Skip to main content
PLOS One logoLink to PLOS One
. 2021 Feb 5;16(2):e0246289. doi: 10.1371/journal.pone.0246289

Impact of gestational low-protein intake on embryonic kidney microRNA expression and in nephron progenitor cells of the male fetus

Letícia de Barros Sene 1, Wellerson Rodrigo Scarano 2, Adriana Zapparoli 1, José Antônio Rocha Gontijo 1, Patrícia Aline Boer 1,*
Editor: Emmanuel A Burdmann3
PMCID: PMC7864410  PMID: 33544723

Abstract

Background

Here, we have demonstrated that gestational low-protein (LP) intake offspring present lower birth weight, reduced nephron numbers, renal salt excretion, arterial hypertension, and renal failure development compared to regular protein (NP) intake rats in adulthood. We evaluated the expression of various miRNAs and predicted target genes in the kidney in gestational 17-days LP (DG-17) fetal metanephros to identify molecular pathways involved in the proliferation and differentiation of renal embryonic or fetal cells.

Methods

Pregnant Wistar rats were classified into two groups based on protein supply during pregnancy: NP (regular protein diet, 17%) or LP diet (6%). Renal miRNA sequencing (miRNA-Seq) performed on the MiSeq platform, RT-qPCR of predicted target genes, immunohistochemistry, and morphological analysis of 17-DG NP and LP offspring were performed using previously described methods.

Results

A total of 44 miRNAs, of which 19 were up and 25 downregulated, were identified in 17-DG LP fetuses compared to age-matched NP offspring. We selected 7 miRNAs involved in proliferation, differentiation, and cellular apoptosis. Our findings revealed reduced cell number and Six-2 and c-Myc immunoreactivity in metanephros cap (CM) and ureter bud (UB) in 17-DG LP fetuses. Ki-67 immunoreactivity in CM was 48% lesser in LP compared to age-matched NP fetuses. Conversely, in LP CM and UB, β-catenin was 154%, and 85% increased, respectively. Furthermore, mTOR immunoreactivity was higher in LP CM (139%) and UB (104%) compared to that in NP offspring. TGFβ-1 positive cells in the UB increased by approximately 30% in the LP offspring. Moreover, ZEB1 metanephros-stained cells increased by 30% in the LP offspring. ZEB2 immunofluorescence, although present in the entire metanephros, was similar in both experimental groups.

Conclusions

Maternal protein restriction changes the expression of miRNAs, mRNAs, and proteins involved in proliferation, differentiation, and apoptosis during renal development. Renal ontogenic dysfunction, caused by maternal protein restriction, promotes reduced reciprocal interaction between CM and UB; consequently, a programmed and expressive decrease in nephron number occurs in the fetus.

Introduction

The lack of nutrients may result in signaling changes in pivotal pathways during various stages of fetal development, which may cause irreversible organ and system disorders in adulthood [1]. Fetal programming refers to any insult during development, which causes long-term effects on an organism’s structure or function [2]. Disruptions in fetal programming result in low birth weight, fewer nephrons, and increased risk of cardiovascular and renal disorders in adulthood [36]. Studies by other authors and us have demonstrated lower birth weight, 28% fewer nephrons, reduced renal salt excretion, chronic renal failure, and arterial hypertension in gestational low-protein (LP) intake compared to standard (NP) protein intake offspring in adulthood [37].

Nephrogenesis involves tight control of gene expression, protein synthesis, and tissue remodeling. Studies have demonstrated that nephron numbers are determined by the interactions between ureter bud (UB) and metanephros mesenchyme (MM) progenitor cells [810]. Signals from MM induce UB stimulated growth and branching of the tubule system. In turn, MM proliferation and differentiation, constituting a mesenchymal cap (CM), is mediated by UB ends [11].

There has been serious interest in the role of epigenetic changes, concerning the long-term effects of prenatal stress, on fetal development [12]. MicroRNAs (miRNAs) are genome-encoded small non-coding RNAs of approximately 22 nucleotides in length and play an essential role in the post-transcriptional regulation of target gene expression [1316]. miRNAs control gene expression post-transcriptionally by regulating mRNA translation or stability in the cytoplasm [17, 18]. Functional studies indicate that miRNAs are involved in critical biological processes during development and in cell physiology [13, 16]. Changes in their expression have been observed in several pathologies [16, 19].

