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











Publication year range
1.
Regen Med ; 10(7): 815-39, 2015.
Article in English | MEDLINE | ID: mdl-26568079

ABSTRACT

AIM: Identification of mechanistic pathways for selected renal cell (SRC) therapeutic bioactivity in rodent models of chronic kidney disease. MATERIALS & METHODS: In vivo and in vitro functional bioassays applied to investigate regenerative outcomes associated with delivery of SRC to diseased rodent kidney. RESULTS: In vivo, SRC reduces chronic infiltration by monocytes/macrophages. SRC attenuates NF-κB and PAI-1 responses while simultaneously promoting host tubular cell expansion through trophic cues. In vitro, SRC-derived conditioned media attenuates TNF-α-induced NF-κB response, TGF-ß-mediated PAI-1 response and increases expression of transcripts associated with cell cycle regulation. Observed bioactive responses were from vesicle and nonvesicle-associated factors, including specific miRNAs. CONCLUSION: We identify a paracrine mechanism for SRC immunomodulatory and trophic cues on host renal tissues, catalyzing long-term functional benefits in vivo.


Subject(s)
Gene Expression Regulation , Kidney Tubules/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Renal Insufficiency, Chronic/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Disease Models, Animal , Kidney Tubules/pathology , Macrophages/pathology , NF-kappa B/genetics , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Transgenic , Rats, Zucker , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta1/genetics
2.
Trends Biotechnol ; 31(9): 505-14, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23932143

ABSTRACT

Methodologies for the rigorous and quantitative evaluation of biological activity or potency are an essential aspect of the developmental pathway for all biologic product candidates. Such assays typically leverage key mechanistic pathways demonstrated to mediate observed therapeutic outcomes. Tissue engineered/regenerative medicine (TE/RM) therapeutics include cell based therapies as well as engineered tissues and neo-organs for which clarity regarding the mechanism or mechanisms of action may not be forthcoming. Here, we discuss how strategies for the development of potency assays for TE/RM product candidates may harness potential mechanisms of action or other therapeutically relevant bioactivity along with cell number and viability. As the pipeline for TE/RM product candidates expands through 2014 and beyond, the establishment of a defined framework for potency assays will facilitate successful translational outcomes.


Subject(s)
Regenerative Medicine/methods , Regenerative Medicine/trends , Tissue Engineering/methods , Tissue Engineering/standards , Animals , Artificial Organs , Biocompatible Materials , Cell- and Tissue-Based Therapy , Humans
3.
Methods Mol Biol ; 1001: 21-33, 2013.
Article in English | MEDLINE | ID: mdl-23494417

ABSTRACT

Presented below is a methodology for the isolation, expansion, and maintenance of urothelial cells derived from human bladder. Such bladder-derived urothelial cells, taken together with bladder or alternately sourced smooth muscle cells, may be complexed with an appropriately shaped biodegradable scaffold to create regenerative constructs capable of seeding formation of new bladder or bladder-like neo-organs upon implantation in human cystectomy patients.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Regenerative Medicine/methods , Tissue Engineering/methods , Urinary Bladder/cytology , Urothelium/cytology , Cryopreservation/methods , Humans
4.
Methods Mol Biol ; 1001: 35-42, 2013.
Article in English | MEDLINE | ID: mdl-23494418

ABSTRACT

Pulsatile cell bodies, three-dimensional cell clusters with satellite streaming cells, can be isolated from -esophageal tissue. One of the key features of these clusters is that they pulsate at rhythmic rates and demonstrate contractility under several in vitro conditions. Their ability to pulsate appears to be due to the presence of interstitial cells of Cajal (ICC), which mediate signal transmission from nerve to muscle cells. As predicted, the cells comprising these clusters express phenotypic and genotypic markers characteristic of smooth and skeletal muscle, neuronal, and epithelial cells. Because of the critical role of ICC in gastrointestinal tract motility, loss of function in these cells can result in a variety of pathologies. Cultures of pulsatile cell bodies may have utility as an in vitro model to study tissue engineering and regenerative medicine approaches to treating defects in gastrointestinal rhythmicity due to disease or injury.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/methods , Esophagus/cytology , Interstitial Cells of Cajal/physiology , Regenerative Medicine/methods , Tissue Engineering/methods , Cell Communication/physiology , Humans
5.
Methods Mol Biol ; 1001: 189-96, 2013.
Article in English | MEDLINE | ID: mdl-23494430

