NF-kappa B activation

TNF and interruption of apoptosis (programmed cell death)

seem to play an important role in persistence of most (all?)

chronic INTRACELLULAR INFECTIONs

 

- if you have additional references or comments, please send a note to Marie Kroun -

 

Hostile takeovers: viral appropriation of the NF-kappaB pathway.

Hiscott J et al. J Clin Invest 2001 Jan;107(2):143-51.   PubMed: 11160127  PDF

Transcriptional regulators of the NF-kB/IkB family promote the expression of well over 100 target genes, the majority of which participate in the host immune response (1). These proteins include a multitude of cytokines and chemokines, receptors required for immune recognition, proteins involved in antigen presentation, and adhesion receptors involved in transmigration across blood vessels walls. Because of this extensive role in immune action, NF-kB has been termed the central mediator of the immune response. Gene knockout and other studies establish roles for NF-kB in the ontogeny of the immune system but also demonstrate that NF-kB participates at multiple steps during oncogenesis (2) and the regulation of programmed cell death (3).
For several reasons, the NF-kB pathway provides an attractive target to viral pathogens. Activation of NF-kB is a rapid, immediate early (IE) event that occurs within minutes after exposure to a relevant inducer, does not require de novo protein synthesis, and results in a strong transcriptional stimulation of several early viral as well as cellular genes. In this review, we will describe strategies that viruses have evolved to modulate the NF-kB pathway, to enhance viral replication, host cell survival, and evasion of immune responses. Activation of NF-kB constitutes an obvious target because many of its target genes — growth factors, cytokines and their receptors, and proto-oncogenes — profoundly influence the host cell cycle. In addition, some viruses exploit the antiapoptotic properties of NF-kB to evade the host defense mechanisms that limit replication by killing infected cells, or conversely to trigger apoptosis as a mechanism to increase virus spread.
Perhaps not surprisingly, the persistent activation of the NF-kB pathway maintained by certain viruses contributes to oncogenic transformation
(
2). In addition to the classic studies with the avian REV-T retrovirus which contains the v-Rel oncoprotein and induces a rapid and fatal B-cell lymphoma in young birds (4), several lines of evidence demonstrate that NF-kB family members contribute to human oncogenesis. Localization of NF-kB–encoding genes at sites of chromosomal translocations and genomic rearrangements in cancer, high levels of NF-kB activity in many breast cancer cells, and constitutive nuclear NF-kB complexes in Hodgkin’s lymphoma cells all support this view (2). Furthermore, as discussed below, viral oncogene products, including human T-cell leukemia virus type 1 (HTLV-1) Tax protein and Epstein-Barr virus latent infection membrane protein 1 (EBV LMP1), each act by unique mechanisms to disrupt NF-kB regulation and initiate viral transformation.

 

It appears, however, that also several bacterial or parasitic intracellular infections activate NF-kB pathway like described above for the viral infections.

 

Searching Medline for NF-kB and several intracellular ‘parasites’, that are all associated with chronic disease in humans and/or animals, certainly give a lot of hits via the formula: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Search&db=PubMed&term=NF-kappaB+AND+*   

- exchange the ‘*’ in the end for the pathogens, if the links below doesn’t work …


Vira:

Cytomegalovirus, Human papilloma virus, Human immunodeficiency virus,

Human T-lymfototrophic virus, Ebstein-Barr virus (mononucleosis), Hepatitis C virus,

Hepatitis B virus, Human herpes virus, Coxsackievirus

 

Bacteria:

Borrelia, Treponema (syphilis), Leptospira (rat bite fever), Chlamydia, Mycoplasma,

Mycobacteria (tuberculosis), Rickettsia, Ehrlichia, Bartonella .... other?

Parasites:

Malaria, Theileria

 

The only intracellular parasite I searched for, which is also known to be associated with chronic persistent infection, that gave no hits at all, was Babesia.

I suppose it is because Babesia hasn’t been investigated regarding NF-kB activation yet, since above two piroplasmatic 'cousins' Malaria and Theileria does induce NF-kB, there is reason to suspect that Babesia also knows the same trick.

I’ve certainly not searched for all , just enough to give a the reader a hint  ….

 

When trying to combat intracellular infections, one cytokine of major importance is TNF, which induces apoptosis (programmed cell death), which can be inhibited by activation of NF-kB:

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Search&db=PubMed&term=NF-kappaB+AND+apoptosis

 

There are so many articles on this interesting subject, but I can only mention a few on selected tickborne infections and similar, with are the infections of special interest to me:

 

NF-kB-dependent inhibition of apoptosis is essential for host cell survival during Rickettsia rickettsii infection

Clifton DR et al. Proc Natl Acad Sci U S A 1998 Apr 14;95(8):4646-51  PMID: 9539792  PDF  [& PMID: 12819104  PDF]

The possibility that bacteria may have evolved strategies to overcome host cell apoptosis was explored by using Rickettsia rickettsii, an obligate intracellular Gram-negative bacteria that is the etiologic agent of Rocky Mountain spotted fever. The vascular endothelial cell, the primary target cell during in vivo infection, exhibits no evidence of apoptosis during natural infection and is maintained for a sufficient time to allow replication and cell-to-cell spread prior to eventual death due to necrotic damage. Prior work in our laboratory demonstrated that R. rickettsii infection activates the transcription factor NF-kappa B and alters expression of several genes under its control. However, when R. rickettsii-induced activation of NF-kappa B was inhibited, apoptosis of infected but not uninfected endothelial cells rapidly ensued. In addition, human embryonic fibroblasts stably transfected with a superrepressor mutant inhibitory subunit Ikappa B that rendered NF-kappa B inactivatable also underwent apoptosis when infected, whereas infected wild-type human embryonic fibroblasts survived. R. rickettsii, therefore, appeared to inhibit host cell apoptosis via a mechanism dependent on NF-kappa B activation. Apoptotic nuclear changes correlated with presence of intracellular organisms and thus this previously unrecognized proapoptotic signal, masked by concomitant NF-kappa B activation, likely required intracellular infection. Our studies demonstrate that a bacterial organism can exert an antiapoptotic effect, thus modulating the host cell's apoptotic response to its own advantage by potentially allowing the host cell to remain as a site of infection.