Thus, miRNAs characterization has helped understand gene regulation and cellular proliferation, differentiation, and apoptosis and explain pathophysiology disorders, including kidney disorders [2022]. Studies have reported that during kidney ontogeny miRNAs are indispensable for nephron development [2326]. Moreover, underexpression of some miRNAs in MM progenitor cells reduces cell proliferation, resulting in early differentiation, and consequently, decreased number of nephrons [27, 28]. This phenomenon is characterized by increased apoptosis and high Bim expression in progenitor cells [27]. Thus, miRNAs modulate the balance between apoptosis and proliferation these metanephric primary cells [29].

We hypothesized that unknown epigenetic changes and miRNA expression profiling are associated with kidney developmental disorder in maternal protein-restricted offspring. Thus, we aimed to evaluate patterns of miRNA and predicted gene expression in the fetal kidney at 17 days of gestational (17-DG) protein-restricted male offspring to identify molecular pathways and disorders involved in renal cell proliferation and differentiation during kidney development.

Material and methodology

Animal and diets

The experiments were conducted as described in detail previously [5, 6] on age-matched female and male rats of sibling-mated Wistar HanUnib rats (250–300 g) originated from a breeding stock supplied by CEMIB/ UNICAMP, Campinas, SP, Brazil. The environment and housing presented the right conditions for managing their health and well-being during the experimental procedure. Immediately after weaning at three weeks of age, animals were maintained under controlled temperature (25°C) and lighting conditions (07:00–19:00h) with free access to tap water and standard laboratory rodent chow (Purina Nuvital, Curitiba, PR, Brazil: Na+ content: 135 ± 3μEq/g; K+ content: 293 ± 5μEq/g), for 12 weeks before breeding. The Institutional Ethics Committee on Animal Use at São Paulo State University (#446-CEUA/UNESP) approved the experimental protocol, and the general guidelines established by the Brazilian College of Animal Experimentation were followed throughout the investigation. It was designated day 1 of pregnancy as the day in which the vaginal smear exhibited sperm. Then, dams were maintained ad libitum throughout the entire pregnancy on an isocaloric rodent laboratory chow with either standard protein content [NP, n = 36] (17% protein) or low protein content [LP, n = 51] (6% protein). The NP and LP maternal food consumption were determined daily (subsequently normalized for body weight), and the bodyweight of dams was recorded weekly in both groups. On 17 days of gestation (17-DG), the dams were anesthetized by ketamine (75mg/kg) and xylazine (10mg/kg), and the uterus was exposed. The fetuses were removed and immediately euthanized by decapitation. The fetuses were weighed and, the tail and limbs were collected for sexing. The metanephros was collected for Next Generation Sequencing (NGS), RT-qPCR, and immunohistochemistry analyses.

Sexing determination

The present study was performed only in male 17-DG offspring, and the sexing was determined by Sry conventional PCR (Polymerase Chain Reaction) sequence analysis. The DNA was extracted by enzymatic lysis with proteinase K and Phenol-Chloroform. For reaction, the Master Mix Colorless—Promega was used, with the manufacturer’s cycling conditions. The Integrated DNA Technologies (IDT) synthesized the primer following sequences bellow:

  • Forward: 5’-TACAGCCTGAGGACATATTA-3’

  • Reverse: 5’-GCACTTTAACCCTTCGATTAG-3’.

Total RNA extraction

RNA was extracted from NP (n = 4) and LP (n = 4) whole kidneys using Trizol reagent (Invitrogen), according to the instructions specified by the manufacturer. Total RNA quantity was determined by the absorbance at 260 nm using a nanoVue spectrophotometer (GE Healthcare, USA). RNA Integrity was ensured by obtaining an RNA Integrity Number—RIN > 8 with Agilent 2100 Bioanalyzer (Agilent Technologies, Germany) [30].

miRNA-Seq and data analysis

Sequencing was performed on the MiSeq platform (Illumina). The protocol followed the manufacturer’s instructions available in (http://www.illumina.com/documents//products/datasheets/datasheet_truseq_sample_prep_kits.pdf). Briefly, the sequencing includes library construction, and this was used 1μg total RNA. In this step, the adapters are connected, the 3 ’and 5’. After ligation of adapters, a reverse transcription reaction was performed to create cDNA. It was then amplified by a standard PCR reaction, which uses primers containing a sequence index for sample identification—this cDNA library, subjected to agarose gel electrophoresis for miRNA isolation. After quantitation, the library concentration was normalized to 2 nM using 10 nM Tris-HCl, pH 8.5, and transcriptome sequencing was performed by MiSeq Reagent Kit v2 (50 cycles).

