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










Publication year range
1.
Stem Cells Transl Med ; 13(1): 14-29, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38071447

ABSTRACT

Perinatal derivatives have been proposed as adjunct therapeutic strategies or innovative treatments. Undoubtedly, perinatal derivatives can offer the opportunity and source material to isolate multipotent stem cells, but both maternal- and fetal-derived tissues can be processed and transformed into engineered tissues or advanced biomedical devices, whose potential remains to be fully elucidated. Promising preclinical and clinical results collected so far clearly foresee an escalation of such novel treatments. Market forecasts predict exponential growth in such advanced medicinal products during the next decade, with a pragmatic innovation for medicine into a more advanced biomedical version, enlarging the portfolio for treating a wide range of congenital and acute conditions. However, all these promising and fascinating therapeutic possibilities cannot gain a solid and recognized role in established medical practice without rigid and harmonized manufacturing strategies. The implementation of strategies according to guidelines and directives compiled by Regulatory Agencies, in conformity to (European) Pharmacopoeia and for Good Manufacturing Practice -conforming production of such products, represent critical steps required to translate perinatal technologies into effective therapeutic approaches. During the past 5 years, a panel of European experts and developers, gathered under the umbrella of the COST Sprint Action, supported by the European Cooperation in Science and Technology action, had the opportunity to revise and summarize experience and recommendations for a fruitful and proficient generation of perinatal biomedical products. In order to facilitate the creation and potential commercialization of perinatal bioengineered and advanced pharmaceutical products and technologies, such a collection of data and recommendations is described and discussed here.


Subject(s)
Medicine , Tissue Engineering , Pregnancy , Female , Humans
2.
Stem Cells Transl Med ; 12(5): 258-265, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37027834

ABSTRACT

Perinatal derivatives (PnD) are drawing growing interest among the scientific community as an unrestricted source of multipotent stem cells, secretome, and biological matrices. They are useful for the treatment of diseases that currently have limited or no effective therapeutic options, but they require the development of regenerative approaches. With this development, the question of regulation of donation, processing, and distribution has therefore become more important. Within the European Cooperation in Science and Technology (COST) community, we compiled a group of international experts on PnD technologies, who revised and compared existing EU national regulations. Notably, despite clear European directives, each EU Country has developed their own implementation and standard levels for cell- and tissue-based therapies. To enable extended applications of PnD treatments within the EU community and worldwide, harmonization is highly recommended. This paper aims to provide an overview of the various options available to introduce PnD into clinical practice. For this purpose, the different aspects resulting from (1) the type of PnD, (2) the amount of available data, (3) the degree of manipulation, and (4) the intended application and the process toward a possible commercialization will be presented. In the future, it will be important to find a balance between regulatory requirements and the best medical quality of the PnD product.


Subject(s)
Cell- and Tissue-Based Therapy , European Union
3.
Int J Mol Sci ; 24(7)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37047181

ABSTRACT

Unsuccessful wound closure in chronic wounds can be linked to altered keratinocyte activation and their inability to re-epithelize. Suggested mechanisms driving this impairment involve unbalanced cytokine signaling. However, the molecular events leading to these aberrant responses are poorly understood. Among cytokines affecting keratinocyte responses, Transforming Growth Factor-ß (TFG-ß) is thought to have a great impact. In this study, we have used a previously characterized skin epidermal in vitro model, HaCaT cells continuously exposed to TGF-ß1, to study the wound recovery capabilities of chronified/senescent keratinocytes. In this setting, chronified keratinocytes show decreased migration and reduced activation in response to injury. Amniotic membrane (AM) has been used successfully to manage unresponsive complicated wounds. In our in vitro setting, AM treatment of chronified keratinocytes re-enabled migration in the early stages of wound healing, also promoting proliferation at later stages. Interestingly, when checking the gene expression of markers known to be altered in TGF-ß chronified cells and involved in cell cycle regulation, early migratory responses, senescence, and chronic inflammation, we discovered that AM treatment seemed to reset back to keratinocyte status. The analysis of the evolution of both the levels of keratinocyte activation marker cytokeratin 17 and the spatial-temporal expression pattern of the proliferation marker Ki-67 in human in vivo biopsy samples suggests that responses to AM recorded in TGF-ß chronified HaCaT cells would be homologous to those of resident keratinocytes in chronic wounds. All these results provide further evidence that sustained TGF-ß might play a key role in wound chronification and postulate the validity of our TGF-ß chronified HaCaT in vitro model for the study of chronic wound physiology.


