Accepted Manuscript Leptin and ghrelin: Sewing metabolism onto neurodegeneration Paola de Candia, Giuseppe Matarese PII:
S0028-3908(17)30622-6
DOI:
10.1016/j.neuropharm.2017.12.025
Reference:
NP 7002
To appear in:
Neuropharmacology
Received Date: 17 October 2017 Revised Date:
11 December 2017
Accepted Date: 13 December 2017
Please cite this article as: de Candia, P., Matarese, G., Leptin and ghrelin: Sewing metabolism onto neurodegeneration, Neuropharmacology (2018), doi: 10.1016/j.neuropharm.2017.12.025. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT 1
Leptin
and
ghrelin:
sewing
2
neurodegeneration.
3
Paola de Candia1* and Giuseppe Matarese2,3*
metabolism
onto
RI PT
4 1IRCCS
6
Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-
7
CNR), 80131 Naples, Italy; 3Dipartimento di Medicina Molecolare e Biotecnologie
8
Mediche, Università di Napoli “Federico II”, 80131 Naples, Italy.
9
* To whom correspondence should be addressed: Prof. Giuseppe Matarese,
10
Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli
11
“Federico
12
[email protected] or Dr. Paola de Candia, PhD, Via Fantoli 16/15, 20138
13
Milan, Italy, Phone: +39 02 55406577, E-mail:
[email protected]
17 18 19 20 21 22 23 24
M AN U
Italy,
Phone/Fax:
+39-0817464580,
TE D
16
Napoli,
EP
15
II”,
Keywords: neurodegeneration, neuroinflammation, metabolism, leptin, ghrelin
AC C
14
MultiMedica, 20138 Milan, Italy; 2Laboratorio di Immunologia, Istituto di
SC
5
E-mail:
ACCEPTED MANUSCRIPT Abstract:
26
Life expectancy has considerably increased over the last decades. The negative
27
consequence of this augmented longevity has been a dramatic increase of age-related
28
chronic neurodegenerative diseases, such as Alzheimer’s, Parkinson’s and multiple
29
sclerosis. Epidemiology is telling us there exists a strong correlation between the
30
neuronal loss characterizing these disorders and metabolic dysfunction. This review
31
aims at presenting the evidence supporting the existence of a molecular system linking
32
metabolism with neurodegeneration, with a specific focus on the role of two hormones
33
with a key role in the regulatory cross talk between metabolic imbalance and the
34
damage of nervous system: leptin and ghrelin.
M AN U
35 36
41 42
EP
40
AC C
39
TE D
37 38
SC
RI PT
25
43 44 45
2
ACCEPTED MANUSCRIPT
1. Introduction
47
The industrialized world population constantly ages with a progressive rise of cancer,
48
cardiovascular diseases, metabolic disorders and neurodegenerative conditions (Niccoli
49
and Partridge 2012). Modern medicine has succeeded to increase human life span, but
50
we now need to develop strategies to suppress/delay aging-dependent pathologies.
51
In this context, a critical clinical question is whether these pathological conditions are
52
independent or instead interconnected. Long known to be associated with diabetes and
53
cardiovascular disease, obesity has been more recently associated with decreased brain
54
size, lower density of the gray matter and cognitive function (Dixon 2010, Debette, Wolf
55
et al. 2014, Climie, Moran et al. 2015). Furthermore, higher weight represents a risk
56
factor for the development of Parkinson’s disease, Alzheimer and vascular dementia
57
(Abbott, Ross et al. 2002, Kivipelto, Ngandu et al. 2005, Xu, Atti et al. 2011). Importantly,
58
an observational study performed in rhesus macaques succeeded to show that caloric
59
restriction can not only lower the incidence of age-related pathologies such as diabetes,
60
cancer and cardiovascular diseases, but it is also able to preserve the gray matter
61
volume in several subcortical regions, the lateral temporal cortex and frontal lobe,
62
critical areas that exert motor and executive functions (Colman, Anderson et al. 2009).
63
Therefore, both human studies and animal models concur to suggest a close
64
relationship between metabolic imbalance (influenced by life style and diet) and the
65
pathological processes leading to neurodegeneration. In this context, we will here
66
examine the role played by leptin and ghrelin. These peptides are not mere regulators of
67
appetite/feeding but are directly involved in neuronal death induced by diversified
68
pathological conditions and play a role in neuroprotection and brain function.
AC C
EP
TE D
M AN U
SC
RI PT
46
69
3
ACCEPTED MANUSCRIPT
2. The vicious cycle of defective metabolism, dysregulated
71
inflammation and neurodegeneration.
72
Neurodegeneration causes the alteration of neuronal structure and function, and
73
consequent increased neuron death in the central nervous system (CNS). In this review,
74
we will mostly focus on the two most frequent neurodegenerative conditions:
75
Alzheimer's and Parkinson's diseases, and the most common cause of neurological
76
disability in young adults, multiple sclerosis.
SC
RI PT
70
77 2.1. Alzheimer’s disease.
79
Alzheimer's disease (AD) is a progressive illness with an estimated prevalence of 10–
80
30% in the population >65 years of age and decades-long preclinical and prodromal
81
phases (Masters, Bateman et al. 2015). In AD, neuronal death begins in the temporal
82
lobe and then in the parietal lobe causing progressive short and long-term memory loss,
83
cognitive impairment and psychiatric disorders (Murray, Kumar et al. 2014, Masters,
84
Bateman et al. 2015). In 1906, Alois Alzheimer identified neuritic plaques and
85
neurofibrillary tangles as prominent neuropathologic features in post-mortem brains of
86
certain patients. Much later, it was discovered that neuritic plaques are formed by the
87
abnormal deposition of amyloid β-peptide (Aβ) at the extracellular level, and
88
intracellular protein aggregates composed of hyper-phosphorylated tau protein (pTau)
89
represent the major components of neurofibrillary tangles (NFTs) (Jeong 2017). Aβ
90
plaques in concert with pTau are believed to be the major drivers of AD
91
neurodegeneration: Aβ deposition starts causing alternations in learning and memory
92
circuitry before clinical AD onset, while aging-associated damage of mitochondria
AC C
EP
TE D
M AN U
78
4
ACCEPTED MANUSCRIPT 93
collaborates with Aβ and pTau to produce synapse loss and cognitive impairment
94
(Jeong 2017).
95 2.2. Parkinson’s disease.
97
In 1817, James Parkinson described a disease that was subsequently named after him as
98
the “Shaking Palsy” and at the end of the 1800s it was proposed that the anatomical
99
substrate of Parkinson’s disease would be a lesion of the substantia nigra. Nowadays, it
100
is well established that the first neurons to degenerate are located the substantia nigra
101
pars compacta (SNc), which produces dopamine (DA), central to motor control
102
(Bernheimer, Birkmayer et al. 1973, Davie 2008).
103
As a consequence, patients suffer from typical motor symptoms such as resting tremor,
104
postural instability and rigidity, inability to move muscles voluntarily (akinesia) and
105
freezing of gait. They also tend to show non-motor symptoms, including sleep disorders,
106
cognitive impairment, apathy, depression and progressive dementia (Antonini, Barone
107
et al. 2012, Kim, So et al. 2014). The classical neuro-pathological feature is the
108
development of intracytoplasmic aggregates of α-synuclein, termed Lewy bodies.
109
Analyzing post-mortem human brain tissue from PD, it was observed that the
110
development of Lewy bodies is associated with microtubule regression, mitochondrial
111
loss and nuclear degradation in neurons. Lewy bodies are also believed to be directly
112
responsible of membrane damage, dysregulation of Ca2+ exchange, oxidative
113
impairment and consequent cell death (Spillantini, Schmidt et al. 1997).
AC C
EP
TE D
M AN U
SC
RI PT
96
114 115
2.3. Multiple Sclerosis.
116
Multiple sclerosis (MS) is a chronic inflammatory disease of the CNS that, differently
117
from AD and PD, mostly affects younger people (onset ranging from 20 to 50 years)
5
ACCEPTED MANUSCRIPT (Milo and Kahana 2010). The course of MS is highly varied and unpredictable. In the
119
majority of affected patients, the disease is initially characterized by reversible
120
neurological symptoms, which are often followed by progressive neurological
121
deterioration over time. The cause of MS is not fully elucidated, but it appears to involve
122
both genetic susceptibility and non-genetic triggers, such infections, life style and
123
environmental factors, that together result in a self-sustaining autoimmune disorder
124
characterized by recurrent immune attacks on the CNS. In particular, MS pathogenic
125
target is the myelin sheath that is progressively destroyed by the action of autoreactive
126
immune cells. Nonetheless, it is now well recognized that the neurological disability in
127
MS patients also directly depends on the progressive axonal loss (Dutta and Trapp
128
2007, Stadelmann, Albert et al. 2008, Trapp and Nave 2008).
M AN U
SC
RI PT
118
129
2.4. The connection between inflammation and brain degeneration.
131
A growing body of observational and experimental data shows that a pivotal factor
132
intervening in the onset and progression of neuronal degeneration is the cascade of
133
processes collectively termed neuroinflammation (Ransohoff 2016). Many studies
134
suggest that normal ageing per se increases inflammatory grade in the brain and
135
systemically and contributes to progressive neural deficits. Basal levels of many pro-
136
inflammatory cytokines increase in older brains, dramatically affecting the survival and
137
the function of neurons, glia, and progenitor cells (Murray and Lynch 1998, Bodles and
138
Barger 2004, Joseph, Shukitt-Hale et al. 2005, Nolan, Maher et al. 2005). In particular,
139
microglia cells, the brain counterpart of macrophages, are neuroprotective in the young
140
brain, but change morphologically and modify their inflammatory profile upon aging-
141
induced senescence. The increased level of neuroinflammatory cytokines and
142
chemokines released by aged over-activated microglia becomes neurotoxic and takes
AC C
EP
TE D
130
6
ACCEPTED MANUSCRIPT active part in promoting neurodegeneration. It has also been hypothesized that aging-
144
related activation of microglia suppresses neurogenesis, known to be profoundly
145
affected by the microenvironment, and thus impairs cognitive function (Luo, Ding et al.
146
2010).
147
In AD, Aβ and NFT accumulation further induces microglia activation, which augments
148
the level of neuroinflammatory mediators, which in turn worsen neurodegenerative
149
conditions (Tuppo and Arias 2005). Also in PD, there exists a vicious cycle in which
150
aggregates of misfolded α-synuclein directly activates glial and other inflammatory cells
151
releasing pro-inflammatory molecules which exacerbate cellular sufferance in the
152
substantia nigra (Rocha, de Miranda et al. 2015, Wang, Liu et al. 2015, Grimmig,
153
Morganti et al. 2016).
154
In conclusion, AD and PD are diseases in which a neurodegenerative-centric view has
155
given the way to including inflammation as a crucial pathogenic promoter. MS, on the
156
other hand, is a condition that has traditionally been considered an autoimmune disease
157
and it has then been re-evaluated in light of the neurodegerative component of the
158
pathogenic process. In other words, these three conditions are prototypical for the
159
intermingling of neuroinflammation and neurodegeneration.
SC
M AN U
TE D
EP
AC C
160
RI PT
143
161
2.5. The association between metabolic imbalance and neurodegeneration.
162
In recent time, it has become clearer and clearer that there exists a link between the
163
metabolic state and the neurological symptoms of neurodegenerative diseases. Several
164
observational studies introduced solid evidence suggesting a direct relation between
165
diet and changes in the brain structure and activity (Solfrizzi, Custodero et al. 2017).
166
Higher body mass index and midlife adiposity increase the risk of later dementia and PD
167
development (Abbott, Ross et al. 2002, Whitmer, Gunderson et al. 2005); subjects with
7
ACCEPTED MANUSCRIPT type 2 diabetes mellitus are more susceptible to develop AD than healthy individuals
169
(Ott, Stolk et al. 1999); and adolescence obesity increases the risk of developing MS
170
(Munger, Chitnis et al. 2009, Hedstrom, Olsson et al. 2012, Langer-Gould, Brara et al.
