The understanding of the dialogue between the brain and the immune system has undergone dramatic changes over the last two decades, with immense impact on the perception of neurodegenerative diseases, mental dysfunction, and many other brain pathologic conditions. Accumulated results have suggested that optimal function of the brain is dependent on support from the immune system, provided that this immune response is tightly controlled. Moreover, in contrast to the previous prevailing dogma, it is now widely accepted that circulating immune cells are needed for coping with brain pathologies and that their optimal effect is dependent on their type, location, and activity. In this perspective, we describe our own scientific journey, reviewing the milestones in attaining this understanding of the brain–immune axis integrated with numerous related studies by others. We then explain their significance in demonstrating the possibility of harnessing the immune system in a well-controlled manner for the treatment of neurodegenerative diseases.

Alzheimer's disease (AD) was first identified in 1906 as a disease characterized by distinctive plaques of amyloid-β and Tau neurofibrillary tangles associated with cognitive and mental dysfunction. Since then, numerous attempts were made to reverse or, at least, to modify or attenuate this disease by targeting the accumulating proteins, with the aim of plaque elimination (16). Unfortunately, such attempts have largely failed (7, 8). Over the years, several other studies suggested that AD, like many other acute and chronic neurodegenerative diseases, is associated with local inflammation (9), and efforts were therefore made to reduce it by systemic administration of steroidal or nonsteroidal anti-inflammatory drugs. Such treatments also failed to affect disease progression (10), leaving the scientific community with the question of what had been missed in the understanding of the disease (7, 1113).

Independent basic research studies carried out by our group and others over the years strived for a deep understanding of the physiological mechanisms that maintain the healthy brain and help repair it. Such mechanisms might be lost or are insufficient under disease conditions, but could be amenable to boosting. To this end, we focused on the potential role of the immune system. When we embarked on this quest 20 years ago, even the possibility that immune cells might not be harmful to the brain and, needless to say, the suggestion that they could be beneficial was considered almost heretical.

Our journey was initiated by a simple assumption that it is inconceivable that the CNS, which is so precious and indispensable, would lack the capacity to be assisted by immune cells for its repair, whereas almost every other tissue depends on such immune support. Since then, we introduced and substantiated the essential role of the blood-borne innate and adaptive immune cells in supporting the brain during normal homeostasis and their vital role in repair following injury or facing neurodegenerative conditions (1428). Based on these works, together with numerous additional studies by others (2942), a new paradigm has emerged, suggesting that tightly regulated brain–immune communication is a necessary component of life-long brain maintenance and is essential for brain repair. More recently, it was found that brain–immune cross-talk is impaired in aging (4347) and in neurodegenerative diseases (23, 4850), and thus a fundamental mechanism of maintenance and support might be lost (23, 51) that could be amenable to restoration by rejuvenating the immune system (5053).

These findings do not negate the original definition of the brain as an immune-privileged organ (54); they refine the understanding of the brain as an organ that, rather than functioning under complete segregation, engages in a unique and tightly regulated relationship with the immune system (55, 56). In this perspective, we describe our own scientific journey alongside with numerous related studies by others, from the original notion of systemic immune cells as destructive to the brain to their current recognition as crucial (if well controlled) (Fig. 1). We further describe the change of perspective from viewing the brain’s borders as absolute barriers between the brain and the circulation, to the current understanding that these borders can also serve as permissive gates for the entry of “reparative” leukocytes, as sites from which immune cells can affect the brain (37, 43, 57, 58), and as sites that can function as a lymphatic system for the brain (40, 41, 5962). Finally, we explain the rationale underlying the shift from attempts to mitigate AD using immunosuppression to our proposal of harnessing the immune system by immunotherapy for the treatment of AD (50, 51).

FIGURE 1.

Milestones (in colors) in the understanding of the communication between the brain and the immune system. The journey started with the common wisdom that the brain is isolated from the immune system (54). Initial studies (I and II) that began to undermine these assumptions highlighted the need for circulating immune cells (both monocyte-derived macrophages and T cells) for brain repair (14, 15, 78, 79), followed by (III) initiation of studies showing that circulating immune cells are needed to support healthy brain plasticity (17, 18, 37, 39). These were also followed by (IV) studies showing the pivotal role of monocyte-derived macrophages (8390) and of T cells (91, 92) in chronic neurodegenerative diseases. The above studies led to intensive research (V) striving to identify the site of communication between the brain and the immune system (20, 22, 23, 43, 49, 59), which led to the most recent approach (VI) of immunotherapy, empowering the peripheral immune system to fight against AD and dementia and, potentially, against additional neurodegenerative diseases (50, 51). (Note that many additional references related to each of these milestones are cited in the text).

FIGURE 1.

Milestones (in colors) in the understanding of the communication between the brain and the immune system. The journey started with the common wisdom that the brain is isolated from the immune system (54). Initial studies (I and II) that began to undermine these assumptions highlighted the need for circulating immune cells (both monocyte-derived macrophages and T cells) for brain repair (14, 15, 78, 79), followed by (III) initiation of studies showing that circulating immune cells are needed to support healthy brain plasticity (17, 18, 37, 39). These were also followed by (IV) studies showing the pivotal role of monocyte-derived macrophages (8390) and of T cells (91, 92) in chronic neurodegenerative diseases. The above studies led to intensive research (V) striving to identify the site of communication between the brain and the immune system (20, 22, 23, 43, 49, 59), which led to the most recent approach (VI) of immunotherapy, empowering the peripheral immune system to fight against AD and dementia and, potentially, against additional neurodegenerative diseases (50, 51). (Note that many additional references related to each of these milestones are cited in the text).

Close modal

Outside the CNS, inflammation is a physiological response that is essential for tissue repair and becomes pathological if not resolved in a timely manner. As part of such peripheral “physiological” inflammation, macrophages were found to be essential players for removing dead cells and cell debris, as well as for facilitating immune resolution and tissue repair (63, 64). It is a multistep reparative process that starts with immune activation and ends with local immune resolution (19, 20, 26, 6568). It is still debatable whether the immune resolution involves the recruitment of an additional wave of macrophages with distinct activities or whether it involves a local conversion of a single population of myeloid cells from an inflammatory to a resolving phenotype (69).

The general view, with respect to the CNS, was that it should be fully protected from immune cell entry to ensure its proper homeostasis and function. Accordingly, the failure of the CNS to recover following acute injury was often attributed to local inflammation, assuming that such inflammation might reflect, at least in part, infiltration of inflammatory cells to the site of damage, a process that should be altogether prevented or blocked (70, 71). Researchers indiscriminately viewed CNS local inflammation as an obstacle to recovery (72, 73) and to cell renewal (7476). Therefore, it was suggested that acute treatment with steroidal anti-inflammatory drugs following spinal cord injury would promote repair (71, 77). In an apparent contrast, in animal models of acute CNS injury, including optic nerve injury (14, 67, 78), spinal cord injury (15, 16, 19, 7982), and brain ischemia (35), it was found by our team, and subsequently by others, that blood-borne macrophages and T cells can facilitate repair. In addition, as will be discussed below, monocyte-derived macrophages were also found by several independent studies to have a role in coping with chronic brain pathologic conditions (37, 39, 78, 79, 8392).

Overall, reparative macrophages were shown to locally display multiple functions, including anti-inflammatory (19, 20, 79), scar degrading activities (93, 94), and the ability to support axonal growth (78).

The finding that peripheral immune cells can facilitate neural tissue protection and repair following CNS injury highlighted the possibility that the peripheral immunity might have a role in supporting CNS homeostasis and functional plasticity. To address this, initial studies focused on the hippocampus, which is known to exhibit life-long neurogenesis (95, 96), a high functional plasticity in its normal day-to-day activity, and is involved in spatial learning and memory (97, 98). It was discovered that neurogenesis, as well as spatial learning and memory, are impaired in young immune-compromised animals, suggesting that systemic immune cells affect healthy brain plasticity (17, 18, 2934, 3639, 42, 99, 100). Over the years, independent studies have identified the meninges as sites through which the immune system can affect the healthy brain. For example, T cell–derived IL-4 was found to enhance cognitive function (37). In addition, meningeal IFN-γ–producing immune cells were found to support social behavior (39). Similarly, deficiency of IFN-γ signaling from middle age onward was found to be linked to cognitive impairment (43). Other studies have suggested that the peripheral immune system supports the ability to cope with mentally stressful conditions (101103). Along the same lines, alterations in T cell number and function have been associated with some cases of autism spectrum disorders (104107). Taken together, these results and others have encouraged studies striving for a better understanding of the relationship between the brain and circulating immune cells.