Subject(s)
Amnion , Keratinocytes , Humans , Amnion/metabolism , Keratinocytes/metabolism , Skin/metabolism , Wound Healing/physiology , Transforming Growth Factor beta/metabolism , Cell Movement
4.
Front Bioeng Biotechnol ; 10: 977590, 2022.
Article in English | MEDLINE | ID: mdl-36304904

ABSTRACT

Perinatal derivatives are drawing growing interest among the scientific community as an unrestricted source of multipotent stromal cells, stem cells, cellular soluble mediators, and biological matrices. They are useful for the treatment of diseases that currently have limited or no effective therapeutic options by means of developing regenerative approaches. In this paper, to generate a complete view of the state of the art, a comprehensive 10-years compilation of clinical-trial data with the common denominator of PnD usage has been discussed, including commercialized products. A set of criteria was delineated to challenge the 10-years compilation of clinical trials data. We focused our attention on several aspects including, but not limited to, treated disorders, minimal or substantial manipulation, route of administration, dosage, and frequency of application. Interestingly, a clear correlation of PnD products was observed within conditions, way of administration or dosage, suggesting there is a consolidated clinical practice approach for the use of PnD in medicine. No regulatory aspects could be read from the database since this information is not mandatory for registration. The database will be publicly available for consultation. In summary, the main aims of this position paper are to show possibilities for clinical application of PnD and propose an approach for clinical trial preparation and registration in a uniform and standardized way. For this purpose, a questionnaire was created compiling different sections that are relevant when starting a new clinical trial using PnD. More importantly, we want to bring the attention of the medical community to the perinatal products as a consolidated and efficient alternative for their use as a new standard of care in the clinical practice.

5.
Front Bioeng Biotechnol ; 10: 965006, 2022.
Article in English | MEDLINE | ID: mdl-35992360

ABSTRACT

Perinatal derivatives (PnD) are birth-associated tissues, such as placenta, umbilical cord, amniotic and chorionic membrane, and thereof-derived cells as well as secretomes. PnD play an increasing therapeutic role with beneficial effects on the treatment of various diseases. The aim of this review is to elucidate the modes of action of non-hematopoietic PnD on inflammation, angiogenesis and wound healing. We describe the source and type of PnD with a special focus on their effects on inflammation and immune response, on vascular function as well as on cutaneous and oral wound healing, which is a complex process that comprises hemostasis, inflammation, proliferation (including epithelialization, angiogenesis), and remodeling. We further evaluate the different in vitro assays currently used for assessing selected functional and therapeutic PnD properties. This review is a joint effort from the COST SPRINT Action (CA17116) with the intention to promote PnD into the clinics. It is part of a quadrinomial series on functional assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer activities, anti-inflammation, wound healing, angiogenesis, and regeneration.

6.
Front Bioeng Biotechnol ; 9: 685128, 2021.
Article in English | MEDLINE | ID: mdl-34178969

ABSTRACT

Due to its intrinsic properties, there has been growing interest in human amniotic membrane (hAM) in recent years particularly for the treatment of ocular surface disorders and for wound healing. Herein, we investigate the potential use of hAM and amnion-chorion membrane (ACM) in oral surgery. Based on our analysis of the literature, it appears that their applications are very poorly defined. There are two options: implantation or use as a cover material graft. The oral cavity is submitted to various mechanical and biological stimulations that impair membrane stability and maintenance. Thus, some devices have been combined with the graft to secure its positioning and protect it in this location. This current opinion paper addresses in detail suitable procedures for hAM and ACM utilization in soft and hard tissue reconstruction in the oral cavity. We address their implantation and/or use as a covering, storage format, application side, size and number, multilayer use or folding, suture or use of additional protective covers, re-application and resorption/fate. We gathered evidence on pre- and post-surgical care and evaluation tools. Finally, we integrated ophthalmological and wound healing practices into the collected information. This review aims to help practitioners and researchers better understand the application of hAM and ACM in the oral cavity, a place less easily accessible than ocular or cutaneous surfaces. Additionally, it could be a useful reference in the generation of new ideas for the development of innovative protective covering, suturing or handling devices in this specific indication. Finally, this overview could be considered as a position paper to guide investigators to fulfill all the identified criteria in the future.

