with less side effects and toxicities, as evidenced with both preclinical and clinical data reviewed at the conference. Several approaches to reduce T cell engagers' toxicities including conditionally active bsAbs and IgM-based bsAbs were also presented and discussed at the conference. For the first time, The Antibody Society and the Chinese Antibody Society jointly held a special session at the AET.

3.
Antib Ther ; 2(4): 79-87, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31844838

ABSTRACT

The 15th Annual Protein Engineering Summit (PEGS) organized by Cambridge Healthtech Institute was held in Boston, USA from April 8th to 12th 2019. This report highlights the presentations in the Oncology Stream of this meeting with a focus on bispecific antibodies (BsAbs). A variety of BsAb formats with different target antigens (CD3, CTLA4, PD-1, PD-L1, EGFR, HER2, BCMA, CD19, CD20, CD38, CD123, TGFß, PSMA, etc.) have been discussed, in which the T-cell engaging (anti-CD3) BsAb is the most studied construct to exhibit promising immunotherapeutic activities. The BsAb formats include IgG-like structures or antibody fragments composed of antigen-binding sites only. Pre-clinical and clinical data from different BsAbs demonstrated the potential therapeutic applications in various solid tumors and hematological malignancies. The ongoing development of BsAb formats will help overcome current clinical issues, such as tumor selectivity and antigen coverage. This report also covers several presentations about emerging targets (e.g. mesothelin, CD47) and new technologies in the field of antibody engineering and therapeutics.

4.
Antib Ther ; 2(4): 99-107, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31942535

ABSTRACT

The Chinese Antibody Society (CAS) convened the third annual conference in Cambridge, Massachusetts, USA on April 7, 2019. More than 600 global members attended the meeting. The theme of this conference was Next-Generation Antibody Therapeutics: Discovery, Development and Beyond. The meeting covered a vast variety of topics including cancer immunotherapy, single-domain antibodies as well as bispecific antibodies, immunotoxins, transgenic mouse platforms for next-generation monoclonal antibody discovery and antibody chemistry, manufacturing and controls (CMCs). Two hot topics were comprehensively discussed by the prestigious panelists and hosts at the panel discussions during the conferences, i.e., bispecific antibodies and antibody CMC.

5.
Antib Ther ; 1(2): 65-74, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30406214

ABSTRACT

The Chinese Antibody Society (CAS) convened the second annual conference in Cambridge, MA, USA on 29 April 2018. More than 600 members from around the world attended the meeting. Invited speakers discussed the latest advancements in therapeutic antibodies with an emphasis on the progress made in China. The meeting covered a vast variety of topics including the current status of therapeutic antibodies, the progress of immuno-oncology, and biosimilars in China. The conference presentations also included the development of several novel antibodies such as antibodies related to weight loss, T-cell receptor-mimicking antibodies that target intracellular antigens, and tumor-targeting antibodies that utilize both innate and adaptive immune pathways. At the meeting, the CAS announced the launch of its official journal-Antibody Therapeutics-in collaboration with Oxford University Press. The conference was concluded by a panel discussion on how to bring a therapeutic drug developed in China to the USA for clinical trials.

6.
Oncotarget ; 8(58): 98051-98067, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228673

ABSTRACT

Forces within the surgical milieu or circulation activate cancer cell adhesion and potentiate metastasis through signaling requiring FAK-Akt1 interaction. Impeding FAK-Akt1 interaction might inhibit perioperative tumor dissemination, facilitating curative cancer surgery without global FAK or AKT inhibitor toxicity. Serial truncation and structurally designed mutants of FAK identified a seven amino acid, short helical structure within FAK that effectively competes with Akt1-FAK interaction. Adenoviral overexpression of this FAK-derived peptide inhibited pressure-induced FAK phosphorylation and AKT-FAK coimmunoprecipitation in human SW620 colon cancer cells briefly exposed to 15mmHg increased pressure, consistent with laparoscopic or post-surgical pressures. Adenoviral FAK-derived peptide expression prevented pressure-activation of SW620 adhesion not only to collagen-I-coated plates but also to murine surgical wounds. A scrambled peptide did not. Finally, we modeled operative shedding of tumor cells before irrigation and closure by transient cancer cell adhesion to murine surgical wounds before irrigation and closure. Thirty minute preincubation of SW620 cells at 15mmHg increased pressure impaired subsequent tumor free survival in mice exposed to cells expressing the scrambled peptide. The FAK-derived sequence prevented this. These results suggest that blocking FAK-Akt1 interaction may prevent perioperative tumor dissemination and that analogs or mimics of this 7 amino acid FAK-derived peptide could impair metastasis.