171
2013). A high-fat diet directly damages brain function via glucotoxicity, i.e. the toxic
172
effects of increased glucose or dysregulated insulin sensitivity (Cai, Cong et al. 2012).
173
Moreover, AD transgenic mice fed with a high fat diet on one side develop insulin
174
resistance, on the other show more severe Aβ accumulation and worse cognitive deficits
175
compared to the same mice fed normally (Ho, Qin et al. 2004). In AD specifically, the
176
connection between high levels of glucose and disease progression has also a direct
177
molecular basis in the role of glucose in the cleavage of tau (Kim, Backus et al. 2009).
178
Notably, T2D and AD diseased mice suffer from similar cognitive impairment, vascular
179
defects and augmented Aβ deposition in the brain (Carvalho, Cardoso et al. 2012,
180
Carvalho, Machado et al. 2013) and AD neurodegeneration is accompanied by a
181
dysfunctional utilization of glucose (Steen, Terry et al. 2005, Schrijvers, Witteman et al.
182
2010).
183
Other sets of studies performed in PD corroborate the hypothesis that drugs used to
184
treat T2D may produce significant neuroprotective effects (Aviles-Olmos, Dickson et al.
185
2013, Aviles-Olmos, Limousin et al. 2013). Similarly to AD, in brain areas affected by PD,
186
it has been observed an impaired expression of insulin receptors and insulin signaling,
187
with altered levels of glucose and glucose tolerance, directly contributing to the
188
formation of α-synuclein pathological aggregates and suggesting that a dysfunction of
189
the insulin/insulin receptor system may precede the decline of the dopaminergic
190
neurons (Sandyk 1993, Moroo, Yamada et al. 1994, Figlewicz, Evans et al. 2003).
191
Interestingly, low levels of insulin in T2D rats correlate with a decrease in messenger
192
RNA for DA transporter and tyrosine hydroxylase in substantia nigra, suggesting an
AC C
EP
TE D
M AN U
SC
RI PT
168
8
ACCEPTED MANUSCRIPT 193
impairment of DA signaling in these animals (Figlewicz, Brot et al. 1996). It has also
194
been described that mouse models of T2D treated with a neurotoxin used to mimic PD-
195
related
196
develop a higher grade of brain inflammation and demonstrate increased accumulation
197
of α-synuclein compared to the non-diabetic controls, suggesting that diabetes causes a
198
worse dopaminergic cell decline in PD conditions (Wang, Zhai et al. 2014). In addition,
199
the decrease of glucose uptake by brain cells can alter the intracellular ratio of ATP and
200
ADP, impacting the capacity of dopaminergic neurons to produce and release DA (Levin
201
2000, Santiago and Potashkin 2013).
202
One striking molecule that seems to be the cornerstone at the crossroad of metabolism,
203
inflammation and neurodegeneration is called leptin.
(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine,
MPTP)
M AN U
SC
RI PT
neurodegeneration
204
3. Leptin: the anorexigenic neuroprotective adipokine with a
206
twisting pro-inflammatory role.
207
3.1. The critical weight of the “thin” gene.
208
Leptin is a 167 amino acid long peptide transcribed by the human ob gene, located on
209
chromosome 7 (Zhang, Proenca et al. 1994). Even if leptin orthologs in non-mammalian
210
animals are significantly different in terms of primary amino-acidic sequence, its
211
function seems to be preserved through the conservation of secondary and tertiary
212
structures. Leptin crystal structure has revealed a four-helix bundle characteristic of the
213
long-chain helical cytokine family (Zhang, Basinski et al. 1997).
214
The crucial role of leptin as “satiety factor” or “fasting hormone” is demonstrated by the
215
phenotype of leptin-deficient mice (ob/ob) that eat excessively and become profoundly
216
obese (Pelleymounter, Cullen et al. 1995). Similarly, mice with a mutation in the leptin
AC C
EP
TE D
205
9
ACCEPTED MANUSCRIPT receptor that renders it unable to sense this factor (db/db) develop hyperphagic obesity
218
and represent a model of spontaneous type 2 diabetes (Chen, Charlat et al. 1996). Leptin
219
is mostly produced and released by the adipose tissue in quantity that is proportional to
220
fat mass, and its circulating concentrations decline rapidly in response to reduced body
221
fat and food intake, providing a dynamic measure of the size of fat storage and acute
222
changes in energy balance (Ahima, Prabakaran et al. 1996). Its primary metabolic
223
function is to inhibit food intake and stimulate energy expenditure, via receptor-
224
mediated actions in the hypothalamus (Jequier 2002). The leptin-leptin receptor axis
225
provides a means for the periphery to communicate the metabolic status of the
226
organism to the CNS. The crucial relevance of the leptin signaling in human energy
227
homeostasis is revealed by the phenotype of individuals who are homozygous for
228
inactivating mutations in leptin or leptin receptor: these people are clearly obese,
229
extremely hyperphagic and closely resemble the metabolic imbalance of the obese and
230
diabetic mice (Montague, Farooqi et al. 1997, Clement, Vaisse et al. 1998). Furthermore,
231
the generalized or partial loss of adipose tissue that characterizes individuals with
232
either genetic or acquired lipodystrophy syndromes results in leptin deficiency, that in
233
turn leads to hyperphagia and insulin resistance. Notably, leptin replacement has
234
become a relevant clinical strategy that results in decreased morbidity for these
235
individuals (Garg 2004, Diker-Cohen, Cochran et al. 2015).
236
Leptin receptors are found in other brain regions than hypothalamus, among which the
237
hippocampus, where leptin may play a role in controlling learning and memory. In
238
addition, leptin was shown to regulate bone metabolism, angiogenesis, hematopoiesis
239
and sexual reproduction (Bennett, Solar et al. 1996, Chehab, Lim et al. 1996, Sierra-
240
Honigmann, Nath et al. 1998, Ducy, Amling et al. 2000). Given its central role in body
AC C
EP
TE D
M AN U
SC
RI PT
217
10
ACCEPTED MANUSCRIPT weight regulation, leptin name comes from the Greek word “leptos” that means “thin”,
242
but this hormone has a much more pleiotropic influence than originally believed.
243
3.2. The neuroprotective effect of leptin
244
Leptin is an important neurotrophic factor, involved in the regulation of neuronal
245
development, function and survival. The actual participation of leptin in the
246
pathogenesis of neurodegeneration has been extensively studies in both ob/ob and
247
db/db mice: these animals show reduced brain volume and a plethora of relevant
248
proteins display an aberrant brain expression pattern such as syntaxin-1,
249
synaptosomal-associated protein-25, synaptobrevin, myelin basic protein and glial
250
fibrillary acidic protein. When the same mice are treated with leptin, the brain weight,
251
overall protein content and locomotor activity get normalized, demonstrating that
252
leptin indeed controls neuronal and glial maturation and myelin development (Ahima,
253
Bjorbaek et al. 1999, Schwartz and Baskin 2013).
254
In human, there exists an association between leptin levels in the circulation and
255
incidence of dementia, with leptin being a protective parameter (Lieb, Beiser et al. 2009,
256
Bigalke, Schreitmuller et al. 2011). Leptin signaling has been extensively analyzed in AD,
257
where it has been shown to diminish the level of tau phosphorylation in neuronal cells
258
and to inhibit Aβ accumulation via activating AMPK and downregulating the expression
259
of the γ-secretase components (Fewlass, Noboa et al. 2004, Greco, Sarkar et al. 2009,
260
Greco, Bryan et al. 2010, Niedowicz, Studzinski et al. 2013). While leptin levels were
261
found increased in both CSF and hippocampal tissue of AD patients, the level of leptin
262
receptor mRNA was instead observed to be decreased in AD brain, with the receptor
263
protein trapped in neurofibrillary tangles, suggesting a dramatic impairment of leptin-
264
dependent signaling (Bonda, Stone et al. 2014). These observations suggest that leptin
265
imbalance can accelerate cognitive decline and AD progression, while AD process, on its
AC C
EP
TE D
M AN U
SC
RI PT
241
11
ACCEPTED MANUSCRIPT part, can block leptin signaling, in a downward loop of progressive neurodegeneration
267
and worsening metabolic defects. Clinical trial data demonstrate that leptin
268
administration is safe for long term use in humans, that suggests a potential application
269
of leptin as a therapy for AD. In mice, chronic administration of leptin to AD-transgenic
270
animals also results in better cognitive performance and behavioral improvements, as
271
early as after 4 weeks of treatment (Greco, Bryan et al. 2010). Moreover, leptin
272
replacement in patients with congenital leptin deficiency leads to a rise in gray matter
273
volume (Matochik, London et al. 2005).
274
3.3 Leptin central role in enhancing the function of the immune system
275
Giving support to the knowledge that leptin exerts pleiotropic effects in periphery, it
276
has been observed that ob/ob and db/db mice are not only obese but also show
277
significant alterations of the immune system (Lord, Matarese et al. 1998, Lord, Matarese
278
et al. 2001). Indeed, several immune cell types are shown to express detectable level of
279
the leptin receptor (Procaccini, Jirillo et al. 2012) and leptin is extremely powerful in
280
regulating the activity of cells of the native and the adaptive immune system. It
281
stimulates neutrophils, regulates phagocytic function of macrophages and the
282
cytotoxicity of natural killer cells and activates dendritic cells (Gainsford, Willson et al.
283
1996, Loffreda, Yang et al. 1998, Caldefie-Chezet, Poulin et al. 2001, Tian, Sun et al.
284
2002, Caldefie-Chezet, Poulin et al. 2003, Macia, Delacre et al. 2006). Leptin also
285
modulates the proliferation of CD4+ T helper (Th) cells (Lord, Matarese et al. 1998). In
286
particular, it activates the proliferation and differentiation of CD4+ T into pro-
287
inflammatory Th-17 cells (Lord, Matarese et al. 1998). To address the cell-intrinsic role
288
of leptin signaling in T cells, the leptin receptor was conditionally targeted in these cells
289
specifically: the study succeed to show that LepR deficiency causes a selective defect in
290
the development of Th17 phenotype and both autoimmune and protective responses
AC C
EP
TE D
M AN U
SC
RI PT
266
12
ACCEPTED MANUSCRIPT (Reis, Lee et al. 2015). At the same time, leptin blocks the activity and growth of the
292
anti-inflammatory T regulatory (Treg) cell subset: it has been reported that level of
293
circulating leptin and frequency of Treg cells indeed inversely correlate (Wagner,
294
Brandhorst et al. 2013).
