ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 5, pp. 853-861 © Pleiades Publishing, Ltd., 2024.
853
MINI-REVIEW
Reverse Genetics Applied to Immunobiology
of Tumor Necrosis Factor, a Multifunctional Cytokine
Sergey A. Nedospasov
1,2,a
*, Andrei A. Kruglov
3
, Alexei V. Tumanov
4
,
Marina S. Drutskaya
1,2
, Irina V. Astrakhantseva
1
, and Dmitry V. Kuprash
2
1
Division of Immunobiology and Biomedicine, Sirius University of Science and Technology,
354340 Federal Territory Sirius, Russia
2
Center for Precision Genome Editing and Genetic Technologies for Biomedicine,
Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
3
Laboratory of Systems Rheumatology, German Rheumatism Research Center (DRFZ), a Leibniz Institute,
10117 Berlin, Germany
4
Department of Microbiology, Immunology and Molecular Genetics,
University of Texas Health Science Center at San Antonio, 79229 San Antonio, TX, USA
a
e-mail: sergei.nedospasov@gmail.com
Received November 21, 2023
Revised December 28, 2023
Accepted February 19, 2024
AbstractTumor necrosis factor (TNF) is one of many cytokines– protein molecules responsible for communi-
cation between the cells of immune system. TNF was discovered and given its grand name because of its striking
antitumor effects in experimental systems, but its main physiological functions in the context of whole organism
turned out to be completely unrelated to protection against tumors. This short review discusses “man-made”
mouse models generated by early genome-editing technologies, which enabled us to establish true functions
ofTNF in health and certain diseases as well as to unravel potential strategies for improving therapy of TNF-de-
pendent diseases.
DOI: 10.1134/S0006297924050067
Keywords: tumor necrosis factor, TNF, conditional mice models, gene knockout, reporter mice, gene overexpression
Abbreviations: ESCs, embryonic stem cells; LT,lymphotoxin; TNF,tumor necrosis factor; UTR,untranslated region.
* To whom correspondence should be addressed.
INTRODUCTION
Almost 50 years ago, Lloyd J. Old and his col-
leagues reported an interesting experiment performed
to study the antitumor effect of bacterial endotoxin
and other substances, which later would be known
as activators of innate immune receptors [1]. As it
was shown, combination of the infection with BCG
(Tb vaccine mycobacterial strain) and subsequent in-
jection of lipopolysaccharide (LPS) from Escherichia
coli leads to the appearance in blood serum of an un-
known “factor” with marked antitumor activity, which
was referred to as Tumor Necrosis Factor (TNF) [2].
This discovery was perceived as further development
of the ideas of William B. Coley, who pointed out the
possibility of using live bacteria or bacterial lysates in
the therapy for some tumors as early as at the begin-
ning of the 20th century [3]. Further studies showed
that TNF had a protein nature and could be produced
by the cells of immune system in response to vari-
ous stimuli, e.g., by myeloid cells in response to LPS.
Molecular cloning and heterologous expression of the
human and mouse Tnf genes made it possible to relate
antitumor activity to a single protein that could be de-
fined as a “cytokine”, i.e., the rapidly increasing super-
family of protein mediators of cell communications.
Administration of the recombinant TNF produced in
E.  coli to mice fully reproduced antitumor activity of
the native TNF[4], and its high doses resulted in man-
ifestation of toxicity associated with anacute inflam-
NEDOSPASOV et al.854
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
matory response in mice (toxicity was not observed in
the L. Old’s experiments [2], probably because it was
impossible to achieve the same high concentrations as
in the experiments with recombinantTNF).
TNF has a molecular weight of 17  kDa; however,
in order to perform its physiological (including anti-
tumor) functions, it has to form a homotrimer, which
is a high-affinity ligand of two different receptors:
TNFR1(p55) and TNFR2(p75)[5]. These receptors dif-
fer both in the type of transmitted intracellular signal
and in the patterns of tissue-specific expression.
Following the molecular description of TNF and
its receptors in the 1980s, other members of the large
families of TNF-like cytokines (most of which are
membrane-bound molecules) and TNFR-like receptors
were soon discovered; they are able to transmit sev-
eral types of intracellular signals, thereby determin-
ing biological activity of the ligands of this family[6].
There were hundreds of papers describing TNF activi-
ty in different situations in  vitro and in vivo; however,
physiological function of TNF in the context of a whole
organism was established in 1990s mainly through
two strategies of reverse genetics: creating mice with
the Tnf gene knockout and generating transgenic mice
with its overexpression. Moreover, several interesting
mouse lines were isolated by the methods of direct ge-
netics. Below we will discuss numerous mouse models
that have been independently created and character-
ized in various laboratories, as well as the so-called
“reporter” mice, which represent additional tools for
studying immunobiology and physiology of this inter-
esting cytokine.
MICE WITH COMPLETE, PARTIAL
OR REGULATED GENETIC DEFICIENCY OF TNF
The knockout techniques that appeared before the
era of genomic nucleases and became widespread in
the 1980s were based on targeting the genes in mouse
embryonic stem cell lines (ESCs) (for other animals,
such lines were created much later). Due to the low ef-
ficiency of this procedure in the mouse ESCs it became
necessary to introduce antibiotic resistance markers to
select knockout clones (usually neomycin-resistance);
as a result, the expression cassette containing the re-
sistance gene (the so-called “neo-cassette”) remained
forever in the structure of the targeted locus and could
influence the activity of the nearby genes. Noteworthy,
it took 10 years from the cloning of the mouse Tnf gene
to the creation of the first Tnf knockout mice. At least
four laboratories obtained such mice almost simulta-
neously and independently (Table  1), but the palm of
victory obviously went to the laboratory of G. Kollias
in Greece. When creating the Tnf knockout mice, one
had to take into consideration two significant peculiar-
ities of this gene. Firstly, the Tnf gene is located very
close to the related lymphotoxin genes Ltα and Ltβ[7];
therefore, combined Tnf/Lt knockouts cannot be ob-
tained by crossbreeding of mice with single knockouts.