7.
Biophys J ; 105(1): 40-7, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23823222

ABSTRACT

Integrins are dynamic transmembrane cation-dependent heterodimers that both anchor cells in position and transduce signals into and out of cells. We used an atomic force microscope (AFM)-based nanorobotic system to measure integrin-binding forces in intact human intestinal epithelial Caco-2 cells. The AFM-based nanorobot enables human-directed, high-accuracy probe positioning and site-specific investigations. Functionalizing the AFM probe with an arginine-glycine-aspartate (RGD)-containing sequence (consensus binding sequence for integrins) allowed us to detect a series of peptide-cell membrane interactions with a median binding force of 115.1 ± 4.9 pN that were not detected in control interactions. Chelating divalent cations from the culture medium abolished these interactions, as did inhibiting intracellular focal adhesion kinase (FAK) using Y15. Adding 1 mM Mg(2+) to the medium caused a rightward shift in the force-binding curve. Adding 1 mM Ca(2+) virtually abolished the RGD-membrane specific interactions and blocked the Mg(2+) effects. Cell adhesion assays demonstrated parallel effects of divalent cations and the FAK inhibitor on cell adhesion. These results demonstrate direct modulation of integrin-binding affinity by both divalent cations and intracellular signal inhibition. Additionally, three binding states (nonspecific, specific inactivated, and specific activated) were delineated from affinity measurements. Although other research has assumed that this process of integrin conformational change causes altered ligand binding, in this work we directly measured these three states in individual integrins in a physiologically based study.


Subject(s)
Integrins/metabolism , Intracellular Space/metabolism , Microscopy, Atomic Force/methods , Nanotechnology/methods , Robotics , Caco-2 Cells , Humans , Intracellular Space/enzymology , Oligopeptides/metabolism , Protein Binding
8.
Anticancer Agents Med Chem ; 11(10): 993-1002, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22023045

ABSTRACT

AKT (or protein kinase B) and focal adhesion kinase (FAK) are two important kinases that regulate various cellular functions. Each is overexpressed and/or aberrantly activated in diverse cancers. Several small molecular inhibitors targeting either AKT or FAK are in development or in clinical trials. It is well established that FAK is an upstream regulator of AKT signaling pathway in various cancer cell lines and in xenograft tumor models. However, very recent reports from our laboratory and others demonstrate that AKT can also directly regulate FAK through direct association and serine phosphorylation. This indicates that AKT and FAK may be dual therapeutic targets for pharmacologic intervention in the treatment of primary and metastatic cancer. FAK-AKT interaction is particularly critical for metastatic adhesion. We review recent developments in AKT and FAK signaling in cancer with the particular emphasis on the novel signaling pathways in which FAK is downstream of AKT. We also provide an update on inhibitors targeting AKT or FAK currently in clinical trials.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Humans , Neoplasms/pathology
9.
Am J Physiol Cell Physiol ; 300(3): C657-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209368

