Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Med Oncol ; 38(8): 96, 2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34273028

ABSTRACT

Targeted cancer therapies based on overexpressed receptors and the fractions containing immunotoxins and bacterial metabolites are one of the well-known methods to overcome the chemotherapy resistance of cancer cells. In this paper, we designed ARA-linker-TGFαL3, using Arazyme, a Serratia proteamaculans metabolite, and a third loop segment of TGFα to target EGFR-expressing breast cancer cells. After cloning in pET28a (+), the expression of recombinant protein was optimized in Escherichia coli strain BL21 (DE3). MDA-MB-468 (EGFR positive) and MDA-MB-453 (EGFR negative) breast cancer cell lines were employed. Also, the chemotherapeutic drug, Taxotere (Docetaxel), was employed to compare cytotoxicity effects. Cell ELISA assessed the binding affinity of recombinant proteins to the receptor, and the cytotoxicity was detected by MTT and lactate dehydrogenase release assays. The interfacing with cancer cell adhesion was evaluated. Furthermore, the induction of apoptosis was examined utilizing flow cytometric analysis, and caspase-3 activity assay. Moreover, RT-PCR was conducted to study the expression of apoptosis (bax, bcl2, and casp3), angiogenesis (vegfr2), and metastasis (mmp2 and mmp9) genes. ARA-linker-TGFαL3 revealed a higher binding affinity, cytotoxicity, and early apoptosis induction in MDA-MB-468 cells compared to the effects of Arazyme while both recombinant proteins showed similar effects on MDA-MB-453. In addition, the Taxotere caused the highest cytotoxicity on cancer cells through induction of late apoptosis. Meanwhile, the expression of angiogenesis and metastasis genes was decreased in both cell lines after treatment with either ARA-linker-TGFαL3 or Arazyme. Our in vitro results indicated the therapeutic effect of ARA-linker-TGFαL3 on breast cancer cells.


Subject(s)
Breast Neoplasms , Multidrug Resistance-Associated Proteins , Transforming Growth Factor alpha , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Multidrug Resistance-Associated Proteins/administration & dosage , Multidrug Resistance-Associated Proteins/metabolism , Protein Binding/physiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/metabolism
2.
Biochem Pharmacol ; 155: 510-523, 2018 09.
Article in English | MEDLINE | ID: mdl-30059675

ABSTRACT

TRAIL, a promising antitumor immuno-agent, exerted limited efficacy in clinical trials. The third disulfide loop of TGF-α (TGF3L peptide) with a very low affinity for EGFR has been reported to enhance the activity of fused antigens or cytokines. We wondered whether fusion of this peptide could enhance TRAIL activity and what the underlying mechanism for this enhancement would be. The TGF3L-TRAIL showed greatly enhanced cytotoxicity in a variety of cancer cell lines while spared normal cells unharmed. Typical apoptosis and cellular caspase activation were potently induced by TGF3L-TRAIL at the concentration levels corresponding to its cytotoxicity. TGF3L-TRAIL was able to activate both DR4 and DR5 the same as TRAIL did. It induced complete cell death in Colo205 through only one receptor when the other one was blocked, different from TRAIL-induced cell death (through DR4 dominantly). TGF3L-TRAIL cytotoxicity was not reduced in some cell lines even if both receptors are blocked simultaneously. Surprisingly, TGF3L-TRAIL self-assembled into stable polymers, which was responsible for its enhanced cytotoxicity. In human tumor xenograft mouse models, TGF3L-TRAIL showed anti-tumor activity similar to or better than TRAIL in different cancer cell types, consistent with its differing enhancement of cytotoxicity in vitro. Taken together, TGF3L fusion of TRAIL obviously enhances the anticancer activity of TRAIL by promoting assembly into polymers, which presents a novel fusion strategy for improving TRAIL function.


Subject(s)
Antineoplastic Agents/metabolism , Drug Carriers/metabolism , Polymers/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transforming Growth Factor alpha/metabolism , Tumor Burden/drug effects , Amino Acid Sequence , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , HeLa Cells , Hep G2 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Polymers/administration & dosage , Polymers/chemistry , Random Allocation , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/chemistry , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/chemistry , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
3.
J Exp Med ; 209(6): 1105-19, 2012 Jun 04.
Article in English | MEDLINE | ID: mdl-22565824

