ArticlePDF Available

A 2-year Double-Blind RCT Follow-up Study with Fermented Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors

Authors:
  • ReGenera R&D International for Aging Intervention, Milano

Abstract and Figures

In recent years a number of studies have reported the significant relationship between metabolic syndrome and neurodegenerative disease. There is accumulating evidence that the interplay of combined genetic and environmental risk factors (from diet to life style to pollutants) to intrinsic age-related oxi-inflammatory changes may be advocated for to explain the pandemic of neurodegenerative diseases. In recent years a specific Fermented Papaya Preparation (FPP) has been shown to significantly affect a number of redox signalling abnormalities in a variety of chronic diseases and as well in aging mechanisms either on experimental and on clinical ground. The aim of the present study was to evaluate FPP use in impending metabolic disease patients with potentially neurodegenerative disease clustered risk factors. The study population consisted of 90 patients aged 45-65 years old, with impending metabolic syndrome and previously selected as to be ApoE4 genotype negative. By applying a RCT, double-blind method, one group received FPP 4.5 g twice a day (the most common dosage utilized in prior clinical studies) while the other received an oral antioxidant cocktail (trans-resveratrol, selenium, vitamin E, vitamin C). Then, after 21 month treatment period, a selected heavy metal chelator was added at the dosage of 3 g/nocte for the final 3 months study treatment. The parameters tested were: routine tests oxidized LDL-cholesterol, anti-oxidised LDL, Cyclophilin-A (CyPA), plasminogen activator inhibitor-1 and CyPA gene expression. From this study it would appear that FPP, unlike the control antioxidant, significantly decreased oxidized-LDL and near normalizing the anti- Ox-LDL/Ox-LDL ratio (p<0.001) although unaffecting the lipid profile per sè. Moreover, only FPP decreased cyclophilin-A plasma level and plasminogen activator-inhibitor (p<0.01) together with downregulating cyclophilin-A gene expression (p<0.01). Insulin resistance was only mildly improved. Heavy metals gut clearance proved to be effectively enhanced by the chelator (p<0.01) and this was not affected by any of the nutraceuticals, nor it added any further benefit to the biological action of FPP.
Content may be subject to copyright.
A 2-year Double-Blind RCT Follow-up Study with Fermented Papaya
Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with
Clustered Neurodegenerative Disease-Risk Factors
Francesco Marotta1*, Massimiliano Marcellino1, Umberto Solimene2, Biagio Cuffari3, Hariom Yadav4, Alexander N Khokhlov5, Aldo Lorenzetti1, Amelie
Mantello6, Joseph Cervi1 and Roberto Catanzaro3
1ReGenera Research Group for Aging Intervention, San Babila Clinic, Milano, Italy
2WHO-Center for Traditional Medicine and Biotechnology, University of Milano, Italy
3Department of Internal Medicine, University of Catania, Catania, Italy
4Center for Diabetes, Obesity and Metabolism, Wake Forest Medical Center, Biotech Place, Winston-Salem, USA
5Evolutionary Cytogerontology Sector, School of Biology, Moscow State University, Moscow, Russia
6Osato Research Institute and Labs, Gifu, Japan
*Corresponding author: Prof. Francesco Marotta, ReGenera Research Group for Aging Intervention, San Babila Clinic, Corso Matteotti 1/A, 20121 Milano, Italy, Tel:
+39-024077243; E-mail: fmarchimede@libero.it
Received date: Apr 28, 2017; Accepted date: May 16, 2017; Published date: May 18, 2017
Copyright: © 2017 Marotta F, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and source are credited.
Abstract
In recent years a number of studies have reported the significant relationship between metabolic syndrome and
neurodegenerative disease. There is accumulating evidence that the interplay of combined genetic and
environmental risk factors (from diet to life style to pollutants) to intrinsic age-related oxi-inflammatory changes may
be advocated for to explain the pandemic of neurodegenerative diseases. In recent years a specific Fermented
Papaya Preparation (FPP) has been shown to significantly affect a number of redox signalling abnormalities in a
variety of chronic diseases and as well in aging mechanisms either on experimental and on clinical ground. The aim
of the present study was to evaluate FPP use in impending metabolic disease patients with potentially
neurodegenerative disease clustered risk factors. The study population consisted of 90 patients aged 45-65 years
old, with impending metabolic syndrome and previously selected as to be ApoE4 genotype negative. By applying a
RCT, double-blind method, one group received FPP 4.5 g twice a day (the most common dosage utilized in prior
clinical studies) while the other received an oral antioxidant cocktail (trans-resveratrol, selenium, vitamin E, vitamin
C). Then, after 21 month treatment period, a selected heavy metal chelator was added at the dosage of 3 g/nocte for
the final 3 months study treatment. The parameters tested were: routine tests oxidized LDL-cholesterol, anti-oxidised
LDL, Cyclophilin-A (CyPA), plasminogen activator inhibitor-1 and CyPA gene expression. From this study it would
appear that FPP, unlike the control antioxidant, significantly decreased oxidized-LDL and near normalizing the anti-
Ox-LDL/Ox-LDL ratio (p<0.001) although unaffecting the lipid profile per . Moreover, only FPP decreased
cyclophilin-A plasma level and plasminogen activator-inhibitor (p<0.01) together with downregulating cyclophilin-A
gene expression (p<0.01). Insulin resistance was only mildly improved. Heavy metals gut clearance proved to be
effectively enhanced by the chelator (p<0.01) and this was not affected by any of the nutraceuticals, nor it added any
further benefit to the biological action of FPP.
Keywords: Fermented Papaya Preparation (FPP); Redox balance;
Antioxidant; Neurodegenerative disease; Caviarlieri
Introduction
Starting in the early 90’ gold standard electron spin resonance
studies had shown that a functional food consisting of fermented
papaya (FPP, ORI, Oxidative Stress laboratory, Gifu, Japan) exhibited a
powerful anti-oxidative activity on
in vitro
cerebral cells [1] as well on
in vivo
epilepsy experimental model, where the epileptogenic
monoamine neural release was consistently reduced [2]. Later on, a
Japanese group proved the capacity of FPP to reduce the derangement
of oxidant/antioxidant balance at the brain level in elderly rats and in
experimental ischemia-reperfusion induced cerebral damage as well
[3,4]. In recent years our group has shown that FPP could benecially
eect on clinical ground a number of redox signaling abnormalities in
a variety of chronic diseases [5,6] as well as some key aging
mechanisms [7,8].
Recent studies using transgenic mice have shown the importance of
proinammatory Cyclophilin A (CypA)-metalloproteinase-9 in blood-
brain-barrier integrity [9] while other have proved that a specic
inhibitor of CyPA could reduce neuroinammation, improve motor
neurons activity and prolong survival in a mouse model of
amyotrophic lateral sclerosis [10]. e potential neuroprotective eects
of FPP are at the moment the target of a clinical study on Parkinson’s
disease patients by the neurology group of Nordera. is group has
reported some preliminary promising results such as a reduction of
motor scores of the Unied Parkinson Disease Rating Scale,
improvement of Activity of Daily Living performance and of redox
biochemistry (G. Nordera, personal communication at Oxidative
Stress in Health and Prevention, September 24, 2014 http://
www.centrostressossidativo.it/video-congresso-stress-ossidativo/). is
latter aspect has received further support by the ne biochemical
Clinical Pharmacology &
Biopharmaceutics Marotta et al., Clin Pharmacol Biopharm 2017, 6:2
DOI: 10.4172/2167-065X.1000170
Research Article OMICS International
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
analysis of same patients cohort by Bolner et al. [11]. Moreover, it has
been recently shown that FPP could dramatically decrease the
oxidative stress parameters in established Alzheimer Disease (AD)
patients [12].
In recent years a number of studies have reported the signicant
relationship between metabolic syndrome and neurodegenerative
disease [13-15]. Indeed, current views on dietary-related diseases and
also neuroimaging data have reported the association between dis-
metabolic patterns and neurodegenerative damage [16] where low-
grade inammation and insulin-resistance seem to be among main
background factors involved [17-19]. As for metabolic syndrome,
dierent groupings of the following metabolic parameters have been
listed in each classication, such as insulin resistance,
hypertriglyceridemia, low HDL-C, obesity/increased waist
circumference, altered glucose tolerance/diabetes mellitus,
hyperinsulinemia, microalbuminuria and hypertension. In the current
study we referred to the denition of MS as outlined by the American
Heart Association and the National Heart, Lung, and Blood Institute
[20].
A further open issue regarding the multifactorial pathophysiology
of neurodegenerative disease is the potentially detrimental role played
by occupational and environmental exposure to heavy metals [21-25].