295
Experimental autoimmune encephalomyelitis (EAE) is the most commonly used
296
experimental model for the human MS (Constantinescu, Farooqi et al. 2011). When
297
ob/ob mice are induced to develop EAE by myelin-specific T cell stimulation, they resist
298
to disease induction and progression, showing low levels of IFN-γ and high levels of the
299
anti-inflammatory IL-4 (Matarese, Di Giacomo et al. 2001, Matarese, Sanna et al. 2001,
300
Sanna, Di Giacomo et al. 2003). Consistently, leptin neutralization in wild type EAE mice
301
is also able to delay disease progression, by blocking the action of auto-reactive
302
lymphocytes (De Rosa, Procaccini et al. 2006, Galgani, Procaccini et al. 2010). On the
303
other hand, leptin replacement in ob/ob mice is sufficient to render them susceptible to
304
disease with a strong shift towards a pro-inflammatory cytokine pattern (Matarese, Di
305
Giacomo et al. 2001, Matarese, Sanna et al. 2001, Sanna, Di Giacomo et al. 2003); and
306
leptin administration to wild type mice increases the severity of EAE (Sanna, Di
307
Giacomo et al. 2003). In addition to its pro-inflammatory effect on lymphocytes, leptin is
308
believed to potentially worsen blood–brain-barrier dysregulation, because it was
309
possible to reduce lymphocyte infiltration into spinal cord by specifically blocking leptin
310
signaling in the endothelium (Ouyang, Hsuchou et al. 2014). Notably, MS patients show
311
high level of leptin expression in active inflammatory lesions of the CNS and in the sera
312
of MS patients before relapses upon treatment with IFN-β (Lock, Hermans et al. 2002,
313
Batocchi, Rotondi et al. 2003, Matarese, Carrieri et al. 2005, Olama, Senna et al. 2012,
314
Emamgholipour, Eshaghi et al. 2013, Carbone, De Rosa et al. 2014). Specific gene
315
expression analysis of T helper lymphocytes from active MS lesions has confirmed the
AC C
EP
TE D
M AN U
SC
RI PT
291
13
ACCEPTED MANUSCRIPT presence of elevated transcriptional level of leptin (Lock, Hermans et al. 2002).
317
Environmental metabolic cues can modify the ratio between inflammation and immune
318
tolerance by skewing T-cell fate decisions toward either the Th1/Th17 pro-
319
inflammatory or Treg immune suppressive cell lineages, because these cell subsets have
320
a different response to leptin levels (MacIver, Michalek et al. 2013).
321
These studies strongly support the notion that leptin represents the crucial in vivo
322
molecular link between metabolic homeostasis and inflammatory dysregulation. Caloric
323
restriction has been described to significantly increase the survival and lifespan in EAE
324
mice, reducing inflammation, demyelination, and axon damage, to the extent that
325
dietary intervention is now considered a potentially efficacious new strategy for MS
326
treatment (Piccio, Stark et al. 2008). One of the beneficial effects of fasting may indeed
327
depend on reduced circulating leptin levels that in turn has a significant beneficial
328
repercussion on the inflammatory state (Liu, Yu et al. 2012). In addition, caloric
329
restriction leads to increased blood concentrations of anti-inflammatory corticosterone
330
and adiponectin (Piccio, Stark et al. 2008); it also augments the level of ghrelin, another
331
pivotal hormone regulating metabolism, but not only (Esquifino, Cano et al. 2004,
332
Esquifino, Cano et al. 2007).
SC
M AN U
TE D
EP
AC C
333
RI PT
316
334
4. Ghrelin: the orexigenic peptide with a consistent
335
neuroprotective and anti-inflammatory function.
336
4.1 The complex ghrelin system.
337
Ghrelin is predominantly produced by specialized endocrine cells of the stomach
338
(Kojima, Hosoda et al. 1999), and circulating ghrelin increases before meals to levels
339
that stimulate food intake and also increases following food deprivation and after
14
ACCEPTED MANUSCRIPT various forms of weight loss; in other words, its rise is expected in conditions of caloric
341
restriction (Cummings, Purnell et al. 2001, Nagaya, Uematsu et al. 2001, Otto, Cuntz et
342
al. 2001, Cummings 2006). Its metabolic effects are opposite to those of leptin, as it
343
stimulates food intake, decreases energy expenditure and regulates growth hormone
344
secretion, glucose-sensing and glucose homeostasis (Cummings and Foster 2003,
345
Nogueiras, Tschop et al. 2008). Ghrelin also influences body weight by promoting food-
346
reward behaviors and increasing the mRNA expression of fat storage–promoting
347
enzymes in the adipose tissue (Perello, Sakata et al. 2010). Diet-induced obesity was
348
shown to dramatically dysregulate ghrelin function in food intake and metabolic control
349
(Briggs, Enriori et al. 2010).
350
The mature form of ghrelin, a 28 amino acid long peptide, results from the proteolytic
351
processing of a precursor peptide named preproghrelin. In humans, the peptide is
352
encoded by a single-copy gene (GHRL) located on the short arm of chromosome 3, that
353
shows high regulatory complexity due to alternative sites for transcription initiation
354
and alternative splicing mRNA variants due to exon skipping or intron retention (Seim,
355
Herington et al. 2009, Sato, Nakamura et al. 2012). In particular, a variant that lacks the
356
coding region of exon 3 (Ex3-deleted ghrelin), and another variant generated by
357
retention of intron 1 (In1-ghrelin) are particularly interesting because their
358
conservation in other mammalian species and/or overexpression in cancer samples
359
suggest that these variants might exert important patho-physiological role, yet to be
360
identified (Yeh, Jeffery et al. 2005, Gahete, Cordoba-Chacon et al. 2011).
361
In addition, the final steps of preproghrelin proteolytic processing generate both the
362
mature native ghrelin peptide, and an additional C-terminal peptide named C-ghrelin,
363
whose further proteolytic process generates a functional peptide, obestatin,
364
hypothesized to be the antagonist hormone for ghrelin (Zhang, Ren et al. 2005, Sato,
AC C
EP
TE D
M AN U
SC
RI PT
340
15
ACCEPTED MANUSCRIPT Nakamura et al. 2012). To add further complexity to the ghrelin system, native ghrelin
366
peptide can be modified through acylation (addition of an octanoyl group) of the highly
367
conserved third serine residue: according to the acylation status, the peptide is named
368
unacylated ghrelin (UAG) or acylated-ghrelin (AG) (Kojima, Hosoda et al. 1999, Nishi,
369
Yoh et al. 2011). The acylation process is mediated by an enzyme known as ghrelin-O-
370
acyl-transferase or GOAT, believed to act before the proteolytic processing (Garg 2007).
371
Notably, while UAG represents the large majority of total blood ghrelin (Patterson,
372
Murphy et al. 2005), it is unable to bind the classic ghrelin receptor (GHSR1a), leaving
373
its potential biological role not yet elucidated. It is also unknown whether UAG is the
374
product of the incomplete acylation of the nascent ghrelin peptide, or is also the product
375
of mature AG de-acylation (Kojima, Hosoda et al. 1999, Nishi, Yoh et al. 2011).
376
4.2 Ghrelin: not just a question of hunger.
377
In addition to mediating opposite effects in the neuro-hypothalamic axis, ghrelin also
378
displays reverse action relative to leptin on immune activity in EAE.
379
administration has been shown to significantly associate with a reduction of pro-
380
inflammatory cytokine expression (TNF-alpha, IL-1β, and IL-6) and diminished cellular
381
infiltrates in the spinal cord (Theil, Miyake et al. 2009). The anti-inflammatory role of
382
ghrelin is striking to the point that it has projected this hormone in the arena as a
383
potential strategy for the management of MS.
384
Ghrelin receptors are expressed in a widespread manner in the CNS, suggesting that this
385
hormone is not only involved in metabolism and inflammation, but plays other essential
386
brain functions, such as learning and memory, anxiety control, reward, mood, and sleep
387
(Muller, Nogueiras et al. 2015). Several beneficial effects of ghrelin on neurons have
388
been described: it protects mitochondria, inhibiting reactive oxygen species formation
389
and cytochrome-c release; it regulates the level of intracellular calcium and nitric oxide,
Ghrelin
AC C
EP
TE D
M AN U
SC
RI PT
365
16
ACCEPTED MANUSCRIPT which has a direct effect on memory function; similarly to leptin, ghrelin also protects
391
neurons from the amyloid-associated toxicity (Chung, Kim et al. 2007, Moon, Choi et al.
392
2011, Gomes, Martins et al. 2014). Ghrelin levels change with ageing and a diminished
393
ghrelin signaling has been shown to precede impairment in cognitive testing and its
394
serum level was found to inversely associate with cognitive function in non-demented
395
elderly people (Rigamonti, Pincelli et al. 2002, Spitznagel, Benitez et al. 2010). A very
396
recent report shows that ghrelin is able to attenuate diabetic-related rat
397
encephalopathy, having a beneficial effect on both neuroinflammation and cognitive
398
impairment. In particular, ghrelin was found to ameliorate astrocytic activation, reduce
399
pro-inflammatory factors such as interleukin-6 and tumor necrosis factor-α and reverse
400
down-regulation of mature nerve growth factor (NGF) in experimental diabetic rats
401
(Zhao, Shen et al. 2017). Ghrelin was also shown to prevent neuronal degeneration and
402
improve motor coordination in a mouse traumatic brain injury model by blocking
403
apoptosis and blood-brain barrier dysfunction (Lopez, Lindsay et al. 2014).
404
Ghrelin dysregulation has been suggested to contribute to the severe cognitive deficit
405
observed in AD, since its expression was found strikingly reduced in one of the cortical
406
regions most affected in AD, the temporal gyrus, when comparing AD to healthy
407
subjects (Gahete, Rubio et al. 2010). Ghrelin level is also diminished in PD, with PD
408
patients showing lower plasma ghrelin concentrations compared to healthy individuals,
409
that demonstrates an impaired ghrelin production in PD conditions (Fiszer,
410
Michalowska et al. 2010). Furthermore, ghrelin production is reduced in pre-motor
411
stage of PD, suggesting a direct implication of this hormone in disease progression
412
(Andrews, Erion et al. 2009), while ghrelin administration is able to protect DA neurons
413
from MPTP-induced death, reduce microglial activation and the levels of TNF-α, IL-1β,
414
and NO (Moon, Kim et al. 2009).
AC C
EP
TE D
M AN U
SC
RI PT
390
17
ACCEPTED MANUSCRIPT Independently of the presence of the acyl group, ghrelin is able to protect primary
416
cultured rat cortical neurons from ischemic injury and glucose deprivation, by
417
increasing phosphorylation of extracellular signal-regulated kinase (ERK)1/2, Akt,
418
glycogen synthase kinase-3β (GSK-3β), Bcl-2/Bax ratio, and β-catenin nuclear
419
translocation and preventing cytochrome c release and caspase-3 activation (Chung,
420
Seo et al. 2008). These results suggest the involvement of a receptor that is distinct from
421
GHS-R1a. Intriguingly, UAG, but not AG, would exert this action by binding and blocking
422
the action of CD36, a scavenger receptor able to recruit and activate microglia and
423
secretion of inflammatory mediators and cause neuronal dysfunction and death
424
(Bulgarelli, Tamiazzo et al. 2009). On the other hand, another study identifies acylated
425
ghrelin as the form responsible for in vivo neuroprotection in a mouse model of PD:
426
while administration of acylated ghrelin in ghrelin KO mice can attenuate the MPTP-
427
induced neuronal loss and reduce glial activation in the substancia nigra, administration
428
of des-acylated ghrelin to the same mice fails to do the same, suggesting that
429
pharmacological approaches impacting the ratio between acyl ghrelin to des-acyl
430
ghrelin may have clinical efficacy to slow PD progression (Bayliss, Lemus et al. 2016).
431
Notably, the dopaminergic receptor D1R and the ghrelin receptor GHSR1a were
432
demonstrated to bind and form heterodimers in vitro and described to co-localize in
433
various areas of the brain (Jiang, Betancourt et al. 2006), and it was subsequently
434
reported that GHSR1a receptor can modify dopaminergic signaling by enhancing DA
435
release and thus reducing PD motor symptoms (Kern, Albarran-Zeckler et al. 2012).
436
It has been observed that the neuroprotective effect of caloric restriction in a mouse
437
experimental model of PD depends on the function of ghrelin, in particular on its
438
capacity to activate AMPK in dopamine neurons (Bayliss, Lemus et al. 2016). AMPK
439
senses cellular energy, increases adenosine triphosphate (ATP) production and
AC C
EP
TE D
M AN U
SC
RI PT
415
18
ACCEPTED MANUSCRIPT suppresses energy consumption in conditions of cell stress (Hardie, Ross et al. 2012).