ABSTRACT

Regenerative medicine and tissue engineering approaches for solving current medical dilemmas such as organ failure, congenital defect, or reconstruction following disease or trauma typically require specific considerations regarding biomaterial selection, identification of key cell types, and applicable surgical techniques (Lanza et al. Principles of tissue engineering, Academic, 2007; Kikuchi, Kanama., Quart Rev 24:51-67, 2007). The ability to evaluate these components in vitro under conditions which simulate relevant in vivo environments can reduce development risks including time and money costs associated with early-stage product development. Similarly, such methods can be useful in making progress in researching features of natural and synthetic biomaterial such as porosity, strength, surface topography, and functionalization, and their singular or collective effects on cell behavior (Kikuchi and Kanama., Quart Rev 24:51-67, 2007; Furth et al. Biomaterials 28:5068-5073, 2007; Mieszawska and Kaplan., BMC Biol 8:59, 2010).Adhesion, migration, and gene and protein expression are all cell behaviors that can be affected by properties of a chosen biomaterial and vary based upon organ system (Cornwell et al. J Biomater Res 71A:55-62, 2004; David et al. Tissue Eng 8(5):787-798, 2002). Understanding of these properties and their role in combination with biomaterial in remodeling is sought in order to fully harness and direct regeneration (Lanza et al. Principles of tissue engineering, Academic Press, 2007; Mieszawska and Kaplan. BMC Biol 8:59, 2010; Matragotri and Lahann J. Nat Mater 8:15-23, 2009).


Subject(s)
Biocompatible Materials/metabolism , Cell Communication/physiology , Cell Movement/physiology , Gene Expression Regulation/physiology , Regenerative Medicine/methods , Tissue Engineering/methods , Cell Adhesion/physiology , Humans
6.
Methods Mol Biol ; 1001: 311-24, 2013.
Article in English | MEDLINE | ID: mdl-23494440

ABSTRACT

Regenerative constructs composed of synthetically sourced, biodegradable biomaterials seeded with smooth muscle-like cells have been leveraged to mediate regeneration of bladder and bladder-like neo-organs. Here, we describe how such constructs may be applied to catalyze regeneration of esophagus and small intestine in preclinical rodent models.


Subject(s)
Esophagus/cytology , Intestine, Small/cytology , Regenerative Medicine/methods , Short Bowel Syndrome/therapy , Tissue Engineering/methods , Animals , Esophagus/injuries , Rats
7.
Tissue Eng Part A ; 18(9-10): 1025-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22136657

ABSTRACT

Urinary pathology requiring urinary diversion, partial or full bladder replacement, is a significant clinical problem affecting ~14,000 individuals annually in the United States alone. The use of gastrointestinal tissue for urinary diversion or bladder reconstruction/replacement surgeries is frequently associated with complications. To try and alleviate or reduce the frequency of these complications, tissue engineering and regenerative medicine strategies have been developed using bio-absorbable materials seeded with cells derived from the bladder. However, bladder-sourced cells may not always be suitable for such applications, especially in patients with bladder cancer. In this study, we describe the isolation and characterization of smooth muscle cells (SMCs) from porcine adipose and peripheral blood that are phenotypically and functionally indistinguishable from bladder-derived SMCs. In a preclinical Good Laboratory Practice study, we demonstrate that autologous adipose- and peripheral blood-derived SMCs may be used to seed synthetic, biodegradable tubular scaffold structures and that implantation of these seeded scaffolds into a porcine cystectomy model leads to successful de novo regeneration of a tubular neo-organ composed of urinary-like neo-tissue that is histologically identical to native bladder. The ability to create urologic structures de novo from scaffolds seeded by autologous adipose- or peripheral blood-derived SMCs will greatly facilitate the translation of urologic tissue engineering technologies into clinical practice.


Subject(s)
Adipose Tissue/cytology , Guided Tissue Regeneration/methods , Tissue Engineering/methods , Urinary Bladder/surgery , Animals , Female , Fluorescent Antibody Technique , Male , Myocytes, Smooth Muscle/cytology , Reverse Transcriptase Polymerase Chain Reaction , Swine , Tissue Scaffolds/chemistry
8.
Med Hypotheses ; 78(2): 231-4, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22100629

ABSTRACT

Recent successes in regenerative medicine and tissue engineering of bladder and bladder-like neo-organs have leveraged regenerative constructs composed of a biodegradable scaffold seeded with a population of smooth muscle cells. We have shown that such smooth muscle cells are isolatable from adipose and other sources alternate to the primary organ. We hypothesize that this regenerative platform is not limited to regeneration of bladder and bladder-like neo-organs, but rather represents a foundational technology platform broadly applicable for regeneration of laminarly organized hollow organs. Using esophagus as an illustrative example in support of this hypothesis, we demonstrate that patch constructs composed of adipose-derived smooth muscle cells seeded on a biodegradable matrix catalyze complete regeneration of the esophageal wall in a rodent model of esophageal injury. By implication, such regenerative constructs may potentially be used to mediate the regeneration of any laminarly organized tubular organ.