Data analysis was performed in collaboration with Tao Chen, Ph.D. from the Division of Genetic and Molecular Toxicological, National Center for Toxicological Research, Jefferson, AR, USA. The data from Next Generation Sequencing (NGS) of miRNAs were generated in FASTAQ format and imported into BaseSpace.com (Illumina, USA). The data quality was evaluated using the base calling CASAVA software developed by the manufacturer (Illumina). The analyzes were done by BaseSpace miRNA Analysis (from the University of Torino, Canada) and the sequence mapping of different miRNAs by Small RNA (Illumina, USA) for rat genome. The differentially expressed miRNA study was analyzed using Ingenuity Pathway Analysis software (Ingenuity, USA).

miRNA expression validation

Four male offspring from different litters were used in each group for the miRNA (miR-127-3p, -144-3p, -298-5p, let-7a-5p, -181a-5p, -181c-3p, and -199a-5p) expression analysis. Briefly, 450 ng RNA was reverse transcribed, without pre-amplification, using the TaqMan® MicroRNA Reverse Transcription Kit, according to the manufacturer’s guidelines. Complementary DNA (cDNA) was amplified using TaqMan MicroRNA Assays (Life Technologies, USA) with TaqMan® Universal PCR Master Mix, No AmpErase® UNG (2x) on StepOnePlusTM Real-Time PCR System (Applied BiosystemsTM), according to the manufacturer’s instructions. Data analysis was performed using relative gene expression evaluated using the comparative quantification method (Pfaffl, 2001). Based on stability analysis, the U6 snRNA and U87 scaRNA was used as a reference gene. All relative quantifications were evaluated using the DataAssist software, v 3.0, using the ΔΔCT method. miRNA data have been generated following the MIQE guidelines [31].

RT-qPCR of predicted target genes

For the cDNA synthesis, the High Capacity cDNA reverse transcription kit (Life Technologies, USA) was used. The RT-qPCR reactions for Bax, Bim, Caspase-3, Collagen 1, GDNF, PCNA, TGFβ-1, Bcl-2, Bcl-6, c-Myc, c-ret, cyclin A, Map2k2, PRDM1, Six-2, Ki-67, MTOR, β-catenin, ZEB1, ZEB2, NOTCH1, and IGF1 gene was performed by SYBR Green Master Mix (Life Technologies, USA) provided by IDT® Integrated DNA Technologies (Table 1). The reactions were done in a total volume of 20 μL using 2 μL of cDNA (diluted 1:30), 10μL SYBER Green Master Mix (Life Technologies, USA), and 4 μL of each specific primer (5 nM). Amplification and detection were performed using the StepOnePlusTM Real-Time PCR System (Applied BiosystemsTM). Ct values were converted to relative expression values using the ΔΔCt method with offspring metanephros data normalized with GAPDH as a reference gene [32].

Table 1. Dilution of antibodies used in immunohistochemistry.

