Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Curr Mol Med ; 13(1): 13-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23176147

ABSTRACT

Podocyte loss plays a key role in the progression of glomerular disorders towards glomerulosclerosis and chronic kidney disease. Podocytes form unique cytoplasmic extensions, foot processes, which attach to the outer surface of the glomerular basement membrane and interdigitate with neighboring podocytes to form the slit diaphragm. Maintaining these sophisticated structural elements requires an intricate actin cytoskeleton. Genetic, mechanic, and immunologic or toxic forms of podocyte injury can cause podocyte loss, which causes glomerular filtration barrier dysfunction, leading to proteinuria. Cell migration and cell division are two processes that require a rearrangement of the actin cytoskeleton; this rearrangement would disrupt the podocyte foot processes, therefore, podocytes have a limited capacity to divide or migrate. Indeed, all cells need to rearrange their actin cytoskeleton to assemble a correct mitotic spindle and to complete mitosis. Podocytes, even when being forced to bypass cell cycle checkpoints to initiate DNA synthesis and chromosome segregation, cannot complete cytokinesis efficiently and thus usually generate aneuploid podocytes. Such aneuploid podocytes rapidly detach and die, a process referred to as mitotic catastrophe. Thus, detached or dead podocytes cannot be adequately replaced by the proliferation of adjacent podocytes. However, even glomerular disorders with severe podocyte injury can undergo regression and remission, suggesting alternative mechanisms to compensate for podocyte loss, such as podocyte hypertrophy or podocyte regeneration from resident renal progenitor cells. Together, mitosis of the terminally differentiated podocyte rather accelerates podocyte loss and therefore glomerulosclerosis. Finding ways to enhance podocyte regeneration from other sources remains a challenge goal to improve the treatment of chronic kidney disease in the future.


Subject(s)
Cytoskeleton/metabolism , Mitosis , Podocytes/pathology , Podocytes/physiology , Actins/genetics , Actins/metabolism , Animals , Cell Cycle , Cell Differentiation , Humans , Kidney Diseases/pathology , Kidney Glomerulus/cytology , Podocytes/cytology , Stem Cells/metabolism
2.
Curr Med Chem ; 17(34): 4134-49, 2010.
Article in English | MEDLINE | ID: mdl-20939819

ABSTRACT

Many diseases and/or physical defects due to injury result in the loss of specialized cells within organ systems and lead to organ system dysfunction. The ultimate goal of cell-based therapies is to regenerate and restore normal function. Populations of embryonic, fetal, adult stem cells and inducible pluripotent stem cells generated by reprogramming of adult cells show promise for the treatment of a variety of diseases. In addition, the recent advancements in adult stem cell biology in both normal and pathological conditions have led to the identification of some intrinsic and extrinsic factors that govern the decision between self renewal versus differentiation of tissue-resident adult stem cells. This is of primary importance for the design of an approach of stem cell-based therapy focused on their in vivo modulation by conventional chemical and biological therapeutics capable to stimulate endogenous cell regeneration. Such therapeutics can act in vivo to promote cell survival, proliferation, differentiation, reprogramming and homing of stem cells or can modulate their niches. In this review, we will highlight the burst of recent literature on novel perspectives of regenerative medicine and their possible clinical applications.


Subject(s)
Organic Chemicals/pharmacology , Regeneration/drug effects , Regenerative Medicine , Stem Cells , Adult , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Mice , Rats , Regenerative Medicine/methods , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Stem Cell Research , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/physiology
3.
Nefrología (Madr.) ; 30(4): 385-393, jul.-ago. 2010. ilus, tab
Article in Spanish | IBECS | ID: ibc-104579