Secondly, the Tnf/Lt locus is within the major histo-
compatibility complex (MHC)[7]; hence, it is very diffi-
cult to obtain congenic mouse lines with modifications
of the Tnf gene and the required alleles of the MHC
genes, so that the knockout mice should be designed
with particular reference to the genetic basis of ESCs
and mouse lines used for subsequent backcrossing.
Pasparakis et al. [8] described new TNF functions
associated with its role in the structural and func-
tional organization of lymphoid tissues, about which
the discoverers of TNF had no idea. It should be not-
ed that the related cytokine, LTα, was considered for
10years to be a functional analog of TNF, produced by
lymphoid rather than by myeloid cells. This view on
lymphotoxin was supported by the fact that the re-
combinant protein demonstrated an antitumor activity
similar to that of TNF in the model of transplantable
sarcoma in mice, as well as in cell cultures sensitive
to cytotoxic effects of TNF [20]. These results indicat-
ed potential redundancy in TNF and LTα functions, in-
spiring creation of a double knockout of both genes.
Table  1 presents some mouse lines with such double
knockouts. Note that earlier the lymphotoxin gene
knockout produced a sensational phenotype [21]: in
mice peripheral lymphoid organs were completely
absent, with the exception of spleen. Further compar-
ison of the phenotypes in the knockout mice made it
possible to differentiate between the functions of TNF
and lymphotoxin, e.g., in the case of organogenesis of
Peyers patches[22], which may be explained by signal
transduction from the membrane lymphotoxin com-
plex L
1
/L
2
[23] through LTβR [24], while a minor
overlap in functions is due to the fact that the soluble
lymphotoxin(L
3
) is able to trigger the signaling cas-
cade in  vivo through TNFR1 and TNFR2[17].
The technique of conditional (tissue-specific or in-
ducible) genetic recombination developed in the early
1990s in mammalian cell culture[25] and then in the
cells of transgenic[26,27] and knockout [28-30] mice
was based on LoxP/Cre recombination system of bacte-
riophage  P1, has truly revolutionized biological stud-
ies. It became possible, firstly, to circumvent the prob-
lem of embryonic lethality typical for a great number
of genes, even those with unknown non-redundant
functions in embryogenesis, and, secondly, to relate
particular functions of the genes and their products to
particular types of producer cells. This strategy proved
to be especially fruitful for studying the genes whose
products manifested pleiotropic functions, which is
typical for cytokines and other regulatory molecules.
Development of a panel of conditional mouse
lines for the Tnf gene began almost 20 years ago[14],
REVERSE GENETICS APPLIED TO TNF IMMUNOBIOLOGY 855
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Table 1. Mouse models with TNF deficiency
Genotype of the Tnf/Lt locus Description References
Mice with the complete TNF gene knockout generated by reverse genetics
Tnf
–/–
complete knockout created by targeting in ESCs of the EK.CCE line
of mice of the 129/Sv lineage, with one backcrossing at C57BL/6
[8]
Tnf
–/–
complete knockout created by targeting in ESCs of the W9.5 line
from mice of the 129/Sv line, with one backcrossing at C57BL/6
[9]
Tnf
–/–
complete knockout created by targeting in ESCs from the hybrid
of the first generation of C57BL/6 and CBA mice, with selection
of the Tnf gene knockout in the C57BL/6 haplotype and 5 rounds
of backcrossing at C57BL/6
[10,11]
Tnf
–/–
complete knockout created by targeting in ESCs from the BL/6-III line
on the genetic basis of C57BL/6
[12]
Tnf
–/–
derived from the mouse line created for conditional targeting of TNF
on the genetic basis of 129/Sv, with multiple backcrossing atC57BL/6;
the targeted locus does not contain neo-cassettes
[13,14]
Mouse models with combined TNF/LT knockout
Tnf
–/–
Lta
–/–
(TNF/LTα double KO)
complete knockout of the Lta and Tnf genes created by targeting
in ESCs of the BL/6-III line on the genetic basis of C57BL/6
[12]
Tnf
–/–
Lta
–/–
(TNF/LTα double KO)
complete knockout of the Lta and Tnf genes created by targeting
in ESCs from the mouse129/Sv line, with one backcrossing atC57BL/6
[15]
Tnf
–/–
Lta
–/–
(TNF/LTα doubleKO)
complete knockout of the Lta and Tnf genes created by targeting
in ESCs from the mouse 129/Sv line, with one backcrossing at C57BL/6
[16]
Tnf
–/–
Ltb
–/–
(TNF/LTβdoubleKO)
complete knockout of the Ltb and Tnf genes created by targeting in ESCs
from the mouse 129/Sv line, with multiple backcrossing at C57BL/6
[17]
Tnf
–/–
Lta
–/–
Ltb
–/–
(TNF/LTα/LTβtriple KO)
complete knockout of the Lta, Tn, and Ltb genes created by targeting in
ESCs from the mouse129/Sv line, with multiple backcrossing at C57BL/6
[18]
Hypomorphic allele of the Tnf gene selected with the help of direct genetics
Tnf
PanR1/PanR1
, Tnf
PanR1/+
in the random mutagenesis experiment using N-ethyl-N-nitrosourea,
a dominant-negative mutant with the P138T amino acid substitution
in the mature TNF protein was found; the mutation prevents TNF
binding to the receptor TNFR1
[19]
Basic “platform” for generating conditional deletions of the Tnf gene in mice
Tnf
flox/flox
using genetic knockout in ESCs from the mouse 129/Sv line,
with multiple backcrossing at C57BL/6, the Tnf gene is framed by LoxP
sites (“floxed”); the modification does not affect activity of the gene
but allows for its subsequent elimination by Cre recombinase
in particular types of cells and/or in inducible fashion
[14]
first for the major populations of immunocytes: my-
eloid cells and lymphocytes. Using these mouse mod-
els, it was shown that the particular homeostatic or
pathogenic functions of TNF are associated with the
particular type of cells producing this cytokine. For ex-
ample, myeloid cells (primarily macrophages and neu-
trophils) are necessary for protection against intra-
cellular infections and formation of granulomas but,
NEDOSPASOV et al.856
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
at the same time, proved to be the major sources of
systemic TNF in different pathological states, e.g.,
LPS-induced toxicity [14]. On the other hand, TNF
produced by T and B cells performs homeostatic func-
tions, including organization of lymphoid tissue [31].