ABSTRACT

ADAM17 (a disintegrin and metalloproteinase 17) is ubiquitously expressed and cleaves membrane proteins, such as epidermal growth factor receptor (EGFR) ligands, l-selectin, and TNF, from the cell surface, thus regulating responses to tissue injury and inflammation. However, little is currently known about its role in skin homeostasis. We show that mice lacking ADAM17 in keratinocytes (A17(ΔKC)) have a normal epidermal barrier and skin architecture at birth but develop pronounced defects in epidermal barrier integrity soon after birth and develop chronic dermatitis as adults. The dysregulated expression of epidermal differentiation proteins becomes evident 2 d after birth, followed by reduced transglutaminase (TGM) activity, transepidermal water loss, up-regulation of the proinflammatory cytokine IL-36α, and inflammatory immune cell infiltration. Activation of the EGFR was strongly reduced in A17(ΔKC) skin, and topical treatment of A17(ΔKC) mice with recombinant TGF-α significantly improved TGM activity and decreased skin inflammation. Finally, we show that mice lacking the EGFR in keratinocytes (Egfr(ΔKC)) closely resembled A17(ΔKC) mice. Collectively, these results identify a previously unappreciated critical role of the ADAM17-EGFR signaling axis in maintaining the homeostasis of the postnatal epidermal barrier and suggest that this pathway could represent a good target for treatment of epidermal barrier defects.


Subject(s)
ADAM Proteins/metabolism , Cell Differentiation/physiology , ErbB Receptors/metabolism , Keratinocytes/cytology , Skin/cytology , ADAM Proteins/genetics , ADAM17 Protein , Administration, Topical , Animals , Animals, Newborn , Dermatitis, Atopic/pathology , Epidermal Cells , Epidermis/metabolism , Epidermis/pathology , ErbB Receptors/genetics , Gene Expression Regulation, Developmental , Interleukin-1/metabolism , Keratinocytes/metabolism , Macrophages/pathology , Mice , Mice, Mutant Strains , Skin/growth & development , Skin/metabolism , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/pharmacology , Transglutaminases/metabolism
4.
Surgery ; 150(2): 339-46, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21801969

ABSTRACT

BACKGROUND: The activation of the epidermal growth factor family of receptors may improve cardiac protection after injury. One epidermal growth factor family ligand, transforming growth factor-alpha, promotes wound healing in multiple tissues in response to oxidative injury and might confer resistance to myocardial depressant factors, although the role of transforming growth factor-alpha in myocardial ischemia/reperfusion injury is unknown. We hypothesized that preischemic infusion of transforming growth factor-alpha would improve myocardial functional recovery after acute ischemia/reperfusion. METHODS: The hearts from adult male rats were isolated and perfused via the Langendorff model. Immediately prior to ischemia, the hearts received an intracoronary infusion of either vehicle or transforming growth factor-alpha (1 ng, 10 ng, or 100 ng). After reperfusion, the hearts were assessed for activation of the prosurvival pathway, Akt. RESULTS: Infusion of transforming growth factor-alpha did not confer any additional functional protection compared with the vehicle, but myocardial tissue analysis revealed significantly increased activation of the Akt pathway in both the 10-ng and 100-ng groups. CONCLUSION: Preischemic infusion of transforming growth factor-alpha does not improve myocardial functional recovery after ischemia/reperfusion injury. Whereas transforming growth factor-alpha treatment does affect actions at the molecular level, these actions do not translate into an observable functional effect. This lack of improvement may point to a relative unimportance of transforming growth factor-alpha in myocardial signaling compared with other epidermal growth factor ligands.


Subject(s)
Antioxidants/administration & dosage , Myocardial Reperfusion Injury/prevention & control , Transforming Growth Factor alpha/administration & dosage , Animals , Disease Models, Animal , Male , Perfusion , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
5.
J Stroke Cerebrovasc Dis ; 19(1): 3-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20123220

ABSTRACT

We previously demonstrated that infusion of transforming growth factor (TGF)-alpha after chronic middle cerebral artery occlusion (MCAO) stimulates stem and progenitor cell proliferation, migration, and neuronal differentiation associated with the amelioration of neurologic impairment. But the use of TGF-alpha in humans is impeded by impracticality of intracranial infusion and the inability of intravenous TGF-alpha to cross the blood-brain barrier. Here we investigated whether intranasal delivery of PEGylated TGF-alpha (PEG-TGF-alpha) is a viable alternative. We found that intranasal PEG-TGF-alpha can also induce the proliferation of neural progenitors and their migration to the damaged striatum, and that this is associated with significant behavioral improvement in the MCAO model. This nonsurgical approach represents a potential therapeutic strategy for human patients.