Indeed, while some metals are involved in physiological enzymatic
reactions, once they accumulate in the brain, especially if overtly acting
as xenobiotic, may give raise to oxidative stress phenomena with
mitochondrial dysfunction, protein misfolding and aberrant
autophagic processes [26-28]. As a matter of fact the update literature
suggests that the aggregation of disease-related proteins during
physiological aging can be advocated for by abnormal protein
homeostasis observed. Such abnormalities may thus represent a
biomarker of aging that could modulate life span and cause
neurodegeneration. Albeit not fully unfolded as yet, it seems that the
interplay between combined genetic and environmental risk factors
(from diet to life style to pollutants) with intrinsic age-related oxi-
inammatory changes may be advocated for to explain the current
increasing surge of neurodegenerative diseases. Given all above, the
current challenging quest of medical community is mostly focused to
track down early biomarkers of likely modiable causative factors
together with possible benecial approaches within a multifunctional
therapeutic armory. e aim of the present study was to evaluate FPP
use in impending metabolic disease patients with potentially higher
risk to develop a neurodegenerative disease.
Methods
Ethics
All procedures were approved by an independent Ethical
Committee for nonpharmacological research (ReGenera Research
Group for Aging Intervention, protocol FPP-MSNEU-10/2015). Each
subject recruited for the study was fully informed and treated in
compliance with the guidelines of the Declaration of Helsinki.
FPP
e FPP used in the present study was obtained from
Carica papaya
L. cultivated in Hawaii, following yeast fermentation for 10 months
and pharmaceutical-grade batch-to-batch control at the Osato
Research Institute (Gifu, Japan). e nal composition of FPP per 100
g is as follows: 90.7 g carbohydrates, 17 μg vitamin B6, 2 μg folic acid,
2.5 mg calcium, 16.9 mg potassium, 240 μg niacin, 4.6 mg magnesium,
14 μg copper, 75 μg zinc, 16 mg arginine, 6 mg lysine, 5 mg histidine,
11 mg phenylalanine, 9 mg tyrosine, 18 mg leucine, 9 mg isoleucine, 5
mg methionine, 13 mg valine, 11 mg glycine, 8 mg proline, 37 mg
glutamic acid, 11 mg serine, 8 mg threonine, 27 mg aspartic acid, and 2
mg tryptophan. is formulation was chosen also to replicate a similar
mixture reported on the market to have antioxidant eect in humans.
Other claimed “fermented papaya” extracts available on the Italian
market were excluded since prior in-house electron spin resonance
testing had ascertained their poor to nil antioxidant eect (Mantello P.
ORI Oxidative Stress Research Lab, Gifu, Japan. In-house data).
Study design
e study population consisted 90 patients ranging from 45 to 65
years old, with impending metabolic syndrome (2 parameters out of
the above-mentioned classication) and 30 age-matched healthy
volunteers (Table 1). Patients with secondary hypertension,
cardiomyopathy, severe abnormalities of liver, thyroid and kidney
function, cerebrovascular diseases, grossly elevated total cholesterol
(>280 mg/dl or LDL >180 mg/dl), malignancies and history of
coronary bypass surgery or on insulin treatment were excluded from
this study. Exclusion criteria were also the consumption of antioxidant
or other supplements in the past 3 months.
All patients were screened for ApoE4 polymorphism to rule out the
presence of a known risk gene pattern for neurodegenerative disease.
is was a RCT, double-blind study, with FPP 4.5 g given twice a day
vs. common antioxidant cocktail (trans-resveratrol 20 mg, selenium 60
mcg, vit E 30 mg, vit C 100 mg, papaya avour) twice a day. en, aer
21 month continuous treatment period, an heavy metal chelator (talc-
size, higher potency chabasite-phillipsite zeolites naturally-occurring
mixture, Novagenics-SOL Ltd., Hong Kong) was added at the dosage of
3 g/nocte for further 3 months to complete the 24 months study
(Figure 1).
At entry 1, 3, 6, 12, 21 and 24 months, biological samples were
collected (BD Vacutainer sampling tubes) to assess biochemical
assessments as below mentioned. Blood serum was isolated by
centrifugation and stored at under −70°C until analysis. Repeated
thawing and freezing were avoided.
Parameters
Pre-recruiting gene testing: Ten millilitres of peripheral whole blood
was withdrawn from an antecubital vein of all recruited subjects.
Genomic DNA was extracted by a modied salting-out method (by
precipitating DNA with the use of 96% ethanol). is was processed to
ApoE genotyping with modications in the multiplex amplication
refractory mutation system PCR (Multi-ARMS PCR). e primer
sequences used in the PCR analysis were as follows: APOE rs7412
F:ATGCCGATGACCTGCAGA R:ACTGGCGCTGCATGTCTT
Product size (bp) 684; rs429358 F:TCGGAACTGGAGGAACAACT
R:TACACTGCCAGGCGCTTCT Product size (bp) 260. Genomic
DNA (50 ng) amplication (ABI Prism 310, Perkin Elmer, USA)
comprised 30 cycles (94ºC for 5 min, 94ºC for 30 s, 65ºC for 30 s, and
72º for 30 s), followed by a cycle of 10 min at 72°C for the nal chain
extension. Moreover, for the analysis of restriction fragment length
polymorphism, the amplication product was digested with HhaI,
followed by 5.0% agarose gel electrophoresis for 2 h and 30 min,
stained with electro-uorescent red stain (ermoFisher, USA) and
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 2 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
UV light-visualization of the DNA fragments. As stated above, ApoE4
subjects were excluded from the study.
Parameters Healthy
Control
FPP-
treated
Antioxidant-
treated
Total no. (male/female) 30 (15/15) 45 (33/18) 45 (28/17)
Mean age 67 71 66
Mean BMI (range) 23 (21-25) 27 (23-35) 26 (24-34)
Family history of diabetes 1/30 14/45 21/45
Family history of hypertension 1/30 17/45 18/45
Overt diabetes NA 6/45 11/45
Duration of diabetes (years) NA 4 3
Dyslipidemia 0/30 28/45 31/45
Smoking 4/30 7/45 12/45
Waist (cm) 82 98 99
Waist/hip ratio 0.82 0.92 0.95
Mean Systolic Blood Pressure
(mmHg)
122 159 161
Mean Diastolic Blood Pressure
(mmHg)
65 78 81
Physical activity 15/30 6/45 12/45
Table 1: Anthropometric, clinical and biological parameters in studied
subjects.
Figure 1: Study design, see text.
Biochemical tests: routine tests (WBC, transaminases, urea,
creatinine, glucose, glycated haemoglobin, uric acid, HOMA test, LDL
and HDL cholesterol) were assayed by using the 7060 Automatic
Biochemical Analyzer (Hitachi, Ltd, Tokyo, Japan). Oxidized
cholesterol, anti-oxidised LDL, ratio of anti-ox-LDL/ox-LDL
cyclophilin-A, plasminogen activator inhibitor-1 were tested as below.
Oxidised LDL and anti-oxidised LDL antibodies assessment: ese
parameters were obtained by fractionation of LDL and preparation of
oxLDL as antigen in the antibody detection assay, according to what
reported by Ketelhuth et al. [29]. For this purpose a commercially
available ELISA kits (ChusaBiotech Co. Ltd., China), was employed.
Briey, microtiter plates were coated with 10 g/ml in PBS of either
native LDL or MDA-LDL to measure anti-oxidized LDL antibodies.
Serum samples were diluted 1:100 in 1% BSA/PBS and incubated for 2
h at 37°C and overnight at 4 C. Aerwards, the plates were washed
four times with alkaline phosphatase-conjugated anti-human IgG
(Sigma Chemical, St. Louis, MO) and le for 3 h at room temperature.
e reaction was halted aer 60 min with 1% BSA/PBS for 2 h at room
temperature. e absorbance was at 450 nm using a Multiskan RC
ELISA reader (ermo Fisher Scientic, USA). e binding of
antibodies to oxidized LDL was determined by subtracting native LDL
binding value from the one of MDA-LDL while the results were
expressed as an Optical Density (OD).
Measurement of cyclophilin A in plasma: Cyclophilin A
concentration in plasma was assayed with quantitative sandwich
enzyme immunoassay kit (Wouhan-UCSN BioScience, Wouhan,
China). Briey, a monoclonal antibody specic for CypA was pre-
coated onto a microplate. Standards and samples were put into the
wells and CypA present is bound by the immobilized antibody. An
enzyme-linked polyclonal antibody specic for CypA is added to the
wells. Plates were washed three times with PBS and a substrate solution
was added to the wells. e following dose-dependent color
appearance was then halted by an acid solution and the color intensity
measured by a reader as above. ere was no evidence of any
detectable cross-reactivity with any other tested proteins. All samples
were analyzed in triplicate and samples with a CV >10% were
discarded.
Determination of plasminogen activator inhibitor-1: PAI-1 serum
concentration was measured by sandwich enzyme ELISA (PAI-1;
DSE100; R&D Systems). Active PAI-1 levels were calculated from a
standard curve constructed by using recombinant human PAI-1. By
this method either active or latent forms of PAI-1 are detected [30].