 

Activation of human monocytic cells by Treponema pallidum and Borrelia burgdorferi lipoproteins and synthetic lipopeptides proceeds via a pathway distinct from that of lipopolysaccharide but involves the transcriptional activator NF-kappa B.
Norgard MV et al. Infect Immun 1996 Sep; 64(9): 3845-52  PMID: 10438943    PDF

There is increasing evidence that lipoproteins of Treponema pallidum and Borrelia burgdorferi are key inflammatory mediators during syphilis and Lyme disease. A principal objective of the present study was to identify more precisely similarities and divergences among lipopolysaccharide (LPS)- and lipoprotein-lipopeptide-induced immune cell signaling events. Like LPS, purified native B. burgdorferi OspA and synthetic analogs of OspA, OspB, and two T. pallidum lipoproteins (Tpp47 and Tpp17) all induced NF-kappa B translocation in THP-1 human monocytoid cells. Acylation of OspA and the synthetic peptides was requisite for cell activation. Polymyxin B abrogated only the response to LPS. By using 70Z/3-derived pre-B-cell lines either lacking or expressing human CD14 (the LPS receptor), it was observed that expression of human CD14 imparted responsiveness to LPS but not to OspA or spirochetal lipopeptides (assessed by induction of NF-kappa B and expression of surface immunoglobulin M). Finally, the biological relevance of the observation that T. pallidum lipoproteins-lipopeptides induce both NF-kappa B and cytokine production in monocytes was supported by the ability of the synthetic analogs to promote human immunodeficiency virus replication in chronically infected U1 monocytoid cells; these observations also suggest a potential mechanism whereby a syphilitic chancre can serve as a cofactor for human immunodeficiency virus transmission. The combined data lend additional support to the proposal that spirochetal lipoproteins and LPS initiate monocyte activation via different cell surface events but that the signaling pathways ultimately converge to produce qualitatively similar cellular responses.

 

Borrelia burgdorferi activates nuclear factor-kappa B and is a potent inducer of chemokine and adhesion molecule gene expression in endothelial cells and fibroblasts.
Ebnet K, Brown KD, Siebenlist UK, Simon MM, Shaw S. J Immunol 1997 Apr 1; 158(7): 3285-92  PMID: 9120285

Lyme disease, caused by the tick-borne spirochete Borrelia burgdorferi, is a systemic infection with preponderance for the skin, joints, heart, and nervous system. Inflammatory lesions of target organs are characterized by the presence of spirochetes and inflammatory leukocytes. We have analyzed the potential of B. burgdorferi to induce gene expression of chemokines and adhesion molecules in human endothelial cells, keratinocytes, and fibroblasts. We find induction of the chemokines RANTES (regulated upon activation, normal T cells expressed and secreted), monocyte chemoattractant protein-1, IL-8, gro-alpha, IFN-inducible protein-10, and mig (monokine induced by gamma-IFN), and of the adhesion molecules E-selectin, ICAM-1, and VCAM-1 in endothelial cells and induction of the same chemokines and ICAM-1 in fibroblasts. This is mediated by the lipid moiety of the outer surface lipoprotein A. Induction of chemokine and adhesion molecule genes by B. burgdorferi occurs rapidly and does not require new protein synthesis. Induction is blocked by inhibitors of nuclear factor (NF)-kappa B. We also find that B. burgdorferi induces nuclear translocation of NF-kappa B and a transient increase in the expression of its inhibitor I kappa B-alpha. These findings indicate that B. burgdorferi is a potent inducer of molecules required for leukocyte recruitment to inflammatory foci, and the data suggest that this biologic activity is due to the ability of the spirochetes to activate the pleiotropic transcription factor NF-kappa B.

 

Cytokines involved in human septic shock--the model of the Jarisch-Herxheimer reaction.
Griffin GE. J Antimicrob Chemother 1998 Jan;41 Suppl A:25-9. PMID: 9511083  PDF 

Free download:

Studies of the cytokine cascade in animal models of infection and human experiments involving endotoxin infusion have contributed fundamentally to understanding the role of cytokines in human sepsis. The complexity of this cytokine cascade has been difficult to unravel in clinical sepsis. However, the Jarisch-Herxheimer reaction has been identified as a model of the cytokine cascade in human sepsis and has provided an excellent model for experiments involving blocking agents. TNF blocking has been shown to be important for protection in animal models of sepsis, but has been somewhat disappointing in humans because adverse events have generally outweighed benefits.

 

Detection of plasma tumor necrosis factor, interleukins 6, and 8 during the Jarisch-Herxheimer Reaction of relapsing fever.
Negussie Y et al. J Exp Med 1992 May 1; 175(5): 1207-12  PMID: 1569394  PDF

The Jarisch-Herxheimer Reaction (J-HR) is a clinical syndrome occurring soon after the first adequate dose of an antimicrobial drug to treat infectious diseases such as Lyme disease, syphilis, and relapsing fever. Previous attempts to identify factors mediating this reaction, that may cause death, have been unsuccessful. We conducted a prospective trial in Addis Ababa, Ethiopia on 17 patients treated with penicillin for proven louse-borne relapsing fever due to Borrelia recurrentis to evaluate the association of symptoms with plasma levels of tumor necrosis factor (TNF), interleukins 6, and 8 (IL-6 and -8). 14 of the 17 (82%) patients experienced a typical J-HR consisting of rigors, a rise in body temperature (1.06 +/- 0.2 degrees C) peaking at 2 h, leukopenia (7.4 +/- 0.6 x 10(-3) cells/mm3) at 4 h, a slight decrease, and then rise of mean arterial blood pressure. Spirochetes were cleared from blood in 5 +/- 1 h after penicillin. There were no fatalities, but constitutional symptoms were severe during J-HR. Plasma TNF, IL-6, and -8 were raised in several patients on admission, but a seven-, six-, and fourfold elevation of these plasma cytokine concentrations over admission levels was detected, respectively, occurring in transient form coincidental with observed pathophysiological changes of J-HR. Elevated plasma cytokine levels were not detected in the three patients who did not suffer J-HR. We conclude that the severe pathophysiological changes characterizing the J-HR occurring on penicillin treatment of louse-borne relapsing fever are closely associated with transient elevation of plasma TNF, IL-6, and -8 concentrations.

 

Prevention of Jarisch-Herxheimer reactions by treatment with antibodies against tumor necrosis factor alpha [see comments]

Fekade D et al. N Engl J Med 1996 Aug 1; 335(5): 311-5  PMID: 8663853   PDF

BACKGROUND: In patients with louse-borne relapsing fever (Borrelia recurrentis infection), antimicrobial treatment is often followed by sudden fever, rigors, and persistent hypotension (Jarisch-Herxheimer reactions) that are associated with increases in plasma concentrations of tumor necrosis factor alpha (TNF-alpha), interleukin-6, and interleukin-8. We attempted to determine whether sheep polyclonal Fab antibody fragments against TNF-alpha (anti-TNF-alpha Fab) could suppress the Jarisch-Herxheimer reaction. METHODS: We conducted a randomized, double-blind, placebo-controlled trial in 49 patients with proven louse-borne relapsing fever. Immediately before the intramuscular injection of penicillin, the patients received an intravenous infusion of either anti-TNF-alpha Fab or a control solution. RESULTS: Ten of the 20 patients given anti-TNF-alpha Fab had Jarisch-Herxheimer reactions with rigors, as compared with 26 of the 29 control patients (P = 0.006). The controls had significantly greater mean maximal increases in temperature (1.5 vs. 0.8 degrees C, P < 0.001), pulse rate (31 vs. 13 per minute, P < 0.001), and systolic blood pressure (25 vs. 15 mm Hg, P < 0.003), as well as higher mean peak plasma concentrations of interleukin-6 (50 vs. 17 micrograms per liter) and interleukin-8 (2000 vs 205 ng per liter) (P < 0.001 for both comparisons). Levels of TNF-alpha were undetectable after treatment with anti-TNF-alpha Fab. CONCLUSIONS: Pretreatment with sheep anti-TNF-alpha Fab suppresses Jarisch-Herxheimer reactions that occur after penicillin treatment for louse-borne relapsing fever, reduces the associated increases in plasma concentrations of interleukin-6 and interleukin-8, and may be useful in other forms of sepsis.