Later, mice with the constitutive or induced deletion
of the Tnf gene in dendritic cells, monocytes, lympho-
cytes[32-34], basophiles, microglia, mast cells[35], ep-
ithelial cells[36, 37], smooth muscle cells[38,  39], etc.,
were created. Interestingly, in all cases there were
unique phenotypic traits associated with the TNF pro-
duction by a particular cellular source.
MOUSE MODELS
BASED ON TNF OVEREXPRESSION
AND/OR “HUMANIZATION”
The first and the best-known transgenic mouse
system utilized the mechanism of posttranscriptional
regulation of the Tnf gene [40], confirmed the hypoth-
esis of a relationship between the overexpression of
TNF and the development of arthritis [41] and be-
came a widespread preclinical model for studying TNF
blockers. It was created in the Kollias’s laboratory[42]
in 1991. This model was an important addition to the
clinical results obtained by Mainiet al. [43] and Feld-
mann et al. [44] and made it possible to substantiate
anti-TNF therapy as an innovative strategy for treating
rheumatoid arthritis. In the original work of Kollias’s
team[42], the number of transgene insertions in mice
was several dozens, which resulted in manifestation
of the TNF-dependent polyarthritis in all mice at the
age of several months. Therefore, the drawbacks of
this model included the early development of only one
type of the TNF-dependent diseases, preventing mod-
eling of other autoimmune diseases. Later, the same
genetic construct was used to create transgenic mice
with a smaller number of transgenic insertions, mak-
ing it possible to ameliorate the pathogenic phenotype
and to extend the range of applications of this preclini-
cal model [45](Table2).
Note that in the works by Keffer et al. [42] and
Hayward et al. [45] the human Tnf gene was overex-
pressed in mice; at the same time, the mouse Tnf gene
was not deleted and could be expressed, while both
TNF receptors remained murine. In essence, these
were partially “humanized” preclinical models allow-
ing treatment of the experimental TNF-mediated pa-
thologies with involvement of the human TNF blockers,
most of them (with the exception of etanercept) being
species-specific and not working against mouse TNF.
Table 2. Mice with TNF overexpression
Mouse model Description References
Tgl278
transgenic line with the human Tnf gene under control of a strong promoter,
with modification of 3′-untranslated region(UTR), which resulted in high
expression of human TNF and caused severe polyarthritis in mice;
preclinical model for studying the effects of TNF blockers invivo
[42]
B6.Cg-Tg(TNF)#Xen
the same but with moderate levels of human TNF expression;
improved preclinical model
[45]
BPSM1
the line with spontaneous insertion of retrotransposon
in the mouse 3′-UTR Tnf, impairing the previously unknown mechanism
of posttranscriptional regulation;
TNF overexpression in this mouse line leads not only to severe
polyarthritis but also to heart valve disease
[46]
TNF
del4
, TNF
del5
, TNF
del6
and their combinations
the panel of mouse lines with impairment of three regions
in the 3′-UTR of the mouse Tnf gene regulating mRNA stability;
it has been shown that impairment of this system may result
in the extremely high TNF production causing death of some embryos
[47]
TNF
ΔARE
mice with deletion of the AT-rich region in 3′-UTR
of the mouse Tnf gene by the LoxP/Cre technique;
phenotypically similar to the Tgl278 and BPSM1 line but, in addition
to arthritis, developed inflammation in the ileum of the small intestine
[48]
Transgenic mice
with the entire human
Tnf/ltα/ltβ-locus
all three genes are under control of the intrinsic promoters/enhancers;
there was a moderate overexpression of genes of the human Tnf/Ltα/Ltβ locus;
cortical atrophy of the thymus
[49]
REVERSE GENETICS APPLIED TO TNF IMMUNOBIOLOGY 857
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Table 3. Reporter mice for visualization of TNF expression
Genetic name of mouse model Description References
TNF-2A-Kat (B6.FRFPK+)
bicistronic expression of TNF and fluorescent protein Katushka
using the viral 2A peptide sequence
[58,59]
hTNF.LucBAC
recombination with the human TNF BAC clone, which resulted
in substitution of the luciferase gene for exon1 of the Tnf gene
[60]
FVB/N-Tg(CAG-EGFP,-Tnf)1Kul/J
mice with the “floxed” eGFP cassette,
which prevents transcription of the mouse Tnf cDNA;
TNF is expressed only after Cre-mediated recombination
[61]
The more advanced “humanized” model included mice
with the human Tnf gene inserted precisely at the site
of the mouse Tnf gene (genetic “knock-in”) and with
conservation of all regulatory elements [50]. These
mice did not demonstrate any abnormalities in the
development of lymphoid tissue microarchitecture; at
the same time, the human TNF performed protective
function in the case of intracellular infections, which
was indicative of normal in  vivo signaling through
the mouse TNFR1 receptor. On the other hand, the hu-
man TNF is unable to effectively activate the mouse
TNFR2[51], which was a disadvantage of this human-
ized model. For quite a number of experimental pa-
thologies (arthritis, acute hepatotoxicity, etc.) such lim-
itation of the model is not a problem, because most of
the biological effects of TNF are mediated by TNFR1.