Intra and inter-assay coecients of variation of PAI-1 antigen were
3.9% and 4.8%, respectively.
Cyclophilin-A gene expression study: Reverse
transcriptionpolymerase chain reaction (RTPCR) was used to
determine their expression at 0, 90 days and at the end of the 24 month
study period. Briey, cells were lysed by Ambion lysis buer (Ambion,
Carlsbad, CA, USA) for 20 min and the lysates were mixed with same
volume of 64% ethanol. e lysate products were put into a column
and centrifuged at 10,000 × g for 1 min. Aerwards, a buer consisting
of 700 μl buer 1 and 500 μl buer 2/3 (Genosolution, Ltd., Shenyang,
China) was used to wash the column. Aer incubation with 50 μl
buer (GenePharma Co., Ltd.), the resulting ow was collected and 50
μl elution buer was added and a further centrifugation was
performed. Next, 1 μl DNAse I was added to 20 μl of RNA solution
with suitable DNAse I buer (GenePharma Co., Ltd.) and incubated at
37˚C for 2 h. e DNAse I was removed using DNAse reagent and
puried RNA obtained by a centrifugation at 10,000 × g for 1 min. e
quantity of total RNA was measured by optical density at 260 nm and
the procedure checked through 1.5% agarose gel electrophoresis. RT
was run in a 20ml solution containing 3 μg total RNA using the Revert
cDNA Synthesis kit (Toyobo Biotech, Co., Ltd., Shanghai China). PCR
was done in a thermal cycler aer prior denaturation at 94˚C for 5
min, followed by amplication for 30 cycles of denaturation at 94˚C
for 40 s, annealing phase at 65˚C for 1 min followed by 72˚C for 1 min.
Aerwards, 5 μl PCR material was isolated by electrophoresis on a
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 3 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
1.5% agarose gel and stained by ethidium bromide. e following
primer was used to carry out the test: CypA forward, 5'GTC AAC CCC
ACC GTG TTC TTC3', and reverse, 5'TTT CTG CTG TCT TTG GGA
CCTTG3' and glyceraldehyde3phosphate dehydrogenase (GAPDH)
forward, 5'ACC ACA GTC CAT GCC ATCAC3', and reverse, 5'TCC
ACC ACC CTG TTG CTGTA3'. e PCR products were subjected to
electrophoresis on a 1.4% agarose gel and the results were
quantitatively assessed by Gelmatrix Scan soware (BioMatrixscan
Inc., Shanghai, China).
Faecal heavy metal measurement
A 15 mg of stool sample was collected and an inductively coupled
argon plasma mass spectrometry (ICP-MS) with an Agilent 7500ce
device was used to test the following heavy metals: lead, arsenic,
mercury, cadmium, nickel and beryllium. According to the IUPAC
guideline [31] the reference value was dened within the 95%
condence interval of the 95th population percentile. e results in the
specimens were expressed as μg/L. Data were expressed on a dry
weight basis so to avoid the variability related to watery component of
the sample.
Results
Routine tests
Blood tests (WBC, creatinine, urea, glucose, glycated haemoglobin,
uric acid, total cholesterol, LDL-cholesterol, HDL-cholesterol,
triglycerides, transaminases, gamma-GT, alkaline phosphatase,
bilirubin) were not aected by any of the nutraceuticals employed,
except a not signicant trend improvement of HOMA test in the FPP-
supplemented group (data not shown).
Oxidized LDL cholesterol and anti-oxidised LDL antibodies
Whereas the group treated with antioxidant cocktail did not show
any statistically signicant dierence in the oxidised lipid asset,
patients treated with FPP showed a signicant improvement of these
parameters (Figure 2). In particular, the oxidized LDL cholesterol was
normalised by this intervention (p<0.001 vs. baseline). is variation
was signicantly detectable starting from the third month observation
period while the anti-Ox-LDL reverted to near to normal level from 12
months observation time onwards (p<0.05, Table 2). ere was no
signicant correlation between these parameter and main clinical
history data (presence or duration of diabetes, hypertension or
dyslipidemia). No signicant gender dierence was detected as for
these parameters are concerned.
Serum level of cyclophilin-A and plasminogen activator
inhibitor-1
At baseline there appeared a signicant correlation between
plasminogen activator inhibitor-1 and uric acid (r: 0.62, p<0.05, not
shown). As compared to baseline and to antioxidant cocktail control,
FPP showed to signicantly decrease serum levels of cyclophillin-A
from 1st month observation (p<0.01, Figure 3) and plasminogen
activator inhibitor-1 from 6 month observation (p<0.01, Figure 4). A
signicant correlation appeared between cyclophillin-A and oxidised-
LDL (r: 0.69, p<0.05, not shown) but not with plasminogen activator
inhibitor-1.
Cyclophilin-A gene expression study
At baseline observation, CyP-A gene expression was comparable
with what observed in healthy controls (Figure 5). However, as
compared to antioxidant cocktail-treated group, FPP enabled a
signicant upregulation at 3 and at 24 months (p<0.05, Figure 5).
Faecal heavy metal assessment
ere was no correlation between this parameter, either when
analysed as a single heavy metal or as a whole pooled value and any of
the biochemical or gene expression values measured.
Irrespective of the group, all patients showed an overall abnormal
faecal metals prole with the exception of nickel and arsenicum.
Unlike FPP or antioxidant control, the chelator-treated group
showed a signicantly greater faecal elimination of heavy metals
(namely: mercury, cadmium, lead and beryllium (Table 3, p<0.05 vs.
baseline and antioxidant combination). ere was a not signicant
trend correlation between cumulative heavy metal values (either at
baseline or aer treatment) with oxidised-LDL or its antibody (data
not shown).
Discussion
Oxidative modification of LDLs may be a prerequisite for the rapid
accumulation of LDLs within macrophages to form foam cells; indeed,
oxidized LDL has been found in extracts from atherosclerotic lesions
[3]. Oxidative modification of LDLs induces also the transformation of
immunogenic epitopes in the LDL molecule, which lead to the
formation of antibodies against oxidized LDLs in the serum [4].
Nevertheless, the clinical importance of these autoantibodies is still
under discussion. For example, in patients with diabetes, no
association has been found between anti-oxidized-LDL antibodies and
microvascular complications [11], nor has an association been found
with the levels of cholesterol in patients with heterozygous
hypercholesterolemia [32] or with the intensity of serum oxidizability
[33]. Others have reported an inverse relation between levels of
cholesterol and anti-oxidized-LDL antibodies in the general
population [34] and this seems to agree with the nding of an inverse
link between IgM autoantibodies to oxidized LDL and carotid artery
atherosclerosis [35]. e preliminary data of our study conrms that
FPP unlike the control antioxidant, specically decreased oxidized-
LDL and, on the long run, also enhanced its antibodies concentration,
although not modifying the lipid prole. Unlike what reported by
Balada et al. [36], we did not notice any signicant gender dierence as
for the levels of anti-oxidized LDL antibodies. While brain cholesterol
metabolism is separated from the systemic circulation, oxidised
cholesterol moieties may damage the functional integrity of blood-
brain barrier with potentially detrimental eects on neuronal
population [37]. e same group has also shown on an experimental
model that oxidised-LDL lipids and 27-OH-cholesterol may trigger Aβ
production by GSH depletion and β-secretase-1 activation in neuronal
cells [38]. It is noteworthy mentioning that a recent experimental study
connects a fat-based/high calorie diet with metabolic syndrome,
oxidative stress and signicant hippocampal and temporal area
degeneration together with memory dysfunction [39]. is evident
histopathological nding is also in agreement with one of the current
clinical hypothesis [40].
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 4 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
Figure 2: Variation of plasma level of oxidised-LDL during treatment with the two nutraceuticals formulas. *p<0.001 vs. baseline values;
**p<0.01 vs. FPP-treated group.
Anti-ox-LDL EU/mL
(18.2 ± 3.4)
Entry 6 months 12 months 21 months 24 months
FPP 28.1 ± 10.2 28.2 ± 9.3 21.4 ± 14.5*21.2 ± 10.7*20.5 ± 7.9*
AntiOx mixture 26.8 ± 12.9 27.1 ± 8.8 27.4 ± 6.4 26.9 ± 13.3 25,7 ± 16.1
Table 2: Comparison of study parameters between the two treatment groups. In brackets: Values in healthy control subjects. *p<0.05 vs. baseline
and vs. Antioxidant cocktail.
