Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Breast Cancer ; 30(4): 559-569, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36977972

ABSTRACT

BACKGROUND: Based on the volume of tissue removed, conservative surgery (BCS) cannot always guarantee satisfactory cosmetic results, unless resorting to more complex oncoplastic approaches. Investigating an alternative to optimize aesthetic outcomes minimizing surgical complexity, was the purpose of this study. We assessed an innovative surgical procedure based on the use of a biomimetic polyurethane-based scaffold intended for regenerating soft-tissue resembling fat, in patients undergoing BCS for non-malignant breast lesions. Safety and performance of the scaffold, and safety and feasibility of the entire implant procedure were evaluated. METHODS: A volunteer sample of 15 female patients underwent lumpectomy with immediate device positioning, performing seven study visits with six-month follow-up. We evaluated incidence of adverse events (AEs), changes in breast appearance (using photographs and anthropomorphic measurements), interference with ultrasound and MRI (assessed by two independent investigators), investigator's satisfaction (through a VAS scale), patient's pain (through a VAS scale) and quality of life (QoL) (using the BREAST-Q© questionnaire). Data reported are the results of the interim analysis on the first 5 patients. RESULTS: No AEs were device related nor serious. Breast appearance was unaltered and the device did not interference with imaging. High investigator's satisfaction, minimal post-operative pain and positive impact on QoL were also detected. CONCLUSIONS: Albeit on a limited number of patients, data showed positive outcomes both in terms of safety and performance, paving the way to an innovative breast reconstructive approach with a potential remarkable impact on clinical application of tissue engineering. TRIAL REGISTRATION: ClinicalTrials.gov (NCT04131972, October 18, 2019).


Subject(s)
Breast Neoplasms , Mammaplasty , Mastectomy, Segmental , Female , Humans , Biomimetics , Breast Neoplasms/surgery , Mammaplasty/adverse effects , Mammaplasty/methods , Patient Satisfaction , Polyurethanes , Quality of Life , Mastectomy, Segmental/adverse effects , Tissue Scaffolds , Tissue Engineering
2.
Sci Rep ; 11(1): 13310, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172806

ABSTRACT

The use of cell-free scaffolds for the regeneration of clinically relevant volumes of soft tissue has been challenged, particularly in the case of synthetic biomaterials, by the difficulty of reconciling the manufacturing and biological performance requirements. Here, we investigated in vivo the importance of biomechanical and biochemical cues for conditioning the 3D regenerative microenvironment towards soft tissue formation. In particular, we evaluated the adipogenesis changes related to 3D mechanical properties by creating a gradient of 3D microenvironments with different stiffnesses using 3D Poly(Urethane-Ester-ether) PUEt scaffolds. Our results showed a significant increase in adipose tissue proportions while decreasing the stiffness of the 3D mechanical microenvironment. This mechanical conditioning effect was also compared with biochemical manipulation by loading extracellular matrices (ECMs) with a PPAR-γ activating molecule. Notably, results showed mechanical and biochemical conditioning equivalency in promoting adipose tissue formation in the conditions tested, suggesting that adequate mechanical signaling could be sufficient to boost adipogenesis by influencing tissue remodeling. Overall, this work could open a new avenue in the design of synthetic 3D scaffolds for microenvironment conditioning towards the regeneration of large volumes of soft and adipose tissue, with practical and direct implications in reconstructive and cosmetic surgery.


Subject(s)
Cellular Microenvironment/physiology , Regeneration/physiology , 3T3-L1 Cells , Adipogenesis/physiology , Adipose Tissue/physiology , Animals , Cell Line , Extracellular Matrix/physiology , Mice , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Wound Healing/physiology
3.
4.
Sci Rep ; 9(1): 12195, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434921

ABSTRACT

IClswell is the chloride current induced by cell swelling, and plays a fundamental role in several biological processes, including the regulatory volume decrease (RVD). ICln is a highly conserved, ubiquitously expressed and multifunctional protein involved in the activation of IClswell. In platelets, ICln binds to the intracellular domain of the integrin αIIb chain, however, whether the ICln/integrin interaction plays a role in RVD is not known. Here we show that a direct molecular interaction between ICln and the integrin α-chain is not restricted to platelets and involves highly conserved amino acid motifs. Integrin α recruits ICln to the plasma membrane, thereby facilitating the activation of IClswell during hypotonicity. Perturbation of the ICln/integrin interaction prevents the transposition of ICln towards the cell surface and, in parallel, impedes the activation of IClswell. We suggest that the ICln/integrin interaction interface may represent a new molecular target enabling specific IClswell suppression in pathological conditions when this current is deregulated or plays a detrimental role.


