You are on page 1of 3

Tumor Necrosis Factor

F Atzeni, Queen Mary University of London, London, UK; Rheumatology Unit, L.Sacco University Hospital, Milan, Italy

P Sarzi-Puttini, Rheumatology Unit, L.Sacco University Hospital, Milan, Italy

© 2013 Elsevier Inc. All rights reserved.

This article is a revision of the previous edition article by TH Rabbitts, volume 4, p 2081, © 2001, Elsevier Inc.

Glossary Osteoclasts and osteoblasts Cells involved in bone


Atherosclerosis and heart failure Cardiovascular remodeling.
involvement. Rheumatoid arthritis Autoimmune rheumatic disease.
Bone damage Bone alterations and resorption. Tumor necrosis factor Proinflammatory cytokine.

Introduction The Role of TNF in the Pathogenesis of RA and Bone


Damage
Tumor necrosis factor (TNF), a 17 kDa protein consisting of
157 amino acids, is a homotrimer in solution, and its bioactiv­
Pathogenesis of RA
ity is mainly regulated by soluble TNFα-binding receptors. In Proinflammatory cytokines such as TNF and interleukin (IL)-1β
humans, the gene maps to chromosome 6. play a key role in the pathogenesis of RA. The synovial mem­
TNF is mainly produced by activated macrophages, T lym­ brane of RA patients is hyperplastic, highly vascularized, and
phocytes, and natural killer (NK) cells, although it is also infiltrated by inflammatory cells. CD4+ T lymphocytes play an
expressed at lower levels by various other cells, including fibro­ important role because, when activated by an antigen, they
blasts, smooth muscle cells, and tumor cells. Cellular TNF is stimulate monocytes, macrophages, and synovial fibroblasts
synthesized as pro-TNF (26 kDa), which is bound to the mem­ to produce IL-1, IL-6, and TNF; they also secrete matrix metal­
brane and released upon the cleavage of its pro-domain by the loproteinases (MMPs) as a result of cell-surface signaling by
TNF-converting enzyme (TACE). TNFα acts via two distinct means of CD69 and CD118, and by means of the release of
receptors (TNFR-1 and TNFR-2) but, although its affinity for soluble mediators such as interferon (IFN)-γ, IL-1, IL-6, IL-17,
TNFR-2 is 5 times higher than that for TNFR-1, the latter and TNF. Activated CD4+ T cells stimulate B cells by means of
initiates most of its biological activities. TNFR-1 (p55) is cell–cell contacts, and bind α sub 1 and β sub 2 integrin, CD 40
expressed on all cell types, whereas the expression of TNFR-2 ligand, and CD28 to produce immunoglobulins (Ig’s), includ­
(p75) is mainly confined to immune cells. The main difference ing rheumatoid factor (RF). They also express the receptor
between the two receptors is that TNFR-2 does not have the activator of NF-κB (RANK), which stimulates osteoclastogen­
death domain (DD) possessed by TNFR-1. TNFR-1 plays a dual esis via the RANK ligand (RANKL). Activated macrophages,
role: it is not only involved in inducing apoptosis, but can also lymphocytes, and fibroblasts can also stimulate angiogenesis,
transduce cell survival signals. The signaling pathways are well which is responsible for the synovial hypervascularity of RA
defined, but the regulation of life/death signaling is still poorly patients. Synovial endothelial cells are activated and express
understood. adhesion molecules that promote the recruitment of inflam­
TNF is involved in a large number of pathological and matory cells. Finally, TNF induces the production of various
physiological pathways: cytokines and stimulates fibroblasts to express adhesion mole­
cules that interact with the ligands on the surface of leukocytes
1. Low levels may help maintain homeostasis by regulating and increase the number of inflammatory cells. TNF has an
the body’s circadian rhythm, and promote the remodeling immunostimulatory role that can alter the balance of regula­
or replacement of injured and senescent tissue by stimu­ tory T cells. RA patients have high TNF levels in their synovial
lating fibroblast growth. fluid, which are localized to the junction between the inflam­
2. It plays a pivotal role in the pathogenesis of rheumatic matory pannus and the healthy cartilage, and associated with
diseases such as rheumatoid arthritis (RA), Crohn’s dis­ bone erosions.
ease (CD), psoriatic arthritis (PsA), and ankylosing
spondylitis (AS).
3. It is involved in bone damage and resorption.
Bone Damage
4. It is involved in host defense and tumor surveillance. Animal studies have provided key evidence that antagonizing
5. It plays a role in heart failure, and promotes the dyslipi­ TNF is a viable therapeutic strategy, and subsequent studies of
demia and insulin resistance involved in initiating the various anti-TNF blockers have shown that blocking TNF
atherosclerosis that leads to plaque rupture and acute improves the symptoms of RA patients and arrests bone
myocardial infarction (AMI). damage by affecting bone remodeling.