In searching for physiological sites of brain–immune communications under “sterile” acute injurious conditions, our team found that monocytes can attain access to the CNS through the leptomeninges adjacent to the lesion site and through the blood–cerebrospinal fluid barrier (BCSFB) at the choroid plexus (CP) (20, 22). Those cells that enter through the BCSFB contribute to the resolution of the inflammation within the site of pathology (20). Investigating the properties of the CP revealed that the trafficking of leukocytes through it is different from the invasion of pathological immune cells through the breached endothelium of the blood–brain barrier in diseases such as multiple sclerosis (108, 109). Thus, for example, following spinal cord injury the epithelial layer of the CP was found to be activated to express increased levels of leukocyte trafficking molecules such as CCL2, ICAM-1, and CXCL10 (22). This activity of the CP was found to be dependent on signals originating from the circulation, such as IFN-γ, and signals emerging from the brain parenchyma, via the cerebrospinal fluid, informing the CP that the brain is in distress (22). Of interest, effector memory T cells were found to populate the CP stroma and to secrete cytokines, such as IFN-γ, IL-4, and IL-10, which can modify the activity of the CP epithelium to express chemokines, cell-adhesion molecules, and growth factors (22, 48). Of note, it is not yet clear whether the cells that reside in the CP and orchestrate its activity for supporting leukocyte trafficking are the same cells that are found in the cerebrospinal fluid and the parenchyma. Over the years, the CP was found to function as a physiological gateway for immune surveillance (20, 43, 101), and a correlation was found between expression of trafficking molecules by the CP and the number of leukocytes in the cerebrospinal fluid (101). The fate of the CP following traumatic injury to the brain has not been studied. Nevertheless, under acute mental stress, the expression of leukocyte trafficking molecules by the CP was found to be affected (101). Taken together, these emerging studies have demonstrated that the meninges and the CP have multiple functions in brain homeostasis, neurogenesis, and repair (20, 37, 43, 49, 110114).

Further studies are needed to fully describe the relationship between T cells that populate the CP, those that pass through the CP, and those that populate the meninges.

Approximately 10 percent of the brain’s cells are myeloid immune cells, known as microglia (115117). They were shown to have a distinct origin from that of macrophages and a slow rate of self-renewal (115). The microglia are maintained under tight regulation, which allows them to act as sentinels and to restore homeostasis following a mild to moderate deviation due to disturbance or damage; however, such a tight regulation restricts their ability to display a robust activity under a severe (118) or persistent pathologic condition, as in the case of chronic neurodegeneration (119). Among the factors controlling microglial activity are cell–cell interactions based on receptor/ligand pairs, such as CD200L-CD200R (120) and CX3CL1-CX3CR1 (121), and soluble factors, such as TGF-β (119, 122, 123).

The role and function of microglia in neurodegenerative diseases have not been fully elucidated. Genome-wide association studies have identified disease-associated risk factors in AD patients, many of which are immune-related genes expressed by microglia (124127) and other immune cells (128131). Among such risk factors is triggering receptor expressed on myeloid cells 2 (TREM2) (132135).

Using single-cell RNA sequencing in a mouse model of AD driven by amyloid-β, a novel subset of disease-associated microglia was identified. These cells develop through a stepwise process in a TREM2-dependent fashion (124). Based on the unique RNA expression profile of these cells, it appears that they may demonstrate protective activity (124). Nevertheless, it is not clear why these activated microglia are not sufficient to mitigate the disease (118, 136). It is possible that their maturation occurs late in the disease course or that these cells are able to function only transiently and become exhausted or even destructive (137). Importantly, there is no consensus, even with respect to the role of TREM2 in microglial activity; however, several findings suggest that the role of TREM2 in microglia, positive or negative, is dependent on the stage of the disease (135, 138143). Overall, the role of microglia in neurodegenerative conditions is still under study, and their function or dysfunction could differ among diseases and their subsets.

In addition to the resident microglia, as mentioned above, monocyte-derived macrophages are also recruited to the CNS under disease pathologic conditions. Although the function of these cells has not been fully elucidated, several studies have suggested that they play an important beneficial role in AD (23, 50, 51, 83, 8589, 144149). Moreover, boosting the number of monocyte-derived macrophages in different models of AD, using various immunological manipulations, was found to be beneficial in reducing the pathology and improving cognitive function (51, 146148). These monocyte-derived macrophages were found to exhibit features associated with enhanced phagocytic activity, along with the expression of scavenger receptors and cytokines that either are not expressed by microglia or are not induced in a timely manner when needed (51, 87, 146149). Notably, the recruitment of monocytes to the CNS was shown to be dependent on the chemokine receptor CCR2 (86). Many of the genetic AD risk factors are associated with immune activity, including factors affecting microglia and monocyte-derived macrophages. Yet, it is evident from several independent studies that in these diseases the myeloid cells within the brain are strongly affected by environmental and nutritional factors, including ω-3 fatty acids (150), curcuminoids (151), vitamin D (152), and resveratrol (153).

It should be noted that in the past, many published studies globally referred to all myeloid cells in the brain as “microglia” with no direct distinction between myeloid cell types, and thus the conclusions might not accurately reflect the specific function of each population.

The disappointing results in many clinical trials aimed at modifying AD by eradication of amyloid-β plaque (14), together with the unclear causal relationships between the levels of plaque load in the brain and cognitive loss (154156), have argued for the presence of additional factors, which may be responsible for the cognitive impairment and thus should be targeted (11, 157159). It is conceivable that adopting a strategy to enhance multiple mechanisms of repair would be more effective than a strategy that directly puts down a single destructive process.

In addition to the monocyte-derived macrophages, regulatory T cells are needed, as well, in restricting inflammatory conditions within the brain (23, 92). In apparent contrast, peripheral effector T cells were also found to be important players in neurodegenerative conditions (91, 160163). Together, these studies suggest that different cell types are displaying distinct functions at the brain's borders and within the brain parenchyma; effector T cells activate the gateway to the brain and allow trafficking through it, whereas monocytes and regulatory T cells are functioning within the site of pathology. Altogether, these results support the notion that aging and exhaustion of the immune system could contribute to the onset and escalation of neurodegenerative diseases. The challenge is how to maintain a balanced response and to orchestrate the activities of all these cell types.

Studies addressing the fate of the CP in animal models of neurodegenerative pathologies, such as AD and amyotrophic lateral sclerosis, revealed that its function as a leukocyte gateway is impaired (23, 48, 49). Restoring the function of the CP could be achieved by different strategies of boosting the peripheral immune system, including transient reduction of systemic Foxp3+ regulatory T cells (23), blocking the PD-1/PD-L1 immune checkpoint pathway (50, 51, 164), or vaccination by CNS Ags (48).

Targeting the PD-1/PD-L1 inhibitory immune checkpoint pathway in the periphery revealed that it is possible to modify disease pathology in animal models driven by amyloidosis and tauopathy (50, 51). For a long-term effect in AD and dementia, an intermittent treatment was found to be sufficient, and thus the potential for adverse immunological effects is likely to be low. Such a treatment (Fig. 2) was shown to reduce brain inflammation, rescue neurons, and reduce brain pathology, although it is not directly targeting any specific disease hallmark. The effect was attributed, at least in part, to the enhanced homing of monocytes to the brain (50, 51), which can augment tissue repair by locally functioning as inflammation-resolving cells, and by expressing scavenger molecules (87, 89, 149). It remains to be determined whether and how resident microglia are affected by such an intervention. Temporarily reducing immunosuppressive mechanisms helps release effector T cell activity, and at the same time, maintains a sufficient number of anti-inflammatory cells in the periphery. Such a treatment approach initiates an immunological chain of events affecting the peripheral immune system as the primary target of the treatment to allow trafficking of inflammation-resolving cells to the sites of brain pathology, in which they play an essential role (23, 92, 165) (Fig. 2).

FIGURE 2.

Schematic representation illustrating the dynamics and the mechanism underlying the disease modification evoked by empowering the peripheral immune system by immune checkpoint blockade. The Abs (e.g., directed to PD-1 or PD-L1) that block inhibitory immune checkpoints reach circulating immune cells, lymphoid organs, and, potentially, the immune cells within the BCSFB’s stroma. The interaction between the Abs and their relevant receptors results in multiple subsequent changes in the peripheral immune system including increased levels of PD-1+ CD4+ effector memory T cells and increased availability of IFN-γ, which activates the choroid plexus epithelium to express leukocyte trafficking molecules. The enhanced ability of the CP to express these molecules supports recruitment of monocyte-derived macrophages and regulatory T cells to the brain. The monocyte-derived macrophages can directly, as well as indirectly, display multiple activities, including suppression of the inflammatory milieu of the brain via local production of IL-10, facilitating removal of amyloid-β oligomers and plaques by expressing unique scavenger receptors (e.g., MSR1). Such an immune modulation is followed by enhanced neuronal survival, rescue of synapses, and a more supportive environment for the functioning of the brain. Altogether, this multistep process leads to disease modification and cognitive improvement.

FIGURE 2.

Schematic representation illustrating the dynamics and the mechanism underlying the disease modification evoked by empowering the peripheral immune system by immune checkpoint blockade. The Abs (e.g., directed to PD-1 or PD-L1) that block inhibitory immune checkpoints reach circulating immune cells, lymphoid organs, and, potentially, the immune cells within the BCSFB’s stroma. The interaction between the Abs and their relevant receptors results in multiple subsequent changes in the peripheral immune system including increased levels of PD-1+ CD4+ effector memory T cells and increased availability of IFN-γ, which activates the choroid plexus epithelium to express leukocyte trafficking molecules. The enhanced ability of the CP to express these molecules supports recruitment of monocyte-derived macrophages and regulatory T cells to the brain. The monocyte-derived macrophages can directly, as well as indirectly, display multiple activities, including suppression of the inflammatory milieu of the brain via local production of IL-10, facilitating removal of amyloid-β oligomers and plaques by expressing unique scavenger receptors (e.g., MSR1). Such an immune modulation is followed by enhanced neuronal survival, rescue of synapses, and a more supportive environment for the functioning of the brain. Altogether, this multistep process leads to disease modification and cognitive improvement.