ABSTRACT

Although focal adhesion kinase (FAK) is typically considered upstream of Akt, extracellular pressure stimulates cancer cell adhesion via Akt-dependent FAK activation. How Akt regulates FAK is unknown. We studied Akt-FAK interaction in colon cancer cells under 15 mmHg increased extracellular pressure. Pressure enhanced Akt-FAK association, blocked by inhibiting FAK or silencing Akt1 but not Akt2, and stimulated FAK serine phosphorylation in Caco-2 and human colon cancer cells from surgical specimens Akt1-dependently. FAK includes three serine (S517/601/695) and one threonine (T600)-containing consensus sequences for Akt phosphorylation. Studying S->A nonphosphorylatable point mutants suggests that these sites coordinately upregulate FAK Y397 tyrosine phosphorylation, which conventionally initiates FAK activation, and mediate pressure-induced cancer cell adhesion. FAK(T600A) mutation did not prevent pressure-induced FAK(Y397) phosphorylation or adhesion. Akt1 appeared to directly bind FAK, and this binding did not depend on the FAK autophosphorylation site (Y397). In addition, our results demonstrated that Akt phosphorylated FAK at three novel serine phosphorylation sites, which were also not required for FAK-Akt binding. This novel interaction suggests that FAK and Akt may be dual kinase targets to prevent cancer cell adhesion and metastasis.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Neoplasm Metastasis/pathology , Pressure/adverse effects , Proto-Oncogene Proteins c-akt/physiology , Serine/metabolism , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Amino Acid Sequence , Caco-2 Cells , Catalytic Domain/genetics , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , Focal Adhesion Protein-Tyrosine Kinases/physiology , Gene Silencing/physiology , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/prevention & control , Phosphorylation/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-akt/metabolism , Serine/genetics
10.
Am J Surg ; 200(5): 610-4, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21056138

ABSTRACT

BACKGROUND: The effect of extracellular pressure on adhesion and adhesiogenic focal adhesion kinase (FAK) and Akt signaling in sarcomas was investigated. METHODS: Human sarcoma cells (HT-1080 fibrosarcoma, KHOS-240S osteosarcoma, and A-673 rhabdomyosarcoma) were subjected to increased pressure followed by adhesion assay. Two cell lines were pretreated with the FAK inhibitor 1,2,4,5-benzenetetraamine tetrahydrochloride (Y15) or Akt IV inhibitor, followed by Western analysis for activated FAK and Akt. Parallel studies were conducted in cells from a resected human fibrous histiosarcoma. RESULTS: Pressure increased adhesion in all 3 sarcoma lines and primary histosarcoma cells by 7% to 18% (n = 6; P < .01 each). Pressure activated FAK and Akt (n = 5; P < .01). Inhibiting FAK or Akt inhibited FAK or Akt phosphorylation and the stimulation of adhesion by increased pressure (n = 5 each; P < .01 each). CONCLUSIONS: Pressure increases sarcoma cell adhesiveness via Akt and FAK. Perioperative manipulation or forces in lymphatic or circulatory systems may potentiate local recurrence or distant metastasis.


Subject(s)
Enzyme Activation/physiology , Extracellular Space/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/enzymology , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Densitometry , Humans , Phosphorylation , Pressure , Sarcoma/pathology , Signal Transduction
11.
J Invest Surg ; 23(6): 303-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21208094

ABSTRACT

Sarcomas are rare tumors, and malignant fibrous histiocytoma (MFH) is the most common soft tissue sarcoma found in adults. Increasing evidence suggests a possible role for AKT activation in soft tissue sarcoma. In the present study, we established a primary human MFH cell line (named MSUMFH cells) from a fresh surgically resected MFH tumor. These cells morphologically resembled human normal fibroblasts that are the presumptive cells of origin of MFH tumor cells. As there is, unfortunately, no standard marker other than morphology to identify MFH at the cellular level, we compared MSUMFH cells to primary nonmalignant fibroblasts and the primary tumor specimen to characterize its signaling. AKT was hyperactivated in both the MSUMFH cell line and original primary MFH tumor cells compared to normal fibroblasts. The AKT hyperactivity in the MSUMFH cell line was not accompanied by activation of focal adhesion kinase (FAK) or downregulated expression of PTEN, each of which is a putative upstream regulator of AKT. In contrast, this AKT hyperactivation required PI-3K and Src in MSUMFH cells. This PI-3K and Src-dependent AKT-activated MSUMFH cell line that we established in this study may be beneficial for the future cell-based study of MFH biology.