Subject(s)
Blood Platelets/metabolism , Cell Membrane/metabolism , Chloride Channels/metabolism , Integrin alpha Chains/metabolism , Animals , Cell Membrane/genetics , Chloride Channels/genetics , Dogs , HEK293 Cells , Humans , Integrin alpha Chains/genetics , Ion Transport , Madin Darby Canine Kidney Cells
5.
Acta Biomater ; 73: 141-153, 2018 06.
Article in English | MEDLINE | ID: mdl-29654992

ABSTRACT

Reconstructive treatment after trauma and tumor resection would greatly benefit from an effective soft tissue regeneration. The use of cell-free scaffolds for adipose tissue regeneration in vivo is emerging as an attractive alternative to tissue-engineered constructs, since this approach avoids complications due to cell manipulation and lack of synchronous vascularization. In this study, we developed a biodegradable polyurethane-based scaffold for soft tissue regeneration, characterized by an exceptional combination between softness and resilience. Exploring the potential as a cell-free scaffold required profound understanding of the impact of its intrinsic physico-chemical properties on the biological performance in vivo. We investigated the effect of the scaffold's hydrophilic character, degradation kinetics, and internal morphology on (i) the local inflammatory response and activation of MGCs (foreign body response); (ii) its ability to promote rapid vascularisation, cell infiltration and migration through the scaffold over time; and (iii) the grade of maturation of the newly formed tissue into vascularized soft tissue in a murine model. The study revealed that soft tissue regeneration in vivo proceeded by gradual infiltration of undifferentiated mesenchymal cells though the periphery toward the center of the scaffold, where the rapid formation of a functional and well-formed vascular network supported cell viability overtime. STATEMENT OF SIGNIFICANCE: Exploring the potential of polyurethane-based soft foam as cell-free scaffold for soft tissue regeneration. In this work, we address the unmet need for synthetic functional soft tissue substitutes that provide adequate biological and mechanical support to soft tissue. We developed a series of flexible cross-linked polyurethane copolymer scaffolds with remarkable fatigue-resistance and tunable physico-chemical properties for soft tissue regeneration in vivo. Accordingly, we could extend the potential of this class of biomaterials, which was so far confined for bone and osteochondral tissue regeneration, to other types of connective tissue.


Subject(s)
Adipose Tissue/blood supply , Neovascularization, Physiologic/drug effects , Polyurethanes , Regeneration/drug effects , Tissue Scaffolds/chemistry , Adipose Tissue/pathology , Animals , Mice , NIH 3T3 Cells , Polyurethanes/chemistry , Polyurethanes/pharmacology
6.
Biomaterials ; 104: 65-77, 2016 10.
Article in English | MEDLINE | ID: mdl-27428768

ABSTRACT

Despite clinical treatments for adipose tissue defects, in particular breast tissue reconstruction, have certain grades of efficacy, many drawbacks are still affecting the long-term survival of new formed fat tissue. To overcome this problem, in the last decades, several scaffolding materials have been investigated in the field of adipose tissue engineering. However, a strategy able to recapitulate a suitable environment for adipose tissue reconstruction and maintenance is still missing. To address this need, we adopted a biologically and mechanically driven design to fabricate an RGD-mimetic poly(amidoamine) oligomer macroporous foam (OPAAF) for adipose tissue reconstruction. The scaffold was designed to fulfil three fundamental criteria: capability to induce cell adhesion and proliferation, support of in vivo vascularization and match of native tissue mechanical properties. Poly(amidoamine) oligomers were formed into soft scaffolds with hierarchical porosity through a combined free radical polymerization and foaming reaction. OPAAF is characterized by a high water uptake capacity, progressive degradation kinetics and ideal mechanical properties for adipose tissue reconstruction. OPAAF's ability to support cell adhesion, proliferation and adipogenesis was assessed in vitro using epithelial, fibroblast and endothelial cells (MDCK, 3T3L1 and HUVEC respectively). In addition, in vivo subcutaneous implantation in murine model highlighted OPAAF potential to support both adipogenesis and vessels infiltration. Overall, the reported results support the use of OPAAF as a scaffold for engineered adipose tissue construct.