Close modal

Several recent studies have suggested additional targets for immunotherapy, based on directly modulating microglia by increasing their phagocytic activity via inhibition of either CD22 (166) or CD33 (167) or by reducing the shedding of TREM2 (168).

Overall, it seems that because of the heterogeneity of AD and dementia conditions and the multiple detrimental factors (environmental and innate) contributing to the cognitive impairment and disease escalation, the targeting of a single factor (e.g., amyloid-β plaque burden, accumulation of aggregated Tau, or local neuroinflammation) is apparently not sufficient to modify such diseases. By empowering the peripheral immune system, it might be possible to overcome the difficulties of coordinately targeting multiple factors that contribute to disease escalation and cognitive impairment, which may differ between patients, and at different timepoints along the course of the disease (169). Immunotherapy, in general, could serve as a means to harness the immune system to fight this disease, independently of its primary etiology, either familial or sporadic and whether it is driven by amyloid-β, Tau, or both. Also, this therapeutic strategy might overcome the need to identify the key precipitating factor(s) leading to the initiation of the pathologic condition or additional factors whose modification is critical to arrest disease progression, or at least change its course. The fact that aging is the major risk factor of all sporadic neurodegenerative diseases supports the notion that rejuvenating the immune system could be a promising therapeutic approach.

On a personal note, the fact that 20 years ago no one would have dared to consider an option as immunotherapy for AD emphasizes how a basic scientific question can lead to a surprising mechanism-based approach (Fig. 1).

M.S. holds the Maurice and Ilse Katz Professorial Chair in Neuroimmunology.

This work was supported by Advanced European Research Council Grant 232835, Israel Science Foundation (ISF) Research Grant 991/16, and ISF Legacy Heritage Biomedical Science Partnership Research Grant 1354/15. We thank the Adelis and Thompson foundations for generous support of our Alzheimer's disease research.

Abbreviations used in this article:

AD

Alzheimer's disease

BCSFB

blood–cerebrospinal fluid barrier

CP

choroid plexus

TREM2

triggering receptor expressed on myeloid cells 2.

