Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cells Tissues Organs ; 174(1-2): 26-33, 2003.
Article in English | MEDLINE | ID: mdl-12784039

ABSTRACT

The host response to infection or injury initiates a cascade of events involving recruitment of leukocytes and the release of multiple inflammatory mediators. One of these mediators, nitric oxide (NO), not only represents an important microbicidal agent in host defense, but also functions as a biological signaling and effector molecule in inflammation and immunity. However, overproduction of NO can be autotoxic and contribute to tissue damage and has been implicated in pathogenesis of tumors, and infectious, autoimmune and chronic degenerative diseases. NO is generated via constitutive and inducible nitric oxide synthases (iNOS) which catalyze the oxidation of a guanidino nitrogen associated with L-arginine. Whereas endothelial NOS (eNOS) and neuronal NOS (nNOS) are constitutively expressed, iNOS is transcriptionally induced by bacterial constituents and inflammatory mediators, including TNF alpha and IL-1. In an experimental model of bacterial component-induced joint inflammation and tissue degradation, functionally distinct roles of the constitutive NOS and iNOS were demonstrated. Following systemic delivery of an arthritogenic dose of streptococcal cell walls (SCW), these bacterial peptidoglycan-polysaccharide complexes disseminate and target the peripheral joints, liver and spleen of the treated animals. Following deposition of the SCW in the peripheral joints, an initial innate inflammatory response to the bacterial components progresses into an adaptive immune response with the recruitment and activation of mononuclear phagocytes and T lymphocytes. With the release of cytokines and inflammatory mediators, there is an upregulation of gene expression for iNOS, but not the constitutive nNOS or eNOS. Nonetheless, the constitutive NOS isoforms, regulated by calcium fluxes and interaction with calmodulin, may also enhance NO production. Increased release of NO was detected not only in the synovium, but also in the circulation, and plasma levels of nitrate plus nitrite, the stable products of NO reactions, correlated with disease progression. Following inhibition of NO production with nonspecific NOS inhibitors, such as N(G)-monomethyl-L-arginine, which target all three isoforms, there is a striking therapeutic benefit with reduced signs and symptoms of erosive arthritis. In contrast, selective targeting of iNOS with N-iminoethyl-L-lysine resulted in exacerbation of the synovial inflammation and degradation of joint structures. Based on these data, it appears that the constitutive isoforms of NOS contribute to the pathophysiology of the arthropathy, and that induced NOS and NO may function, in part, in a protective pathway. Moreover, the suppression of NO following treatment with TNF alpha antagonists results in reduced inflammation and the associated synovial pathology. Collectively, these data implicate discrete roles for the NOS isoforms in the emergence of local tissue pathology and underscore the need to define the specific pathways that are being targeted for interventional strategies.


Subject(s)
Nitric Oxide/metabolism , Temporomandibular Joint Disorders/immunology , Temporomandibular Joint Disorders/metabolism , Animals , Humans , Nitric Oxide Synthase/metabolism , Temporomandibular Joint Disorders/pathology , Tumor Necrosis Factor-alpha/immunology
2.
J Immunol ; 166(4): 2734-40, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160339