Subject(s)
Esophagus/physiology , Regeneration/physiology , Tissue Engineering/methods , Tissue Scaffolds , Urinary Bladder/physiology , Absorbable Implants , Animals , Female , Myocytes, Smooth Muscle/pathology , Rats , Rats, Inbred Lew , Regenerative Medicine , Tissue Engineering/instrumentation
9.
Regen Med ; 6(6): 721-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22050524

ABSTRACT

AIMS: To apply an organ regeneration platform technology of autologous smooth muscle cell/biomaterial combination products, previously demonstrated to be successful for urinary tissue regeneration, to the regeneration of the small intestine. MATERIALS & METHODS: Patch and tubular constructs were implanted in rodent small intestines and histologically evaluated over a time course for evidence of regeneration of the laminarly organized neo-mucosa and muscle layers. RESULTS: Laminarly organized neo-mucosa and muscle layer bundles are demonstrated as early as 8 weeks postimplantation. CONCLUSION: An organ regeneration technology platform of autologous smooth muscle cell/biomaterial combination products can be extended to the regeneration of the small intestine.


Subject(s)
Intestine, Small/physiology , Myocytes, Smooth Muscle/cytology , Prosthesis Implantation , Regeneration/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Anastomosis, Surgical , Animals , Female , Gene Expression Regulation , Intestine, Small/cytology , Intestine, Small/surgery , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/transplantation , Myocytes, Smooth Muscle/ultrastructure , Rats , Rats, Inbred Lew
10.
Lipids Health Dis ; 10: 171, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21957910

ABSTRACT

BACKGROUND: Therapeutically bioactive cell populations are currently understood to promote regenerative outcomes in vivo by leveraging mechanisms of action including secretion of growth factors, site specific engraftment and directed differentiation. Constitutive cellular populations undoubtedly participate in the regenerative process. Adipose tissue represents a source of therapeutically bioactive cell populations. The potential of these cells to participate in various aspects of the regenerative process has been demonstrated broadly. However, organ association of secretory and developmental markers to specific peri-organ adipose depots has not been investigated. To characterize this topographical association, we explored the potential of cells isolated from the stromal vascular fraction (SVF) of kidney sourced adipose to express key renal associated factors. RESULTS: We report that renal adipose tissue is a novel reservoir for EPO expressing cells. Kidney sourced adipose stromal cells demonstrate hypoxia regulated expression of EPO and VEGF transcripts. Using iso-electric focusing, we demonstrate that kidney and non-kidney sourced adipose stromal cells present unique patterns of EPO post-translational modification, consistent with the idea that renal and non-renal sources are functionally distinct adipose depots. In addition, kidney sourced adipose stromal cells specifically express the key renal developmental transcription factor WT1. CONCLUSIONS: Taken together, these data are consistent with the notion that kidney sourced adipose stromal (KiSAS) cells may be primed to recreate a regenerative micro-environment within the kidney. These findings open the possibility of isolating solid-organ associated adipose derived cell populations for therapeutic applications in organ-specific regenerative medicine products.


Subject(s)
Adipocytes, White/metabolism , Gene Expression Regulation , Intra-Abdominal Fat/cytology , Kidney/cytology , Regenerative Medicine/methods , Adipocytes, White/cytology , Animals , Biomarkers , Cell Hypoxia , Cell Separation , Cells, Cultured , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Intra-Abdominal Fat/metabolism , Kidney/metabolism , Male , Organ Specificity , Protein Processing, Post-Translational , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
11.
J Cell Physiol ; 226(10): 2702-11, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21792927