Subject(s)
Behavior, Animal/drug effects , Drug Carriers , Infarction, Middle Cerebral Artery/drug therapy , Motor Activity/drug effects , Neurons/drug effects , Polyethylene Glycols/chemistry , Transforming Growth Factor alpha/administration & dosage , Administration, Intranasal , Animals , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Neurogenesis/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function , Stem Cells/drug effects , Stem Cells/pathology , Time Factors , Transforming Growth Factor alpha/chemistry
6.
Neurotherapeutics ; 6(3): 539-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19560743

ABSTRACT

Treatment of malignant gliomas represents one of the most formidable challenges in oncology. Despite treatment with surgery, radiation therapy, and chemotherapy, the prognosis remains poor, particularly for glioblastoma, which has a median survival of 12 to 15 months. An important impediment to finding effective treatments for malignant gliomas is the presence of the blood brain barrier, which serves to prevent delivery of potentially active therapeutic compounds. Multiple efforts are focused on developing strategies to effectively deliver active drugs to brain tumor cells. Blood brain barrier disruption and convection-enhanced delivery have emerged as leading investigational delivery techniques for the treatment of malignant brain tumors. Clinical trials using these methods have been completed, with mixed results, and several more are being initiated. In this review, we describe the clinically available methods used to circumvent the blood brain barrier and summarize the results to date of ongoing and completed clinical trials.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Glioma/drug therapy , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain/blood supply , Brain/drug effects , Catheterization/instrumentation , Catheterization/methods , Drug Implants , Exotoxins/administration & dosage , Genetic Vectors , Glioblastoma/drug therapy , Humans , Immunotoxins/administration & dosage , Interleukins/administration & dosage , Transferrin/administration & dosage , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor beta/administration & dosage , Ultrasonic Therapy/methods
7.
Neuroscience ; 160(2): 470-83, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19248822

ABSTRACT

Transforming growth factor-alpha (TGFalpha) is a powerful endogenous mitogen and neurotrophic factor, which has previously been shown to induce a massive proliferative response in the brains of Parkinson's disease model rats injured by an acute neurotoxic lesion. We now show that TGFalpha can also produce a massive proliferative response in rat brains subjected to stroke caused by a middle cerebral artery occlusion (MCAO), even when the growth factor is administered as late as 4 weeks after injury. This combination of stimuli provokes DNA synthesis, shown by 5'-bromo-2-deoxyuridine incorporation, throughout the ependymal layer and subventricular zone (SVZ) of the forebrain during the 4 weeks of growth factor administration. The newly generated cells migrate preferentially along and ventral to the corpus callosum (CC) and external capsule to the site of the injury where many of them differentiate into several site-appropriate neuronal phenotypes in association with near complete (99%) behavioral recovery. We conclude that the injury response of endogenous neural stem cells as well as behavioral recovery can be significantly enhanced by application of TGFalpha, and that this approach represents a potential therapeutic strategy for chronic stroke and other neurological damage in human patients.


Subject(s)
Adult Stem Cells/physiology , Neurogenesis/physiology , Neurons/physiology , Stroke/pathology , Transforming Growth Factor alpha/physiology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Analysis of Variance , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Chronic Disease , Disease Models, Animal , Motor Activity/physiology , Neostriatum/cytology , Neostriatum/pathology , Neostriatum/physiology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Recovery of Function/physiology , Spatial Behavior/physiology , Statistics, Nonparametric , Stroke/physiopathology , Time Factors , Transforming Growth Factor alpha/administration & dosage
8.
Behav Brain Res ; 198(2): 440-8, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19110003

ABSTRACT

The growth factor, transforming growth factor-alpha (TGF-alpha) is strongly expressed in the hypothalamic circadian pacemaker, the suprachiasmatic nucleus (SCN). TGF-alpha is one of several SCN peptides recently suggested to function as a circadian output signal for the regulation of locomotor activity rhythms in nocturnal rodents. When infused in the brain, TGF-alpha suppresses activity. TGF-alpha suppresses other behaviors as well including feeding, resulting in weight loss. Elevated TGF-alpha is correlated with some cancers, and it is possible the TGF-alpha and its receptor, the epidermal growth factor receptor (EGFR), mediate fatigue and weight loss associated with cancer. If true for cancers outside of the brain, then systemic TGF-alpha should also affect behavior. We tested this hypothesis in hamsters with intraperitoneal injections or week-long subcutaneous infusions of TGF-alpha. Both treatments suppressed activity and infusions caused reduced food consumption and weight loss. To identify areas of the brain that might mediate these effects of systemic TGF-alpha, we used immunohistochemistry to localize cells with an activated MAP kinase signaling pathway (phosphorylated ERK1). Cells were activated in two hypothalamic areas, the paraventricular nucleus and a narrow region surrounding the third ventricle. These sites could not only be targets of TGF-alpha produced in the SCN but could also mediate effects of elevated TGF-alpha from tumors both within and outside the central nervous system.


