You are on page 1of 3

comment | SERIES

comment | SERIES

The immunology of blood: connecting the dots at


the neurovascular interface
Katerina Akassoglou recounts how her work on the mechanisms and functions of blood leaks in the brain led to the
discovery of fibrin-targeting immunotherapy.

Katerina Akassoglou

I
n 1883, Eduard von Rindfleisch identified were not the mediators of inflammatory
multiple sclerosis (MS) lesions around demyelination in the TNF-transgenic mice,
vessels that were “engorged with blood”, what was triggering the disease?
providing the first link between blood
leaks and brain inflammation1. Since then, Finding fibrin in Vienna
blood extravasation has been identified as a I presented my findings from the
common thread in many devastating brain TNF-transgenic mice to the Demyelination
and peripheral diseases and correlates with and Remyelination INSERM Conference
disease severity. However, a chicken-and-egg Philippe Laudat at Aix-Les-Bains in
question has posed a long-standing 1995. After a talk in which I faced more
dilemma: does more blood leak into the questions than I could answer, I realized
brain as a consequence of disease, or does how essential it would be to master
the influx of blood proteins enable disease neuropathology to understand how innate
pathogenesis? Here, I focus on my 26-year immune activation induced brain disease.
journey to connect the dots between blood, During the meeting social, a boat tour of
the inflammatory response and brain the Rhône river, I met Hans Lassmann, an
disease. Together with an international team expert MS neuropathologist at the Medical
of scientists and collaborators, our work University of Vienna. Dr. Lassmann was
shed light on the causal role of blood leaks in intrigued by my presentation, and we
toxic inflammation and tissue damage and brainstormed ways to experimentally
revealed a unique niche at the neurovascular Katerina Akassoglou in her laboratory at
determine the mechanism of disease in the
interface for the development of imaging Gladstone Institutes, University of California,
TNF-transgenic mice. In the fall of 1996,
tools and new therapeutics2–4. San Francisco in 2019.
with support from the European Molecular
Biology Organization, I moved to his
TNF days laboratory in Vienna to analyze hundreds
In 1994, I started my PhD work studying and paralysis without immunization of brains from the TNF-transgenic mice.
tumor necrosis factor (TNF) in the with myelin antigens was so unexpected A key finding from these studies was that
laboratory of George Kollias at the Hellenic that my short oral presentation at the IV axonal damage and oligodendrogliopathy
Pasteur Institute in Athens, Greece. International Society of Neuroimmunology are induced by innate immune activation,
The Kollias lab had just discovered that (ISNI) Congress in Amsterdam in 1994 was suggesting that TNF-transgenic mice
transgenic mice expressing TNF in the featured in the meeting highlights7. could be a new model for studying MS11.
synovium spontaneously developed severe A year later, our paper reporting To find out how this pathology started,
inflammation in their joints, indicating a spontaneous inflammatory demyelination we analyzed the brains early, before any
direct involvement of TNF in rheumatoid by TNF expression in the brain was symptoms occurred. Using electron
arthritis (RA)5. I was fortunate to participate communicated by Rita Levi-Montalcini to microscopy, I noticed major changes in
in the preclinical testing of anti-TNF the Proceedings of the National Academy blood vessels and perivascular deposits,
monoclonal antibodies that were later of Sciences8. Building on these findings, I which Dr. Lassmann identified as massive
approved for the treatment of RA6. However, generated multiple transgenic lines that blood–brain barrier (BBB) disruptions
as all the arthritis-related PhD projects revealed that TNF produced by astrocytes with fibrin deposits. We could detect BBB
were taken, I was tasked with resolving the but not neurons was responsible for the leaks as early as one week of age, before
enigmatic phenotype of a TNF-transgenic neurologic disease9. We attempted to any other symptom or cell infiltration were
mouse line that became paralyzed without genetically rescue the TNF-transgenic mice present. Had we found the smoking gun? As
signs of arthritis. In the Kollias lab we by depleting T cells but, to our surprise, fibrinogen is a marker for BBB disruption,
found that, in this mouse model, TNF was found that depletion of T cells exacerbated we looked for fibrinogen immunostaining
expressed only in the brain and spinal cord, features of the disease10. This result led in the brains of MS patients. I was awed
resulting in features reminiscent of MS us to conclude that innate immunity was to discover an abundance of fibrin
such as neuroinflammation, demyelination responsible for demyelination, and it made deposits, which often colocalized with
and paralysis. At the time, the finding us question the prevailing hypothesis that microglia and macrophages. Eager to
that expression of a cytokine in the brain T cell suppression would be therapeutic learn more about fibrin, I found a paper
was sufficient to induce demyelination in progressive disease10. But if T cells that had just been published reporting
710 Nature Immunology | VOL 21 | July 2020 | 710–712 | www.nature.com/natureimmunology
SERIES | comment
| SERIES comment