ABSTRACT

Cuando se produce un daño en un tejido adulto, el proceso de renovación celular continuada es crítico y crucial para la reparación del mismo y, en determinados órganos, se facilita por la presencia de células madre o progenitoras. El riñón, a diferencia de otros órganos como el hígado, es de regeneración lenta. Incluso ha sido considerado durante años como incapaz de regenerarse. Sin embargo, varios estudios han demostrado que existen posibles nichos de células madre renales en la papila renal, progenitores tubulares o progenitores renales CD24+CD133+ localizados en el polo urinario de la cápsula de Bowman. Estas células podrían participar teóricamente en la reparación de la lesión renal. Sin embargo, todavía no se ha demostrado de forma precisa cuál sería su papel ni cómo actuarían después del daño. Aún así, estas células madre renales podrían ser dianas terapéuticas para el remodelado del tejido renal dañado. Por otro lado, se ha postulado que las células madre derivadas de la médula ósea podrían participar en la regeneración renal, especialmente las de estirpe mesenquimal. Sin embargo, tampoco se conoce con exactitud el modo en que actuarían. Hay estudios que sugieren la existencia de fusión celular entre estas células y células residentes, otros apuntan a su diferenciación en células renales, mientras que otros sugieren una acción paracrina responsable del efecto reparador a través de la secreción de factores de crecimiento como HGF, VEGF y IGF-1. Todas estas moléculas secretadas proporcionarían un entorno regenerativo que limitaría el área del daño y que facilitaría la migración de las células madre (AU)


Cell replenishment is critical for adult tissue repair after damage. In some organs this process is facilitated by stem cells. In contrast to the liver, the kidney has limited capacity for regeneration. Nevertheless, there are several recent studies suggesting the presence of stem cells in the adult kidney. Stem cell renal niches have been identified in the renal papilla in animals as well as in the urinary pole of the Bowman capsule in humans (CD24 + CD133 + stem cells).Although these cells may contribute to organ regeneration, how these cells exert this effect and their role after kidney damage is not known. Nevertheless, renal stem cells maybe therapeutic targets for treatment of renal diseases. On the other hand, bone marrow derived stem cells may also contribute in renal repair, particularly mesenchymal stem cells. However, the mechanism for producing such effect has not been elucidated. Some studies suggest there is cell fusion between bone marrow and resident tubular cells; others suggest bone marrow cells are able to differentiate in resident cells, while some authors propose bone marrow cells facilitate organ regeneration by a paracrine action; that is by secreting growth factors as hepatocyte growth factor, vascular endothelial growth factor and insulin growth factor 1. All these secreted molecules would provide a regenerative milieu able to constrain renal damage and to amplify migration of stem cells to the damaged organ (AU)


Subject(s)
Humans , Guided Tissue Regeneration/methods , Acute Kidney Injury/therapy , Intercellular Signaling Peptides and Proteins/therapeutic use , Adult Stem Cells , Hepatocyte Growth Factor/therapeutic use
4.
Nefrologia ; 30(4): 385-93, 2010.
Article in Spanish | MEDLINE | ID: mdl-20651879

ABSTRACT

Cell replenishment is critical for adult tissue repair after damage. In some organs this process is facilitated by stem cells. In contrast to the liver, the kidney has limited regeneration capacity and has even been considered over several years as not being able to regenerate itself. Nevertheless, there are several recent studies suggesting the presence of stem cells in the adult kidney. Stem cell renal niches have been identified in the renal papillae in animals as well as in the urinary pole of the Bowman's capsule in humans (CD24+CD133+ stem cells). Although these cells may contribute to organ regeneration, how these cells exert this effect and their role after kidney injury is not known. Nevertheless, renal stem cells may be therapeutic targets for treatment of renal diseases. On the other hand, bone-marrow-derived stem cells may also contribute to renal repair, particularly mesenchymal stem cells. However, the mechanism for producing such effect has not been elucidated. Some studies suggest there is cell fusion between bone marrow and resident tubular cells; others suggest that bone marrow cells are able to differentiate in resident cells, while some authors propose bone marrow cells facilitate organ regeneration by a paracrine action; that is by secreting growth factors such as HGF, VEGF and IGF1. All these secreted molecules would provide a regenerative milieu able to constrain renal damage and to amplify stem cells migration to the damaged organ.