Subject(s)
Eating/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/metabolism , Phosphorylation/drug effects , Transforming Growth Factor alpha/physiology , Weight Loss/drug effects , Animals , Cricetinae , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , Infusions, Subcutaneous , Injections, Intraperitoneal , Male , Mesocricetus , Signal Transduction/drug effects , Third Ventricle/cytology , Third Ventricle/metabolism , Time Factors , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/pharmacology
9.
Clin Cancer Res ; 14(20): 6531-7, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927293

ABSTRACT

PURPOSE: Neuroblastomas frequently show expression of the epidermal growth factor receptor (EGFR) and may therefore be susceptible to EGFR-targeted therapies. Here, EGFR expression and functionality was investigated in parental chemosensitive neuroblastoma cell lines (UKF-NB-3, IMR-32, NLF, SH-SY5Y) and their cisplatin-resistant sublines (UKF-NB-3(r)CDDP(1000), IMR-32(r)CDDP(1000), NLF(r)CDDP(1000), and SH-SY5Y(r)CDDP(500)). Moreover, the EGFR antibody cetuximab, the EGFR tyrosine kinase inhibitor Tyrphostin B46, and recombinant EGFR-targeted toxins were investigated for their influence on the viability and growth of neuroblastoma cells. EXPERIMENTAL DESIGN: EGFR expression and function was measured by flow cytometry or Western blot. Cell viability was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Apoptosis was examined by immunostaining for active caspase-3 or cleaved poly(ADP-ribose) polymerase. Cellular binding of FITC-labeled immunotoxins was studied by flow cytometry, and cellular uptake was studied by confocal laser scanning microscopy. RESULTS: The EGFR-targeted antibody and growth factor toxins scFv(14E1)- Pseudomonas exotoxin A (ETA) and TGF-alpha-ETA exerted anti-cancer effects in neuroblastoma cell lines that were insensitive to cetuximab or EGFR tyrosine kinase inhibitors. Furthermore, adaptation of chemosensitive neuroblastoma cells to cisplatin increased EGFR expression and sensitivity to both recombinant toxins. Treatment of chemosensitive neuroblastoma cells with cisplatin reversibly increased EGFR expression, whereas cisplatin-resistant cells showed enhanced EGFR expression independent of the presence of cisplatin. Combination treatment with scFv(14E1)-ETA or TGF-alpha-ETA and cisplatin exerted significantly improved anticancer effects compared with either single treatment in parental neuroblastoma cells, cisplatin-resistant sublines, and primary cultures. CONCLUSIONS: EGFR-targeted cytotoxic reagents such as scFv(14E1)-ETA and TGF-alpha-ETA represent promising candidates for further development as antineuroblastoma agents, especially in combination with cisplatin.


Subject(s)
ADP Ribose Transferases/therapeutic use , Antineoplastic Agents/pharmacology , Bacterial Toxins/therapeutic use , Cell Proliferation/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Exotoxins/therapeutic use , Neuroblastoma/drug therapy , Virulence Factors/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Survival/drug effects , Cetuximab , ErbB Receptors/genetics , ErbB Receptors/metabolism , Flow Cytometry , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinase Inhibitors/adverse effects , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/genetics , Tumor Cells, Cultured , Tyrphostins/adverse effects , Pseudomonas aeruginosa Exotoxin A
10.
Exp Neurol ; 214(1): 10-24, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18647603

ABSTRACT

Astrocytes respond to environmental cues and play a multifaceted role in the response to trauma in the central nervous system. As the most prevalent contributors to the glial scar, astrocytes are targeted as barriers to regeneration. However, there is also strong evidence that astrocytes are vital for neuroprotection and metabolic support after injury. In addition, consistent with their role during development, astrocytes may be capable of supporting the growth of injured axons. Therefore, we hypothesized that with appropriate stimulation, the reparative functions of endogenous astrocytes could be harnessed to promote axon growth and recovery after spinal cord injury. Transforming growth factor-alpha (TGF-alpha) is a mitogenic growth factor that is active on astrocytes and is poised to contribute to such a strategy. Recombinant TGF-alpha was administered intrathecally to adult C57BL/6 mice for two weeks following a moderate mid-thoracic spinal cord contusion. By three weeks post-injury, TGF-alpha infusion had not affected locomotor recovery, but promoted extensive axon growth and altered the composition of the lesion site. The center of the lesion in the treated mice contained greater numbers of new cells and increased astrocyte invasion. Despite the expression of inhibitory proteoglycans, there was a marked increase in axons expressing neurofilament and GAP-43 immunoreactivity, and the new axons were closely associated with increased laminin expression within and beyond the astrocyte matrix. The results demonstrate that astrocytes are dynamic players in the response to spinal cord injury, and the growth-supportive role of these cells can be enhanced by TGF-alpha infusion.