Nevertheless, in some cases, TNFR2 may play a signif-
icant role; therefore, the mice with “humanization” of
both TNF and the extracellular domain of TNFR2 with
the possibility of conditional activation of TNFR2 were
developed to eliminate this limitation and to specify
the role of TNFR2 [52]. Transgenic mice expressing
Cre recombinase in FoxP3-positive cells were used to
demonstrate that signal transduction via TNFR2 in the
T-regulatory cells contributes to protection of the cen-
tral nervous system in the model of autoimmune pa-
thology[52]. One more interesting line contained the
modified Tnf gene, the product of which had certain
amino acid variations and, as a result, almost no sol-
uble TNF was formed[53]. The experiments with this
mouse line have demonstrated for the first time that
many TNF functions in vivo are mediated by the mem-
brane-bound but not soluble form.
Several mouse models with overexpression of the
endogenous mouse TNF due to impaired 3′-UTR re-
gions of the Tnf gene, which are responsible for mRNA
stability, were described(Table  2). In the BPSM1 mice,
there was spontaneous integration of retrotransposon
in the 3′-UTR[46] leading to the development of severe
polyarthritis, as well as heart valve disease. The panel
of mouse lines with combinatorial damage to several
regions of 3′-UTR involved in the control of mRNA sta-
bility demonstrates not only polyarthritis and heart
diseases but, in some cases, also embryonic lethality
as a result of extremely high TNF production  [47]. All
these regions are affected by the Cre/LoxP-mediated
deletion in the TNF
ΔARE
line developed in the Kollias’s
laboratory [48] 25years ago; therefore, with regard to
arthritis this model is equivalent to the BPSM1 line and
resembles the Tgl278 line, but with more pronounced
symptoms of systemic inflammation.
Taking into consideration complexities in regu-
lation of the genes encoding TNF and lymphotoxins,
detailed elucidation of the mechanism of accelerated
atrophy of the thymus in the mice carrying the entire
human locus Tnf/Lt as a transgene insertion (Table  2)
might need further investigation[49].
“REPORTER” MICE ARE AN IMPORTANT TOOL
FOR STUDYING TNF FUNCTIONS
Genetic constructs encoding different luminescent
or fluorescent proteins considerably expanded the pos-
sibilities of detection and visualization of gene activity
in  vitro and in vivo[54]. Transgenic mice, with expres-
sion of the protein under study being accompanied
by expression of the reporter protein, have become a
widespread tool for such studies [55]. In most cases,
transgenesis utilized bicistronic constructs that control
constitutive or conditional expression of the studied
protein and the fluorescent reporter protein. However,
it is also possible to design more complex constructs
that allow monitoring cells of a particular lineage of
cellular differentiation, with the fluorescence of one
protein switching to the florescence of another protein
in the course of differentiation[56].
Several variants of the reporter mice for TNF stud-
ies are known (Table  3). Luciferase, green fluorescent
protein GFP, and far-red fluorescent protein Katushka
were used as reporter proteins [57]. Detection of re-
porter proteins is possible not only by flow cytometry
and histological identification of TNF-producing cells,
but also by in vivo visualization.
NEDOSPASOV et al.858
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
CONCLUSIONS
TNF proved to be a surprisingly complex cytokine
with numerous functions both in homeostasis of the im-
mune system and in some nonimmune organs as well
as in different pathologies. Mouse models have made
it possible to link the protective and homeostatic func-
tions of TNF and its proinflammatory effects (which
may facilitate the development of certain pathologies),
to the particular types of the TNF-producing cells. One
conclusion that can be drawn from a large number
of studies discussed in this review is that the system-
ic TNF blockade in vivo will inevitably demonstrate
side effects due to neutralization of homeostatic and
protective functions of TNF. Consequently, elucidating
how to neutralize TNF only from a “pathogenic” cellu-
lar source[62], may allow us to apply improved thera-
peutic strategies to a variety of TNF-dependent diseases.
Acknowledgments. The authors are grateful to their
colleagues S.  I.  Grivennikov, L.  Tessarollo, A.  A.  Kuchmiy,
A.  R.  Galimov, S.  V.  Kozlov, I.  R.  Mufazalov, and R.  Nau-
mann for their contributions to creation and study of
some mouse models discussed in this review.
Contributions. S.A.N. the concept; S.A.N., I.V.A.,
and D.V.K. writing the manuscript; A.A.K., A.V.T., and
M.S.D. editing the manuscript and making important
additions. All authors participated in creation or study
of the mouse lines discussed in the review.
Funding. The work was supported by the Ministry
of Science and Higher Education of the Russian Feder-
ation, project “Development of Bioresource Collection
‘Collection of Laboratory Rodents with SPF-status for
Fundamental, Biomedical and Pharmacological Re-
search’ of the Institute of Biological Chemistry, Russian
Academy of Sciences” (agreement no.075-15-2021-1067).
The study of the phenotype of some genetically modi-
fied mouse lines was supported by the Russian Science
Foundation (project no.19-75-30032).
Ethics declarations. All manipulations were
carried out in accordance with the guideline of the
Federation of European Laboratory Animals Science
Associations. The experiments were approved by the
Bioethics Committee of the Institute of Molecular Biol-
ogy, Russian Academy of Sciences. The authors of this
work declare that they have no conflicts of interest.
REFERENCES
1. Parr, I., Wheeler, E., and Alexander, P. (1973) Similar-
ities of the anti-tumour actions of endotoxin, lipid A
and double-stranded RNA, Br. J. Cancer, 27, 370-389,
doi:10.1038/bjc.1973.45.
2. Carswell, E. A., Old, L. J., Kassel, R. L., Green, S.,
Fiore, N., and Williamson, B. (1975) An endotoxin-
induced serum factor that causes necrosis of tumors,
Proc. Natl. Acad. Sci. USA, 72, 3666-3670, doi:10.1073/
pnas.72.9.3666.
3. Coley, W. B. (1910) The treatment of inoperable
sarcoma by bacterial toxins (the mixed toxins of
the Streptococcus erysipelas and the Bacillus pro-
digiosus), Proc. R. Soc. Med., 3, 1-48, doi: 10.1177/
003591571000301601.