Subject(s)
Kidney/physiology , Regeneration , Animals , Bone Marrow Cells , Humans , Intercellular Signaling Peptides and Proteins/physiology , Stem Cells/physiology
5.
G Ital Nefrol ; 26(6): 660-9, 2009.
Article in Italian | MEDLINE | ID: mdl-19918748

ABSTRACT

Converging evidence suggests that damage to podocytes plays a key role in progression towards glomerulosclerosis, in particular as the primary cause of all forms of focal segmental glomerulosclerosis (FSGS), the most common glomerular disease leading to end-stage renal disease. Any damage occurring to the complex architecture of specialized proteins that constitute the podocyte foot processes, essential to the highly specialized functions of podocytes, leads inevitably to loss of function in the glomerular filtration barrier, and ultimately to proteinuria. Recent studies have also highlighted that a reduction of the podocyte number in a damaged glomerulus is a critical factor for the development of proteinuria and glomerulosclerosis. As long as the podocyte loss is limited, restitution or repair is possible, which shows that the glomerular architecture can be remodeled. However, mature podocytes have limited capacity to divide and display all the phenotypic and functional features of highly specialized, terminally differentiated cells. A potential mechanism for podocyte replacement might be stem-cell-based regeneration, since it has been established that the developmental source of podocytes are resident renal progenitors. Podocyte damage could then be potentially repaired by a stem cell population resident in the kidney.


Subject(s)
Glomerulosclerosis, Focal Segmental/etiology , Podocytes/pathology , Humans , Podocytes/physiology , Regeneration
6.
Exp Cell Res ; 315(2): 264-73, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19014933

ABSTRACT

Current immunosuppressive protocols have reduced rejection occurrence in heart transplantation; nevertheless, management of heart transplant recipients is accompanied by major adverse effects, due to drug doses close to toxic range. In allograft rejection, characterized by T-helper 1 (Th1) cell-mediated response, the CXCL10-CXCR3 axis plays a pivotal role in triggering a self-promoting inflammatory loop. Indeed, CXCL10 intragraft production, required for initiation and development of graft failure, supports organ infiltration by Th1 cells. Thus, targeting the CXCL10-CXCR3 axis while avoiding generalized immunosuppression, may be of therapeutic significance. Based on preclinical evidence for immunoregulatory properties of vitamin D receptor agonists, we propose that a less hypercalcemic vitamin D analogue, BXL-01-0029, might have the potential to contribute to rejection management. We investigated the effect of BXL-01-0029 on CXCL10 secretion induced by proinflammatory stimuli, both in human isolated cardiomyocytes (Hfcm) and purified CD4+ T cells. Mycophenolic acid (MPA), the active agent of mycophenolate mofetil, was used for comparison. BXL-01-0029 inhibited IFNgamma and TNFalpha-induced CXCL10 secretion by Hfcm more potently than MPA, impairing cytokine synergy and pathways. BXL-01-0029 reduced also CXCL10 protein secretion and gene expression by CD4+ T cells. Furthermore, BXL-01-0029 did not exert any toxic effect onto both cell types, suggesting its possible use as a dose-reducing agent for conventional immunosuppressive drugs in clinical transplantation.


Subject(s)
Cholecalciferol/pharmacology , Immunosuppressive Agents/pharmacology , Myocytes, Cardiac/drug effects , T-Lymphocytes/drug effects , Active Transport, Cell Nucleus/drug effects , Blotting, Western , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Cholecalciferol/analogs & derivatives , Gene Expression/drug effects , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Ionomycin/pharmacology , Microscopy, Fluorescence , Mycophenolic Acid/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Receptors, Calcitriol/agonists , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Interferon gamma Receptor
7.
J Endocrinol Invest ; 31(9): 809-19, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18997494