Subject(s)
Astrocytes/drug effects , Axons/drug effects , Nerve Regeneration/drug effects , Spinal Cord Injuries/drug therapy , Transforming Growth Factor alpha/administration & dosage , Analysis of Variance , Animals , Astrocytes/pathology , Axons/pathology , Cell Count , Female , Immunohistochemistry , Injections, Spinal , Mice , Microscopy, Confocal , Motor Activity/drug effects , Random Allocation , Recovery of Function/drug effects , Spinal Cord Injuries/pathology , Thoracic Vertebrae
11.
Neuro Oncol ; 10(3): 320-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18403491

ABSTRACT

The purpose of this study is to determine the maximum tolerated dose (MTD), dose-limiting toxicity (DLT), and intracerebral distribution of a recombinant toxin (TP-38) targeting the epidermal growth factor receptor in patients with recurrent malignant brain tumors using the intracerebral infusion technique of convection-enhanced delivery (CED). Twenty patients were enrolled and stratified for dose escalation by the presence of residual tumor from 25 to 100 ng/ml in a 40-ml infusion volume. In the last eight patients, coinfusion of (123)I-albumin was performed to monitor distribution within the brain. The MTD was not reached in this study. Dose escalation was stopped at 100 ng/ml due to inconsistent drug delivery as evidenced by imaging the coinfused (123)I-albumin. Two DLTs were seen, and both were neurologic. Median survival after TP-38 was 28 weeks (95% confidence interval, 26.5-102.8). Of 15 patients treated with residual disease, two (13.3%) demonstrated radiographic responses, including one patient with glioblastoma multiforme who had a nearly complete response and remains alive >260 weeks after therapy. Coinfusion of (123)I-albumin demonstrated that high concentrations of the infusate could be delivered >4 cm from the catheter tip. However, only 3 of 16 (19%) catheters produced intraparenchymal infusate distribution, while the majority leaked infusate into the cerebrospinal fluid spaces. Intracerebral CED of TP-38 was well tolerated and produced some durable radiographic responses at doses

Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Transforming Growth Factor alpha/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Exotoxins/adverse effects , Humans , Injections, Intraventricular , Magnetic Resonance Imaging , Maximum Tolerated Dose , Middle Aged , Tomography, Emission-Computed, Single-Photon , Transforming Growth Factor alpha/adverse effects
12.
Skin Res Technol ; 14(3): 370-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-19159386

ABSTRACT

BACKGROUND/PURPOSE: The skin has a functional and active phosphocreatine (PCR)/creatine kinase (CPK) system that regenerates adenosine triphosphate energy reserves during periods of ischemia. The objective of this study was to evaluate how topically applied growth factors affect CPK activity and distribution, and what histological changes growth factors induce in murine skin. METHODS: Epidermal growth factor (EGF), transforming growth factor alpha (TGF-alpha) and suramin (growth factor inhibitor) were applied to murine skin for nine days before mice were sacrificed and CPK level and distributions were measured. RESULTS: TGF-alpha considerably increased CPK activity. Both EGF and TGF-alpha induced a CPK MM to CPK BB transition and histologically induced abnormal differentiation of keratinocytes. CONCLUSION: The skin PCR/CPK system is affected by growth factors. Furthermore, this system appears to play an important role, both in the normal physiology of skin and pathophysiological conditions such as psoriasis and carcinogenesis.


Subject(s)
Creatine Kinase/metabolism , Epidermal Growth Factor/administration & dosage , Keratinocytes/cytology , Keratinocytes/enzymology , Skin/cytology , Skin/enzymology , Transforming Growth Factor alpha/administration & dosage , Administration, Topical , Animals , Cell Differentiation/drug effects , Cells, Cultured , Cytoplasm/enzymology , Enzyme Activation/drug effects , Intercellular Signaling Peptides and Proteins/administration & dosage , Keratinocytes/drug effects , Male , Mice , Skin/drug effects , Tissue Distribution/drug effects
13.
Syst Biol (Stevenage) ; 153(1): 22-33, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16983832

ABSTRACT

Members of the ErbB receptor family are associated with several cancers and appear to be providing useful targets for pharmacological therapeutics for tumours of the lung and breast. Further improvements of these therapies may be guided by a quantitative, dynamic integrative systems understanding of the complexities of ErbB dimerisation, trafficking and activation, for it is these complexities that render difficult intuiting how perturbations such as drug intervention will affect ErbB signalling activities. Towards this goal, we have developed a computational model implementing commonly accepted principles governing ErbB receptor interaction, trafficking, phosphorylation and dephosphorylation. Using this model, we are able to investigate several hypotheses regarding the compartmental localisation of dephosphorylation. Model results applied to experimental data on ErbB 1, ErbB2 and ErbB3 phosphorylation in H292 human lung carcinoma cells support a hypothesis that key dephosphorylation activity for these receptors occurs largely in an intracellular, endosomal compartment rather than at the cell surface plasma membrane. Thus, the endocytic trafficking-related compartmentalisation of dephosphorylation may define a critical aspect of the ErbB signalling response to ligand.