4. Pennica, D., Nedwin, G. E., Hayflick, J. S., Seeburg,
P.H., Derynck,R., Palladino, M.A., Kohr, W.J., Aggar-
wal, B. B., and Goeddel, D. V. (1984) Human tumour
necrosis factor: precursor structure, expression and
homology to lymphotoxin, Nature, 312, 724-729,
doi:10.1038/312724a0.
5. Brenner, D., Blaser, H., and Mak, T. W. (2015) Regu-
lation of tumour necrosis factor signalling: live or
let die, Nat. Rev. Immunol., 15, 362-374, doi: 10.1038/
nri3834.
6. Locksley, R. M., Killeen,N., and Lenardo, M. J. (2001)
The TNF and TNF receptor superfamilies, Cell, 104,
487-501, doi:10.1016/S0092-8674(01)00237-9.
7. Nedospasov, S. A., Shakhov, A. N., Turetskaya, R. L.,
Mett, V.A., and Georgiyev, G.P. (1985) Molecular clon-
ing of the genes encoding human tumor necrosis fac-
tors: tandem localization of the alpha and beta genes
in a short segment (6 k.b.p.) of the human genome,
Dokl. Akad. Nauk SSSR, 285, 1487-1490.
8. Pasparakis, M., Alexopoulou, L., Episkopou, V., and
Kollias,G. (1996) Immune and inflammatory respons-
es in TNF alpha-deficient mice: a critical requirement
for TNF alpha in the formation of primary Bcell fol-
licles, follicular dendritic cell networks and germinal
centers, and in the maturation of the humoral immune
response, J. Exp. Med., 184, 1397-1411, doi: 10.1084/
jem.184.4.1397.
9. Marino, M. W., Dunn, A., Grail,D., Inglese, M., Nogu-
chi,Y., and Richards,E. (1997) Characterization of tu-
mor necrosis factor-deficient mice, Proc. Natl. Acad.
Sci. USA, 94, 8093-8098, doi:10.1073/pnas.94.15.8093.
10. Taniguchi,T., Takata,M., Ikeda,A., Momotani,E., and
Sekikawa,K. (1997) Failure of germinal center forma-
tion and impairment of response to endotoxin in tu-
mor necrosis factor alpha-deficient mice, Lab. Invest.,
77, 647-658.
11. Iraqi, F., Sekikawa, K., Rowlands, J., and Teale, A.
(2001) Susceptibility of tumour necrosis factor-alpha
genetically deficient mice to Trypanosoma congolense
infection, Paras. Immunol., 23, 445-451, doi: 10.1046/
j.1365-3024.2001.00401.x.
12. Körner,H., Cook,M., and Riminton, D.S. (1997) Distinct
roles for lymphotoxin-α and tumor necrosis factor in
organogenesis and spatial organization of lymphoid
tissue, Eur. J. Immunol., 27, 2600-2609, doi: 10.1002/
eji.1830271020.
13. Kuprash, D. V., Tumanov, A. V., Liepinsh, D. J., Lem-
ckert, F. A., Hoek, R. M., Ledermann, B., Köntgen, F.,
REVERSE GENETICS APPLIED TO TNF IMMUNOBIOLOGY 859
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
De St Groth, B.F., and Sedgwick, J.D. (2005) Novel tu-
mor necrosis factor‐knockout mice that lack Peyer’s
patches, Eur. J. Immunol., 35, 1592-1600, doi: 10.1002/
eji.200526119.
14. Grivennikov, S.I., Tumanov, A.V., Liepinsh, D.J., Kru-
glov, A. A., Marakusha, B.I., Shakhov, A.N., Muraka-
mi,T., Drutskaya, L.N., Förster,I., Clausen, B.E., Tes-
sarollo,L., Ryffel,B., Kuprash, D.V., and Nedospasov,
S. A. (2005) Distinct and nonredundant in vivo func-
tions of TNF produced by T cells and macrophages/
neutrophils, Immunity, 22, 93-104, doi: 10.1016/
j.immuni.2004.11.016.
15. Eugster, H-P., Muller, M., Karrer, U., Car, B. D., Schny-
der, B., Eng, V. M., Woerly, G., Hir, M. L., Padova,
F.D., Aguet, M., Zinkernagel, R., Bluethmann,H., and
Ryffel, B. (1996) Multiple immune abnormalities in
tumor necrosis factor and lymphotoxin-α double-
deficient mice, Int. Immunol., 8, 23-36, doi: 10.1093/
intimm/8.1.23.
16. Amiot,F., Bellkaid,Y., Lebastard,M., Ave,P., Dautry,F.,
and Milon, G. (1996) Abnormal organisation of the
splenic marginal zone and the correlated leukocytosis
in lymphotoxin-alpha and tumor necrosis factor alpha
double deficient mice, Eur. Cytokine Netw., 7, 733-739.
17. Kuprash, D. V., Alimzhanov, M. B., Tumanov, A. V.,
Anderson, A. O., Pfeffer, K., and Nedospasov, S. A.
(1999) TNF and lymphotoxin beta cooperate in the
maintenance of secondary lymphoid tissue microar-
chitecture but not in the development of lymph nodes,
J. Immunol., 15, 6575-6580, doi: 10.4049/jimmunol.
163.12.6575.
18. Kuprash, D. V., Alimzhanov, M. B., Tumanov, A. V.,
Grivennikov, S. I., Shakhov, A. N., Drutskaya, L. N.,
Marino, M. W., Turetskaya, R. L., Anderson, A. O.,
Rajewsky,K., Pfeffer,K., and Nedospasov, S.A. (2002)
Redundancy in tumor necrosis factor (TNF) and lym-
photoxin (LT) signaling in vivo: mice with inactiva-
tion of the entire TNF/LT locus versus single-knockout
mice, Mol. Cell. Biol., 22, 8626-8634, doi:10.1128/MCB.