ABSTRACT

Chemokines exert their multifunctional role in several physiologic and pathologic processes through interaction with their specific receptors. Much evidence have revealed that metastatic spread tumor cells may use chemokine-mediated mechanisms. In particular, an involvement of stromal cell-derived factor-1 (SDF-1) in growth of primary tumors and in metastatic process has been demonstrated. Indeed, it has been suggested that CXCR4 expression by tumor cells, plays a critical role in cell metastasis by a chemotactic gradient to organs expressing the ligand SDF-1. Moreover, CXCR4 overexpression correlated with poor prognosis in many types of cancer. In physiologic condition, SDF-1 also plays an essential role modulating stem cell proliferation, survival, and homing through its canonical receptor CXCR4. Recently, several studies have demonstrated the existence of a small subset of cancer cells which share many characteristics with stem cells and named cancer stem cells (CSC). They constitute a reservoir of self-sustaining cells with the ability to maintain the tumor growth. In particular, most of them express CXCR4 receptor and respond to a chemotactic gradient of its specific ligand SDF-1, suggesting that CSC probably represent a subpopulation capable of initiating metastasis. This review focuses on the role of SDF-1/CXCR4 axis in cancer and in the metastatic progression by tumoral cells, as well as the role of CSC in tumor pathogenesis and in metastatic process. A better understanding of migratory mechanism involving cancer cells and CSC provides a powerful tool for developing novel therapies reducing both local and distant recurrences.


Subject(s)
Chemokine CXCL12/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Humans , Neoplasm Metastasis
8.
G Ital Nefrol ; 25(5): 537-46, 2008.
Article in Italian | MEDLINE | ID: mdl-18828116

ABSTRACT

Recent evidence suggests that injury to the renal vasculature may play an important role in the pathogenesis of both chronic and acute ischemic kidney injury. Early alterations in peritubular capillary blood flow during reperfusion have been documented and associated with loss of normal endothelial cell function. In addition, ischemia induces alterations in endothelial cells that may promote inflammation and procoagulant activity, thus contributing to vascular congestion. Reduction of the microvasculature density increases hypoxia-mediated fibrosis and alters proper hemodynamics, which may lead to hypertension. This may play a critical role in the progression of chronic kidney disease following initial recovery from ischemia/reperfusion-induced acute kidney injury. The turnover and replacement of endothelial cells is therefore an important mechanism in the maintenance of vascular integrity also in the kidney. It is becoming clear that impaired vascular repair mechanisms as a result of a reduced number and/or impaired function of endothelial progenitor cells may contribute to renal disease. Moreover, investigators have begun to identify potential mechanisms responsible for the loss of function of endothelial progenitors in renal disease. In allografts, persistent injury results in excessive turnover of graft vascular endothelial cells. Moreover, chronic damage elicits a response that is associated with the recruitment of both leukocytes and endothelial progenitors, facilitating an overlapping process of inflammation and angiogenesis. In conclusion, angiogenesis and endothelial cell turnover play a pivotal role in renal disease and allograft rejection. Manipulation of these processes might have important implications for the development of novel therapeutic strategies in the near future.


Subject(s)
Endothelial Cells/physiology , Kidney Diseases/etiology , Stem Cells/physiology , Chronic Disease , Disease Progression , Humans , Ischemia/etiology , Kidney/blood supply , Kidney Diseases/surgery , Kidney Transplantation
10.
G Ital Nefrol ; 24(3): 212-20, 2007.
Article in Italian | MEDLINE | ID: mdl-17554733