 

Suppression of acute Ixodes scapularis-induced Borrelia burgdorferi infection using tumor necrosis factor-alpha, interleukin-2, and interferon-gamma.
Zeidner N et al. J Infect Dis 1996 Jan; 173(1): 187-95  PMID: 8537658

Down-regulation of mammalian cytokine production has been demonstrated during tick feeding. To examine the hypothesis that reconstitution of cytokines during tick feeding could facilitate immune containment of Borrelia burgdorferi, the following experiments were done. C3H/HeJ mice were given cytokines for 10 days after Ixodes scapularis attachment. At day 21, ear biopsies were analyzed for B. burgdorferi. Polymerase chain reaction analysis indicated a protection rate of 95% in mice receiving tumor necrosis factor (TNF)-alpha. Mice that received interleukin (IL)-2 or interferon (IFN)-gamma had infection rates of 30%-45% compared with 83% for untreated controls. No correlation was noted between neutralizing antibody, reactivity by Western blot, and subsequent protection. Culture of B. burgdorferi in cytokine-conditioned media indicated that TNF-alpha, IFN-gamma, and IL-2 were not cytotoxic for B. burgdorferi. These data suggest that cytokine-induced protection from B. burgdorferi infection was immune-mediated and that cellular immunity may be associated with protection from I. scapularis-induced infection.              

 

Synergistic effects of interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha: central monoamine, corticosterone, and behavioral variations.

Brebner K et al. Neuropsychopharmacology 2000 Jun;22(6):566-80  PMID: 10788757

The proinflammatory cytokines interleukin-1beta (IL-1beta), IL-6, and tumor necrosis factor-alpha (TNF-alpha) influence neuroendocrine activity, promote central neurotransmitter alterations, and induce a constellation of symptoms collectively referred to as sickness behaviors. These cytokines may also elicit anxiety and anhedonia, and have been associated with psychological disturbances in humans. In the present investigation, systemic IL-1beta and TNF-alpha dose-dependently and synergistically disrupted consumption of a highly palatable food source (chocolate milk), possibly reflecting anorexia or anhedonia engendered by the treatments. As well, these cytokines synergistically increased plasma corticosterone levels. Although IL-1beta and TNF-alpha provoked variations of amine turnover in the hypothalamus, locus coeruleus, and central amygdala, synergistic effects were not evident in this respect. Nevertheless, in view of the central amine variations induced by the cytokines, it is suggested that immune activation may come to influence complex behavioral processes, as well as affective state.

 

Role of T cells and cytokines in fatal and resolving experimental babesiosis: protection in TNFRp55-/- mice infected with the human Babesia WA1 parasite. Hemmer RM et al. J Parasitol 2000 Aug;86(4):736-42   PMID: 10958449

The full discussion section from this reference is cited below, because the authors give a thorough explanations for the role of TNF-a and other cytokines (including a lot of references) - and this is possibly a common pathogenetic 'pathway' for several (all?) intracellular parasitic infections ?

DISCUSSION:

TNF-a is a pleiotropic cytokine that appears to be an integral component of the immunologic defense network. TNF-a seems to be required for resistance to several intracellular facultative bacteria and parasites, including Listeria monocytogenes and Leishmania major (Havell, 1989; Green et al., 191990; Kaufmann, 1993). In contrast, other evidence demonstrates the detrimental effects of TNF-a on the host. TNF-a is involved in triggering the lethal effects of cachexia, septic shock syndrome, inflammation, and other systemic manifestations of disease (Tracey and Cerami, 1994). The fact that this cytokine is crucial in organizing the events involved in both immunity and disease demonstrates the complexity of TNF-a function. Previous studies from this laboratory demonstrated that WA1 babesial infection results in pulmonary edema, infiltration and adhesion of mononuclear cells to the pulmonary veins, and endothelial cell activation, as demonstrated by hypertrophy and upregulation of intracellular adhesion molecule-1 (Hemmer et al., 1999). There is much evidence that these inflammatory responses can be mediated by TNF-a (Mulligan et al., 1993; Urquhart, 1994; Luca et al., 1997). The effector functions of TNF-a include increased vascular permeability, upregulation of adhesion molecule expression on endothelial cells, and stimulation of other changes in vascular endothelium that contribute to tissue injury. Thus, the pathology observed in WA1-infected mice correlates with known biological effects of TNF-a and suggests a role for its activity in the WA1-associated disease.

In the present study we examined the difference in TNF-a production by CD4+, CD8+, and gd+ splenic T-cell subpopulations in fatal and resolving Babesia infections. Upregulation of TNF-a production by gd+ T cells in fatal WA1 infections, but not in resolving infections, suggested that TNF-a production was detrimental to the host. Additional support for the role of TNF-a in the pathogenesis of WA1 came from infections of TNFRp55-/- mice. These mice are resistant to either lipopolysaccharide or bacterial superantigen-induced septic shock, a syndrome mediated by TNF-a (Pfeffer et al., 1993). Furthermore, in the characterization of TNFRp55-/- mice, it was shown that inoculation of TNF-a triggered mononuclear cell infiltration of lung, liver, and kidney in C57BL/6 mice, but not in TNFRp55-/- mice (Neumann et al., 1996). This provides evidence that the TNFRp55 receptor is necessary for TNF-a mediated leukocyte adhesion to activated endothelium. In our study, the TNFRp55-/- mice survived a lethal WA1 infection, thus demonstrating that abolishing TNFRp55 function eliminated an important pathogenic mechanism of the WA1 parasite. This result suggests that during a WA1 infection, signaling through TNFRp55 promotes the recruitment of leukocytes to the lungs, and the outcome is activation of the vascular endothelium, pulmonary edema, respiratory distress, and death. Taken together, these results implicate TNF-a as an essential component of the WA1-associated pathology.