22.24.8626-8634.2002.
19. Rutschmann, S., Hoebe, K., Zalevsky, J., Du, X.,
Mann, N., Dahiyat, B. I., Steed, P., and Beutler, B.
(2006) PanR1, a dominant negative missense allele
of the gene encoding TNF-α (Tnf), does not impair
lymphoid development, J. Immunol., 176, 7525-7532,
doi:10.4049/jimmunol.176.12.7525.
20. Gray, P. W., Aggarwal, B. B., Benton, C. V., Bringman,
T.S., Henzel, W.J., Jarrett, J.A., Leung, D.W., Moffat,B.,
Ng,P., Svedersky, L.P., Palladino, M.A., and Nedwin,
G. E. (1984) Cloning and expression of cDNA for hu-
man lymphotoxin, a lymphokine with tumour necrosis
activity, Nature, 312, 721-724, doi:10.1038/312721a0.
21. De Togni, P., Goellner, J., Ruddle, N.H., Streeter, P. R.,
Fick, A., Mariathasan, S., Smith, S. C., Carlson, R.,
and Shornick, L. P. (1994) Abnormal development
of peripheral lymphoid organs in mice deficient in
lymphotoxin, Science, 264, 703-707, doi: 10.1126/
science.8171322.
22. Gogoleva, V. S., Kuprash, D.V., Grivennikov, S. I., Tu-
manov, A. V., Kruglov, A. A., and Nedospasov, S. A.
(2022) LTα, TNF, and ILC3 in Peyers patch organogen-
esis, Cells, 11, 1970, doi:10.3390/cells11121970.
23. Browning, J., Ngam-ek, A., Lawton, P., DeMarinis, J.,
Tizard, R., Chow, E. P., Hession, C., O’Brine-Greco, B.,
Foley, S.F., and Ware, C.F. (1993) Lymphotoxin beta, a
novel member of the TNF family that forms a hetero-
meric complex with lymphotoxin on the cell surface,
Cell, 72, 847-856, doi:10.1016/0092-8674(93)90574-A.
24. Crowe, P. D., VanArsdale, T. L., Walter, B. N., Ware,
C. F., Hession, C., Ehrenfels, B., Browning, J. L., Din,
W. S., Goodwin, R. G., and Smith, C. A. (1994) A lym-
photoxin-β-specific receptor, Science, 264, 707-710,
doi:10.1126/science.8171323.
25. Sauer,B., and Henderson, N. (1988) Site-specific DNA
recombination in mammalian cells by the Cre recom-
binase of bacteriophageP1, Proc. Natl. Acad. Sci. USA,
85, 5166-5170, doi:10.1073/pnas.85.14.5166.
26. Lakso,M., Sauer,B., Mosinger,B., Lee, E.J., Manning,
R.W., Yu, S.H., Mulder, K.L., and Westphal,H. (1992)
Targeted oncogene activation by site-specific recombi-
nation in transgenic mice, Proc. Natl. Acad. Sci. USA,
89, 6232-6236, doi:10.1073/pnas.89.14.6232.
27. Orban, P. C., Chui, D., and Marth, J. D. (1992) Tis-
sue- and site-specific DNA recombination in trans-
genic mice, Proc. Natl. Acad. Sci. USA, 89, 6861-6865,
doi:10.1073/pnas.89.15.6861.
28. Gu, H., Zou, Y. R., and Rajewsky, K. (1993) Indepen-
dent control of immunoglobulin switch recombina-
tion at individual switch regions evidenced through
Cre-loxP-mediated gene targeting, Cell, 73, 1155-1164,
doi:10.1016/0092-8674(93)90644-6.
29. Kühn, R., Schwenk, F., Aguet, M., and Rajewsky, K.
(1995) Inducible gene targeting in mice, Science,
269, 1427-1429, doi:10.1126/science.7660125.
30. Gu, H., Marth, J. D., Orban, P. C., Mossmann, H., and
Rajewsky, K. (1994) Deletion of a DNA polymerase
beta gene segment in T cells using cell type-specif-
ic gene targeting, Science, 265, 103-106, doi: 10.1126/
science.8016642.
31. Tumanov, A.V., Grivennikov, S.I., Kruglov, A.A., Sheb-
zukhov, Y.V., Koroleva, E.P., Piao,Y., Cui, C.-Y., Kuprash,
D.V., and Nedospasov, S.A. (2010) Cellular source and
molecular form of TNF specify its distinct functions
in organization of secondary lymphoid organs, Blood,
116, 3456-3464, doi:10.1182/blood-2009-10-249177.
32. Wolf, Y., Shemer, A., Polonsky, M., Gross, M., Mild-
ner, A., Yona, S., David, E., Kim, K.-W., Goldmann, T.,
Amit, I., Heikenwalder, M., Nedospasov, S., Prinz, M.,
Friedman, N., and Jung, S. (2017) Autonomous TNF
is critical for in vivo monocyte survival in steady
state and inflammation, J. Exp. Med., 214, 905-917,
doi:10.1084/jem.20160499.
NEDOSPASOV et al.860
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
33. Kruglov,A., Drutskaya,M., Schlienz,D., Gorshkova,E.,
Kurz,K., Morawietz,L., and Nedospasov,S. (2020) Con-
trasting contributions of TNF from distinct cellular
sources in arthritis, Ann. Rheum. Dis., 79, 1453-1459,
doi:10.1136/annrheumdis-2019-216068.
34. Wen, Y., Rudemiller, N. P., Zhang, J., Robinette, T.,
Lu,X., Ren,J., Privratsky, J.R., Nedospasov, S.A., and
Crowley, S. D. (2020) TNF-α in Tlymphocytes attenu-
ates renal injury and fibrosis during nephrotoxic ne-
phritis, Am.J. Physiol. Renal. Physiol., 318, F107-F116,
doi:10.1152/ajprenal.00347.2019.