441
Through AMPK, ghrelin is known to activate peroxisome proliferator-activated receptor
442
γ coactivator-1α (PGC-1α) (Canto and Auwerx 2009), a master regulator of
443
mitochondrial biogenesis, known to be dysfunctional during PD progression and
444
pinpointed to be significantly decreased in DA neurons from PD patients (Zheng, Liao et
445
al. 2010). The positive effect of ghrelin on PGC-1 determines enhanced mitochondrial
446
health and may reduce dopaminergic cell loss. The relevance of this ghrelin target in
447
preventing neurodegeneration is demonstrated by the phenotype of PGC-1α null mice
448
that result to be more susceptible to MPTP-induced PD (St-Pierre, Drori et al. 2006). On
449
the other hand, ghrelin target AMPK is central in the induction of autophagy, the
450
process
451
removed/destroyed/digested. When the energy balance is low, AMPK activation leads
452
to mTOR inhibition and subsequent autophagy induction.
453
4.2 mTOR and autophagy
454
The mechanistic target of rapamycin (mTOR) protein is a highly conserved serine–
455
threonine kinase that senses the energy status and regulates anabolism, cell-cycle
456
progression, growth and autophagy. Two different complexes of mTOR (mTORC1 and
457
mTORC2) show different sensitivity to rapamycin and associate with different protein
458
regulatory partners. The nutrient-sensitive complex mTORC1 is central for the
459
induction of protein synthesis through phosphorylation of ribosomal S6 kinase (S6K).
460
In recent times, the crucial role of mTORC1 as signaling target of both leptin and ghrelin
461
is being unveiled. In rodent hypothalamus, leptin has been shown to increase mTORC1
462
activity, and mTOR signaling to mediate the effect of leptin in food intake regulation.
463
Either mTORC1 inhibition by rapamycin, or the deletion of mTOR target S6K is able to
damaged
or
unnecessary
cellular
components
are
AC C
EP
TE D
whereby
M AN U
SC
RI PT
440
19
ACCEPTED MANUSCRIPT disrupt leptin's anorectic effect, demonstrating that this intracellular signaling pathway
465
is required for the appetite-suppressing effects of this hormone (Cota, Proulx et al.
466
2006, Cota, Matter et al. 2008). Ghrelin has also been shown to promote
467
phosphorylation of S6K1 in the rat hypothalamus, an effect that is suppressed by
468
rapamycin, suggesting that ghrelin role in food intake, adiposity and insulin secretion is
469
actually controlled by the mTORC1/S6K1 pathway in the central nervous system
470
(Stevanovic, Trajkovic et al. 2013). Since ghrelin and leptin play opposite roles in food
471
intake, it has been hypothesized that mTORC1 is a switching balance that mediates the
472
distinct effects of these two peptides in metabolic regulation, depending on nutritional
473
status and neuronal activity (Haissaguerre, Saucisse et al. 2014).
474
Interestingly, a systemic dysregulation of the mTOR signaling cascade is present in the
475
brain but also in peripheral lymphocytes of Alzheimer's disease patients before the
476
onset of the disease, suggesting this specific pathway may represent a risk factor for
477
Alzheimer (Yates, Zafar et al. 2013). Indeed, mTOR signaling pathway has been shown
478
to be involved in the balance of phosphorylated/non-phosphorylated tau, and its
479
localization and aggregation, suggesting an mTOR direct effect in tau deposition (Kang
480
and Cho 2015, Tang, Ioja et al. 2015). In neuronal cell lines and primary neurons, PD
481
inducers such as 6-hydroxydopamine or N-methyl-4-phenylpyridine reduce cell
482
viability in parallel with a suppression of mTOR function and activation of caspase-3
483
and poly-ADP ribose polymerase (PARP) cleavage. Furthermore, overexpression of
484
mTOR reduced cell death in response to the PD toxins. It was also found that the
485
suppression of Akt and the activation of AMPK were important contributors of neuronal
486
cell death (Xu, Liu et al. 2014). Another report has shown that mTOR activation can
487
prevent oxidative stress-mediated cell death, while treatment with rapamycin
488
potentiated this type of cell death in dopamine neurons. Importantly, a crucial
AC C
EP
TE D
M AN U
SC
RI PT
464
20
ACCEPTED MANUSCRIPT mechanism involved in the capacity of mTOR to protect the neurons was found to be
490
autophagy, mTOR being the key regulator of the autophagic process in response to
491
growth factors and cell stress (Saxton and Sabatini 2017). Recent work suggests that
492
dysregulated autophagy is implicated in different neurodegenerative diseases with the
493
common denominator of cellular toxicity associated with misfolded protein
494
accumulation. The formation of α-synuclein aggregates in PD is inhibited by the
495
induction of autophagy, leading to the hypothesis that the correction of autophagic
496
process may rescue from disease progression (Mishra, ur Rasheed et al. 2015).
497
Consistently, treatment with an autophagy inhibitor is able to protect the dopamine
498
cells (Choi, Kim et al. 2010). Other studies, though, tell a different story. Rapamycin has
499
been shown to protect against neuron death in in vivo models of PD and the
500
downregulation of mTOR signaling pathway and the consequent recovery of
501
macroautophagy secondary to curcumin treatment has a protective effect against the
502
neurodegenerative pathology in a missense mutation of α-synuclein model of
503
hereditary PD (Malagelada, Jin et al. 2010, Jiang, Zhang et al. 2013).
SC
M AN U
TE D
EP
504
RI PT
489
5. Conclusion and Perspectives
506
Animal models and human studies strongly suggest that there is a close interconnection
507
between metabolism, inflammation and neurodegeneration. Several observations
508
indicate that therapies targeting systemic inflammation may provide benefit for those
509
afflicted by neurodegenerative disorders; and that the two conditions of
510
neurodegenerative disorders and metabolic dysregulation share specific molecular
511
mechanisms to the point that agents with proven efficacy in one may be useful against
512
the other. In other words, therapies aimed at re-establishing metabolic homeostasis
AC C
505
21
ACCEPTED MANUSCRIPT may have also an efficacy in contrasting cognitive decline. In this context, hormones like
514
leptin or ghrelin, originally believed to be primarily regulators of feeding and appetite,
515
have been later demonstrated to play important roles in brain function,
516
neuroprotection and neuroinflammation, linking metabolic regulation with brain health
517
and disease (Figure 1).
518
It is important to point out that leptin and ghrelin have been discovered to play a role in
519
CNS pathologies other than neurodegeneration. By regulating the hypothalamic-
520
pituitary-adrenal axis, ghrelin affects anxiety and mood disorders, depression, fear and
521
alcohol dependence (Leggio 2010, Spencer, Emmerzaal et al. 2015, Zarouna, Wozniak et
522
al. 2015). At the same time, many human studies have associated leptin levels with
523
depression and anxiety, sleep disturbances, behavioral disorders, social isolation and
524
even suicide attempts (Westling, Ahren et al. 2004, Atmaca, Tezcan et al. 2005, Lu, Kim
525
et al. 2006, Lawson, Miller et al. 2012, Zarouna, Wozniak et al. 2015). Still, the role of
526
ghrelin and leptin in mood regulation has not been yet clearly dissected, and further
527
studies are needed to reveal potential common pattern of emotional changes and
528
level/biological impact of these two hormones. All these complex and pleiotropic effects
529
of leptin and ghrelin on CNS and the periphery must be taken into account in future
530
clinical trials evaluating the beneficial and adverse consequences of their
531
administration in humans with the final goal of identifying novel therapeutic
532
approaches that, combined with lifestyle modifications, can target neuroinflammation
533
and synergistically combat neurodegeneration.
534
Moreover, we cannot ignore that, at the systemic level, leptin and ghrelin act in concert
535
with a plethora of other signals. Beside leptin, many different molecules differently
536
impact metabolism and immunological functions, such as adipose-derived adipokines
537
resistin, angiopoietin-like protein 2, adiponectin, visfatin and adipsin (Bluher and
AC C
EP
TE D
M AN U
SC
RI PT
513
22
ACCEPTED MANUSCRIPT Mantzoros 2015). In particular, the concomitant influence of leptin on eating and
539
energy expenditure is believed to work in concert with the short-term effects induced
540
by amylin, a pancreatic β-cell hormone co-released with insulin in response to food
541
intake. Importantly amylin precursor is also expressed by hypothalamic neurons and
542
positively regulated by leptin; amylin blockade decreases the effects of leptin on
543
neurons in vivo and in vitro, suggesting hypothalamic amylin acts in concert with leptin
544
(Li, Kelly et al. 2015). Regarding the biological role of ghrelin, we need to acquire more
545
knowledge on the overlapping or distinct effect of the acylated, un-acylated and de-
546
acylated forms of ghrelin, but also on the contribution of the multiple different mRNA
547
variants. Beside the above-mentioned Ex3-deleted ghrelin and In1-ghrelin, some of the
548
splicing variants generate peptides with small changes in their sequences compared
549
with native ghrelin, such as des-Gln14-ghrelin, which is identical to native ghrelin
550
except for the deletion of one glutamine residue (Gln14), whose difference in function is
551
still totally obscure (Sato, Nakamura et al. 2012).
552
Finally, a fuller understanding of the grid composed by the myriad of these molecules
553
and their reciprocal interaction, and a better knowledge of the effect of leptin and
554
ghrelin on intracellular central regulatory hubs, such as mTOR, may provide not only
555
new approaches to improve these two hormones signaling and sensitivity, but also a
556
more in-depth comprehension of the strings that link metabolism and brain health.
SC
M AN U
TE D
EP
AC C
557
RI PT
538
558 559 560
23
ACCEPTED MANUSCRIPT
Acknowledgements
562
This work was supported by funds from the European Foundation for the Study of
563
Diabetes/Juvenile Diabetes Research Foundation/Lilly program 2015. PdC is also
564
supported by the National Multiple Sclerosis Society NMSS (PP-1606-24687) and
565
Fondazione Italiana Sclerosi Multipla FISM (2016/R/10). GM is supported by the Italian
566
Space Agency (ASI, 2014-033-R.O) and Fondazione Italiana Sclerosi Multipla FISM
567
(2016/R/18).
568
The authors declare that they have no conflicts of interest with the contents of this
569
article.