7.
Cells ; 9(2)2020 01 28.
Article in English | MEDLINE | ID: mdl-32012802

ABSTRACT

Chronic wounds are characterized for their incapacity to heal within an expected time frame. Potential mechanisms driving this impairment are poorly understood and current hypotheses point to the development of an unbalanced milieu of growth factor and cytokines. Among them, TGF-ß is considered to promote the broadest spectrum of effects. Although it is known to contribute to healthy skin homeostasis, the highly context-dependent nature of TGF-ß signaling restricts the understanding of its roles in healing and wound chronification. Historically, low TGF-ß levels have been suggested as a pattern in chronic wounds. However, a revision of the available evidence in humans indicates that this could constitute a questionable argument. Thus, in chronic wounds, divergences regarding skin tissue compartments seem to be characterized by elevated TGF-ß levels only in the epidermis. Understanding how this aspect affects keratinocyte activities and their capacity to re-epithelialize might offer an opportunity to gain comprehensive knowledge of the involvement of TGF-ß in chronic wounds. In this review, we compile existing evidence on the roles played by TGF-ß during skin wound healing, with special emphasis on keratinocyte responses. Current limitations and future perspectives of TGF-ß research in chronic wounds are discussed.


Subject(s)
Keratinocytes/pathology , Skin/metabolism , Skin/pathology , Transforming Growth Factor beta/metabolism , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Animals , Chronic Disease , Humans , Wound Healing
8.
Cells ; 9(1)2020 01 20.
Article in English | MEDLINE | ID: mdl-31968599

ABSTRACT

Defects in wound closure can be related to the failure of keratinocytes to re-epithelize. Potential mechanisms driving this impairment comprise unbalanced cytokine signaling, including Transforming Growth Factor-ß (TFG-ß). Although the etiologies of chronic wound development are known, the relevant molecular events are poorly understood. This lack of insight is a consequence of ethical issues, which limit the available evidence to humans. In this work, we have used an in vitro model validated for the study of epidermal physiology and function, the HaCaT cells to provide a description of the impact of sustained exposure to TGF-ß. Long term TGF-ß1 treatment led to evident changes, HaCaT cells became spindle-shaped and increased in size. This phenotype change involved conformational re-arrangements for actin filaments and E-Cadherin cell-adhesion structures. Surprisingly, the signs of consolidated epithelial-to-mesenchymal transition were absent. At the molecular level, modified gene expression and altered protein contents were found. Non-canonical TGF-ß pathway elements did not show relevant changes. However, R-Smads experienced alterations best characterized by decreased Smad3 levels. Functionally, HaCaT cells exposed to TGF-ß1 for long periods showed cell-cycle arrest. Yet, the strength of this restraint weakens the longer the treatment, as revealed when challenged by pro-mitogenic factors. The proposed setting might offer a useful framework for future research on the mechanisms driving wound chronification.


Subject(s)
Cell Cycle Checkpoints , Epithelial-Mesenchymal Transition/drug effects , Keratinocytes/cytology , Skin/cytology , Transforming Growth Factor beta/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial-Mesenchymal Transition/genetics , HaCaT Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Phenotype , Signal Transduction , Smad3 Protein/metabolism , Transcription, Genetic/drug effects
9.
J Wound Care ; 27(12): 806-815, 2018 12 02.
Article in English | MEDLINE | ID: mdl-30557111