Moreover, only FPP decreased cyclophilin-A plasma level and
plasminogen activator-inhibitor (PAI-1) (p<0.01). During
inammatory processes and following oxidative stress, CyPA is
released into the extracellular compartment by inamed cells [41,42]
as a tentative protection response but this factor ends up being a potent
chemoattractant for human monocytes and neutrophils. Cyclophilin A
has indeed been shown to be more strongly associated with oxidative
stress and inammation than C-reactive protein [43]. Interestingly,
prior small open pilot in-house studies testing same cohort of patients
with an omega-3-rich sturgeon-based supplement (Caviarlieri, Swiss
cap) did not benecially aect the lipid prole, oxidised lipid
parameters or CyPA. On the contrary, this latter variable showed a
worsening change either as plasma level or as gene expression, making
it a not advisable choice in metabolic syndrome or diabetes (J. Cervi,
manuscript in preparation). e brain is characterized by a low content
of antioxidant systems and the damage caused by oxidative stress is
one of the earliest pathophysiological events in the development of
Alzheimer's disease where plasma oxidized-LDL level has been
positively correlated with disease severity [44].
A further independent cardiovascular risk factor [45] as well co-
aggravating parameter in metabolic syndrome [46] is represented by
PAI-1, the major regulator of brinolysis and a stress-related factor. As
mentioned above, unlike the antioxidant cocktail and the recent
negative study testing high dose tocotrienol [47], FPP supplementation
brought about a signicant normalization of altered values of this
parameter.
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 5 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
Figure 3: Variation of plasma level of Cyclophillin-A during
treatment with the two nutraceuticals formulas. *p<0.01 vs. baseline
values; **p<0.01 vs. FPP-treated group.
Figure 4: Variation of plasma level of plasminogen-activator
inhibitor-1 during treatment with the two nutraceuticals formulas.
*p<0.01 vs. baseline values; **p<0.01 vs. FPP-treated group.
Figure 5: Cyclophillin-A gene expression at baseline and during 24 months follow up under treatment with two dierent nutraceuticals. Only
FPP was eective in upregulating this parameter (*p<0.05 vs. baseline and vs. Antioxidant cocktail).
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 6 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
Months Normal range FPP AntiOx mixture Chelator
Mercury
mg/kg dry wt.
21
24
<0.5 (0.3 ± 0.2)
0.4 ± 0.3
(0.4 ± 0.1)
0.4 ± 0.2
(0.3 ± 0.2)
1.1 ± 0.3*
Cadmium
mg/kg dry wt.
21
24
<0.5 (0.2 ± 0.3)
0,5 ± 0.14
(0.4 ± 0.3)
0.5 ± 0.12
(0.3 ± 0.1)
1.4 ± 0.31*
Lead
mg/kg dry wt.
21
24
<0.5 (0.2 ± 0.1)
0.2 ± 0.6
(0.2 ± 0.12)
0.3 ± 0.09
(0.3 ± 0.02)
1.1 ± 0.41*
Arsenicum
mg/kg dry wt.
21
24
<0.3 (0.1 ± 0.04)
0.09 ± 0.032
(0.1 ± 0.08)
0.1 ± 0.03
(0.3 ± 0.04)
0.2 ± 0.12
Beryllium
mg/kg dry wt.
21
24
<0.009 (0.004 ± 0.003)
0.009 ± 0.004
(0.007 ± 0.004)
0.004 ± 0.003
(0.009 ± 0.004)
0.015 ± 0.002*
Nickel
mg/kg dry wt.
21
24
<8 (5.8 ± 0.8)
5.6 ± 0.3
(6.4 ± 1.2)
6.2 ± 1.3
(6.1 ± 0.14)
6.9 ± 0.38
Table 3: Faecal heavy metal test: Eect of nutraceuticals with and without selected chelator. In brackets: baseline values corresponding to time 21
months and 24 month observation period of the trial. *p<0.05 vs. baseline and vs. antioxidant cocktail.
On the other hand, a one-year consumption of a nutraceutical rich
in resveratrol, one of the ingredient of our antioxidant cocktail, had
shown to improve PAI-1, but CyPA and oxidised lipid markers were
not tested [48]. A vascular component of neurodegenerative disease
pathophysiology is credited by several epidemiological studies [49] and
Oh et al. [50] has shown that patients with MCI and AD had
signicantly higher plasma PAI-1 levels, ese appeared to correlate
with cognitive function thus being a potentially early biomarker for
AD. e upregulation of PAI-1 has also been suggested to explain
insucient BDNF neurotrophic support and increased
neurodegeneration while suppressing PAI-1 expression/activity
increases degradation [51]. To the vascular hypothesis is likely to
cooperate also an increased CyPA which may aect endothelial cells
and vascular smooth muscle cells [52,53]. Whatever the main
mechanisms, it seems that by matching the metabolic proling and
AD, there might be three subtypes; an inammatory one, a non-
inammatory type and a zinc deciency-associated type aecting
younger population [54]. In a prior study using a sturgeon eggs-
derived compound of putative anti-inammatory eect (Caviarlieri,
Swiss cap, Switzerland) we had shown to increase circulating level of
BDNF in healthy occupationally-stressed individuals [55]. However,
internal and follow up data showed that this was a rather short-lived
eect and disappointingly unaecting PAI-1, OxLDL, Cyp-A or
proBDNF. So, its initially suggested neuroprotective eect
in vitro
(but
not at brain gene expression level in animals) had to be abandoned on
clinical ground by our group.
On the other hand, the downregulating eect played by FPP on
CyPA gene expression, but not from the antioxidant control, may be of
relevance, when considering that it has become clear that a
hyperglicemia and oxidative stress milieu may stimulate this molecule
[56] which, on its turn, drives the ox-LDL-mediated dierentiation
and activation of monocytes to foam cells [57,58].
Heavy metals gut clearance was not aected by any of the
nutraceuticals. However, the chelator, by itself remarkably increased
fecal gut discharge of heavy metals but only in those patients with
abnormal baseline values. e addition of a chelator to the control
nutraceuticals didn’t prove to yield better results while the addition to
FPP showed a trend increase of fecal discharge of cadmium (p<0.07 vs.
oral chelator alone, n.s.).
e identication and application of novel biomarkers in nutritional
and life-style interventional plans may foster healthy longevity and
benecially aect the occurrence of concomitant illnesses. Our
preliminary data suggest that FPP might play a noteworthy role within
a wider and comprehensive preventive medicine strategy plan for
impending metabolic diseases which are potential co-factors of
neurodegenerative disease. Environmental neurotoxins disrupt protein
processing as shown in experimental studies [26-28] while eective
heavy metals chelators remain a further additive avenue to possibly
pursue in selected cases.
Conclusion
As overall platform, virtuous dietary regimen such as the nutritional
model of Mediterranean diet still maintains its importance to counter
ght the cellular events involved in the atherosclerosis and
neurodegenerative process [59,60]. However, given these promising
results, longer studies addressing the issue of specic long term
evidence-based nutraceutical implementations are highly awaited.
Lastly, most recent research work points out the crucial importance of
mitochondrial dynamics in metabolic and neurodegenerative diseases
[61]. Indeed, exposure of environmental toxin, high-calorie intake,
glucolipotoxicity brings about dysfunctional mitochondria which
perpetuate ROS spilling over. Interestingly, the work of Collard et al.
[62] in diabetic rats has shown how FPP optimised mitochondrial
energetics. Such “cellular energetic” hypothesis as one of the possible
FPP mechanisms also crossing the blood-brain-barrier has been
recently envisaged by Hayashi Y. (in: Mantello A, Catanzaro C, He F,
Cuari B, Bissi L, Milazzo M, Lorenzetti A. Marotta F. Novel
Nutrigenomics Avenues in Nutraceuticals Use: e Current Status of
Fermented Papaya Preparation–Ed. E. Aguilar, Bioactive compounds:
At the frontier between nutrition and pharmacology, 2016; 94-119).
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 7 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
Acknowledgments
is study was supported by an unbiased grant from Osato Research
Institute, Japan (91%) and with a minor grant (9%) rom San Babila
Clinic, Milano.
References
1. Marotta F, Celep GS, Cabeca A, Polimeni P (2012) Novel concepts on
functional foods and nutrigenomics in healthy aging and chronic
diseases: a review of fermented papaya preparation research progress.
Functional Foods in Health and Disease 2: 120-136.
2. Santiago LA, Osato JA, Hiramatsu M, Mori A (1992) Fermented papaya
preparation quenched free radicals and inhibited lipids peroxidation in
iron- induced epileptic focus in rats. Oxygen Rad 4: 405-408.
3. Aruoma OI, Hayashi Y, Marotta F, Mantello P, Rachmilewitz E, et al.
(2010) Applications and bioecacy of the functional food supplement
fermented papaya preparation. Toxicology 278: 6-16.
4. Zhang J, Mori A, Chen Q, Zhao B (2006) Fermented papaya preparation
attenuates β-amyloid precursor protein: β-amyloid-mediated copper
neurotoxicity in β-amyloid precursor protein and β-amyloid precursor
protein Swedish mutation overexpressing SH-SY5Y cells. Neuroscience
143: 63-72.
5. Marotta F, Chui DH, Jain S, Koike K, Zhou L, et al. (2011) Eect of a
fermented nutraceutical on thioredoxin level and TNF-alpha signaling in
cirrhotic patients. J Biol Regul Homeost Agents 25: 37-45.