was confirmed in mouse models of


Blood vessel Alzheimer’s disease, pericyte deficiency
Fibrinogen and brain injury20–22. But how did fibrin
have these profound effects on the brain’s
immune and repair functions?

Fibrin-targeting Fibrin Zeroing in on innate immunity


Nervous system immunotherapy In 2002, in a review article in Biological
Chemistry entitled “Nervous system
pathology: the fibrin perspective”, we
Macrophages first proposed the hypothesis that fibrin
interacts with receptors on nervous
MCP1 NADPH oxidase
CXCL10 Microglia ROS system cells23. In my laboratory at the
T cells University of California San Diego and
Gladstone Institutes, we discovered the
Inflammation Oxidative stress cell targets for fibrinogen and fibrin in
the brain and identified a causal role for
BBB disruption in glial activation and
Inhibition of repair Demyelination Neurodegeneration
neuronal dysfunction24–27. We discovered
that culturing microglia on fibrin-coated
plates induced dramatic activation27.
Multiple sclerosis, brain injury, Alzheimer’s disease
Fibrinogen binds the CD11b/CD18 receptor
via the fibrin γ377–397 peptide28, and genetic
mutation of that peptide (producing
Fggγ390–396A mice) results in fully coagulable
fibrinogen that does not bind CD11b/
Fig. 1 | Fibrin mechanisms and functions in neuroinflammation. Upon BBB disruption, fibrinogen leaks CD1829. This allowed us to investigate the
into the brain, and, by activation of the coagulation cascade, it is converted to insoluble fibrin deposits. role of fibrin-induced inflammation in
Fibrin activates microglia and induces the release of CXCL10 and MCP1 to promote recruitment of the brain without disrupting hemostasis.
peripheral immune cells, as well as activation of NADPH oxidase to induce oxidative injury, leading to We showed that Fggγ390–396A mice had
tissue damage. Inhibition of the interaction of fibrin with its cellular receptors or supression of fibrin’s reduced neurological signs, inflammation,
downstream signaling pathways in cells could enable the discovery of selective therapies to block the demyelination and perivascular microglia
toxic effects of blood leaks in a wide range of diseases that cause vascular damage and inflammation. clusters in experimental autoimmune
encephalomyelitis (EAE, a mouse model
of MS)27,30. Using in vivo two-photon
that fibrin mediates inflammation via the many assumed I had changed fields. In imaging, we showed that fibrin deposition
CD11b/CD18 integrin receptor12. These Dr. Strickland’s laboratory, I first found and coagulation activity occurred early and
observations and results, in combination that tPA- and plasminogen-deficient mice correlated with microglial activation, while
with our earlier finding that astrocytes, had exacerbated demyelination after sciatic fibrin was also present during sustained
which regulate the BBB, were the primary nerve injury, which could be rescued by inflammation in chronic disease30–32.
source of neuroinflammation-promoting fibrin depletion14. In collaboration with Jay Fibrin signaling via CD11b/CD18 was
TNF9, led me to hypothesize that BBB Degen at Cincinnati Children’s Hospital, sufficient to induce demyelination,
disruption is an upstream activator of we performed the first genetic validation CXCL10 and MCP1 chemokine release,
innate-immune-mediated demyelination of the role of fibrinogen in the nervous and peripheral cell recruitment33, as
and neurodegeneration. system15. Mice lacking fibrinogen had well as oxidative stress via activation of
At the time, fibrin was primarily increased remyelination after nerve injury NADPH oxidase4 — a mechanism that
studied by hematologists for its role and decreased neuroinflammation and also contributes to microglia-mediated
in blood coagulation, which made my demyelination in the context of transgenic synapse loss in Alzheimer’s disease
search for a laboratory in which to study TNF expression14,16,17. These results models34. Blood clotting and oxidative
its role in brain inflammation unfruitful. were in line with the protective effects stress genes were mechanistically coupled
However, in 1997, I met Sidney Strickland of anticoagulants in autoimmune MS in neuroinflammation, suggesting a positive
at the 17th Blankenese Conference models18. In addition, fibrinogen-deficient feedback loop between fibrin, inflammation,
on Neurodegeneration in Hamburg, mice had increased expression of the coagulation and oxidative injury35. These
shortly after his landmark paper on the p75 neurotrophin receptor (p75NTR) after results established that the interaction
role of the fibrinolytic enzyme tissue nerve injury16. In the laboratory of Moses between fibrin and an inflammatory
plasminogen activator (tPA) in excitotoxic Chao at New York University, I made the receptor was necessary and sufficient to
neuronal death13. I joined his laboratory serendipitous discovery that tPA expression promote neuroinflammation (Fig. 1). Could
as a postdoctoral fellow, first at the State and fibrinolysis were suppressed by p75NTR, we develop an approach to selectively target
University of New York at Stony Brook thus identifying an unanticipated role for the inflammatory functions of fibrin without
and later at The Rockefeller University, to neurotrophin receptor signaling in the affecting its beneficial effects in hemostasis?
determine the role of the tPA/fibrin pathway removal of fibrin from the nervous system19.
in neuroinflammation. Integrating vascular Fibrinogen-deficient mice were later studied The making of an antibody
and BBB research with neuroimmunology in other laboratories, and their protection During my postdoc at The Rockefeller
was unconventional at the time, and from neuroinflammation and demyelination University, we occasionally had joint lab