In addition to TNF-a, other cytokines were involved in Babesia infections. IFN-g, another proinflammatory cytokine, was shown to have significanfly higher peak expression by CD8+ T cells in WA1-infected mice compared to mice infected with B. microti. IFN-g is the principal macrophage-activating cytokine and central in the regulation of TNF-a (Rudin et al., 1997). There is evidence that IFN-g synergizes with TNF-a to enhance an inflammatory response (Tracey and Cerami, 1994). Upregulation of both of these cytokines during the WA1 infection could account for the severity of the WA1-associated disease.

In resolving B. microti infections, increased production of both IL-10 and IL-4 by gd+ T cells occurred as the parasite load was decreasing. Thus. in B. microti infections, increased expression of IL-10 and IL-4 was important for resolution of the parasitemia.

One striking observation was that gd+ T cells were the major cytokine-producing cells in both WA1 and B. microti infections. gd+ T cells act as a first line of defense against a pathogen and response of gd+ T cells to antigen happens faster than that ab+ [?, hard to read on my copy] T cells (Boismenu and Havran, 1997). These responses indicate that the cytokine profile of  gd+ T cells may regulate the functional activities of both innate and acquired immune responses. Production of IFN-g by  gd+ T cells has been shown to prime macrophages for TNF-a expression (Nishimura et al., 1995). Other studies of intracellular organisms imply that control of pathogen dissemination and host tissue damage requires the regulatory influence of gd+ T cells (Rosat et al., 1993; Fu et al.. 1994). These studies cast  gd+ T cells as an important regulatory cell for both innate and acquired immune responses. Despite the fact that  gd+ T cells were the predominant cytokine-producing cells detected in our experiments, gd-/- mice behaved similar to controls when they were infected with either WA1 or B. microti. This result is not entirely surprising in light of the fact that  gd+ T cells make up a small percentage of all T cells, approximately 2-5% in the mouse spleen. Certainly other cell types are major producers of cytokines. Macrophages and natural killer cells that produce TNF-a and IFN-g, respectively, could contribute to the WA1-associated pathology. The flow cytometry results showed limited involvement of cytokine production by CD4+ and CD8+ T cells. This was probably not due to technical difficulties, because IFN-g production was observed in CD8+ T cells, and IL-2 expression was detected in CD4+ T cells (data not shown). It is possible that the cytokine expression was below the level of detection for this assay, or that cytokines other than the ones we tested are involved in Babesia infections. It is important to note that in this study, the T cells were not restimulated in vitro, and this may explain the paucity of cytokine expression by CD4+ T cells. Because it was well established that CD4+ and CD8+ T cells have many important functions in defense against intracellular pathogens, we used CD4-/- and CD8-/- mice to study the effects of an absence of these T-cell subpopulations on the outcome of WA1 and B. microti infections. There was a significant delay in parasite clearance in both WA1- and B. microti-infected CD4-/- mice. A similar persistence in parasitemia was observed in mice depleted of CD4+ T cells by monoclonal antibody treatment (Shimada et al., 1996). These results demonstrate a role for CD4+ T cells in the elimination of parasites from the blood. Following WA1 inoculation, CD8-/- mice had a moderately increased survival rate. One possibility is that because CD8+ T cells are a major source of lFN-g, the absenec of CD8+ T cells reduces the overall availabilty of IFN-g (Boehm et al., 1997). Consequently, the synergistic effects of IFN-g and TNF-a are inhibited, leading to a reduced inflammatory response and increased survival. The protection in B. microti-infected CD4-/- and CD8-/- mice suggests that either CD4+ or CD8+ T cells, without the other subset present, but in the presence of B cells, gd+ T cells, and innate immune cells can be sufficient for immunity. Our results emphasize the plasticity and redundancy of CD4+ and CD8+ T-cell functions in response to intracellular pathogens.

These studies were undertaken to gain a more complete understanding of the role of T cells and cytokines in the pathogenesis of WA1 infections and the resolution of B. microti infections. We have shown that cytokine responses of splenic T cells were different during fatal and resolving infections and that cytokines contribute to both host pathology and host immunity. TNF-a activity through TNFRp55 was essential to the WA1-associated disease. Additional studies of T cells, B cells, endothelial cells, and innate immune responses are needed to elucidate the different mechanisms involved in fatal and resolving Babesia infections. Future experiments will be directed at studying the role of cytokine-stimulated endothelial cells and macrophages in WA1 pathology.

 

In vitro and in vivo induction of tumor necrosis factor alpha by Borrelia burgdorferi.

Defosse DL, Johnson RC.  Infect Immun 1992 Mar; 60(3): 1109-13. PMID: 1541526   PDF 

Tumor necrosis factor alpha (TNF-alpha) is an immunoregulatory cytokine with many biological activities including the mediation of inflammation. We examined sera and synovial fluids from patients seropositive for infection with Borrelia burgdorferi using a radioimmunoassay specific for TNF-alpha. Significant elevation of TNF-alpha was found in the sera and synovial fluids of patients examined, while controls showed no elevation. Sera of mice infected with B. burgdorferi contained elevated levels of TNF-alpha which varied during the course of a 24-day infection. To determine whether B. burgdorferi is capable of inducing TNF-alpha production, spirochetes were added to adherent human peripheral blood mononuclear cells or mouse peritoneal exudate cells and 24 h later supernatants were assayed. TNF-alpha induction occurred in a dose-dependent manner. The maximum stimulation occurred when a ratio of 1 to 10 spirochetes per mononuclear cell was used. At optimal concentrations, induction was not diminished by inactivation of spirochetes or pretreatment with polymyxin B. These results suggest that an increase in TNF-alpha production may occur as a result of infection with B. burgdorferi.

 

Microbial Lipopeptides Induce the Production of IL-17 in Th Cells.

Infante-Duarte C, Horton HF. Byrne MC, Kamradt T. J Immunol 2000 Dec 1;165(11):6107-6115.  PMID: 11086043   PDF

Naive Th cells can be directed in vitro to develop into Th1 or Th2 cells by IL-12 or IL-4, respectively. In vivo, chronic immune reactions lead to polarized Th cytokine patterns. We found earlier that Borrelia burgdorferi, the spirochaete that causes Lyme disease, induces Th1 development in alphass TCR-transgenic Th cells. Here, we used TCR-transgenic Th cells and oligonucleotide arrays to analyze the differences between Th1 cells induced by IL-12 vs those induced by B. burgdorferi. Transgenic Th cells primed with peptide in the presence of B. burgdorferi expressed several mRNAs, including the mRNA encoding IL-17, at significantly higher levels than Th cells primed with peptide and IL-12. Cytometric single-cell analysis of Th cell cytokine production revealed that IL-17 cannot be categorized as either Th1 or Th2 cytokine. Instead, almost all IL-17-producing Th cells simultaneously produced TNF-alpha and most IL-17(+) Th cells also produced GM-CSF. This pattern was also observed in humans. Th cells from synovial fluid of patients with Lyme arthritis coexpressed IL-17 and TNF-alpha upon polyclonal stimulation. The induction of IL-17 production in Th cells is not restricted to B. burgdorferi. Priming of TCR-transgenic Th cells in the presence of mycobacterial lysates also induced IL-17/TNF-alpha coproduction. The physiological stimulus for IL-17 production was hitherto unknown. We show here for the first time that microbial stimuli induce the expression of IL-17 together with TNF-alpha in both murine and human T cells. Chronic IL-17 expression induced by microbes could be an important mediator of infection-induced immunopathology.             