ABSTRACT

NO is an essential cytotoxic agent in host defense, yet can be autotoxic if overproduced, as evidenced in inflammatory lesions and tissue destruction in experimental arthritis models. Treatment of streptococcal cell wal1-induced arthritis in rats with N:(G)-monomethyl-L-arginine (L-NMMA), a competitive nonspecific inhibitor of both constitutive and inducible isoforms of NO synthase (NOS), prevents intraarticular accumulation of leukocytes, joint swelling, and bone erosion. Because increased inducible NOS (iNOS) expression and NO generation are associated with pathogenesis of chronic inflammation, we investigated whether a selective inhibitor of iNOS, N:-iminoethyl-L-lysine (L-NIL), would have more directed anti-arthritic properties. Whereas both L-NMMA and L-NIL inhibited nitrite production by streptococcal cell wall-stimulated rat mononuclear cells in vitro and systemic treatment of arthritic rats with L-NMMA ablated synovitis, surprisingly L-NIL did not mediate resolution of inflammatory joint lesions. On the contrary, daily administration of L-NIL failed to reduce the acute response and exacerbated the chronic inflammatory response, as reflected by profound tissue destruction and loss of bone and cartilage. Although the number of iNOS-positive cells within the synovium decreased after treatment with L-NIL, immunohistochemical analyses revealed a distinct pattern of endothelial and neuronal NOS expression in the arthritic synovium that was unaffected by the isoform-specific L-NIL treatment. These studies uncover a contribution of the constitutive isoforms of NOS to the evolution of acute and chronic inflammation pathology which may be important in the design of therapeutic agents.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Administration, Oral , Animals , Arthritis, Experimental/prevention & control , Enzyme Activation/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Hindlimb , Injections, Intraperitoneal , Isoenzymes/antagonists & inhibitors , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/metabolism , Leukocytosis/enzymology , Leukocytosis/pathology , Leukocytosis/prevention & control , Lysine/administration & dosage , Lysine/analogs & derivatives , Lysine/pharmacology , Neutrophils/enzymology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Polysaccharides, Bacterial/administration & dosage , Rats , Rats, Inbred Lew , omega-N-Methylarginine/administration & dosage , omega-N-Methylarginine/pharmacology
3.
Arthritis Rheum ; 42(6): 1119-27, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10366104

ABSTRACT

OBJECTIVE: To investigate the ability of hemoglobin (Hgb), a nitric oxide (NO) scavenger, to deplete excess NO and reduce inflammation and injury in synovial tissue from joints with inflammatory arthritis. METHODS: The severity of streptococcal cell wall-induced arthritis was monitored following administration of Hgb. Plasma nitrite and nitrate levels were measured, and inducible NO synthase (iNOS) and cytokine messenger RNA (mRNA) expression in peripheral blood mononuclear cells (PBMC) and joint tissue were evaluated. RESULTS: Following systemic administration of Hgb to arthritic rats, plasma levels of nitrite and nitrate as well as iNOS mRNA expression in the joints and PBMC were significantly reduced. Moreover, inflammatory cell accumulation and disease pathology in the joint tissue were dramatically attenuated without obvious side effects. Consistent with this reduction in the inflammatory response, cytokine gene expression was decreased in the synovium of Hgb-treated rats. CONCLUSION: Modulation of NO levels through the use of a NO scavenger, Hgb, influences the development and severity of arthritis. These findings suggest that depletion of excess NO by NO scavengers provides a prototype for further exploration of potential treatments for chronic arthritis.


Subject(s)
Arthritis, Infectious/prevention & control , Hemoglobins/pharmacology , Streptococcal Infections/prevention & control , Animals , Arthritis, Infectious/blood , Arthritis, Infectious/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cytokines/biosynthesis , Cytokines/genetics , Female , Germ-Free Life , Immunohistochemistry , Joints/drug effects , Joints/metabolism , Joints/pathology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Monocytes/drug effects , Monocytes/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nitrates/blood , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitrites/blood , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Streptococcal Infections/blood , Streptococcal Infections/pathology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology
4.
Int Rev Immunol ; 16(5-6): 553-80, 1998.
Article in English | MEDLINE | ID: mdl-9646177

ABSTRACT

Regulation of developmental processes as well as host defense and repair mechanisms requires the maintenance of a delicate balance of positive and negative regulatory signals. TGF-beta, a molecule known for its many diverse activities, can promote or inhibit cell growth and function. Disruption of the balance between these opposing activities can contribute to aberrant development, malignancy, or pathogenic immune and inflammatory responses. TGF-beta transgenic mouse studies highlight the essential function(s) of TGF-beta and its receptors and provide insight to potential therapeutic approaches to manipulate TGF-beta expression.