ABSTRACT

OBJECTIVE: The amniotic membrane (AM) is a tissue with low immunogenity and high therapeutic potential due to its anti-inflammatory, anti-fibrotic and antimicrobial effects. This paper describes the use of cryopreserved amniotic membrane allografts to treat diabetic foot ulcers (DFUs) in patients with diabetes. METHOD: In this case series, AM was processed to obtain a final medicinal product: cryopreserved amniotic membrane. cryopreserved AM was applied every 7-10 days until total epithelialisation of the DFUs. RESULTS: A total of 14 patients with DFUs (median size: 12.30cm, (range: 0.52-42.5cm2) were treated and followed up until complete closure (median time: 20 weeks, range: 7-56 weeks). Patients received 4-40 AM applications. All patients in this study achieved complete epithelialisation of the wound. No adverse events were observed. CONCLUSION: AM is a feasible and safe treatment in complex DFUs. Furthermore, the treatment is successful in achieving epithelialisation of long-evolution, unhealed wounds resistant to conventional therapies.


Subject(s)
Allografts/transplantation , Amnion/transplantation , Cryopreservation/methods , Diabetic Foot/surgery , Wound Healing/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prospective Studies , Spain , Treatment Outcome , Young Adult
10.
Sci Rep ; 7(1): 15262, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127427

ABSTRACT

During wound healing, the migration of keratinocytes onto newly restored extracellular matrix aims to reestablish continuity of the epidermis. The application of amniotic membrane (AM) to chronic, deep traumatic, non-healing wounds has proven successful at stimulating re-epithelialization. When applied on epithelial cell cultures, AM activates extracellular signal-regulated kinases 1/2 (ERK1/2) and c-Jun N-terminal kinases 1/2 (JNK1/2), with the overexpression and phosphorylation of c-Jun along the wound edge. The effect of AM on the migration of cells was investigated by studying critical proteins involved in the focal adhesions turn-over: Focal Adhesion Kinase (FAK), Paxillin and Vinculin. In Mv1Lu and HaCaT cells, validated models for cell migration and wound healing, AM affected the expression and activation of Paxillin, but did not affect Vinculin expression, both factors which integrate into focal adhesions. Moreover, AM regulation also affected FAK activity through phosphorylation. Finally, we have determined that AM regulation of focal adhesions involves both JNK and MEK MAP kinase signaling pathways. This data provides a molecular background to understand how AM regulates critical cell and molecular aspects of cell migration, organizing and directing the movement of cells by the continuous formation, maturation, and turnover of focal adhesion structures at the migration leading edge.


Subject(s)
Amnion/chemistry , Cell Movement , Epithelial Cells/metabolism , Focal Adhesions/metabolism , MAP Kinase Signaling System , Wound Healing , Animals , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Mink , Paxillin/metabolism , Vinculin/metabolism
11.
Placenta ; 59: 146-153, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28413063

ABSTRACT

Wound healing usually follows a predictable sequence and prognosis of events. Its evolutionary process is the result of a complicated interaction between patient-related factors, the wound, the treatment used and the skills and knowledge of the professionals who treat them. Only through a meticulous initial assessment of the wound is it possible to identify the factors that contribute to its complexity. The challenge for professionals will be to implement efficient therapies at the right time and in the most cost-efficient way in order to reduce associated problems, treat the symptoms and expectations of the patients and achieve adequate wound healing whenever possible. This is particularly evident in big chronic wounds with considerable tissue loss, which become senescent in the process of inflammation or proliferation losing the ability to epithelialize. Generally, chronic wounds do not respond to current treatments, therefore they need special interventions. AM is a tissue of particular interest as a biological dressing and it has well-documented reepithelialization effects which are in part related to its capacity to synthesize and release biological active factors. Our studies have demonstrated that amniotic membrane (AM) is able to induce epithelialization in chronic wounds that were unable to epithelialize. AM induces several signaling pathways that are involved in cell migration and/or proliferation. Additionally, AM is able to selectively antagonize the anti-proliferative effect of transforming growth factor-ß (TGF-ß) by modifying the genetic program that TGF-ß induces on keratinocytes. The combined effect of AM on keratinocytes, promoting cell proliferation/migration and antagonizing the effect of TGF-ß is the perfect combination, allowing chronic wounds to move out of their non-healing state and progress into epithelialization.