Nature Immunology | VOL 21 | July 2020 | 710–712 | www.nature.com/natureimmunology 711


comment | SERIES
comment | SERIES

meetings with Barry Coller, the inventor it was known that multiple cytokines are 5. Keffer, J. et al. EMBO J. 10, 4025–4031 (1991).
6. Vilcek, J. Love and Science: A Memoir (Seven Stories Press,
of abciximab — a monoclonal antibody involved in the inflammatory process6.” 2016).
used as a thrombolytic drug that potently Despite this complexity, targeting individual 7. Cross, A. H., Vincent, A. & Racke, M. K. J. Neuroimmunol. 58,
inhibits fibrinogen binding to platelet αIIbβ3 proteins within the cytokine or angiogenic 117–120 (1995).
8. Probert, L., Akassoglou, K., Pasparakis, M., Kontogeorgos, G. &
integrin36. Thinking back on his work, I networks, such as TNF or the growth factor Kollias, G. Proc. Natl Acad. Sci. USA 92, 11294–11298 (1995).
wondered: if an antibody can specifically VEGF, has indeed succeeded in the clinic. 9. Akassoglou, K., Probert, L., Kontogeorgos, G. & Kollias, G. J.
inhibit the binding of fibrin to platelets Therefore, fibrin-targeting immunotherapy, Immunol. 158, 438–445 (1997).
10. Kassiotis, G. et al. Eur. J. Immunol. 29, 912–917 (1999).
and reduce thrombus formation, could we a novel approach to selectively suppress 11. Akassoglou, K. et al. Am. J. Pathol. 153, 801–813 (1998).
develop an antibody to specifically inhibit pathogenic innate immunity, may have 12. Tang, L., Ugarova, T. P., Plow, E. F. & Eaton, J. W. J. Clin. Invest.
binding of fibrin to immune cells and thus therapeutic benefits in clinical applications. 97, 1329–1334 (1996).
13. Tsirka, S. E., Gualandris, A., Amaral, D. G. & Strickland, S. Nature
stop inflammation without interfering with As fibrin is a global activator of innate 377, 340–344 (1995).
hemostasis? The fibrinogen γ chain contains immunity at sites of vascular damage3, these 14. Akassoglou, K., Kombrinck, K. W., Degen, J. L. & Strickland, S. J.
the CD11b/CD18 binding γ377–395 peptide28, approaches could have therapeutic benefits Cell Biol. 149, 1157–1166 (2000).
15. Bugge, T. H. et al. Cell 87, 709–719 (1996).
which is adjacent to but distinct from the in both brain and peripheral diseases.
16. Akassoglou, K., Yu, W.-M., Akpinar, P. & Strickland, S. Neuron 33,
motif that mediates platelet engagement. 861–875 (2002).
The γ377–395 peptide is considered cryptic What the future holds 17. Akassoglou, K. et al. Proc. Natl Acad. Sci. USA 101,
in soluble fibrinogen, and binding to The interface of the brain, immune and 6698–6703 (2004).