Subject(s)
Transforming Growth Factor beta/physiology , Adjuvants, Immunologic , Animals , Antineoplastic Agents , Growth Inhibitors , Humans , Immunosuppression Therapy , Mice , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
5.
Am J Pathol ; 151(5): 1281-8, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9358754

ABSTRACT

Mice homozygous for a nonfunctional transforming growth factor-beta 1 gene develop rampant inflammation in vital organs that contributes to a shortened life span. The presence of circulating anti-nuclear anti-bodies, immune deposits in tissues, leukocyte infiltration, and increased major histocompatibility complex antigen expression resembles an autoimmune-like syndrome. One of the overt symptoms that appears in these mice lacking transforming growth factor-beta 1 is the development of dry crusty eyes that close persistently as their health declines. Histologically, the eyes appear normal with little or no inflammation. However, inflammatory lesions, predominantly lymphocytic, develop in the lacrimal glands, disrupting their structure and function and severely limiting their ability to generate tears. This histopathology and aberrant function mimic that of Sjögren's syndrome, a human autoimmune disease characterized by dry eyes and dry mouth. Impeding the leukocyte infiltration into the glands with synthetic fibronectin peptides, which block adhesion, not only prevents the inflammatory pathology but also prevents the persistent eye closure characteristic of these mice.


Subject(s)
Dacryocystitis/genetics , Dacryocystitis/pathology , Eye/pathology , Mice, Knockout/genetics , Transforming Growth Factor beta/physiology , Animals , Cell Adhesion Molecules/metabolism , Cell Adhesion Molecules/physiology , Cytokines/genetics , Dacryocystitis/physiopathology , Isomerism , Lacrimal Apparatus/metabolism , Lacrimal Apparatus/pathology , Mice , Mice, Knockout/anatomy & histology , Mice, Mutant Strains , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
6.
J Infect Dis ; 175(4): 1004-7, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9086170

ABSTRACT

Since certain functions mediated by nitric oxide (NO) decline with age, the age dependence of NO production by macrophages from BALB/c mice was investigated. Lipopolysaccharide-, peptidoglycan-polysaccharide-, or interferon-gamma-stimulated splenic and peritoneal macrophages from young (1 month old), middle-aged (4-5 months old), and old (6-20 months old) BALB/c mice showed a progressive and marked decline in NO production. This age-related decline in inducible NO extended to C57/BL6 and CB6F1 mice. mRNA for inducible NO synthase (iNOS), the enzyme responsible for inducible NO production by macrophages, also declined with age. Importantly, the reduced NO production by macrophages from old mice could be up-regulated by pretreating the mice with either cholera toxin or concanavalin A. These findings indicate that reduced production of NO by murine macrophages correlates directly with advancing age, likely due to deficient signals or signal transduction responsible for iNOS mRNA and protein generation.


Subject(s)
Macrophages/metabolism , Nitric Oxide/biosynthesis , Age Factors , Animals , Blotting, Northern , Female , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/metabolism
7.
J Immunol ; 157(3): 1306-12, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8757639

ABSTRACT

The targeted disruption of the TGF-beta1 gene in mice (TGF-beta1 -/-) leads to extensive inflammation in vital organs, cachexia, and death within 3 to 4 wk. Significant inflammatory lesions develop initially in the periductal regions of the salivary glands and escalate as the animals become symptomatic. These inflammatory sites, characterized by lymphocytic infiltration and increased proliferation, cytokine mRNA expression, and IgG-positive cells, resemble lesions of Sjögren's syndrome. Moreover, the inflammatory pathology, enhanced MHC expression, and Ab production are consistent with an autoimmune-like etiology. Glandular atrophy and loss of acini with reduced saliva production appear to contribute to the wasting syndrome characteristic of the TGF-beta1 -/- mice. To determine whether the structural and functional defects were developmental due to the absence of TGF-beta1 or secondary to the inflammation, TGF-beta1 -/- mice were treated with synthetic fibronectin peptides, which block leukocyte infiltration. Daily systemic injections of RGD, CS-1, and/or peptides derived from the heparin-binding region of the A chain not only prevented leukocyte infiltration in the salivary glands of the TGF-beta1 -/- mice, but also reversed the acinar and ductal derangements. These data suggested that salivary gland development is not jeopardized in the absence of TGF-beta1, but that the extensive infiltration of inflammatory cells compromises glandular structure and function. The essential nature of TGF-beta1 in controlling inflammatory and immune processes is confirmed by these studies. Moreover, these TGF-beta1 -/- mice provide an important model of autoimmune disease that can be used in the design of therapeutic interventions.