M AN U
SC
RI PT
561
570 571 572
576 577 578 579 580 581
EP
575
AC C
574
TE D
573
582 583 584
24
ACCEPTED MANUSCRIPT 585 586 587 588
590
RI PT
589 References
591
EP
TE D
M AN U
SC
Abbott, R. D., G. W. Ross, L. R. White, J. S. Nelson, K. H. Masaki, C. M. Tanner, J. D. Curb, P. L. Blanchette, J. S. Popper and H. Petrovitch (2002). "Midlife adiposity and the future risk of Parkinson's disease." Neurology 59(7): 1051-1057. Ahima, R. S., C. Bjorbaek, S. Osei and J. S. Flier (1999). "Regulation of neuronal and glial proteins by leptin: implications for brain development." Endocrinology 140(6): 27552762. Ahima, R. S., D. Prabakaran, C. Mantzoros, D. Qu, B. Lowell, E. Maratos-Flier and J. S. Flier (1996). "Role of leptin in the neuroendocrine response to fasting." Nature 382(6588): 250-252. Andrews, Z. B., D. Erion, R. Beiler, Z. W. Liu, A. Abizaid, J. Zigman, J. D. Elsworth, J. M. Savitt, R. DiMarchi, M. Tschoep, R. H. Roth, X. B. Gao and T. L. Horvath (2009). "Ghrelin promotes and protects nigrostriatal dopamine function via a UCP2-dependent mitochondrial mechanism." J Neurosci 29(45): 14057-14065. Antonini, A., P. Barone, R. Marconi, L. Morgante, S. Zappulla, F. E. Pontieri, S. Ramat, M. G. Ceravolo, G. Meco, G. Cicarelli, M. Pederzoli, M. Manfredi, R. Ceravolo, M. Mucchiut, G. Volpe, G. Abbruzzese, E. Bottacchi, L. Bartolomei, G. Ciacci, A. Cannas, M. G. Randisi, A. Petrone, M. Baratti, V. Toni, G. Cossu, P. Del Dotto, A. R. Bentivoglio, M. Abrignani, R. Scala, F. Pennisi, R. Quatrale, R. M. Gaglio, A. Nicoletti, M. Perini, T. Avarello, A. Pisani, A. Scaglioni, P. E. Martinelli, F. Iemolo, L. Ferigo, P. Simone, P. Soliveri, B. Troianiello, D. Consoli, A. Mauro, L. Lopiano, G. Nastasi and C. Colosimo (2012). "The progression of non-motor symptoms in Parkinson's disease and their contribution to motor disability and quality of life." J Neurol 259(12): 2621-2631. Atmaca, M., E. Tezcan, M. Kuloglu and B. Ustundag (2005). "Serum leptin levels in obsessive-compulsive disorder." Psychiatry Clin Neurosci 59(2): 189-193. Aviles-Olmos, I., J. Dickson, Z. Kefalopoulou, A. Djamshidian, P. Ell, T. Soderlund, P. Whitton, R. Wyse, T. Isaacs, A. Lees, P. Limousin and T. Foltynie (2013). "Exenatide and the treatment of patients with Parkinson's disease." J Clin Invest 123(6): 2730-2736. Aviles-Olmos, I., P. Limousin, A. Lees and T. Foltynie (2013). "Parkinson's disease, insulin resistance and novel agents of neuroprotection." Brain 136(Pt 2): 374-384. Batocchi, A. P., M. Rotondi, M. Caggiula, G. Frisullo, F. Odoardi, V. Nociti, C. Carella, P. A. Tonali and M. Mirabella (2003). "Leptin as a marker of multiple sclerosis activity in patients treated with interferon-beta." J Neuroimmunol 139(1-2): 150-154. Bayliss, J. A., M. Lemus, V. V. Santos, M. Deo, J. D. Elsworth and Z. B. Andrews (2016). "Acylated but not des-acyl ghrelin is neuroprotective in an MPTP mouse model of Parkinson's disease." J Neurochem 137(3): 460-471.
AC C
592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626
25
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Bayliss, J. A., M. B. Lemus, R. Stark, V. V. Santos, A. Thompson, D. J. Rees, S. Galic, J. D. Elsworth, B. E. Kemp, J. S. Davies and Z. B. Andrews (2016). "Ghrelin-AMPK Signaling Mediates the Neuroprotective Effects of Calorie Restriction in Parkinson's Disease." J Neurosci 36(10): 3049-3063. Bennett, B. D., G. P. Solar, J. Q. Yuan, J. Mathias, G. R. Thomas and W. Matthews (1996). "A role for leptin and its cognate receptor in hematopoiesis." Curr Biol 6(9): 1170-1180. Bernheimer, H., W. Birkmayer, O. Hornykiewicz, K. Jellinger and F. Seitelberger (1973). "Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations." J Neurol Sci 20(4): 415-455. Bigalke, B., B. Schreitmuller, K. Sopova, A. Paul, E. Stransky, M. Gawaz, K. Stellos and C. Laske (2011). "Adipocytokines and CD34 progenitor cells in Alzheimer's disease." PLoS One 6(5): e20286. Bluher, M. and C. S. Mantzoros (2015). "From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century." Metabolism 64(1): 131-145. Bodles, A. M. and S. W. Barger (2004). "Cytokines and the aging brain - what we don't know might help us." Trends Neurosci 27(10): 621-626. Bonda, D. J., J. G. Stone, S. L. Torres, S. L. Siedlak, G. Perry, R. Kryscio, G. Jicha, G. Casadesus, M. A. Smith, X. Zhu and H. G. Lee (2014). "Dysregulation of leptin signaling in Alzheimer disease: evidence for neuronal leptin resistance." J Neurochem 128(1): 162172. Briggs, D. I., P. J. Enriori, M. B. Lemus, M. A. Cowley and Z. B. Andrews (2010). "Dietinduced obesity causes ghrelin resistance in arcuate NPY/AgRP neurons." Endocrinology 151(10): 4745-4755. Bulgarelli, I., L. Tamiazzo, E. Bresciani, D. Rapetti, S. Caporali, D. Lattuada, V. Locatelli and A. Torsello (2009). "Desacyl-ghrelin and synthetic GH-secretagogues modulate the production of inflammatory cytokines in mouse microglia cells stimulated by betaamyloid fibrils." J Neurosci Res 87(12): 2718-2727. Cai, H., W. N. Cong, S. Ji, S. Rothman, S. Maudsley and B. Martin (2012). "Metabolic dysfunction in Alzheimer's disease and related neurodegenerative disorders." Curr Alzheimer Res 9(1): 5-17. Caldefie-Chezet, F., A. Poulin, A. Tridon, B. Sion and M. P. Vasson (2001). "Leptin: a potential regulator of polymorphonuclear neutrophil bactericidal action?" J Leukoc Biol 69(3): 414-418. Caldefie-Chezet, F., A. Poulin and M. P. Vasson (2003). "Leptin regulates functional capacities of polymorphonuclear neutrophils." Free Radic Res 37(8): 809-814. Canto, C. and J. Auwerx (2009). "PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure." Curr Opin Lipidol 20(2): 98-105. Carbone, F., V. De Rosa, P. B. Carrieri, S. Montella, D. Bruzzese, A. Porcellini, C. Procaccini, A. La Cava and G. Matarese (2014). "Regulatory T cell proliferative potential is impaired in human autoimmune disease." Nat Med 20(1): 69-74. Carvalho, C., S. Cardoso, S. C. Correia, R. X. Santos, M. S. Santos, I. Baldeiras, C. R. Oliveira and P. I. Moreira (2012). "Metabolic alterations induced by sucrose intake and Alzheimer's disease promote similar brain mitochondrial abnormalities." Diabetes 61(5): 1234-1242. Carvalho, C., N. Machado, P. C. Mota, S. C. Correia, S. Cardoso, R. X. Santos, M. S. Santos, C. R. Oliveira and P. I. Moreira (2013). "Type 2 diabetic and Alzheimer's disease mice present similar behavioral, cognitive, and vascular anomalies." J Alzheimers Dis 35(3): 623-635.
AC C
627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675
26
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Chehab, F. F., M. E. Lim and R. Lu (1996). "Correction of the sterility defect in homozygous obese female mice by treatment with the human recombinant leptin." Nat Genet 12(3): 318-320. Chen, H., O. Charlat, L. A. Tartaglia, E. A. Woolf, X. Weng, S. J. Ellis, N. D. Lakey, J. Culpepper, K. J. Moore, R. E. Breitbart, G. M. Duyk, R. I. Tepper and J. P. Morgenstern (1996). "Evidence that the diabetes gene encodes the leptin receptor: identification of a mutation in the leptin receptor gene in db/db mice." Cell 84(3): 491-495. Choi, K. C., S. H. Kim, J. Y. Ha, S. T. Kim and J. H. Son (2010). "A novel mTOR activating protein protects dopamine neurons against oxidative stress by repressing autophagy related cell death." J Neurochem 112(2): 366-376. Chung, H., E. Kim, D. H. Lee, S. Seo, S. Ju, D. Lee, H. Kim and S. Park (2007). "Ghrelin inhibits apoptosis in hypothalamic neuronal cells during oxygen-glucose deprivation." Endocrinology 148(1): 148-159. Chung, H., S. Seo, M. Moon and S. Park (2008). "Phosphatidylinositol-3kinase/Akt/glycogen synthase kinase-3 beta and ERK1/2 pathways mediate protective effects of acylated and unacylated ghrelin against oxygen-glucose deprivation-induced apoptosis in primary rat cortical neuronal cells." J Endocrinol 198(3): 511-521. Clement, K., C. Vaisse, N. Lahlou, S. Cabrol, V. Pelloux, D. Cassuto, M. Gourmelen, C. Dina, J. Chambaz, J. M. Lacorte, A. Basdevant, P. Bougneres, Y. Lebouc, P. Froguel and B. GuyGrand (1998). "A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction." Nature 392(6674): 398-401. Climie, R. E., C. Moran, M. Callisaya, L. Blizzard, J. E. Sharman, A. Venn, T. G. Phan, R. Beare, J. Forbes, N. B. Blackburn and V. Srikanth (2015). "Abdominal Obesity and Brain Atrophy in Type 2 Diabetes Mellitus." PLoS One 10(11): e0142589. Colman, R. J., R. M. Anderson, S. C. Johnson, E. K. Kastman, K. J. Kosmatka, T. M. Beasley, D. B. Allison, C. Cruzen, H. A. Simmons, J. W. Kemnitz and R. Weindruch (2009). "Caloric restriction delays disease onset and mortality in rhesus monkeys." Science 325(5937): 201-204. Constantinescu, C. S., N. Farooqi, K. O'Brien and B. Gran (2011). "Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS)." Br J Pharmacol 164(4): 1079-1106. Cota, D., E. K. Matter, S. C. Woods and R. J. Seeley (2008). "The role of hypothalamic mammalian target of rapamycin complex 1 signaling in diet-induced obesity." J Neurosci 28(28): 7202-7208. Cota, D., K. Proulx, K. A. Smith, S. C. Kozma, G. Thomas, S. C. Woods and R. J. Seeley (2006). "Hypothalamic mTOR signaling regulates food intake." Science 312(5775): 927930. Cummings, D. E. (2006). "Ghrelin and the short- and long-term regulation of appetite and body weight." Physiol Behav 89(1): 71-84. Cummings, D. E. and K. E. Foster (2003). "Ghrelin-leptin tango in body-weight regulation." Gastroenterology 124(5): 1532-1535. Cummings, D. E., J. Q. Purnell, R. S. Frayo, K. Schmidova, B. E. Wisse and D. S. Weigle (2001). "A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans." Diabetes 50(8): 1714-1719. Davie, C. A. (2008). "A review of Parkinson's disease." Br Med Bull 86: 109-127. De Rosa, V., C. Procaccini, A. La Cava, P. Chieffi, G. F. Nicoletti, S. Fontana, S. Zappacosta and G. Matarese (2006). "Leptin neutralization interferes with pathogenic T cell autoreactivity in autoimmune encephalomyelitis." J Clin Invest 116(2): 447-455.