Subject(s)
Amnion , Biological Dressings , Skin Ulcer/therapy , Wound Healing , Humans
12.
Cell Transplant ; 22(8): 1351-67, 2013.
Article in English | MEDLINE | ID: mdl-23031712

ABSTRACT

Human amniotic epithelial cells (hAECs) have been the object of intense research due to their potential therapeutic use. In this paper, we present molecular evidence of a bona fide epithelial to mesenchymal transition (EMT) undergone by hAECs. Amniotic membrane (AM)-derived hAECs showed the presence of typical epithelial markers such as E-cadherin and cytokeratins. hAECs in culture, however, underwent morphological changes acquiring a mesenchymal shape. Epithelial cell markers were lost and typical mesenchymal markers, such as vimentin and α-SMA, appeared. Several genes associated with EMT, such as SNAI1, MMP9, PAI1, or ACTA2, increased their expression. The expression of the transcription activators KLF4 or MTA3 was consistent with the downregulation of CDH1. We have shown that hAECs undergo EMT due to the autocrine production of TGF-ß. Furthermore, the addition of the TGF-ß receptor I (ALK5) inhibitor SB-431542 or TGF-ß neutralizing antibody to hAECs prevented EMT and preserved the hAECs' epithelial phenotype. Altogether, these results suggest that cultured hAECs undergo EMT through the autocrine production of TGF-ß.


Subject(s)
Amnion/cytology , Autocrine Communication , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta/metabolism , Antibodies, Neutralizing/pharmacology , Autocrine Communication/drug effects , Benzamides , Biomarkers/metabolism , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Dioxoles , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Epithelium/drug effects , Epithelium/metabolism , Gene Expression Regulation/drug effects , Humans , Kruppel-Like Factor 4 , Mesoderm/drug effects , Mesoderm/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics
13.
Stem Cells Dev ; 21(2): 260-72, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21504358

ABSTRACT

The density gradient centrifugation method was originally designed for the isolation of mononuclear peripheral blood cells and rapidly adapted to fractionate bone marrow (BM) cells. This method involves the use of gradient density solutions with low viscosity and low osmotic pressure that allows erythrocytes and more mature cells gravitate to the bottom at a density fraction superior to 1.080 g/dL; mononuclear cells (MNCs) held in the plasma-solution to interphase at a density between 1.053 and 1.073 g/dL; plasma, dilution medium and anticoagulant to occupy a density less than 1.050 g/dL and the fat cells to float due to their very low density. BM-mesenchymal stem cells (MSCs) are usually obtained after the separation and cultures of BM-MNCs from the plasma-solution interphase, which is traditionally considered the only source of progenitor cells (hematopoietic and nonhematopoietic). In this study evidences that MSCs could be isolated from the very low-density cells of the fat layer are presented. In addition, we demonstrated that the MSCs obtained from these cells have similar immunophenotypic characteristics, and similar proliferative and differentiation potential to those obtained from the MNCs at plasma-solution interphase. The method represents a simple and cost effective way to increase the MSCs yield from each BM donor, without the need to look for other sources, additional manipulation of cells, and risks of contamination or disturbances of the potential of differentiation. These cells might serve as a complementary source of MSCs to facilitate preclinical and clinical application in tissue engineering and cell therapy.


Subject(s)
Bone Marrow Cells/cytology , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Bone Marrow Cells/physiology , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Centrifugation, Density Gradient , Child , Female , Humans , Immunophenotyping , Male , Mesenchymal Stem Cells/physiology , Middle Aged
14.
Wound Repair Regen ; 18(4): 368-77, 2010.
Article in English | MEDLINE | ID: mdl-20636551