1
Egan
,
M. F.
,
J.
Kost
,
P. N.
Tariot
,
P. S.
Aisen
,
J. L.
Cummings
,
B.
Vellas
,
C.
Sur
,
Y.
Mukai
,
T.
Voss
,
C.
Furtek
, et al
.
2018
.
Randomized trial of verubecestat for mild-to-moderate Alzheimer’s disease
.
N. Engl. J. Med.
378
:
1691
1703
.
2
Doody
,
R. S.
,
R.
Raman
,
M.
Farlow
,
T.
Iwatsubo
,
B.
Vellas
,
S.
Joffe
,
K.
Kieburtz
,
F.
He
,
X.
Sun
,
R. G.
Thomas
, et al
Alzheimer’s Disease Cooperative Study Steering Committee
; 
Semagacestat Study Group
.
2013
.
A phase 3 trial of semagacestat for treatment of Alzheimer’s disease
.
N. Engl. J. Med.
369
:
341
350
.
3
Gauthier
,
S.
,
H. H.
Feldman
,
L. S.
Schneider
,
G. K.
Wilcock
,
G. B.
Frisoni
,
J. H.
Hardlund
,
H. J.
Moebius
,
P.
Bentham
,
K. A.
Kook
,
D. J.
Wischik
, et al
.
2016
.
Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial
.
Lancet
388
:
2873
2884
.
4
Mullard
,
A.
2016
.
Pharma pumps up anti-tau Alzheimer pipeline despite first Phase III failure
.
Nat. Rev. Drug Discov.
15
:
591
592
.
5
Masters
,
C. L.
,
R.
Bateman
,
K.
Blennow
,
C. C.
Rowe
,
R. A.
Sperling
,
J. L.
Cummings
.
2015
.
Alzheimer’s disease
.
Nat. Rev. Dis. Primers
1
:
15056
.
6
Selkoe
,
D. J.
,
J.
Hardy
.
2016
.
The amyloid hypothesis of Alzheimer’s disease at 25 years
.
EMBO Mol. Med.
8
:
595
608
.
7
Panza
,
F.
,
M.
Lozupone
,
V.
Dibello
,
A.
Greco
,
A.
Daniele
,
D.
Seripa
,
G.
Logroscino
,
B. P.
Imbimbo
.
2019
.
Are antibodies directed against amyloid-β (Aβ) oligomers the last call for the Aβ hypothesis of Alzheimer’s disease?
Immunotherapy
11
:
3
6
.
8
Egan
,
M. F.
,
J.
Kost
,
T.
Voss
,
Y.
Mukai
,
P. S.
Aisen
,
J. L.
Cummings
,
P. N.
Tariot
,
B.
Vellas
,
C. H.
van Dyck
,
M.
Boada
, et al
.
2019
.
Randomized trial of verubecestat for prodromal Alzheimer’s disease
.
N. Engl. J. Med.
380
:
1408
1420
.
9
Heneka
,
M. T.
,
M. J.
Carson
,
J.
El Khoury
,
G. E.
Landreth
,
F.
Brosseron
,
D. L.
Feinstein
,
A. H.
Jacobs
,
T.
Wyss-Coray
,
J.
Vitorica
,
R. M.
Ransohoff
, et al
.
2015
.
Neuroinflammation in Alzheimer’s disease
.
Lancet Neurol.
14
:
388
405
.
10
Meyer
,
P. F.
,
J.
Tremblay-Mercier
,
J.
Leoutsakos
,
C.
Madjar
,
M. É.
Lafaille-Maignan
,
M.
Savard
,
P.
Rosa-Neto
,
J.
Poirier
,
P.
Etienne
,
J.
Breitner
;
PREVENT-AD Research Group
.
2019
.
INTREPAD: a randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. [Published erratum appears in 2019 Neurology 93: 371.]
Neurology
92
:
e2070
e2080
.
11
Wang
,
J.
,
B. J.
Gu
,
C. L.
Masters
,
Y. J.
Wang
.
2017
.
A systemic view of Alzheimer disease - insights from amyloid-β metabolism beyond the brain. [Published erratum appears in 2017 Nat. Rev. Neurol. 13: 703.]
Nat. Rev. Neurol.
13
:
612
623
.
12
Stower
,
H.
2018
.
Searching for Alzheimer’s disease therapies
.
Nat. Med.
24
:
894
897
.
13
Verheijen
,
J.
,
K.
Sleegers
.
2018
.
Understanding Alzheimer disease at the interface between genetics and transcriptomics
.
Trends Genet.
34
:
434
447
.
14
Moalem
,
G.
,
R.
Leibowitz-Amit
,
E.
Yoles
,
F.
Mor
,
I. R.
Cohen
,
M.
Schwartz
.
1999
.
Autoimmune T cells protect neurons from secondary degeneration after central nervous system axotomy
.
Nat. Med.
5
:
49
55
.
15
Rapalino
,
O.
,
O.
Lazarov-Spiegler
,
E.
Agranov
,
G. J.
Velan
,
E.
Yoles
,
M.
Fraidakis
,
A.
Solomon
,
R.
Gepstein
,
A.
Katz
,
M.
Belkin
, et al
.
1998
.
Implantation of stimulated homologous macrophages results in partial recovery of paraplegic rats
.
Nat. Med.
4
:
814
821
.
16
Hauben
,
E.
,
E.
Agranov
,
A.
Gothilf
,
U.
Nevo
,
A.
Cohen
,
I.
Smirnov
,
L.
Steinman
,
M.
Schwartz
.
2001
.
Posttraumatic therapeutic vaccination with modified myelin self-antigen prevents complete paralysis while avoiding autoimmune disease
.
J. Clin. Invest.
108
:
591
599
.
17
Kipnis
,
J.
,
H.
Cohen
,
M.
Cardon
,
Y.
Ziv
,
M.
Schwartz
.
2004
.
T cell deficiency leads to cognitive dysfunction: implications for therapeutic vaccination for schizophrenia and other psychiatric conditions. [Published erratum appears in 2005 Proc. Natl. Acad. Sci. USA 102: 14122]
Proc. Natl. Acad. Sci. USA
101
:
8180
8185
.
18
Ziv
,
Y.
,
N.
Ron
,
O.
Butovsky
,
G.
Landa
,
E.
Sudai
,
N.
Greenberg
,
H.
Cohen
,
J.
Kipnis
,
M.
Schwartz
.
2006
.
Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood
.
Nat. Neurosci.
9
:
268
275
.
19
Shechter
,
R.
,
A.
London
,
C.
Varol
,
C.
Raposo
,
M.
Cusimano
,
G.
Yovel
,
A.
Rolls
,
M.
Mack
,
S.
Pluchino
,
G.
Martino
, et al
.
2009
.
Infiltrating blood-derived macrophages are vital cells playing an anti-inflammatory role in recovery from spinal cord injury in mice
.
PLoS Med.
6
:
e1000113
.
20
Shechter
,
R.
,
O.
Miller
,
G.
Yovel
,
N.
Rosenzweig
,
A.
London
,
J.
Ruckh
,
K. W.
Kim
,
E.
Klein
,
V.
Kalchenko
,
P.
Bendel
, et al
.
2013
.
Recruitment of beneficial M2 macrophages to injured spinal cord is orchestrated by remote brain choroid plexus
.
Immunity
38
:
555
569
.
21
London
,
A.
,
E.
Itskovich
,
I.
Benhar
,
V.
Kalchenko
,
M.
Mack
,
S.
Jung
,
M.
Schwartz
.
2011
.
Neuroprotection and progenitor cell renewal in the injured adult murine retina requires healing monocyte-derived macrophages
.
J. Exp. Med.
208
:
23
39
.
22
Kunis
,
G.
,
K.
Baruch
,
N.
Rosenzweig
,
A.
Kertser
,
O.
Miller
,
T.
Berkutzki
,
M.
Schwartz
.
2013
.
IFN-γ-dependent activation of the brain’s choroid plexus for CNS immune surveillance and repair
.
Brain
136
:
3427
3440
.
23
Baruch
,
K.
,
N.
Rosenzweig
,
A.
Kertser
,
A.
Deczkowska
,
A. M.
Sharif
,
A.
Spinrad
,
A.
Tsitsou-Kampeli
,
A.
Sarel
,
L.
Cahalon
,
M.
Schwartz
.
2015
.
Breaking immune tolerance by targeting Foxp3(+) regulatory T cells mitigates Alzheimer’s disease pathology
.
Nat. Commun.
6
:
7967
.
24
Raposo
,
C.
,
N.
Graubardt
,
M.
Cohen
,
C.
Eitan
,
A.
London
,
T.
Berkutzki
,
M.
Schwartz
.
2014
.
CNS repair requires both effector and regulatory T cells with distinct temporal and spatial profiles
.
J. Neurosci.
34
:
10141
10155
.
25
Schwartz
,
M.
,
R.
Shechter
.
2010
.
Systemic inflammatory cells fight off neurodegenerative disease
.
Nat. Rev. Neurol.
6
:
405
410
.
26
Shechter
,
R.
,
A.
London
,
M.
Schwartz
.
2013
.
Orchestrated leukocyte recruitment to immune-privileged sites: absolute barriers versus educational gates
.
Nat. Rev. Immunol.
13
:
206
218
.
27
Shechter
,
R.
,
M.
Schwartz
.
2013
.
CNS sterile injury: just another wound healing?
Trends Mol. Med.
19
:
135
143
.
28
Shechter
,
R.
,
M.
Schwartz
.
2013
.
Harnessing monocyte-derived macrophages to control central nervous system pathologies: no longer ‘if’ but ‘how’
.
J. Pathol.
229
:
332
346
.
29
Clark
,
S. M.
,
C. N.
Vaughn
,
J. A.
Soroka
,
X.
Li
,
L. H.
Tonelli
.
2018
.
Neonatal adoptive transfer of lymphocytes rescues social behaviour during adolescence in immune-deficient mice
.
Eur. J. Neurosci.
47
:
968
978
.
30
Song
,
E. J.
,
S. G.
Jeon
,
K. A.
Kim
,
J. I.
Kim
,
M.
Moon
.
2017
.
Restricted CD4+ T cell receptor repertoire impairs cognitive function via alteration of Th2 cytokine levels
.
Neurogenesis (Austin)
4
:
e1256856
.
31
Qi
,
F.
,
Z.
Zuo
,
J.
Yang
,
S.
Hu
,
Y.
Yang
,
Q.
Yuan
,
J.
Zou
,
K.
Guo
,
Z.
Yao
.
2017
.
Combined effect of BCG vaccination and enriched environment promote neurogenesis and spatial cognition via a shift in meningeal macrophage M2 polarization
.
J. Neuroinflammation
14
:
32
.
32
Coder
,
B.
,
W.
Wang
,
L.
Wang
,
Z.
Wu
,
Q.
Zhuge
,
D. M.
Su
.
2017
.
Friend or foe: the dichotomous impact of T cells on neuro-de/re-generation during aging
.
Oncotarget
8
:
7116
7137
.
33
Song
,
C.
,
J. D.
Nicholson
,
S. M.
Clark
,
X.
Li
,