ABSTRACT

Myocardin (MYOCD) is a smooth and cardiac muscle-specific transcriptional coactivator that is required for the proper expression of contraction-related genes. Through its function to transactivate effector genes, MYOCD plays an essential role in mediating the switch between contractile and non-contractile phenotypes, particularly in smooth muscle cells (SMC). There are at least two known transcript variants of MYOCD that are expressed in SMC, differing only by the presence (+) or absence (Δ) of Exon 11. To date, no functional role has been assigned to the domain encoded by Exon 11, nor have any notable differences between the ability of each isoform to activate contraction-related genes been observed. In this study we compared sequences for Exon 11 among several mammalian species and identified a highly conserved, putative target sequence for glycogen synthase kinase 3 (GSK3) phosphorylation, suggesting a regulatory role for Exon 11 that can be modulated by alternative splicing. The function of Exon 11 was investigated by altering MYOCD splice selection in cultured porcine SMC with small interfering RNAs (siRNA) and specific chemical inhibitors, resulting in a relative increase in expression of ΔExon 11 variants in the endogenous pool of MYOCD mRNA. The relative increase in ΔExon 11 mRNAs correlated with a reduction of contractile phenotype in the porcine SMC as evidenced by morphological assessment and molecular analysis of effector genes. Together, these data suggest that MYOCD ΔExon 11 may participate in modulating SMC phenotype, potentially acting as a dominant-negative repressor of contraction-related genes.


Subject(s)
Alternative Splicing/physiology , Myocytes, Smooth Muscle/physiology , Nuclear Proteins/genetics , Trans-Activators/genetics , Amino Acid Sequence , Animals , Aorta/cytology , Carotid Arteries/cytology , Conserved Sequence , Genetic Variation , In Vitro Techniques , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenotype , Swine , Trans-Activators/chemistry , Trans-Activators/metabolism , Urinary Bladder/cytology
12.
Tissue Eng Part C Methods ; 17(8): 843-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21595545

ABSTRACT

Adipose tissue contains a heterogeneous cell population composed of endothelial cells, adipocytes, smooth muscle cells (SMC), and mesenchymal progenitors and stromal cells that meet the criteria put forth by the International Society for Cellular Therapy as defining mesenchymal stem cells (MSC). In this study, we expanded the stromal vascular fraction (SVF) of human adipose tissue and characterized the resulting adherent primary cell cultures by quantitative reverse transcription-polymerase chain reaction, antigen expression, protein fingerprinting, growth kinetics, in vitro tri-lineage differentiation bioactivity, and functional responses to small molecules modulating SMC-related developmental pathways and compared the results to those obtained with functionally validated MSC cultures. SVF-derived initial cultures (P0) were expanded in a defined medium that was not optimized for MSC growth conditions, neither were recombinant cytokines or growth factors added to the media to direct differentiation. The adherent cell cultures derived from SVF expansion under these conditions had markedly distinct phenotypic and biological properties relative to functionally validated MSC cultures. SVF-derived adherent cell cultures retained characteristics consistent with the SMC subpopulation within adipose tissue--phenotype, gene, and protein expression--that were independent of passage number and source of SVF (n=4 independent donors). SVF-derived cells presented significantly less robust in vitro tri-lineage differentiation bioactivity relative to validated MSC. Expanded SVF cells and MSC had opposite responses to the thromboxane A2 mimetic U46619, demonstrating an unambiguous functional distinction between the two cell types. Taken together, these data support the conclusions that SVF cells expanded under the conditions described in these studies are accurately described as adipose-derived SMC and represent a cellular subpopulation of adipose SVF that is separate and distinct from other classes of adipose-derived cells.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Stromal Cells/cytology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adipocytes/cytology , Biopsy , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Humans , Phenotype , Thromboxane A2/metabolism
13.
Cell Transplant ; 20(11-12): 1771-90, 2011.
Article in English | MEDLINE | ID: mdl-21439130