Subject(s)
Histiocytoma, Malignant Fibrous/physiopathology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction , Histiocytoma, Malignant Fibrous/pathology , Humans , Oncogene Protein pp60(v-src)/physiology , Phosphatidylinositol 3-Kinases/metabolism
12.
Cell Oncol ; 31(4): 273-89, 2009.
Article in English | MEDLINE | ID: mdl-19633364

ABSTRACT

Cell adhesion is important in cancer metastasis. Malignant cells in cancer patients may be exposed to physical forces such as extracellular pressure and shear, that stimulate their adhesion to matrix proteins, endothelium and surgical wounds. Pressure induces phosphorylation of AKT and focal adhesion kinase (FAK), which are required for pressure-stimulated cancer cell adhesion, but what mediates this effect is unknown. ILK may influence cell adhesion and FAK and AKT phosphorylation in other settings. We therefore hypothesized that ILK might also regulate pressure-stimulated cancer cell adhesion through AKT and FAK phosphorylation. Silencing ILK by siRNA reduced basal cancer cell adhesion and prevented the stimulation of adhesion by pressure. ILK mediated pressure-stimulated adhesion through specifically regulating phosphorylation of AKT at Ser473 and FAK at Tyr397 and 576 as well as ILK association with FAK and AKT. The siRNA-mediated loss of function of ILK in regulating increase in adhesion by pressure was not rescued by overexpression of alpha-parvin, an important ILK binding partner, although pressure promoted ILK-alpha-parvin association and translocated both ILK and alpha-parvin from cytosol to membrane/cytoskeleton. ILK may be a key mediator of mechanotransduced signals in cancer cells and an important therapeutic target to inhibit metastatic cancer cell adhesion.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mechanotransduction, Cellular , Neoplasms/physiopathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Adhesion , Cell Line, Tumor , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Neoplasms/chemistry , Neoplasms/enzymology , Phosphorylation , Pressure , Protein Binding , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
13.
Am J Physiol Gastrointest Liver Physiol ; 296(4): G876-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19179620

ABSTRACT

Repetitive strain stimulates intestinal epithelial migration across fibronectin via focal adhesion kinase (FAK), Src, and extracellular signal-related kinase (ERK) although how these signals act and interact remains unclear. We hypothesized that PI3K is central to this pathway. We subjected Caco-2 and intestinal epithelial cell-6 cells to 10 cycles/min deformation on flexible fibronectin-coated membranes, assayed migration by wound closure, and signaling by immunoblots. Strain stimulated PI3K, AKT, glycogen synthase kinase (GSK), and p38 phosphorylation. Blocking each kinase prevented strain stimulation of migration. Blocking PI3K prevented strain-stimulated ERK and p38 phosphorylation. Blocking AKT did not. Downstream, blocking PI3K, AKT, or ERK inhibited strain-induced GSK-Ser9 phosphorylation. Upstream of AKT, reducing FAK or Rac1 by siRNA blocked strain-stimulated AKT phosphorylation, but inhibiting Src by PP2 or siRNA did not. Transfection with FAK point mutants at Tyr397, Tyr576/577, or Tyr925 demonstrated that only FAK925 phosphorylation is required for strain-stimulated AKT phosphorylation. Myosin light chain activation by strain required FAK, Rac1, PI3K, AKT, GSK, and ERK but not Src or p38. Finally, blebbistatin, a nonmuscle myosin II inhibitor, blocked the motogenic effect of strain downstream of myosin light chain. Thus strain stimulates intestinal epithelial migration across fibronectin by a complex pathway including Src, FAK, Rac1, PI3K, AKT, GSK, ERK, p38, myosin light chain, and myosin II.