Subject(s)
Autoimmunity , Fibronectins/pharmacology , Salivary Glands/pathology , Sjogren's Syndrome/pathology , Transforming Growth Factor beta/deficiency , Animals , Base Sequence , Cell Division , Interferon-gamma/genetics , Interleukin-2/genetics , Interleukin-6/genetics , Mice , Molecular Sequence Data , RNA, Messenger/analysis , Receptors, Interleukin-2/analysis , Saliva/chemistry , Sjogren's Syndrome/immunology
8.
J Immunol ; 153(5): 1936-46, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-8051399

ABSTRACT

Approximately 2 wk after birth, mice having a TGF-beta 1 null mutation (TGF-beta 1(-/-)) exhibit a progressive wasting syndrome and death. Associated with this phenotype is a multifocal infiltration of lymphocytes and macrophages into target organs, especially the heart, lungs, and salivary glands. To explore the consequences of TGF-beta 1 deficiency on the immune system, lymphocyte phenotype and function were analyzed. Initially, lymphoid organ architecture seemed to be normal and, as symptoms developed, the thymus decreased in size, whereas lymph nodes were enlarged. Phenotypically, the TGF-beta 1(-/-) lymphoid cells seemed to be more differentiated in the thymus and activated in the lymph nodes, but remarkably unaffected in the spleen. Moreover, TGF-beta 1(-/-) spleen and lymph nodes displayed enhanced numbers of proliferating cells, as measured by proliferating cell nuclear Ag and/or cyclin-dependent kinase levels. Consistent with this hyperproliferative response, constitutive levels of IL-2 mRNA were elevated in the thymus and both IL-2 and IL-2R mRNA were increased in the lymph nodes. In contrast with the activation profile of TGF-beta 1(-/-) lymphoid cells in vivo, mitogen challenge of these cells in vitro revealed suppressed proliferation that was associated with a defect in inducible IL-2 mRNA expression and IL-2 secretion. Moreover, the addition of rIL-2 restored the deficient mitogen-induced proliferation. The mechanism leading to T cell anergy remains unclear; however, these data confirm the essential role for TGF-beta 1 in maintaining normal immune function.


Subject(s)
Lymphocyte Activation , Transforming Growth Factor beta/physiology , Animals , Base Sequence , DNA Primers/chemistry , Gene Expression , Immunophenotyping , Interleukin-2/genetics , Interleukin-2/pharmacology , Lymphoid Tissue/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , RNA, Messenger/genetics , Receptors, Interleukin-2/genetics
9.
Proc Natl Acad Sci U S A ; 91(11): 5187-91, 1994 May 24.
Article in English | MEDLINE | ID: mdl-8197206

ABSTRACT

Pronounced mononuclear leukocyte (MNL) infiltration occurs in multiple organs of mice homozygous for a transforming growth factor beta 1 (TGF-beta 1) loss-of-function gene mutation [TGF-beta 1 (-/-)], followed by cachexia and eventually death. Consistent with the increased leukocyte adhesion and tissue infiltration, MNLs isolated from spleen, thymus, and peripheral blood of symptomatic TGF-beta 1 (-/-) mice, as compared to littermate controls, exhibited increased adhesion to extracellular matrix proteins and to endothelial cells in vitro. Incubation of TGF-beta 1 (-/-) MNLs with selected synthetic peptides corresponding to cell- and heparin-binding sequences of fibronectin (FN) significantly attenuated adhesion of these cells not only to FN but also to endothelial cells in vitro. Based on these observations, mice were treated with the FN peptides in an attempt to rescue them from tissue inflammation and cardiopulmonary failure. Daily injections of a combination of four synthetic FN peptides that interact with beta 1-integrins and/or cell surface proteoglycans blocked the massive infiltration of MNLs into the heart and lungs of TGF-beta 1 (-/-) mice. Peptide treatment initiated on day 8, coincident with the first evidence of increased leukocyte-endothelial cell interactions, not only blocked tissue infiltration but also moderated the lethal wasting syndrome.