GENE FORWARD SEQUENCE REVERSE SEQUENCE
Six-2 5’-GCCGAGGCCAAGGAAAGGGAG-3’ 5’-GAGTGGTCTGGCGTCCCCGA-3’
c-myc 5’-AGCGTCCGAGTGCATCGACC-3’ 5’-ACGTTCCAAGACGTTGTGTG-3’
c-ret 5’-GTTTCCCTGATGAGAAGAAGTG-3’ 5’-GTGGACAGCAGGACAGATA-3’
Bcl-2 5’-ACGGTGGTGGAGGAACTCTT-3’ 5’-GTCATCCACAGAGCGATGTTG-3’
Col-1 5`-ACCTGTGTGTTCCCCACT-3` 5`-CTTCTCCTTGGGGTTTGGGC-3`
TGFβ-1 5`-GGACTCTCCACCTGCAAGAC-3` 5`-GACTGGCGAGCCTTAGTTTG-3`
Ciclin A 5’-GCC TTCACCATTCATGTGGAT-3’ 5’-TTGCTGCGGGTAAAGAGACAG-3’
Bax 5’-TTCAGTGAGACAGGAGCTGG-3’ 5’-GCATCTTCCTTGCCTGTGAT-3’
Bim 5’-CAATGAGACTTACACGAGGAGG-3’ 5’CCAGACCAGACGGAAGATGAA-3’
Casp 3 5’-ACGGGACTTGGAAAGCATC-3’ 5’-TAAGGAAGCCTGGAGCACAG-3’
GDNF 5’-CAGAGGGAAAGGTCGCAGAG-3’ 5’-TCGTAGCCCAAACCCAAGTC-3’
Ki-67 5’- GTCTCTTGGCACTCACAG-3’ 5’-TGGTGGAGTTACTCCAGGAGAC-3’
mTOR 5`-ACGCCTGCCATACTTGAGTC-3` 5`-TGGATCTCCAGCTCTCCGAA-3`
VEGF 5`-CGGGCCTCTGAAACCATGAA-3` 5`-GCTTTCTGCTCCCCTTCTGT-3`
GAPDH 5`-CAACTCCCTCAAGATTGTCAGCAA-3` 5`-GGCATGGACTGTGGTCATGA-3`
Β-catenin 5’-AGTCCTTTATGAGTGGGAGCAA-3’ 5’- GTTTCAGCATCTGTGACGGTTC-3’
Map2K2 5’- ACCGGCACTCACTATCAACC-3’ 5’-TTGAGCTCACCGACCTTAGC-3’
Bcl-6 5’-CCAACCTGAAGACCCACACTC-3’ 5’-GCGCAGATGGCTCTTCAGAGTC-3’
PCNA 5’-TTTGAGGCACGCCTGATCC-3’ 5’-GGAGACGTGAGACGAGTCCAT-3′
PRDM1 5’-CTTGTGTGGTATTGTCGGGAC-3’ 5’-CACGCTGTACTCTCTCTTGG-3’
NOTCH1 5`-ACTGCCCTCTGCCCTATACA-3` 5`-GACACGGGCTTTTCACACAC-3`
IGF1 5`-AAGCCTACAAAGTCAGCTCG-3` 5`-GGTCTTGTTTCCTGCACTTC-3`
ZEB1 5’-CATTTGATTGAGCACATGCG-3’ 5’-AGCGGTGATTCATGTGTTGAG-3’
ZEB2 5’-CCCTTCTGCGACATAAATACGA-3’ 5’-TGTGATTCATGTGCTGCGAGT-3’

Immunohistochemistry

The fetus (n = 4 per group) was removed and immediately fixed in 4% paraformaldehyde (0.1 M phosphate, pH 7.4). The materials were dehydrated, diaphanized, and included in paraplast, and the blocks were cut into 5-μm-thickness sections. Histological sections were deparaffinized and processed for immunofluorescence and immunoperoxidase. The sections were incubated with a blocking solution (8% fetal bovine serum, 2.5% bovine albumin, and 2% skimmed milk powder in PBS). Subsequently, set with the primary antibody (anti-Six-2) diluted in PBS containing 1% skim milk overnight under refrigeration. After washing with PBS, the sections were incubated with a specific secondary antibody, conjugated to the Alexa 488 fluorophore, diluted in the same buffer, containing 1% milk for 2 hours at room temperature. After successive washes with PBS, the slides were mounted with coverslips using the Vectashield fluorescent assembly medium (Vector Laboratories, Inc. Burlingame). The fluorescence in the specimen was detected by laser confocal microscopy. The images were obtained using the Focus Imagecorder Plus system. For the c-Myc, Ki-67, Bcl-2, TGFβ-1, β-catenin, ZEB1, ZEB2, Caspase 3 cleaved, cyclin A and WT1 proteins, immunohistochemistry was performed. The slides were hydrated, and after being washed in PBS pH 7.2 for 5 minutes, the antigenic recovery was made with citrate buffer pH 6.0 for 25 minutes in the pressure cooker. The slides were washed in PBS. Subsequently, endogenous peroxidase blockade with hydrogen peroxide and methanol was performed for 10 minutes in the dark. The sections were rewashed in PBS. Blocking of non-specific binding was then followed, and the slides were incubated with a blocking solution (5% skimmed milk powder, in PBS) for 1 hour. The sections were incubated with the primary antibody (Table 2), diluted in 1% BSA overnight in the refrigerator. After washing with PBS, the sections were exposed to the specific secondary antibody for 2 hours at room temperature. The slides were washed with PBS. The slices were revealed with DAB (3,3’- diaminobenzidine tetrahydrochloride, Sigma—Aldrich CO®, USA). After successive washing in running water, the slides were counterstained with hematoxylin, dehydrated, and mounted with a coverslip, using Entellan®. The images were obtained using the photomicroscope (Olympus BX51) or a Zeiss LSM 780-NLO confocal on an Axio Observer Z.1 microscope (Carl Zeiss AG, Germany) from the National Institute of Science and Technology on Photonics Applied to Cell Biology (INFABIC) at the State University of Campinas.