ABSTRACT

Chemokines are a family of small, structurally related cytokines that regulate trafficking of different subsets of leukocytes, thus critically regulating inflammation. The chemokine system influences allograft biology at 3 main levels: 1) the process of ischemia-reperfusion injury, 2) the induction of transplant tolerance, and 3) the pathogenesis of acute rejection and chronic allograft nephropathy. Accordingly, following ischemia/reperfusion in a rat model, CXCR2 produced at the graft level attracts and activates granulocytes, which in turn promotes graft damage. Moreover, in some experimental models CCR4 recruits T regulatory cells and mediates transplant tolerance. Furthermore, the discovery of the involvement of CXCR3 in the induction of the alloresponse to transplant suggests that this chemokine receptor might represent an important target for treatment of both acute rejection and chronic allograft nephropathy. Indeed, CXCR3 ligands play a pivotal role in the initiation and amplification of host alloresponses and also alter vascular cell functions, which explains their critical role not only in the development of acute rejection, but also in the pathogenesis of chronic allograft nephropathy, where both immune- and nonimmune- mediated mechanisms are involved. Finally, we have recently demonstrated that the pretransplant serum level of the CXCR3 ligand IP-10/CXCL10 is a clinically useful parameter for the identification of subjects with a high risk of acute rejection, chronic allograft nephropathy, and graft failure. This simple test could contribute to the prevention of acute rejections and the individualization of immunosuppressive therapies.


Subject(s)
Chemokines/drug effects , Chemokines/physiology , Graft Rejection/prevention & control , Kidney Transplantation , Postoperative Complications/prevention & control , Renal Insufficiency, Chronic/prevention & control , Graft Rejection/etiology , Humans , Postoperative Complications/etiology , Renal Insufficiency, Chronic/etiology
11.
Curr Med Chem ; 14(10): 1129-39, 2007.
Article in English | MEDLINE | ID: mdl-17456026

ABSTRACT

The discovery of stem cells (SC) has shed new light on the understanding of mechanisms responsible for ischemic and degenerative disorders, and opened a new field for regenerative medicine. Furthermore, dysregulation of SC self-renewal and their transformation seem to be involved also in the development of cancer, suggesting that pharmacological treatment devoted to regulate SC genomic and phenotypic functions might represent a potential new strategy even for the treatment of neoplastic disorders. SC display a promiscuous set of transcription factors and an open chromatin structure which are required to maintain their multipotentiality, while they are progressively quenched during differentiation into specific multiple lineages. The mechanisms that govern stem cell fate decisions are under tight control but remain potentially alterable. Recent studies have shown that several currently used drugs such as colony stimulating factors, statins, angiotensin-II receptor antagonists/ACE-inhibitors, Erythropoietin, nitric oxide donors, estrogens and glitazones, have modulatory activity on SC functions. These drugs mostly enhance SC survival and mobilization. Furthermore, a series of new pharmacological agents such as the chemokine receptor antagonist AMD3100, glycogen synthase kinase-3 (GSK-3) inhibitors and histone deacetylase inhibitors (HDACi), that modulate the growth, differentiation and mobilization of SC, have been recently discovered and are currently under evaluation in both in vivo experimental models and preliminary clinical trials. Thus, modulation of SC properties through pharmacological treatment represents a new field of investigation which may lead to the development of novel strategies for the treatment not only of ischemic and degenerative disorders, but also of cancer.


Subject(s)
Stem Cells/drug effects , Animals , Cell Count , Cell Differentiation/physiology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hematopoietic Stem Cell Mobilization , Humans , Receptors, CXCR4/drug effects
12.
Gut ; 55(7): 961-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16368781