Brenner’s Encyclopedia of Genetics, 2nd edition, Volume 7 doi:10.1016/B978-0-12-374984-0.01594-1 229


230 Tumor Necrosis Factor

It regulates bone marrow levels of osteoclast precursors administration of anti-TNF agents can lead to their regression
directly by upregulating c-fms expression, and activates osteo­ and enable the reactivation of latent TB. However, such treat­
clasts by enhancing RANK signaling mechanisms. The RANKL ment is only one of many risk factors that predispose to the
pathway regulates osteoclast differentiation and function dur­ development of TB.
ing physiological bone remodeling, and RANKL promotes Finally, TNF is involved in leukocyte trafficking and the
osteoclastogenesis by binding to its cognate receptor on osteo­ clearance of immune complex (IC). Bacterial infections have
clast precursor cells. Osteoprotegerin (OPG) is a soluble decoy always been the most common infections in our immunocom­
RANKL receptor that blocks RANKL’s pro-osteoclastogenic promised patients receiving anti-TNF therapy.
activity. The RANK/RANKL pathway is also activated in patients
with inflammatory arthritis, thus leading to dysregulated bone
remodeling. The RANKL:OPG messenger RNA (mRNA) expres­
Tumor Surveillance
sion ratio is increased in RA synovial tissue, which indicates The exact relationship between TNF and cancer is unclear:
that pro-osteoclastogenic conditions dominate within the joint high-dose TNF has been used to treat some malignancies,
microenvironment. including melanoma and sarcoma, but it has also been sug­
TNF indirectly influences osteoclastogenesis by inducing gested that cancer-related cachexia could be treated using
the expression of RANKL and macrophage colony-stimulating anti-TNF agents, which indicates that TNF may play different
factor (M-CSF) in bone marrow stromal cells of osteoblast roles at different concentrations in the different stages of carci­
lineage. nogenesis. Similarly, anti-TNF therapy may have a spectrum of
TNF is considered an ‘upstream’ dominant proinflamma­ effects on de novo malignancies, in situ carcinomas (CIS), and
tory cytokine because anti-TNF antibodies significantly reduce pre-existing tumors, and so, given hypothetically increased risk
the IL-1 production of cultured synovial cells taken from of new or recurrent malignancies in already affected patients,
patients with RA. It also affects osteoblasts by inhibiting their researchers initially excluded them from randomized clinical
differentiation and maturation, thus leading to a decrease in trials of anti-TNF therapy for RA. However, since then, a num­
alkaline phosphatase and osteocalcin expression. Mice and ber of studies have shown that anti-TNF therapy does not
wild-type mice treated with exogenous TNF show increased increase the risk of incident malignancy in RA patients,
numbers of CD11b+ osteoclast precursor cells, an effect that although further investigations are still necessary to confirm
is independent of RANKL. In human disease, data from clinical these findings.
trials confirm that inhibiting TNF activity in RA patients effec­
tively protects against bone erosion. Although this is largely
due to the anti-inflammatory effects of TNF inhibition, other
The Role of TNF in Heart Failure and Atherosclerosis
possible mechanisms by which TNF inhibition decreases struc­
tural damage include a direct reduction of osteoclast-mediated Heart Failure
bone loss and an increase in osteoblast-mediated bone
The expression of TNF is highly tissue-specific. Under normal
formation.
conditions, it is expressed in the spleen, liver, thymus, and
kidney but, following various types of stimulation (e.g., bacter­
ial sepsis), it can be expressed at extremely high levels. It has
The Role of TNF in Host Defense and Tumor Surveillance also been shown that myocardial stress, and pressure and
Host Defense volume overload, leads to large quantities of TNF in the
heart. The mechanisms by which TNF mediates cardiac injury
TNF is released by activated macrophages, T lymphocytes, and are not entirely clear, but it has been demonstrated that circu­
other immune cells in response to various infectious stimuli and, lating TNF levels inversely correlate with peak blood flow in
among its other effects, it has antitumoral and antiviral activity, patients with heart failure, regardless of their age, ejection frac­
mediates systemic inflammatory responses to infection and sep­ tion, or peak oxygen consumption. This suggests that TNF may
sis, and plays a crucial role in host responses to a number of contribute to peripheral skeletal muscle weakness/fatigue in
infections, especially those caused by Mycobacterium tuberculosis such patients. With these theories in mind, a number of clinical
and other intracellular pathogens. trials have studied the effects of TNFα inhibition in patients
TNF is essential for controlling intracellular pathogens as it with heart failure. However, the published data showed that
stimulates inflammatory cell recruitment to the infectious area, anti-TNF agents offer no benefit for various reasons, including
and the formation and maintenance of the granulomas that the fact that they have untoward effects in the setting of heart
physically contain the infection. It acts as part of the immune failure and the possibility that the biological agents used in the
response to M. tuberculosis infection (TB) in conjunction with trials were intrinsically toxic.
IFN-γ by inducing macrophage activation, enhancing immune
cell recruitment, and increasing the chemokine expression of
macrophages by activating the NF-κB signaling pathway. TNF
Atherosclerosis
also mediates cell death by inducing the caspase-mediated
apoptotic pathway. TNF promotes dyslipidemia and insulin resistance (both of
These roles have been elucidated by murine models. In which are traditional risk factors for atherosclerosis), upregu­
TNF-deficient mice, inflammatory cell recruitment is delayed lates adhesion molecules, which leads to the formation of fatty
and, even after their recruitment, the leukocytes fail to form streaks and the beginning of atherosclerosis, and is involved in
the organized granulomas necessary to contain infection. the inflammation that leads to plaque rupture, which is the
Furthermore, once the granulomas have formed, the most frequent cause of acute myocardial infarct (AMI).
Tumor Necrosis Factor 231