Table 2. Sequence of the primers used for RT-qPCR, designed by the company IDT.

Antibody Dilution Company
Anti-Six-2 (11562-1-AP) 1:50 Proteintech
Anti-c-Myc (NBP1-19671) 1:150 Novus Biologicals
Anti-Ki-67 (ab16667) 1:100 Abcam
Anti-Bcl-2 (ab7973) 1:100 Abcam
Anti-TGFβ-1 (sc-146) 1:50 Santa Cruz
Anti-Β-catenina (ab32572) 1:500 Abcam
Anti-ZEB1 (sc-10572) 1:50 Santa Cruz
Anti-ZEB2 (sc-48789) 1:50 Santa Cruz
Anti-VEGF (NB100-664) 1:50 Novus Biologicals
Anti-Caspase-3 clivada (9664) 1:200 Cell Signaling
Anti-Ciclina A (sc-31085) 1:50 Santa Cruz
Anti-WT1 (sc-192) 1:50 Santa Cruz
Anti-mTOR (cs-7c10) 1:200 Cell Signaling

Morphology quantification

Paraffin 5 μm kidney sections were analyzed using CellSens Dimension software from a photomicroscope (Olympus BX51). The kidney slices were accessed to determine the nephrogenic area, CM and UB protein and cell number, hematoxylin-eosin stained in 17-DG LP fetus (n = 5) compared to age-matched NP offspring (n = 5) from different mothers. We quantified all CM and UB of each metanephros analyzed (4NP and 4LP from different mothers), and statistical analysis was performed by t-test, and the values were expressed as mean ± SD. The p≤0.05 was considered significant. GraphPad Prism v01 Software, Inc., USA, was used for statistical analysis and figure construction.

Statistical analysis

The t-test was used, and the values were expressed as mean ± standard deviation (SD). P≤0.05 was considered significant. GraphPad Prisma v. 01 software (GraphPad Software, Inc., USA) was used for statistical analysis and figure construction.

Results

Expression of miRNAs by miRNA-Seq

To understand the microRNA changes associated with maternal low-protein renal programming, we performed the expression of a global miRNA profiling analysis. It was identified 44 deregulated miRNAs (p ≤ 0.05), of which 19 and 25 miRNAs, respectively, were up-or down-regulated (Table 3). The top expressed miRNAs and their functions, pathways, and networks were identified using Ingenuity Software (Table 4).