Subject(s)
Adipocytes/physiology , Adipogenesis/physiology , Adipose Tissue/growth & development , Mechanotransduction, Cellular/physiology , Oligopeptides/chemistry , Tissue Scaffolds , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Biomimetic Materials/chemical synthesis , Cell Adhesion/physiology , Cell Line , Cell Proliferation/physiology , Compressive Strength , Elastic Modulus , Extracellular Matrix/chemistry , Female , Gases/chemistry , Mice , Neovascularization, Physiologic/physiology , Porosity , Stress, Mechanical , Tissue Engineering/instrumentation , Tissue Engineering/methods
7.
J Nanobiotechnology ; 14: 18, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26955876

ABSTRACT

BACKGROUND: Thanks to mechanotransductive components cells are competent to perceive nanoscale topographical features of their environment and to convert the immanent information into corresponding physiological responses. Due to its complex configuration, unraveling the role of the extracellular matrix is particularly challenging. Cell substrates with simplified topographical cues, fabricated by top-down micro- and nanofabrication approaches, have been useful in order to identify basic principles. However, the underlying molecular mechanisms of this conversion remain only partially understood. RESULTS: Here we present the results of a broad, systematic and quantitative approach aimed at understanding how the surface nanoscale information is converted into cell response providing a profound causal link between mechanotransductive events, proceeding from the cell/nanostructure interface to the nucleus. We produced nanostructured ZrO2 substrates with disordered yet controlled topographic features by the bottom-up technique supersonic cluster beam deposition, i.e. the assembling of zirconia nanoparticles from the gas phase on a flat substrate through a supersonic expansion. We used PC12 cells, a well-established model in the context of neuronal differentiation. We found that the cell/nanotopography interaction enforces a nanoscopic architecture of the adhesion regions that affects the focal adhesion dynamics and the cytoskeletal organization, which thereby modulates the general biomechanical properties by decreasing the rigidity of the cell. The mechanotransduction impacts furthermore on transcription factors relevant for neuronal differentiation (e.g. CREB), and eventually the protein expression profile. Detailed proteomic data validated the observed differentiation. In particular, the abundance of proteins that are involved in adhesome and/or cytoskeletal organization is striking, and their up- or downregulation is in line with their demonstrated functions in neuronal differentiation processes. CONCLUSION: Our work provides a deep insight into the molecular mechanotransductive mechanisms that realize the conversion of the nanoscale topographical information of SCBD-fabricated surfaces into cellular responses, in this case neuronal differentiation. The results lay a profound cell biological foundation indicating the strong potential of these surfaces in promoting neuronal differentiation events which could be exploited for the development of prospective research and/or biomedical applications. These applications could be e.g. tools to study mechanotransductive processes, improved neural interfaces and circuits, or cell culture devices supporting neurogenic processes.


Subject(s)
Cell Differentiation/drug effects , Mechanotransduction, Cellular/drug effects , Nanoparticles/administration & dosage , Nanostructures/administration & dosage , Zirconium/administration & dosage , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cytoskeleton/drug effects , Down-Regulation/drug effects , Extracellular Matrix/drug effects , PC12 Cells , Rats , Surface Properties/drug effects , Up-Regulation/drug effects
8.
Macromol Biosci ; 16(6): 870-81, 2016 06.
Article in English | MEDLINE | ID: mdl-26900107

ABSTRACT

This study presents a custom-made in situ gelling polymeric precursor for cell encapsulation. Composed of poly((2-hydroxyethyl)methacrylate-co-(3-aminopropyl)methacrylamide) (P(HEMA-co-APM) mother backbone and RGD-mimicking poly(amidoamine) (PAA) moiteis, the comb-like structured polymeric precursor is tailored to gather the advantages of the two families of synthetic polymers, i.e., the good mechanical integrity of PHEMA-based polymers and the biocompatibility and biodegradability of PAAs. The role of P(HEMA-co-APM) in the regulation of the chemico-physical properties of P(HEMA-co-APM)/PAA hydrogels is thoroughly investigated. On the basis of obtained results, namely the capability of maintaining vital NIH3T3 cell line in vitro for 2 d in a 3D cell culture, the in vivo biocompatibility in murine model for 16 d, and the ability of finely tuning mechanical properties and degradation kinetics, it can be assessed that P(HEMA-co-APM)/PAAs offer a cost-effective valid alternative to the so far studied natural polymer-based systems for cell encapsulation.