ABSTRACT

BACKGROUND: We previously demonstrated the existence of two distinct subsets of T cell receptor (TCR)alphabeta+CD8alphabeta+ single positive (SP) cells in human postnatal thymus which express the chemokine receptor CCR7 or CXCR3 and migrate in vitro in response to their specific ligands. AIM: To investigate whether these two CD8+ thymocyte subsets had distinct peripheral colonisation. METHODS: TCRalphabeta+CD8+ SP cells were obtained from normal postnatal thymus, mesenteric lymph node (LNs), small bowel, and peripheral blood (PB) specimens. Cells were then evaluated for expression of surface molecules, cytolytic potential, telomere length, and profile of cytokine production. RESULTS: CD8+CCR7+CXCR3- thymocytes exhibited CD62L, in common with those which localise to LNs. In contrast, CD8+CCR7-CXCR3+ thymocytes lacked CD62L but exhibited CD103, similar to intraepithelial lymphocytes (IELs) present in the gut mucosa where the CXCR3 ligand, CXCL10, and the CD103 ligand, E-cadherin, are highly and consistently expressed. In addition, thymocytes and gut CD8+CXCR3+CD103+ cells showed comparable telomere length, which was higher than that of PB CXCR3+CD8+ T cells. However, both of these populations contained perforin and granzyme A, and displayed the ability to produce interferon gamma and interleukin 2. Of note, CXCR3 deficient, in comparison with wild-type C57Black/6, mice showed decreased proportions of CD3+CD8alphabeta+ and increased proportions of CD3+CD8alphaalpha+ lymphocytes at gut level. Moreover, adoptive transfer of CD3+CD8alphabeta+ thymocytes from wild-type into CXCR3 deficient mice resulted in a significant increase in CD3+CD8alphabeta+ T cells in the gut mucosa but not in other tissues. CONCLUSIONS: The results of this study demonstrate the existence of a previously unrecognised subset of TCRalphabeta+CD8alphabeta+ SP CXCR3+CD103+ thymocytes which share phenotypic and functional features with CD8+ IELs, thus suggesting the possibility of their direct colonisation of the gut mucosa.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Integrins/analysis , Intestinal Mucosa/immunology , Receptors, Chemokine/analysis , Adoptive Transfer , Adult , Analysis of Variance , Animals , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/ultrastructure , Cell Separation/methods , Child, Preschool , Flow Cytometry , Humans , Immunohistochemistry/methods , Infant , Infant, Newborn , Interleukins/biosynthesis , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, CCR7 , Receptors, CXCR3 , Receptors, Chemokine/genetics , Telomere/ultrastructure
13.
Article in English | MEDLINE | ID: mdl-15777209

ABSTRACT

The goal to attenuate inflammation without inducing generalized immunosuppression has focused the attention on chemokines, a family of chemotactic peptides that regulate the leukocyte traffick into tissues. However, the development of drugs that block ckemokine activity may be hampered by the observation that some chemokines display pleiotropic biologic functions. For example, the chemokines CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC exhibit the ability to recruit different leukocytes subsets, the capacity to induce the proliferation of vascular pericytes as well as powerful anti-tumor effects, which are mediated by a common receptor, named CXCR3. Because of their pleiotropic biologic effects, these chemokines have been proposed as possible therapeutic targets in cancer, allograft rejection, glomerulonephritis, diabetes, multiple sclerosis, and autoimmune disorders of the thyroid. The chemokine CXCL4/PF4 shares several activities with CXCL9, CXCL10, and CXCL11, including angiostatic effects, although its specific receptor has remained unknown for a long time. Recently, we provided evidence that the different functions of CXCL9, CXCL10, and CXCL11 on distinct cell types can be at least partly explained by the interaction of these chemokines with two distinct receptors. Indeed, in addition to the classic form of CXCR3 receptor, which we have renamed as CXCR3-A, a novel CXCR3 receptor variant (CXCR3-B) was identified, that not only mediates the angiostatic activity of CXCR3 ligands, but also acts as functional receptor for CXCL4. In this review, we focus on the accumulating evidence demonstrating the pivotal role of CXCR3-binding chemokines in several human diseases. Studies based on CXCR3 targeting have shown its importance in different pathologic conditions and orally active small molecules capable of inhibiting this receptor are now being developed in order to be tested for their activity in humans.


Subject(s)
Drug Delivery Systems/methods , Receptors, Chemokine/metabolism , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Humans , Protein Binding/physiology , Receptors, CXCR3 , Receptors, Chemokine/immunology , Transplantation, Homologous/immunology , Transplantation, Homologous/methods
15.
J Endocrinol Invest ; 26(2): 177-80, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12739748