Subject(s)
Lung Neoplasms/metabolism , Models, Biological , Neuregulin-1/administration & dosage , Oncogene Proteins v-erbB/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction/drug effects , Transforming Growth Factor alpha/administration & dosage , Cell Line , Computer Simulation , Dose-Response Relationship, Drug , Drug Combinations , Enzyme Activation/drug effects , Humans , Metabolic Clearance Rate/drug effects , Phosphorylation/radiation effects
14.
Technol Cancer Res Treat ; 5(3): 201-13, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16700617

ABSTRACT

Despite advances in our knowledge about the genesis, molecular biology, and natural history of malignant gliomas and the use of a multi-disciplinary approach to their treatment, patients harboring this diagnosis continue to face a grim prognosis. At the time of diagnosis, patients typically undergo surgery for the establishment of a histologic diagnosis, the reduction of tumor burden, and the relief of mass effect, with the maintenance of the patient's neurological function in mind. This is followed by the administration of adjuvant therapeutics, including radiation therapy and chemotherapy. Many investigational agents with laboratory evidence of efficacy against malignant gliomas have not met their promise in the clinical setting, largely due to the barriers that they must overcome to reach the tumor at a therapeutically meaningful concentration for a durable period of time. The relevant aspects of the blood-brain barrier, blood-tumor barrier, and blood-cerebrospinal fluid barrier, as they pertain to the delivery of agents to the tumor, will be discussed along with the strategies devised to circumvent them. This discussion will be followed by a description of agents currently in preclinical and clinical development, many of which are the result of intense ongoing research into the molecular biology of gliomas.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Convection , Drug Delivery Systems , Glioma/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Bacterial Toxins/administration & dosage , Bacterial Toxins/pharmacokinetics , Biological Transport , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/pharmacokinetics , Exotoxins/administration & dosage , Exotoxins/pharmacokinetics , Glioma/metabolism , Humans , Interleukin-13/administration & dosage , Interleukin-13/pharmacokinetics , Interleukin-4/administration & dosage , Interleukin-4/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Transferrin/administration & dosage , Transferrin/analogs & derivatives , Transferrin/pharmacokinetics , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/pharmacokinetics
15.
Cell Death Differ ; 13(4): 576-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16179940

ABSTRACT

The serine protease granzyme B (GrB) of cytotoxic lymphocytes efficiently induces apoptosis by direct activation of caspases and cleavage of central caspase substrates. We employed human GrB as an effector function in chimeric fusion proteins that also contain the EGFR ligand TGFalpha or an ErbB2-specific single-chain antibody fragment (scFv) for selective targeting to tumor cells. GrB-TGFalpha (GrB-T) and GrB-scFv(FRP5) (GrB-5) molecules expressed in the yeast Pichia pastoris were bifunctional, cleaving synthetic and natural GrB substrates, and binding specifically to cells expressing EGFR or ErbB2 target receptors. Upon cell binding the chimeric molecules were internalized into intracellular vesicles, but could be released into the cytosol by the endosomolytic reagent chloroquine. Treatment with picomolar to nanomolar concentrations of GrB-5 and GrB-T resulted in selective and rapid tumor cell killing, accompanied by clear signs of apoptosis such as chromatin condensation, membrane blebbing, formation of apoptotic bodies and activation of endogenous initiator and effector caspases.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis , Drug Delivery Systems , Recombinant Fusion Proteins/administration & dosage , Serine Endopeptidases/administration & dosage , Antibodies/administration & dosage , Antibodies/genetics , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival/drug effects , Chloroquine/pharmacology , Cytoplasmic Vesicles/drug effects , Cytoplasmic Vesicles/enzymology , Dose-Response Relationship, Drug , Endocytosis , Enzyme Activation/drug effects , ErbB Receptors/metabolism , Granzymes , Humans , Inhibitory Concentration 50 , Pichia/genetics , Pichia/metabolism , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/biosynthesis , Serine Endopeptidases/genetics , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/genetics
16.
Brain Res ; 1038(2): 171-82, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15757633