ABSTRACT

Large-surface or deep wounds often become senescent in the inflammatory or proliferation stages and cannot progress to reepithelialization. This failure makes intervention necessary to provide the final sealing epithelial layer. The best current treatment is autologous skin graft, although there are other choices such as allogenic or autologous skin substitutes and synthetic dressings. Amniotic membrane (AM) is a tissue of interest as a biological dressing due to its biological properties and immunologic characteristics. It has low immunogenicity and beneficial reepithelialization effects, with antiinflammatory, antifibrotic, antimicrobial, and nontumorigenic properties. These properties are related to its capacity to synthesize and release cytokines and growth factors. We report the use of AM as a wound dressing in two patients with large and deep traumatic wounds. Negative pressure wound therapy followed by AM application was capable of restoring skin integrity avoiding the need for skin graft reconstruction. AM induced the formation of a well-structured epidermis. To understand this effect, we designed some assays on human keratinocyte-derived HaCaT cells. AM treatment of HaCaT induced ERK1/2 and SAP/JNK kinases phosphorylation and c-jun expression, a gene critical for keratinocytes migration; however, it did not affect cell cycle distribution. These data suggest that AM substantially modifies the behavior of keratinocytes in chronic wounds, thereby allowing effective reepithelialization.


Subject(s)
Amnion/transplantation , Biological Dressings , Wound Healing/physiology , Wounds, Penetrating/therapy , Aged , Biopsy , Cell Cycle/physiology , Cell Line/physiology , Combined Modality Therapy , Female , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Keratinocytes/physiology , Middle Aged , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Negative-Pressure Wound Therapy , Phosphorylation/physiology , Proto-Oncogene Proteins c-jun/physiology , Treatment Outcome , Wounds, Penetrating/pathology
15.
J Cell Sci ; 117(Pt 18): 4113-25, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15280432

ABSTRACT

Transforming growth factor beta (TGF-beta) signalling leads to phosphorylation and activation of receptor-regulated Smad2 and Smad3, which form complexes with Smad4 and accumulate in the nucleus. The Smads, however, do not seem to reside statically in the cytoplasm in the absence of signalling or in the nucleus upon TGF-beta stimulation, but have been suggested to shuttle continuously between these cellular compartments in both the absence and presence of TGF-beta. Here we investigate this nucleocytoplasmic shuttling in detail in living cells using fusions of Smad2 and Smad4 with enhanced GFP. We first establish that the GFPSmad fusions behave like wild-type Smads in a variety of cellular assays. We go on to demonstrate directly, using photobleaching experiments, that Smad2 and Smad4 shuttle between the cytoplasm and nucleus in both TGF-beta-induced cells and in uninduced cells. In uninduced cells, GFPSmad2 is less mobile in the cytoplasm than is GFPSmad4, suggesting that it may be tethered there. In addition, we show that both GFPSmad2 and GFPSmad4 undergo a substantial decrease in mobility in the nucleus upon TGF-beta stimulation, suggesting that active complexes of Smads are tethered in the nucleus, whereas unactivated Smads are more freely diffusible. We propose that regulated cytoplasmic and nuclear retention may play a role in determining the distribution of Smads between the cytoplasm and the nucleus in both uninduced cells and upon TGF-beta induction.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , DNA-Binding Proteins/drug effects , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins , HeLa Cells , Humans , Macromolecular Substances , Protein Transport/drug effects , Protein Transport/physiology , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Smad2 Protein , Smad4 Protein , Trans-Activators/drug effects , Transforming Growth Factor beta/pharmacology
16.
Biochem J ; 379(Pt 1): 209-16, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14715079

ABSTRACT

Smad4 is an essential signal transducer of the transforming growth factor beta (TGF-beta) signalling pathway and has been identified as a tumour suppressor, being mutated in approx. 50% of pancreatic cancers and approx. 15% of colorectal cancers. Two missense mutations in the C-terminal domain of Smad4, D351H (Asp351-->His) and D537Y (Asp537-->Tyr), have been described recently in the human colorectal cancer cell lines CACO-2 and SW948 respectively [Woodford-Richens, Rowan, Gorman, Halford, Bicknell, Wasan, Roylance, Bodmer and Tomlinson (2001) Proc. Natl. Acad. Sci. U.S.A. 98, 9719-9723]. Previous work in vitro suggested that only Asp-351 was required for interaction with Smad2 [Wu, Fairman, Penry and Shi (2001) J. Biol. Chem. 276, 20688-20694]. In the present study, we investigate the functional consequences of these point mutations in vivo. We demonstrate that neither of these colorectal cancer cells undergo growth arrest in response to TGF-beta, which can be explained, at least in part, by their inability to up-regulate cyclin-dependent kinase inhibitors p21 (CIP1 ) or p15 ( INK4b) after TGF-beta stimulation. Although the point-mutated Smad4s are expressed at normal levels in these colorectal cancer cells, they cannot interact with either TGF-beta-induced phosphorylated Smad2 or Smad3. As a result, these Smad4 mutants do not accumulate in the nucleus after TGF-beta stimulation, are not recruited to DNA by relevant Smad-binding transcription factors and cannot generate transcriptionally active DNA-bound complexes. Therefore both these colorectal tumour cells completely lack functional Smad4 activity owing to the missense mutations. Given the location of these mutations in the three-dimensional structure of the Smad4 C-terminal domain, the results also give us significant insights into Smad complex formation.