ABSTRACT

The chemoattractant cytokines (chemokines) have been classified into 4 major sub-families in relation to the position of the cysteine residues in their NH2 terminal portion. Interferon-gamma inducible chemokines (CXCL9/Mig, CXCL10/IP-10, CXCL11/I-TAC), strongly associated to Th1-mediated immune responses, belong to the CXC sub-family. They represent an exception among chemokines in that they specifically interact with a single type of receptor, named CXCR3. A statistically significant increase of CXCL10/IP-10 and CXCL9/Mig expression, in thyroid tissue specimens obtained from subjects affected by Hashimoto's thyroiditis and recent onset Graves' disease has been reported. Furthermore, a statistically significant increase in serum CXCL10/IP-10 levels has been found in newly diagnosed Graves' patients when compared to healthy subjects as well as patients with long standing disease and a strong statistically significant inverse correlation between circulating CXCL10/IP-10 levels and disease duration has been demonstrated. Similar findings have been obtained when Type 1 autoimmune diabetes affected patients have been taken into account. In conclusion, such experiences have demonstrated an important role played by interferon-gamma inducible CXC chemokines in the pathogenesis of glandular autoimmunity. In fact, it is reasonable to assume that glandular epithelial cells may modulate the autoimmune process at least in its initial phase, through the production of chemokines which induce migration of Th1 lymphocytes into the gland. Interferon-gamma secretion by lymphocytes would, in turn, stimulate chemokines production by follicular cells, thus perpetuating the autoimmune cascade.


Subject(s)
Autoimmunity/immunology , Chemokines, CXC/immunology , Graves Disease/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/immunology , Thyroiditis, Autoimmune/immunology , Chemokine CXCL10 , Chemokine CXCL9 , Humans , Receptors, CXCR3 , Receptors, Chemokine/immunology
16.
G Ital Nefrol ; 19(6): 641-9, 2002.
Article in Italian | MEDLINE | ID: mdl-12508169

ABSTRACT

Cytokines are soluble factors that are critical for the pathophysiology of the immune system and exhibit other important functions. Cytokines produced by type 1 helper T (Th1) lymphocytes, such as interferon (IFN)-g, play a pathogenic role in proliferative glomerulonephrites (GN), as well as in the acute rejection of kidney allografts. Cytokines produced by type 2 Th (Th2) lymphocytes, such as interleukin (IL)-4, IL-5, and IL-13), predominate in membranous GN and in minimal change disease. More recently, the pathogenic role of some members of the family of chemotactic cytokines (chemokines) in different nephropathies and in the acute and chronic rejection of kidney allografts has also been demonstrated. In particular, the chemokine MCP1/CCL2 has been found to be expressed in the kidneys of subjects with tubulo-interstitial nephritis and seems to play an important role in the sclerotic evolution of both inflammatory and metabolic nephropathies. Interactions between IP-10/CXCL10, Mig/CXCL9 and I-TAC/CXCL11 and their shared receptor, CXCR3, seem to be responsible not only for Th1 cell infiltration in acute allograft rejection and in proliferative GN, but also for mesangial cell proliferation typical of the latter condition. In proliferative GN, mesangial cells indeed express both these chemokines and their receptor. Moreover, in the kidneys of subjects suffering from chronic allograft nephropathy, IP-10/CXCL10, Mig/CXCL9 and I-TAC/CXCL11 have been found to be produced by and to act on the proxymal tubular epithelial cells, endothelial cells and smooth muscle vessel cells, suggesting their possible role in both the genesis of tubular atrophy and allograft artheriosclerosis.


Subject(s)
Chemokines/immunology , Cytokines/immunology , Kidney Diseases/immunology , Kidney Transplantation/immunology , Graft Rejection/immunology , Humans , Kidney Diseases/physiopathology , T-Lymphocytes/immunology
19.
Eur J Immunol ; 31(8): 2431-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11500827