ABSTRACT

Transforming growth factor-alpha (TGF-alpha) is a candidate output signal of the hypothalamic circadian pacemaker. TGF-alpha is expressed in the suprachiasmatic nucleus (SCN) of rats, hamsters, and rhesus macaques [A. Kramer, F.C. Yang, P. Snodgrass, X. Li, T.E. Scammell, F.C. Davis and C.J. Weitz, Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling, Science, 294 (2001) 2511-5., X. Li, N. Sankrithi and F.C. Davis, Transforming growth factor-alpha is expressed in astrocytes of the suprachiasmatic nucleus in hamster: role of glial cells in circadian clocks, Neuroreport, 13 (2002) 2143-7., Y.J. Ma, M.E. Costa and S.R. Ojeda, Developmental expression of the genes encoding transforming growth factor alpha and its receptor in the hypothalamus of female rhesus macaques, Neuroendocrinology, 60 (1994) 346-59., Y.J. Ma, M.P. Junier, M.E. Costa and S.R. Ojeda, Transforming growth factor-alpha gene expression in the hypothalamus is developmentally regulated and linked to sexual maturation, Neuron, 9 (1992) 657-70.]. TGF-alpha reversibly inhibits wheel-running activity during long-term infusions into the third ventricle of hamsters (2 weeks, intracerebroventricular or ICV) [A. Kramer, F.C. Yang, P. Snodgrass, X. Li, T.E. Scammell, F.C. Davis and C.J. Weitz, Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling, Science, 294 (2001) 2511-5.], and this effect appears to be mediated by the epidermal growth factor receptor (EGFR or ErbB-1) [A. Kramer, F.C. Yang, P. Snodgrass, X. Li, T.E. Scammell, F.C. Davis and C.J. Weitz, Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling, Science, 294 (2001) 2511-5.]. Here, we demonstrate that this inhibitory effect is not restricted to wheel-running behavior or to mediation by the EGFR. Using direct observation, we found the effects of long-term TGF-alpha infusion (ICV, 12 microl/day, 3.3 microM) to be more general than previously reported. Other active behaviors such as grooming and feeding were reversibly inhibited and hamsters showed dramatic weight loss as a result of reduced feeding (34% of body weight over 19 days). TGF-alpha did not disrupt a non-behavioral rhythm, the rhythm in pineal melatonin. Wheel-running activity was also inhibited by another epidermal growth factor-like (EGF-like) peptide, neuregulin (NRG-1), that binds to different ErbB receptors. Like TGF-alpha, NRG-1 caused a significant weight loss. We also show that an acute injection of TGF-alpha inhibits activity (ICV, 5 microl, 3.3 microM over 2 min), with inhibition and recovery occurring over a few hours. Although the results are consistent with the proposed [A. Kramer, F.C. Yang, P. Snodgrass, X. Li, T.E. Scammell, F.C. Davis and C.J. Weitz, Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling, Science, 294 (2001) 2511-5.] role for EGF-like peptides in the daily regulation of activity, the actions of these peptides might also contribute to the behavioral etiology of diseases in which EGF-like peptides are expressed.


Subject(s)
Motor Activity/drug effects , Neuregulin-1/pharmacology , Transforming Growth Factor alpha/pharmacology , Weight Loss/drug effects , Animals , Cricetinae , Eating/drug effects , ErbB Receptors/drug effects , Injections, Intraventricular , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/pharmacology , Male , Mesocricetus , Neuregulin-1/administration & dosage , Neuropeptides/administration & dosage , Neuropeptides/pharmacology , Orexin Receptors , Orexins , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Transforming Growth Factor alpha/administration & dosage
17.
Neuro Oncol ; 7(1): 90-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15701286

ABSTRACT

Glioblastoma multiforme remains refractory to conventional therapy, and novel therapeutic modalities are desperately needed. TP-38 is a recombinant chimeric protein containing a genetically engineered form of the cytotoxic Pseudomonas exotoxin fused to transforming growth factor (TGF)-alpha. TGF-alpha binds with high affinity to the epidermal growth factor receptor, which is uniformly overexpressed in malignant gliomas, often because of gene amplification. Prior to therapy with TP-38, the patient described here was completely refractory to multiple other therapies, with radiographic and pathologic evidence of tumor progression. After therapy, she improved clinically, was weaned off steroids and anti-convulsants, and experienced a progressive decrease in enhancing tumor volume. Despite multiple prior recurrences, she has not progressed for >43 months after TP-38 therapy. Small remaining areas of enhancement demonstrate no evidence of tumor histologically and are hypometabolic on positron emission tomography. This report describes a dramatic and sustained clinical and radiographic response in a patient with a bifrontal glioblastoma multiforme treated with intratumoral infusion of a novel targeted toxin, TP-38.