6. Tomella C, Catanzaro R, Illuzzi N, Cabeca A, Celep G, et al. (2014) e
hidden phenomenon of oxidative stress during treatment of subclinical-
mild hypothyroidism: a protective nutraceutical intervention.
Rejuvenation Res 17: 180-183.
7. Marotta F, Weksler M, Naito Y, Yoshida C, Yoshioka M, et al. (2006)
Nutraceutical supplementation: eect of a fermented papaya preparation
on redox status and DNA damage in healthy elderly individuals and
relationship with GSTM1 genotype: a randomized, placebo-controlled,
cross-over study. Ann N Y Acad Sci 1067: 400-407.
8. Marotta F, Pavasuthipaisit K, Yoshida C, Albergati F, Marandola P (2006)
Relationship between aging and susceptibility of erythrocytes to oxidative
damage: in view of nutraceutical interventions. Rejuvenation Res 9:
227-230.
9. Bell RD, Winkler EA, Singh I, Sagare AP, Deane R, et al. (2012)
Apolipoprotein E controls cerebrovascular integrity via cyclophilin A.
Nature 485: 512-516.
10. Pasetto L, Pozzi S, Castelnovo M, Basso M, Estevez AG, et al. (2016)
Targeting Extracellular Cyclophilin A Reduces Neuroinammation and
Extends Survival in a Mouse Model of Amyotrophic Lateral Sclerosis. J
Neurosci 37: 1413-1427.
11. Bolner A, Micciolo R, Ottavio Bosello O, Nordera G (2016) Eect of
papaya supplementation on oxidative stress markers in Parkinson’s
disease. Oxid Antioxid Med Sci 5: 49-55.
12. Barbagallo M, Marotta F, Dominguez LJ (2015) Oxidative stress in
patients with Alzheimer's disease: eect of extracts of fermented papaya
powder. Mediators Inamm.
13. Matsuzaki T, Sasaki K, Tanizaki Y, Hata J, Fujimi K, et al. (2010) Insulin
resistance is associated with the pathology of Alzheimer disease: the
Hisayama study. Neurology 75: 764-770.
14. Schrijvers EM, Witteman JC, Sijbrands EJ, Hofman A, Koudstaal PJ, et al.
(2010) Insulin metabolism and the risk of Alzheimer disease: the
Rotterdam Study. Neurology 75: 1982-1987.
15. Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, et al. (2011)
Insulin resistance and Alzheimer-like reductions in regional cerebral
glucose metabolism for cognitively normal adults with prediabetes or
early type 2 diabetes. Arch Neurol 68: 51-57.
16. Pistollato F, Cano SS, Elio I, Vergar MM, Giampieri F, Battino M. (2015)
e Use of Neuroimaging to Assess Associations Among Diet, Nutrients,
Metabolic Syndrome, and Alzheimer's Disease. J Alzheimers Dis 48:
303-318.
17. Minihane AM, Vinoy S, Russell WR, Baka A, Roche HM, et al. (2015)
Low-grade inammation, diet composition and health: current research
evidence and its translation. Br J Nutr 114: 999-1012.
18. Li X, Song D, Leng SX (2015) Link between type 2 diabetes and
Alzheimer's disease: from epidemiology to mechanism and treatment.
Clin Interv Aging 10: 549-560.
19. Kim B, Feldman EL (2015) Insulin resistance as a key link for the
increased risk of cognitive impairment in the metabolic syndrome. Exp
Mol Med 47: e149.
20. National Cholesterol Education Program (NCEP) Expert Panel on
Detection, Evaluation, and Treatment of High Blood Cholesterol in
Adults (Adult Treatment Panel III), “ird report of the National
Cholesterol Education Program (NCEP) (2002) Expert panel on
detection, evaluation, and treatment of high blood cholesterol in adults
(Adult Treatment Panel III) nal report. Circulation 106: 3143-3421.
21. Sun YH, Nfor ON, Huang JY, Liaw YP (2015) Association between dental
amalgam llings and Alzheimer's disease: a population-based cross-
sectional study in Taiwan. Alzheimers Res er 7: 65-69.
22. Hare DJ, Rembach A, Roberts BR (2016) e Emerging Role of
Metalloproteomics in Alzheimer's Disease Research. Methods Mol Biol
1303: 379-389.
23. Souza-Talarico JN, Marcourakis T, Barbosa F, Moraes Barros SB, Rivelli
DP, et al. (2017) Association between heavy metal exposure and poor
working memory and possible mediation eect of antioxidant defenses
during aging. Sci Total Environ 575: 750-757.
24. Maya S, Prakash T, Madhu KD, Goli D (2016) Multifaceted eects of
aluminium in neurodegenerative diseases: A review. Biomed
Pharmacother 83: 746-754.
25. Escudero-Lourdes C (2016) Toxicity mechanisms of arsenic that are
shared with neurodegenerative diseases and cognitive impairment: Role
of oxidative stress and inammatory responses. Neurotoxicology 53:
223-235.
26. Malecki EA (2001) Manganese toxicity is associated with mitochondrial
dysfunction and DNA fragmentation in rat primary striatal neurons.
Brain Res Bull 55: 225-228.
27. Zhang Z, Miah M, Culbreth M, Aschner M (2016) Autophagy in
Neurodegenerative Diseases and Metal Neurotoxicity. Neurochem Res 41:
409-422.
28. Karri V, Schuhmacher M, Kumar V (2016) Heavy metals (Pb, Cd, As and
MeHg) as risk factors for cognitive dysfunction: A general review of metal
mixture mechanism in brain. Environ Toxicol Pharmacol 48: 203-213.
29. Ketelhuth DF, Tonini GC, Carvalho MD, Ramos RF, Boschcov P, et al.
(2008) Autoantibody response to chromatographic fractions from
oxidized LDL in unstable angina patients and healthy controls. Scand J
Immunol 68: 456-462.
30. Declerck PJ, Moreau H, Jespersen J (1993) Multicenter evaluation of
commercially available methods for the immunological determination of
plasminogen activator inhibitor-1 (PAI-1). romb Haemost 70: 858-863.
31. Cornelis R, Fuentes-Arderiu X, Bruunshuus I, Templeton D (1997)
International Union of Pure and Applied Chemistry and International
Federation of Clinical Chemistry, Committee on Nomenclature.
Properties and Units (C-NPU): properties and units in the clinical
laboratory sciences. IX. Properties and units in trace elements (IFCC-
IUPAC technical report 1997). Eur J Clin Chem Clin Biochem 35:
833-843.
32. Delimaris I, Georgopoulos S, Kroupis C, Zachari A, Liberi M, et al. (2008)
Serum oxidizability, total antioxidant status and albumin serum levels in
patients with aneurysmal or arterial occlusive disease. Clin Biochem 41:
706-711.
33. Craig WY, Poulin SE, Neveux LM, Palomaki GE, Dostal DA (1995) Anti-
oxidized LDL antibodies and antiphospholipid antibodies in healthy
subjects: relationship with lipoprotein- and oxidation-related analytes. J
Autoimmun 8: 713-726.
34. Tinahones FJ, Gomez-Zumaquero JM, Rojo-Martinez G, Cardona F,
Esteva de Antonio IE, et al. (2002) Increased levels of anti-oxidized low-
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 8 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
density lipoprotein antibodies are associated with reduced levels of
cholesterol in the general population. Metabolism 51: 429-431.
35. Karvonen J, Paivansalo M, Kesaniemi YA, Horkko S (2003)
Immunoglobulin M type of autoantibodies to oxidized low-density
lipoprotein has an inverse relation to carotid artery atherosclerosis.
Circulation 108: 2107-2112.
36. Balada E, Ordi-Ros J, Matas L, Mauri M, Bujan S, et al. (2002)
Atherosclerosis and anti-oxidized low density lipoprotein antibodies in an
elderly population. Med Clin (Barc) 119: 161-165.
37. Dias IH, Polidori MC, Griths HR (2014) Hypercholesterolaemia-
induced oxidative stress at the blood-brain barrier. Biochem Soc Trans
42: 1001-1005.
38. Dias IH, Mistry J, Fell S, Reis A, Spickett CM, et al. (2014) Oxidized LDL
lipids increase β-amyloid production by SH-SY5Y cells through
glutathione depletion and lipid ra formation. Free Radic Biol Med 75:
48-59.
39. Treviño S, Aguilar-Alonso P, Flores Hernandez JA, Brambila E, Guevara J,
et al. (2015) A high calorie diet causes memory loss, metabolic syndrome
and oxidative stress into hippocampus and temporal cortex of rats.
Synapse 69: 421-433.
40. van Dijk G, van Heijningen S, Reijne AC, Nyakas C, van der Zee EA, et al.