18. Paterson, P. Y. Fed. Proc. 35, 2428–2434 (1976).
CD11b occurs only after the conversion vascular systems represents a new frontier of 19. Sachs, B. D. et al. J. Cell Biol. 177, 1119–1132 (2007).
of fibrinogen to insoluble fibrin28. I scientific exploration, with the potential to 20. Cortes-Canteli, M. et al. Neuron 66, 695–709 (2010).
hypothesized that a monoclonal antibody change the way we think about fundamental 21. Montagne, A. et al. Nat. Med. 24, 326–337 (2018).
22. Pous, L. et al. Nat. Commun. 11, 630 (2020).
against the γ377–395 epitope would selectively mechanisms of neurological diseases and 23. Akassoglou, K. & Strickland, S. Biol. Chem. 383, 37–45 (2002).
inhibit the interaction between fibrin and the discovery of new therapies2. ‘Connecting 24. Schachtrup, C. et al. Proc. Natl Acad. Sci. USA 104,
CD11b without affecting coagulation. This the dots’ will continue to depend on an 11814–11819 (2007).
25. Petersen, M. A. et al. Neuron 96, 1003–1012.e7 (2017).
hypothesis was supported by our finding unwavering focus on the discovery of new 26. Schachtrup, C. et al. J. Neurosci. 30, 5843–5854 (2010).
that either mutating the γ377–395 epitope (in mechanisms, pursuing the integration 27. Adams, R. A. et al. J. Exp. Med. 204, 571–582 (2007).
Fggγ390–396A mice) or administering the γ377–395 of new technologies and promoting 28. Ugarova, T. P. et al. Biochemistry 42, 9365–9373 (2003).
29. Flick, M. J. et al. J. Clin. Invest. 113, 1596–1606 (2004).
peptide reduced neuroinflammation without collaborations within a community of 30. Davalos, D. et al. Nat. Commun. 3, 1227 (2012).
interfering with in vivo clotting time27. In experts in neuroscience, immunology, 31. Davalos, D. et al. Ann. Neurol. 75, 303–308 (2014).
my laboratory, we generated antibodies vascular biology, drug discovery and clinical 32. Lee, N. J. et al. Brain 141, 1637–1649 (2018).
against the γ377–395 peptide, and the lead development, all the while welcoming 33. Ryu, J. K. et al. Nat. Commun. 6, 8164 (2015).
34. Merlini, M. et al. Neuron 101, 1099–1108.e6 (2019).
clone was 5B8, a highly selective monoclonal serendipity. Developing safe and efficacious 35. Mendiola, A. S. et al. Nat. Immunol. 21, 513–524 (2020).
antibody that bound fibrin and blocked drugs against well-defined necessary and 36. Coller, B. S., Anderson, K. M. & Weisman, H. F. Haemostasis 26
fibrin-induced, CD11b-mediated activation sufficient targets, such as fibrin, could (Suppl. 4), 285–293 (1996).