Subject(s)
Brain Neoplasms/drug therapy , Exotoxins/administration & dosage , Glioblastoma/drug therapy , Recombinant Fusion Proteins/administration & dosage , Transforming Growth Factor alpha/administration & dosage , Brain Neoplasms/pathology , Female , Glioblastoma/pathology , Humans , Injections, Intraventricular , Magnetic Resonance Imaging , Middle Aged , Pseudomonas aeruginosa , Treatment Outcome
18.
J Neurosci ; 24(41): 8924-31, 2004 Oct 13.
Article in English | MEDLINE | ID: mdl-15483111

ABSTRACT

We examined the cell proliferative, neurogenic, and behavioral effects of transforming growth factor alpha (TGFalpha) in a 6-OHDA Parkinson's disease model when compared with naive rats. Intrastriatal TGFalpha infusion induced significant proliferation, hyperplastic nodules, and substantial migratory waves of nestin-positive progenitor cells from the adult subventricular zone (SVZ) of dopamine-denervated rats. Interestingly, SVZ cells in naive rats displayed proliferation but minimal migration in response to the TGFalpha infusion. The cells in the expanded SVZ accumulated cytoplasmic beta-catenin, indicating activation of classical Wnt signaling. However, no evidence of any neuronal differentiation was found of these recruited progenitor cells anywhere examined in the brain. Consequently, no evidence of dopaminergic (DA) neurogenesis was found in the striatum or substantia nigra in any experimental group, and amphetamine-induced behavioral rotations did not improve. In summary, the cells in the TGFalpha-induced migratory cellular wave remain undifferentiated and do not differentiate into midbrain-like DA neurons.


Subject(s)
Cell Movement/drug effects , Corpus Striatum/drug effects , Neurons/drug effects , Parkinsonian Disorders/drug therapy , Stem Cells/drug effects , Transforming Growth Factor alpha/administration & dosage , Animals , Behavior, Animal/drug effects , Bromodeoxyuridine , Cell Proliferation/drug effects , Corpus Striatum/pathology , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Dopamine/metabolism , Drug Administration Routes , Intermediate Filament Proteins/biosynthesis , Lateral Ventricles/cytology , Lateral Ventricles/drug effects , Lateral Ventricles/metabolism , Male , Nerve Tissue Proteins/biosynthesis , Nestin , Neurons/metabolism , Neurons/pathology , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Rats , Rats, Sprague-Dawley , Stem Cells/pathology , Trans-Activators/metabolism , Tyrosine 3-Monooxygenase/biosynthesis , beta Catenin
19.
Urology ; 64(3): 409-21, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15351555

Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Transitional Cell/therapy , Cytosine/analogs & derivatives , Immunologic Factors/therapeutic use , Immunotherapy , Urinary Bladder Neoplasms/therapy , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Administration, Intravesical , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/therapeutic use , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/pathology , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cytosine/administration & dosage , Cytosine/adverse effects , Cytosine/therapeutic use , Exotoxins/administration & dosage , Exotoxins/therapeutic use , Heart Diseases/chemically induced , Hemocyanins/administration & dosage , Hemocyanins/therapeutic use , Humans , Immunologic Factors/administration & dosage , Immunotherapy/methods , Immunotherapy/trends , Interferons/administration & dosage , Interferons/therapeutic use , Interleukins/administration & dosage , Interleukins/therapeutic use , Randomized Controlled Trials as Topic , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/therapeutic use , Treatment Outcome , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology
20.
Int J Pharm ; 277(1-2): 163-72, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15158979

ABSTRACT

The present study was designed to investigate the effects of microemulsion and aqueous solution containing transforming growth factor alpha (TGF-alpha) and/or aprotinin administered intragastrically (i.g.) on healing of acute gastric ulcers induced by acetylsalicylic acid (ASA). The microemulsion was prepared by modification of the microemulsion formulation described in our previous study. Acute gastric lesions were induced by the application of ASA (150 mg/kg in 1.5 ml of 0.2N HCl i.g.). TGF-alpha in solution or microemulsion formulations were administered at a dose of 10 microg/kg per 24h i.g. for 2 days. The effects of TGF-alpha on the healing was evaluated with the measurement of ulcer score, basal gastric acid secretion, total protein content of gastric fluid, gastric mucus level and histological analysis. The results indicated that the highest decrease in ulcer area was observed in group treated with microemulsion containing TGF-alpha plus aprotinin (TA-ME). TGF-alpha in microemulsion formulation was more effective than TGF-alpha in solution formulation in the increase of gastric mucus secretion, in the decrease of gastric acid secretions and ulcer scores. Histological evaluation of the gastric mucosa samples revealed that, best recovery was obtained in the TA-ME treated group.


Subject(s)
Aspirin/toxicity , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Transforming Growth Factor alpha/therapeutic use , Animals , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Injections, Intralesional , Male , Mice , Rats , Rats, Wistar , Stomach Ulcer/pathology , Transforming Growth Factor alpha/administration & dosage , Transforming Growth Factor alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...