Subject(s)
Cell Movement/physiology , Epithelial Cells/physiology , Fibronectins/physiology , Intestinal Mucosa/cytology , Animals , Cell Line , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glycogen Synthase Kinases/antagonists & inhibitors , Glycogen Synthase Kinases/metabolism , Humans , Mechanotransduction, Cellular , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Biol Chem ; 284(4): 2001-11, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19047055

ABSTRACT

The intestinal epithelium is repetitively deformed by shear, peristalsis, and villous motility. Such repetitive deformation stimulates the proliferation of intestinal epithelial cells on collagen or laminin substrates via ERK, but the upstream mediators of this effect are poorly understood. We hypothesized that the phosphatidylinositol 3-kinase (PI3K)/AKT cascade mediates this mitogenic effect. PI3K, AKT, and glycogen synthase kinase-3beta (GSK-3beta) were phosphorylated by 10 cycles/min strain at an average 10% deformation, and pharmacologic blockade of these molecules or reduction by small interfering RNA (siRNA) prevented the mitogenic effect of strain in Caco-2 or IEC-6 intestinal epithelial cells. Strain MAPK activation required PI3K but not AKT. AKT isoform-specific siRNA transfection demonstrated that AKT2 but not AKT1 is required for GSK-3beta phosphorylation and the strain mitogenic effect. Furthermore, overexpression of AKT1 or an AKT chimera including the PH domain and hinge region of AKT2 and the catalytic domain and C-tail of AKT1 prevented strain activation of GSK-3beta, but overexpression of AKT2 or a chimera including the PH domain and hinge region of AKT1 and the catalytic domain and C-tail of AKT2 did not. These data delineate a role for PI3K, AKT2, and GSK-3beta in the mitogenic effect of strain. PI3K is required for both ERK and AKT2 activation, whereas AKT2 is sequentially required for GSK-3beta. Furthermore, AKT2 specificity requires its catalytic domain and tail region. Manipulating this pathway may prevent mucosal atrophy and maintain the mucosal barrier in conditions such as ileus, sepsis, and prolonged fasting when peristalsis and villous motility are decreased and the mucosal barrier fails.


Subject(s)
Glycogen Synthase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stress, Physiological/physiology , Cell Line , Cell Proliferation , Collagen/metabolism , Glycogen Synthase Kinases/antagonists & inhibitors , Humans , Isoenzymes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Myosins/antagonists & inhibitors , Myosins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Stress, Mechanical , src-Family Kinases/metabolism
15.
Exp Cell Res ; 314(2): 286-96, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17825284

ABSTRACT

Cell adhesion is a critical step in cancer metastasis, activated by extracellular forces such as pressure and shear. Reducing AKT1, but not AKT2, ablates the increase in cancer cell adhesion associated with 15 mm Hg increased extracellular pressure. To identify the determinants of this AKT isoform specificity, we exchanged the pleckstrin homology (PH) domains and/or hinge regions of AKT1 and AKT2. Wild type isoforms or these chimeras were overexpressed in Caco-2 cells in the absence or presence of isoform-specific siRNA to suppress endogenous AKT1. Pressure-induced AKT translocation and phosphorylation to the membrane were compared, along with the stimulation of cell adhesion by pressure. Pressure stimulated translocation of AKT1, but not AKT2 to the plasma membrane. Among our chimeras, only the chimeric AKT2 (chimera2), in which both the AKT2 PH domain and hinge region had been replaced by those of AKT1, translocated to the membrane in response to pressure. Similarly, only chimera2 rescued the function of AKT1 in mediating pressure-stimulated adhesion after endogenous AKT1 had been reduced. Pressure also promoted phosphorylation of AKT1 but not AKT2, and expression of a nonphosphorylatable double point mutant prevented pressure-stimulated adhesion. Among the chimeras, pressure promoted only chimera2 phosphorylation. These results identify the AKT1 PH domain and hinge region as functional domains which jointly permit AKT1 translocation and phosphorylation in response to extracellular pressure and distinguish determine the specificity of AKT1 in mediating the effects of extracellular pressure on cancer cell adhesion. These may be useful targets for interventions to inhibit metastasis.