Subject(s)
Alanine/analogs & derivatives , Hydrogels/chemistry , Polyhydroxyethyl Methacrylate/chemistry , Acrylamides/chemistry , Alanine/chemistry , Alanine/pharmacology , Animals , Cell Culture Techniques/methods , Hydrogels/pharmacology , Kinetics , Materials Testing , Methacrylates/chemistry , Mice , NIH 3T3 Cells/drug effects , Polyhydroxyethyl Methacrylate/pharmacology
9.
Mol Imaging ; 142015.
Article in English | MEDLINE | ID: mdl-26044881

ABSTRACT

The main scientific issue hindering the development of tissue engineering technologies is the lack of proper vascularization. Among the various approaches developed for boosting vascularization, scaffold design has attracted increasing interest over the last few years. The aim of this article is to illustrate a scaffold design strategy for enhancing vascularization based on sacrificial microfabrication of embedded microchannels. This approach was combined with an innovative poly(ether urethane urea) (PEUtU) porous scaffold to provide an alternative graft substitute material for the treatment of tissue defects. Fluorescent and chemiluminescent imaging combined with computed tomography were used to study the behavior of the scaffold composition within living subjects by analyzing angiogenesis and inflammation processes and observing the variation in x-ray absorption, respectively. For this purpose, an IntegriSense 680 probe was used in vivo for the localization and quantification of integrin αvß3, due to its critical involvement in angiogenesis, and a XenoLight RediJect Inflammation Probe for the study of the decline in inflammation progression during healing. Overall, the collected data suggest the advantages of embedding a synthetic vascular network into a PEUtU porous matrix to enhance in vivo tissue integration, maturation, and regeneration. Moreover, our imaging approach proved to be an efficient and versatile tool for scaffold in vivo testing.


Subject(s)
Diagnostic Imaging/methods , Neovascularization, Physiologic , Tissue Scaffolds/chemistry , Animals , Female , Inflammation/pathology , Mechanical Phenomena , Mice , Porosity , Tomography, X-Ray Computed
10.
Acta Biomater ; 18: 144-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25724444

ABSTRACT

The potential of the 3D cell culture approach for creating in vitro models for drug screening and cellular studies, has led to the development of hydrogels that are able to mimic the in vivo 3D cellular milieu. To this aim, synthetic polymer-based hydrogels, with which it is possible to fine-tune the chemical and biophysical properties of the cell microenvironment, are becoming more and more acclaimed. Of all synthetic materials, poly(amidoamine)s (PAAs) hydrogels are known to have promising properties. In particular, PAAs hydrogels containing the 2,2-bisacrylamidoacetic acid-agmatine monomeric unit are capable of enhancing cellular adhesion by interacting with the RGD-binding αVß3 integrin. The synthesis of a new photocrosslinkable, biomimetic PAA-Jeffamine®-PAA triblock copolymer (PJP) hydrogel is reported in this paper with the aim of improving the optical, biocompatibility and cell-adhesion properties of previously studied PAA hydrogels and providing an inexpensive alternative to the RGD peptide based hydrogels. The physicochemical properties of PJP hydrogels are extensively discussed and the behavior of 2D and 3D cell cultures was analyzed in depth with different cell types. Moreover, cell-laden PJP hydrogels were patterned with perfusable microchannels and seeded with endothelial cells, in order to investigate the possibility of using PJP hydrogels for fabricating cell laden tissue-like micro constructs and microfluidic devices. Overall the data obtained suggest that PJP could ultimately become a useful tool for fabricating improved in vitro models in order to potentially enhance the effectiveness of drug screening and clinical treatments.