Subject(s)
Colorectal Neoplasms/metabolism , DNA-Binding Proteins/physiology , Mutation, Missense , Neoplasm Proteins/physiology , Trans-Activators/physiology , Xenopus Proteins , ADP-Ribosylation Factor 1/metabolism , Active Transport, Cell Nucleus , Amino Acid Substitution , Cell Division/drug effects , Cell Nucleus/metabolism , Colorectal Neoplasms/pathology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dimerization , Forkhead Transcription Factors , Gene Expression Regulation, Neoplastic , Humans , Macromolecular Substances , Models, Molecular , Neoplasm Proteins/chemistry , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Nerve Growth Factors , Point Mutation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Signal Transduction , Smad Proteins , Smad2 Protein , Smad3 Protein , Smad4 Protein , Structure-Activity Relationship , Trans-Activators/chemistry , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/pharmacology
17.
Oncogene ; 22(24): 3698-711, 2003 Jun 12.
Article in English | MEDLINE | ID: mdl-12802277

ABSTRACT

We have investigated the mechanism whereby tumor cells become resistant to the antiproliferative effects of transforming growth factor (TGF)-beta, while maintaining other responses that can lead to increased malignancy and invasiveness. TGF-beta signaling results in nuclear accumulation of active Smad complexes which regulate transcription of target genes. Here we show that in two pancreatic carcinoma cell lines, PT45 and Panc-1, that are resistant to TGF-beta-induced growth arrest, the TGF-beta-Smad signaling pathway is attenuated compared with epithelial cells that are sensitive to the antiproliferative effects of TGF-beta (HaCaT and Colo-357). In PT45 and Panc-1 cells, active Smad complexes remain nuclear for only 1-2 h compared with more than 6 h in HaCaT and Colo-357 cells. The attenuated pathway in PT45 and Panc-1 cells correlates with low levels of TGF-beta type I receptor and results in an altered expression profile of TGF-beta-inducible genes required for cell cycle arrest. Most significantly, expression of the CDK inhibitor, p21(Cip1/WAF1), which is required for TGF-beta-induced growth arrest in these cells, is not maintained. Moreover, we show that artificially attenuating the TGF-beta-Smad signaling pathway in HaCaT cells is sufficient to prevent TGF-beta-induced growth arrest. Our results demonstrate that the duration of TGF-beta-Smad signaling is a critical determinant of the specificity of the TGF-beta response.


Subject(s)
DNA-Binding Proteins/physiology , Pancreatic Neoplasms/pathology , Trans-Activators/physiology , Transforming Growth Factor beta/pharmacology , Activin Receptors, Type I/analysis , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/analysis , Humans , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/analysis , Smad4 Protein , Transcription, Genetic , Tumor Cells, Cultured
18.
J Biol Chem ; 278(5): 3251-6, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12435725