Subject(s)
Fibronectins/metabolism , Leukocytes, Mononuclear/cytology , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Cell Adhesion , Cells, Cultured , Chemotaxis, Leukocyte , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Fibronectins/chemical synthesis , Fluorescent Antibody Technique , Mice , Mice, Knockout , Molecular Sequence Data
10.
J Leukoc Biol ; 55(3): 401-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8120457

ABSTRACT

A wide variety of functions, many of which represent opposing activities, have been attributed to TGF-beta, a molecule implicated in embryogenesis, development, and immune and inflammatory processes. This paradoxical behavior of promoting or inhibiting cell growth and function, while important in normal physiology and homeostasis, can contribute to or interrupt pathologic sequelae, making TGF-beta a particularly intriguing molecule for study. New transgenic mouse models displaying targeted alterations in TGF-beta 1 expression offer novel and unique opportunities to determine the essential function(s) of TGF-beta.


Subject(s)
Transforming Growth Factor beta/physiology , Animals , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Models, Biological , Transforming Growth Factor beta/genetics
11.
J Exp Med ; 178(2): 749-54, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-7688035

ABSTRACT

Nitric oxide (NO), a toxic radical gas produced during the metabolism of L-arginine by NO synthase (NOS), has been implicated as a mediator of immune and inflammatory responses. A single injection of streptococcal cell wall fragments (SCW) induces the accumulation of inflammatory cells within the synovial tissue and a cell-mediated immune response that leads destructive lesions. We show here that NO production is elevated in the inflamed joints of SCW-treated rats. Administration of NG-monomethyl-L-arginine, an inhibitor of NOS, profoundly reduced the synovial inflammation and tissue damage as measured by an articular index and reflected in the histopathology. These studies implicate the NO pathway in the pathogenesis of an inflammatory arthritis and demonstrate the ability of a NOS inhibitor to modulate the disease.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Arginine/analogs & derivatives , Arthritis/drug therapy , Acute Disease , Animals , Arginine/therapeutic use , Arthritis/etiology , Cell Wall , Culture Techniques , Female , Nitric Oxide/metabolism , Nitric Oxide Synthase , Rats , Rats, Sprague-Dawley , Streptococcus , Synovial Membrane/metabolism , Synovial Membrane/pathology , omega-N-Methylarginine
12.
Biochem Biophys Res Commun ; 194(2): 819-25, 1993 Jul 30.
Article in English | MEDLINE | ID: mdl-8343166

ABSTRACT

Rat migration inhibitory factor-related protein 8 (MRP8) and MRP14 were identified during screening of a subtracted cDNA library generated to identify differences in gene expression between LEW/N and F344/N rats. The predicted amino acid sequence of rat MRP8 and MRP14 is 60-80% identical with that from human and mouse. Expression of these genes correlated with chronic inflammation in LEW/N rats, but were absent in F344/N rats which do not develop arthritis in response to streptococcal cell wall peptidoglycan-polysaccharide complexes (SCW). These differences suggest a role for MRP8 and MRP14 in susceptibility to SCW-induced chronic disease.