AC C
676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723
27
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Debette, S., C. Wolf, J. C. Lambert, F. Crivello, A. Soumare, Y. C. Zhu, S. Schilling, C. Dufouil, B. Mazoyer, P. Amouyel, C. Tzourio and A. Elbaz (2014). "Abdominal obesity and lower gray matter volume: a Mendelian randomization study." Neurobiol Aging 35(2): 378386. Diker-Cohen, T., E. Cochran, P. Gorden and R. J. Brown (2015). "Partial and generalized lipodystrophy: comparison of baseline characteristics and response to metreleptin." J Clin Endocrinol Metab 100(5): 1802-1810. Dixon, J. B. (2010). "The effect of obesity on health outcomes." Mol Cell Endocrinol 316(2): 104-108. Ducy, P., M. Amling, S. Takeda, M. Priemel, A. F. Schilling, F. T. Beil, J. Shen, C. Vinson, J. M. Rueger and G. Karsenty (2000). "Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass." Cell 100(2): 197-207. Dutta, R. and B. D. Trapp (2007). "Pathogenesis of axonal and neuronal damage in multiple sclerosis." Neurology 68(22 Suppl 3): S22-31; discussion S43-54. Emamgholipour, S., S. M. Eshaghi, A. Hossein-nezhad, K. Mirzaei, Z. Maghbooli and M. A. Sahraian (2013). "Adipocytokine profile, cytokine levels and foxp3 expression in multiple sclerosis: a possible link to susceptibility and clinical course of disease." PLoS One 8(10): e76555. Esquifino, A. I., P. Cano, V. Jimenez, R. A. Cutrera and D. P. Cardinali (2004). "Experimental allergic encephalomyelitis in male Lewis rats subjected to calorie restriction." J Physiol Biochem 60(4): 245-252. Esquifino, A. I., P. Cano, V. Jimenez-Ortega, M. P. Fernandez-Mateos and D. P. Cardinali (2007). "Immune response after experimental allergic encephalomyelitis in rats subjected to calorie restriction." J Neuroinflammation 4: 6. Fewlass, D. C., K. Noboa, F. X. Pi-Sunyer, J. M. Johnston, S. D. Yan and N. Tezapsidis (2004). "Obesity-related leptin regulates Alzheimer's Abeta." FASEB J 18(15): 18701878. Figlewicz, D. P., M. D. Brot, A. L. McCall and P. Szot (1996). "Diabetes causes differential changes in CNS noradrenergic and dopaminergic neurons in the rat: a molecular study." Brain Res 736(1-2): 54-60. Figlewicz, D. P., S. B. Evans, J. Murphy, M. Hoen and D. G. Baskin (2003). "Expression of receptors for insulin and leptin in the ventral tegmental area/substantia nigra (VTA/SN) of the rat." Brain Res 964(1): 107-115. Fiszer, U., M. Michalowska, B. Baranowska, E. Wolinska-Witort, W. Jeske, M. Jethon, M. Piascik-Gromada and E. Marcinowska-Suchowierska (2010). "Leptin and ghrelin concentrations and weight loss in Parkinson's disease." Acta Neurol Scand 121(4): 230236. Gahete, M. D., J. Cordoba-Chacon, M. Hergueta-Redondo, A. J. Martinez-Fuentes, R. D. Kineman, G. Moreno-Bueno, R. M. Luque and J. P. Castano (2011). "A novel human ghrelin variant (In1-ghrelin) and ghrelin-O-acyltransferase are overexpressed in breast cancer: potential pathophysiological relevance." PLoS One 6(8): e23302. Gahete, M. D., A. Rubio, J. Cordoba-Chacon, F. Gracia-Navarro, R. D. Kineman, J. Avila, R. M. Luque and J. P. Castano (2010). "Expression of the ghrelin and neurotensin systems is altered in the temporal lobe of Alzheimer's disease patients." J Alzheimers Dis 22(3): 819-828. Gainsford, T., T. A. Willson, D. Metcalf, E. Handman, C. McFarlane, A. Ng, N. A. Nicola, W. S. Alexander and D. J. Hilton (1996). "Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells." Proc Natl Acad Sci U S A 93(25): 1456414568.
AC C
724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772
28
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Galgani, M., C. Procaccini, V. De Rosa, F. Carbone, P. Chieffi, A. La Cava and G. Matarese (2010). "Leptin modulates the survival of autoreactive CD4+ T cells through the nutrient/energy-sensing mammalian target of rapamycin signaling pathway." J Immunol 185(12): 7474-7479. Garg, A. (2004). "Acquired and inherited lipodystrophies." N Engl J Med 350(12): 12201234. Garg, A. (2007). "The ongoing saga of obestatin: is it a hormone?" J Clin Endocrinol Metab 92(9): 3396-3398. Gomes, S., I. Martins, A. C. Fonseca, C. R. Oliveira, R. Resende and C. M. Pereira (2014). "Protective effect of leptin and ghrelin against toxicity induced by amyloid-beta oligomers in a hypothalamic cell line." J Neuroendocrinol 26(3): 176-185. Greco, S. J., K. J. Bryan, S. Sarkar, X. Zhu, M. A. Smith, J. W. Ashford, J. M. Johnston, N. Tezapsidis and G. Casadesus (2010). "Leptin reduces pathology and improves memory in a transgenic mouse model of Alzheimer's disease." J Alzheimers Dis 19(4): 11551167. Greco, S. J., S. Sarkar, J. M. Johnston and N. Tezapsidis (2009). "Leptin regulates tau phosphorylation and amyloid through AMPK in neuronal cells." Biochem Biophys Res Commun 380(1): 98-104. Grimmig, B., J. Morganti, K. Nash and P. C. Bickford (2016). "Immunomodulators as Therapeutic Agents in Mitigating the Progression of Parkinson's Disease." Brain Sci 6(4). Haissaguerre, M., N. Saucisse and D. Cota (2014). "Influence of mTOR in energy and metabolic homeostasis." Mol Cell Endocrinol 397(1-2): 67-77. Hardie, D. G., F. A. Ross and S. A. Hawley (2012). "AMPK: a nutrient and energy sensor that maintains energy homeostasis." Nat Rev Mol Cell Biol 13(4): 251-262. Hedstrom, A. K., T. Olsson and L. Alfredsson (2012). "High body mass index before age 20 is associated with increased risk for multiple sclerosis in both men and women." Mult Scler 18(9): 1334-1336. Ho, L., W. Qin, P. N. Pompl, Z. Xiang, J. Wang, Z. Zhao, Y. Peng, G. Cambareri, A. Rocher, C. V. Mobbs, P. R. Hof and G. M. Pasinetti (2004). "Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer's disease." FASEB J 18(7): 902904. Jeong, S. (2017). "Molecular and Cellular Basis of Neurodegeneration in Alzheimer's Disease." Mol Cells. Jequier, E. (2002). "Leptin signaling, adiposity, and energy balance." Ann N Y Acad Sci 967: 379-388. Jiang, H., L. Betancourt and R. G. Smith (2006). "Ghrelin amplifies dopamine signaling by cross talk involving formation of growth hormone secretagogue receptor/dopamine receptor subtype 1 heterodimers." Mol Endocrinol 20(8): 1772-1785. Jiang, T. F., Y. J. Zhang, H. Y. Zhou, H. M. Wang, L. P. Tian, J. Liu, J. Q. Ding and S. D. Chen (2013). "Curcumin ameliorates the neurodegenerative pathology in A53T alphasynuclein cell model of Parkinson's disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy." J Neuroimmune Pharmacol 8(1): 356-369. Joseph, J. A., B. Shukitt-Hale, G. Casadesus and D. Fisher (2005). "Oxidative stress and inflammation in brain aging: nutritional considerations." Neurochem Res 30(6-7): 927935.
AC C
773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819
29
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Kang, E. B. and J. Y. Cho (2015). "Effect of treadmill exercise on PI3K/AKT/mTOR, autophagy, and Tau hyperphosphorylation in the cerebral cortex of NSE/htau23 transgenic mice." J Exerc Nutrition Biochem 19(3): 199-209. Kern, A., R. Albarran-Zeckler, H. E. Walsh and R. G. Smith (2012). "Apo-ghrelin receptor forms heteromers with DRD2 in hypothalamic neurons and is essential for anorexigenic effects of DRD2 agonism." Neuron 73(2): 317-332. Kim, B., C. Backus, S. Oh, J. M. Hayes and E. L. Feldman (2009). "Increased tau phosphorylation and cleavage in mouse models of type 1 and type 2 diabetes." Endocrinology 150(12): 5294-5301. Kim, S. R., H. Y. So, E. Choi, J. H. Kang, H. Y. Kim and S. J. Chung (2014). "Influencing effect of non-motor symptom clusters on quality of life in Parkinson's disease." J Neurol Sci 347(1-2): 310-315. Kivipelto, M., T. Ngandu, L. Fratiglioni, M. Viitanen, I. Kareholt, B. Winblad, E. L. Helkala, J. Tuomilehto, H. Soininen and A. Nissinen (2005). "Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease." Arch Neurol 62(10): 15561560. Kojima, M., H. Hosoda, Y. Date, M. Nakazato, H. Matsuo and K. Kangawa (1999). "Ghrelin is a growth-hormone-releasing acylated peptide from stomach." Nature 402(6762): 656-660. Langer-Gould, A., S. M. Brara, B. E. Beaber and C. Koebnick (2013). "Childhood obesity and risk of pediatric multiple sclerosis and clinically isolated syndrome." Neurology 80(6): 548-552. Lawson, E. A., K. K. Miller, J. I. Blum, E. Meenaghan, M. Misra, K. T. Eddy, D. B. Herzog and A. Klibanski (2012). "Leptin levels are associated with decreased depressive symptoms in women across the weight spectrum, independent of body fat." Clin Endocrinol (Oxf) 76(4): 520-525. Leggio, L. (2010). "Role of the ghrelin system in alcoholism: Acting on the growth hormone secretagogue receptor to treat alcohol-related diseases." Drug News Perspect 23(3): 157-166. Levin, B. E. (2000). "Glucose-regulated dopamine release from substantia nigra neurons." Brain Res 874(2): 158-164. Li, Z., L. Kelly, M. Heiman, P. Greengard and J. M. Friedman (2015). "Hypothalamic Amylin Acts in Concert with Leptin to Regulate Food Intake." Cell Metab 22(6): 10591067. Lieb, W., A. S. Beiser, R. S. Vasan, Z. S. Tan, R. Au, T. B. Harris, R. Roubenoff, S. Auerbach, C. DeCarli, P. A. Wolf and S. Seshadri (2009). "Association of plasma leptin levels with incident Alzheimer disease and MRI measures of brain aging." JAMA 302(23): 25652572. Liu, Y., Y. Yu, G. Matarese and A. La Cava (2012). "Cutting edge: fasting-induced hypoleptinemia expands functional regulatory T cells in systemic lupus erythematosus." J Immunol 188(5): 2070-2073. Lock, C., G. Hermans, R. Pedotti, A. Brendolan, E. Schadt, H. Garren, A. Langer-Gould, S. Strober, B. Cannella, J. Allard, P. Klonowski, A. Austin, N. Lad, N. Kaminski, S. J. Galli, J. R. Oksenberg, C. S. Raine, R. Heller and L. Steinman (2002). "Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis." Nat Med 8(5): 500-508. Loffreda, S., S. Q. Yang, H. Z. Lin, C. L. Karp, M. L. Brengman, D. J. Wang, A. S. Klein, G. B. Bulkley, C. Bao, P. W. Noble, M. D. Lane and A. M. Diehl (1998). "Leptin regulates proinflammatory immune responses." FASEB J 12(1): 57-65.