ABSTRACT

Development of a tissue-engineered neo-kidney augment (NKA) requires evaluation of defined, therapeutically relevant cell and cell/biomaterial composites (NKA constructs) for regenerative potential in mammalian kidney. Previous work identified primary renal cell populations that extended survival and improved renal function in a rodent model of chronic kidney disease (CKD). This study extends that work toward the goal of developing NKA by (i) screening in vivo inflammatory and fibrotic responses to acellular biomaterials delivered to healthy rodent renal parenchyma, (ii) evaluating the functionality of renal cell/biomaterial combinations in vitro, (iii) generating NKA constructs by combining therapeutically relevant cell populations with biocompatible biomaterial, and (iv) evaluating in vivo neokidney tissue development in response to NKA constructs delivered to healthy rodent renal parenchyma. Gelatin and hyaluronic acid (HA)-based hydrogels elicited the least inflammatory and fibrotic responses in renal parenchyma relative to polycaprolactone (PCL) and poly(lactic-co-glycolic acid) (PLGA) beads or particles and were associated with neovascularization and cellular infiltration by 4 weeks postimplantation. Renal cell populations seeded onto gelatin or HA-based hydrogels were viable and maintained a tubular epithelial functional phenotype during an in vitro maturation of 3 days as measured by transcriptomic, proteomic, secretomic, and confocal immunofluorescence assays. In vivo delivery of cell-seeded NKA constructs (bioactive renal cells + gelatin hydrogels) to healthy rodent renal parenchyma elicited neokidney tissue formation at 1 week postimplantation. To investigate a potential mechanism by which NKA constructs could impact a disease state, the effect of conditioned media on TGF-ß signaling pathways related to tubulo-interstitial fibrosis associated with CKD progression was evaluated. Conditioned medium was observed to attenuate TGF-ß-induced epithelial-mesenchymal transition (EMT) in vitro in a human proximal tubular cell line (HK2).


Subject(s)
Kidney/cytology , Tissue Engineering , Animals , Cell Adhesion , Cell Survival , Cells, Cultured , Dogs , Epithelial-Mesenchymal Transition/drug effects , Gelatin/chemistry , Gene Expression Profiling , Humans , Hydrogels/chemistry , Kidney/metabolism , Kidney/pathology , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Proteome/analysis , Rats , Rats, Inbred Lew , Transforming Growth Factor beta/pharmacology
14.
Appl Immunohistochem Mol Morphol ; 19(2): 184-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20930617

ABSTRACT

Bladder tissue has been regenerated in humans with neurogenic bladder using an implant produced from autologous urothelial (UC) and smooth muscle cells (SMC) expanded from bladder biopsies seeded onto a biodegradable synthetic scaffold. As the majority of bladder cancers are urothelial carcinomas (aka, transitional cell carcinoma), this 2-cell type autologous sourcing strategy presents significant challenges to product development. Entire bladders have been regenerated in cystectomized animals using a single-cell-type sourcing strategy: implants were seeded with bladder-derived SMC-only. Applying the bladder SMC-only sourcing strategy to produce clinical implants for bladder replacement or urinary diversion in bladder cancer patients requires methods for screening SMC cultures for the presence of potentially cancerous UC cells to provide evidence of SMC culture purity before seeding the scaffold. In this report, we show a 10-fold to 100-fold improvement in the sensitivity of qualitative and quantitative reverse-transcription PCR (qRT-PCR)-based assays for detecting UC positive for Cytokeratin 5 (CK5) in mixed SMC/UC cultures when the cell population was first subjected to magnetic activated cell sorting to enrich for cells expressing the epithelial cell adhesion molecule (known as EPCAM or CD326), a marker known to be present in normal UC and upregulated in the cancerous UC.


Subject(s)
Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Cell Adhesion Molecules/analysis , Keratin-5/analysis , Myocytes, Smooth Muscle/pathology , Urothelium/pathology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Culture Techniques , Epithelial Cell Adhesion Molecule , Flow Cytometry , Humans , Keratin-5/genetics , Keratin-5/metabolism , Magnetics , Myocytes, Smooth Muscle/metabolism , Organ Culture Techniques , Regeneration , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Engineering/methods , Tissue Scaffolds , Transplantation, Autologous , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder, Neurogenic/genetics , Urinary Bladder, Neurogenic/metabolism , Urinary Bladder, Neurogenic/pathology , Urothelium/metabolism
15.
Tissue Eng Part C Methods ; 16(1): 33-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19351240

ABSTRACT

Suspensions of viable urothelial cells (UC) isolated from patient bladder biopsies often contain considerable amounts of extraneous materials comprised of cellular debris, dead and dying UC, and red blood cells. We have consistently observed an inversely proportional relationship between UC attachment efficiency and the amount of extraneous materials in the suspension; viable UC cell attachment efficiency decreases as the amount of extraneous materials in the cell suspension increases. Processing the initial cell isolate to reduce the amount of extraneous materials can enrich for viable UC capable of attaching and proliferating in ex vivo cultures. In this report, we describe the isolation of an enriched population of viable UC from nonneurogenic and neurogenic bladder tissue biopsies using iodixanol self-generated density gradients (OptiPrep), and characterization by trypan blue exclusion, fluorescence-activated cell sorting, immunofluorescence, and growth kinetics.