Subject(s)
Arthritis, Experimental/genetics , Calcium-Binding Proteins/genetics , Gene Expression , Amino Acid Sequence , Animals , Base Sequence , Calcium-Binding Proteins/biosynthesis , Calgranulin A , Calgranulin B , Cloning, Molecular , DNA , Female , Genetic Predisposition to Disease , Humans , Mice , Molecular Sequence Data , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Rhamnose , Sequence Homology, Amino Acid , Species Specificity
13.
J Exp Med ; 173(4): 981-91, 1991 Apr 01.
Article in English | MEDLINE | ID: mdl-2007861

ABSTRACT

The multifunctional cytokine, transforming growth factor beta (TGF-beta), was identified by immunocytochemistry in the brain tissues of four patients with acquired immune deficiency syndrome (AIDS), but not in control brain tissue. The TGF-beta staining was localized to cells of monocytic lineage as well as astrocytes, especially in areas of brain pathology. In addition, the brain tissues from the AIDS patients contained transcripts for human immunodeficiency virus 1 (HIV-1) by in situ hybridization, suggesting a correlation between the presence of HIV-1 in the brain and the expression of TGF-beta. However, the expression of TGF-beta was not limited to HIV-1-positive cells, raising the possibility of alternative mechanisms for the induction of TGF-beta in these AIDS patients' brains. To investigate these mechanisms, purified human monocytes were infected in vitro with HIV-1 and were shown to secrete increased levels of TGF-beta. In addition, HIV-1-infected monocytes released a factor(s) capable of triggering cultured astrocytes that are not infected with HIV-1 to secrete TGF-beta. The release of TGF-beta, which is an extremely potent chemotactic factor, may contribute to the recruitment of HIV-1-infected monocytic cells, enabling viral spread to and within the central nervous system (CNS). Moreover, TGF-beta augments cytokine production, including cytokines known to be neurotoxic. The identification of TGF-beta within the CNS implicates this cytokine in the immunopathologic processes responsible for AIDS-related CNS dysfunction.


Subject(s)
AIDS Dementia Complex/physiopathology , Astrocytes/physiology , Macrophages/physiology , Transforming Growth Factor beta/physiology , Astrocytes/metabolism , Blotting, Northern , Brain/microbiology , HIV-1/growth & development , Humans , In Vitro Techniques , Male , Monocytes/metabolism , RNA, Messenger/genetics , Transforming Growth Factor beta/genetics
14.
J Clin Invest ; 85(1): 192-9, 1990 Jan.
Article in English | MEDLINE | ID: mdl-2295695

ABSTRACT

A population of circulating mononuclear cells from patients with AIDS was identified which expressed interleukin 2 receptors (IL-2R). By dual-fluorescence flow microfluorometry, the patients' IL-2R+ cells were further identified as Leu M3+ monocytes (29.4 +/- 5.2% of the Leu M3+ cells were IL-2R+, n = 15), whereas Leu M3+ monocytes from normal subjects were IL-2R negative (2.0 +/- 0.42%; P less than 0.001). By Northern analysis, monocytes from AIDS patients, but not control subjects, constitutively expressed steady-state levels of IL-2R mRNA. Functionally, the IL-2R+ monocytes were capable of depleting IL-2 from culture supernatants, suggesting a mechanism for the reduced IL-2 levels commonly seen in AIDS patients. IL-2R+ monocytes also expressed increased levels of surface HLA-DR which may favor monocyte T-cell interactions and the transmission of human immunodeficiency virus (HIV). In additional studies, normal monocytes were infected with a macrophage-tropic HIV isolate in vitro and monitored for IL-2R and HLA-DR expression. Within 24-48 h after exposure to HIV in vitro, but before evidence of productive infection, greater than 25% of the monocytes became IL-2R+ with increasing numbers of IL-2R+ cells and HLA-DR levels through day 6. These early signaling effects of HIV could be mimicked by adding purified HIV envelope glycoprotein gp120 to the monocytes. This stimulation of monocytes before or independent of productive infection of the cells by HIV is consistent with in vivo observations of activated and/or abnormal functions by monocytes that do not appear to be infected with HIV in AIDS patients.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , Cell Transformation, Viral , Gene Expression , HIV/genetics , Monocytes/immunology , Receptors, Interleukin-2/genetics , Acquired Immunodeficiency Syndrome/blood , Acquired Immunodeficiency Syndrome/complications , Antigens, Surface/analysis , Cells, Cultured , Homosexuality , Humans , Lipopolysaccharides/pharmacology , Male , Monocytes/drug effects , RNA, Messenger/genetics , Receptors, Interleukin-2/biosynthesis , Reference Values , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/immunology , Transcription, Genetic/drug effects
15.
Growth Factors ; 4(1): 27-35, 1990.
Article in English | MEDLINE | ID: mdl-1707635