AC C
820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868
30
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Lopez, N. E., G. Lindsay, L. R. Karina, H. A. Mary, J. Putnam, B. Eliceiri, R. Coimbra and V. Bansal (2014). "Ghrelin decreases motor deficits after traumatic brain injury." J Surg Res 187(1): 230-236. Lord, G. M., G. Matarese, J. K. Howard, R. J. Baker, S. R. Bloom and R. I. Lechler (1998). "Leptin modulates the T-cell immune response and reverses starvation-induced immunosuppression." Nature 394(6696): 897-901. Lord, G. M., G. Matarese, J. K. Howard and R. I. Lechler (2001). "The bioenergetics of the immune system." Science 292(5518): 855-856. Lu, X. Y., C. S. Kim, A. Frazer and W. Zhang (2006). "Leptin: a potential novel antidepressant." Proc Natl Acad Sci U S A 103(5): 1593-1598. Luo, X. G., J. Q. Ding and S. D. Chen (2010). "Microglia in the aging brain: relevance to neurodegeneration." Mol Neurodegener 5: 12. Macia, L., M. Delacre, G. Abboud, T. S. Ouk, A. Delanoye, C. Verwaerde, P. Saule and I. Wolowczuk (2006). "Impairment of dendritic cell functionality and steady-state number in obese mice." J Immunol 177(9): 5997-6006. MacIver, N. J., R. D. Michalek and J. C. Rathmell (2013). "Metabolic regulation of T lymphocytes." Annu Rev Immunol 31: 259-283. Malagelada, C., Z. H. Jin, V. Jackson-Lewis, S. Przedborski and L. A. Greene (2010). "Rapamycin protects against neuron death in in vitro and in vivo models of Parkinson's disease." J Neurosci 30(3): 1166-1175. Masters, C. L., R. Bateman, K. Blennow, C. C. Rowe, R. A. Sperling and J. L. Cummings (2015). "Alzheimer's disease." Nat Rev Dis Primers 1: 15056. Matarese, G., P. B. Carrieri, A. La Cava, F. Perna, V. Sanna, V. De Rosa, D. Aufiero, S. Fontana and S. Zappacosta (2005). "Leptin increase in multiple sclerosis associates with reduced number of CD4(+)CD25+ regulatory T cells." Proc Natl Acad Sci U S A 102(14): 5150-5155. Matarese, G., A. Di Giacomo, V. Sanna, G. M. Lord, J. K. Howard, A. Di Tuoro, S. R. Bloom, R. I. Lechler, S. Zappacosta and S. Fontana (2001). "Requirement for leptin in the induction and progression of autoimmune encephalomyelitis." J Immunol 166(10): 5909-5916. Matarese, G., V. Sanna, A. Di Giacomo, G. M. Lord, J. K. Howard, S. R. Bloom, R. I. Lechler, S. Fontana and S. Zappacosta (2001). "Leptin potentiates experimental autoimmune encephalomyelitis in SJL female mice and confers susceptibility to males." Eur J Immunol 31(5): 1324-1332. Matochik, J. A., E. D. London, B. O. Yildiz, M. Ozata, S. Caglayan, A. M. DePaoli, M. L. Wong and J. Licinio (2005). "Effect of leptin replacement on brain structure in genetically leptin-deficient adults." J Clin Endocrinol Metab 90(5): 2851-2854. Milo, R. and E. Kahana (2010). "Multiple sclerosis: geoepidemiology, genetics and the environment." Autoimmun Rev 9(5): A387-394. Mishra, A. K., M. S. ur Rasheed, S. Shukla, M. K. Tripathi, A. Dixit and M. P. Singh (2015). "Aberrant autophagy and parkinsonism: does correction rescue from disease progression?" Mol Neurobiol 51(3): 893-908. Montague, C. T., I. S. Farooqi, J. P. Whitehead, M. A. Soos, H. Rau, N. J. Wareham, C. P. Sewter, J. E. Digby, S. N. Mohammed, J. A. Hurst, C. H. Cheetham, A. R. Earley, A. H. Barnett, J. B. Prins and S. O'Rahilly (1997). "Congenital leptin deficiency is associated with severe early-onset obesity in humans." Nature 387(6636): 903-908. Moon, M., J. G. Choi, D. W. Nam, H. S. Hong, Y. J. Choi, M. S. Oh and I. Mook-Jung (2011). "Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-beta1-42 oligomer-injected mice." J Alzheimers Dis 23(1): 147-159.
AC C
869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917
31
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Moon, M., H. G. Kim, L. Hwang, J. H. Seo, S. Kim, S. Hwang, S. Kim, D. Lee, H. Chung, M. S. Oh, K. T. Lee and S. Park (2009). "Neuroprotective effect of ghrelin in the 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease by blocking microglial activation." Neurotox Res 15(4): 332-347. Moroo, I., T. Yamada, H. Makino, I. Tooyama, P. L. McGeer, E. G. McGeer and K. Hirayama (1994). "Loss of insulin receptor immunoreactivity from the substantia nigra pars compacta neurons in Parkinson's disease." Acta Neuropathol 87(4): 343-348. Muller, T. D., R. Nogueiras, M. L. Andermann, Z. B. Andrews, S. D. Anker, J. Argente, R. L. Batterham, S. C. Benoit, C. Y. Bowers, F. Broglio, F. F. Casanueva, D. D'Alessio, I. Depoortere, A. Geliebter, E. Ghigo, P. A. Cole, M. Cowley, D. E. Cummings, A. Dagher, S. Diano, S. L. Dickson, C. Dieguez, R. Granata, H. J. Grill, K. Grove, K. M. Habegger, K. Heppner, M. L. Heiman, L. Holsen, B. Holst, A. Inui, J. O. Jansson, H. Kirchner, M. Korbonits, B. Laferrere, C. W. LeRoux, M. Lopez, S. Morin, M. Nakazato, R. Nass, D. PerezTilve, P. T. Pfluger, T. W. Schwartz, R. J. Seeley, M. Sleeman, Y. Sun, L. Sussel, J. Tong, M. O. Thorner, A. J. van der Lely, L. H. van der Ploeg, J. M. Zigman, M. Kojima, K. Kangawa, R. G. Smith, T. Horvath and M. H. Tschop (2015). "Ghrelin." Mol Metab 4(6): 437-460. Munger, K. L., T. Chitnis and A. Ascherio (2009). "Body size and risk of MS in two cohorts of US women." Neurology 73(19): 1543-1550. Murray, C. A. and M. A. Lynch (1998). "Evidence that increased hippocampal expression of the cytokine interleukin-1 beta is a common trigger for age- and stress-induced impairments in long-term potentiation." J Neurosci 18(8): 2974-2981. Murray, P. S., S. Kumar, M. A. Demichele-Sweet and R. A. Sweet (2014). "Psychosis in Alzheimer's disease." Biol Psychiatry 75(7): 542-552. Nagaya, N., M. Uematsu, M. Kojima, Y. Date, M. Nakazato, H. Okumura, H. Hosoda, W. Shimizu, M. Yamagishi, H. Oya, H. Koh, C. Yutani and K. Kangawa (2001). "Elevated circulating level of ghrelin in cachexia associated with chronic heart failure: relationships between ghrelin and anabolic/catabolic factors." Circulation 104(17): 2034-2038. Niccoli, T. and L. Partridge (2012). "Ageing as a risk factor for disease." Curr Biol 22(17): R741-752. Niedowicz, D. M., C. M. Studzinski, A. M. Weidner, T. L. Platt, K. N. Kingry, T. L. Beckett, A. J. Bruce-Keller, J. N. Keller and M. P. Murphy (2013). "Leptin regulates amyloid beta production via the gamma-secretase complex." Biochim Biophys Acta 1832(3): 439444. Nishi, Y., J. Yoh, H. Hiejima and M. Kojima (2011). "Structures and molecular forms of the ghrelin-family peptides." Peptides 32(11): 2175-2182. Nogueiras, R., M. H. Tschop and J. M. Zigman (2008). "Central nervous system regulation of energy metabolism: ghrelin versus leptin." Ann N Y Acad Sci 1126: 14-19. Nolan, Y., F. O. Maher, D. S. Martin, R. M. Clarke, M. T. Brady, A. E. Bolton, K. H. Mills and M. A. Lynch (2005). "Role of interleukin-4 in regulation of age-related inflammatory changes in the hippocampus." J Biol Chem 280(10): 9354-9362. Olama, S. M., M. K. Senna and M. Elarman (2012). "Synovial/serum leptin ratio in rheumatoid arthritis: the association with activity and erosion." Rheumatol Int 32(3): 683-690. Ott, A., R. P. Stolk, F. van Harskamp, H. A. Pols, A. Hofman and M. M. Breteler (1999). "Diabetes mellitus and the risk of dementia: The Rotterdam Study." Neurology 53(9): 1937-1942. Otto, B., U. Cuntz, E. Fruehauf, R. Wawarta, C. Folwaczny, R. L. Riepl, M. L. Heiman, P. Lehnert, M. Fichter and M. Tschop (2001). "Weight gain decreases elevated plasma
AC C
918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966
32
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
ghrelin concentrations of patients with anorexia nervosa." Eur J Endocrinol 145(5): 669-673. Ouyang, S., H. Hsuchou, A. J. Kastin, P. K. Mishra, Y. Wang and W. Pan (2014). "Leukocyte infiltration into spinal cord of EAE mice is attenuated by removal of endothelial leptin signaling." Brain Behav Immun 40: 61-73. Patterson, M., K. G. Murphy, C. W. le Roux, M. A. Ghatei and S. R. Bloom (2005). "Characterization of ghrelin-like immunoreactivity in human plasma." J Clin Endocrinol Metab 90(4): 2205-2211. Pelleymounter, M. A., M. J. Cullen, M. B. Baker, R. Hecht, D. Winters, T. Boone and F. Collins (1995). "Effects of the obese gene product on body weight regulation in ob/ob mice." Science 269(5223): 540-543. Perello, M., I. Sakata, S. Birnbaum, J. C. Chuang, S. Osborne-Lawrence, S. A. Rovinsky, J. Woloszyn, M. Yanagisawa, M. Lutter and J. M. Zigman (2010). "Ghrelin increases the rewarding value of high-fat diet in an orexin-dependent manner." Biol Psychiatry 67(9): 880-886. Piccio, L., J. L. Stark and A. H. Cross (2008). "Chronic calorie restriction attenuates experimental autoimmune encephalomyelitis." J Leukoc Biol 84(4): 940-948. Procaccini, C., E. Jirillo and G. Matarese (2012). "Leptin as an immunomodulator." Mol Aspects Med 33(1): 35-45. Ransohoff, R. M. (2016). "How neuroinflammation contributes to neurodegeneration." Science 353(6301): 777-783. Reis, B. S., K. Lee, M. H. Fanok, C. Mascaraque, M. Amoury, L. B. Cohn, A. Rogoz, O. S. Dallner, P. M. Moraes-Vieira, A. I. Domingos and D. Mucida (2015). "Leptin receptor signaling in T cells is required for Th17 differentiation." J Immunol 194(11): 52535260. Rigamonti, A. E., A. I. Pincelli, B. Corra, R. Viarengo, S. M. Bonomo, D. Galimberti, M. Scacchi, E. Scarpini, F. Cavagnini and E. E. Muller (2002). "Plasma ghrelin concentrations in elderly subjects: comparison with anorexic and obese patients." J Endocrinol 175(1): R1-5. Rocha, N. P., A. S. de Miranda and A. L. Teixeira (2015). "Insights into Neuroinflammation in Parkinson's Disease: From Biomarkers to Anti-Inflammatory Based Therapies." Biomed Res Int 2015: 628192. Sandyk, R. (1993). "The relationship between diabetes mellitus and Parkinson's disease." Int J Neurosci 69(1-4): 125-130. Sanna, V., A. Di Giacomo, A. La Cava, R. I. Lechler, S. Fontana, S. Zappacosta and G. Matarese (2003). "Leptin surge precedes onset of autoimmune encephalomyelitis and correlates with development of pathogenic T cell responses." J Clin Invest 111(2): 241250. Santiago, J. A. and J. A. Potashkin (2013). "Shared dysregulated pathways lead to Parkinson's disease and diabetes." Trends Mol Med 19(3): 176-186. Sato, T., Y. Nakamura, Y. Shiimura, H. Ohgusu, K. Kangawa and M. Kojima (2012). "Structure, regulation and function of ghrelin." J Biochem 151(2): 119-128. Saxton, R. A. and D. M. Sabatini (2017). "mTOR Signaling in Growth, Metabolism, and Disease." Cell 169(2): 361-371. Schrijvers, E. M., J. C. Witteman, E. J. Sijbrands, A. Hofman, P. J. Koudstaal and M. M. Breteler (2010). "Insulin metabolism and the risk of Alzheimer disease: the Rotterdam Study." Neurology 75(22): 1982-1987. Schwartz, M. W. and D. G. Baskin (2013). "Leptin and the brain: then and now." J Clin Invest 123(6): 2344-2345.