 

Excerpts:

IL-12 is a dominant factor for Th1-induction both in vitro (6, 7) and in vivo (24). Several microorganisms. including Borrelia burgdorferi can induce IL-12 expression in macrophages (7, 25-29). ….

B. burgdorferi induces not only IL-12 but also other cytokines in host macrophages, including IL-1b (29), TNF-a , IL-6 (30), and IL-10 (31). Whereas IL-1b and TNF-a can support Th1 phenotype development (27, 32,33), IL-6 and IL-10 would be considered antagonistic to Th1 induction (34,35). Therefore, the increased IFN-g production and decreased IL-4 production induced by B. burgdorferi is the net effect og diverse signals, some of which antagonize each other. This complicated situation, rather than Th priming in the presence of selected polarizing cytokines in vitro, is likely to be typical of differentiation in vivo induced by microbial pathogens. …

Thus, IL-17, which has been found in inflammatory lesions (38-41), is induced by microbial stimuli and coexpressed with other proinflammatory cytokines. The failure to down-regulate microbe-induced expression of IL-17 could therefore be one link between infection and autoimmunity.

 

From ‘Results’:

Up-regulation of several cytokine and chemokine mRNA’s by B. burgdorferi or IL-12.

B. burgdorferi and synthetic lipopeptides induce enhanced IL-17 production …. Thus, the presence of B. burgdorferi or the synthetic lipopeptide derived from B. burgdorferi OspA during T cell priming induced enhanced IL-17 production.

IL-17 is produced by activated CD4+, TCR-transgenic T cells.

IL-6 and IL-18 induce enhanced IL-17 production. … Exposure to microbes such as B. burgdorferi or M. bovis BCG strain Danish induces the production of multiple cytokines in different host cells including APC.

IL-17 is coexpressed with TNF-a and GM-CSF, but not with type 1 or 2 cytokines.

IL-17 does not exert an obvious effect on Th-phenotype development.

Coproduction of IL-17 and TNF-a by synovial fluid Th cells from patients with Lyme arthritis and reactive arthritis. [Seven patients met established clinical and serological criteria for the diagnosis of Lyme arthritis. Their clinical characteristics have been described betore (47) One patient suffered from Chlamydia-associated acute reactive arthritis.]

 

Discussion (all text included):

To date, the development, stability, and plasticity of Th subsets have been studied mostly in T cells that were cultured in vitro in the presence of cytokines and Abs (6, 8, 12, 13, 16-20). In addition, Th cells activated in vivo with highly differentiated patterns of cytokine secretion have been isolated from patients or in various models of chronic inflammatory diseases or in chronic infection (14, 15, 23). Little is known about the physiological pathways of Th differentiation and the stability or plasticity of Th cytokine production induced by microbes. Here, we used oligonucleotide arrays to compare the effects of B. burgdorferi on the cytokine production of Th cells with the well-documented effects of exogenously added IL-12 and anti-IL-4. This analysis revealed that a number of genes were exclusively or predominantly induced by B. burgdorferi but not by IL-12. One such gene was that for IL-17, which we decided to analyze in more detail.

 

IL-17 originally had been described as a cytokine produced by activated murine cytotoxic T cells and called CTLA-8 (48). Murine IL-17 is a 21-kDa glycoprotein consisting of 147 amino acids that has 57% sequence identity with a herpesvirus Saimiri protein (HVS 13, also called vIL-l7) (49). It shares 63% amino acid sequence homology with the 155-aa huIL-17 protein, but has no obvious homologies with other cytokines (37, 50). Similar to other mRNAs encoding proinflammatory cytokines, the human and murine IL-17 mRNAs possess several ATTTA motifs (37, 48) associated with rapid mRNA decay (51,52). A receptor for IL-l7 (IL-l7R) with ubiquitous tissue distribution has been described. This IL-17R has no homology with any of the known cytokine receptor families. Furthermore, there are no obvious motifs in the receptor's intracellular domain, which would suggest how signal transduction is mediated following ligation of the IL-l7R (36, 50). Similar to TNF-a , with which it is coexpressed as described here, IL-17 activates the transcription factors NF-k B and AP-1 (36, 50, 53, 54), both of which are involved in the regulation of expression of proinflammatory cytokines. Accordingly, IL-17 exhibits pleiotrophic activities on various parenchymal cells. including the induction of IL-6. IL-8, G-CSF, leukemia inhibitory factor, PGE2, ICAM-l. and matrix metalloproteinase expression in fibroblasts and other stromal cells (36, 37, 50, 55-57), production of NO, iNOS, and cyclooxygenase-2 by chondrocytes (53, 58), induction of osteoclast differentiation factor in osteoblasts (39), stimulation of granulopoiesis (37, 59), maturation of dendritic cells (60), and costimulation of T cell proliferation (36). IL-17 mRNA had been detected in activated murine T cells (36) and human CD4 memory cells (37, 39, 50). Nothing is known about its regulation or the stimuli that naturally induce IL-17 production. Here, we show that IL-17 production by activated Th cells is enhanced 6-fold by microbial lipopeptides.