35. Dudeck, J., Kotrba, J., Immler, R., Hoffmann, A.,
Voss,M., Alexaki, V.I., Morton,L., Jahn, S.R., Katsoulis-
Dimitriou,K., Winzer,S., Kollias,G., Fischer,T., Nedo-
spasov, S. A., Dunay, I. R., Chavakis, T., Müller, A. J.,
Schraven, B., Sperandio, M., and Dudeck, A. (2021)
Directional mast cell degranulation of tumor necro-
sis factor into blood vessels primes neutrophil ex-
travasation, Immunity, 54, 468-483.e5, doi: 10.1016/
j.immuni.2020.12.017.
36. Ninnemann, J., Winsauer, C., Bondareva, M., Kühl,
A. A., Lozza, L., Durek, P., Lissner, D., Siegmund, B.,
Kaufmann, S. H. E., Mashreghi, M.-F., Nedospasov,
S. A., and Kruglov, A. A. (2022) TNF hampers intesti-
nal tissue repair in colitis by restricting IL-22 bioavail-
ability, Mucosal. Immunol., 15, 698-716, doi: 10.1038/
s41385-022-00506-x.
37. Lakin,R., Polidovitch,N., Yang,S., Parikh,M., Liu, X.,
Debi, R., Gao, X., Chen, W., Guzman, C., Yakobov, S.,
Izaddoustdar, F., Wauchop, M., Lei, Q., Xu, W., Nedo-
spasov, S.A., Christoffels, V.M., and Backx, P.H. (2023)
Cardiomyocyte and endothelial cells play distinct
roles in the tumour necrosis factor (TNF)-dependent
atrial responses and increased atrial fibrillation vul-
nerability induced by endurance exercise training in
mice, Cardiovasc. Res., 119, 2607-2622, doi: 10.1093/
cvr/cvad144.
38. Kroetsch, J.T., Levy, A.S., Zhang,H., Aschar-Sobbi,R.,
Lidington,D., Offermanns,S., Nedospasov, S.A., Backx,
P.H., Heximer, S.P., and Bolz, S.-S. (2017) Constitutive
smooth muscle tumour necrosis factor regulates mi-
crovascular myogenic responsiveness and systemic
blood pressure, Nat. Commun., 8, 14805, doi:10.1038/
ncomms14805.
39. Dinh, D. D., Lidington, D., Kroetsch, J. T., Ng, C.,
Zhang,H., Nedospasov, S.A., Heximer, S.P., and Bolz,
S.-S. (2020) Experimental subarachnoid hemorrhage
drives catecholamine-dependent cardiac and periph-
eral microvascular dysfunction, Front. Physiol., 11,
402, doi:10.3389/fphys.2020.00402.
40. Han,J., Brown,T., and Beutler,B. (1990) Endotoxin-re-
sponsive sequences control cachectin/tumor necrosis
factor biosynthesis at the translational level, J. Exp.
Med., 171, 465-475, doi:10.1084/jem.171.2.465.
41. Yocum, D. E., Esparza, L., Dubry, S., Benjamin, J. B.,
Volz, R., and Scuderi, P. (1989) Characteristics of
tumor necrosis factor production in rheumatoid ar-
thritis, Cell. Immunol., 122, 131-145, doi: 10.1016/
0008-8749(89)90154-8.
42. Keffer, J., Probert, L., Cazlaris, H., Georgopoulos, S.,
Kaslaris, E., Kioussis, D., and Kollias, G. (1991)
Transgenic mice expressing human tumour necro-
sis factor: a predictive genetic model of arthritis,
EMBO J., 10, 4025-4031, doi: 10.1002/j.1460-2075.
1991.tb04978.x.
43. Maini, R.N., Elliott,M., Brennan, F.M., Williams, R.O.,
and Feldmann,M. (1994) Targeting TNF alpha for the
therapy of rheumatoid arthritis, Clin. Exp. Rheumatol.,
12, S63-S66.
44. Feldmann, M., Elliott, M. J., Maini, R. N., and Woody,
J. N. (1997) Anti-tumor necrosis factor-α therapy of
rheumatoid arthritis, Adv. Immunol., 64, 283-350,
doi:10.1016/S0065-2776(08)60891-3.
45. Hayward, M.D., Jones, B. K., Saparov, A., Hain, H.S.,
Trillat, A.-C., Bunzel, M. M., Corona, A., Li-Wang, B.,
Strenkowski,B., Giordano,C., Shen,H., Arcamone,E.,
Weidlick, J., Vilensky, M., Tugusheva, M., Felkner,
R. H., Campbell, W., Rao, Y., Grass, D. S., and
Buiakova, O. (2007) An extensive phenotypic charac-
terization of the hTNFα transgenic mice, BMC Physiol.
,
7, 13, doi:10.1186/1472-6793-7-13.
46. Lacey, D., Hickey, P., Arhatari, B. D., O’Reilly, L. A.,
Rohrbeck, L., Kiriazis, H., Du, X.-J., and Bouil-
let, P. (2015) Spontaneous retrotransposon inser-
tion into TNF 3′UTR causes heart valve disease and
chronic polyarthritis, Proc. Natl. Acad. Sci. USA, 112,
9698-9703, doi:10.1073/pnas.1508399112.
47. Clayer, E., Dalseno, D., Kueh, A., Lacey, D., Tsai, M.,
Arvell, E. H., Wimmer, V. C., and Bouillet, P. (2020)
Severe impairment of TNF post-transcriptional regu-
lation leads to embryonic death, iScience, 23, 101726,
doi:10.1016/j.isci.2020.101726.
48. Kontoyiannis, D., Pasparakis, M., Pizarro, T. T., Com-
inelli, F., and Kollias, G. (1999) Impaired on/off reg-
ulation of TNF biosynthesis in mice lacking TNF
AU-rich elements: implications for joint and gut-as-
sociated immunopathologies, Immunity, 10, 387-398,
doi:10.1016/s1074-7613(00)80038-2.