ABSTRACT

In normal epithelial cells, transforming growth factor-beta (TGF-beta) typically causes growth arrest in the G(1) phase of the cell cycle and may eventually lead to apoptosis. However, transformed cells lose these inhibitory responses and often instead show an increase in malignant character following TGF-beta treatment. In the canine kidney-derived epithelial cell line, MDCK, synergism between activation of the Raf/MAPK pathway and the resulting autocrine production of TGF-beta triggers transition from an epithelial to a mesenchymal phenotype. During this process, these cells become refractive to TGF-beta-induced cell cycle arrest and apoptosis. TGF-beta signals are primarily transduced to the nucleus through complexes of receptor-regulated Smads, Smad2 and Smad3 with the common mediator Smad, Smad4. Here we show that the transition from an epithelial to mesenchymal phenotype is accompanied by gradual down-regulation of expression of Smad3. Restoration of Smad3 to previous levels of expression restores the cell cycle arrest induced by TGF-beta without reverting the cells to an epithelial phenotype or impacting on the MAPK pathway. Regulation of apoptosis is not affected by Smad3 levels. These data attribute to Smad3 a critical role in the control of cell proliferation by TGF-beta, which is lost following an epithelial to mesenchymal transition.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Cell Division/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Mesoderm/cytology , Trans-Activators/genetics , Transforming Growth Factor beta/pharmacology , Urothelium/cytology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line , Dogs , Gene Expression Regulation/drug effects , Kidney , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Phenotype , Smad3 Protein
19.
Mol Cell ; 10(2): 283-94, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12191474

ABSTRACT

Transforming growth factor (TGF)-beta stimulation leads to phosphorylation and activation of Smad2 and Smad3, which form complexes with Smad4 that accumulate in the nucleus and regulate transcription of target genes. Here we demonstrate that, following TGF-beta stimulation of epithelial cells, receptors remain active for at least 3-4 hr, and continuous receptor activity is required to maintain active Smads in the nucleus and for TGF-beta-induced transcription. We show that continuous nucleocytoplasmic shuttling of the Smads during active TGF-beta signaling provides the mechanism whereby the intracellular transducers of the signal continuously monitor receptor activity. Our data therefore explain how, at all times, the concentration of active Smads in the nucleus is directly dictated by the levels of activated receptors in the cytoplasm.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Trans-Activators/metabolism , 3T3 Cells , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HeLa Cells , Humans , Mice , Models, Biological , Phosphorylation , Protein Binding , Protein Transport , Signal Transduction/drug effects , Smad2 Protein , Smad3 Protein , Smad4 Protein , Time Factors , Transforming Growth Factor beta/pharmacology
20.
Mol Pharmacol ; 62(1): 65-74, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065756

ABSTRACT

Small molecule inhibitors have proven extremely useful for investigating signal transduction pathways and have the potential for development into therapeutics for inhibiting signal transduction pathways whose activities contribute to human diseases. Transforming growth factor beta (TGF-beta) is a member of a large family of pleiotropic cytokines that are involved in many biological processes, including growth control, differentiation, migration, cell survival, adhesion, and specification of developmental fate, in both normal and diseased states. TGF-beta superfamily members signal through a receptor complex comprising a type II and type I receptor, both serine/threonine kinases. Here, we characterize a small molecule inhibitor (SB-431542) that was identified as an inhibitor of activin receptor-like kinase (ALK)5 (the TGF-beta type I receptor). We demonstrate that it inhibits ALK5 and also the activin type I receptor ALK4 and the nodal type I receptor ALK7, which are very highly related to ALK5 in their kinase domains. It has no effect on the other, more divergent ALK family members that recognize bone morphogenetic proteins (BMPs). Consistent with this, we demonstrate that SB-431542 is a selective inhibitor of endogenous activin and TGF-beta signaling but has no effect on BMP signaling. To demonstrate the specificity of SB-431542, we tested its effect on several other signal transduction pathways whose activities depend on the concerted activation of multiple kinases. SB-431542 has no effect on components of the ERK, JNK, or p38 MAP kinase pathways or on components of the signaling pathways activated in response to serum.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Proteins , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transcription, Genetic/drug effects , 3T3 Cells , Activating Transcription Factor 2 , Activins/pharmacology , Animals , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/pharmacology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Drug Interactions , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinases/metabolism , Osmotic Pressure/drug effects , Phosphorylation/drug effects , Protein Serine-Threonine Kinases , Rats , Receptor, Transforming Growth Factor-beta Type I , Signal Transduction/drug effects , Smad Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...