Integrative neurobiology of metabolic diseases, neuroinammation, and
neurodegeneration. Front Neurosci 9: 173.
41. Jin ZG, Melaragno MG, Liao DF, Yan C, Haendeler J, et al. (2000)
Cyclophilin A is a secreted growth factor induced by oxidative stress. Circ
Res 87: 789-796.
42. Arora K, Gwinn WM, Bower MA, Watson A, Okwumabua I, et al. (2005)
Extracellular cyclophilins contribute to the regulation of inammatory
responses. J Immunol 175: 517-522.
43. Taguchi I, Abe S, Inoue T (2013) Cyclophilin A is a promising predictor
of coronary artery disease. Circ J 77: 321-232.
44. Zhao Z, Zhou H, Peng Y, Qiu CH, Sun QY, et al. (2014) Expression and
signicance of plasma 3-NT and ox-LDL in patients with Alzheimer's
disease. Genet Mol Res 13: 8428-8435.
45. Nikolopoulos GK, Bagos PG, Tsangaris I, Tsiara CG, Kopterides P, et al.
(2014) e association between plasminogen activator inhibitor type 1
(PAI-1) levels, PAI-1 4G/5G polymorphism, and myocardial infarction: a
Mendelian randomization meta-analysis. Clin Chem Lab Med 52:
937-950.
46. Aburto-Mejía E, Santiago-Germán D, Martínez-Marino M, María
Eugenia Galván-Plata, Almeida-Gutiérrez E, et al. (2017)
Hypobrinolytic State in Subjects with Type 2 Diabetes Mellitus
Aggravated by the Metabolic Syndrome before Clinical Manifestations of
Atherothrombotic Disease. Biomed Res Int 2017: 6519704.
47. Che HL, Kanthimathi MS, Loganathan R, Yuen KH, Tan AT, et al. (2017)
Acute eects of a single dose of tocotrienols on insulinemic and
inammatory responses in metabolic syndrome subjects aer a high-fat
challenge. Eur J Clin Nutr 71: 107-114.
48. Tomé-Carneiro J, Gonzálvez M, Larrosa M, Yáñez-Gascón MJ, García-
Almagro FJ, et al. (2012) I One-year consumption of a grape nutraceutical
containing resveratrol improves the inammatory and brinolytic status
of patients in primary prevention of cardiovascular disease.Am J Cardiol
110: 356-363.
49. Safouris A, Psaltopoulou T, Sergentanis TN, Boutati E, Kapaki E, et al.
(2015) Vascular risk factors and Alzheimer's disease pathogenesis: are
conventional pharmacological approaches protective for cognitive decline
progression? CNS Neurol Disord Drug Targets 14: 257-269.
50. Oh J, Lee HJ, Song JH, Park SI, Kim H (2014) Plasminogen activator
inhibitor-1 as an early potential diagnostic marker for Alzheimer's
disease. Exp Gerontol 60: 87-91.
51. Gerenu G, Martisova E, Ferrero H, Carracedo M, Rantamäki T, et al.
(2017) Modulation of BDNF cleavage by plasminogen-activator
inhibitor-1 contributes to Alzheimer's neuropathology and cognitive
decits. Biochim Biophys Acta 1863: 991-1001.
52. Jin ZG, Melaragno MG, Liao DF, Yan C, Haendeler J, et al. (2000)
Cyclophilin A is a secreted growth factor induced by oxidative stress. Circ
Res 87: 789-796.
53. Suzuki J, Jin ZG, Meoli DF, Matoba T, Berk BC (2006) Cyclophilin A is
secreted by a vesicular pathway in vascular smooth muscle cells. Circ Res
98: 811-817.
54. Bredesen DE (2015) Metabolic proling distinguishes three subtypes of
Alzheimer's disease. Aging (Albany NY) 7: 595-600.
55. Chui DH, Marcellino M, Marotta F, Sweed H, Solimene U, et al. (2014) A
double-blind, rct testing benecial modulation of BDNF in middle-aged,
life style-stressed subjects: a clue to brain protection? J Clin Diagn Res 8:
MC01-MC06.
56. Tsai SF, Hsieh CC, Wu MJ, Chen CH, Lin TH, et al. (2016) Novel ndings
of secreted cyclophilin A in diabetic nephropathy and its association with
renal protection of dipeptidyl peptidase 4 inhibitor. Clin Chim Acta 463:
181-192.
57. Xue Z, Yuan W, Li J, Zhou H, Xu L, et al. (2017) Cyclophilin A mediates
the ox-LDL-induced activation and apoptosis of macrophages via
autophagy. Int J Cardiol 230: 142-148.
58. Ramachandran S, Vinitha A, Kartha CC (2016) Cyclophilin A enhances
macrophage dierentiation and lipid uptake in high glucose conditions: a
cellular mechanism for accelerated macro vascular disease in diabetes
mellitus. Cardiovasc Diabetol 15: 152-158.
59. Casas R, Sacanella E, Estruch R (2014) e immune protective eect of
the Mediterranean diet against chronic low-grade inammatory diseases.
Endocr Metab Immune Disord Drug Targets 14: 245-254.
60. Claassen JA (2015) New cardiovascular targets to prevent late onset
Alzheimer disease. Eur J Pharmacol 763: 131-134.
61. Jha SK, Jha NK, Kumar D, Ambasta RK, Kumar P (2016) Linking
mitochondrial dysfunction, metabolic syndrome and stress signaling in
Neurodegeneration. Biochim Biophys Acta pii: S0925-S4439.
62. Collard E, Roy S (2010) Improved function of diabetic wound-site
macrophages and accelerated wound closure in response to oral
supplementation of a fermented papaya preparation. Antioxid Redox
Signal 13: 599-606.
Citation: Marotta F, Marcellino M, Solimene U, Cuffari B, Yadav H, et al. (2017) A 2-year Double-Blind RCT Follow-up Study with Fermented
Papaya Preparation (FPP) Modulating Key Markers in Middle-Age Subjects with Clustered Neurodegenerative Disease-Risk Factors.
Clin Pharmacol Biopharm 6: 170. doi:10.4172/2167-065X.1000170
Page 9 of 9
Clin Pharmacol Biopharm, an open access journal
ISSN: 2167-065X
Volume 6 • Issue 2 • 1000170
... In fact, FPP ® has shown a powerful in vitro anti-oxidative activity on brain cells [39], as well on in vivo experimental model of epilepsy consistently reducing neural release of epileptogenic monoamine [40]. Moreover, FPP ® showed a clear action in reducing the derangement of oxidant/antioxidant balance at the brain level in elderly rats and in experimental ischemia-reperfusion model [20,41,42]; FPP ® modulates oxidative DNA damage, protecting brain from oxidative damage in hypertensive rats and reducing genotoxic effect of H2O2 [43], and protecting the body from the aging-related diseases [44][45][46][47], including neurodegenerative diseases [47][48][49]. However, a clear in vivo action of FPP ® on the molecular signature of aging, such as telomerase activity and telomeres length has not been provided yet. ...
... In fact, FPP ® has shown a powerful in vitro anti-oxidative activity on brain cells [39], as well on in vivo experimental model of epilepsy consistently reducing neural release of epileptogenic monoamine [40]. Moreover, FPP ® showed a clear action in reducing the derangement of oxidant/antioxidant balance at the brain level in elderly rats and in experimental ischemia-reperfusion model [20,41,42]; FPP ® modulates oxidative DNA damage, protecting brain from oxidative damage in hypertensive rats and reducing genotoxic effect of H2O2 [43], and protecting the body from the aging-related diseases [44][45][46][47], including neurodegenerative diseases [47][48][49]. However, a clear in vivo action of FPP ® on the molecular signature of aging, such as telomerase activity and telomeres length has not been provided yet. ...
... Previous reports have shown that FPP ® is able to modulate characteristic phenomena of elderly people, such as the pro-inflammatory profile [31] and the oxidative damage [44,45]. Moreover, FPP ® supplementation induced several beneficial effects in patients with neurodegenerative diseases and Electromagnetic Field Intolerance Syndrome [21,43,47,48]. Supporting these reports we have shown that plasmatic telomerase concentration and the telomeres length in bone marrow and ovarian germ cells, are significantly increased by the daily FPP ® administration. ...
Article
Full-text available
In recent decades much attention has been paid to how dietary antioxidants may positively affect the human health, including the beneficial effects of fermented foods and beverages. Fermented Papaya Preparation (FPP®) has been shown to represent a valuable approach to obtain systemic antioxidants effect. In this study, we wanted to verify whether FPP® had a clear and scientifically supported in vivo anti-aging effect together with the induction of a systemic antioxidant reaction. To this purpose we daily treated a mouse model suitable for aging studies (C57BL/6J) with FPP®-supplemented water from either the 6th weeks (early treatment) or the 51th weeks (late treatment) of age as compared to mice receiving only tap water. After 10 months of FPP® treatment, we evaluated the telomerase activity, antioxidants and Reactive Oxygen Species ROS plasmatic levels and the telomeres length in the bone marrow and ovaries in both mice groups. The results showed that the daily FPP® assumption induced increase in telomeres length in bone marrow and ovary, together with an increase in the plasmatic levels of telomerase activity, and antioxidant levels, with a decrease of ROS. Early treatment resulted to be more effective, suggesting a potential key role of FPP® in preventing the age-related molecular damages.