ABSTRACT

Transforming growth factor beta 1 (TGF-beta 1) and its closely related homologue, TGF-beta 2, rapidly induce growth factor gene expression by freshly isolated human peripheral blood monocytes. Within 3 h of exposure to TGF-beta, mRNA species specific for interleukin-1 (IL-1 beta), tumor necrosis factor-alpha (TNF-alpha), platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF) were observed. By 14-18 h, cytokine bioactivity and protein were detected in the culture supernatants. Furthermore, not only TGF-beta 1, but also TGF-beta 2 mRNA are expressed constitutively in unstimulated monocytes. However, in response to exogenous TGF-beta (beta 1 or beta 2), only TGF-beta 1 gene expression is upregulated, and the expression of TGF-beta 2 mRNA is unchanged. This selective autoinduction of TGF-beta 1 appears to be controlled at both transcriptional and post-transcriptional levels. These paracrine and autocrine activities of TGF-beta suggest potential mechanisms through which an inflammatory response can be initiated and amplified. In addition, the TGF-beta enhancement of growth factor generation may promote fibrosis and angiogenesis relevant to physiological tissue repair as well as pathological fibrotic sequelae.


Subject(s)
Growth Substances/biosynthesis , Leukocytes, Mononuclear/metabolism , Transforming Growth Factor beta/physiology , Blotting, Northern , Cells, Cultured , Cycloheximide/pharmacology , DNA Probes , Gene Expression Regulation , Growth Substances/genetics , Half-Life , Humans , Interleukin-1/biosynthesis , Leukocytes, Mononuclear/drug effects , RNA/metabolism , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
17.
Immunol Today ; 10(8): 258-61, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2478145

ABSTRACT

Transforming growth factors (TGFs) are small polypeptides that were initially defined by their ability to induce transformation of non-neoplastic cells in culture. However, it has become increasingly clear that TGFs are not restricted in function to promoting cell growth. One type of transforming growth factor, TGF-beta, is a multifunctional molecule which has unique and potent effects on many target cells and tissues. In this article, Sharon Wahl, Nancy McCartney-Francis and Stephan Mergenhagen focus on the evolving role of TGF-beta in regulating inflammation, immune responses and tissue repair.


Subject(s)
Transforming Growth Factors/immunology , Animals , Inflammation/etiology , Mice , Neovascularization, Pathologic
18.
J Cell Biochem ; 40(2): 193-9, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2670976

ABSTRACT

Mononuclear cells generate a variety of hormone-like proteins termed growth factors that are instrumental in the evolution and resolution of inflammatory reactions. Many of these growth regulatory molecules have multifunctional properties. For example, the mononuclear cell-derived growth factors, platelet-derived growth factor (PDGF), and transforming growth factor beta (TGF-beta), are potent leukocyte chemoattractants. In addition, TGF-beta, a product of platelets, T lymphocytes, and monocytes, appears to induce the transcription of other monocyte-derived growth hormone genes. In this regard, picomolar concentrations of TGF-beta stimulate peripheral blood monocytes to transcribe the genes for PDGF (c-sis), basic fibroblast growth factor (FGF), interleukin 1 (IL-1), and tumor necrosis factor (TNF). Furthermore, levels of mRNA for TGF-beta, which is constitutively expressed in resting monocytes, are also increased by exogenous TGF-beta. Each of these monocyte products exhibits a plethora of biological activities on other cell types. T lymphocytes, in response to antigen, contribute to this network by secreting growth factors and lymphokines that regulate monocyte growth factor production.