 

Microbial lipopeptides did not enhance the IL-17 production of T cells primed with plate-bound Abs against CD3 and CD28 in the absence of APC [antigen presenting cell]. Therefore, the microbial lipopeptides act on APC rather than directly on T cells. Bacterial (lipo) proteins, such as OspA or the mycobacterial 19-kDa protein. activate APC after binding to Toll-like receptors (61, 62). Our findings that both spirochaetal lipopeptides and mycobacterial lysates induce IL-17 production in Th cells in an APC-dependent manner are compatible with the hypothesis that Toll-like receptor-mediated activation induces APC to produce a currently unidentified factor(s) that induces IL-17 production in Th cells. In support of this hypothesis, we found that exposure to IL-6 or IL-18, two cytokines which are produced by activated APC, also increased the IL-17 production of activated Th cells. The fact that exogenously added IL-6 erihanced IL-17 production of Th cells is of interest because one of the major in vivo effects of IL-17 is the induction of IL-6, e.g., in rheumatoid synovial cells (36, 37, 50, 55, 56). Furthermore, both TNF-a and IFN-g have additive effects on the IL-17-induced expression of IL-6 (37). The IL-17 mRNA is equipped with eight ATTTA motifs (37, 48), which should normally prevent long-term effects of mirobe-induced IL-17 (51, 52). Chronic infection., e.g.. with B. burgdorferi or Chlamydia trachomatis (63), could maintain an IL-17-inducing stimulus. The reciprocal induction/enhancement of IL-17 and IL-6 could then contribute to the pathogenesis of chronic inflammatory diseases such as antibiotic-resistant Lyme arthritis (42, 63). Thus, IL-17-producing Th cells could become an important therapeutic target for the suppression of chronic inflammation. The role of infections in the other chronic inflammatory conditions in which IL-17 has been detected (38-40, 55, 64, 65) remains to be determined.

 

There is ample evidence for IL-17 as a mediator of inflammation in humans. Several studies have detected IL-17-expressing human T cells in inflammatory lesions. Rheumatoid arthritis is the best-characterized example: IL-17 is expressed, secreted, and functional in rheumatoid synovia (38, 39, 55) but not in osteoarthritic synovia (38, 39). CD45RO+CD4+ T lymphocytes are the source of IL-17 in the synovia as determined by immunohistochemistry (38, 39). Within the inflamed rheumatoid synovium, IL-17 is a major stimulus for the secretion of IL-6 by synoviocytes. When pieces of synovium were cultured for 1 wk in the presence or absence of Abs against IL-17, the production of both IL-6 and leukemia inhibitory factor were reduced by up to 70% in the presence of anti-IL-17 (38, 55). Of note, these highly effective concentrations of IL-17 are detected in rheumatoid synovium, although only ~1% of synovial T cells were found to express IL-17 mRNA by in situ hybridization (38). This is similar to the ~2% synovial fluid Th cells, which we found to produce IL-17 upon polyclonal stimulation, and it is reasonable to assume that this relatively small population may exert significant physiological effects in vivo. Supporting this hypothesis, IL-17 mRNA-expressing cells were found in higher numbers during clinical exacerbations in patients with multiple sclerosis compared with remission, and IL-17 mRNA-expressing MNC were found more frequently in cerebrospinal fluid as compared with peripheral blood in these patients (40). Finally, IL-17 message has been detected in T cells from various inflammatory skin lesions (64, 65) and in rejected but not in functional kidney transplants (66, 67).

 

We show here for the first time that microbial stimuli induce IL-17 production in murine CD4+ Th cells and that the percentage of IL-17 producing synovial fluid T cells from patients with Lyme arthritis or Chlamydia-induced reactive arthritis is significantly higher than in controls. IL-17 production is induced by B. burgdorferi both in mice and in patients and may be of importance for the chronic inflammatory conditions, e.g., antibiotic-resistant arthritis, sometimes induced by infection with B. burgdorferi. The highly coordinate expression of IL-17 with TNF-a , together with its presence and function in chronic inflammatory lesions, argue for an important role of IL-17 in infection-induced inflammatory responses.

 

Our analyses of IL-l7 coexpression with other cytokines revealed that it is neither a typical Th1 nor Th2 cytokine. Accordingly, the in vitro conditions that induce either Th1 or Th2 development had no effect on the frequencies of IL-17-producing Th cells or the amount of IL-17 secreted into the culture supernatants. Vice versa, exogenously added IL-17 did neither affect Thl/Th2 phenotype differentiation nor did it induce IL-17 production. Thus, IL-17 cannot be classified as typical Th1 or Th2 cytokine. Similarly, when the cytokine production of human T cell clones obtained from rheumatoid synovium (68) or from inflammatory skin lesions (64, 65) had been examined, no correlation between IL-17 production and the production of either IL-4 or IFN-g (68) bad been detected such that human IL-17 had previously not been classified as Th1 or Th2 cytokine. Instead, we here describe that almost all IL-17-producing Th cells, murine and human, also produce TNF-a , a potent proinflammatory cytokine that is an effective therapeutic target in chronic inflammatory diseases, such as rheumatoid arthritis (69) and chronic inflammatory bowel disease (70). In both murine and human Th cells. IL-17 production was also positively correlated with that of another proinflammatory cytokine, GM-CSF.

 

In summary, we have shown that microbial lipopeptides induce the production of IL-17 together with TNF-a and GM-CSF. We demonstrated T cells coexpressing IL-17 and TNF-a in the synovial fluid of patients with Lyme arthritis and suggest microbe-induced IL-17 expression as a possible mediator of infection-induced immunopathology.

 

 

The pattern and level of cytokines secreted by Th1 and Th2 lymphocytes of syphilitic patients correlate to the progression of the disease.

Podwinska J, Lusiak M, Zaba R, Bowszyc J. FEMS Immunol Med Microbiol 2000 May;28(1):1-14.  PMID: 10767602    

The results of our previous work indicated that cell-mediated immune response, of importance in protection against Treponema pallidum, is distinctly inhibited at certain periods of syphilitic infection. Considering that cytokines, produced by Th1 lymphocytes, take part in this response and that their secretion may be regulated by cytokines of Th2 lymphocytes, we examined if, and in which stages of syphilis, such a regulation may exist. In this study we have examined the ability of cells to produce IL-2, IFN and TNF (Th1 or Th1 like cytokines) as well as IL-6 and IL-10 (Th2 or Th2 like cytokines). It was found that cells of syphilitic patients were able to produce IL-2, IFN, TNF, IL-10 and weakly IL-6 already in primary seronegative syphilis. At the same stage of syphilis, but seropositive, ability of Th1 lymphocytes to produce cytokines reached the highest values, whereas the cells producing IL-10 lost this ability. The cells producing IL-6 and MIF had the highest ability in secondary early syphilis. In this stage of syphilis again slightly increased the ability of cells to secrete IL-10, which reached the highest value in early latent syphilis. The growing ability to produce IL-6 and IL-10 was accompanied with a diminished production of IL-2, IFN and TNF nearly in all stages of syphilis. Only MIF, in contrast to other cytokines, was produced in late syphilis without distinct changes. The greatest suppression of Th1 lymphocytes to produce cytokines and cells to secretion of MIF was found in early latent syphilis when the level of IL-10 in cell culture supernates was the highest. High ability of Th2 lymphocytes to cytokines secretion in late syphilis and low ability of Th1 ones, which are very important for cell-mediated immune response, may be the reason for facilitating T. pallidum multiplication and development of latent stages of disease despite presence of immunologically competent cells.