... Overweight condition as well as overt obesity is one the most expanding medical health threat worldwide [1]. Such excess weight, at different degree, is frequently associated to a number of comorbidities represented by metabolic syndrome, insulin resistance, diabetes, cardiovascular diseases and atherosclerosis and other health disorders linked to chronic inflammatory process [2][3][4]. As a matter of fact, overweight and obesity show the typical feature of low-grade inflammation where the fat tissue, and namely, visceral adipose tissue (VAT), being the largest endocrine gland in the body, plays the main role in releasing a wide array of inflammatory mediators (cytokines, hormones, acute-phase proteins, chemokimes, Retinol-binding protein 4, growth factors) [5,6]. ...
Article
Full-text available
We confirmed that either mild over-weight and normal-weight obese subjects harbor a relentless low-grade inflammation which may pave the way to silent progression of cardiovascular and metabolic diseases. A nattokinasemeriva formulation proved to yield a therapeutic and likely preventative efficacy for its robust anti-inflammatory and microcirculatory properties
Chapter
Energy restriction (ER) or calorie restriction (CR) refers to adequate intake of essential nutrients while being accompanied by energy restriction. Aging is known to be a risk factor for neurodegeneration, vascular disease and heart failure, and increased general inflammation that leads to type 2 diabetes by aging mechanisms. Energy restriction has been proven to prevent age-related chronic diseases, cancers, and increase average and maximum lifespan, and protect against neurodegenerative diseases, diabetes, hypertension, and vascular disease.The effects of CR on lifespan may be caused by various mechanisms, including changes and enhancements in metabolism energy, oxidative stress, insulin sensitivity, and structural changes in the sympathetic and neuroendocrine systems.CR with mechanisms such as apoptosis reduces damage to stem cells and protects them, maintains balance and tissue size and homeostasis, and prevents the occurrence of diseases related to aging.Energy restriction is beneficial to promote mitochondrial turnover by increasing the biogenesis of new mitochondria and the destruction of damaged mitochondria that generate superoxide radicals. Mitochondrial biogenesis reduces the production of ROS (reactive oxygen species), and in addition to preventing the loss of mitochondrial function in muscles, protecting fat tissue and affecting the energy metabolism of lipids on carbohydrates, glucose production, and glycolysis.We also elucidated the characteristics of the senescence-associated secretory phenotype (SASP) by which senescent cells provoke self-immune antagonism. The SASP response expresses instructions at the transcriptional level. Post-translational modifications activate a number of SASP genes, especially interleukins.Agingomics studies RNAs groups together that affect transcription and transcription can classify aging. Age-related gene expression is used to classify aging biomarkers.Cellular fitness evolves as a final result of entropy through chromosome organization, transcriptional regulation, and nuclear export/import in the nucleus to protein translation, autophagy recycling of organelles, cytoskeleton suspension, and subsequently extracellular matrix suspension and extracellular signaling.In addition, we mentioned the studies on the antioxidant and anti-aging properties of some plants, which were confirmed by Professor Marotta and his colleagues, and the effect of functional foods by Professor Barbagallo et al and the Mediterranean diet in the use of foods with oily polyphenols, as well as ketone therapy, by some other scientists and we described their effect on the cell with the molecular mechanisms of pathways such as NADH and P450 cytochrome enzymes.KeywordsER (energy restriction)CR (calorie restriction)AgeingOxidative stressSASP (senescence-associated secretory phenotype)AgingomicsFunctional foodsP450 cytochrome
Article
Scope: Reactive oxygen species production by innate immune cells plays a central role in host defense against invading pathogens at wound-site. A weakened hos-defense results in persistent infection leading to wound chronicity. Fermented Papaya Preparation (FPP), a complex sugar matrix, bolstered respiratory burst activity and improved wound healing outcomes in chronic wound patients. The objective of the current study was to identify underlying molecular factor/s responsible for augmenting macrophage host defense mechanisms following FPP supplementation. Methods and results: In depth LC-MS/MS analysis of cells supplemented with FPP led to identification of myo-inositol as a key determinant of FPP activity towards improving macrophage function. Myo-inositol, in quantities that is present in FPP, significantly improved macrophage respiratory burst and phagocytosis via de novo synthesis pathway of ISYNA1. Additionally, myo-inositol transporters, HMIT and SMIT1, played a significant role in such activity. Blocking these pathways using siRNA attenuated FPP-induced improved macrophage host defense activities. FPP supplementation emerges as a novel approach to increase intracellular myo-inositol levels. Such supplementation also modified wound microenvironment in chronic wound patients to augment myo-inositol levels in wound fluid. Conclusion: These observations indicate that myo-inositol in FPP influences multiple aspects of macrophage function critical for host defense against invading pathogens. This article is protected by copyright. All rights reserved.
Article
Full-text available
More that half-century of skeletal muscle research is continuing at the Padua University (Italy) under the auspices of the Interdepartmental Research Centre of Myology (CIR-Myo), the European Journal of Translational Myology (EJTM) and recently also with the support of the A&C M-C Foundation for Translational Myology, Padova, Italy. The Volume 30 (1), 2020 of EJTM opens with the collection of abstracts of the conference “2020 Padua Muscle Days: Mobility Medicine 30 years of Translational Research”, an International Conference, that will be held March 18-21, 2020 in Euganei Hills and Padova (Italy). These abstracts are excellent examples of translational research and of the multidimensional approaches that are needed to classify and manage in both the acute and chronic phases Mobility Medicine diseases that span from neurologic, metabolic, traumatic syndromes to the biological process of aging. Their excellent contents grant the high scientific profiles of researchers and clinicians who are eager to present their results at the PaduaMuscleDays. A series of EJTM Communications will add to this preliminary evidence. Here you can find the Announcement details.
Article
Full-text available
Background . Metabolic and genetic factors induce plasminogen activator inhibitor type-1 (PAI-1) overexpression; higher PAI-1 levels decrease fibrinolysis and promote atherothrombosis. Aim . To assess PAI-1 antigen levels among subjects with type 2 diabetes mellitus (T2DM) plus Metabolic Syndrome (MetS) before clinical manifestations of atherothrombosis and the contribution of metabolic factors and 4G/5G polymorphism of PAI-1 gene on the variability of PAI-1. Methods . We conducted an observational, cross-sectional assay in a hospital in Mexico City from May 2010 to September 2011. MetS was defined by the International Diabetes Federation criteria. PAI-1 levels and 4G/5G polymorphism were determined by ELISA and PCR-RFLP analysis. Results . We enrolled 215 subjects with T2DM plus MetS and 307 controls. Subjects with T2DM plus MetS had higher PAI-1 levels than the reference group (58.4 ± 21 versus 49.9 ± 16 ng/mL, p=0.026 ). A model with components of MetS explained only 12% of variability on PAI-1 levels ( R2 = 0.12; p=0.001 ), with β=0.18 ( p=0.03 ) for hypertension, β=-0.16 ( p=0.05 ) for NL HDL-c, and β=0.15 ( p=0.05 ) for NL triglycerides. Conclusion . Subjects with T2DM plus MetS have elevated PAI-1 levels before clinical manifestations of atherothrombotic disease. Metabolic factors have a more important contribution than 4G/5G polymorphism on PAI-1 plasma variability.
Article
Full-text available
Significance statement: We provide evidence that extracellular PPIA is a mediator of the neuroinflammatory reaction in ALS and is toxic for motor neurons. Supporting this, a specific extracellular PPIA inhibitor reduced neuroinflammation, rescued motor neurons and extended survival in the SOD1(G93A) mouse model of familial ALS. Our findings suggest selective pharmacological inhibition of extracellular PPIA as a novel therapeutic strategy not only for SOD1-linked, but possibly also for sporadic ALS. This approach aims at tackling the neuroinflammatory reaction that is a major hallmark of ALS. However, given the complexity of the disease, a combination of therapeutic approaches may be necessary.