Inhibiting TNF in patients with RA may therefore reduce MI Desai SB and Furst DE (2006) Problems encountered during anti-tumor necrosis factor
rates by inhibiting one or more of these mechanisms, particu­ therapy. Best Practice & Research: Clinical Rheumatology 20: 759–760.
Dixon WG, Hyrich KL, Watson KD, et al. BSRBR Control Centre Consortium, Symmons
larly in those with a good clinical response.
DP, BSR Biologics Register (2010) Drug-specific risk of tuberculosis in patients with
TNF may also promote thrombophilia by encouraging rheumatoid arthritis treated with anti-TNF therapy: Results from the British Society
thrombotic events. for Rheumatology Biologics Register (BSRBR). Annals of the Rheumatic Diseases
69: 522–528.
Dixon WG, Watson K, Lunt K, et al. British Society for Rheumatology Biologics Register
Control Centre Consortium, Silman AJ, Symmons DPM (2006) Rates of serious
Conclusions infection, including site-specific and bacterial intracellular infection, in rheumatoid
arthritis patients receiving anti-tumor necrosis factor therapy. Arthritis &
TNF is involved in various pathological and physiological path­ Rheumatism 54: 2368–2376.
ways. The use of anti-TNF drugs to treat rheumatic diseases has Ferraccioli G and Gremese E (2004) Autoantibodies and thrombophilia in RA: TNF and
TNFa blockers. Annals of the Rheumatic Diseases 63: 613–615.
confirmed the relevance of this cytokine to their pathogenesis,
Gremese E and Ferraccioli G (2011) The metabolic syndrome: The crossroads between
as well as to host defenses and cancer control. However, further rheumatoid arthritis and cardiovascular risk. Autoimmunity Reviews 10: 582–589.
studies are required to improve our understanding of its role Grivennikov SI and Karin M (2011) Inflammatory cytokines in cancer: Tumour necrosis
and to answer some still open questions. factor and interleukin 6 take the stage. Annals of the Rheumatic Diseases 70
(supplement 1): i104–i108.
Karmakar S, Kay J, and Gravallese EM (2010) Bone damage in rheumatoid arthritis:
See also: Autoimmune Diseases; Cytokinesis; Immunological Mechanistic insights and approaches to prevention. Rheumatic Disease Clinics of
North America 36: 385–404.
Tolerance; Psoriasis.
Nurmohamed MT and Kitas G (2011) Cardiovascular risk in rheumatoid arthritis and
diabetes: How does it compare and when does it start? Annals of the Rheumatic
Diseases 70: 881–883.
Sarzi-Puttini P, Atzeni F, Gerli R, et al. (2010) Cardiac involvement in systemic rheumatic
Further Reading diseases: An update. Autoimmunity Reviews 9: 849–852.
Sarzi-Puttini P, Atzeni F, Shoenfeld Y, and Ferraccioli G (2005) TNF-alpha, rheumatoid
Choy EHS and Panayi GS (2001) Cytokine pathways and joint inflammation in arthritis, and heart failure: A rheumatological dilemma. Autoimmunity Reviews 4(3):
rheumatoid arthritis. The New England Journal of Medicine 344: 907–916. 153–161. Review.
Crum NF, Lederman ER, and Wallace MR (2005) Infections associated with tumor Scott DL and Kingsley GH (2006) Tumor necrosis factor inhibitors for rheumatoid
necrosis factor-α antagonists. Medicine (Baltimore) 84: 291–302. arthritis. The New England Journal of Medicine 335: 704–712.

You might also like