A. D.
Keegan
,
L. H.
Tonelli
.
2016
.
Expansion of brain T cells in homeostatic conditions in lymphopenic Rag2(-/-) mice
.
Brain Behav. Immun.
57
:
161
172
.
34
Möhle
,
L.
,
N.
Israel
,
K.
Paarmann
,
M.
Krohn
,
S.
Pietkiewicz
,
A.
Müller
,
I. N.
Lavrik
,
J. S.
Buguliskis
,
B. H.
Schott
,
D.
Schlüter
, et al
.
2016
.
Chronic toxoplasma gondii infection enhances β-amyloid phagocytosis and clearance by recruited monocytes
.
Acta Neuropathol. Commun.
4
:
25
.
35
Wattananit
,
S.
,
D.
Tornero
,
N.
Graubardt
,
T.
Memanishvili
,
E.
Monni
,
J.
Tatarishvili
,
G.
Miskinyte
,
R.
Ge
,
H.
Ahlenius
,
O.
Lindvall
, et al
.
2016
.
Monocyte-derived macrophages contribute to spontaneous long-term functional recovery after stroke in mice
.
J. Neurosci.
36
:
4182
4195
.
36
Pape
,
K.
,
R.
Tamouza
,
M.
Leboyer
,
F.
Zipp
.
2019
.
Immunoneuropsychiatry - novel perspectives on brain disorders
.
Nat. Rev. Neurol.
15
:
317
328
.
37
Derecki
,
N. C.
,
A. N.
Cardani
,
C. H.
Yang
,
K. M.
Quinnies
,
A.
Crihfield
,
K. R.
Lynch
,
J.
Kipnis
.
2010
.
Regulation of learning and memory by meningeal immunity: a key role for IL-4
.
J. Exp. Med.
207
:
1067
1080
.
38
Filiano
,
A. J.
,
S. P.
Gadani
,
J.
Kipnis
.
2017
.
How and why do T cells and their derived cytokines affect the injured and healthy brain?
Nat. Rev. Neurosci.
18
:
375
384
.
39
Filiano
,
A. J.
,
Y.
Xu
,
N. J.
Tustison
,
R. L.
Marsh
,
W.
Baker
,
I.
Smirnov
,
C. C.
Overall
,
S. P.
Gadani
,
S. D.
Turner
,
Z.
Weng
, et al
.
2016
.
Unexpected role of interferon-γ in regulating neuronal connectivity and social behaviour
.
Nature
535
:
425
429
.
40
Louveau
,
A.
,
B. A.
Plog
,
S.
Antila
,
K.
Alitalo
,
M.
Nedergaard
,
J.
Kipnis
.
2017
.
Understanding the functions and relationships of the glymphatic system and meningeal lymphatics
.
J. Clin. Invest.
127
:
3210
3219
.
41
Louveau
,
A.
,
I.
Smirnov
,
T. J.
Keyes
,
J. D.
Eccles
,
S. J.
Rouhani
,
J. D.
Peske
,
N. C.
Derecki
,
D.
Castle
,
J. W.
Mandell
,
K. S.
Lee
, et al
.
2015
.
Structural and functional features of central nervous system lymphatic vessels. [Published erratum appears in 2016 Nature 533: 278.]
Nature
523
:
337
341
.
42
Walsh
,
J. T.
,
S.
Hendrix
,
F.
Boato
,
I.
Smirnov
,
J.
Zheng
,
J. R.
Lukens
,
S.
Gadani
,
D.
Hechler
,
G.
Gölz
,
K.
Rosenberger
, et al
.
2015
.
MHCII-independent CD4+ T cells protect injured CNS neurons via IL-4
.
J. Clin. Invest.
125
:
2547
.
43
Baruch
,
K.
,
A.
Deczkowska
,
E.
David
,
J. M.
Castellano
,
O.
Miller
,
A.
Kertser
,
T.
Berkutzki
,
Z.
Barnett-Itzhaki
,
D.
Bezalel
,
T.
Wyss-Coray
, et al
.
2014
.
Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function
.
Science
346
:
89
93
.
44
Yousef
,
H.
,
C. J.
Czupalla
,
D.
Lee
,
M. B.
Chen
,
A. N.
Burke
,
K. A.
Zera
,
J.
Zandstra
,
E.
Berber
,
B.
Lehallier
,
V.
Mathur
, et al
.
2019
.
Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1
.
Nat. Med.
25
:
988
1000
.
45
Matt
,
S. M.
,
R. W.
Johnson
.
2016
.
Neuro-immune dysfunction during brain aging: new insights in microglial cell regulation
.
Curr. Opin. Pharmacol.
26
:
96
101
.
46
Lucin
,
K. M.
,
T.
Wyss-Coray
.
2009
.
Immune activation in brain aging and neurodegeneration: too much or too little?
Neuron
64
:
110
122
.
47
Villeda
,
S. A.
,
J.
Luo
,
K. I.
Mosher
,
B.
Zou
,
M.
Britschgi
,
G.
Bieri
,
T. M.
Stan
,
N.
Fainberg
,
Z.
Ding
,
A.
Eggel
, et al
.
2011
.
The ageing systemic milieu negatively regulates neurogenesis and cognitive function
.
Nature
477
:
90
94
.
48
Kunis
,
G.
,
K.
Baruch
,
O.
Miller
,
M.
Schwartz
.
2015
.
Immunization with a myelin-derived antigen activates the brain’s choroid plexus for recruitment of immunoregulatory cells to the CNS and attenuates disease progression in a mouse model of ALS
.
J. Neurosci.
35
:
6381
6393
.
49
Mesquita
,
S. D.
,
A. C.
Ferreira
,
F.
Gao
,
G.
Coppola
,
D. H.
Geschwind
,
J. C.
Sousa
,
M.
Correia-Neves
,
N.
Sousa
,
J. A.
Palha
,
F.
Marques
.
2015
.
The choroid plexus transcriptome reveals changes in type I and II interferon responses in a mouse model of Alzheimer’s disease
.
Brain Behav. Immun.
49
:
280
292
.
50
Baruch
,
K.
,
A.
Deczkowska
,
N.
Rosenzweig
,
A.
Tsitsou-Kampeli
,
A. M.
Sharif
,
O.
Matcovitch-Natan
,
A.
Kertser
,
E.
David
,
I.
Amit
,
M.
Schwartz
.
2016
.
PD-1 immune checkpoint blockade reduces pathology and improves memory in mouse models of Alzheimer’s disease
.
Nat. Med.
22
:
135
137
.
51
Rosenzweig
,
N.
,
R.
Dvir-Szternfeld
,
A.
Tsitsou-Kampeli
,
H.
Keren-Shaul
,
H.
Ben-Yehuda
,
P.
Weill-Raynal
,
L.
Cahalon
,
A.
Kertser
,
K.
Baruch
,
I.
Amit
, et al
.
2019
.
PD-1/PD-L1 checkpoint blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model
.
Nat. Commun.
10
:
465
.
52
Villeda
,
S. A.
,
K. E.
Plambeck
,
J.
Middeldorp
,
J. M.
Castellano
,
K. I.
Mosher
,
J.
Luo
,
L. K.
Smith
,
G.
Bieri
,
K.
Lin
,
D.
Berdnik
, et al
.
2014
.
Young blood reverses age-related impairments in cognitive function and synaptic plasticity in mice
.
Nat. Med.
20
:
659
663
.
53
Wyss-Coray
,
T.
2016
.
Ageing, neurodegeneration and brain rejuvenation
.
Nature
539
:
180
186
.
54
Medawar
,
P. B.
1948
.
Immunity to homologous grafted skin; the fate of skin homografts transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye
.
Br. J. Exp. Pathol.
29
:
58
69
.
55
Schwartz
,
M.
,
J.
Kipnis
,
S.
Rivest
,
A.
Prat
.
2013
.
How do immune cells support and shape the brain in health, disease, and aging?
J. Neurosci.
33
:
17587
17596
.
56
Kipnis
,
J.
2016
.
Multifaceted interactions between adaptive immunity and the central nervous system
.
Science
353
:
766
771
.
57
Kipnis
,
J.
,
S.
Gadani
,
N. C.
Derecki
.
2012
.
Pro-cognitive properties of T cells
.
Nat. Rev. Immunol.
12
:
663
669
.
58
Radjavi
,
A.
,
I.
Smirnov
,
N.
Derecki
,
J.
Kipnis
.
2014
.
Dynamics of the meningeal CD4(+) T-cell repertoire are defined by the cervical lymph nodes and facilitate cognitive task performance in mice
.
Mol. Psychiatry
19
:
531
533
.
59
Da Mesquita
,
S.
,
A.
Louveau
,
A.
Vaccari
,
I.
Smirnov
,
R. C.
Cornelison
,
K. M.
Kingsmore
,
C.
Contarino
,
S.
Onengut-Gumuscu
,
E.
Farber
,
D.
Raper
, et al
.
2018
.
Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. [Published erratum appears in 2018 Nature 564: E7.]
Nature
560
:
185
191
.
60
Sandrone
,
S.
,
D.
Moreno-Zambrano
,
J.
Kipnis
,
J.
van Gijn
.
2019
.
A (delayed) history of the brain lymphatic system
.
Nat. Med.
25
:
538
540
.
61
Antila
,
S.
,
S.
Karaman
,
H.
Nurmi
,
M.
Airavaara
,
M. H.
Voutilainen
,
T.
Mathivet
,
D.
Chilov
,
Z.
Li
,
T.
Koppinen
,
J. H.
Park
, et al
.
2017
.
Development and plasticity of meningeal lymphatic vessels
.
J. Exp. Med.
214
:
3645
3667
.
62
Absinta
,
M.
,
S. K.
Ha
,
G.
Nair
,
P.
Sati
,
N. J.
Luciano
,
M.
Palisoc
,
A.
Louveau
,
K. A.
Zaghloul
,
S.
Pittaluga
,
J.
Kipnis
,
D. S.
Reich
.
2017
.
Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI
.
eLife
6
:
e29738
.
63
Vargas
,
M. E.
,
B. A.
Barres
.
2007
.
Why is Wallerian degeneration in the CNS so slow?
Annu. Rev. Neurosci.
30
:
153
179
.
64
Perry
,
V. H.
,
P. B.
Andersson
,
S.
Gordon
.
1993
.
Macrophages and inflammation in the central nervous system
.
Trends Neurosci.
16
:
268
273
.
65
Buckley
,
C. D.
,
D. W.
Gilroy
,
C. N.
Serhan
,
B.
Stockinger
,
P. P.
Tak
.
2013
.
The resolution of inflammation
.
Nat. Rev. Immunol.
13
:
59
66
.
66
Gronert
,
K.
2010
.
Resolution, the grail for healthy ocular inflammation
.
Exp. Eye Res.
91
:
478
485
.
67
London
,
A.
,
I.
Benhar
,
M. J.
Mattapallil
,
M.
Mack
,
R. R.
Caspi
,
M.
Schwartz
.
2013
.
Functional macrophage heterogeneity in a mouse model of autoimmune central nervous system pathology
.
J. Immunol.
190
:
3570
3578
.
68
Schwartz
,
M.
,
K.
Baruch
.
2014
.
The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus
.
EMBO J.
33
:
7
22
.
69
Mills
,
C. D.
,
K.
Ley
.
2014
.
M1 and M2 macrophages: the chicken and the egg of immunity
.
J. Innate Immun.
6
:
716
726
.
70
Popovich
,
P. G.
,
T. B.
Jones
.
2003
.
Manipulating neuroinflammatory reactions in the injured spinal cord: back to basics
.
Trends Pharmacol. Sci.
24
:
13
17
.
71
Young
,
W.
2002
.
Methylprednisolone and spinal cord injury
.
J. Neurosurg.
96
(
1
Suppl.
):
141
142
.
72
Trivedi
,
A.
,
A. D.
Olivas
,
L. J.
Noble-Haeusslein
.
2006
.
Inflammation and spinal cord injury: infiltrating leukocytes as determinants of injury and repair processes
.
Clin. Neurosci. Res.
6
:
283
292
.
73
Tran
,
A. P.
,
P. M.
Warren
,
J.
Silver
.
2018
.
The biology of regeneration failure and success after spinal cord injury
.
Physiol. Rev.
98
:
881
917
.
74
Fan
,
L. W.
,
Y.
Pang
.
2017
.
Dysregulation of neurogenesis by neuroinflammation: key differences in neurodevelopmental and neurological disorders
.
Neural Regen. Res.
12
:
366
371
.
75
Ekdahl
,
C. T.
,
J. H.
Claasen
,
S.
Bonde
,
Z.
Kokaia
,
O.
Lindvall
.
2003
.
Inflammation is detrimental for neurogenesis in adult brain
.
Proc. Natl. Acad. Sci. USA
100
:
13632
13637
.
76
Monje
,
M. L.
,
H.
Toda
,
T. D.
Palmer
.
2003
.
Inflammatory blockade restores adult hippocampal neurogenesis
.
Science
302
:
1760
1765
.
77
Bracken
,
M. B.
,
M. J.
Shepard
,
W. F.
Collins
,
T. R.
Holford
,
W.
Young
,
D. S.
Baskin
,
H. M.
Eisenberg
,
E.
Flamm
,
L.
Leo-Summers
,
J.
Maroon
, et al
.
1990
.
A randomized, controlled trial of methylprednisolone or naloxone in the treatment of acute spinal-cord injury. Results of the second national acute spinal cord injury study
.
N. Engl. J. Med.
322
:
1405
1411
.
78
Yin
,
Y.
,
M. T.
Henzl
,
B.
Lorber
,
T.
Nakazawa
,
T. T.
Thomas
,
F.
Jiang
,
R.
Langer
,
L. I.
Benowitz
.
2006
.
Oncomodulin is a macrophage-derived signal for axon regeneration in retinal ganglion cells
.
Nat. Neurosci.
9
:
843
852
.
79
Kigerl
,
K. A.
,
J. C.
Gensel
,
D. P.
Ankeny
,
J. K.
Alexander
,
D. J.
Donnelly
,
P. G.
Popovich
.
2009
.
Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord
.
J. Neurosci.
29
:
13435
13444
.
80
Ishii
,
H.
,
X.
Jin
,
M.
Ueno
,
S.
Tanabe
,
T.
Kubo
,
S.
Serada
,
T.
Naka
,
T.
Yamashita
.
2012
.
Adoptive transfer of Th1-conditioned lymphocytes promotes axonal remodeling and functional recovery after spinal cord injury
.
Cell Death Dis.
3
:
e363
.
81
Greenhalgh
,
A. D.
,
S.
David
.
2014
.
Differences in the phagocytic response of microglia and peripheral macrophages after spinal cord injury and its effects on cell death
.
J. Neurosci.
34
:
6316
6322
.
82
Cusimano
,
M.
,
D.
Biziato
,
E.
Brambilla
,
M.
Donegà
,
C.
Alfaro-Cervello
,
S.
Snider
,
G.
Salani
,
F.
Pucci
,
G.
Comi
,
J. M.
Garcia-Verdugo
, et al
.
2012
.
Transplanted neural stem/precursor cells instruct phagocytes and reduce secondary tissue damage in the injured spinal cord
.
Brain
135
:
447
460
.
83
Butovsky
,
O.
,
M.
Koronyo-Hamaoui
,
G.
Kunis
,
E.
Ophir
,
G.
Landa
,
H.
Cohen
,
M.
Schwartz
.
2006
.
Glatiramer acetate fights against Alzheimer’s disease by inducing dendritic-like microglia expressing insulin-like growth factor 1
.
Proc. Natl. Acad. Sci. USA
103
:
11784
11789
.
84
Butovsky
,
O.
,
G.
Kunis
,
M.
Koronyo-Hamaoui
,
M.
Schwartz
.
2007
.
Selective ablation of bone marrow-derived dendritic cells increases amyloid plaques in a mouse Alzheimer’s disease model
.
Eur. J. Neurosci.
26
:
413
416
.
85
Simard
,
A. R.
,
D.
Soulet
,
G.
Gowing
,
J. P.
Julien
,
S.
Rivest
.
2006
.
Bone marrow-derived microglia play a critical role in restricting senile plaque formation in Alzheimer’s disease
.
Neuron
49
:
489
502
.
86
Naert
,
G.
,
S.
Rivest
.
2013
.
A deficiency in CCR2+ monocytes: the hidden side of Alzheimer’s disease
.
J. Mol. Cell Biol.
5
:
284
293
.
87
Town
,
T.
,
Y.
Laouar
,
C.
Pittenger
,
T.
Mori
,
C. A.
Szekely
,
J.
Tan
,
R. S.
Duman
,
R. A.
Flavell
.
2008
.
Blocking TGF-beta-Smad2/3 innate immune signaling mitigates Alzheimer-like pathology
.
Nat. Med.
14
:
681
687
.
88
Koronyo-Hamaoui
,
M.
,
M. K.
Ko
,
Y.
Koronyo
,
D.
Azoulay
,
A.
Seksenyan
,
G.
Kunis
,
M.
Pham
,
J.
Bakhsheshian
,
P.
Rogeri
,
K. L.
Black
, et al
.
2009
.
Attenuation of AD-like neuropathology by harnessing peripheral immune cells: local elevation of IL-10 and MMP-9
.
J. Neurochem.
111
:
1409
1424
.
89
Koronyo
,
Y.
,
B. C.
Salumbides
,
J.
Sheyn
,
L.
Pelissier
,
S.
Li
,
V.
Ljubimov
,
M.
Moyseyev
,
D.
Daley
,
D. T.
Fuchs
,
M.
Pham
, et al
.
2015
.
Therapeutic effects of glatiramer acetate and grafted CD115+ monocytes in a mouse model of Alzheimer’s disease
.
Brain
138
:
2399
2422
.
90
El Khoury
,
J.
,
M.
Toft
,
S. E.
Hickman
,
T. K.
Means
,
K.
Terada
,
C.
Geula
,
A. D.
Luster
.
2007
.
Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease
.
Nat. Med.
13
:
432
438
.
91
Beers
,
D. R.
,
J. S.
Henkel
,
W.
Zhao
,
J.
Wang
,
S. H.
Appel
.
2008
.
CD4+ T cells support glial neuroprotection, slow disease progression, and modify glial morphology in an animal model of inherited ALS
.
Proc. Natl. Acad. Sci. USA
105
:
15558
15563
.
92
Henkel
,
J. S.
,
D. R.
Beers
,
S.
Wen
,
A. L.
Rivera
,
K. M.
Toennis
,
J. E.
Appel
,
W.
Zhao
,
D. H.
Moore
,
S. Z.
Powell
,
S. H.
Appel
.
2013
.
Regulatory T-lymphocytes mediate amyotrophic lateral sclerosis progression and survival. [Published erratum appears in 2013 EMBO Mol. Med. 5: 326.]
EMBO Mol. Med.
5
:
64
79
.
93
Rolls
,
A.
,
R.
Shechter
,
A.
London
,
Y.
Segev
,
J.
Jacob-Hirsch
,
N.
Amariglio
,
G.
Rechavi
,
M.
Schwartz
.
2008
.
Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation
.
PLoS Med.
5
:
e171
.
94
Shechter
,
R.
,
C.
Raposo
,
A.
London
,
I.
Sagi
,
M.
Schwartz
.
2011
.
The glial scar-monocyte interplay: a pivotal resolution phase in spinal cord repair
.
PLoS One
6
:
e27969
.
95
Yao
,
J.
,
Y.
Mu
,
F. H.
Gage
.
2012
.
Neural stem cells: mechanisms and modeling. [Published erratum appears in 2012 Protein Cell 3: 559.]
Protein Cell
3
:
251
261
.
96
van Praag
,
H.
,
A. F.
Schinder
,
B. R.
Christie
,
N.
Toni
,
T. D.
Palmer
,
F. H.
Gage
.
2002
.
Functional neurogenesis in the adult hippocampus
.
Nature
415
:
1030
1034
.
97
Santarelli
,
L.
,
M.
Saxe
,
C.
Gross
,
A.
Surget
,
F.
Battaglia
,
S.
Dulawa
,
N.
Weisstaub
,
J.
Lee
,
R.
Duman
,
O.
Arancio
, et al
.
2003
.
Requirement of hippocampal neurogenesis for the behavioral effects of antidepressants
.
Science
301
:
805
809
.
98
Abrous
,
D. N.
,
J. M.
Wojtowicz
.
2015
.
Interaction between neurogenesis and hippocampal memory system: new vistas
.
Cold Spring Harb. Perspect. Biol.
7
:
a018952
.
99
Brynskikh
,
A.
,
T.
Warren
,
J.
Zhu
,
J.
Kipnis
.
2008
.
Adaptive immunity affects learning behavior in mice
.
Brain Behav. Immun.
22
:
861
869
.
100
Kokaia
,
Z.
,
G.
Martino
,
M.
Schwartz
,
O.
Lindvall
.
2012
.
Cross-talk between neural stem cells and immune cells: the key to better brain repair?
Nat. Neurosci.
15
:
1078
1087
.
101
Kertser, A., K. Baruch, A. Deczkowska, A. Weiner, T. Croese, M. Kenigsbuch, I. Cooper, M. Tsoory, S. Ben-Hamo, I. Amit, and M. Schwartz. 2019. Corticosteroid signaling at the brain-immune interface impedes coping with severe psychological stress. Sci. Adv. 5: eaav4111
102
Cohen
,
H.
,
Y.
Ziv
,
M.
Cardon
,
Z.
Kaplan
,
M. A.
Matar
,
Y.
Gidron
,
M.
Schwartz
,
J.
Kipnis
.
2006
.
Maladaptation to mental stress mitigated by the adaptive immune system via depletion of naturally occurring regulatory CD4+CD25+ cells
.
J. Neurobiol.
66
:
552
563
.
103
Lewitus
,
G. M.
,
H.
Cohen
,
M.
Schwartz
.
2008
.
Reducing post-traumatic anxiety by immunization
.
Brain Behav. Immun.
22
:
1108
1114
.
104
Warren
,
R. P.
,
L. J.
Yonk
,
R. A.
Burger
,
P.
Cole
,
J. D.
Odell
,
W. L.
Warren
,
E.
White
,
V. K.
Singh
.
1990
.
Deficiency of suppressor-inducer (CD4+CD45RA+) T cells in autism
.
Immunol. Invest.
19
:
245
251
.
105
Yonk
,
L. J.
,
R. P.
Warren
,
R. A.
Burger
,
P.
Cole
,
J. D.
Odell
,
W. L.
Warren
,
E.
White
,
V. K.
Singh
.
1990
.
CD4+ helper T cell depression in autism
.
Immunol. Lett.
25
:
341
345
.
106
Ashwood
,
P.
,
P.
Krakowiak
,
I.
Hertz-Picciotto
,
R.
Hansen
,
I. N.
Pessah
,
J.
Van de Water
.
2011
.
Associations of impaired behaviors with elevated plasma chemokines in autism spectrum disorders
.
J. Neuroimmunol.
232
:
196
199
.
107
Filiano
,
A. J.
,
S. P.
Gadani
,
J.
Kipnis
.
2015
.
Interactions of innate and adaptive immunity in brain development and function
.
Brain Res.
1617
:
18
27
.
108
Bartholomäus
,
I.
,
N.
Kawakami
,
F.
Odoardi
,
C.
Schläger
,
D.
Miljkovic
,
J. W.
Ellwart
,
W. E.
Klinkert
,
C.
Flügel-Koch
,
T. B.
Issekutz
,
H.
Wekerle
,
A.
Flügel
.
2009
.
Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions
.
Nature
462
:
94
98
.
109
Reboldi
,
A.
,
C.
Coisne
,
D.
Baumjohann
,
F.
Benvenuto
,
D.
Bottinelli
,
S.
Lira
,
A.
Uccelli
,
A.
Lanzavecchia
,
B.
Engelhardt
,
F.
Sallusto
.
2009
.
C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE
.
Nat. Immunol.
10
:
514
523
.
110
Silva-Vargas
,
V.
,
A. R.
Maldonado-Soto
,
D.
Mizrak
,
P.
Codega
,
F.
Doetsch
.
2016
.
Age-dependent niche signals from the choroid plexus regulate adult neural stem cells
.
Cell Stem Cell
19
:
643
652
.
111
Emerich
,
D. F.
,
S. J.
Skinner
,
C. V.
Borlongan
,
A. V.
Vasconcellos
,
C. G.
Thanos
.
2005
.
The choroid plexus in the rise, fall and repair of the brain
.
Bioessays
27
:
262
274
.
112
Redzic
,
Z.
2011
.
Molecular biology of the blood-brain and the blood-cerebrospinal fluid barriers: similarities and differences
.
Fluids Barriers CNS
8
:
3
.
113
Meeker
,
R. B.
,
K.
Williams
,
D. A.
Killebrew
,
L. C.
Hudson
.
2012
.
Cell trafficking through the choroid plexus
.
Cell Adh. Migr.
6
:
390
396
.
114
Korin
,
B.
,
T. L.
Ben-Shaanan
,
M.
Schiller
,
T.
Dubovik
,
H.
Azulay-Debby
,
N. T.
Boshnak
,
T.
Koren
,
A.
Rolls
.
2017
.
High-dimensional, single-cell characterization of the brain’s immune compartment
.
Nat. Neurosci.
20
:
1300
1309
.
115
Ginhoux
,
F.
,
M.
Prinz
.
2015
.
Origin of microglia: current concepts and past controversies
.
Cold Spring Harb. Perspect. Biol.
7
:
a020537
.
116
Wolf
,
S. A.
,
H. W.
Boddeke
,
H.
Kettenmann
.
2017
.
Microglia in physiology and disease
.
Annu. Rev. Physiol.
79
:
619
643
.
117
Streit
,
W. J.
,
M. B.
Graeber
,
G. W.
Kreutzberg
.
1988
.
Functional plasticity of microglia: a review
.
Glia
1
:
301
307
.
118
Deczkowska
,
A.
,
I.
Amit
,
M.
Schwartz
.
2018
.
Microglial immune checkpoint mechanisms. [Published erratum appears in 2018 Nat. Neurosci. 21: 1137.]
Nat. Neurosci.
21
:
779
786
.
119
Cohen
,
M.
,
O.
Matcovitch
,
E.
David
,
Z.
Barnett-Itzhaki
,
H.
Keren-Shaul
,
R.
Blecher-Gonen
,
D. A.
Jaitin
,
A.
Sica
,
I.
Amit
,
M.
Schwartz
.
2014
.
Chronic exposure to TGFβ1 regulates myeloid cell inflammatory response in an IRF7-dependent manner
.
EMBO J.
33
:
2906
2921
.
120
Xie
,
X.
,
X.
Luo
,
N.
Liu
,
X.
Li
,
F.
Lou
,
Y.
Zheng
,
Y.
Ren
.
2017
.
Monocytes, microglia, and CD200-CD200R1 signaling are essential in the transmission of inflammation from the periphery to the central nervous system
.
J. Neurochem.
141
:
222
235
.
121
Eyo, U. B., J. Peng, M. Murugan, M. Mo, A. Lalani, P. Xie, P. Xu, D. J. Margolis, and L. J. Wu. 2017. Regulation of physical microglia-neuron interactions by fractalkine signaling after status epilepticus. eNeuro. DOI: 10.1523/ENEURO.0209-16.2016.
122
Butovsky
,
O.
,
M. P.
Jedrychowski
,
C. S.
Moore
,
R.
Cialic
,
A. J.
Lanser
,
G.
Gabriely
,
T.
Koeglsperger
,
B.
Dake
,
P. M.
Wu
,
C. E.
Doykan
, et al
.
2014
.
Identification of a unique TGF-β-dependent molecular and functional signature in microglia. [Published erratum appears in 2014 Nat. Neurosci. 17: 1286.]
Nat. Neurosci.
17
:
131
143
.
123
Kierdorf
,
K.
,
M.
Prinz
.
2013
.
Factors regulating microglia activation
.
Front. Cell. Neurosci.
7
:
44
.
124
Keren-Shaul, H., A. Spinrad, A. Weiner, O. Matcovitch-Natan, R. Dvir-Szternfeld, T. K. Ulland, E. David, K. Baruch, D. Lara-Astaiso, B. Toth, et al. 2017. A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169: 1276–1290.e17.
125
Sala Frigerio, C., L. Wolfs, N. Fattorelli, N. Thrupp, I. Voytyuk, I. Schmidt, R. Mancuso, W. T. Chen, M. E. Woodbury, G. Srivastava, et al. 2019. The major risk factors for Alzheimer’s disease: age, sex, and genes modulate the microglia response to Aβ plaques. Cell Rep. 27: 1293–1306.e6.
126
Masuda
,
T.
,
R.
Sankowski
,
O.
Staszewski
,
C.
Böttcher
,
L.
Amann
,
Sagar
,
C.
Scheiwe
,
S.
Nessler
,
P.
Kunz
,
G.
van Loo
, et al
.
2019
.
Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. [Published erratum appears in 2019 Nature 568: E4.]
Nature
566
:
388
392
.
127
Böttcher
,
C.
,
S.
Schlickeiser
,
M. A. M.
Sneeboer
,
D.
Kunkel
,
A.
Knop
,
E.
Paza
,
P.
Fidzinski
,
L.
Kraus
,
G. J. L.
Snijders
,
R. S.
Kahn
, et al
NBB-Psy
.
2019
.
Human microglia regional heterogeneity and phenotypes determined by multiplexed single-cell mass cytometry
.
Nat. Neurosci.
22
:
78
90
.
128
Ajami
,
B.
,
N.
Samusik
,
P.
Wieghofer
,
P. P.
Ho
,
A.
Crotti
,
Z.
Bjornson
,
M.
Prinz
,
W. J.
Fantl
,
G. P.
Nolan
,
L.
Steinman
.
2018
.
Single-cell mass cytometry reveals distinct populations of brain myeloid cells in mouse neuroinflammation and neurodegeneration models
.
Nat. Neurosci.
21
:
541
551
.
129
Mrdjen, D., A. Pavlovic, F. J. Hartmann, B. Schreiner, S. G. Utz, B. P. Leung, I. Lelios, F. L. Heppner, J. Kipnis, D. Merkler, et al. 2018. High-dimensional single-cell mapping of central nervous system immune cells reveals distinct myeloid subsets in health, aging, and disease. [Published erratum appears in 2018 Immunity 48: 599.] Immunity 48: 380–395.e6.
130
Jordão, M. J. C., R. Sankowski, S. M. Brendecke, Sagar, G. Locatelli, Y. H. Tai, T. L. Tay, E. Schramm, S. Armbruster, N. Hagemeyer, et al. 2019. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 363: eaat7554.
131
Van Hove
,
H.
,
L.
Martens
,
I.
Scheyltjens
,
K.
De Vlaminck
,
A. R.
Pombo Antunes
,
S.
De Prijck
,
N.
Vandamme
,
S.
De Schepper
,
G.
Van Isterdael
,
C. L.
Scott
, et al
.
2019
.
A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment
.
Nat. Neurosci.
22
:
1021
1035
.
132
Guerreiro
,
R.
,
A.
Wojtas
,
J.
Bras
,
M.
Carrasquillo
,
E.
Rogaeva
,
E.
Majounie
,
C.
Cruchaga
,
C.
Sassi
,
J. S.
Kauwe
,
S.
Younkin
, et al
Alzheimer Genetic Analysis Group
.
2013
.
TREM2 variants in Alzheimer’s disease
.
N. Engl. J. Med.
368
:
117
127
.
133
Jonsson
,
T.
,
H.
Stefansson
,
S.
Steinberg
,
I.
Jonsdottir
,
P. V.
Jonsson
,
J.
Snaedal
,
S.
Bjornsson
,
J.
Huttenlocher
,
A. I.
Levey
,
J. J.
Lah
, et al
.
2013
.
Variant of TREM2 associated with the risk of Alzheimer’s disease
.
N. Engl. J. Med.
368
:
107
116
.
134
Kleinberger
,
G.
,
Y.
Yamanishi
,
M.
Suárez-Calvet
,
E.
Czirr
,
E.
Lohmann
,
E.
Cuyvers
,
H.
Struyfs
,
N.
Pettkus
,
A.
Wenninger-Weinzierl
,
F.
Mazaheri
, et al
.
2014
.
TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis
.
Sci. Transl. Med.
6
:
243ra86
.
135
Jay
,
T. R.
,
C. M.
Miller
,
P. J.
Cheng
,
L. C.
Graham
,
S.
Bemiller
,
M. L.
Broihier
,
G.
Xu
,
D.
Margevicius
,
J. C.
Karlo
,
G. L.
Sousa
, et al
.
2015
.
TREM2 deficiency eliminates TREM2+ inflammatory macrophages and ameliorates pathology in Alzheimer’s disease mouse models
.
J. Exp. Med.
212
:
287
295
.
136
Deczkowska
,
A.
,
H.
Keren-Shaul
,
A.
Weiner
,
M.
Colonna
,
M.
Schwartz
,
I.
Amit
.
2018
.
Disease-associated microglia: a universal immune sensor of neurodegeneration
.
Cell
173
:
1073
1081
.
137
Norris
,
G. T.
,
J.
Kipnis
.
2019
.
Immune cells and CNS physiology: microglia and beyond
.
J. Exp. Med.
216
:
60
70
.
138
Carmona
,
S.
,
K.
Zahs
,
E.
Wu
,
K.
Dakin
,
J.
Bras
,
R.
Guerreiro
.
2018
.
The role of TREM2 in Alzheimer’s disease and other neurodegenerative disorders
.
Lancet Neurol.
17
:
721
730
.
139
Krasemann, S., C. Madore, R. Cialic, C. Baufeld, N. Calcagno, R. El Fatimy, L. Beckers, E. O’Loughlin, Y. Xu, Z. Fanek, et al. 2017. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47: 566–581.e9.
140
Konishi
,
H.
,
H.
Kiyama
.
2018
.
Microglial TREM2/DAP12 signaling: a double-edged sword in neural diseases
.
Front. Cell. Neurosci.
12
:
206
.
141
Leyns
,
C. E. G.
,
J. D.
Ulrich
,
M. B.
Finn
,
F. R.
Stewart
,
L. J.
Koscal
,
J.
Remolina Serrano
,
G. O.
Robinson
,
E.
Anderson
,
M.
Colonna
,
D. M.
Holtzman
.
2017
.
TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy
.
Proc. Natl. Acad. Sci. USA
114
:
11524
11529
.
142
Jay
,
T. R.
,
A. M.
Hirsch
,
M. L.
Broihier
,
C. M.
Miller
,
L. E.
Neilson
,
R. M.
Ransohoff
,
B. T.
Lamb
,
G. E.
Landreth
.
2017
.
Disease progression-dependent effects of TREM2 deficiency in a mouse model of Alzheimer’s disease
.
J. Neurosci.
37
:
637
647
.
143
Deming
,
Y.
,
Z.
Li
,
B. A.
Benitez
,
C.
Cruchaga
.
2018
.
Triggering receptor expressed on myeloid cells 2 (TREM2): a potential therapeutic target for Alzheimer disease?
Expert Opin. Ther. Targets
22
:
587
598
.
144
Simard
,
A. R.
,
S.
Rivest
.
2006
.
Neuroprotective properties of the innate immune system and bone marrow stem cells in Alzheimer’s disease
.
Mol. Psychiatry
11
:
327
335
.
145
Boissonneault
,
V.
,
M.
Filali
,
M.
Lessard
,
J.
Relton
,
G.
Wong
,
S.
Rivest
.
2009
.
Powerful beneficial effects of macrophage colony-stimulating factor on beta-amyloid deposition and cognitive impairment in Alzheimer’s disease
.
Brain
132
:
1078
1092
.
146
Thériault
,
P.
,
A.
ElAli
,
S.
Rivest
.
2015
.
The dynamics of monocytes and microglia in Alzheimer’s disease
.
Alzheimers Res. Ther.
7
:
41
.
147
Herz
,
J.
,
A. J.
Filiano
,
A.
Smith
,
N.
Yogev
,
J.
Kipnis
.
2017
.
Myeloid cells in the central nervous system
.
Immunity
46
:
943
956
.
148
Li
,
Q.
,
B. A.
Barres
.
2018
.
Microglia and macrophages in brain homeostasis and disease
.
Nat. Rev. Immunol.
18
:
225
242
.
149
Lampron
,
A.
,
P. M.
Pimentel-Coelho
,
S.
Rivest
.
2013
.
Migration of bone marrow-derived cells into the central nervous system in models of neurodegeneration
.
J. Comp. Neurol.
521
:
3863
3876
.
150
Hjorth
,
E.
,
M.
Zhu
,
V. C.
Toro
,
I.
Vedin
,
J.
Palmblad
,
T.
Cederholm
,
Y.
Freund-Levi
,
G.
Faxen-Irving
,
L. O.
Wahlund
,
H.
Basun
, et al
.
2013
.
Omega-3 fatty acids enhance phagocytosis of Alzheimer’s disease-related amyloid-β42 by human microglia and decrease inflammatory markers
.
J. Alzheimers Dis.
35
:
697
713
.
151
Yu
,
Y.
,
Q.
Shen
,
Y.
Lai
,
S. Y.
Park
,
X.
Ou
,
D.
Lin
,
M.
Jin
,
W.
Zhang
.
2018
.
Anti-inflammatory effects of curcumin in microglial cells
.
Front. Pharmacol.
9
:
386
.
152
Lue
,
L. F.
,
Y. M.
Kuo
,
T.
Beach
,
D. G.
Walker
.
2010
.
Microglia activation and anti-inflammatory regulation in Alzheimer’s disease
.
Mol. Neurobiol.
41
:
115
128
.
153
Moussa
,
C.
,
M.
Hebron
,
X.
Huang
,
J.
Ahn
,
R. A.
Rissman
,
P. S.
Aisen
,
R. S.
Turner
.
2017
.
Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease
.
J. Neuroinflammation
14
:
1
.
154
Karran
,
E.
,
M.
Mercken
,
B.
De Strooper
.
2011
.
The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics
.
Nat. Rev. Drug Discov.
10
:
698
712
.
155
Castello
,
M. A.
,
S.
Soriano
.
2014
.
On the origin of Alzheimer’s disease. Trials and tribulations of the amyloid hypothesis
.
Ageing Res. Rev.
13
:
10
12
.
156
Selkoe
,
D.
2019
.
β-secretase inhibitors for Alzheimer’s disease: heading in the wrong direction?
Lancet Neurol.
18
:
624
626
.
157
Ricciarelli
,
R.
,
E.
Fedele
.
2017
.
The amyloid cascade hypothesis in Alzheimer’s disease: it’s time to change our mind
.
Curr. Neuropharmacol.
15
:
926
935
.
158
Dong
,
Y.
,
X.
Li
,
J.
Cheng
,
L.
Hou
.
2019
.
Drug development for Alzheimer’s disease: microglia induced neuroinflammation as a target?
Int. J. Mol. Sci.
20
:
E558
.
159
Mathys
,
H.
,
J.
Davila-Velderrain
,
Z.
Peng
,
F.
Gao
,
S.
Mohammadi
,
J. Z.
Young
,
M.
Menon
,
L.
He
,
F.
Abdurrob
,
X.
Jiang
, et al
.
2019
.
Single-cell transcriptomic analysis of Alzheimer’s disease. [Published erratum appears in 2019 Nature 571: E1.]
Nature
570
:
332
337
.
160
Marsh
,
S. E.
,
E. M.
Abud
,
A.
Lakatos
,
A.
Karimzadeh
,
S. T.
Yeung
,
H.
Davtyan
,
G. M.
Fote
,
L.
Lau
,
J. G.
Weinger
,
T. E.
Lane
, et al
.
2016
.
The adaptive immune system restrains Alzheimer’s disease pathogenesis by modulating microglial function
.
Proc. Natl. Acad. Sci. USA
113
:
E1316
E1325
.
161
Appel
,
S. H.
,
D. R.
Beers
,
J. S.
Henkel
.
2010
.
T cell-microglial dialogue in Parkinson’s disease and amyotrophic lateral sclerosis: are we listening?
Trends Immunol.
31
:
7
17
.
162
Monsonego
,
A.
,
A.
Nemirovsky
,
I.
Harpaz
.
2013
.
CD4 T cells in immunity and immunotherapy of Alzheimer’s disease
.
Immunology
139
:
438
446
.
163
Liu
,
J.
,
Y.
Ma
,
S.
Tian
,
L.
Zhang
,
M.
Zhao
,
Y.
Zhang
,
D.
Xu
.
2014
.
T cells promote the regeneration of neural precursor cells in the hippocampus of Alzheimer’s disease mice
.
Neural Regen. Res.
9
:
1541
1547
.
164
Pardoll
,
D. M.
2012
.
The blockade of immune checkpoints in cancer immunotherapy
.
Nat. Rev. Cancer
12
:
252
264
.
165
Beers
,
D. R.
,
J. S.
Henkel
,
W.
Zhao
,
J.
Wang
,
A.
Huang
,
S.
Wen
,
B.
Liao
,
S. H.
Appel
.
2011
.
Endogenous regulatory T lymphocytes ameliorate amyotrophic lateral sclerosis in mice and correlate with disease progression in patients with amyotrophic lateral sclerosis
.
Brain
134
:
1293
1314
.
166
Pluvinage
,
J. V.
,
M. S.
Haney
,
B. A. H.
Smith
,
J.
Sun
,
T.
Iram
,
L.
Bonanno
,
L.
Li
,
D. P.
Lee
,
D. W.
Morgens
,
A. C.
Yang
, et al
.
2019
.
CD22 blockade restores homeostatic microglial phagocytosis in ageing brains
.
Nature
568
:
187
192
.
167
Griciuc
,
A.
,
A.
Serrano-Pozo
,
A. R.
Parrado
,
A. N.
Lesinski
,
C. N.
Asselin
,
K.
Mullin
,
B.
Hooli
,
S. H.
Choi
,
B. T.
Hyman
,
R. E.
Tanzi
.
2013
.
Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta
.
Neuron
78
:
631
643
.
168
Schlepckow
,
K.
,
G.
Kleinberger
,
A.
Fukumori
,
R.
Feederle
,
S. F.
Lichtenthaler
,
H.
Steiner
,
C.
Haass
.
2017
.
An Alzheimer-associated TREM2 variant occurs at the ADAM cleavage site and affects shedding and phagocytic function
.
EMBO Mol. Med.
9
:
1356
1365
.
169
Schwartz
,
M.
2017
.
Can immunotherapy treat neurodegeneration?
Science
357
:
254
255
.

M.S. is an inventor of the intellectual property that forms the basis for development of PD-L1 immunotherapy for AD.