Article
Full-text available
Background Vascular disease in diabetes is initiated by monocyte adhesion to vascular endothelium, transmigration and formation of foam cells. Increasing clinical evidence supports a role for the secretory protein, cyclophilin A in diabetic vascular disease. The means by which cyclophilin A contributes to vascular lesion development in diabetes is however largely unknown. Methods In this study we investigated using THP1 cells and human monocytes whether cyclophilin A under hyperglycemic conditions, functions in the inflammatory cascade as a chemoattractant and increases lipid uptake by formation of foam cells invitro. We developed an invitro model of monocytes cultured in 20 mm glucose (high glucose) equivalent to 360 mg/dL of plasma glucose levels. These monocytes were then differentiated into macrophages using PMA and subsequently transformed to lipid laden foam cells using oxidized low density lipoproteins in the presence and absence of cyclophilin A. This cellular model was used to study monocyte to macrophage differentiation, transmigration and foam cell formation. A similar cellular model using siRNA mediated transient elimination of the cyclophilin A gene as well as chemical inhibitors were used to further confirm the role of cyclophilin A in the differentiation and foam cell formation process. ResultsCyclophilin A effectively increased migration of high glucose treated monocytes to the endothelial cell monolayer (p < 0.0001). In the presence of cyclophilin A, differentiated macrophages, when treated with oxLDL had a 36 percent increase in intracellular lipid accumulation (p = 0.01) when compared to cells treated with oxLDL alone. An increased flux of reactive oxygen species was also observed (p = 0.01). Inflammatory cytokines such as TNF-α, MCP-1 and cyclophilin A were significantly increased. Silencing cyclophilin A in THP-1 cells and human monocytes using siRNA or chemical inhibitor, TMN355 resulted in decrease in lipid uptake by 65–75% even after exposure to oxidized LDL. The expression of scavenger receptors expressed during differentiation process, CD36 and LOX-1 were decreased (p < 0.0001). Levels of extracellular cyclophilin A and other inflammatory cytokines such as TNF-α and MCP-1also significantly reduced. Conclusions Taken together, we describe here a possible cellular basis by which cyclophilin A may accelerate atherogenesis in diabetes mellitus.
Article
Full-text available
Background/objectives: Evidence shows that tocotrienols potentially reverse various chronic disease progressions caused by the metabolic syndrome. We aimed to investigate the acute effects of a single-dose supplementation of gamma and delta tocotrienols (γδ-T3, 1:4 ratio) compared with those in placebo on the insulinemic, anti-inflammatory and anti-thrombogenic responses in metabolic syndrome subjects. Subjects/methods: Thirty metabolic syndrome subjects (15 men and 15 women) were recruited to a randomized, double-blinded and crossover study. The subjects were administered a single dose of 200 mg or 400 mg γδ-T3 emulsions or placebo incorporated into a glass of strawberry-flavored milkshake, consumed together with a high-fat muffin. Blood samples were collected at 0, 5, 15, 30, 60, 90, 120, 180, 240, 300 and 360 min after meal intake. Results: Plasma vitamin E levels reflected the absorption of γδ-T3 after treatments. Postprandial changes in serum C-peptide, serum insulin, plasma glucose, triacylglycerol, non-esterified fatty acid and adiponectin did not differ between treatments, with women displaying delayed increase in the aforementioned markers. No significant difference between treatments was observed for plasma cytokines (interleukin-1 beta, interleukin-6 and tumor necrosis factor alpha) and thrombogenic markers (plasminogen activator inhibitor type 1 and D-dimer). Conclusions: Supplementation of a single dose of γδ-T3 did not change the insulinemic, anti-inflammatory and anti-thrombogenic responses in metabolic syndrome subjects.European Journal of Clinical Nutrition advance online publication, 19 October 2016; doi:10.1038/ejcn.2016.200.
Article
Brain-derived neurotrophic factor (BDNF) plays pivotal roles in neuronal function. The cleaved – mature – form of BDNF (mBDNF), predominantly expressed in adult brains, critically determines its effects. However, insufficient proteolytic processing under pathology may lead to the precursor form of BDNF (proBDNF) and thereby increased neuronal apoptosis and synaptic weakening. Previous findings in our lab showed that cognitive stimulation (CS) delayed memory decline in Tg2576 mouse model of Alzheimer's disease (AD), an effect that was tightly associated with augmented levels of mBDNF. In view of this association, the present study explored whether altered cleavage of BDNF could be involved in AD-related traits triggered by excessive amyloid-β (Aβ) pathology and whether this process could be therapeutically targeted. Aβ pathology, both in AD patient samples and experimental models, triggered the upregulation of plasminogen-activator inhibitor-1 (PAI-1) via JNK/c-Jun. This led to inhibition of plasmin-regulated conversion of mBDNF. Pharmacological inhibition of PAI-1 with PAI-039 sufficiently reverted Aβ-induced tau hyperphosphorylation and neurotoxicity. Chronic treatment of 15 old-month Tg2576 mice with oral administration of PAI-039 resulted in improved BDNF maturation and cognitive function without inducing significant changes in amyloid burden. In conclusion, upregulation of PAI-1 may be a critical mechanism underlying insufficient neurotrophic support and increased neurodegeneration associated with AD. Thus, targeting BDNF maturation through pharmacological inhibition of PAI-1 might become a potential treatment for AD.
Article
Background: Oxidized low-density lipoprotein (ox-LDL) is the most common inflammatory factor that mediates the activation and apoptosis of macrophages. Cyclophilin A (CyPA) is expressed following oxidative stress, hypoxia, and infection. However, the role of CyPA in the activation and apoptosis of macrophages is unclear. The aims of the study were to determine whether CyPA mediates the ox-LDL-induced activation and apoptosis in RAW264.7 cells and to analyze potential mechanisms. Methods and results: Through Western blot and ELISA test, the expression of CyPA induced by ox-LDL is time-dependent in RAW264.7 cells. Gene silencing of CyPA reduced the generation of lipid droplets in the cytoplasm and downregulated the expression of the surface markers of macrophage activation, namely, CD80, CD86, and major histocompatibility complex class 2 antigen. Cell apoptosis is significantly decreased and the level of anti-apoptosis protein bcl-2 is increased in CyPA silent cells compared with the control group. Finally, autophagy-related protein LC3-II/LC3-I ratio level significantly decreased in CyPA silent cells with less autophagosome formation while the blocked autophagy flux was recovered. The differences in the activation and apoptosis between CyPA silent cells and the control cells were inhibited by pre-treatment with class III PI 3-kinase inhibitor 3-MA. Conclusions: These results indicate that CyPA mediates the ox-LDL-induced activation and apoptosis in RAW264.7 cells by regulating autophagy.
Article
Background: Inverse associations have been observed between memory performance and blood concentrations of cadmium (Cd) and lead (Pb). Low antioxidant cell activity has also been linked to decline in memory due to aging. However, it has not yet been established whether the heavy metal-memory relationship is mediated by differences in antioxidant activity. Methods: We examined Cd and Pb levels, as well as oxidative stress parameters, in blood samples from 125 older adults (age range 50-82years). The Counting Span Test (CST) was used to evaluate working memory capacity (WMC). The Monte Carlo Method for Assessing Mediation (MCMAM) was used to analyze the mediation role of antioxidant activity in the heavy metals-memory association. Results: High blood Cd (BCd) concentration alone, and in combination with elevated blood Pb (BPb) concentration, was associated with poor WMC (p≤0.001) and low enzymatic antioxidant defenses (p≥0.006). The variance in WMC accounted for by BCd or by BCd combine with BPb was 20.6% and 18.6%, respectively. The MCMAM revealed that the influence of BCd and BPb concentrations on WMC was mediated by low antioxidant capacity (confidence interval - CI: 0.072 to -0.064 for BCd; CI: -0.062 to -0.045 for BPb). Conclusion: These findings showed Pb and Cd blood concentration in older adults, even at levels below the current recommended threshold, was negatively associated with WMC and that this relationship may be partly mediated by low antioxidant defenses. Knowledge on the environmental factors that negatively influence brain and cognition during aging can help inform public policy strategies to prevent and control the adverse effects of environmental contaminant exposure during aging.
Article
Background: Our previous clinical indicated that urinary cyclophilin A was a a good marker for diabetic nephropathy. Methods: We used animal and cell models of diabetic nephropathy to examine the role of cyclophilin A in disease progression. Results: Significantly increased urinary cyclophilin A could be detected in db/db at the 8th week. Linagliptin (3mg/kg/day and 15mg/kg/day) could suppress urinary 8-hydroxy-2' -deoxyguanosine at the 8th and 16th week but only the high dose Linagliption could suppress cyclophilin A at the 8th week. Compared to 8-hydroxy-2' -deoxyguanosine, cyclophilin A was a stronger, earlier, and more sensitive marker. Immunohistochemical staining for cyclophilin A was also positive for db/db. In cell studies, oxidative stress and hyperglycemia could stimulate MES-13 and HK-2 cells to secrete cyclophilin A. Hyperglycemia stimulated HK-2 cells to secrete TGFβ1, which caused secretion of cyclophilin A. The secreted cyclophilin A further stimulated CD 147 to move outward from cytosol onto cell membrane in confocal microscopy, which was associated with the p38 MAPK pathway in the downstream. Conclusions: Secreted cyclophilin A may play an important role in diabetic nephropathy in the mouse model and is associated with TGFβ1, CD 147, and the p38 MAPK pathway.