49. Liepinsh, D.J., Kruglov, A.A., Galimov, A.R., Shakhov,
A.N., Shebzukhov, Y.V., Kuchmiy, A.A., Grivennikov,
S.I., Tumanov, A.V., Drutskaya, M.S., Feigenbaum,L.,
Kuprash, D.V., and Nedospasov, S.A. (2009) Accelerat-
ed thymic atrophy as a result of elevated homeostatic
expression of the genes encoded by the TNF/lympho-
toxin cytokine locus, Eur. J. Immunol., 39, 2906-2915,
doi:10.1002/eji.200839191.
50. Olleros, M. L., Chavez-Galan, L., Segueni, N.,
Bourigault, M.L., Vesin,D., Kruglov, A.A., Drutskaya,
M. S., Bisig,R., Ehlers, S., Aly, S., Walter, K., Kuprash,
D.V., Chouchkova,M., Kozlov, S.V., Erard,F., Ryffel,B.,
Quesniaux, V. F. J., Nedospasov, S. A., and Garcia, I.
(2015) Control of mycobacterial infections in mice ex-
REVERSE GENETICS APPLIED TO TNF IMMUNOBIOLOGY 861
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
pressing human tumor necrosis factor (TNF) but not
mouse TNF, Infect. Immun., 83, 3612-3623, doi:10.1128/
IAI.00743-15.
51. Ameloot, P., Fiers, W., De Bleser, P., Ware, C. F., Van-
denabeele,P., and Brouckaert,P. (2001) Identification
of tumor necrosis factor (TNF) amino acids crucial for
binding to the murine p75 TNF receptor and construc-
tion of receptor-selective mutants, J.Biol. Chem., 276,
37426-37430, doi:10.1074/jbc.M102020200.
52. Atretkhany, K-S. N., Mufazalov, I. A., Dunst, J., Kuch-
miy, A., Gogoleva, V.S., Andruszewski, D., Drutskaya,
M. S., Faustman, D. L., Schwabenland, M., Prinz, M.,
Kruglov, A. A., Waisman, A., and Nedospasov, S. A.
(2018) Intrinsic TNFR2 signaling in T regulatory
cells provides protection in CNS autoimmunity, Proc.
Natl. Acad. Sci. USA, 115, 13051-13056, doi: 10.1073/
pnas.1807499115.
53. Ruuls, S. R., Hoek, R. M., and Ngo, V.N. (2001) Mem-
brane-bound TNF supports secondary lymphoid organ
structure but is subservient to secreted TNF in driv-
ing autoimmune inflammation, Immunity, 15, 533-543,
doi:10.1016/s1074-7613(01)00215-1.
54. Chudakov, D. M., Matz, M. V., Lukyanov, S., Lukyan-
ov, K. A., Zambito, G., and Mezzanotte, L. (2010) Flu-
orescent proteins and their applications in imaging
living cells and tissues, Physiol. Rev., 90, 1103-1163,
doi:10.1152/physrev.00038.2009.
55. Chawda, C., McMorrow, R., Gaspar, N., Zambito, G.,
and Mezzanotte, L. (2022) Monitoring immune cell
function through optical imaging: a review high-
lighting transgenic mouse models, Mol. Imaging Biol.,
24, 250-263, doi:10.1007/s11307-021-01662-5.
56. Rubtsov, Y.P., Niec, R.E., Josefowicz,S., Li,L., Darce,J.,
Mathis,D., Benoist,C., and Rudensky, A.Y. (2010) Sta-
bility of the regulatory Tcell lineage invivo, Science,
329, 1667-1671, doi:10.1126/science.1191996.
57. Shcherbo,D., Merzlyak, E.M., Chepurnykh, T.V., Frad-
kov, A.F., Ermakova, G.V., Solovieva, E.A., Lukyanov,
K. A., Bogdanova, E.A., Zaraisky, A. G., Lukyanov, S.,
and Chudakov, D.M. (2007) Bright far-red fluorescent
protein for whole-body imaging, Nat. Methods, 4, 741-
746, doi:10.1038/nmeth1083.
58. Shebzukhov, Y. V., Kuchmiy, A. A., Kruglov, A. A.,
Zipp, F., Siffrin, V., and Nedospasov, S. A. (2014) Ex-
perimental applications of TNF-reporter mice with
far-red fluorescent label, in The TNF Superfamily
(Bayry,J., ed.) New York, Springer New York, pp.151-
162, doi:10.1007/978-1-4939-0669-7_13.
59. Kuchmiy, A. A., Kruglov, A. A., Galimov, A. R., Sheb-
zukhov, Yu. V., Chudakov, D.M., Lukyanov, S.A., and
Nedospasov, S. A. (2011) A new line oftransgenic re-
porter mice for the study of expression of the tumor
necrosis factor, Ross. Immunol. Zh., 5, 3-4.
60. Minshawi, F., White, M. R. H., Muller, W., Hum-
phreys,N., Jackson,D., Campbell, B.J., Adamson,A., and
Papoutsopoulou, S. (2019) Human TNF-Luc reporter
mouse: a new model to quantify inflammatory respons-
es, Sci. Rep., 9, 193, doi:10.1038/s41598-018-36969-x.
61. Hall, B. E., Zhang, L., Sun, Z. J., Utreras, E., Prochaz-
kova, M., Cho, A., Terse, A., Arany, P., Dolan, J. C.,
Schmidt, B.L., and Kulkarni, A.B. (2016) Conditional
TNF-α overexpression in the tooth and alveolar bone
results in painful pulpitis and osteitis, J. Dent. Res.,
95, 188-195, doi:10.1177/0022034515612022.
62. Drutskaya, M. S., Efimov, G. A., Astrakhantseva, I. V.,
Kruglov, A. A., and Nedospasov, S. A. (2018) Making
anti-cytokine therapy more selective: studies in mice,
Cytokine, 101, 33-38, doi:10.1016/j.cyto.2016.08.022.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.