ABSTRACT

The nature and the functional activity of immunocytes present in the cumulus oophorus, a mass of cells surrounding the oocyte, were examined here for the first time. The cumuli oophorus were obtained from women who had taken part in an in vitro fertilization program and were suffering from blocked fallopian tubes. Both macrophages and CD4(+) T cells were detected in all cumuli. CD4(+) T cell clones, generated from T cells of these cumuli, showed higher potential to produce IL-4 and leukemia inhibitory factor (LIF) than CD4(+) T cell clones generated from peripheral blood or ovary specimens from the same women. More importantly, IL-4 and LIF, but not IFN-gamma mRNA was found to be constitutively expressed in vivo by cumulus oophorus cells. Progesterone is highly produced by the cumulus oophorus/oocyte complex. We recently showed that progesterone up-regulates the production of LIF by T cells and that the progesterone-induced LIF production is mediated by IL-4. Progesterone produced by cumulus granulosa cells may favor IL-4 production by T cells, which in turn can produce LIF. As the treatment with LIF enhances the in vitro growth and development of mammalian embryos, our data suggest that T cells present in the cumulus oophorus produce cytokines that may provide a microenvironment suitable for pre-implantation development of the mammalian embryo.


Subject(s)
Blastocyst/physiology , CD4-Positive T-Lymphocytes/metabolism , Interleukin-6 , Oocytes/cytology , Oocytes/immunology , Adult , Blastocyst/cytology , Blastocyst/immunology , Cells, Cultured , Female , Gene Expression , Growth Inhibitors/biosynthesis , Growth Inhibitors/genetics , Humans , Immunohistochemistry , Interferon-gamma/genetics , Interleukin-4/biosynthesis , Interleukin-4/genetics , Leukemia Inhibitory Factor , Lymphokines/biosynthesis , Lymphokines/genetics , Macrophages/immunology , Ovary/cytology , Ovary/immunology , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Int Arch Allergy Immunol ; 124(1-3): 146-50, 2001.
Article in English | MEDLINE | ID: mdl-11306952

ABSTRACT

BACKGROUND: The aim of this study was to investigate whether human mast cells express functional active CCR3 receptors, which are activated by CC chemokines. These ligands include the CCR3-selective chemokines eotaxin and eotaxin-2 and the more promiscuous CC chemokines, MCP-4, MCP-3, MCP-2 and RANTES. METHODS: Immunohistochemical analysis was performed on skin, gut and lung specimens. Double immunostaining was performed with anti-CCR3 and antitryptase, and anti-CCR3 and antichymase antibody (Ab) by using the avidin-biotin-peroxidase system with two different substrates. Mast cells were isolated and purified from human lung parenchyma (HLMC) by countercurrent elutriation followed by discontinuous Percoll density gradient. Flow-cytometric analysis of HLMC surface CCR3 expression was performed with the monoclonal Ab anti-CCR3 (7B11). Functional activation of HLMC was verified by the ability of cells to release histamine and/or migrate in response to eotaxin. RESULTS: High percentages (>70%) of tryptase-positive cells showing CCR3 expression were found in the skin and in the intestinal submucosa, whereas much lower percentages (< or = 20%) were found in the intestinal mucosa and in the lung interstitium. Eotaxin (1-100 nM) neither induced histamine release from HLMC nor enhanced anti-IgE-induced histamine release. In contrast, eotaxin (10-100 nM) and RANTES (10-100 nM) induced HLMC chemotaxis in vitro. Preincubation of HLMC with antibody anti-CCR3 (5 microg/ml) before loading into the chemotaxis chamber abrogated chemotaxis elicited by eotaxin. Double immunostaining with anti-CCR3 and anti-chymase antibody showed that the vast majority of CCR3-expressing mast cells in the various human tissues examined were tryptase-chymase double-positive. CONCLUSIONS: These results indicate that CCR3 is expressed on human mast cells and that these cells are attracted by CCR3-binding chemokines.


Subject(s)
Chemokines, CC , Mast Cells/metabolism , Receptors, Chemokine/metabolism , Cells, Cultured , Chemokine CCL11 , Chemokine CCL5/pharmacology , Chemotaxis , Cytokines/pharmacology , Humans , Intestinal Mucosa/metabolism , Lung/cytology , Lung/metabolism , Mast Cells/drug effects , Mast Cells/physiology , Receptors, CCR3 , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...