Subject(s)
Biomimetic Materials/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Oligopeptides/pharmacology , Polyamines/pharmacology , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Elastic Modulus/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Mice , Microfluidics , NIH 3T3 Cells , Optical Phenomena
11.
Biomaterials ; 45: 124-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662502

ABSTRACT

Despite significant progresses were achieved in tissue engineering over the last 20 years, a number of unsolved problems still remain. One of the most relevant issues is the lack of a proper vascularization that is limiting the size of the engineered tissues to smaller than clinically relevant dimensions. Sacrificial molding holds great promise to engineered construct with perfusable vascular architectures, but there is still the need to develop more versatile approaches able to be independent of the nature and dimensions of the construct. In this work we developed a versatile sacrificial molding technique for fabricating bulk, cell-laden and porous scaffolds with embedded vascular fluidic networks. These branched fluidic architectures are created by highly resistant thermoplastic sacrificial templates, made of poly(vinyl alcohol), representing a remarkable progress in manufacturability and scalability. The obtained architecture, when perfused in bioreactor, has shown to prevent the formation of a necrotic core in thick cell-laden constructs and enabled the rapid fabrication of hierarchically branched endothelium. In conclusion we demonstrate a novel strategy towards the engineering of vascularized thick tissues through the integration of the PVA-based microfabrication sacrificial approach and perfusion bioreactors. This approach may be able to scale current engineered tissues to clinically relevant dimensions, opening the way to their widespread clinical applications.


Subject(s)
Bioreactors , Neovascularization, Physiologic , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cell Survival , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Gels , Mice , Microfluidics , Microtechnology , NIH 3T3 Cells , Optical Imaging , Porosity
12.
Acta Biomater ; 10(8): 3463-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24793656

ABSTRACT

Nucleus pulposus (NP) tissue damage can induce detrimental mechanical stresses and strains on the intervertebral disc, leading to disc degeneration. This study demonstrates the potential of a novel, photo-curable, injectable, synthetic polymer hydrogel (pHEMA-co-APMA grafted with polyamidoamine (PAA)) to encapsulate and differentiate human mesenchymal stem cells (hMSC) towards a NP phenotype under hypoxic conditions which could be used to restore NP tissue function and mechanical properties. Encapsulated hMSC cultured in media (hMSC and chondrogenic) displayed good cell viability up to day 14. The genotoxicity effects of ultraviolet (UV) on hMSC activity confirmed the acceptability of 2.5min of UV light exposure to cells. Cytotoxicity investigations revealed that hMSC cultured in media containing p(HEMA-co-APMA) grafted with PAA degradation product (10% and 20%v/v concentration) for 14days significantly decreased the initial hMSC adhesion ability and proliferation rate from 24hrs to day 14. Successful differentiation of encapsulated hMSC within hydrogels towards chondrogenesis was observed with elevated expression levels of aggrecan and collagen II when cultured in chondrogenic media under hypoxic conditions, in comparison with culture in hMSC media for 14days. Characterization of the mechanical properties revealed a significant decrease in stiffness and modulus values of cellular hydrogels in comparison with acellular hydrogels at both day 7 and day 14. These results demonstrate the potential use of an in vivo photo-curable injectable, synthetic hydrogel with encapsulated hMSC for application in the repair and regeneration of NP tissue.


Subject(s)
Absorbable Implants , Hydrogels/chemistry , Intervertebral Disc/growth & development , Mesenchymal Stem Cell Transplantation/instrumentation , Mesenchymal Stem Cells/physiology , Regeneration/physiology , Cell Hypoxia/physiology , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Guided Tissue Regeneration/instrumentation , Humans , Hydrogels/radiation effects , Injections , Intervertebral Disc/cytology , Materials Testing , Mesenchymal Stem Cells/cytology , Photochemistry/methods , Prosthesis Design , Ultraviolet Rays
13.
Acta Biomater ; 10(3): 1206-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24361426

ABSTRACT

Poly(amido-amine) (PAA) hydrogels containing the 2,2-bisacrylamidoacetic acid-4-amminobutyl guanidine monomeric unit have a known ability to enhance cellular adhesion by interacting with the arginin-glycin-aspartic acid (RGD)-binding αVß3 integrin, expressed by a wide number of cell types. Scientific interest in this class of materials has traditionally been hampered by their poor mechanical properties and restricted range of degradation rate. Here we present the design of novel biocompatible, RGD-mimic PAA-based hydrogels with wide and tunable degradation rates as well as improved mechanical and biological properties for biomedical applications. This is achieved by radical polymerization of acrylamide-terminated PAA oligomers in both the presence and absence of 2-hydroxyethylmethacrylate. The degradation rate is found to be precisely tunable by adjusting the PAA oligomer molecular weight and acrylic co-monomer concentration in the starting reaction mixture. Cell adhesion and proliferation tests on Madin-Darby canine kidney epithelial cells show that PAA-based hydrogels have the capacity to promote cell adhesion up to 200% compared to the control. Mechanical tests show higher compressive strength of acrylic chain containing hydrogels compared to traditional PAA hydrogels.