Subject(s)
Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/physiology , Caco-2 Cells , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Chimera/genetics , Chimera/metabolism , Humans , Models, Biological , Phosphatidylinositols/pharmacology , Phosphorylation , Pressure , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/metabolism , Serine/genetics , Serine/metabolism
16.
J Fluoresc ; 17(2): 215-21, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17279335

ABSTRACT

Agkisacutacin isolated from the venom of Agkistrodon acutus is a coagulation factor IX / coagulation factor X-binding protein with marked anticoagulant- and platelet-modulating activities. Ca(2+) ion-induced stabilization and refolding of Agkisacutacin have been studied by following fluorescent measurements. Ca(2+) ions not only increase the structural stability of agkisacutacin against GdnHCl denaturation, but also induce its refolding. The GdnHCl-induced unfolding of the apo-agkisacutacin and the purified agkisacutacin is a single-step process with no detectable intermediate state. Ca(2+) ions play an important role in the stabilization of the structure of agkisacutacin. Ca(2+)-stabilized agkisacutacin exhibits higher resistance to GdnHCl denaturation than the apo-agkisacutacin. It is possible to induce refolding of the unfolded apo-agkisacutacin merely by adding 1 mM Ca(2+) ions without changing the concentration of the denaturant. The kinetic result of Ca(2+)-induced refolding provides evidences for that agkisacutacin consists of at least two refolding phases and the first phase of Ca(2+)-induced refolding should involve the formation of the compact Ca(2+)-binding site regions, and subsequently, the protein undergoes further conformational rearrangements to form the native structure.


Subject(s)
Agkistrodon/metabolism , Calcium/chemistry , Crotalid Venoms/chemistry , Animals , Cations, Divalent/chemistry , Protein Denaturation , Protein Folding , Spectrometry, Fluorescence
17.
Biochem Biophys Res Commun ; 345(2): 803-8, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16701558

ABSTRACT

The oxidative stress induced by photodynamic therapy using the phthalocyanine Pc 4 (PDT) can lead to apoptosis, and is accompanied by photodamage to Bcl-2 and accumulation of de novo ceramide. Similar to PDT, the oxidative stress inducer and Bcl-2 inhibitor HA14-1 triggers apoptosis. To test the specificity of the ceramide response, Jurkat cells were exposed to an equitoxic dose of HA14-1. Unlike PDT, HA14-1 did not induce accumulation of de novo ceramide, although levels of sphingomyelin, phosphatidylserine and phosphatidylethanolamine were below control values after either treatment. In contrast to PDT, (i) the transient inhibition of serine palmitoyltransferase induced by HA14-1 was associated with the initial decrease in de novo ceramide, and (ii) HA14-1-initiated inhibition of sphingomyelin synthase and glucosylceramide synthase did not result in accumulation of de novo ceramide. These results show that the ceramide response to PDT is not induced by another pro-apoptotic stimulus, and may be unique to PDT as described here.


Subject(s)
Apoptosis/drug effects , Benzopyrans/pharmacology , Ceramides/metabolism , Nitriles/pharmacology , Photochemotherapy/adverse effects , Apoptosis/radiation effects , Cells, Cultured , Glucosyltransferases/pharmacology , Humans , Jurkat Cells , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Serine C-Palmitoyltransferase/pharmacology , Sphingomyelins/metabolism , Time Factors , Transferases (Other Substituted Phosphate Groups)/pharmacology
18.
Article in English | MEDLINE | ID: mdl-12174265

ABSTRACT

It has been determined that each FP molecular contains one Ca(2+)-binding site. By the use of fluorescence probe Tb(3+), the distance between Tb(3+) and tryptophan (Trp) residue was obtained to be 0.375. Tb(3+) ion is coordinated with FP more strongly than Ca(2+) ion, and can bind to FP and replace the Ca(2+) ion in FP completely.

19.
Article in English | MEDLINE | ID: mdl-12174266

ABSTRACT

The conformation and the properties of the fibrinolytic principle (FP) from Agkistrodon acutus venom were studied by chemical modification and fluorescence spectroscopy. Results showed that there are more than one tryptophan (Trp) residue in the FP molecule and they are located in the more hydrophobic core, could be quenched by acrylamide (Acr), a polarized quencher without electric charge. The collisional quenching constants of FP at different concentrations of Acr were calculated in terms of Stern-Volmer equation, and the fraction of the Trp quenched was obtained by the modified Stern-Volmer equation as 83%.

SELECTION OF CITATIONS
SEARCH DETAIL
...