Subject(s)
Triiodobenzoic Acids/pharmacology , Urinary Bladder, Neurogenic/pathology , Urinary Bladder/pathology , Urothelium/pathology , Biopsy , Cell Separation , Cell Survival , Cells, Cultured , Centrifugation, Density Gradient , Contrast Media/pharmacology , Flow Cytometry , Humans , Kinetics , Microscopy, Fluorescence/methods , Regenerative Medicine , Tissue Engineering/methods , Triiodobenzoic Acids/chemistry , Trypan Blue/pharmacology
16.
Am J Pathol ; 166(6): 1841-50, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15920168

ABSTRACT

Dynamic instability of microtubules is critical for mitotic spindle assembly and disassembly during cell division, especially in rapidly dividing tumor cells. Microtubule-associated proteins (MAPs) are a family of proteins that influence this property. We showed previously that MAP2, a neuron-specific protein that stabilizes microtubules in the dendrites of postmitotic neurons, is induced in primary cutaneous melanoma but is absent in metastatic melanomas. We proposed that induction of a microtubule-stabilizing protein in primary melanoma could disrupt the dynamic instability of microtubules, inhibit cell division and prevent or delay tumor progression. Here we show, by Kaplan-Meier survival and multivariate Cox regression analysis, that patients diagnosed with MAP2+ primary melanomas have significantly better metastatic disease-free survival than those with MAP2- disease. Investigation of the mechanisms that underlie the effect of MAP2 on melanoma progression showed that MAP2 expression in metastatic melanoma cell lines leads to microtubule stabilization, cell cycle arrest in G2-M phase and growth inhibition. Disruption of microtubule dynamics by MAP2 resulted in multipolar mitotic spindles, defects in cytokinesis and accumulation of cells with large nuclei, similar to those seen in vivo in MAP2+ primary melanomas cells. These data suggest that ectopic activation of a neuronal differentiation gene in melanoma during early tumor progression inhibits cell division and correlates with inhibition or delay of metastasis.


Subject(s)
Biomarkers, Tumor/analysis , Cell Proliferation , Melanoma/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Invasiveness/pathology , Skin Neoplasms/metabolism , Apoptosis/physiology , Blotting, Western , Cell Differentiation , Cell Line, Tumor , Flow Cytometry , Humans , Immunohistochemistry , Melanoma/mortality , Melanoma/pathology , Mitosis/physiology , Neurons/cytology , Prognosis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Survival Analysis
17.
Melanoma Res ; 14(6): 509-16, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15577322

ABSTRACT

Melastatin 1 (MLSN1), originally identified as melanoma metastasis suppressor, represents a TRPM subfamily of transient receptor potential (TRP) proteins which serve diverse biological roles in a wide variety of cell types. Down-regulation of MLSN1 expression in human cutaneous melanoma, as indicated by in situ hybridization, appears to be a prognostic marker for melanoma metastasis. However, the exact physiological function(s) of MLSN1, the mechanism(s) involved in the regulation of its expression and its role in melanoma tumour progression are not yet clear. In this study, we identified a 654 bp upstream sequence of MLSN1, containing four E boxes (E1-E4), including an 11 bp M box (E4), that is sufficient for melanocyte-specific transcription and activation by the melanocyte transcription factor MITF (a bHLH-zip factor). Deletion analysis showed that the two distal E boxes (E3 and E4) in the MLSN1 promoter are required for both its activation by MITF and its constitutive activity in melanoma cells. Western blot analysis using polyclonal rabbit anti-human MLSN1 antibodies identified several polypeptides, presumably generated by both alternative splicing of MLSN1 messenger RNA (mRNA) and proteolytic cleavage, in both melanocytes and metastatic melanoma cells. Thus, multiple mechanisms appear to regulate MLSN1 expression in melanocytes and melanoma cells.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Melanocytes/physiology , Melanoma/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Alternative Splicing , Animals , Base Sequence , Blotting, Western , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , HeLa Cells , Helix-Loop-Helix Motifs , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/metabolism , Mice , Microphthalmia-Associated Transcription Factor , Molecular Sequence Data , NIH 3T3 Cells , Neoplasm Proteins/metabolism , Protein Isoforms , RNA, Messenger , Rabbits , Sequence Deletion , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , TRPM Cation Channels , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL