You are on page 1of 25

Cellular and Molecular Life Sciences (2019) 76:3167–3191

https://doi.org/10.1007/s00018-019-03178-2 Cellular and Molecular Life Sciences

REVIEW

Metalloproteinases and their tissue inhibitors in Alzheimer’s disease


and other neurodegenerative disorders
Santiago Rivera1   · Laura García‑González1 · Michel Khrestchatisky1 · Kévin Baranger1

Received: 22 May 2019 / Revised: 22 May 2019 / Accepted: 29 May 2019 / Published online: 13 June 2019
© Springer Nature Switzerland AG 2019

Abstract
As life expectancy increases worldwide, age-related neurodegenerative diseases will increase in parallel. The lack of effec-
tive treatment strategies may soon lead to an unprecedented health, social and economic crisis. Any attempt to halt the
progression of these diseases requires a thorough knowledge of the pathophysiological mechanisms involved to facilitate
the identification of new targets and the application of innovative therapeutic strategies. The metzincin superfamily of met-
alloproteinases includes matrix metalloproteinases (MMP), a disintegrin and metalloproteinase (ADAM) and ADAM with
thrombospondin motifs (ADAMTS). These multigenic and multifunctional proteinase families regulate the functions of an
increasing number of signalling and scaffolding molecules involved in neuroinflammation, blood–brain barrier disruption,
protein misfolding, synaptic dysfunction or neuronal death. Metalloproteinases and their physiological inhibitors, the tissue
inhibitors of metalloproteinases (TIMPs), are therefore, at the crossroads of molecular and cellular mechanisms that support
neurodegenerative processes, and emerge as potential new therapeutic targets. We provide an overview of current knowledge
on the role and regulation of metalloproteinases and TIMPs in four major neurodegenerative diseases: Alzheimer’s disease,
Parkinson’s disease, amyotrophic lateral sclerosis and Huntington’s disease.

Keywords  Parkinson’s disease · Huntington’s disease · Amyotrophic lateral sclerosis · ADAM · TIMP · Neurodegenerative
brain disease

Abbreviations CSF Cerebrospinal fluid


6-OHDA 6-Hydroxydopamine CTF C-terminal fragment
5xFAD Transgenic mice bearing 5 familial C99 APP-CTF of 99 amino acids
mutations on human App and Psen1 DAPT  N-[N-(3,5-Difluorophenacetyl)-
genes l-alanyl]-S-phenylglycine t-butyl
AD Alzheimer’s disease ester, γ-secretase inhibitor
Aβ Amyloid beta peptide DLB Dementia with Lewy bodies
ADAM A disintegrin and metalloproteinase ECM Extracellular matrix
ADAMTS ADAMs with thrombospondin motifs  FADD Fas-associated protein with death
ALS Amyotrophic lateral sclerosis domain
APOE Apolipoprotein E HD Huntington’s disease
APP Amyloid precursor protein HD-NSCs Neural stem cells from HD patients
BACE-1 Beta-site APP cleaving enzyme 1 HEKswe Human embryonic kidney cells that
BBB Blood–brain barrier express App gene with the Swedish
C3 BACE-1 inhibitor IV mutation
CAA​ Cerebral amyloid angiopathy Htt Huntingtin
CNS Central nervous system ICV Intracerebroventricular
IDE Insulin degrading enzyme
* Santiago Rivera IL-1 Interleukin-1
santiago.rivera@univ‑amu.fr LBs Lewy bodies
LDLR Low-density lipoprotein receptor
1
Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol,
Marseille, France

13
Vol.:(0123456789)

3168 S. Rivera et al.

LRP-1 Low-density lipoprotein receptor- neurodegenerative processes, matrix metalloproteinases


related protein 1 (MMPs) are of particular interest because: (1) MMPs are
LRRK2 Leucine-rich repeat kinase 2 expressed by all types of central nervous system (CNS) cells;
LTP Long-term potentiation (2) MMPs expression is modulated by potentially neuro-
MCP-1 Monocyte chemoattractant protein 1 toxic proteins, which can in turn undergo cleavage and func-
MMP Matrix metalloproteinase tional regulation by these same MMPs; (3) MMPs modulate
MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydro- chronic neuroinflammation through cleavage of inflamma-
pyridine tory mediators (i.e., cytokines and chemokines) and tight
MSC Mesenchymal stem cells junction proteins that ensure blood–brain barrier (BBB)
MT-MMP Membrane-type matrix impermeability; (4) MMPs control the processing of extra-
metalloproteinase cellular matrix (ECM) and transmembrane proteins involved
MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-di- in cell–cell interactions; (5) MMPs can initiate or be part of
phenyl tetrazolium bromide proteolytic cascades that amplify cellular responses in a web
NFT Neurofibrillary tangles of interactions between numerous enzymes and substrates.
NSC Neural stem cell In this review, we will discuss the most relevant knowl-
NTF N-terminal fragment edge on MMPs and their physiological inhibitors, the tissue
PD Parkinson’s disease inhibitors of metalloproteinases (TIMPs), in four neurode-
PGRN Progranulin generative diseases that strike in mid–late-life: Alzheimer’s
Psen 1 and Psen 2 Presenilin 1 and 2 disease (AD), Parkinson’s disease (PD), amyotrophic lat-
RAGE Receptor for advanced glycation end eral sclerosis (ALS) and Huntington’s disease (HD). We will
products also discuss to a lesser extent the contribution of two other
sAPPα/β Soluble APPα/β metalloproteinase subfamilies, which, like MMPs, belong
TIMP Tissue inhibitor of metalloproteinases to the superfamily of metzincins: a disintegrin and metal-
TNF-α Tumour necrosis factor α loproteinase (ADAM) and ADAMs with thrombospondin
TREM2 Triggering receptor expressed on motifs (ADAMTS). While ADAMs are membrane-anchored
myeloid cells 2 enzymes, chiefly considered as sheddases of membrane
proteins, ADAMTS are mostly extracellular matrix (ECM)-
degrading enzymes. ADAMs and ADAMTS in the nervous
Introduction system are extensively discussed in this CMLS special issue
by Hsia and colleagues [1] and elsewhere [2].
Humans have not evolved to last long in nature. As the forces
of natural selection disappear with the improvement of life
and health conditions, average life expectancy has exceeded Metalloproteinases and natural tissue
80 years in many countries of the world. The concomitant inhibitors of metalloproteinases
consequence is the soaring of aging-related neurodegenera-
tive disorders that cannot be cured or significantly slowed Matrix metalloproteinases
down, thereby affecting millions of patients and even more
caregivers. These disorders generally start with mild symp- Twenty-four proteinases constitute the MMP family in
toms that alter motor, behavioural and cognitive functions. humans. According to their structure and substrate specific-
The gradual loss of large amounts of neurons inevitably ity, this multigenic family have been traditionally classified
leads to a worsening of these symptoms, which often end into five categories: gelatinases (-2 and -9), stromelysins
up with a loss of global autonomy before the patient’s death. (MMP-3 and -10), collagenases (MMP-1, -8 and -13), mem-
Neurodegenerative diseases are associated with genetic and brane-type MMPs (MT-MMPs, MMP-14, -15, -16, -17, -24
environmental stimuli that act in synergy to elicit disease- and -25) and the other MMPs, e.g., MMP-7, MMP-12 or
specific dysfunctions, but also share fundamental common- MMP-28 (see for reviews [2–4]). Widely expressed in all
alities: free radicals generation, mitochondrial dysfunction, human cells, MMPs have significant structural homology
immunosenescence, protein misfolding or inflammation. and this is one of the main reasons that hinder the develop-
The confluence of these pathogenic driving forces makes ment of specific inhibitors against each of them. All MMPs
it difficult to identify a single cause that could be more eas- possess a 17–29 amino acid hydrophobic signal peptide that
ily targeted. It is of the utmost importance to elucidate the targets the proteinase to the secretory pathway, followed by
underlying mechanisms of the pathology to clearly iden- a pro-domain sequence of 77–87 amino acids that interacts
tify molecular targets and help design effective therapeu- with a conserved cysteine residue and the Z ­ n2+ cation of
tic strategies. Among the biofactors that can contribute to the catalytic site, thereby maintaining the enzyme under

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3169

a zymogen inactive state. The catalytic domain of ~ 170 general, TIMPs bind all MMPs and some ADAMs, but
amino acids, harbours a ­Zn2+ binding consensus sequence only TIMP-3 appears to have inhibitory activity against
HExxHxxGxxH with three histidine residues that confer ADAMTS [15]. The N-terminal domain of TIMPs is suf-
­ n2+. Redox-mediated break-up of the interac-
stability to Z ficient to inhibit MMPs, but in the case of ADAM10,
tion between the cysteine and the metal cation as well as TIMP-1 and TIMP-3 also require the interaction of their
propeptide removal by autocatalysis or by other proteinases, C-terminal domain with the proteinase. ADAM10 is not
triggers the conversion of inactive pro-MMPs into active inhibited by TIMP-2 and -4 [16]. The binding affinities
MMPs by a mechanism known as “cysteine switch”. Down- between TIMPs and MMPs or their modes of interaction
stream of the catalytic site, a short hinge region followed differ significantly. For instance, TIMP-1 inhibits soluble
by the hemopexin domain (~ 200 amino acids) exhibits a MMPs and ADAM10/17, but it is a very poor inhibitor of
relatively variable amino acid sequence, which is crucial for MT-MMPs. Also, TIMP-1 is able to specifically bind the
proteinase-substrate specificity. Of note, MMP-7, MMP-23 hemopexin domain of pro-MMP-9, although the biological
and MMP-26 lack the hemopexin domain. In addition to this function of this complex is unknown [17]. TIMP-2 inhibits
multidomain common structure, the six members of the MT- both soluble and membrane-bound MMPs. In addition,
MMP subfamily contain membrane anchoring domains; a TIMP-2 specifically binds to the hemopexin domain of
glycosylphosphatidylinositol in the case of MMP-17 (MT4- pro-MMP-2, paradoxically leading to the activation of the
MMP) and MMP-25 (MT6-MMP) or a transmembrane enzyme on the plasma membrane; indeed, the N-terminal
domain with a short intracytoplasmic moiety in the case of domain of TIMP-2 interacts with MT1-MMP, whereas
MMP-14 (MT1-MMP), MMP-15 (MT2-MMP), MMP-16 the C-terminal domain binds to the hemopexin domain of
(MT3-MMP) and MMP-24 (MT5-MMP). Moreover, MT- pro-MMP-2, thus fixing the pro-MMP-2 on the membrane
MMPs, MMP-11, MMP-23 and MMP-28 contain a specific and allowing its activation by cleavage of the pro-peptide
di-basic sequence at the end of the pro-domain, which is by an adjacent MT1-MMP [18]. TIMP-3 inhibits MMPs,
recognized by the intracellular ­Ca2+-dependent serine pro- ADAMs and ADAMTS. TIMP-4 has the same inhibitory
teinase furin, leading to activation of these enzymes in the profile as TIMP-2. All TIMPs are ubiquitously distributed,
trans-Golgi network [4]. with the exception of TIMP-4, which is predominantly
MMPs have been usually considered as ECM-degrading expressed in heart, ovary and brain [19].
enzymes. This view is now changing with the growing Timp-1, Timp-3 and Timp-4 genes are nested in introns of
number of non-matrix substrates identified among signal- genes coding for synaptic proteins synapsins and Timp-2 is
ling molecules, trophic factors, receptors, cell adhesion close to the synapsin IV gene [20–22]. The intriguing genetic
molecules, and even nuclear proteins [5–7]. It must be link between TIMPs and synapsins likely underlines func-
emphasized that MMPs are more and more involved in the tional relations of these proteins in the nervous system that
modulation of neuroinflammatory processes common to need to be explored, as many neuropathological conditions
many nervous system pathologies through the regulation of: show significant alterations of their expression.
(1) ECM proteolysis [8]; (2) cell proliferation, differentia-
tion and migration [9–11]; (3) activation and inactivation of
inflammatory mediators [12]; (4) BBB permeability [13].
If we add that proteolytic-independent effects of MMPs are Alzheimer’s disease
beginning to be unveiled [14], we can conclude that MMPs
represent a paradigm of extraordinary and fascinating func- Alzheimer’s disease (AD) is the most common type of neu-
tional diversity. rodegenerative disorder for which we still lack efficient cur-
ing, slowing or preventing treatments. Prototypic symptoms
Tissue inhibitors of metalloproteinases of AD start with short-term mild memory deficits that dete-
riorate with time, progressively leading to severe memory
TIMPs are pleiotropic secreted proteins, mostly known for loss, changes in mood, anxiety, depression, agitation, spatial
their reversible MMP inhibitory activities. The four TIMPs disorientation, motor impairment, and finally dementia. AD
(TIMP-1 to TIMP-4) share 40% of sequence homology affects 45 million people currently worldwide and this figure
and a backbone with twelve conserved cysteine residues is expected to rise to 131 million in 2050 unless efficient
and six disulfide bonds that are essential for their biologi- treatments are found in the meantime [23]. Only a handful of
cal activities. Besides the signal peptide, TIMPs structure available drugs can transiently and moderately limit clinical
comprises a N-terminal inhibitory domain of MMPs and symptoms in the early stages of the disease (https:​ //www.alz.
a C-terminal domain of interaction. TIMP-1, -2 and -4 org/alzhe​imers​-demen​tia/resea​rch_progr​ess/treat​ment-horiz​
are found as soluble or cell surface-associated proteins, on). The difficulty in finding effective treatments for AD is
whereas TIMP-3 preferentially binds to ECM proteins. In due to a multitude of factors, including:

13

3170 S. Rivera et al.

• The multifactorial etiology of AD. Familial forms of the APP cleavage by β- and γ-secretases occurs following APP
disease represent ~ 1% of cases and are caused by muta- endocytosis into the endosomes or during APP targeting to
tions in 3 genes: amyloid precursor protein (App), and the cell surface through the trans-Golgi network, thereby
presenilins 1 (Psen1) and 2 (Psen2). The overwhelming highlighting the importance of APP cellular trafficking in
majority of patients suffer from sporadic forms of the amyloidogenesis (reviewed in [29]). Aβ accumulation results
disease of unknown etiology, where genetic and environ- from a balance between its formation and its degradation/
mental risk factors likely synergize with age to prepare a clearance. Several metalloproteinases, including insulin
fertile ground for the development of the disease. degrading enzyme (IDE), angiotensin converting enzyme,
• The difficulty of successfully reproducing valuable pre- endothelin converting enzyme and neprilysin, play a prime
clinical data in human clinical trials. role in Aβ degradation (reviewed in [30]). Moreover, some
• Comorbidity with other disorders/injuries may add con- MMPs (i.e., MMP-2, -3, -9 and MT1-MMP) have also been
founding factors from an etiological standpoint. reported to display Aβ-degrading activity (see [3]). Com-
• A relatively poor knowledge of the pathophysiological plementary to proteolytic degradation, the neurovascular
mechanisms involved. To date, most research efforts have hypothesis of AD suggests that tuning down Aβ levels in
focused on studying two pathological characteristics of the brain parenchyma can also be achieved by scavenging
the disease: the accumulation of the amyloid beta peptide systems mostly operating in the BBB microenvironment.
(Aβ) resulting from the proteolytic processing of APP, Several reports indicate that one of these systems is the
and the hyperphosphorylation of the tau protein (p-tau). apolipoprotein E (APOE) receptor, low-density lipoprotein
Unfortunately, the results of clinical trials targeting Aβ receptor (LDLR). Thus, overexpression of LDLR in the
or p-tau have been well below expectations [24–26], mouse brain limits amyloid plaque formation and enhances
prompting researchers to discover alternative targets. Aβ clearance from brain to blood [31, 32], as well as Aβ
• Clinical trials are conducted on older people after diag- uptake and degradation by astrocytes through a mechanism
nosis, but AD starts 10 or 15 years earlier, meaning that where Aβ directly binds to LDLR, even in the absence of
treatments against relevant targets (e.g., Aβ, p-tau) occur APOE [33]. The low-density lipoprotein receptor-related
too late, when the aging brain has already lost much of protein 1 (LRP-1), mainly expressed by glial and endothe-
its healing ability and resilience. lial cells, also contributes to Aβ clearance. This is confirmed
by experiments in which the decrease/abrogation of LRP-1
Despite the diversity of potentially pathogenic mecha- expression using antisense oligonucleotides [34] or LRP-
nisms in AD, metalloproteinases have been mainly involved 1-conditional knockout [35], interferes with Aβ clearance
in the metabolism of APP and Aβ. It is in this context that via the BBB, leading to its accumulation in the brain and the
we will address the following sections. consequent learning deficits. Selective knockdown of LRP-1
in astrocytes also reduces Aβ uptake and degradation, which
Main pathogenic pathways linked to APP processing correlates with a downregulation of major Aβ-degrading
and amyloidogenesis enzymes MMP-2, MMP-9 and IDE [36]. Unlike, LDLR and
LRP-1, the receptor for advanced glycation end products
Almost 30 years after its publication, the amyloid hypothesis (RAGE) promotes Aβ influx from blood-to-brain (reviewed
of AD remains a valuable tool to study the underlying patho- in [37]). Accordingly, silencing or eliminating RAGE has
physiological mechanisms, despite the failure of clinical tri- been shown to preserve BBB integrity in vitro [38] and
als based on its principles. The amyloid hypothesis essen- reduce brain Aβ levels in transgenic AD mice by decreas-
tially asserts that excessive accumulation of Aβ is a major ing β- and γ-secretase activities [39].
pathogenic determinant (see for an updated review [27]). Aβ It is clear that neurotoxicity derived from APP cleavage
results from sequential cleavage of APP by two canonical can no longer be considered exclusively on the basis of Aβ
secretases; β-secretase (beta-site APP cleaving enzyme-1, accumulation. Other APP metabolites generated by different
BACE-1) generates a transmembrane fragment of 99 amino proteinases (e.g., MT-MMPs, δ-secretase) also contribute to
acids (C99), which is then cleaved by the γ-secretase com- Alzheimer’s pathology (reviewed in [40]), paving the way
plex, thus releasing 4 kDa Aβ monomers, mainly 40 (Aβ40) for an assessment of APP breakdown products as compo-
and 42 (Aβ42) amino acids. Aβ42 aggregates more easily nents of pathogenic pathways, and therefore, as potential
than Aβ40 and it is, therefore, more toxic. Monomers can new therapeutic targets.
assemble into oligomers, protofibrils and fibrils, which will
eventually give rise to amyloid plaques. Although the lat- Non‑pathogenic pathways linked to APP processing
ter are a hallmark of AD and provide bona fide post mor-
tem diagnostic, small size oligomers (e.g., dimers, trimers) In physiological conditions, APP is mainly processed at
are increasingly seen as the most toxic forms of Aβ [28]. the plasma membrane level by α-secretases of the ADAM

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3171

family. This subfamily of metzincins present structural and exhibit increased levels of sAPPα associated with enhanced
functional similarities with MMPs (see for review [2]). LTP and enhanced learning and memory capabilities [58].
ADAM10 is the main physiological/constitutive α-secretase Other authors have corroborated in  vitro the ability of
[41, 42], while ADAM17 has been identified as responsi- MMP-9 to cleave soluble Aβ at the α-site, but the enzyme
ble of the regulated α-secretase activity [43]. α-secretase seems to be more efficient at cleaving in the C-terminus with
cleavage in the middle of the Aβ sequence prevents its for- decreasing processing efficiency towards the N-terminus to
mation and concomitantly generates a soluble N-terminal generate fragments ending at positions 34, 33, 30, 23, 20 and
APP fragment (sAPPα) with neurotrophic and neuroprotec- 16 [59, 60]. In addition, MMP-9 exhibits a rather uncom-
tive properties [44, 45]. Aβ oligomers stimulate transient mon feature among Aβ-degrading enzymes because it can
increase of sAPPα levels and α-secretase activity in cultured cleave preformed Aβ fibrils and compact amyloid plaques, as
cortical neurons, which could be interpreted as a cell self- shown on brain slices from APP/PS1 transgenic mice [59].
defence mechanism [46]. While there is little doubt about the This further adds to the possibility that promoting MMP-9
neurotoxicity of Aβ accumulation, the physiological roles activity may hold therapeutic potential. However, this pos-
of the peptide are still poorly understood. Several pieces tulate is subject to caution as it cannot be excluded that
of evidence converge to show that Aβ is part of the innate proteolytic degradation of plaques could release toxic Aβ
immunity arsenal to fight against bacterial, fungal and viral assemblages and other neurotoxins contained in their mesh.
pathogens in the brain [47–50]. Moreover, physiological Aβ MMP-2 presents Aβ-degrading profiles roughly similar to
is involved in maintaining basal neurotransmitter release and those reported for MMP-9, although MMP-2 appears to be
synaptic activity [51]. It has been shown that picomolar con- more efficient in cleaving Aβ42 [60].
centrations of Aβ are sufficient to stimulate learning and Reactive astrocytes produce MMP-2 and MMP-9 and are
memory in healthy rodents as well as long-term potentiation systematically associated with Aβ deposits in transgenic
(LTP)—a form of synaptic plasticity considered to be a cel- mouse models of AD and in post mortem AD brains. In
lular substrate for learning—through the activation of nico- support of a functional link between both observations, the
tinic receptors [52–54]. N-terminal fragments of Aβ found in incubation of recombinant Aβ in conditioned media from
the cerebrospinal fluid (CSF) of healthy and AD individuals cultured mouse astrocytes yields breakdown Aβ C-terminal
appear to be determinant for these physiological effects, with fragments, which can be prevented by selective chemical
the YEVHHQ motif located before the α-cleaving site in the inhibitors of MMP-2 and MMP-9, and by TIMP-1 [61].
Aβ sequence playing a prominent role [55]. It has also been Furthermore, MMP-9 and MMP-2 knockout mice exhibit
shown that cGMP nucleotide stimulates the release of Aβ higher levels of Aβ in the brain compared to wild-type mice,
by facilitating the approximation in endolysosomal compart- thereby supporting that both MMPs degrade Aβ in vivo [61].
ments between APP and BACE-1, the main β-secretase. The In the same vein, a recent study conducted to investigate
blockade of Aβ with antibodies in these conditions inter- the impact of physical activity on Alzheimer’s pathology
feres with cGMP-induced enhancement of LTP and memory reported that exercise training in Tg2576 transgenic mice
[56]. Taken together, these data indicate that the proteolytic significantly decreases the levels of Aβ40 in the cortex, and
regulation of Aβ transcends the idea of its neurotoxicity and Aβ42 in the cortex and hippocampus. Of note, decreased
raises the question of the potential clinical consequences of Aβ concentration was concomitant with a rise in the lev-
lowering Aβ levels below a physiological threshold. els of five proteins involved in Aβ clearance, among which
MMP-9 [62].
MMPs are mainly considered as Aβ‑degrading In addition to MMP-2 and MMP-9, other MMPs can also
enzymes, but not only degrade Aβ. For instance, metal ligand clioquinol stimulates
Aβ degradation in a cell culture system through a mecha-
MMPs with Aβ‑degrading activity might be beneficial nism involving the activation of MMP-3 [63]. MMP-7 was
recently reported to cleave Aβ40 and Aβ42, a process that
Pioneer work in the 90’s linked for the first time AD with is inhibited by binding of Aβ to C ­ u2+ [64]. Concerning
MMPs. Tökés and collaborators found upregulated expres- membrane-anchored MMPs, it was shown that COS cells
sion levels of inactive pro-MMP-9 in hippocampal neurons overexpressing MT1-MMP degrade exogenous soluble Aβ40
of post mortem AD patients. They also demonstrated by and Aβ42, and release 6 different Aβ-derived peptides with
mass spectrometry the ability of active MMP-9 to cleave cleavage sites mostly located around the α-site, between
Aβ40 on 3 sites of the membrane-spanning domain, but also residues 12 and 18 of the Aβ sequence. Moreover, a soluble
at ­Lys16–Leu17, the so-called α-cleavage site [57]. In the first variant of MT1-MMP lacking the transmembrane domain
case, MMP-9 could prevent the formation of the neurotoxic was able to degrade in situ parenchymal amyloid plaques
β-sheet. In the second, MMP-9 could act as an α-secretase. in brain slices of Tg2576 mice [65], a feature shared only
Consistent with this idea, MMP-9 overexpressing mice with MMP-9, neprilysin and IDE among metalloproteinases.

13

3172 S. Rivera et al.

MMPs can be pathogenic in connection with inflammatory by diapedesis or through a disrupted BBB. Evidences in this
processes direction are rather limited at the moment. The monocytic
cell line THP-1 treated with Aβ42 releases more MMP-9,
Although the degrading action of MMPs on Aβ could in along with tumour necrosis factor (TNF-α), interleukin-
principle be considered beneficial, MMPs are multifunc- 1beta (IL-1β) and monocyte chemoattractant protein-1
tional enzymes, whose roles may depend not only on the cell (MCP-1), compared to untreated cells [74]. Purified neu-
types that express them, but also on the substrates involved trophils dramatically increase the production of MMP-9
or the pathological and spatio-temporal context (see for in response to incubation with highly toxic Aβ25-35 [75].
review [2]). In the case of BBB disruption, MMPs have a Interestingly, neutrophils infiltrate the brain of AD patients
prominent role as they can proteolyse major components of and are also observed in the brain parenchyma of transgenic
the basement membranes and tight junctions (see for review AD mice in areas with Aβ deposition [76].
[66] in this special CMLS issue). The impermeability of
BBB, which ensures selective exchanges between the cen- Regulation of MMPs in Alzheimer’s animal models
tral and peripheral nervous systems, is compromised in AD and Alzheimer’s patients
patients [67] and in transgenic AD mice [68]. This may con-
tribute to inflammatory and neurodegenerative processes, The data above convincingly show that Aβ is a substrate
probably with the participation of resident brain immune of MMPs. In turn, Aβ modulates the expression of MMPs,
cells, as suggested by a study using a co-culture of endothe- probably revealing the existence of feedback regulatory
lial cells and astrocytes exposed to Aβ42. In this work, mechanisms. Since early work by Gottschall and collabo-
astrocyte-induced BBB opening was related to the activa- rators showing that Aβ40 could trigger the expression of
tion of MMP-9 and the concomitant decrease in claudin-5, MMP-2, -3 and -9 in rat mixed neuronal/astrocyte cultures
a MMP-9 substrate that contributes to the maintenance of [77], other authors have confirmed these data. Thus, astro-
functional tight junctions [69]. Another important source cytes surrounding Aβ plaques in transgenic mice brains
of MMP, reactive microglia, may stimulate Aβ accumula- show increased levels of MMP-2 and -9 [61]. The increase
tion through a MMP-9-dependent mechanism. In this case, in the mRNA of both gelatinases is already detected at
microglial MMP-9 generates C-terminal truncated Aβ frag- 4 months of age in the hippocampus of 5xFAD mice, and
ments that interfere with Aβ clearance across the BBB and even earlier (2 months) in some astrocytes close to incipient
promote cerebral aggregation of intracranial-injected Aβ42 amyloid plaques [78]. On the other hand, MMP-9 immu-
in wild-type mice. It is noteworthy that the seeding prop- nostaining is upregulated in the soma and dendrites of cor-
erties of these C-terminal fragments generated by MMP-9 tical neurons in 2-month-old 5xFAD mice [78], indicating
could, therefore, contribute to the development of sporadic altogether that the upregulation of MMP-2 and MMP-9 in
forms of the disease [70]. Recently, another report pointed Aβ enriched areas starts at pre-symptomatic stages of the
out that disruption of the blood–CSF barrier 6 h after intrac- pathology and persists over the prodromal-like and symp-
erebral injection of Aβ oligomers was linked to conspicuous tomatic phases, as they have been defined in the 5xFAD
inflammation, and was prevented by broad spectrum MMP model [79]. Whether this conveys beneficial or detrimental
inhibitor GM6001 and by MMP-3 deficiency in mice, sug- effects cannot be systematically anticipated, as functional
gesting that the control of MMP activity may have therapeu- duality is a characteristic of both gelatinases. These might
tic potential [71]. Likewise, Aβ42 oligomers dowregulated be beneficial based on their Aβ-degrading properties, but
tight junction scaffold proteins in an in vitro BBB model they might also promote BBB disruption and neuroinflam-
using bEnd.3 endothelial cells. The weakening effects of mation [80] or exert direct neurotoxicity in the hippocampus,
oligomers on BBB were blocked by either knocking down as reported for MMP-9 [81]. Along this line, AβE22Q and
RAGE, which was induced by Aβ42, or by GM6001 treat- AβL34V mutations associated with hemorrhagic symptoms
ment, which inhibited the activity of concomitantly upregu- in early onset CAA stimulate the expression and activation
lated MMP-2 and MMP-9 [38]. GM6001 and another MMP of MMP-2 by endothelial cells in a rather selective manner,
inhibitor, minocycline, also  proved to efficiently reduce which is linked to possible BBB dysfunctions [82]. Moreo-
inflammation and oxidative stress associated with cerebral ver, Mizoguchi and collaborators demonstrated that intracer-
amyloid angiopathy (CAA) in AD mice [72]. ebroventricular (ICV) injections of Aβ upregulated MMP-9
Blood-derived immune cells (e.g., macrophages, lympho- expression in neurons and astrocytes and triggered cogni-
cytes, granulocytes) are suspected to influence AD patho- tive deficits. Such deficits were efficiently prevented in mice
genesis, but the extent of their contribution to disease and lacking MMP-9 and also after treatment with chemical MMP
the underlying mechanisms remain elusive (for review [73]). inhibitors [83]. In addition to the extensive work on gelati-
The question arises as to the role of immune cell MMPs, nases, other MMPs have also attracted attention in the field.
provided that these cells can reach the brain parenchyma MT1-MMP content was found increased in neurons and

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3173

amyloid plaques of 5xFAD mice at 6 months of age—dur- elevated plasma levels of MMP-3, its substrate gelsolin was
ing the symptomatic phase of the pathology—this increase decreased in these samples, suggesting that the combined
affecting both the active and inactive forms of MT1-MMP use of gelsolin and MMP-3 could, therefore, hold potential
[78]. Other authors have shown that MT1-MMP expression as a plasma biomarker in AD [98]. Even accepting that cor-
is mainly associated with reactive microglial cells in 5xFAD relative data do not allow for a causal link between MMP
mice [84] and reactive astrocytes in Tg-SwDI transgenic AD levels and disease progression, it has been proposed that pro-
mice [65]. Morevoer, Aβ42 increases the mRNA levels of teolysis limits the amount of circulating Aβ that could reach
MMP-3, -12 and -13 in a microglial cell line and in primary the brain. This idea is based on the ability of MMPs and
murine microglia in a PI3/Akt dependent manner [85]. The ADAMs (e.g., MT1-MMP, MMP-9, ADAM10, ADAM12
expression of MMP-1, for which no involvement in APP/Aβ and ADAM17) to shed RAGE or LRP-1 [99–103], with the
metabolism is known, correlates with neuronal degeneration consequent release of the soluble truncated receptors that
in the rat hippocampus 1 month after ICV injection of Aβ25- can bind circulating Aβ; it is estimated that 70% of circulat-
35 [86]. Taken together, these data consistently indicate the ing Aβ40 and 90% of Aβ42 is bound to soluble LRP-1 [104].
upregulation of MMP levels after exposure to exogenous Most reports associate MMPs to APP metabolism in AD,
Aβ or in the vicinity of Aβ deposits in transgenic mice, with however, recent work also unveils the possibility of func-
spatial distributions that may vary depending on the cell type tional links between MMPs and tau protein. For example,
or animal model studied. increased levels of active MMP-2 colocalize with p-tau in
Studies in humans mostly reflect the upward trend in neurofibrillary tangles (NFT) and dystrophic neurites in
MMP levels, although some declines have been noted. the entorhinal cortex of AD patients at early disease stages
Human cerebrovascular smooth muscle cells, constituents [105]. The same study reports that MMP-2 can cleave
of the BBB, show increased expression of MMP-2 and its recombinant tau in vitro, but fails to cleave tau contained in
activating enzyme MT1-MMP after Aβ treatment in culture, NFT, leading to the speculation that inactivation of MMP-2
which eventually results in increased MMP-2 activity and in NFT might prevent the formation of toxic truncated tau
reduced cell viability [87]. Oligomers of Aβ42 positively assemblies. MMP-9 and MMP-3 are also described as tau-
regulate the expression of several MMPs (i.e., MMP-1, -3, degrading enzymes, but only MMP-9 can generate fragments
-9, -10, -12 and -19) in microglia isolated from the brain prone to generate tau oligomers, which are increasingly con-
of AD patients post mortem, compared to controls without sidered as the most toxic forms of tau [106]. MMP-3 and
dementia [88]. ELISA assay revealed MMP-1 levels signifi- MMP-10 levels correlate with tau or p-tau levels in the CSF
cantly elevated by more than 50% in different cortical areas of AD patients, further stressing the potential interest of
of AD patients [89]. Also in post mortem brain tissue, MT5- MMPs as biomarkers of AD pathology [97, 107].
MMP immunostaining was detected in amyloid plaques [90] Overall, these data illustrate a wide range of mechanisms
and MT1-MMP in microglial cells neighbouring plaques by which MMPs can have beneficial or detrimental effects
[84], while MMP-3 was found in both amyloid plaques and on AD. Particular mention is given in the following section
the interstitium between myelinated axons and astrocytes, in to MT-MMPs, which are relatively new enzymes in the field
particular, in the parietal cortex [91]. The concentrations of of AD and open new avenues for a better understanding of
MMP-3 are higher in the CSF of AD patients compared to pathophysiology.
healthy controls, in contrast with MMP-9 whose levels are
lowered in the AD condition [92]. Similarly, Horstmann and Alternative processing of APP by MT‑MMPs
collaborators reported an increase in the levels of MMP-3 and contribution to Alzheimer’s pathogenesis
in CSF and plasma of AD patients, whereas MMP-2 and
MMP-9 decrease in plasma [93]. Interestingly, Lorenzl and Early work from Higashi and Miyazaki showed that MT1-
collaborators found consistent increased MMP-9 levels in MMP could process APP outside the Aβ sequence [108]. In
plasma in two studies [94, 95] and another study also showed this study, MT1-MMP activated by concanavalin A in the
increased MMP-9/TIMP-1 ratios in CSF from AD patients HT1080 fibrosarcoma cell line was shown to cleave at the
[96]. Noticeably, this study reported increased MMP-3 and ­Asn579 and M­ et580 site of A
­ PP770 ­(VLAN579-M580ISEPR),
MMP-9 levels, as well as increased MMP-3/TIMP-1 ratio, releasing a soluble truncated APP fragment lacking the
in the CSF of non-demented individuals carrying AD risk MMP-2 inhibitory domain (ISYGNDALMP). This deca-
markers (e.g., p-tau, Aβ42 or APOE ε4 allele). A negative peptide was previously reported in a study where MMP-2
correlation was found between plasma levels of MMP-3 was originally identified as a putative α-secretase capable
and the mini-mental state examination score that assesses of hydrolizing the ­Lys16-Leu17 bond within the Aβ sequence
cognitive performance [97]. Such negative correlation was [109]. The decapeptide, known as APP-derived peptide
also confirmed in the plasma of AD patients for MMP-3 inhibitor (APP-IP), specifically inhibits MMP-2 with an
activity measured by casein zymography. Consistent with ­IC50 value of 30 nM, well below the 2 µM for MT1-MMP

13

3174 S. Rivera et al.

and > 10 µM for MMP-3, -7 and -9. The APP-IP sequence, [114, 115]. Functionally, 5xFAD mice exhibit impaired LTP
spanning from amino acids 579–601 (βAPP770 numbering), [116, 117] and impaired learning and memory [118–120].
is present in sAPPα and sAPPβ. The authors concluded that Both deficits are reversed in 5xFAD/MT5-MMP-/- bigenic
MT1-MMP processing of APP would be a way for the tan- mice, which show preservation of LTP and olfactory and
dem MMP-2/MT1-MMP to regulate ECM proteolysis [108]. spatial learning and memory, compared to 5xFAD controls
Additional work on HEK293T cells demonstrated that MT1-, [114, 115]. It is also important to note that the lack of MT5-
MT3-, and MT5-MMP could release 5 major APP fragments MMP strongly decreases the release of a soluble N-terminal
after being co-expressed with ­APP770 [110]. In this study, APP fragment of ~ 95 kDa (sAPP95), complementary of
the cleavage sites for MT3-MMP were identified by mass the transmembrane CTF-30 fragment [114], supporting the
spectrometry at positions ­Ala463-Met464, ­Asn579-Met580 notion that APP is an in vivo substrate of MT5-MMP. In
­(VLAN579-M580ISEPR), ­His622-Ser623 and H ­ is685-Gln686. It addition to these in vivo data, MT5-MMP co-immunopre-
579 580
is noticeable that cleavage at A­ sn -Met by MT3-MMP cipitates with APP following MT5-MMP overexpression in
matches the MT1-MMP-mediated cleavage previously iden- HEK293 cells expressing App with the Swedish mutation
tified by Higashi and Miyazaki [108]. It is also interesting (HEKswe), resulting in the release of sAPP95 and increases
to note that although ­His685-Gln686 cleavage is one residue in Aβ40 levels [114, 115]. Although the precise molecular
upstream of the α-cleavage site in the Aβ sequence, MT3- mechanism behind MT5-MMP actions is still elusive, it is
MMP does not modify Aβ levels in HEK293T cells [110]. known that the proteinase is internalized in the endosomal
The authors suggested that MT5-MMP could have cleavage system [121] and favours APP sorting in endosomes [115],
profiles similar to those of MT1- and MT3-MMP, which a major site of Aβ formation. In addition, it has been shown
has been confirmed by a recent study showing that MT5- that N2a Swedish cells release exosomes containing η-CTF
MMP can effectively cleave the shorter neuronal isoform of that can be uptaken by neurons [122]. Overall, MT5-MMP
­APP696 at the ­VLAN504-M505ISEPR, also known as eta (η) could promote trafficking of APP or its metabolites (i.e.,
site [111]. According to this study, MT5-MMP (η-secretase) CTF-30/η-CTF) into intracellular organelles for more effi-
generates a CTF fragment of ~ 30 kDa (CTF-30/η-CTF) that cient amyloidogenic processing by β- and γ-secretases. This
can be further cleaved by α-secretase to produce a peptide way of cooperative cleavage has been recently shown for
(Aη-α, for amyloid eta-alpha) of ~ 12 kDa, which impairs asparagine endopeptidase (δ-secretase), whose cleavage of
LTP in cultured rat neurons. On the contrary, the concerted APP upstream the β-cleavage site favours subsequent pro-
action of η- and β-secretase generates the Aη-β peptide, cessing by β-secretase [123]. Not exclusively, it is also con-
surprisingly lacking neurotoxicity. Together, these data ceivable that η-CTF could be used as an exosomal cargo for
challenge the traditional view of α- and β-secretase as ben- intercellular propagation (see Fig. 1 for schematic represen-
eficial and detrimental enzymes, respectively. Interestingly, tation of MT5-MMP interactions with APP).
η-cleavage by MT5-MMP is strongly potentiated in the pres- The structurally close MT1-MMP shares about 60%
ence of BACE-1 inhibitors, raising concerns about possible sequence homology with MT5-MMP. While MT5-MMP
side effects mediated by the MMP in the case of therapeutic is primarily expressed in the nervous system, MT1-MMP
inhibition of BACE-1. The putative Aη-α peptide was found is ubiquitously distributed in the organism, including the
in the CSF of Alzheimer’s patients and healthy individuals, brain. We found that MT1-MMP expression is upregulated
but the concentrations did not differ between the two groups, in the brains of 5xFAD mice at symptomatic stages of the
making it difficult for the moment to assess the importance pathology and its overexpression in HEKswe cells stimulates
of Aη-α in AD pathology. Of note, despite that MT5- and C99 and Aβ production [78]. Moreover, MT1-MMP inter-
MT1-MMP cleave at the V ­ LAN504-M505ISEPR site, only acts with APP in HEKswe cells, and such interaction gener-
MT5-MMP and not MT1-MMP was identified in this study ates sAPP95 and the complementary transmembrane CTF-
as having η-secretase activity in vivo at 10 days postnatal 30, eventually leading to twofold increase of Aβ40 levels
[111]. [124]. Consistent with its prominent β-secretase role, it was
By the same time, our group tested the implication of shown that BACE-1 inhibition with C3 almost suppresses
MT5-MMP in AD using a different approach; we crossed basal Aβ production. Interestingly, such drastic effect of C3
transgenic 5xFAD mice carrying familial AD mutations in is prevented in the presence of MT1-MMP, which restores
human APP and Psen1 genes [112] with MT5-MMP knock- Aβ basal levels, suggesting that MT1-MMP may function
out mice [113]. The resulting bigenic 5xFAD/MT5-MMP-/- as a surrogate β-secretase upon BACE-1 inhibition [124].
strain exhibits dramatic reductions of Aβ burden (e.g., The β-secretase-like activity of MT1-MMP may reveal
plaques, oligomers, soluble Aβ40 and Aβ42) and C99 levels functional redundancies with BACE-1, which could be use-
in cortical and hippocampal regions [114]. The fall in Aβ/ ful for maintaining the physiological levels of Aβ in situa-
C99 levels is concomitant with reduced glial reactivity and tions where, for example, BACE-1 would be therapeutically
reduced levels of IL-1β and TNF-α in different brain areas inhibited. From these studies, it appears that MT5-MMP

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3175

Fig. 1  Schematic model summarizing the potential roles of MT1- and bodies (MVB) (6), followed by γ-secretase cleavage (7) to generate
MT5-MMP in APP  processing and the amyloidogenic pathway. 1: beta amyloid peptide (Aβ) and APP intracellular domain (AICD). The
Canonical amyloid precursor protein (APP) and MT-MMPs are tar- latter may be also generated in the lysosomes (8) and translocated
geted from the trans-Golgi network (TGN) to the plasma membrane into the nucleus (9). Aβ and sAPPβ can be degraded by lysosomes
or directly to the endosomal pathway. 2: On the plasma membrane, (10) or released into the extracellular space (11). CTF-30/η-CTF
MT-MMPs generate a soluble APP fragment of 95 kDa (sAPP95), and Aβ can also be released in exosomes (12). Note that generation
either alone or in concert with MMP-2, and the residual transmem- of η-fragments has only been reported for MT5-MMP so far. The
brane fragment of 30 kDa (CTF-30/η-CTF). 3: Also at the membrane contribution of MMP-2 to sAPP95 production has been described in
level, APP can be processed by α-secretase through the canonical association with MT1-MMP, and it remains to be determined whether
pathway that generates sAPPα and the transmembrane α-CTF/C83. this could also be the case for MT5-MMP. For the sake of simplicity,
In addition, the combined action of MT-MMPs and α-secretase could the cartoon does not show late and recycling endosomes, the possible
generate a synaptotoxic peptide (Aη-α). 4: MT-MMPs could promote location of γ-secretase on  the plasma membrane or APP trafficking
the internalization of CTF-30/η-CTF or unprocessed APP in early between the endosomal system and the TGN. This scheme includes
endosomes (5), where β-secretase generates sAPPβ, β-CTF/C99 or freely available objects from Servier Medical Art templates (https​://
Aη-β. β-secretase processing of APP may also occur in multivesicular smart​.servi​er.com)

13

3176 S. Rivera et al.

and MT1-MMP behave differently in response to BACE-1 cell surface TREM2 results in reduction of microglia phago-
inhibition; while MT5-MMP would generate synaptotoxic cytic activity, thereby possibly contributing to Aβ accumu-
peptides (i.e., Aη-α) leveraging on increased APP bioavail- lation [131]. Similarly, the shedding of LRP-1 by different
ability for α-secretase, MT1-MMP would rather compensate ADAMs (e.g., ADAM12) [99] might affect the ability of
for a physiological deficiency in Aβ. Another interesting fea- this receptor to promote endocytic-mediated clearance of
ture is that sAPP95 release by MT1-MMP partially involves Aβ. Finally, ADAMs could also contribute to the control of
MMP-2 when activated by MT1-MMP. In the absence of chronic inflammation by regulating cytokine activity. This
MT1-MMP, MMP-2 loses its ability to generate sAPP95, could be the case of ADAM17, which sheds membrane-
indicating that MT1-MMP is the step-limiting enzyme bound TNF-α and releases its soluble form [135]. Increasing
[124]. It remains to be elucidated whether MMP-2 can also experimental evidence suggests that TNF-α contributes to
cleave at ­VLAN579-M580ISEPR, which would indicate the AD pathogenesis and TNF-α inhibitors, some of which are
existence of a consensus cleavage site for APP shared by in clinical trials, may improve the pathological and cognitive
several MT-MMPs and MMP-2. These data highlight a new outcome (reviewed in [136]).
function for MMP-2—in tandem with MT1-MMP—in addi-
tion to its Aβ-degrading properties, thus providing a new TIMPs in Alzheimer’s disease
example of the dual/multifunctional nature of MMPs.
Like MT5-MMP, MT1-MMP also favours endosomal Along with the accumulation of data demonstrating the
APP sorting [124]. Gaining insight into endocytosis and involvement of metalloproteinases in neurodegenerative
recycling of MT-MMPs between the endosomal system and processes , the interest in their inhibitors has increased in
the membrane may be essential to understand the patho- parallel. The list of endogenous MMP inhibitors and their
physiological features of these enzymes. In this context, both physiological implications in the nervous system is rather
MT1-MMP [125] and APP [126] are internalized through a extensive and has already been examined elsewhere [3]. We
clathrin-dependent mechanism that involves specific short will focus on TIMPs in the context of this review.
sequences of their cytoplasmic tails. MT5-MMP internali- Early work reported the presence of TIMP-1 in neu-
zation and recycling is mediated by interactions between ritic lesions of AD and colocalization with APP, as well as
the last three residues at carboxyl terminus and Mint3, a with abnormally phosphorylated and truncated tau [137].
protein that contains two type III PDZ domains [121]. Since then, numerous attempts to describe a clear profile
Mint3 has also been identified as an adaptor protein that of expression for TIMPs have been undertaken with even-
promotes the export of APP from the Golgi complex [127]. tually somehow discordant results. It has been shown that
Together, these data may reflect interdependent trafficking TIMP-1 and TIMP-2 levels remain stable when comparing
mechanisms between MT-MMPs and APP that enable func- patients suffering from AD or mild cognitive impairment
tional interactions eventually leading to C99/Aβ production (MCI) with non-demented controls [92]. While plasma lev-
(summarized in Fig. 1). els of MMP-9 were higher in AD, PD and ALS patients
Overall, MT-MMPs appear as new actors in AD patho- compared to healthy individuals, no changes in TIMP-1
genesis, with multiple influences on pathways where proteo- and TIMP-2 levels were observed in the same work [138].
lytic and non-proteolytic interactions with APP can coexist. A pilot study on a small cohort of AD patients reported
A better understanding of such interactions is needed to shed moderately elevated levels of TIMP-1 in plasma [139].
light on the underlying pathogenic mechanisms. The comparison of different types of dementia reveals that
plasma levels of TIMP-1 and TIMP-2 respectively decrease
Singular contributions of ADAMs to Alzheimer’s in vascular and frontotemporal dementia, compared to
disease AD and MCI [95]. However, TIMP-2 levels are increased
in the CSF of Alzheimer patients [138]. Another study
Although the importance of ADAMs in AD has long been showed increased levels of TIMP-1 in bigenic APPSwDI/
recognized for their contribution to α-secretase activity [128, NOS2-/- mice compared to APPSwDI controls, along with
129], recent research extends their scope to mechanisms decreased MMP activity and increased Aβ levels, suggest-
other than those involving a direct cleavage of APP. For ing that TIMP-1 could negatively influence the pathological
instance, it has been shown that ADAM30 stimulates APP outcome as a consequence of lowered nitric oxide produc-
sorting to lysosomes and its degradation via the activation tion [140]. It has been proposed that the concentration of
of cathepsin D [130]. It is also noteworthy that ADAM10 TIMP-1 in the CSF is a distinctive biomarker to differentiate
[131, 132] and ADAM17 [133] can shed the triggering AD patients from those with subcortical vascular disease,
receptor expressed on myeloid cells 2 (TREM2), a protein the latter having higher levels than the former [107]. In the
for which some genetic variants are linked to an increased same vein, Duits and collaborators reported decreased lev-
risk of AD (see [134] for review). Mechanistically, lowering els of TIMP-1 and TIMP-2 in the CSF of AD patients with

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3177

cerebral microbleeds, arguing for an imbalance in favour of its inhibitory potential on MMPs suggests that it could con-
MMP proteolytic activity, which could eventually contribute tribute to modulate the progression of neurodegenerative
to the generation of such lesions  [141]. diseases.
TIMP-1 was found to be secreted by neurons in response
to MMP-9 release by mixed neuron/astrocyte cultures when
exposed to neurotoxic Aβ25-35, and to stimulate astrocyte Parkinson’s disease
proliferation [142], in agreement with our previous work
[143]. Coincident with gliosis, TIMP-1 mRNA expres- PD is the second most common neurodegenerative disorder
sion is strongly upregulated at early stages (between 4 and after AD,  characterised by motor symptoms, including rest
6 months) of the pathology in the brain of 5xFAD mice, tremor, bradykinesia, muscle stiffness, impaired posture and
whereas TIMP-2 and TIMP-3 mRNA levels show only mod- balance, and loss of automatic movement. Motor impair-
erate changes during the same period [78]. It remains to be ment is associated with selective loss of dopaminergic neu-
assessed  whether TIMP-1-mediated gliosis represents a rons in the substantia nigra and the degeneration of their
self-defence mechanism to eliminate Aβ and/or a conse- striatal projections. Although PD is an incurable disease,
quence of chronic inflammation. Increased levels of TIMP- a number of drugs and surgical interventions have shown
1, along with the absence of MMP-9, have been observed to be effective and significantly improve symptoms for a
in microvessels of AD compared to non-demented controls. limited period of time. A major feature of PD is the pres-
The authors correlated this observation with the ability of ence of fibrillar intracellular aggregates called Lewy bodies
thrombin to induce TIMP-1 expression in cultured brain (LBs) in different brain areas. LBs are mainly composed
endothelial cells. Altogether, these data suggest that changes of α-synuclein (α-Syn), a presynaptic protein of 140 amino
in angiogenesis in AD brains may involve the modulation of acids for which three missense mutations (A30P, E46K and
the MMP-9/TIMP-1 balance [144]. A53T) as well as wild-type gene multiplications, have been
Although not directly associated with AD, TIMP-2 associated with familial forms of PD [150–154]. α-Syn
mRNA and protein have been proposed as biomarkers of aggregation has also been reported in sporadic forms of PD
aging in the heart and cerebellum [145]. These results are of unknown etiology, which constitute the majority of cases
particularly significant in the context of recent findings from [155, 156]. It should be noted that α-Syn neuronal aggre-
Wyss–Coray’s group showing that TIMP-2 is an essential gates released into the extracellular space can be internalized
anti-aging factor as it promotes neuroplasticity, including by other neurons, reminiscent of a prion-like mechanism
LTP and hippocampal-dependent cognition. These conclu- that can promote PD progression through the self-propa-
sions were drawn after elegant parabiotic experiments in gation of α-Syn [157]. In addition to α-Syn, ninety other
which plasma from young donor mice enriched with TIMP-2 molecules have been identified in LBs, including parkin,
compensated for cognitive deficits in elderly recipient mice, tau, leucine-rich repeat kinase 2 (LRRK2), DJ-1 and APP
evoking the effect of an “elixir of eternal youth” [146]. With [158]. Even though a clear correlation exists between PD
aging being the main risk factor for AD, TIMP-2, as a reju- and the presence of LBs, the causal role of these aggregates
venating factor, could certainly play a role in the prevention has been questioned by recent studies in which early signs of
of AD and other neurodegenerative diseases. neuronal dysfunction/death in the substantia nigra precede
TIMP-3 inhibits APP cleavage by α-secretases ADAM10 LBs formation [159]. In this context, other authors have sug-
and ADAM17, resulting in increased levels of C99 and Aβ, gested that α-Syn oligomers and protofibrils are true toxic
probably due to increased cleavage of APP by β-secretase assemblies as opposed to fibrillar structures contained in
[147]. The proposed mechanism implies that α-secretase LBs, which would rather convey neuroprotective roles [158,
inhibition by TIMP-3 could stimulate APP trafficking to 160–162]. Alternatively, there is also experimental evidence
endosomes, where β- and γ-secretases convert APP to Aβ. that truncated forms of α-Syn contribute to PD and dementia
Consitent with this hypothesis, TIMP-3 levels are upregu- with Lewy bodies (DLB) pathology. Early work reported
lated in brains from both AD patients and transgenic AD the presence of truncated α-Syn in isolated LBs from DLB
mice [147]. In addition, TIMP-3 expression is correlated brains [163] and in transgenic mice harbouring the A53T
with NFT in AD patients compared to non-demented indi- familial mutation [164]. Subsequently, 3 other C-terminal
viduals [148]. truncated major peptides (residues 1-78, 1-91 and 1-93)
TIMP-4 has been less studied in neurodegenerative dis- were identified, with the particularity that they were more
eases than the other three TIMPs. Only one study reports abundant after digestion of A53T α-Syn compared to wild-
that plasma levels of TIMP-4 are higher in AD patients than type  [165]. When the 1-93 peptide was overexpressed in
in healthy controls and TIMP-4 levels positively correlate the mouse substantia nigra it selectively killed dopaminer-
with the severity of the disease [149]. While we are still gic neurons independently of LB-like formation. Crowther
waiting for clear evidence of TIMP-4 functions in the CNS, and colleagues demonstrated in vitro the ability of these

13

3178 S. Rivera et al.

truncated α-Syn forms to aggregate into filaments [166], MMP-1, -2, -8 and -9 [180], indicating that DJ-1 is a sub-
while other authors found that stable expression of C-ter- strate of several MMPs and that these interactions may have
minal truncated α-Syn increases neuroblastoma cell vulner- pathophysiological significance in PD.
ability to oxidative stress [167]. These data clearly indicate The functional and regulatory interplay between various
that α-Syn fragments contribute to PD and DLB pathogen- MMPs and PD markers further strengthens the implica-
esis, and several reports involve MMPs in α-Syn truncation. tion of MMPs in PD pathogenesis. Thus, α-Syn stimulates
One study showed that, in this order of efficacy, MMP-3, the expression of MMP-9 by cultured microglia and astro-
MT1-MMP, MMP-2, MMP-1 and MMP-9 cleaved recom- cytes [181]. Moreover, α-Syn promotes the expression of
binant α-Syn in vitro [168], a cleavage that was confirmed MT1-MMP and cell adhesion molecule CD44 in microglial
in another study for MMP-1, MMP-3 and MMP-9 [169]. cells. Shedding of CD44 by MT1-MMP facilitates micro-
α-Syn fragments generated by MMP-3 are prone to form glia detachment from the ECM and promotes its migra-
amyloidogenic aggregates that exhibit higher cytotoxicity tion in vitro [182]. Accordingly, siRNA-mediated silenc-
compared to intact α-Syn. Further support for a pathogenic ing of MT1-MMP dramatically decreases the migration
role of MMP-3 in PD comes from reports showing elevated of the microglial cell line BV-2 when transplanted in the
expression of the enzyme in dopaminergic neurons of the injured substantia nigra of 6-OHDA treated mice, suggest-
substantia nigra in a rat model of PD induced by 6-hydroxy- ing that MT1-MMP stimulates microglia recruitment into
dopamine (6-OHDA) [168], as well as in the brains of PD injured brain areas and thus accelerates the degenerative
patients [165]. In addition, in another PD model induced by process [182]. Mutations in the LRRK2 gene are the most
the injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyr- common cause of autosomal dominantly inherited PD [183,
idine (MPTP), the production of reactive oxygen species 184]. One study on microglia and leukocytes from LRRK2
and the degeneration of dopamine neurons are attenuated in mutant mice showed that increased levels of inflamma-
MMP-3-deficient mice [170], these effects being associated tory mediators and decreased levels of anti-inflammatory
with the prevention of BBB disruption and the improvement progranulin (PGRN) were associated with increased and
of locomotor activity [171]. Along this line, intracellular decreased levels of MMP-2 and MMP-9, respectively. Also
activation of MMP-3 triggers neuronal apoptosis [172] and interesting, hippocampal neurons of LRRK2 mutant mice
the subsequent release of active MMP-3, which stimulates show high levels of MMP-12 [185], an enzyme that cleaves
the production of inflammatory cytokines by neighbouring microglial PGRN, thereby possibly interfering with its anti-
microglia and eventually the exacerbation of dopaminergic inflammatory properties [186]. Together, these data are par-
neuron death [173]. Cultured microglia undergoes activa- ticularly sound because microglia is a critical regulator of
tion in response to α-Syn by a mechanism that involves the the inflammatory process that accompanies PD pathogenesis
upregulation of MMP expression (i.e., MMP-1, -3, -8 and (reviewed in [187]).
-9), and the consequent proteolytic activation of protease- In order to implement cell-based neuroprotective strat-
activated receptor-1 [174]. Under these conditions, the inhi- egies against PD, conditioned media from cultured mes-
bition of MMP-8 effectively reduces the levels of reactive enchymal stem cells (MSC) have been used to disaggre-
oxygen species and inflammatory mediators such as TNF-α gate preformed α-Syn fibrils in vitro, with the consequent
and IL-1β. MMP-3 implication in PD pathology has also increase in cell viability. Conditioned media of cells treated
been related to its ability to cleave DJ-1. The latter is a pro- with MMP-2 siRNA prevented the beneficial effects of
tein deglycase with protective antioxidant properties that normal conditioned media, suggesting that MMP-2 is one
mediates pleiotropic cellular responses, and whose mutation of the soluble factors that confer neuroprotection [188].
is associated with autosomal recessive early-onset PD [175]. In line with these observations, reduced levels of MMP-2
MMP-3-mediated cleavage of DJ-1 impairs its antioxidant in MPTP-treated mice can be restored by the inoculation
function in cultured dopamine-producing CATH.a cells upon of MSC. In addition, injection of MMP-2 in the brain of
oxidative stress [176]. The same study provided ground for mice pre-inoculated with α-Syn degrades extracellu-
in vivo cleavage, as the decrease in DJ-1 content observed lar α-Syn aggregates and oligomers [188]. Also consist-
after MPTP treatment was prevented in MMP-3 knockout ent with these data, MMP-2 levels were decreased in the
mice. Interestingly, one of the DJ-1 mutations (L166P) pre- substantia nigra of post mortem PD brains, while those of
vents its dimerization and causes its proteolytic digestion TIMP-1 were increased [189]. Whether the MMP/TIMP
by the proteasome [177, 178]. Also noteworthy, the L1669 ratio may be used as a marker of PD progression is still
mutation abrogates the anti-aggregation properties of wild- unclear, despite studies associating reduced TIMP-2 levels
type DJ-1 with respect to α-Syn [179]. From these data, it and increased vulnerability of neuroblastoma cells to MPTP
follows that MMP-3 processing of DJ-1 could harm its anti- or 6-OHDA, which is consistent with TIMP-2 preventing
oxidant and anti-α-Syn aggregation properties. In addition, the effects of these neurotoxins [190]. It appears, therefore,
DJ-1 can be cleaved effectively by MT1-MMP as well as by that different MMPs display opposite actions in PD; while

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3179

MMP-3 cleavage of α-Syn could promote its aggregation Many studies on ALS have used transgenic mice express-
into neurotoxic fibrils, MMP-2 could rather degrade them, ing ­C u 2+/Zn 2+ superoxide dismutase (SOD1) carrying
thus providing neuroprotection. Therefore, the distinction the G93A familial mutation (­ SOD1G93A). In these mice,
between both MMPs should be considered in prospective the involvement of MMP-9 in ALS pathogenesis is still
anti-PD therapies with MMPs as potential targets in PD. In under debate because different laboratories have reported
this line, it has been shown that the inhibition of MMP-8 conflicting data. One study shows that MMP-9 deletion
in the ­LRRK2G2019S mouse model reduces the levels of in ­SOD1G93A mice accelerates motor neuron disease and
inflammatory mediators and improves locomotor activity increases cell death [195], whereas another study reports
during systemic inflammatory LPS challenge [191]. Like- decreased neuron loss and greater survival in MMP-9 knock-
wise, hydroxamate-based MMP inhibitor Ro28-2653, which out mice in correlation with decreased TNF-α and FasL
targets MMP-2, -9 and MT1-MMP, significantly reduces immunostaining [196]. Selective expression of MMP-9 in
neuronal loss and MMP-9 levels in the substantia nigra of fast-fatigable motor neurons, which are highly vulnerable in
MPTP-treated mice [192]. ­SOD1G93A mice compared to the fast-fatigue resistant and
There are no reports yet linking ADAM function to PD. slow motor neuron subtypes, is concomitant with the activa-
However, since the contribution of reactive microglia to tion of the unfolded protein response and endoplasmic retic-
pathology is increasingly supported by experimental data, ulum stress mark characteristic of early pathological stages
it would be justified to explore in this context the modulation of ­SOD1G93A. Genetic deletion of MMP-9 in these mice, as
by ADAMs of proteins involved in microglial function, such well as pharmacological inhibition or viral gene therapy,
as TREM2. Of note, some authors have reported association delays muscle denervation [197]. In an attempt to interpret
between TREM2 variants and increased risk of PD, although selective motor neuron vulnerability, it has been shown that
the data are not conclusive for the moment [134]. osteopontin, an ECM protein, is selectively expressed in
fast-fatigue resistant and slow motor neurons, and stimulates
late expression of MMP-9 in these neuronal subtypes, which
could contribute to the second wave of degeneration in ALS
Amyotrophic lateral sclerosis by a mechanism controlled by MMP-9 [198]. Another study
showed that early administration of MMP inhibitor Ro28-
Amyotrophic lateral sclerosis (ALS) is a rare rapidly pro- 2653 increases the survival of S ­ OD1G93A mice [199], sug-
gressive neurodegenerative disease that selectively affects gesting a powerful detrimental activity of MMPs in ALS.
motor neurons in the brain, brainstem and spinal cord, lead- in vitro studies further support the possible contribution of
ing to severe disability, muscle atrophy, and also cognitive MMP-9 to ALS, as MMP-9 inhibition with glycoursodeoxy-
deficits in about 40% of ALS patients. Poor prognosis is ­ OD1G93A prevents cell degen-
cholic acid in a cell model of S
a distinctive clinical feature and life expectancy after the eration [200]. Although these data suggest that MMP-9 can
diagnosis is less than 20 months in 50% of patients, who be a candidate for therapeutic intervention in ALS, caution
often die from ventilatory failure (see for review [193]). Cur- is required; indeed, in a sporadic model of ALS using rNLS8
rently, there are no drugs that can cure or prevent ALS and mice, MMP-9 knockdown leads to the preservation of motor
the rapid course of the disease, as well as the lack of reliable neurons and muscle function, even if in the long run the
biomarkers, make therapeutic strategies particularly chal- reduction of MMP-9 results in premature death of a subset of
lenging. As with many other degenerative diseases, aging is rNLS8 mice [201]. It is then possible that targeting MMP-9
a major risk factor, with a rapid increase in prevalence after outside the neuromotor system may cause adverse effects
age 55 [193]. While the majority of patients have sporadic due to the inhibition of more general physiological processes
ALS of unknown multifactorial origin, genetically linked controlled by this MMP. In addition, MMP-9 could also be
ALS accounts for between 5 and 10% of cases. Deleterious considered as an early biomarker of the disease. MMP-9
genetic mutations affect genes coding for proteins implicated (and also MMP-2) levels measured by gelatin zymography
in RNA processing, and protein transport and clearance, like were found to increase in the serum of ­SOD1G93A mice at
C9orf72, SOD1, TDP-43 and FUS (reviewed in [194]). the beginning of the symptomatic phases of the pathology,
The pathogenesis of ALS is likely the result of a combi- as opposed to significantly lower levels observed during the
nation of genetic and environmental factors that may influ- pre-symptomatic and late-end stages [202].
ence key biological processes such as excitatory/inhibitory Beyond a neurocentric interpretation of ALS patho-
neurotransmitter balance, BBB integrity, protein folding, genesis, astrocytes could also play a deleterious role,
oxidative stress or neuroinflammation. All of these events thereby justifying attempts to replace diseased astrocytes
are commonly linked with MMP activity, and a number of by healthy astrocytes. Izrael and colleagues intrathecally
evidences discussed below support the role of MMPs in the injected astrocytes derived from human embryonic stem
disease. cells into transgenic ­SOD1G93A mice and rats, and observed

13

3180 S. Rivera et al.

an improvement of motor performance and delayed disease that encodes a chain of polyglutamine (polyQ) residues close
onset linked to astrocytic secretion of TIMP-1 and TIMP-2 to the N-terminus of Htt. This results in the aggregation of
[203]. On the contrary, a positive correlation has been put Htt in the nucleus and cytoplasm of neurons and the conse-
forward between TIMP-3 levels and neurodegeneration. quent cell demise that affects mainly the striatum and cortex
Degenerating neurons of S ­ OD1 G93A mice exhibit aug- [212–214]. The underlying neurotoxic mechanism is still
mented TIMP-3 levels in parallel with increased interactions elusive, but several lines of evidence point to Htt proteolysis
between Fas and Fas-associated protein with death domain by caspases and calpains, which releases N-terminal small
(FADD). Mechanistically, TIMP-3 inhibition of FADD fragments thought to be particularly toxic [215–217]. More
cleavage by MMP-3 would stabilize Fas–FADD interactions, recently, Miller and collaborators expanded the repertoire of
thereby promoting Fas-mediated apoptosis [204]. proteinases involved in this process to MMPs [218]. They
Few studies have been conducted in patients to explore screened 514 siRNA targeting all the proteinases encoded
possible changes in MMP content. Pioneer work in CNS tis- by the human genome to determine those whose inhibi-
sue from ALS patients localized increased immunostaining tion would reduce Htt processing, and more precisely the
of MMP-2 in astrocytes and MMP-9 in pyramidal neurons production of small N-terminal fragments. Among 11 con-
of the motor cortex and in spinal motor neurons [205]. In firmed proteinases, they found MMP-10, MT1-MMP and
addition, one study reported increased levels of MMP-9 in MMP-23B. They confirmed the upregulation of MMP-10
plasma of ALS patients but not in CSF [206], while another and MT1-MMP expression in striatal H ­ dh111Q/111Q cells as
7Q/7Q
study also reported an increase in MMP-9 levels in CSF compared to ­Hdh cells, and established a good correla-
[207]. It has been shown by ELISA assay that the concen- tion between the knockdown of these MMPs and a signifi-
tration of MT1-MMP, MMP-2 and TIMP-1 increases in the cant reduction of caspase activation in ­Hdh111Q/111Q cells.
CSF of ALS patients compared to healthy controls [208], In the same study it was shown that TIMP-1 and TIMP-3
thus confirming previous work showing increased TIMP-1 inhibited Htt cytotoxicity in ­Hdh111Q/111Q cells. Moreover,
levels in the CSF as well [138]. striatal MMP activity was also increased in Htt YAC128
There is scarce information on ADAMs in ALS. One (glutamine repeat expansion model) and R/62 (N-terminal
study described enhanced secretion of TNF-α by microglia fragment model) mouse models of HD. While MT1-MMP
from ­SOD1G93A, ­SOD1L8Q, or S ­ OD1G10V mutants upon and MMP-2 do not directly cleave Htt, MMP-10 generates
shedding by ADAM10 and ADAM17, thereby contribut- a N-terminal fragment of 48 kDa after cleavage at residue
ing to microglia inflammatory response [209]. In the same 402, very close to the N-terminus. Importantly, H ­ dh111Q/111Q
vein, it was shown that iron accumulation in ventral motor cell death was prevented upon knockdown of MMP-10. Fur-
neurons of S ­ OD1G93A mice induced oxidative stress and ther work from the same laboratory confirmed significant
the secretion of TNF-α mediated by increased ADAM17 alterations of MMPs expression in induced pluripotent stem
activity [210]. It has been suggested that the reduction in cell-derived neural stem cells from HD patients (HD-NSCs)
ADAMTS-4 observed in the spinal cord of S ­ OD1G93A mice with an expanded CAG repeat, compared to the isogenic
may be an adaptive mechanism to counteract the poten- corrected control [219]. The study shows that the expres-
tial neurotoxic effect of this metalloproteinase [211]. The sion of MMP-3 and -10 increases in HD-NSCs compared
authors reached this conclusion after observing significant to control NSCs, in contrast with the decrease observed
motor neuron death following the injection of recombinant in MT1-MMP, TIMP-1 and TIMP-2. TIMP-1 levels were
ADAMTS-4 in the spinal cord of S ­ OD1G93A mice. Collec- restored by TGF-β treatment, in pace with reduced neurotox-
tively, different ADAMs could, therefore, contribute to the icity of Htt, suggesting overall a beneficial effect of TIMP-
degenerative process that takes place in ALS. 1-mediated inhibition of Htt proteolysis by MMP-3 and -10
[219]. However, this idea is questionable, as other authors
have reported increased levels of TIMP-1 and -2 in the CSF
Huntington’s disease of HD patients [138].
MMP-9 levels are increased in post mortem tissues of
HD (also known as Huntington’s chorea) is the most fre- HD patients, as well as the content of pro-inflammatory
quent among inherited neurodegenerative polyglutamine dis- mediators MCP-1, IL-6, IL-8 and the anti-inflammatory
orders. It is still an incurable disease that causes changes in IL-10 [220]. Likewise, plasma MMP-9 and IL-6 levels are
mood and mental abilities, as well as deficits in motor coor- higher in HD patients compared to healthy controls and this
dination and ultimately dementia. HD is not properly speak- is also observed in R6/2 HD mice compared to wild-type
ing an aging-related disease since symptoms may appear littermates [221]. Moreover, MMP-3 and MMP-9 levels
at the age of 40 or even earlier. The cause of the disease increase in CSF from HD patients in correlation with dis-
lies on the mutation of the gene coding for huntingtin (Htt), ease severity [222]. The same authors showed that cultured
which presents an expanded CAG triplet repeat in exon 1 microglia from YAC128 mice (transgenic mouse model

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3181

with a 128-glutamine stretch) exhibits increased secretion formation. They also found that ADAM10 activity increased
of cytokines upon stimulation with MMP-3, an activator in the Htt-deficient adult brain, suggesting a role for Htt in
of microglia. Interestingly, the pro-inflammatory effect of synapse plasticity and remodelling. The question arises as
MMP-3 is equivalent to that caused by LPS treatment, link- to whether deficient regulation of ADAM10/N-cadherin in
ing the action of MMPs in HD to their pro-inflammatory HD brains with dysfunctional Htt could possibly contribute
properties. Seemingly, in a rat model of HD induced by to the disease.
intraperitoneal administration of a natural toxin, 3-Nitro-
propionic acid, MMP-9 expression and gelatinolytic activity
are upregulated in injured striatal blood vessels, suggesting
the possible implication of MMP-9 in BBB disruption in Concluding remarks and perspectives
HD. Noticeably, MMP-9 expression has been reported to be
upregulated in subsets of astrocytes and microglial cells in The traditional view of metalloproteinases as merely non-
the vicinity of lesions [223]. In contrast with the aforemen- specific degrading enzymes is definitely obsolete. Their
tioned studies, other authors reported recently in HD patients implications in homeostatic or detrimental pathways in
no changes in CSF MMP-9 levels and the lack of neuroin- many neurodegenerative diseases mobilize finely tuned
flammation at premanifest stages of the pathology [224]. proteolytic interactions with substrates and the generation
With regard to ADAMs, one study reports the physiologi- of biologically active cleavage products. A plethora of sub-
cal interaction between ADAM10 and Htt during neurulation strates has been identified and we can anticipate that their
[225]. The authors found that Htt inhibits N-cadherin shed- number will grow in the coming years in the light of new
ding by ADAM10, which promotes neurulation and rosette high-throughput proteomic techniques. Table 1 summarizes,

Table 1  Substrates that undergo direct or indirect cleavage under the influence of metalloproteinase actions
Alzheimer’s disease Parkinson’s disease Amyotrophic lateral sclerosis Huntington’s disease

MMP-1 – α-Syn [168, 169] – –


DJ-1 [180]
MMP-2 APP [109] α-Syn [168, 169] – Htt [218, 219]
Aβ [60, 61, 78, 82] DJ-1 [180]
tau [105]
MMP-3 Aβ [63] α-Syn [168, 169] FADD [204] –
tau [106] DJ-1 [176]
gelsolin [98]
MMP-7 Aβ [64] – – –
MMP-8 – DJ-1 [180] – –
MMP-9 APP [58, 61] α-Syn [168, 169] – –
Aβ [57, 59, 60, 70] DJ-1 [180]
tau [106]
claudin-5 [69]
RAGE [102, 103]
MMP-10 – – – Htt [218, 219]
MMP-12 – PGRN [186] – –
MMP-14 APP [78, 108–110, 124] α-Syn [168] – Htt [218, 219]
(MT1-MMP) Aβ [65] DJ-1 [180]
LRP-1 [99] CD44 [182]
MMP-16 APP [110] – – –
(MT3-MMP)
MMP-24 APP [110, 111, 114] – – –
(MT5-MMP)
ADAM10 APP [41, 42, 45] – TNF-α [209] N-cadherin [225]
LRP-1 [99]
RAGE [101, 102]
TREM2 [131, 132]
ADAM12 LRP-1 [99] – – –
ADAM17 APP [43] – TNF-α [209, 210] –
LRP-1 [99]
TREM2 [133]

13

3182 S. Rivera et al.

in a non-exhaustive manner, some of principal substrates the Initiative d’Excellence of Aix-Marseille University - A*MIDEX,
that have been cited in this review. a French “Investissements d’Avenir” programme.
In some cases, metalloproteinases have a dual effect, as
it is the case for MT1-MMP in AD experimental models
by degrading Aβ and also contributing to its formation. References
Likewise, α-secretases (e.g., ADAM10) may generate neu-
1. Hsia H-E, Tüshaus J, Brummer T, Zheng Y, Scilabra SD,
roprotective sAPPα and give rise to synaptotoxic C-terminal
Lichtenthaler SF (2019) Functions of ‘a disintegrin and metal-
fragments in synergy with MT5-MMP. We are witnessing loproteases (ADAMs)’ in the mammalian nervous system. Cell
the expansion of APP processing beyond prototypical con- Mol Life Sci. https​://doi.org/10.1007/s0001​8-019-03173​-7
trol by canonical secretases. In this scenario, MT-MMPs 2. Rivera S, Khrestchatisky M, Kaczmarek L, Rosenberg GA,
Jaworski DM (2010) Metzincin proteases and their inhibi-
appear to play an important role in the generation of new
tors: foes or friends in nervous system physiology? J Neu-
truncated APP fragments, whose functions are still poorly rosci 30(46):15337–15357. https​: //doi.org/10.1523/JNEUR​
understood. Similar challenges arise when considering the OSCI.3467-10.2010
physiological and pathological roles of breakdown products 3. Baranger K, Rivera S, Liechti FD, Grandgirard D, Bigas J, Seco
J, Tarrago T, Leib SL, Khrestchatisky M (2014) Endogenous and
resulting from the metalloproteinase-mediated process-
synthetic MMP inhibitors in CNS physiopathology. Prog Brain
ing of tau, α-Syn or Htt in AD, PD or HD, respectively. Res 214:313–351. https​://doi.org/10.1016/B978-0-444-63486​
Although proteolysis is clearly the main functional char- -3.00014​-1
acteristic of metalloproteinases, the field now opens up to 4. Okada Y (2017) Proteinases and matrix degradation. In: Firestein
GS, Budd RC, Gabriel SE, McInnes IB, O’Dell JR (eds) Kelley
study proteolysis-independent protein–protein interactions,
and Firestein’s textbook of rheumatology, vol 1, 10th edn. Else-
such as those reported for ADAM30, which bring to light vier, Philadelphia, pp 106–125. https​://doi.org/10.1016/B978-0-
new ways in which metalloproteinases can influence disease 323-31696​-5.00008​-5
outcomes. Therefore, it is plausible that new strategies will 5. Overall CM, Tam EM, Kappelhoff R, Connor A, Ewart T, Mor-
rison CJ, Puente X, Lopez-Otin C, Seth A (2004) Protease degra-
emerge based on the modulation non-proteolytic interactions
domics: mass spectrometry discovery of protease substrates and
between metalloproteinases and partner proteins. In addi- the CLIP-CHIP, a dedicated DNA microarray of all human pro-
tion, this could offer alternatives to targeting the proteolytic teases and inhibitors. Biol Chem 385(6):493–504. https​://doi.
activity of metalloproteinases, which often suffers from the org/10.1515/BC.2004.058
6. Starr AE, Dufour A, Maier J, Overall CM (2012) Biochemical
lack of specificity due to highly conserved catalytic domains
analysis of matrix metalloproteinase activation of chemokines
between family members. CCL15 and CCL23 and increased glycosaminoglycan binding of
Inflammation is a common event in neurodegenerative CCL16. J Biol Chem 287(8):5848–5860. https:​ //doi.org/10.1074/
diseases and many metalloproteinases are produced by jbc.M111.31460​9
7. Hsu TI, Lin SC, Lu PS, Chang WC, Hung CY, Yeh YM, Su
brain and peripheral immune cells. Gaining insight into
WC, Liao PC, Hung JJ (2015) MMP7-mediated cleavage of
the disease will also require a better understanding of how nucleolin at Asp255 induces MMP9 expression to promote tumor
the metalloproteinases released by these cells contribute to malignancy. Oncogene 34(7):826–837. https​://doi.org/10.1038/
pathology in a given spatio-temporal context and/or how onc.2014.22
8. Rosenberg GA (2017) Extracellular matrix inflammation in vas-
metalloproteinases can activate/inactivate immune cells by
cular cognitive impairment and dementia. Clin Sci 131(6):425–
controlling the cleavage of key receptors and cytokines (e.g., 437. https​://doi.org/10.1042/CS201​60604​
TREM2, TNF-α, IL-1β, etc). 9. Wojcik L, Sawicka A, Rivera S, Zalewska T (2009) Neurogen-
From all the above, it appears that the study of metal- esis in gerbil hippocampus following brain ischemia: focus on
the involvement of metalloproteinases. Acta Neurobiol Exp
loproteinases warrants a more global approach in which we
69(1):52–61
can learn from their conserved functions as well as their 10. Ould-yahoui A, Tremblay E, Sbai O, Ferhat L, Bernard A, Char-
specificities. These concepts should inspire future research rat E, Gueye Y, Lim NH, Brew K, Risso JJ, Dive V, Khrest-
on neurodegenerative diseases, where a thorough knowledge chatisky M, Rivera S (2009) A new role for TIMP-1 in modulat-
ing neurite outgrowth and morphology of cortical neurons. PLoS
of biological processes should pave the way for exciting dis-
One 4(12):e8289. https​://doi.org/10.1371/journ​al.pone.00082​89
coveries that will help to design more effective diagnostic 11. Ould-Yahoui A, Sbai O, Baranger K, Bernard A, Gueye Y, Char-
and therapeutic strategies. rat E, Clement B, Gigmes D, Dive V, Girard SD, Feron F, Khrest-
chatisky M, Rivera S (2013) Role of matrix metalloproteinases
Acknowledgements  This work was supported by funding from the in migration and neurotrophic properties of nasal olfactory stem
CNRS and Aix-Marseille Université and by public grants overseen and ensheathing cells. Cell Transplant 22(6):993–1010. https​://
by the French National Research Agency (ANR), MAD5 to SR, doi.org/10.3727/09636​8912X​65746​8
and PREVENTAD to MK, as part of the second “Investissements 12. Schonbeck U, Mach F, Libby P (1998) Generation of biologically
d’Avernir” program. The work was also supported by the DHUNE active IL-1 beta by matrix metalloproteinases: a novel caspase-
Centre of Excellence and grants from CoEN, “Fondation Plan Alz- 1-independent pathway of IL-1 beta processing. J Immunol
heimer”, France Alzheimer and Vaincre l’Alzheimer to SR. KB was 161(7):3340–3346
granted a research associate fellowship (Management of Talents) by 13. Rosenberg GA (2002) Matrix metalloproteinases in neuroinflam-
mation. Glia 39(3):279–291. https​://doi.org/10.1002/glia.10108​

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3183

14. Marchant DJ, Bellac CL, Moraes TJ, Wadsworth SJ, Dufour A, 29. Tan JZA (1861) Gleeson PA (2019) The role of membrane
Butler GS, Bilawchuk LM, Hendry RG, Robertson AG, Cheung trafficking in the processing of amyloid precursor protein and
CT, Ng J, Ang L, Luo Z, Heilbron K, Norris MJ, Duan W, Bucyk production of amyloid peptides in Alzheimer’s disease. BBA-
T, Karpov A, Devel L, Georgiadis D, Hegele RG, Luo H, Gran- Biomembranes 4:697–712. https​://doi.org/10.1016/j.bbame​
ville DJ, Dive V, McManus BM, Overall CM (2014) A new m.2018.11.013
transcriptional role for matrix metalloproteinase-12 in antiviral 30. De Strooper B (2010) Proteases and proteolysis in Alzheimer
immunity. Nat Med 20(5):493–502. https​://doi.org/10.1038/ disease: a multifactorial view on the disease process. Physiol Rev
nm.3508 90(2):465–494. https​://doi.org/10.1152/physr​ev.00023​.2009
15. Nagase H, Brew K (2003) Designing TIMP (tissue inhibitor of 31. Kim J, Castellano JM, Jiang H, Basak JM, Parsadanian M, Pham
metalloproteinases) variants that are selective metalloproteinase V, Mason SM, Paul SM, Holtzman DM (2009) Overexpression
inhibitors. Bioch Soc Symp 70:201–212 of low-density lipoprotein receptor in the brain markedly inhib-
16. Amour A, Knight CG, Webster A, Slocombe PM, Stephens PE, its amyloid deposition and increases extracellular A beta clear-
Knauper V, Docherty AJ, Murphy G (2000) The in vitro activity ance. Neuron 64(5):632–644. https​://doi.org/10.1016/j.neuro​
of ADAM-10 is inhibited by TIMP-1 and TIMP-3. FEBS Lett n.2009.11.013
473(3):275–279 32. Castellano JM, Deane R, Gottesdiener AJ, Verghese PB, Stewart
17. Vandooren J, Van den Steen PE, Opdenakker G (2013) Biochem- FR, West T, Paoletti AC, Kasper TR, DeMattos RB, Zlokovic
istry and molecular biology of gelatinase B or matrix metallo- BV, Holtzman DM (2012) Low-density lipoprotein receptor over-
proteinase-9 (MMP-9): the next decade. Crit Rev Biochem Mol expression enhances the rate of brain-to-blood Abeta clearance
48(3):222–272. https​://doi.org/10.3109/10409​238.2013.77081​9 in a mouse model of beta-amyloidosis. Proc Natl Acad Sci USA
18. Shofuda K, Moriyama K, Nishihashi A, Higashi S, Mizushima H, 109(38):15502–15507. https:​ //doi.org/10.1073/pnas.120644​ 6109​
Yasumitsu H, Miki K, Sato H, Seiki M, Miyazaki K (1998) Role 33. Basak JM, Verghese PB, Yoon H, Kim J, Holtzman DM (2012)
of tissue inhibitor of metalloproteinases-2 (TIMP-2) in regula- Low-density lipoprotein receptor represents an apolipopro-
tion of pro-gelatinase A activation catalyzed by membrane-type tein E-independent pathway of Abeta uptake and degradation
matrix metalloproteinase-1 (MT1-MMP) in human cancer cells. by astrocytes. J Biol Chem 287(17):13959–13971. https​://doi.
J Biochem 124(2):462–470 org/10.1074/jbc.M111.28874​6
19. Leco KJ, Apte SS, Taniguchi GT, Hawkes SP, Khokha R, Schultz 34. Jaeger LB, Dohgu S, Hwang MC, Farr SA, Murphy MP, Fleegal-
GA, Edwards DR (1997) Murine tissue inhibitor of metallopro- DeMotta MA, Lynch JL, Robinson SM, Niehoff ML, Johnson
teinases-4 (Timp-4): cDNA isolation and expression in adult SN, Kumar VB, Banks WA (2009) Testing the neurovascular
mouse tissues. FEBS Lett 401(2–3):213–217 hypothesis of Alzheimer’s disease: LRP-1 antisense reduces
20. Derry JM, Barnard PJ (1992) Physical linkage of the A-raf-1, blood-brain barrier clearance, increases brain levels of amyloid-
properdin, synapsin I, and TIMP genes on the human and mouse beta protein, and impairs cognition. J Alzheimers Dis 17(3):553–
X chromosomes. Genomics 12(4):632–638 570. https​://doi.org/10.3233/JAD-2009-1074
21. Pohar N, Godenschwege TA, Buchner E (1999) Invertebrate 35. Storck SE, Meister S, Nahrath J, Meissner JN, Schubert N, Di
tissue inhibitor of metalloproteinase: structure and nested Spiezio A, Baches S, Vandenbroucke RE, Bouter Y, Prikulis I,
gene organization within the synapsin locus is conserved from Korth C, Weggen S, Heimann A, Schwaninger M, Bayer TA, Pie-
Drosophila to human. Genomics 57(2):293–296. https​://doi. trzik CU (2016) Endothelial LRP1 transports amyloid-beta(1-42)
org/10.1006/geno.1999.5776 across the blood-brain barrier. J Clin Invest 126(1):123–136.
22. Rahkonen OP, Koskivirta IM, Oksjoki SM, Jokinen E, Vuorio EI https​://doi.org/10.1172/JCI81​108
(2002) Characterization of the murine Timp4 gene, localization 36. Liu CC, Hu J, Zhao N, Wang J, Wang N, Cirrito JR, Kane-
within intron 5 of the synapsin 2 gene and tissue distribution of kiyo T, Holtzman DM, Bu G (2017) Astrocytic LRP1 medi-
the mRNA. Biochim Biophys Acta 1577(1):45–52 ates brain abeta clearance and impacts amyloid deposition. J
23. Prince M, Comas-Herrera A, Knapp M, Guerchet M, Karagi- Neurosci 37(15):4023–4031. https​://doi.org/10.1523/JNEUR​
annidou M (2016) World Alzheimer Report 2016. Improving OSCI.3442-16.2017
healthcare for people living with dementia. Coverage, quality and 37. Cai Z, Liu N, Wang C, Qin B, Zhou Y, Xiao M, Chang L, Yan
costs now and in the future. Alzheimer’s Disease International, LJ, Zhao B (2016) Role of RAGE in Alzheimer’s disease. Cell
London Mol Neurobiol 36(4):483–495. https​://doi.org/10.1007/s1057​
24. Mullane K, Williams M (2013) Alzheimer’s therapeutics: contin- 1-015-0233-3
ued clinical failures question the validity of the amyloid hypoth- 38. Wan W, Cao L, Liu L, Zhang C, Kalionis B, Tai X, Li Y, Xia
esis-but what lies beyond? Biochem Pharmacol 85(3):289–305. S (2015) Abeta(1-42) oligomer-induced leakage in an in vitro
https​://doi.org/10.1016/j.bcp.2012.11.014 blood-brain barrier model is associated with up-regulation of
25. Mikulca JA, Nguyen V, Gajdosik DA, Teklu SG, Giunta EA, RAGE and metalloproteinases, and down-regulation of tight
Lessa EA, Tran CH, Terak EC, Raffa RB (2014) Potential novel junction scaffold proteins. J Neurochem 134(2):382–393. https​
targets for Alzheimer pharmacotherapy: II Update on secretase ://doi.org/10.1111/jnc.13122​
inhibitors and related approaches. J Clin Pharm Ther 39(1):25– 39. Fang F, Yu Q, Arancio O, Chen D, Gore SS, Yan SS, Yan SF
37. https​://doi.org/10.1111/jcpt.12112​ (2018) RAGE mediates Abeta accumulation in a mouse model
26. Mehta D, Jackson R, Paul G, Shi J, Sabbagh M (2017) Why do of Alzheimer’s disease via modulation of beta- and gamma-
trials for Alzheimer’s disease drugs keep failing? A discontin- secretase activity. Hum Mol Genet 27(6):1002–1014. https​://
ued drug perspective for 2010-2015. Expert Opin Investig Drugs doi.org/10.1093/hmg/ddy01​7
26(6):735–739. https:​ //doi.org/10.1080/135437​ 84.2017.132386​ 8 40. Muller UC, Deller T, Korte M (2017) Not just amyloid: physi-
27. Selkoe DJ, Hardy J (2016) The amyloid hypothesis of Alzhei- ological functions of the amyloid precursor protein family.
mer’s disease at 25 years. EMBO Mol Med 8(6):595–608. https​ Nat Rev Neurosci 18(5):281–298. https​: //doi.org/10.1038/
://doi.org/10.15252​/emmm.20160​6210 nrn.2017.29
28. Lesne SE, Sherman MA, Grant M, Kuskowski M, Schneider JA, 41. Jorissen E, Prox J, Bernreuther C, Weber S, Schwanbeck
Bennett DA, Ashe KH (2013) Brain amyloid-beta oligomers in R, Serneels L, Snellinx A, Craessaerts K, Thathiah A, Tes-
ageing and Alzheimer’s disease. Brain 136(Pt 5):1383–1398. seur I, Bartsch U, Weskamp G, Blobel CP, Glatzel M, De
https​://doi.org/10.1093/brain​/awt06​2 Strooper B, Saftig P (2010) The disintegrin/metalloproteinase

13

3184 S. Rivera et al.

ADAM10 is essential for the establishment of the brain cortex. protein:enhancement of learning and memory. J Alzheimers Dis
J Neurosci 30(14):4833–4844. https​://doi.org/10.1523/JNEUR​ 19(2):441–449. https​://doi.org/10.3233/JAD-2009-1230
OSCI.5221-09.2010 54. Puzzo D, Privitera L, Palmeri A (2012) Hormetic effect of
42. Kuhn PH, Wang H, Dislich B, Colombo A, Zeitschel U, Ellwart amyloid-beta peptide in synaptic plasticity and memory. Neu-
JW, Kremmer E, Rossner S, Lichtenthaler SF (2010) ADAM10 robiol Aging 33(7):1484. https​://doi.org/10.1016/j.neuro​biola​
is the physiologically relevant, constitutive alpha-secretase of ging.2011.12.020
the amyloid precursor protein in primary neurons. EMBO J 55. Lawrence JL, Tong M, Alfulaij N, Sherrin T, Contarino M,
29(17):3020–3032. https​://doi.org/10.1038/emboj​.2010.167 White MM, Bellinger FP, Todorovic C, Nichols RA (2014)
43. Buxbaum JD, Liu KN, Luo Y, Slack JL, Stocking KL, Pes- Regulation of presynaptic ­Ca2+, synaptic plasticity and contex-
chon JJ, Johnson RS, Castner BJ, Cerretti DP, Black RA tual fear conditioning by a N-terminal beta-amyloid fragment. J
(1998) Evidence that tumor necrosis factor alpha convert- Neurosci 34(43):14210–14218. https​://doi.org/10.1523/JNEUR​
ing enzyme is involved in regulated alpha-secretase cleav- OSCI.0326-14.2014
age of the Alzheimer amyloid protein precursor. J Biol Chem 56. Palmeri A, Ricciarelli R, Gulisano W, Rivera D, Rebosio C,
273(43):27765–27767 Calcagno E, Tropea MR, Conti S, Das U, Roy S, Pronzato MA,
44. Chasseigneaux S, Allinquant B (2012) Functions of Abeta, Arancio O, Fedele E, Puzzo D (2017) Amyloid-beta peptide is
sAPPalpha and sAPPbeta : similarities and differences. J needed for cGMP-induced long-term potentiation and memory.
Neurochem 120(Suppl 1):99–108. https ​ : //doi.org/10.111 J Neurosci 37(29):6926–6937. https​://doi.org/10.1523/JNEUR​
1/j.1471-4159.2011.07584​.x OSCI.3607-16.2017
45. Hick M, Herrmann U, Weyer SW, Mallm JP, Tschape JA, Borgers 57. Backstrom JR, Lim GP, Cullen MJ, Tokes ZA (1996) Matrix
M, Mercken M, Roth FC, Draguhn A, Slomianka L, Wolfer DP, metalloproteinase-9 (MMP-9) is synthesized in neurons of the
Korte M, Muller UC (2015) Acute function of secreted amyloid human hippocampus and is capable of degrading the amyloid-
precursor protein fragment APPsalpha in synaptic plasticity. beta peptide (1-40). J Neurosci 16(24):7910–7919
Acta Neuropathol 129(1):21–37. https​://doi.org/10.1007/s0040​ 58. Fragkouli A, Tsilibary EC, Tzinia AK (2014) Neuroprotec-
1-014-1368-x tive role of MMP-9 overexpression in the brain of Alzhei-
46. Rose C, Dorard E, Audrain M, Gorisse-Hussonnois L, Cartier mer’s 5xFAD mice. Neurobiol Dis 70:179–189. https​://doi.
N, Braudeau J, Allinquant B (2018) Transient increase in sAP- org/10.1016/j.nbd.2014.06.021
Palpha secretion in response to Abeta1-42 oligomers: an attempt 59. Yan P, Hu X, Song H, Yin K, Bateman RJ, Cirrito JR, Xiao Q,
of neuronal self-defense? Neurobiol Aging 61:23–35. https:​ //doi. Hsu FF, Turk JW, Xu J, Hsu CY, Holtzman DM, Lee JM (2006)
org/10.1016/j.neuro​biola​ging.2017.09.008 Matrix metalloproteinase-9 degrades amyloid-beta fibrils in vitro
47. Soscia SJ, Kirby JE, Washicosky KJ, Tucker SM, Ingelsson M, and compact plaques in situ. J Biol Chem 281(34):24566–24574.
Hyman B, Burton MA, Goldstein LE, Duong S, Tanzi RE, Moir https​://doi.org/10.1074/jbc.M6024​40200​
RD (2010) The Alzheimer’s disease-associated amyloid beta- 60. Hernandez-Guillamon M, Mawhirt S, Blais S, Montaner J, Neu-
protein is an antimicrobial peptide. PLoS One 5(3):e9505. https​ bert TA, Rostagno A, Ghiso J (2015) Sequential amyloid-beta
://doi.org/10.1371/journ​al.pone.00095​05 degradation by the matrix metalloproteases MMP-2 and MMP-9.
48. Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, J Biol Chem 290(24):15078–15091. https​://doi.org/10.1074/jbc.
Ghofrani J, Lefkowitz A, McColl G, Goldstein LE, Tanzi RE, M114.61093​1
Moir RD (2016) Amyloid-beta peptide protects against microbial 61. Yin KJ, Cirrito JR, Yan P, Hu X, Xiao Q, Pan X, Bateman R,
infection in mouse and worm models of Alzheimer’s disease. Sci Song H, Hsu FF, Turk J, Xu J, Hsu CY, Mills JC, Holtzman DM,
Transl Med 8(340):340ra372. https​://doi.org/10.1126/scitr​anslm​ Lee JM (2006) Matrix metalloproteinases expressed by astro-
ed.aaf10​59 cytes mediate extracellular amyloid-beta peptide catabolism. J
49. Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK, Rod- Neurosci 26(43):10939–10948. https​://doi.org/10.1523/JNEUR​
riguez AS, Mitchell T, Washicosky KJ, Gyorgy B, Breakefield OSCI.2085-06.2006
XO, Tanzi RE, Moir RD (2018) Alzheimer’s disease-associated 62. Moore KM, Girens RE, Larson SK, Jones MR, Restivo JL, Holtz-
beta-amyloid is rapidly seeded by herpesviridae to protect man DM, Cirrito JR, Yuede CM, Zimmerman SD, Timson BF
against brain infection. Neuron 100(6):1527–1532. https​://doi. (2016) A spectrum of exercise training reduces soluble Abeta
org/10.1016/j.neuro​n.2018.11.043 in a dose-dependent manner in a mouse model of Alzheimer’s
50. Dominy SS, Lynch C, Ermini F, Benedyk M, Marczyk A, Kon- disease. Neurobiol Dis 85:218–224. https​://doi.org/10.1016/j.
radi A, Nguyen M, Haditsch U, Raha D, Griffin C, Holsinger LJ, nbd.2015.11.004
Arastu-Kapur S, Kaba S, Lee A, Ryder MI, Potempa B, Mydel 63. White AR, Du T, Laughton KM, Volitakis I, Sharples RA, Xili-
P, Hellvard A, Adamowicz K, Hasturk H, Walker GD, Reynolds nas ME, Hoke DE, Holsinger RM, Evin G, Cherny RA, Hill
EC, Faull RLM, Curtis MA, Dragunow M, Potempa J (2019) AF, Barnham KJ, Li QX, Bush AI, Masters CL (2006) Degrada-
Porphyromonas gingivalis in Alzheimer’s disease brains: Evi- tion of the Alzheimer disease amyloid beta-peptide by metal-
dence for disease causation and treatment with small-molecule dependent up-regulation of metalloprotease activity. J Biol Chem
inhibitors. Sci Adv 5(1):33. https:​ //doi.org/10.1126/sciadv​ .aau33​ 281(26):17670–17680. https:​ //doi.org/10.1074/jbc.M60248​ 7200​
33 64. Taniguchi M, Matsuura K, Nakamura R, Kojima A, Konishi
51. Abramov E, Dolev I, Fogel H, Ciccotosto GD, Ruff E, Slut- M, Akizawa T (2017) MMP-7 cleaves amyloid beta fragment
sky I (2009) Amyloid-beta as a positive endogenous regulator peptides and copper ion inhibits the degradation. Biometals
of release probability at hippocampal synapses. Nat Neurosci 30(5):797–807. https​://doi.org/10.1007/s1053​4-017-0048-4
12(12):1567–1576. https​://doi.org/10.1038/nn.2433 65. Liao MC, Van Nostrand WE (2010) Degradation of soluble
52. Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski A, Palm- and fibrillar amyloid beta-protein by matrix metalloproteinase
eri A, Arancio O (2008) Picomolar amyloid-beta positively (MT1-MMP) in vitro. Biochemistry 49(6):1127–1136. https​://
modulates synaptic plasticity and memory in hippocampus. J doi.org/10.1021/bi901​994d
Neurosci 28(53):14537–14545. https​://doi.org/10.1523/JNEUR​ 66. Montaner J, Ramiro L, Simats A, Hernández-Guillamon M,
OSCI.2692-08.2008 Delgado P, Bustamante A, Rosell A (2019) Matrix metallopro-
53. Morley JE, Farr SA, Banks WA, Johnson SN, Yamada teinases and ADAMs in stroke. Cell Mol Life Sci. https​://doi.
KA, Xu L (2010) A physiological role for amyloid-beta org/10.1007/s0001​8-019-03175​-5

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3185

67. Sweeney MD, Sagare AP, Zlokovic BV (2018) Blood-brain 80. Seo JH, Guo S, Lok J, Navaratna D, Whalen MJ, Kim KW, Lo
barrier breakdown in Alzheimer disease and other neurodegen- EH (2012) Neurovascular matrix metalloproteinases and the
erative disorders. Nat Rev Neurol 14(3):133–150. https​://doi. blood–brain barrier. Curr Pharm Design 18(25):3645–3648
org/10.1038/nrneu​rol.2017.188 81. Jourquin J, Tremblay E, Decanis N, Charton G, Hanessian S,
68. Giannoni P, Arango-Lievano M, Neves ID, Rousset MC, Chollet AM, Le Diguardher T, Khrestchatisky M, Rivera S
Baranger K, Rivera S, Jeanneteau F, Claeysen S, Marchi N (2003) Neuronal activity-dependent increase of net matrix met-
(2016) Cerebrovascular pathology during the progression of alloproteinase activity is associated with MMP-9 neurotoxicity
experimental Alzheimer’s disease. Neurobiol Dis 88:107–117. after kainate. Eur J Neurosci 18(6):1507–1517
https​://doi.org/10.1016/j.nbd.2016.01.001 82. Hernandez-Guillamon M, Mawhirt S, Fossati S, Blais S, Pares
69. Spampinato SF, Merlo S, Sano Y, Kanda T, Sortino MA (2017) M, Penalba A, Boada M, Couraud PO, Neubert TA, Montaner J,
Astrocytes contribute to Abeta-induced blood-brain barrier Ghiso J, Rostagno A (2010) Matrix metalloproteinase 2 (MMP-
damage through activation of endothelial MMP9. J Neurochem 2) degrades soluble vasculotropic amyloid-beta E22Q and L34V
142(3):464–477. https​://doi.org/10.1111/jnc.14068​ mutants, delaying their toxicity for human brain microvascular
70. Mazzitelli S, Filipello F, Rasile M, Lauranzano E, Starvaggi- endothelial cells. J Biol Chem 285(35):27144–27158. https:​ //doi.
Cucuzza C, Tamborini M, Pozzi D, Barajon I, Giorgino T, org/10.1074/jbc.M110.13522​8
Natalello A, Matteoli M (2016) Amyloid-beta 1-24 C-terminal 83. Mizoguchi H, Takuma K, Fukuzaki E, Ibi D, Someya E, Akazawa
truncated fragment promotes amyloid-beta 1-42 aggregate for- KH, Alkam T, Tsunekawa H, Mouri A, Noda Y, Nabeshima T,
mation in the healthy brain. Acta Neuropathol Comm 4(1):110. Yamada K (2009) Matrix metalloprotease-9 inhibition improves
https​://doi.org/10.1186/s4047​8-016-0381-9 amyloid beta-mediated cognitive impairment and neurotoxic-
71. Brkic M, Balusu S, Van Wonterghem E, Gorle N, Benilova I, ity in mice. J Pharmacol Exp Ther 331(1):14–22. https​://doi.
Kremer A, Van Hove I, Moons L, De Strooper B, Kanazir S, Lib- org/10.1124/jpet.109.15472​4
ert C, Vandenbroucke RE (2015) Amyloid beta oligomers disrupt 84. Langenfurth A, Rinnenthal JL, Vinnakota K, Prinz V, Carlo
blood-CSF barrier integrity by activating matrix metalloprotein- AS, Stadelmann C, Siffrin V, Peaschke S, Endres M, Heppner
ases. J Neurosci 35(37):12766–12778. https​://doi.org/10.1523/ F, Glass R, Wolf SA, Kettenmann H (2014) Membrane-type 1
JNEUR​OSCI.0006-15.2015 metalloproteinase is upregulated in microglia/brain macrophages
72. Garcia-Alloza M, Prada C, Lattarulo C, Fine S, Borrelli LA, in neurodegenerative and neuroinflammatory diseases. J Neuro-
Betensky R, Greenberg SM, Frosch MP, Bacskai BJ (2009) sci Res 92(3):275–286. https​://doi.org/10.1002/jnr.23288​
Matrix metalloproteinase inhibition reduces oxidative stress 85. Ito S, Koshikawa N, Mochizuki S, Takenaga K (2007) 3-Methy-
associated with cerebral amyloid angiopathy in vivo in trans- ladenine suppresses cell migration and invasion of HT1080
genic mice. J Neurochem 109(6):1636–1647. https​://doi.org/10. fibrosarcoma cells through inhibiting phosphoinositide
1111/j.1471-4159.2009.06096​.x 3-kinases independently of autophagy inhibition. Int J Oncol
73. Prinz M, Priller J (2017) The role of peripheral immune cells in 31(2):261–268
the CNS in steady state and disease. Nat Neurosci 20(2):136– 86. Ierusalimsky VN, Kuleshova EP, Balaban PM (2013) Expression
144. https​://doi.org/10.1038/nn.4475 of the type 1 metalloproteinase in the rat hippocampus after the
74. Jhang KA, Lee EO, Kim HS, Chang KA, Suh YH, Chong YH intracerebroventricular injection of beta-amyloid peptide (25-35).
(2016) S100A9 exacerbates the Abeta1-42-mediated innate Acta Neurobiol Exp 73(4):571–578
immunity in human THP-1 monocytes. CNS Neurol Disord-DR 87. Jung SS, Zhang W, Van Nostrand WE (2003) Pathogenic A beta
15(8):910–917 induces the expression and activation of matrix metalloprotein-
75. Achilli C, Ciana A, Minetti G (2014) Amyloid-beta (25-35) pep- ase-2 in human cerebrovascular smooth muscle cells. J Neuro-
tide induces the release of pro-matrix metalloprotease 9 (pro- chem 85(5):1208–1215
MMP-9) from human neutrophils. Mol Cell Biochem 397(1– 88. Walker DG, Link J, Lue LF, Dalsing-Hernandez JE, Boyes BE
2):117–123. https​://doi.org/10.1007/s1101​0-014-2178-0 (2006) Gene expression changes by amyloid beta peptide-stim-
76. Zenaro E, Pietronigro E, Della Bianca V, Piacentino G, Marongiu ulated human postmortem brain microglia identify activation of
L, Budui S, Turano E, Rossi B, Angiari S, Dusi S, Montresor multiple inflammatory processes. J Leuk Biol 79(3):596–610.
A, Carlucci T, Nani S, Tosadori G, Calciano L, Catalucci D, https​://doi.org/10.1189/jlb.07053​77
Berton G, Bonetti B, Constantin G (2015) Neutrophils promote 89. Leake A, Morris CM, Whateley J (2000) Brain matrix metallo-
Alzheimer’s disease-like pathology and cognitive decline via proteinase 1 levels are elevated in Alzheimer’s disease. Neurosci
LFA-1 integrin. Nat Med 21(8):880–886. https:​ //doi.org/10.1038/ Lett 291(3):201–203
nm.3913 90. Sekine-Aizawa Y, Hama E, Watanabe K, Tsubuki S, Kanai-
77. Deb S, Gottschall PE (1996) Increased production of matrix Azuma M, Kanai Y, Arai H, Aizawa H, Iwata N, Saido TC (2001)
metalloproteinases in enriched astrocyte and mixed hippocam- Matrix metalloproteinase (MMP) system in brain: identification
pal cultures treated with beta-amyloid peptides. J Neurochem and characterization of brain-specific MMP highly expressed in
66(4):1641–1647 cerebellum. Eur J Neurosci 13(5):935–948
78. Py NA, Bonnet AE, Bernard A, Marchalant Y, Charrat E, Che- 91. Yoshiyama Y, Asahina M, Hattori T (2000) Selective distribution
cler F, Khrestchatisky M, Baranger K, Rivera S (2014) Differ- of matrix metalloproteinase-3 (MMP-3) in Alzheimer’s disease
ential spatio-temporal regulation of MMPs in the 5xFAD mouse brain. Acta Neuropathol 99(2):91–95
model of Alzheimer’s disease: evidence for a pro-amyloidogenic 92. Mroczko B, Groblewska M, Zboch M, Kulczynska A, Koper OM,
role of MT1-MMP. Front Aging Neurosci 6:247. https​://doi. Szmitkowski M, Kornhuber J, Lewczuk P (2014) Concentrations
org/10.3389/fnagi​.2014.00247​ of matrix metalloproteinases and their tissue inhibitors in the
79. Girard SD, Jacquet M, Baranger K, Migliorati M, Escoffier G, cerebrospinal fluid of patients with Alzheimer’s disease. J Alz-
Bernard A, Khrestchatisky M, Feron F, Rivera S, Roman FS, heimers Dis 40(2):351–357. https​://doi.org/10.3233/JAD-13163​
Marchetti E (2014) Onset of hippocampus-dependent memory 4
impairments in 5XFAD transgenic mouse model of Alzheimer’s 93. Horstmann S, Budig L, Gardner H, Koziol J, Deuschle M,
disease. Hippocampus 24(7):762–772. https​://doi.org/10.1002/ Schilling C, Wagner S (2010) Matrix metalloproteinases
hipo.22267​ in peripheral blood and cerebrospinal fluid in patients with

13

3186 S. Rivera et al.

Alzheimer’s disease. Int Psychogeriatr 22(6):966–972. https​:// and Alzheimer’s disease. J Alzheimers Dis 27(3):665–676. https​
doi.org/10.1017/S1041​61021​00008​27 ://doi.org/10.3233/JAD-2011-11056​6
94. Lorenzl S, Albers DS, Relkin N, Ngyuen T, Hilgenberg SL, Chir- 108. Higashi S, Miyazaki K (2003) Novel processing of beta-amyloid
ichigno J, Cudkowicz ME, Beal MF (2003) Increased plasma precursor protein catalyzed by membrane type 1 matrix metal-
levels of matrix metalloproteinase-9 in patients with Alzheimer’s loproteinase releases a fragment lacking the inhibitor domain
disease. Neurochem Int 43(3):191–196 against gelatinase A. Biochemistry 42(21):6514–6526. https​://
95. Lorenzl S, Buerger K, Hampel H, Beal MF (2008) Profiles of doi.org/10.1021/bi020​643m
matrix metalloproteinases and their inhibitors in plasma of 109. Miyazaki K, Hasegawa M, Funahashi K, Umeda M (1993)
patients with dementia. Int Psychogeriatr 20(1):67–76. https​:// A metalloproteinase inhibitor domain in Alzheimer amyloid
doi.org/10.1017/S1041​61020​70057​90 protein precursor. Nature 362(6423):839–841. https ​ : //doi.
96. Stomrud E, Bjorkqvist M, Janciauskiene S, Minthon L, Hansson org/10.1038/36283​9a0
O (2010) Alterations of matrix metalloproteinases in the healthy 110. Ahmad M, Takino T, Miyamori H, Yoshizaki T, Furukawa
elderly with increased risk of prodromal Alzheimer’s disease. M, Sato H (2006) Cleavage of amyloid-beta precursor protein
Alz Res Ther 2(3):20. https​://doi.org/10.1186/alzrt​44 (APP) by membrane-type matrix metalloproteinases. J Biochem
97. Hanzel CE, Iulita MF, Eyjolfsdottir H, Hjorth E, Schultzberg M, 139(3):517–526. https​://doi.org/10.1093/jb/mvj05​4
Eriksdotter M, Cuello AC (2014) Analysis of matrix metallo- 111. Willem M, Tahirovic S, Busche MA, Ovsepian SV, Chafai M,
proteases and the plasminogen system in mild cognitive impair- Kootar S, Hornburg D, Evans LD, Moore S, Daria A, Hampel H,
ment and Alzheimer’s disease cerebrospinal fluid. J Alzheimers Muller V, Giudici C, Nuscher B, Wenninger-Weinzierl A, Krem-
Dis 40(3):667–678. https​://doi.org/10.3233/JAD-13228​2 mer E, Heneka MT, Thal DR, Giedraitis V, Lannfelt L, Muller U,
98. Peng M, Jia J, Qin W (2015) Plasma gelsolin and matrix metal- Livesey FJ, Meissner F, Herms J, Konnerth A, Marie H, Haass C
loproteinase 3 as potential biomarkers for Alzheimer disease. (2015) eta-Secretase processing of APP inhibits neuronal activ-
Neurosci Lett 595:116–121. https​://doi.org/10.1016/j.neule​ ity in the hippocampus. Nature 526(7573):443–447. https​://doi.
t.2015.04.014 org/10.1038/natur​e1486​4
99. Selvais C, D’Auria L, Tyteca D, Perrot G, Lemoine P, Troeberg 112. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-
L, Dedieu S, Noel A, Nagase H, Henriet P, Courtoy PJ, Marbaix Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar
E, Emonard H (2011) Cell cholesterol modulates metallopro- R (2006) Intraneuronal beta-amyloid aggregates, neurodegen-
teinase-dependent shedding of low-density lipoprotein receptor- eration, and neuron loss in transgenic mice with five familial
related protein-1 (LRP-1) and clearance function. FASEB J Alzheimer’s disease mutations: potential factors in amyloid
25(8):2770–2781. https​://doi.org/10.1096/fj.10-16950​8 plaque formation. J Neurosci 26(40):10129–10140. https​://doi.
100. Liu Q, Zhang J, Tran H, Verbeek MM, Reiss K, Estus S, Bu org/10.1523/JNEUR​OSCI.1202-06.2006
G (2009) LRP1 shedding in human brain: roles of ADAM10 113. Komori K, Nonaka T, Okada A, Kinoh H, Hayashita-Kinoh H,
and ADAM17. Mol Neurodegener 4:17. https ​ : //doi. Yoshida N, Yana I, Seiki M (2004) Absence of mechanical allo-
org/10.1186/1750-1326-4-17 dynia and Abeta-fiber sprouting after sciatic nerve injury in mice
101. Raucci A, Cugusi S, Antonelli A, Barabino SM, Monti L, Bier- lacking membrane-type 5 matrix metalloproteinase. FEBS Lett
haus A, Reiss K, Saftig P, Bianchi ME (2008) A soluble form 557(1–3):125–128
of the receptor for advanced glycation endproducts (RAGE) is 114. Baranger K, Marchalant Y, Bonnet AE, Crouzin N, Carrete A,
produced by proteolytic cleavage of the membrane-bound form Paumier JM, Py NA, Bernard A, Bauer C, Charrat E, Moschke K,
by the sheddase a disintegrin and metalloprotease 10 (ADAM10). Seiki M, Vignes M, Lichtenthaler SF, Checler F, Khrestchatisky
FASEB J 22(10):3716–3727. https:​ //doi.org/10.1096/fj.08-10903​ M, Rivera S (2016) MT5-MMP is a new pro-amyloidogenic pro-
3 teinase that promotes amyloid pathology and cognitive decline in
102. Zhang L, Bukulin M, Kojro E, Roth A, Metz VV, Fahrenholz F, a transgenic mouse model of Alzheimer’s disease. Cell Mol Life
Nawroth PP, Bierhaus A, Postina R (2008) Receptor for advanced Sci 73(1):217–236. https​://doi.org/10.1007/s0001​8-015-1992-1
glycation end products is subjected to protein ectodomain shed- 115. Baranger K, Bonnet AE, Girard SD, Paumier JM, Garcia-Gon-
ding by metalloproteinases. J Biol Chem 283(51):35507–35516. zalez L, Elmanaa W, Bernard A, Charrat E, Stephan D, Bauer
https​://doi.org/10.1074/jbc.M8069​48200​ C, Moschke K, Lichtenthaler SF, Roman FS, Checler F, Khrest-
103. Metz VV, Kojro E, Rat D, Postina R (2012) Induction of RAGE chatisky M, Rivera S (2017) MT5-MMP Promotes Alzheimer’s
shedding by activation of G protein-coupled receptors. PLoS One Pathogenesis in the Frontal Cortex of 5xFAD Mice and APP
7(7):e41823. https​://doi.org/10.1371/journ​al.pone.00418​23 Trafficking in  vitro. Front Mol Neurosci 9:163. https​://doi.
104. Sagare A, Deane R, Bell RD, Johnson B, Hamm K, Pendu R, org/10.3389/fnmol​.2016.00163​
Marky A, Lenting PJ, Wu Z, Zarcone T, Goate A, Mayo K, Per- 116. Kimura R, Ohno M (2009) Impairments in remote memory sta-
lmutter D, Coma M, Zhong Z, Zlokovic BV (2007) Clearance bilization precede hippocampal synaptic and cognitive failures in
of amyloid-beta by circulating lipoprotein receptors. Nat Med 5XFAD Alzheimer mouse model. Neurobiol Dis 33(2):229–235.
13(9):1029–1031. https​://doi.org/10.1038/nm163​5 https​://doi.org/10.1016/j.nbd.2008.10.006
105. Terni B, Ferrer I (2015) Abnormal expression and distribution of 117. Crouzin N, Baranger K, Cavalier M, Marchalant Y, Cohen-Solal
MMP2 at initial stages of Alzheimer’s disease-related pathology. C, Roman FS, Khrestchatisky M, Rivera S, Feron F, Vignes M
J Alzheimers Dis 46(2):461–469. https​://doi.org/10.3233/JAD- (2013) Area-specific alterations of synaptic plasticity in the
14246​0 5XFAD mouse model of Alzheimer’s disease: dissociation
106. Nubling G, Levin J, Bader B, Israel L, Botzel K, Lorenzl S, Giese between somatosensory cortex and hippocampus. PLoS One
A (2012) Limited cleavage of tau with matrix-metalloproteinase 8(9):e74667. https​://doi.org/10.1371/journ​al.pone.00746​67
MMP-9, but not MMP-3, enhances tau oligomer formation. 118. Jawhar S, Trawicka A, Jenneckens C, Bayer TA, Wirths O (2012)
Exp Neurol 237(2):470–476. https​://doi.org/10.1016/j.expne​ Motor deficits, neuron loss, and reduced anxiety coinciding with
urol.2012.07.018 axonal degeneration and intraneuronal Abeta aggregation in the
107. Bjerke M, Zetterberg H, Edman A, Blennow K, Wallin A, 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging
Andreasson U (2011) Cerebrospinal fluid matrix metalloprotein- 33(1):196. https:​ //doi.org/10.1016/j.neurob​ iolag​ ing.2010.05.027
ases and tissue inhibitor of metalloproteinases in combination 119. Girard SD, Baranger K, Gauthier C, Jacquet M, Bernard A,
with subcortical and cortical biomarkers in vascular dementia Escoffier G, Marchetti E, Khrestchatisky M, Rivera S, Roman

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3187

FS (2013) Evidence for early cognitive impairment related to A, Mazaheri F, Tahirovic S, Lleo A, Alcolea D, Fortea J, Wil-
frontal cortex in the 5XFAD mouse model of Alzheimer’s dis- lem M, Lammich S, Molinuevo JL, Sanchez-Valle R, Antonell
ease. J Alzheimers Dis 33(3):781–796. https​://doi.org/10.3233/ A, Ramirez A, Heneka MT, Sleegers K, van der Zee J, Martin
JAD-2012-12098​2 JJ, Engelborghs S, Demirtas-Tatlidede A, Zetterberg H, Van
120. Baranger K, Giannoni P, Girard SD, Girot S, Gaven F, Stephan D, Broeckhoven C, Gurvit H, Wyss-Coray T, Hardy J, Colonna
Migliorati M, Khrestchatisky M, Bockaert J, Marchetti-Gauthier M, Haass C (2014) TREM2 mutations implicated in neuro-
E, Rivera S, Claeysen S, Roman FS (2017) Chronic treatments degeneration impair cell surface transport and phagocytosis.
with a 5-HT4 receptor agonist decrease amyloid pathology in the Sci Transl Med 6(243):243ra286. https​://doi.org/10.1126/scitr​
entorhinal cortex and learning and memory deficits in the 5xFAD anslm​ed.30090​93
mouse model of Alzheimer’s disease. Neuropharmacology 132. Thornton P, Sevalle J, Deery MJ, Fraser G, Zhou Y, Stahl S,
126:128–141. https:​ //doi.org/10.1016/j.neurop​ harm.​ 2017.08.031 Franssen EH, Dodd RB, Qamar S, Gomez Perez-Nievas B, Nicol
121. Wang P, Wang X, Pei D (2004) Mint-3 regulates the retrieval LS, Eketjall S, Revell J, Jones C, Billinton A, St George-Hyslop
of the internalized membrane-type matrix metalloproteinase, PH, Chessell I, Crowther DC (2017) TREM2 shedding by cleav-
MT5-MMP, to the plasma membrane by binding to its carboxyl age at the H157-S158 bond is accelerated for the Alzheimer’s
end motif EWV. J Biol Chem 279(19):20461–20470. https​://doi. disease-associated H157Y variant. EMBO Mol Med 9(10):1366–
org/10.1074/jbc.M4002​64200​ 1378. https​://doi.org/10.15252​/emmm.20170​7673
122. Laulagnier K, Javalet C, Hemming FJ, Chivet M, Lachenal G, 133. Feuerbach D, Schindler P, Barske C, Joller S, Beng-Louka E,
Blot B, Chatellard C, Sadoul R (2018) Amyloid precursor protein Worringer KA, Kommineni S, Kaykas A, Ho DJ, Ye C, Welzen-
products concentrate in a subset of exosomes specifically endocy- bach K, Elain G, Klein L, Brzak I, Mir AK, Farady CJ, Aich-
tosed by neurons. Cell Mol Life Sci 75(4):757–773. https​://doi. holz R, Popp S, George N, Neumann U (2017) ADAM17 is the
org/10.1007/s0001​8-017-2664-0 main sheddase for the generation of human triggering receptor
123. Zhang Z, Song M, Liu X, Su Kang S, Duong DM, Seyfried NT, expressed in myeloid cells (hTREM2) ectodomain and cleaves
Cao X, Cheng L, Sun YE, Ping YuS, Jia J, Levey AI, Ye K TREM2 after Histidine 157. Neurosci Lett 660:109–114. https​
(2015) Delta-secretase cleaves amyloid precursor protein and ://doi.org/10.1016/j.neule​t.2017.09.034
regulates the pathogenesis in Alzheimer’s disease. Nat Comm 134. Carmona S, Zahs K, Wu E, Dakin K, Bras J, Guerreiro R (2018)
6:8762. https​://doi.org/10.1038/ncomm​s9762​ The role of TREM2 in Alzheimer’s disease and other neurode-
124. Paumier JM, Py NA, Garcia-Gonzalez L, Bernard A, Stephan generative disorders. Lancet Neurol 17(8):721–730. https​://doi.
D, Louis L, Checler F, Khrestchatisky M, Baranger K, Rivera org/10.1016/S1474​-4422(18)30232​-1
S (2018) Proamyloidogenic effects of membrane type 1 matrix 135. Black RA, Rauch CT, Kozlosky CJ, Peschon JJ, Slack JL, Wolf-
metalloproteinase involve MMP-2 and BACE-1 activities, son MF, Castner BJ, Stocking KL, Reddy P, Srinivasan S, Nel-
and the modulation of APP trafficking. FASEB J. https​://doi. son N, Boiani N, Schooley KA, Gerhart M, Davis R, Fitzner
org/10.1096/fj.20180​1076r​ JN, Johnson RS, Paxton RJ, March CJ, Cerretti DP (1997) A
125. Uekita T, Itoh Y, Yana I, Ohno H, Seiki M (2001) Cytoplasmic metalloproteinase disintegrin that releases tumour-necrosis
tail-dependent internalization of membrane-type 1 matrix met- factor-alpha from cells. Nature 385(6618):729–733. https​://doi.
alloproteinase is important for its invasion-promoting activity. J org/10.1038/38572​9a0
Cell Biol 155(7):1345–1356. https​://doi.org/10.1083/jcb.20010​ 136. Decourt B, Lahiri DK, Sabbagh MN (2017) Targeting tumor
8112 necrosis factor alpha for Alzheimer’s disease. Curr Alz Res
126. Perez RG, Soriano S, Hayes JD, Ostaszewski B, Xia W, Selkoe 14(4):412–425. https:​ //doi.org/10.2174/156720​ 50136​ 66160​ 9301​
DJ, Chen X, Stokin GB, Koo EH (1999) Mutagenesis identi- 10551​
fies new signals for beta-amyloid precursor protein endocytosis, 137. Peress N, Perillo E, Zucker S (1995) Localization of tissue inhibi-
turnover, and the generation of secreted fragments, including tor of matrix metalloproteinases in Alzheimer’s disease and nor-
Abeta42. J Biol Chem 274(27):18851–18856 mal brain. J Neuropathol Exp Neurol 54(1):16–22
127. Caster AH, Kahn RA (2013) Recruitment of the Mint3 adaptor is 138. Lorenzl S, Albers DS, LeWitt PA, Chirichigno JW, Hilgenberg
necessary for export of the amyloid precursor protein (APP) from SL, Cudkowicz ME, Beal MF (2003) Tissue inhibitors of matrix
the Golgi complex. J Biol Chem 288(40):28567–28580. https​:// metalloproteinases are elevated in cerebrospinal fluid of neuro-
doi.org/10.1074/jbc.M113.48110​1 degenerative diseases. J Neurol Sci 207(1–2):71–76
128. Vincent B, Checler F (2012) alpha-Secretase in Alzheimer’s 139. Marksteiner J, Imarhiagbe D, Defrancesco M, Deisenhammer
disease and beyond: mechanistic, regulation and function in the EA, Kemmler G, Humpel C (2014) Analysis of 27 vascular-
shedding of membrane proteins. Curr Alz Res 9(2):140–156 related proteins reveals that NT-proBNP is a potential biomarker
129. Saftig P, Lichtenthaler SF (2015) The alpha secretase for Alzheimer’s disease and mild cognitive impairment: a pilot-
ADAM10: a metalloprotease with multiple functions in the study. Exp Gerontol 50:114–121. https​://doi.org/10.1016/j.exger​
brain. Prog Neurobiol 135:1–20. https​://doi.org/10.1016/j.pneur​ .2013.12.001
obio.2015.10.003 140. Ridnour LA, Dhanapal S, Hoos M, Wilson J, Lee J, Cheng RY,
130. Letronne F, Laumet G, Ayral AM, Chapuis J, Demiautte F, Laga Brueggemann EE, Hines HB, Wilcock DM, Vitek MP, Wink DA,
M, Vandenberghe ME, Malmanche N, Leroux F, Eysert F, Sot- Colton CA (2012) Nitric oxide-mediated regulation of beta-amy-
tejeau Y, Chami L, Flaig A, Bauer C, Dourlen P, Lesaffre M, loid clearance via alterations of MMP-9/TIMP-1. J Neurochem
Delay C, Huot L, Dumont J, Werkmeister E, Lafont F, Mendes 123(5):736–749. https​://doi.org/10.1111/jnc.12028​
T, Hansmannel F, Dermaut B, Deprez B, Herard AS, Dhenain M, 141. Duits FH, Hernandez-Guillamon M, Montaner J, Goos JD, Mon-
Souedet N, Pasquier F, Tulasne D, Berr C, Hauw JJ, Lemoine Y, tanola A, Wattjes MP, Barkhof F, Scheltens P, Teunissen CE, van
Amouyel P, Mann D, Deprez R, Checler F, Hot D, Delzescaux T, der Flier WM (2015) Matrix metalloproteinases in Alzheimer’s
Gevaert K, Lambert JC (2016) ADAM30 Downregulates APP- disease and concurrent cerebral microbleeds. J Alzheimers Dis
Linked defects through cathepsin D activation in Alzheimer’s 48(3):711–720. https​://doi.org/10.3233/JAD-14318​6
disease. EBioMedicine 9:278–292. https​://doi.org/10.1016/j. 142. Hernandez-Guillamon M, Delgado P, Ortega L, Pares M, Rosell
ebiom​.2016.06.002 A, Garcia-Bonilla L, Fernandez-Cadenas I, Borrell-Pages M,
131. Kleinberger G, Yamanishi Y, Suarez-Calvet M, Czirr E, Lohm- Boada M, Montaner J (2009) Neuronal TIMP-1 release accom-
ann E, Cuyvers E, Struyfs H, Pettkus N, Wenninger-Weinzierl panies astrocytic MMP-9 secretion and enhances astrocyte

13

3188 S. Rivera et al.

proliferation induced by beta-amyloid 25-35 fragment. J Neuro- Parkinson and Lewy body dementia. Ann Neurol 55(2):164–
sci Res 87(9):2115–2125. https​://doi.org/10.1002/jnr.22034​ 173. https​://doi.org/10.1002/ana.10795​
143. Ogier C, Creidy R, Boucraut J, Soloway PD, Khrestchatisky M, 155. Ghosh D, Mehra S, Sahay S, Singh PK, Maji SK (2017) alpha-
Rivera S (2005) Astrocyte reactivity to Fas activation is attenu- synuclein aggregation and its modulation. Int J Biol Macromol
ated in TIMP-1 deficient mice, an in vitro study. BMC Neurosci 100:37–54. https​://doi.org/10.1016/j.ijbio​mac.2016.10.021
6:68. https​://doi.org/10.1186/1471-2202-6-68 156. Longhena F, Faustini G, Missale C, Pizzi M, Spano P, Bellucci
144. Thirumangalakudi L, Samany PG, Owoso A, Wiskar B, Gram- A (2017) The contribution of alpha-synuclein spreading to Par-
mas P (2006) Angiogenic proteins are expressed by brain blood kinson’s disease synaptopathy. Neural Plast 2017:5012129.
vessels in Alzheimer’s disease. J Alzheimers Dis 10(1):111–118 https​://doi.org/10.1155/2017/50121​29
145. Park SK, Hwang YS, Park KK, Park HJ, Seo JY, Chung WY 157. Olanow CW, Brundin P (2013) Parkinson’s disease and alpha
(2009) Kalopanaxsaponin A inhibits PMA-induced invasion synuclein: is Parkinson’s disease a prion-like disorder? Mov
by reducing matrix metalloproteinase-9 via PI3K/Akt- and Dis 28(1):31–40. https​://doi.org/10.1002/mds.25373​
PKCdelta-mediated signaling in MCF-7 human breast cancer 158. Wakabayashi K, Tanji K, Odagiri S, Miki Y, Mori F, Takahashi
cells. Carcinogenesis 30(7):1225–1233. https​://doi.org/10.1093/ H (2013) The Lewy body in Parkinson’s disease and related
carci​n/bgp11​1 neurodegenerative disorders. Mol Neurobiol 47(2):495–508.
146. Castellano JM, Mosher KI, Abbey RJ, McBride AA, James ML, https​://doi.org/10.1007/s1203​5-012-8280-y
Berdnik D, Shen JC, Zou B, Xie XS, Tingle M, Hinkson IV, 159. Milber JM, Noorigian JV, Morley JF, Petrovitch H, White
Angst MS, Wyss-Coray T (2017) Human umbilical cord plasma L, Ross GW, Duda JE (2012) Lewy pathology is not the first
proteins revitalize hippocampal function in aged mice. Nature sign of degeneration in vulnerable neurons in Parkinson dis-
544(7651):488–492. https​://doi.org/10.1038/natur​e2206​7 ease. Neurology 79(24):2307–2314. https​://doi.org/10.1212/
147. Hoe HS, Cooper MJ, Burns MP, Lewis PA, van der Brug M, WNL.0b013​e3182​78fe3​2
Chakraborty G, Cartagena CM, Pak DT, Cookson MR, Rebeck 160. Ding TT, Lee SJ, Rochet JC, Lansbury PT Jr (2002) Annu-
GW (2007) The metalloprotease inhibitor TIMP-3 regulates amy- lar alpha-synuclein protofibrils are produced when spherical
loid precursor protein and apolipoprotein E receptor proteolysis. protofibrils are incubated in solution or bound to brain-derived
J Neurosci 27(40):10895–10905. https:​ //doi.org/10.1523/JNEUR​ membranes. Biochemistry 41(32):10209–10217
OSCI.3135-07.2007 161. Lashuel HA, Hartley D, Petre BM, Walz T, Lansbury PT
148. Dunckley T, Beach TG, Ramsey KE, Grover A, Mastroeni D, Jr (2002) Neurodegenerative disease: amyloid pores from
Walker DG, LaFleur BJ, Coon KD, Brown KM, Caselli R, Kukull pathogenic mutations. Nature 418(6895):291. https​: //doi.
W, Higdon R, McKeel D, Morris JC, Hulette C, Schmechel D, org/10.1038/41829​1a
Reiman EM, Rogers J, Stephan DA (2006) Gene expression cor- 162. Lashuel HA, Petre BM, Wall J, Simon M, Nowak RJ, Walz T,
relates of neurofibrillary tangles in Alzheimer’s disease. Neuro- Lansbury PT Jr (2002) Alpha-synuclein, especially the Parkin-
biol Aging 27(10):1359–1371. https​://doi.org/10.1016/j.neuro​ son’s disease-associated mutants, forms pore-like annular and
biola​ging.2005.08.013 tubular protofibrils. J Mol Biol 322(5):1089–1102
149. Qin W, Jia X, Wang F, Zuo X, Wu L, Zhou A, Li D, Min B, Wei 163. Baba M, Nakajo S, Tu PH, Tomita T, Nakaya K, Lee VM,
C, Tang Y, Xing Y, Dong X, Wang Q, Gao Y, Li Y, Jia J (2015) Trojanowski JQ, Iwatsubo T (1998) Aggregation of alpha-
Elevated plasma angiogenesis factors in Alzheimer’s disease. J synuclein in Lewy bodies of sporadic Parkinson’s disease and
Alzheimers Dis 45(1):245–252. https​://doi.org/10.3233/JAD- dementia with Lewy bodies. Am J Pathol 152(4):879–884
14240​9 164. Lee MK, Stirling W, Xu Y, Xu X, Qui D, Mandir AS, Daw-
150. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, son TM, Copeland NG, Jenkins NA, Price DL (2002) Human
Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, alpha-synuclein-harboring familial Parkinson’s disease-linked
Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Ala-53 –> Thr mutation causes neurodegenerative disease with
Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe alpha-synuclein aggregation in transgenic mice. Proc Natl
LI, Nussbaum RL (1997) Mutation in the alpha-synuclein Acad Sci USA 99(13):8968–8973. https​: //doi.org/10.1073/
gene identified in families with Parkinson’s disease. Science pnas.13219​7599
276(5321):2045–2047 165. Choi DH, Kim YJ, Kim YG, Joh TH, Beal MF, Kim YS
151. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, (2011) Role of matrix metalloproteinase 3-mediated alpha-
Przuntek H, Epplen JT, Schols L, Riess O (1998) Ala30Pro muta- synuclein cleavage in dopaminergic cell death. J Biol
tion in the gene encoding alpha-synuclein in Parkinson’s disease. Chem 286(16):14168–14177. https​: //doi.org/10.1074/jbc.
Nat Genet 18(2):106–108. https​://doi.org/10.1038/ng029​8-106 M111.22243​0
152. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kach- 166. Crowther RA, Jakes R, Spillantini MG, Goedert M (1998) Syn-
ergus J, Hulihan M, Peuralinna T, Dutra A, Nussbaum R, Lincoln thetic filaments assembled from C-terminally truncated alpha-
S, Crawley A, Hanson M, Maraganore D, Adler C, Cookson MR, synuclein. FEBS Lett 436(3):309–312
Muenter M, Baptista M, Miller D, Blancato J, Hardy J, Gwinn- 167. Kanda S, Bishop JF, Eglitis MA, Yang Y, Mouradian MM
Hardy K (2003) alpha-Synuclein locus triplication causes Parkin- (2000) Enhanced vulnerability to oxidative stress by alpha-
son’s disease. Science 302(5646):841. https​://doi.org/10.1126/ synuclein mutations and C-terminal truncation. Neuroscience
scien​ce.10902​78 97(2):279–284
153. Chartier-Harlin MC, Kachergus J, Roumier C, Mouroux V, 168. Sung JY, Park SM, Lee CH, Um JW, Lee HJ, Kim J, Oh YJ, Lee
Douay X, Lincoln S, Levecque C, Larvor L, Andrieux J, Hulihan ST, Paik SR, Chung KC (2005) Proteolytic cleavage of extracel-
M, Waucquier N, Defebvre L, Amouyel P, Farrer M, Destee A lular secreted {alpha}-synuclein via matrix metalloproteinases.
(2004) Alpha-synuclein locus duplication as a cause of familial J Biol Chem 280(26):25216–25224. https​://doi.org/10.1074/jbc.
Parkinson’s disease. Lancet 364(9440):1167–1169. https​://doi. M5033​41200​
org/10.1016/S0140​-6736(04)17103​-1 169. Levin J, Giese A, Boetzel K, Israel L, Hogen T, Nubling G,
154. Zarranz JJ, Alegre J, Gomez-Esteban JC, Lezcano E, Ros R, Kretzschmar H, Lorenzl S (2009) Increased alpha-synuclein
Ampuero I, Vidal L, Hoenicka J, Rodriguez O, Atares B, Llo- aggregation following limited cleavage by certain matrix
rens V, Gomez Tortosa E, del Ser T, Munoz DG, de Yebenes metalloproteinases. Exp Neurol 215(1):201–208. https​://doi.
JG (2004) The new mutation, E46K, of alpha-synuclein causes org/10.1016/j.expne​urol.2008.10.010

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3189

170. Kim YS, Choi DH, Block ML, Lorenzl S, Yang L, Kim YJ, Sug- 183. Paisan-Ruiz C, Jain S, Evans EW, Gilks WP, Simon J, van der
ama S, Cho BP, Hwang O, Browne SE, Kim SY, Hong JS, Beal Brug M, Lopez de Munain A, Aparicio S, Gil AM, Khan N,
MF, Joh TH (2007) A pivotal role of matrix metalloproteinase-3 Johnson J, Martinez JR, Nicholl D, Carrera IM, Pena AS, de
activity in dopaminergic neuronal degeneration via microglial Silva R, Lees A, Marti-Masso JF, Perez-Tur J, Wood NW, Sin-
activation. FASEB J 21(1):179–187. https​://doi.org/10.1096/ gleton AB (2004) Cloning of the gene containing mutations that
fj.06-5865c​om cause PARK8-linked Parkinson’s disease. Neuron 44(4):595–
171. Chung YC, Kim YS, Bok E, Yune TY, Maeng S, Jin BK (2013) 600. https​://doi.org/10.1016/j.neuro​n.2004.10.023
MMP-3 contributes to nigrostriatal dopaminergic neuronal loss, 184. Cookson MR (2010) The role of leucine-rich repeat kinase 2
BBB damage, and neuroinflammation in an MPTP mouse model (LRRK2) in Parkinson’s disease. Nat Rev Neurosci 11(12):791–
of Parkinson’s disease. Mediat Inflamm 2013:370526. https:​ //doi. 797. https​://doi.org/10.1038/nrn29​35
org/10.1155/2013/37052​6 185. Caesar M, Felk S, Zach S, Bronstad G, Aasly JO, Gasser T,
172. Choi DH, Kim EM, Son HJ, Joh TH, Kim YS, Kim D, Flint Gillardon F (2014) Changes in matrix metalloprotease activity
Beal M, Hwang O (2008) A novel intracellular role of matrix and progranulin levels may contribute to the pathophysiological
metalloproteinase-3 during apoptosis of dopaminergic function of mutant leucine-rich repeat kinase 2. Glia 62(7):1075–
cells. J Neurochem 106(1):405–415. https​://doi.org/10.111 1092. https​://doi.org/10.1002/glia.22663​
1/j.1471-4159.2008.05399​.x 186. Suh HS, Choi N, Tarassishin L, Lee SC (2012) Regulation of
173. Kim YS, Kim SS, Cho JJ, Choi DH, Hwang O, Shin DH, Chun progranulin expression in human microglia and proteolysis of
HS, Beal MF, Joh TH (2005) Matrix metalloproteinase-3: a novel progranulin by matrix metalloproteinase-12 (MMP-12). PLoS
signaling proteinase from apoptotic neuronal cells that activates One 7(4):e35115. https​://doi.org/10.1371/journ​al.pone.00351​15
microglia. J Neurosci 25(14):3701–3711. https:​ //doi.org/10.1523/ 187. Bachiller S, Jimenez-Ferrer I, Paulus A, Yang Y, Swanberg M,
JNEUR​OSCI.4346-04.2005 Deierborg T, Boza-Serrano A (2018) Microglia in neurological
174. Lee EJ, Woo MS, Moon PG, Baek MC, Choi IY, Kim WK, Junn diseases: a road map to brain-disease dependent-inflammatory
E, Kim HS (2010) Alpha-synuclein activates microglia by induc- response. Front Cell Neurosci 12:488. https​://doi.org/10.3389/
ing the expressions of matrix metalloproteinases and the sub- fncel​.2018.00488​
sequent activation of protease-activated receptor-1. J Immunol 188. Oh SH, Kim HN, Park HJ, Shin JY, Kim DY, Lee PH (2017) The
185(1):615–623. https​://doi.org/10.4049/jimmu​nol.09034​80 cleavage effect of mesenchymal stem cell and its derived matrix
175. Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, metalloproteinase-2 on extracellular alpha-synuclein aggregates
Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van in Parkinsonian models. Stem Cells Translat Med 6(3):949–961.
Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van https​://doi.org/10.5966/sctm.2016-0111
Duijn CM, Oostra BA, Heutink P (2003) Mutations in the DJ-1 189. Lorenzl S, Albers DS, Narr S, Chirichigno J, Beal MF (2002)
gene associated with autosomal recessive early-onset parkinson- Expression of MMP-2, MMP-9, and MMP-1 and their endog-
ism. Science 299(5604):256–259. https​://doi.org/10.1126/scien​ enous counterregulators TIMP-1 and TIMP-2 in postmortem
ce.10772​09 brain tissue of Parkinson’s disease. Exp Neurol 178(1):13–20
176. Choi DH, Hwang O, Lee KH, Lee J, Beal MF, Kim YS (2011) 190. Kim SY, Woo MS, Park JS, Hyun JW, Kim YS, Kim HS (2010)
DJ-1 cleavage by matrix metalloproteinase 3 mediates oxida- The neuroprotective role of tissue inhibitor of metalloprotein-
tive stress-induced dopaminergic cell death. Antiox Redox Sign ase-2 in MPP+- or 6-OHDA-treated SK-N-BE(2)C and SH-
14(11):2137–2150. https​://doi.org/10.1089/ars.2009.3059 SY5Y human neuroblastoma cells. Neurosci Lett 468(2):136–
177. Miller DW, Ahmad R, Hague S, Baptista MJ, Canet-Aviles 140. https​://doi.org/10.1016/j.neule​t.2009.10.084
R, McLendon C, Carter DM, Zhu PP, Stadler J, Chandran J, 191. Kim J, Jeong YH, Lee EJ, Park JS, Seo H, Kim HS (2017) Sup-
Klinefelter GR, Blackstone C, Cookson MR (2003) L166P pression of neuroinflammation by matrix metalloproteinase-8
mutant DJ-1, causative for recessive Parkinson’s disease, is inhibitor in aged normal and LRRK2 G2019S Parkinson’s dis-
degraded through the ubiquitin-proteasome system. J Biol Chem ease model mice challenged with lipopolysaccharide. Biochem
278(38):36588–36595. https:​ //doi.org/10.1074/jbc.M30427​ 2200​ Biophys Res Comm 493(2):879–886. https​://doi.org/10.1016/j.
178. Kim YS, Joh TH (2012) Matrix metalloproteinases, new insights bbrc.2017.09.129
into the understanding of neurodegenerative disorders. Bio- 192. Lorenzl S, Calingasan N, Yang L, Albers DS, Shugama S, Gre-
mol Ther 20(2):133–143. https​://doi.org/10.4062/biomo​lther​ gorio J, Krell HW, Chirichigno J, Joh T, Beal MF (2004) Matrix
.2012.20.2.133 metalloproteinase-9 is elevated in 1-methyl-4-phenyl-1,2,3,6-
179. Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich tetrahydropyridine-induced parkinsonism in mice. Neuromol
A (2004) DJ-1 is a redox-dependent molecular chaperone Med 5(2):119–132. https​://doi.org/10.1385/nmm:5:2:119
that inhibits alpha-synuclein aggregate formation. PLoS Biol 193. Logroscino G, Tortelli R, Rizzo G, Marin B, Preux P, Malaspina
2(11):e362. https​://doi.org/10.1371/journ​al.pbio.00203​62 A (2015) Amyotrophic lateral sclerosis: an aging-related disease.
180. Butler GS, Dean RA, Tam EM, Overall CM (2008) Pharma- Curr Geri Rep 4:142. https:​ //doi.org/10.1007/s13670​ -015-0127-8
coproteomics of a metalloproteinase hydroxamate inhibitor in 194. Lukaszewicz-Zajac M, Mroczko B, Slowik A (2014) Matrix
breast cancer cells: dynamics of membrane type 1 matrix met- metalloproteinases (MMPs) and their tissue inhibitors (TIMPs)
alloproteinase-mediated membrane protein shedding. Mol Cell in amyotrophic lateral sclerosis (ALS). J Neural Trans
Biol 28(15):4896–4914. https​://doi.org/10.1128/MCB.01775​-07 121(11):1387–1397. https:​ //doi.org/10.1007/s00702​ -014-1205-3
181. Joo SH, Kwon KJ, Kim JW, Kim JW, Hasan MR, Lee HJ, Han 195. Dewil M, Schurmans C, Starckx S, Opdenakker G, Van Den
SH, Shin CY (2010) Regulation of matrix metalloproteinase-9 Bosch L, Robberecht W (2005) Role of matrix metalloprotein-
and tissue plasminogen activator activity by alpha-synuclein in ase-9 in a mouse model for amyotrophic lateral sclerosis. Neu-
rat primary glial cells. Neurosci Lett 469(3):352–356. https:​ //doi. roReport 16(4):321–324
org/10.1016/j.neule​t.2009.12.026 196. Kiaei M, Kipiani K, Calingasan NY, Wille E, Chen J, Heissig
182. Kim S, Cho SH, Kim KY, Shin KY, Kim HS, Park CH, Chang B, Rafii S, Lorenzl S, Beal MF (2007) Matrix metalloprotein-
KA, Lee SH, Cho D, Suh YH (2009) Alpha-synuclein induces ase-9 regulates TNF-alpha and FasL expression in neuronal, glial
migration of BV-2 microglial cells by up-regulation of CD44 and cells and its absence extends life in a transgenic mouse model of
MT1-MMP. J Neurochem 109(5):1483–1496. https​://doi.org/10. amyotrophic lateral sclerosis. Exp Neurol 205(1):74–81. https​://
1111/j.1471-4159.2009.06075​.x doi.org/10.1016/j.expne​urol.2007.01.036

13

3190 S. Rivera et al.

197. Kaplan A, Spiller KJ, Towne C, Kanning KC, Choe GT, Geber 209. Liu Y, Hao W, Dawson A, Liu S, Fassbender K (2009) Expres-
A, Akay T, Aebischer P, Henderson CE (2014) Neuronal matrix sion of amyotrophic lateral sclerosis-linked SOD1 mutant
metalloproteinase-9 is a determinant of selective neurodegen- increases the neurotoxic potential of microglia via TLR2. J Biol
eration. Neuron 81(2):333–348. https​://doi.org/10.1016/j.neuro​ Chem 284(6):3691–3699. https​://doi.org/10.1074/jbc.M8044​
n.2013.12.009 46200​
198. Morisaki Y, Niikura M, Watanabe M, Onishi K, Tanabe S, Mori- 210. Lee JK, Shin JH, Gwag BJ, Choi EJ (2015) Iron accumulation
waki Y, Okuda T, Ohara S, Murayama S, Takao M, Uchida S, promotes TACE-mediated TNF-alpha secretion and neurodegen-
Yamanaka K, Misawa H (2016) Selective expression of osteo- eration in a mouse model of ALS. Neurobiol Dis 80:63–69. https​
pontin in ALS-resistant motor neurons is a critical determinant ://doi.org/10.1016/j.nbd.2015.05.009
of late phase neurodegeneration mediated by matrix metallopro- 211. Lemarchant S, Pomeshchik Y, Kidin I, Karkkainen V, Valonen
teinase-9. Sci Rep 6:27354. https​://doi.org/10.1038/srep2​7354 P, Lehtonen S, Goldsteins G, Malm T, Kanninen K, Koistinaho
199. Lorenzl S, Narr S, Angele B, Krell HW, Gregorio J, Kiaei M, J (2016) ADAMTS-4 promotes neurodegeneration in a mouse
Pfister HW, Beal MF (2006) The matrix metalloproteinases model of amyotrophic lateral sclerosis. Mol Neurodegener 11:10.
inhibitor Ro 28-2653 [correction of Ro 26-2853] extends survival https​://doi.org/10.1186/s1302​4-016-0078-3
in transgenic ALS mice. Exp Neurol 200(1):166–171. https:​ //doi. 212. DiFiglia M, Sapp E, Chase KO, Davies SW, Bates GP, Vonsat-
org/10.1016/j.expne​urol.2006.01.026 tel JP, Aronin N (1997) Aggregation of huntingtin in neuronal
200. Vaz AR, Cunha C, Gomes C, Schmucki N, Barbosa M, Brites intranuclear inclusions and dystrophic neurites in brain. Science
D (2015) Glycoursodeoxycholic acid reduces matrix metal- 277(5334):1990–1993
loproteinase-9 and caspase-9 activation in a cellular model of 213. Sapp E, Schwarz C, Chase K, Bhide PG, Young AB, Penney J,
superoxide dismutase-1 neurodegeneration. Mol Neurobiol Vonsattel JP, Aronin N, DiFiglia M (1997) Huntingtin localiza-
51(3):864–877. https​://doi.org/10.1007/s1203​5-014-8731-8 tion in brains of normal and Huntington’s disease patients. Ann
201. Spiller KJ, Khan T, Dominique MA, Restrepo CR, Cotton- Neurol 42(4):604–612. https​://doi.org/10.1002/ana.41042​0411
Samuel D, Levitan M, Jafar-Nejad P, Zhang B, Soriano A, Rigo 214. Wheeler VC, White JK, Gutekunst CA, Vrbanac V, Weaver M,
F, Trojanowski JQ, Lee VM (2019) Reduction of matrix met- Li XJ, Li SH, Yi H, Vonsattel JP, Gusella JF, Hersch S, Auer-
alloproteinase 9 (MMP-9) protects motor neurons from TDP- bach W, Joyner AL, MacDonald ME (2000) Long glutamine
43-triggered death in rNLS8 mice. Neurobiol Dis 124:133–140. tracts cause nuclear localization of a novel form of huntingtin in
https​://doi.org/10.1016/j.nbd.2018.11.013 medium spiny striatal neurons in HdhQ92 and HdhQ111 knock-
202. Soon CP, Crouch PJ, Turner BJ, McLean CA, Laughton KM, in mice. Hum Mol Genet 9(4):503–513
Atkin JD, Masters CL, White AR, Li QX (2010) Serum matrix 215. Wellington CL, Singaraja R, Ellerby L, Savill J, Roy S, Leavitt B,
metalloproteinase-9 activity is dysregulated with disease progres- Cattaneo E, Hackam A, Sharp A, Thornberry N, Nicholson DW,
sion in the mutant SOD1 transgenic mice. Neuromusc Disord Bredesen DE, Hayden MR (2000) Inhibiting caspase cleavage of
20(4):260–266. https​://doi.org/10.1016/j.nmd.2009.11.015 huntingtin reduces toxicity and aggregate formation in neuronal
203. Izrael M, Slutsky SG, Admoni T, Cohen L, Granit A, Hasson A, and nonneuronal cells. J Biol Chem 275(26):19831–19838. https​
Itskovitz-Eldor J, Krush Paker L, Kuperstein G, Lavon N, Yehez- ://doi.org/10.1074/jbc.M0014​75200​
kel Ionescu S, Solmesky LJ, Zaguri R, Zhuravlev A, Volman E, 216. Hermel E, Gafni J, Propp SS, Leavitt BR, Wellington CL, Young
Chebath J, Revel M (2018) Safety and efficacy of human embry- JE, Hackam AS, Logvinova AV, Peel AL, Chen SF, Hook V, Sin-
onic stem cell-derived astrocytes following intrathecal transplan- garaja R, Krajewski S, Goldsmith PC, Ellerby HM, Hayden MR,
tation in SOD1(G93A) and NSG animal models. Stem Cell Res Bredesen DE, Ellerby LM (2004) Specific caspase interactions
Ther 9(1):152. https​://doi.org/10.1186/s1328​7-018-0890-5 and amplification are involved in selective neuronal vulnerability
204. Lee JK, Shin JH, Suh J, Choi IS, Ryu KS, Gwag BJ (2008) in Huntington’s disease. Cell Death Diff 11(4):424–438. https​://
Tissue inhibitor of metalloproteinases-3 (TIMP-3) expression doi.org/10.1038/sj.cdd.44013​58
is increased during serum deprivation-induced neuronal apop- 217. Gafni J, Hermel E, Young JE, Wellington CL, Hayden MR,
tosis in vitro and in the G93A mouse model of amyotrophic Ellerby LM (2004) Inhibition of calpain cleavage of hunting-
lateral sclerosis: a potential modulator of Fas-mediated apop- tin reduces toxicity: accumulation of calpain/caspase fragments
tosis. Neurobiol Dis 30(2):174–185. https​://doi.org/10.1016/j. in the nucleus. J Biol Chem 279(19):20211–20220. https​://doi.
nbd.2008.01.004 org/10.1074/jbc.M4012​67200​
205. Lim GP, Backstrom JR, Cullen MJ, Miller CA, Atkinson RD, 218. Miller JP, Holcomb J, Al-Ramahi I, de Haro M, Gafni J, Zhang
Tokes ZA (1996) Matrix metalloproteinases in the neocortex and N, Kim E, Sanhueza M, Torcassi C, Kwak S, Botas J, Hughes
spinal cord of amyotrophic lateral sclerosis patients. J Neuro- RE, Ellerby LM (2010) Matrix metalloproteinases are modifi-
chem 67(1):251–259 ers of huntingtin proteolysis and toxicity in Huntington’s dis-
206. Beuche W, Yushchenko M, Mader M, Maliszewska M, Felgen- ease. Neuron 67(2):199–212. https​://doi.org/10.1016/j.neuro​
hauer K, Weber F (2000) Matrix metalloproteinase-9 is elevated n.2010.06.021
in serum of patients with amyotrophic lateral sclerosis. NeuroRe- 219. Naphade S, Embusch A, Madushani KL, Ring KL, Ellerby
port 11(16):3419–3422 LM (2017) Altered expression of matrix metalloproteinases
207. Fang L, Huber-Abel F, Teuchert M, Hendrich C, Dorst J, Schat- and their endogenous inhibitors in a human isogenic stem cell
tauer D, Zettlmeissel H, Wlaschek M, Scharffetter-Kochanek model of Huntington’s disease. Front Neurosci 11:736. https​://
K, Tumani H, Ludolph AC, Brettschneider J (2009) Linking doi.org/10.3389/fnins​.2017.00736​
neuron and skin: matrix metalloproteinases in amyotrophic lat- 220. Silvestroni A, Faull RL, Strand AD, Moller T (2009) Distinct
eral sclerosis (ALS). J Neurol Sci 285(1–2):62–66. https​://doi. neuroinflammatory profile in post-mortem human Hunting-
org/10.1016/j.jns.2009.05.025 ton’s disease. NeuroReport 20(12):1098–1103. https​: //doi.
208. Niebroj-Dobosz I, Janik P, Sokolowska B, Kwiecinski H (2010) org/10.1097/WNR.0b013​e3283​2e34e​e
Matrix metalloproteinases and their tissue inhibitors in serum 221. Chang KH, Wu YR, Chen YC, Chen CM (2015) Plasma inflam-
and cerebrospinal fluid of patients with amyotrophic lateral matory biomarkers for Huntington’s disease patients and
sclerosis. Eur J Neurol 17(2):226–231. https​://doi.org/10.111 mouse model. Brain Behav Immun 44:121–127. https​://doi.
1/j.1468-1331.2009.02775​.x org/10.1016/j.bbi.2014.09.011

13
Metalloproteinases and their tissue inhibitors in Alzheimer’s disease and other… 3191

222. Connolly C, Magnusson-Lind A, Lu G, Wagner PK, Southwell Neuroinfln 3(6):e287. https​://doi.org/10.1212/NXI.00000​00000​


AL, Hayden MR, Bjorkqvist M, Leavitt BR (2016) Enhanced 00028​7
immune response to MMP3 stimulation in microglia express- 225. Lo Sardo V, Zuccato C, Gaudenzi G, Vitali B, Ramos C, Tar-
ing mutant huntingtin. Neuroscience 325:74–88. https​://doi. tari M, Myre MA, Walker JA, Pistocchi A, Conti L, Valenza M,
org/10.1016/j.neuro​scien​ce.2016.03.031 Drung B, Schmidt B, Gusella J, Zeitlin S, Cotelli F, Cattaneo E
223. Duran-Vilaregut J, del Valle J, Manich G, Camins A, Pallas (2012) An evolutionary recent neuroepithelial cell adhesion func-
M, Vilaplana J, Pelegri C (2011) Role of matrix metallopro- tion of huntingtin implicates ADAM10-Ncadherin. Nat Neurosci
teinase-9 (MMP-9) in striatal blood-brain barrier disruption in 15(5):713–721. https​://doi.org/10.1038/nn.3080
a 3-nitropropionic acid model of Huntington’s disease. Neu-
ropathol Appl Neuro 37(5):525–537. https​://doi.org/10.111 Publisher’s Note Springer Nature remains neutral with regard to
1/j.1365-2990.2010.01157​.x jurisdictional claims in published maps and institutional affiliations.
224. Vinther-Jensen T, Bornsen L, Budtz-Jorgensen E, Ammitzboll
C, Larsen IU, Hjermind LE, Sellebjerg F, Nielsen JE (2016)
Selected CSF biomarkers indicate no evidence of early neu-
roinflammation in Huntington disease. Neurol Neuroimmun

13

You might also like