 

Treatment of psoriatic arthritis with antitumour necrosis factor-alpha antibody clears skin lesions of psoriasis resistant to treatment with methotrexate.
Ogilvie AL et al. Br J Dermatol. 2001 Mar;144(3):587-9. PMID: 11260020

BACKGROUND: In inflamed skin, keratinocytes and inflammatory cells both produce large amounts of tumour necrosis factor (TNF) -alpha, a cytokine with broad effects that are relevant to inflammation. Blockade of this proinflammatory cytokine by a monoclonal anti-TNF-alpha antibody might be effectively used in the treatment of inflammatory skin diseases. OBJECTIVES: To gather information about the efficacy of an anti-TNF-alpha antibody (infliximab) in the treatment of skin lesions of psoriatic arthritis. METHODS: Six patients with progressive joint disease and psoriatic skin lesions unresponsive to methotrexate therapy were treated with anti-TNF-alpha antibody. The Psoriasis Area and Severity Index was determined before and 10 weeks after initiation of therapy. RESULTS: Improvement of psoriatic skin lesions was observed in all patients. In addition, a marked improvement of the joint disease was noted. CONCLUSIONS: Therapy with anti-TNF-alpha antibody may be an effective treatment regimen for both psoriatic arthritis and psoriatic skin lesions.

 

Tumor necrosis factor-alpha (TNF alpha) in cerebrospinal fluid from patients with meningitis of different etiologies: high levels of TNF alpha indicate bacterial meningitis.
Glimaker M et al. J Infect Dis 1993 Apr; 167(4): 882-9. PMID: 8450254         

The levels of tumor necrosis factor (TNF)-alpha in cerebrospinal fluid (CSF) were analyzed in 139 patients with meningitis and in 20 control subjects. Elevated concentrations were observed in 42 (82%) of 51 patients with purulent bacterial meningitis (18/24 Haemophilus influenzae, 13/14 Streptococcus pneumoniae, 7/7 Neisseria meningitidis, and 4/6 with other purulent bacterial etiology). In contrast, elevated levels were found in only 5 of 78 individuals with nonbacterial meningitis (2/8 with herpes simplex type 2, 3/3 with varicella-zoster virus). Thus, the positive and negative predictive values were 0.89 for indicating a purulent bacterial meningitis. Raised CSF TNF alpha levels were observed in 7 of 8 patients with purulent bacterial meningitis in whom the routinely used parameters did not unequivocally indicate the diagnosis. Moderately increased levels were seen in 5 of 6 patients with Mycobacterium tuberculosis meningitis and in 1 of 4 cases of Borrelia burgdorferi. Thus, the present study indicates that concentrations of TNF alpha in CSF usually can discriminate between purulent bacterial and nonbacterial meningitis. These findings may contribute diagnostic guidance with routinely used CSF parameters.

 

 

Interestingly both arsenium and selenium also interact with NF-kB and its respective inhibitor – can that perhaps explain the since ancient times empirically demonstrated effect of these – in higher dose very toxic minerals - is there a need for giving these as nutrition supplements in small doses in order to enhance immune function ?

 

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=search&term=NF-kappaB+AND+Arsen*

http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=search&term=NF-kappaB+AND+Selen*

 

Arsenic suppresses necrosis induced by selenite in human leukemia HL-60 cells.
Zeng H. Biol Trace Elem Res 2001 Oct;83(1):1-15.  PMID: 11693998

Selenium, an essential trace element for humans, has been shown to have anticancer effects. Arsenic, a possibly essential ultratrace element for humans, has been used in the treatment of leukemia. Anticancer effects of selenium and arsenic have been related to their ability to induce apoptosis. Because humans are exposed to diverse trace elements simultaneously, it is important to learn their interrelationship. In this study, we demonstrate that sodium selenite (Na2SeO3) causes apoptosis at 3 microM and necrosis at high concentrations (> 3 microM) in HL-60 cells. Similarly, both sodium arsenite (NaAsO2) at 50 microM and sodium arsenate (Na2HAsO4) induce apoptosis at 500 microM and necrosis at higher concentrations (> 50 microM and > 500 microM, respectively) in HL-60 cells. Arsenite/arsenate, but not selenite, enhances AP-1 DNA-binding activity. This finding indicates different mechanisms through which apoptosis is induced by these two elements. Interestingly, we observed that HL-60 cell necrosis induced by a high concentration (> 3 microM) of selenite was essentially inhibited by arsenic (50 microM of NaAsO2 or 500 microM of Na2HAsO4), which resulted in a net effect of apoptosis. Because AP-1 DNA-binding activity was not induced in the presence of a combination of necrotic amount of selenite and apoptotic amount of arsenite/arsenate, the observed apoptosis apparently was through the mechanism used by selenite. Our results suggest, for the first time, that the toxic necrotic effect of selenite can be neutralized by arsenite/arsenate at the cellular level.

 

A previously commonly used ‘tonikum’ used in Denmark to treat ‘chronic fatigue’ after infection used to contain a small doses of arsenic, which was removed when the toxicity of arsenic was described, then the ‘tonikum’ lost its effect and went out of use probably because of it no longer had the same beneficial effect ? (according to an old pharmacist).

In animals arsenic was still used now and then as late as in the 1970’ies, because it was known to be able to raise a lame animal for a while, so it could be sold by the horse-trader as seemingly healthy and is still used sometimes in other countries, for instance by Tarello (according to veterinarians).

 

Until about 1950 – when penicillin arrived on the scene – arsenic was commonly used for treating  infections – including spirochetoses like syphilis (and borreliosis). Arsenic was used for and had very good effect on a patient described by Garin & Bujadoux. Paralysie par les Tiques. J Med Lyon 1922;71:765-767  – a case which in retrospect expressed almost all the most characteristic features of what is today called Lyme borreliosis - Abbreviated case-story translated by M. Kroun  from French – pardon me in advance for any translation errors, my french could certainly be better:

 

June 14, 1922: JMB, 58 years old, got a tickbite by Ixodes hexagonus on his left buttock. He knew the tick well from bites on his dog and sheep, and removed it without damaging its head. Three weeks later, he was hit by a very rapidly developing, painful disease, with alarming stabbing pains.

After this, irradiating pain developed in his left ischiadicus and he consulted dr. HINGLAIS, who saw a red inflamed area on the left buttock, with the tickbite in its center, the size of a 5 Franc coin. It was red, warm, painful and with palpable inguinal lymphglands on the same side. Later the rash increased in size into a wide, confluent one, without vesicles, which ultimately covered his left and right buttock, hypochondriac area, lower part of truncus and left leg down to the knee.

No fever. Urine normal. In the same area as the rash, the diseased suffered from severe pain, including a belt around the thorax and in right plexus brachialis area with irradiating pain down to the elbow. He suffered terribly, without any relief from 'antineurotique' medicine and also morphica had no or only transient effect.