Subject(s)
Growth Substances/physiology , Inflammation/physiopathology , Wound Healing , Humans
19.
J Exp Med ; 168(4): 1403-17, 1988 Oct 01.
Article in English | MEDLINE | ID: mdl-2971758

ABSTRACT

Group A streptococcal cell wall (SCW)-injected rats exhibit a profound immunosuppression that persists for months after the initial intraperitoneal injection of SCW. The goal of this study was to determine the mechanisms for the suppressed T lymphocyte proliferative responses in this experimental model of chronic inflammation. When spleen cell preparations were depleted of adherent cells, restoration of T cell proliferative responses to Con A and PHA occurred, implicating adherent macrophages in the regulation of immunosuppression. Furthermore, macrophages from SCW-treated animals, when cocultured with normal spleen cells in the presence of Con A or PHA, effectively inhibited the proliferative response. Supernatants from suppressed spleen cell cultures were found to inhibit normal T cell mitogenesis. Taken together, these results implicated a soluble macrophage-derived suppressor factor in the down regulation of T cell proliferation after exposure to SCW in vivo. Subsequent in vitro studies to identify this suppressor molecule(s) revealed the activity to be indistinguishable from the polypeptide transforming growth factor beta (TGF-beta). Furthermore, TGF-beta was identified by immunolocalization within the spleens of SCW-injected animals. The cells within the spleen that stained positively for TGF-beta were phagocytic cells that had ingested, and were presumably activated by, the SCW. These studies document that TGF-beta, previously shown to be a potent immunosuppressive agent in vitro, also effectively inhibits immune function in chronic inflammatory lesions in vivo.


Subject(s)
Immune Tolerance , Lymphocyte Activation , Macrophages/immunology , Streptococcus pyogenes/immunology , Transforming Growth Factors/immunology , Animals , Blotting, Northern , Cell Adhesion , Cell Wall/immunology , Female , Gene Expression Regulation , Immunohistochemistry , Interleukin-1/biosynthesis , Interleukin-2/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Specific Pathogen-Free Organisms , Spleen/analysis , Spleen/immunology , Streptococcus pyogenes/ultrastructure , Transforming Growth Factors/analysis , Transforming Growth Factors/genetics
20.
J Immunol ; 140(9): 3026-32, 1988 May 01.
Article in English | MEDLINE | ID: mdl-3129508

ABSTRACT

Transforming growth factor-beta (TGF-beta), a product of neoplastic and hemopoietic cells, is a bifunctional regulator of the immune response. At femtomolar concentrations, TGF-beta stimulates monocyte migration, and picomolar quantities induce synthesis of monocyte growth factors, including IL-1, that may promote tissue repair by regulating fibrosis and angiogenesis. Paradoxically, TGF-beta at picomolar concentrations also blocks the ability of IL-1 to stimulate lymphocyte proliferation. At 0.01 to 1.0 ng/ml, TGF-beta 1 and its homologue, TGF-beta 2, suppress the IL-1-dependent murine thymocyte proliferation assay. TGF-beta also inhibits human peripheral blood T lymphocyte mitogenesis. Inhibition of cell division appears to occur after activation of the lymphocytes inasmuch as neither gene expression nor translation of IL-2R is suppressed. Furthermore, TGF-beta does not block synthesis of IL-2. Therefore, TGF-beta 1 and TGF-beta 2 likely act at a site distal to IL-1 to block lymphocyte DNA synthesis. These findings suggest that TGF-beta secreted in an inflammatory site may be beneficial in diminishing lymphocyte function while promoting fibrosis and tissue repair. However, TGF-beta generated by neoplastic tissues may provide a mechanism for unrestricted tumor cell growth through its selective immunosuppressive effects.


Subject(s)
Immunosuppressive Agents , Interleukin-1/antagonists & inhibitors , Lymphocyte Activation , Monocytes/physiology , Peptides/pharmacology , Humans , In Vitro Techniques , Interleukin-2/biosynthesis , Receptors, Immunologic/metabolism , Receptors, Interleukin-2 , Receptors, Transferrin/metabolism , Transforming Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...