of innate immunity without affecting expand the toolbox of therapies that target
fibrin polymerization4. As drug discovery neurovascular pathology. Implementing Acknowledgements
requires broad expertise, I assembled a functional multiomics approaches will I am especially grateful to all my current and former
laboratory members for their creativity, grit and
multidisciplinary team of 34 scientists from further reveal new mechanisms and
exceptional contributions. I thank my mentors,
three academic institutions and from the therapeutic targets at the neurovascular collaborators and colleagues for their enthusiasm and
pharmaceutical industry to fully characterize interface35. As the neurovascular interface continued support. Thanks to Hans Lassmann, Sidney
this fibrin-targeting immunotherapy4. Jae is a unique niche for the development Strickland, Jay L. Degen, George Kollias, Moses V. Chao,
Kyu Ryu and Anke Meyer-Franke led the of imaging tools and new therapies, Mark A. Petersen and Kathryn Claiborn for critical
reading of this manuscript. K.A. is currently supported
development of biochemical and cell assays drug discovery that selectively targets
by NIH/NINDS R35 NS097976, NIH/NIA RF1 AG064926,
to study fibrin in vitro, which enabled the pathogenic functions of blood proteins Department of Defense MS160082, the Dagmar Dolby
screening of fibrin-targeting antibody clones could change the diagnosis and treatment Family Fund, the Edward and Pearl Fein and the
and their prioritization for in vivo studies4. of neurodegenerative, autoimmune and Simon Family Trust. Lastly, I am grateful to the University
In accordance with the genetic evidence inflammatory diseases. ❐ of California, San Diego; Gladstone Institutes; and the
University of California, San Francisco for promoting a
in Fggγ390–396A mice27,34, we found that the culture of discovery and innovation.
fibrin-targeting immunotherapy protected Katerina Akassoglou    ✉
mouse models of MS and Alzheimer’s Gladstone Institutes and Department of Neurology,
Competing interests
disease from neuroinflammation and Weill Institute for Neurosciences, University of K.A. is the scientific founder and advisor of Therini Bio,
neurodegeneration4 (Fig. 1). California, San Francisco, San Francisco, CA, USA. Inc. K.A. is an inventor on The Regents of the University
As neurovascular interactions are complex ✉e-mail: kakassoglou@gladstone.ucsf.edu of California issued patents US7807645, US8569242,
and multifactorial, one might ask whether US8877195 and US8980836 covering fibrin antibodies;
targeting a single blood protein would be Published online: 23 June 2020 a co-inventor on The Regents of the University of
https://doi.org/10.1038/s41590-020-0671-z California and Gladstone Institutes pending patent
enough to protect from neuroinflammation. applications U.S. 15/943,474 and EP 15735033.1 covering
A similar question was raised in the early References fibrin assays of microglia activation; and a co-inventor on
nineties regarding anti-TNF therapy, as noted 1. Rindfleisch, E. Archiv Pathol. Anat. Physiol. 26, 474–483 (1863). Gladstone Institutes issued patent US9669112 covering
by Jan Vilcek, the inventor of infliximab: 2. Petersen, M. A., Ryu, J. K. & Akassoglou, K. Nat. Rev. Neurosci. fibrin in vivo models and US10451611 and pending
19, 283–301 (2018). patent applications U.S. 16/572,365 and EP 15735284.05
“To most scientists and physicians it seemed 3. Davalos, D. & Akassoglou, K. Semin. Immunopathol. 34, covering in vitro fibrin assays. Her interests are managed
inconceivable that blocking a single cytokine 43–62 (2012). in accordance with the Gladstone Institutes’ conflict of
could be beneficial for RA patients, when 4. Ryu, J. K. et al. Nat. Immunol. 19, 1212–1223 (2018). interest policy.

712 Nature Immunology | VOL 21 | July 2020 | 710–712 | www.nature.com/natureimmunology

You might also like