In July and August, i.e. during 2 1/2 month, he suffered tremendously.

On Sep. 19, he traveled by automobile to dr. GARIN, who noted the following:

Lungs and heart normal. Puls 96. Temp. 37.5. Liver not palpable. Leftsided neck adenitis, as a chain of small glands along m. sternocleidomestoideus, but none in axillae, genitalia or inguinal regions. Nervous systeme: paresis of right m. deltoideus with atrophy, it was impossible for him to lift his arm, otherwise normal movements. No objective sensory disturbances, but the patient describe 2 very painful spots at the basis of thorax and belt-formed pain corresponding to the 2 lowest intercostal nerves, causing breathing difficulty. These pains were alleviated after the first injection of novarsenobezol. No trophic disturbances on the lower extremities. Normal tendon reflexes. Normal pupil-reaction. The sick man is very disturbed, having chills and being restless.

Oct. 3. Same findings, but when asked to sit up, a slight positive Kernig.

Lumbarpuncture the same day reveals, spinal fluid under pressure, glucose normal, increased albumin, 75 WBCs mostly polymorphonuclear cells. No microbes, no spirochetes visible. Wassermann reaction slightly positive.

Oct. 17. The symptoms have improved and the patient is discharged, but he still have reduced movement in his right arm and atrophy of the deltoideus muscle and he still suffers some pains at the basis of the thorax.

Treatment: from Sep. 26 - Oct. 16 he was given 4 injections of neo-billon, 0.10, 0.30, 0.45, 0.60 cgr. - the last three times preceded by cyan Hg. (Mercury)

Notably the most severe pain was relieved after the first injection of novarsenobenzol.

Seen in the outpatient clinic on Oct. 30 and Nov. 8, where he does not suffer much, but still have reduced movement of the right arm.

Authors discussion:

This case seem to be a case of 'tick paralysis'. The Wassermann reaction was slightly positive, but that is sometimes the case in other tick-borne diseases like Rocky Mountain Spotted fever and relapsing fever. On the other hand, does this man not have any sign of syphilis.

In experiments, HAWDEN, in Columbia, have observed:

It is possible to infect lambs and pheasants via a tickbite. The illness shows about 6-7 days after the bite.

It is not possible to reproduce the illness via injection of blood from a sick into an animal.

He did not find the pathologic agent.

Regarding the etiology, the english authors suspects that an infectious agent is transmitted by the tick, or a toxin is being inoculated.

We also admit voluntarily that it must be an agent ('virus') transported by the tick.

This hypothesis is supported by the adminstration of novarsenobenzol to our patient, that was very effective in alleviating the severe pain.

Our observation differs from others in the english literature, that our patient had a rash after the tickbite, and it also differs regarding the tick that was responsible for causing this disease, in our case it was an Ixodes hexagonus tick.

We propose to collect ticks from the patients village and try to produce the illness in an animal via tickbite.

 

Arsenite inhibit NF-kB activity by stable expression of a kinase mutated form of IKKb (inhibitor of KappaB):

 

Opposite effect of NF-kB and c-Jun-N-terminal kinase on p53-independent GADD45 induction by arsenite.

Chen F et al. J Biol Chem 2001 Jan 9. PMID: 11150309  PDF

Cell cycle checkpoint, a major genomic surveillance mechanism, is an important step in maintaining genomic stability and integrity in response to environmental stresses. Using cells derived from human bronchial epithelial cells, we demonstrate that NF-kB and c-Jun-N-terminal kinase (JNK) reciprocally regulated arsenic trioxide (arsenite)-induced, p53 independent expression of GADD45 protein, a cell cycle checkpoint protein that arrests cells at G2/M phase transition. Inhibition of NF-kB activation by stable expression of a kinase mutated form of IKKb (IKKb-KM) caused increased and prolonged induction of GADD45 by arsenite. In contrast, the induction of GADD45 by arsenite was transient and less potent in cells where the NF-kB activation pathway was normal. Analysis of cell cycle profile by flow cytometry indicated that NF-kB inhibition potentiates arsenite-induced G2/M cell cycle arrest. Abrogation of JNK activation, on the other hand, decreased GADD45 expression induced by arsenite, suggesting a role for JNK activation in GADD45 induction. These results may indicate a molecular mechanism that NF-kB and JNK differentially contribute to cell cycle regulation in response to arsenite.

 

Arsenic-interferon-alpha-triggered apoptosis in HTLV-I transformed cells is associated with tax down-regulation and reversal of NF-kappa B activation.

El-Sabban ME et al. Blood. 2000 Oct 15;96(8):2849-55.  PMID: 11023521   PDF

Human T-cell lymphotropic virus type I (HTLV-I)-associated adult T-cell leukemia/lymphoma (ATL) is a malignancy of mature activated T cells resistant to conventional chemotherapy. The viral transactivator protein Tax plays a critical role in HTLV-I-induced transformation and apoptosis resistance by inducing I kappa B-alpha degradation, resulting in the activation of the NF-kappa Bpathway. In these HTLV-I-transformed cells, arsenic trioxide (As) and interferon (IFN)-alpha synergize to induce cell cycle arrest and apoptosis. We demonstrate that cell death induction is only partly dependent upon caspase activation and is not associated with modulation of bcl-2, bax, or p53 expression. However, combined As and IFN induce the degradation of Tax, associated with an up-regulation of I kappa B-alpha resulting in a sharp decrease in RelA DNA binding nuclear factor (NF)-kappa B complexes because of the cytoplasmic retention of RelA. Taken the role of Tax in HTLV-I-induced transformation, its down-regulation probably accounts for cell death induction through inactivation of the NF-kappa B pathway. Such specific targeting of the viral oncoprotein by As-IFN treatment, reminiscent of As targeting of promyelocytic leukemia/retinoic acid receptor-alpha in acute promyelocytic leukemia, provides strong rational for combined As-IFN therapy in ATL patients. (Blood. 2000;96:2849-2855)

 

AIDS and the "Selenium-CD4 T Cell Tailspin" The Geography of a Pandemic.
Foster HD . Townsend Letter for Doctors & Patients - december 2000: 94-99

Abstract: AIDS involves slow but relentless declines in serum selenium and CD4 T-cells, both of which are independent predictors of mortality. This relationship between selenium and the immune system is one of positive feedback, in which a decline in either of these two variables causes further depression of the other. Termed the "selenium-CD4 T Cell Tailspin" by the author, this feedback loop plays a crucial role in both the diffusion of HIV-1 and the associated development of AIDS. Manipulating this tailspin opens new avenues for both prevention and treatment.

 

 

Review not finished ….

Marie Kroun

april 2003

……………………….

Journal of the International Society for Orthomolecular Medicine

http://www.healthy.net/library/journals/ortho/index.html