AC C
967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015
33
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Seim, I., A. C. Herington and L. K. Chopin (2009). "New insights into the molecular complexity of the ghrelin gene locus." Cytokine Growth Factor Rev 20(4): 297-304. Sierra-Honigmann, M. R., A. K. Nath, C. Murakami, G. Garcia-Cardena, A. Papapetropoulos, W. C. Sessa, L. A. Madge, J. S. Schechner, M. B. Schwabb, P. J. Polverini and J. R. Flores-Riveros (1998). "Biological action of leptin as an angiogenic factor." Science 281(5383): 1683-1686. Solfrizzi, V., C. Custodero, M. Lozupone, B. P. Imbimbo, V. Valiani, P. Agosti, A. Schilardi, A. D'Introno, M. La Montagna, M. Calvani, V. Guerra, R. Sardone, D. I. Abbrescia, A. Bellomo, A. Greco, A. Daniele, D. Seripa, G. Logroscino, C. Sabba and F. Panza (2017). "Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer's Disease and Late-Life Cognitive Disorders: A Systematic Review." J Alzheimers Dis 59(3): 815-849. Spencer, S. J., T. L. Emmerzaal, T. Kozicz and Z. B. Andrews (2015). "Ghrelin's Role in the Hypothalamic-Pituitary-Adrenal Axis Stress Response: Implications for Mood Disorders." Biol Psychiatry 78(1): 19-27. Spillantini, M. G., M. L. Schmidt, V. M. Lee, J. Q. Trojanowski, R. Jakes and M. Goedert (1997). "Alpha-synuclein in Lewy bodies." Nature 388(6645): 839-840. Spitznagel, M. B., A. Benitez, J. Updegraff, V. Potter, T. Alexander, E. Glickman and J. Gunstad (2010). "Serum ghrelin is inversely associated with cognitive function in a sample of non-demented elderly." Psychiatry Clin Neurosci 64(6): 608-611. St-Pierre, J., S. Drori, M. Uldry, J. M. Silvaggi, J. Rhee, S. Jager, C. Handschin, K. Zheng, J. Lin, W. Yang, D. K. Simon, R. Bachoo and B. M. Spiegelman (2006). "Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators." Cell 127(2): 397-408. Stadelmann, C., M. Albert, C. Wegner and W. Bruck (2008). "Cortical pathology in multiple sclerosis." Curr Opin Neurol 21(3): 229-234. Steen, E., B. M. Terry, E. J. Rivera, J. L. Cannon, T. R. Neely, R. Tavares, X. J. Xu, J. R. Wands and S. M. de la Monte (2005). "Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes?" J Alzheimers Dis 7(1): 63-80. Stevanovic, D., V. Trajkovic, S. Muller-Luhlhoff, E. Brandt, W. Abplanalp, C. Bumke-Vogt, B. Liehl, P. Wiedmer, K. Janjetovic, V. Starcevic, A. F. Pfeiffer, H. Al-Hasani, M. H. Tschop and T. R. Castaneda (2013). "Ghrelin-induced food intake and adiposity depend on central mTORC1/S6K1 signaling." Mol Cell Endocrinol 381(1-2): 280-290. Tang, Z., E. Ioja, E. Bereczki, K. Hultenby, C. Li, Z. Guan, B. Winblad and J. J. Pei (2015). "mTor mediates tau localization and secretion: Implication for Alzheimer's disease." Biochim Biophys Acta 1853(7): 1646-1657. Theil, M. M., S. Miyake, M. Mizuno, C. Tomi, J. L. Croxford, H. Hosoda, J. Theil, S. von Horsten, H. Yokote, A. Chiba, Y. Lin, S. Oki, T. Akamizu, K. Kangawa and T. Yamamura (2009). "Suppression of experimental autoimmune encephalomyelitis by ghrelin." J Immunol 183(4): 2859-2866. Tian, Z., R. Sun, H. Wei and B. Gao (2002). "Impaired natural killer (NK) cell activity in leptin receptor deficient mice: leptin as a critical regulator in NK cell development and activation." Biochem Biophys Res Commun 298(3): 297-302. Trapp, B. D. and K. A. Nave (2008). "Multiple sclerosis: an immune or neurodegenerative disorder?" Annu Rev Neurosci 31: 247-269. Tuppo, E. E. and H. R. Arias (2005). "The role of inflammation in Alzheimer's disease." Int J Biochem Cell Biol 37(2): 289-305.
AC C
1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063
34
ACCEPTED MANUSCRIPT
EP
TE D
M AN U
SC
RI PT
Wagner, N. M., G. Brandhorst, F. Czepluch, M. Lankeit, C. Eberle, S. Herzberg, V. Faustin, J. Riggert, M. Oellerich, G. Hasenfuss, S. Konstantinides and K. Schafer (2013). "Circulating regulatory T cells are reduced in obesity and may identify subjects at increased metabolic and cardiovascular risk." Obesity (Silver Spring) 21(3): 461-468. Wang, L., Y. Q. Zhai, L. L. Xu, C. Qiao, X. L. Sun, J. H. Ding, M. Lu and G. Hu (2014). "Metabolic inflammation exacerbates dopaminergic neuronal degeneration in response to acute MPTP challenge in type 2 diabetes mice." Exp Neurol 251: 22-29. Wang, Q., Y. Liu and J. Zhou (2015). "Neuroinflammation in Parkinson's disease and its potential as therapeutic target." Transl Neurodegener 4: 19. Westling, S., B. Ahren, L. Traskman-Bendz and A. Westrin (2004). "Low CSF leptin in female suicide attempters with major depression." J Affect Disord 81(1): 41-48. Whitmer, R. A., E. P. Gunderson, E. Barrett-Connor, C. P. Quesenberry, Jr. and K. Yaffe (2005). "Obesity in middle age and future risk of dementia: a 27 year longitudinal population based study." BMJ 330(7504): 1360. Xu, W. L., A. R. Atti, M. Gatz, N. L. Pedersen, B. Johansson and L. Fratiglioni (2011). "Midlife overweight and obesity increase late-life dementia risk: a population-based twin study." Neurology 76(18): 1568-1574. Xu, Y., C. Liu, S. Chen, Y. Ye, M. Guo, Q. Ren, L. Liu, H. Zhang, C. Xu, Q. Zhou, S. Huang and L. Chen (2014). "Activation of AMPK and inactivation of Akt result in suppression of mTOR-mediated S6K1 and 4E-BP1 pathways leading to neuronal cell death in in vitro models of Parkinson's disease." Cell Signal 26(8): 1680-1689. Yates, S. C., A. Zafar, P. Hubbard, S. Nagy, S. Durant, R. Bicknell, G. Wilcock, S. Christie, M. M. Esiri, A. D. Smith and Z. Nagy (2013). "Dysfunction of the mTOR pathway is a risk factor for Alzheimer's disease." Acta Neuropathol Commun 1: 3. Yeh, A. H., P. L. Jeffery, R. P. Duncan, A. C. Herington and L. K. Chopin (2005). "Ghrelin and a novel preproghrelin isoform are highly expressed in prostate cancer and ghrelin activates mitogen-activated protein kinase in prostate cancer." Clin Cancer Res 11(23): 8295-8303. Zarouna, S., G. Wozniak and A. I. Papachristou (2015). "Mood disorders: A potential link between ghrelin and leptin on human body?" World J Exp Med 5(2): 103-109. Zhang, F., M. B. Basinski, J. M. Beals, S. L. Briggs, L. M. Churgay, D. K. Clawson, R. D. DiMarchi, T. C. Furman, J. E. Hale, H. M. Hsiung, B. E. Schoner, D. P. Smith, X. Y. Zhang, J. P. Wery and R. W. Schevitz (1997). "Crystal structure of the obese protein leptin-E100." Nature 387(6629): 206-209. Zhang, J. V., P. G. Ren, O. Avsian-Kretchmer, C. W. Luo, R. Rauch, C. Klein and A. J. Hsueh (2005). "Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin's effects on food intake." Science 310(5750): 996-999. Zhang, Y., R. Proenca, M. Maffei, M. Barone, L. Leopold and J. M. Friedman (1994). "Positional cloning of the mouse obese gene and its human homologue." Nature 372(6505): 425-432. Zhao, Y., Z. Shen, D. Zhang, H. Luo, J. Chen, Y. Sun and Q. Xiao (2017). "Ghrelin ameliorates nerve growth factor Dysmetabolism and inflammation in STZ-induced diabetic rats." Metab Brain Dis 32(3): 903-912. Zheng, B., Z. Liao, J. J. Locascio, K. A. Lesniak, S. S. Roderick, M. L. Watt, A. C. Eklund, Y. Zhang-James, P. D. Kim, M. A. Hauser, E. Grunblatt, L. B. Moran, S. A. Mandel, P. Riederer, R. M. Miller, H. J. Federoff, U. Wullner, S. Papapetropoulos, M. B. Youdim, I. CantutiCastelvetri, A. B. Young, J. M. Vance, R. L. Davis, J. C. Hedreen, C. H. Adler, T. G. Beach, M. B. Graeber, F. A. Middleton, J. C. Rochet, C. R. Scherzer and P. D. G. E. C. Global (2010).
AC C
1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111
35
ACCEPTED MANUSCRIPT 1112 1113 1114
"PGC-1alpha, a potential therapeutic target for early intervention in Parkinson's disease." Sci Transl Med 2(52): 52ra73.
1115 1116
RI PT
1117 1118 1119
SC
1120 1121
M AN U
1122 1123 1124 1125
1129
EP
1128
AC C
1127
TE D
1126
36
ACCEPTED MANUSCRIPT
Ghrelin
Lep n
Health
Pro-inflammatory: - Ac vates pro-inflammatory T cells - Reduces Treg func on - Worsens blood-brain barrier dysregula on
Balanced immune/metabolic homeostasis allows: - The neuroprotec ve role of lep n and ghrelin; - The proper control of mTOR pathway and autophagy.
1130 1131
SC
Disease
Altered control of metabolic func ons and chronic inflamma on: - Impairs the neuroprotec ve role of lep n and ghrelin; - Dysregulates mTOR signaling and leads to defec ve autophagy.
M AN U
An -inflammatory: - Reduces microglial ac va on and the pro-inflammatory cytokines Diminishes spinal cord cell infiltrates and auto-immune severity in EAE
Neuroprotec ve: - Controls brain size and a correct protein expression pa ern - Reduces the forma on of Aβ and NFT - Promotes survival of dopaminergic neurons
RI PT
Neuroprotec ve: - Protects dopaminergic neurons - Reduces ROS produc on and cytochrome c release - Ameliorates blood-brain barrier dysfunc on - Improves motor coordina on
Figure 1. The multiple roles played by ghrelin and leptin in the crosstalk between metabolic
1133
imbalance, neuroinflammation and neurodegeneration. In healthy conditions, regulated levels of
1134
leptin and ghrelin exert a beneficial effect (green) on the central nervous system. When metabolic balance
1135
is disrupted, their neuroprotective role is lost and high leptin levels aggravate neuroinflammation and
1136
consequent neurodegeneration (red).
AC C
EP
TE D
1132
37
ACCEPTED MANUSCRIPT
HIGHLIGHTS 1. Leptin exerts a neuroprotective role by blocking the formation of Aß and NFT and promoting survival of dopaminergic neurons.
RI PT
2. Leptin, on the other hand, also plays a pro-inflammatory role by activating T cells and worsening blood-brain barrier dysregulation.
SC
3. Ghrelin exerts a beneficial role by protecting dopaminergic neurons, improving motor coordination and ameliorating blood-brain barrier dysfunction.
M AN U
4. Ghrelin also plays an anti-inflammatory role by reducing microglial activation and pro-inflammatory cytokines.
AC C
EP
TE D
5. The altered control of metabolic functions impairs the neuroprotective role of leptin and ghrelin; and worsens the pro-inflammatory role of leptin.