Subject(s)
Hydrogels/pharmacology , Mechanical Phenomena/drug effects , Polyamines/pharmacology , Tissue Engineering/methods , Actins/metabolism , Animals , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Dogs , Hydrogels/chemical synthesis , Hydrogels/chemistry , Madin Darby Canine Kidney Cells , Polyamines/chemical synthesis , Polyamines/chemistry , Polyhydroxyethyl Methacrylate/pharmacology
14.
J Nanobiotechnology ; 11: 35, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24119372

ABSTRACT

BACKGROUND: Substrate nanoscale topography influences cell proliferation and differentiation through mechanisms that are at present poorly understood. In particular the molecular mechanism through which cells 'sense' and adapt to the substrate and activate specific intracellular signals, influencing cells survival and behavior, remains to be clarified. RESULTS: To characterize these processes at the molecular level we studied the differentiation of PC12 cells on nanostructured TiO2 films obtained by supersonic cluster beam deposition.Our findings indicate that, in PC12 cells grown without Nerve Growth Factor (NGF), the roughness of nanostructured TiO2 triggers neuritogenesis by activating the expression of nitric oxide synthase (NOS) and the phospho-extracellular signal-regulated kinase 1/2 (pERK1/2) signaling. Differentiation is associated with an increase in protein nitration as observed in PC12 cells grown on flat surfaces in the presence of NGF. We demonstrate that cell differentiation and protein nitration induced by topography are not specific for PC12 cells but can be regarded as generalized effects produced by the substrate on different neuronal-like cell types, as shown by growing the human neuroblastoma SH-SY5Y cell line on nanostructured TiO2. CONCLUSION: Our data provide the evidence that the nitric oxide (NO) signal cascade is involved in the differentiation process induced by nanotopography, adding new information on the mechanism and proteins involved in the neuritogenesis triggered by the surface properties.


Subject(s)
Biocompatible Materials/chemistry , Mechanotransduction, Cellular , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Titanium/chemistry , Animals , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factor/pharmacology , Neurites/metabolism , Neurites/ultrastructure , Nitric Oxide Synthase Type II/genetics , PC12 Cells , Rats , Surface Properties , Titanium/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/metabolism
15.
Respir Physiol Neurobiol ; 188(2): 223-8, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23619193

ABSTRACT

The causes and development of lung fluid, as well as the integrity of the alveolar-capillary membrane at high altitude, are undefined. This study was conceived to see whether fluid accumulates within the lung with acute high altitude exposure, and whether this is associated with alveolar capillary membrane damage. We studied lung carbon monoxide diffusion (DLCO), its components - membrane diffusion (DM) and capillary volume (VC) and alveolar volume (VA) measured in 43 healthy subjects in Milan (122 m) and after 1 and 3 days at Capanna Regina Margherita (4559 m). DLCO measurement was adjusted for hemoglobin and inspired oxygen. We also measured plasma surfactant derived protein B (SPB) and Receptor of Advanced Glycation End-products (RAGE) as markers of alveolar-capillary membrane damage, and ultrasound lung comets as a marker of extravascular lung water. 21 subjects received acetazolamide and 22 placebo. DLCO was lower at Capanna Regina Margherita (day 1: 24.3 ± 4.7 and day 3: 23.6 ± 5.4 mL/mmHg/min), than in Milan (25.8 ± 5.5; p<0.001 vs. day 1 and 3) due to DM reduction (Milan: 50.5 ± 14.6 mL/mmHg/min, Capanna Regina Margherita day 1: 45.1 ± 11.5 mL/mmHg/min, day 3: 43.2 ± 13.9 mL/mmHg/min; p<0.05 Milan vs. day 3) with a partially compensatory VC increase (Milan: 96 ± 37 mL, Capanna Regina Margherita day 1: 152 ± 66 mL, day 3: 153 ± 59 mL; p<0.001 Milan vs. day 1 and day 3). Acetazolamide did not prevent the fall in DLCO albeit, between day 1 and 3, such a trend was observed. Regardless of treatment lung comets increased from 0 to 7.2 ± 3.6 (p<0.0001). SPB and RAGE were unchanged. Lung fluid increased at high altitude without evidence from plasma measurements, supporting alveolar-capillary damage.


Subject(s)
Altitude , Lung/physiology , Pulmonary Diffusing Capacity/physiology , Adult , Blood Pressure/physiology , Carbon Monoxide/metabolism , Female , Heart Rate/physiology , Humans , Male , Middle Aged , Pulmonary Alveoli/physiology , Respiratory Function Tests , Statistics, Nonparametric
16.
J Biol Chem ; 286(47): 40659-70, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21917931

ABSTRACT

Identifying functional partners for protein/protein interactions can be a difficult challenge. We proposed the use of the operon structure of the Caenorhabditis elegans genome as a "new gene-finding tool" (Eichmüller, S., Vezzoli, V., Bazzini, C., Ritter, M., Fürst, J., Jakab, M., Ravasio, A., Chwatal, S., Dossena, S., Bottà, G., Meyer, G., Maier, B., Valenti, G., Lang, F., and Paulmichl, M. (2004) J. Biol. Chem. 279, 7136-7146) that could be functionally translated to the human system. Here we show the validity of this approach by studying the predicted functional interaction between ICln and HSPC038. In C. elegans, the gene encoding for the ICln homolog (icln-1) is embedded in an operon with two other genes, Nx (the human homolog of Nx is HSPC038) and Ny. ICln is a highly conserved, ubiquitously expressed multifunctional protein that plays a critical role in the regulatory volume decrease after cell swelling. Following hypotonic stress, ICln translocates from the cytosol to the plasma membrane, where it has been proposed to participate in the activation of the swelling-induced chloride current (ICl(swell)). Here we show that the interaction between human ICln and HSPC038 plays a role in volume regulation after cell swelling and that HSPC038 acts as an escort, directing ICln to the cell membrane after cell swelling and facilitating the activation of ICl(swell). Assessment of the NMR structure of HSPC038 showed the presence of a zinc finger motif. Moreover, NMR and additional biochemical techniques enabled us to identify the putative ICln/HSPC038 interacting sites, thereby explaining the functional interaction of both proteins on a molecular level.


Subject(s)
Cell Size , Ion Channels/metabolism , Proteins/metabolism , Cell Membrane/metabolism , Chlorides/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Ion Channels/chemistry , Ion Channels/genetics , Models, Molecular , Osmotic Pressure , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Transport , Proteins/chemistry , Proteins/genetics
17.
Cell Physiol Biochem ; 22(5-6): 579-90, 2008.
Article in English | MEDLINE | ID: mdl-19088440

ABSTRACT

ICln is a ubiquitous, multifunctional protein with functions in cell volume regulation and RNA processing, and is thus part of an intricate protein network critically involved in the homoeostasis of cells. To better understand this vital protein network in health and disease it is fundamental to characterize the interactions between the physiological pathways in which ICln is involved, as well as the spatio-temporal regulation of these interactions. In this study, we focused on the interaction between the two best studied pathways in which ICln is involved--regulatory volume decrease and RNA processing--and asked, whether or not the RNA processing factor and ICln interaction partner LSm4 may also have a function in cell volume regulation in NIH3T3 fibroblasts or HEK293 Phoenix cells. To address this question, we studied in isotonic and hypotonic conditions by FRET, biochemistry and electrophysiology, the intracellular distribution of the RNA processing factor LSm4, its interaction with ICln, as well as the involvement of LSm4 in the activation of the swelling dependent anion and osmolyte channel IClswell. In isotonic conditions, LSm4 associates with ICln, and the plasma membrane. Hypotonic cell swelling leads to the dissociation of LSm4 from the plasma membrane, and from ICln. Over-expression of LSm4 affects the translocation of ICln to the cell membrane and markedly inhibits the activation kinetics and current density of IClswell. These findings indicate that LSm4 not only acts in RNA processing, but also as a co-factor in cell volume regulation.


Subject(s)
Cell Membrane/metabolism , Cell Size , Ribonucleoproteins, Small Nuclear/metabolism , Animals , Cell Line , Humans , Hypotonic Solutions/metabolism , Ion Channels/metabolism , Isotonic Solutions/metabolism , Mice , Protein Binding , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...