Table 3. Lists of the deregulated miRNAs obtained by miRNA-Seq.

miRNAs up-regulated FC miRNAs down-regulated FC
83_ACCACCAACCGTTGACTGTACC_rno-mir-181a-2 1.55 69_ACAGTAGTCTGCACATTGGTT_rno-mir-199a 0.76
38_AACATTCAACGCTGTCGGTG_rno-mir-181a-2 2.08 rno-miR-136-3p 0.53
10_GGCAGAGGAGGGCTGTTCTTCC_rno-mir-298 1.44 rno-let-7g-5p 0.78
rno-miR-298-5p 1.47 rno-miR-144-3p 0.49
rno-miR-183-5p 1.46 13_AAGGGATTCTGATGTTGGTCACACTC_rno-mir-541 0.52
rno-miR-181d-5p 1.36 15_TCCCTGAGGAGCCCTTTGAGCCTGAAA_rno-mir-351-2 0.50
rno-miR-455-3p 1.93 56_TCGGATCCGTCTGAGCTTGGC_rno-mir-127 0.53
35_AACATTCATTGCTGTCGGTGGGA_rno-mir-181b-1 1.89 9_ATATAATACAACCTGCTAAGTGT_rno-mir-374 0.54
50_CAGTGCAATGATGAAAGGGC_rno-mir-130b 1.85 56_TCGGATCCGTCTGAGCTTGG_rno-mir-127 0.52
83_ACCACCAACCGTTGACTGTACCT_rno-mir-181a-2 1.81 rno-miR-320-3p 0.55
rno-miR-151-3p 1.31 rno-miR-376b-3p 0.56
rno-miR-181c-3p 1.36 16_AAACCGTTACCATTACTGAGTTT_rno-mir-451 0.56
69_TACAGCAGGCACAGACAGGCAGT_rno-mir-214 1.53 56_TCGGTCGATCGGTCGGTCGGTT_rno-mir-341 0.56
rno-miR-195-3p 1.58 16_AAACCGTTACCATTACTGAGTTTAGT_rno-mir-451 0.54
21_TACCCTGTAGATCCGAATTTGTGA_rno-mir-10a 1.2 12_GGATATCATCATATACTGTAAG_rno-mir-144 0.59
rno-miR-1298 1.52 60_TCAGTGCATCACAGAACTTTGTTT_rno-mir-148b 0.72
rno-miR-92b-3p 1.36 15_TCCCTGAGGAGCCCTTTGAGCCTGT_rno-mir-351-2 0.77
61_ACCACAGGGTAGAACCACGGAA_rno-mir-140 1.60 13_AAGGGATTCTGATGTTGGTCACAC_rno-mir-541 0.58
50_CAGTGCAATGATGAAAGGGCATA_rno-mir-130b 1.31 15_CTGAGAACTGAATTCCATGGGTT_rno-mir-146a 0.58
  9_GACCCTGGTCTGCACTCTGTCT_rno-mir-504 0.58
  rno-let-7b-5p 0.69
  rno-let-7f-5p 081
  14_TCCCTGAGACCCTTTAACCTG_rno-mir-125a 0.58
  rno-miR-410-3p 0.63
  rno-miR-541-5p 0.68

Table 4. Top canonical pathways affected by differentially expressed miRNAs in 17 DG LP metanephros.

17 DG Pathway analysis results (IPA) Number of miRNAs p-value/score
NP vs LP Top Molecular and Cellular Functions
Cellular Development 12 4.71E-02–4.31E-05
Cellular Growth and Proliferation 12 3.41E-02–1.60E-04
DNA Replication, Recombination, and Repair 04 3.12E-02–4.83E-04
Cell Cycle 04 3.49E-02–7.49E-04
Cellular Movement 06 3.49E-02–1.11E-03
Top Networks
Organismal Injury and Abnormalities, Reproductive System Disease, Cancer 34
Top Tox Lists
Renal Ischemia-Reperfusion Injury microRNA Biomarker Panel (Mouse)
Top 10 highly expressed miRNAs
miR-199a-5p; miR-136-3p
miR-181a-5p; miR-298-5p
miR-144-3p; miR-541-5p
miR-127-3p; miR-374b-5p
miR-183-5p; Let-7a-5p
4.31E-05

Validation of miRNA expression

In the LP group’s animals, Let-7a-5p, miR-181a-5p, miR-181c-3p were upregulated, while the miR-127-3p, miR-144-3p, and miR-199a-5p were downregulated relative to NP animals. The results do not show any difference in miR-298 expression, comparing both groups (Fig 1). Table 5 revealed the values obtained by miRNAs sequencing with the RT-qPCR validation data. Although significant miRNA expression difference was observed in LP relative to NP offspring, the fold change (FC) of the validated miRNAs was similar to both techniques.

Fig 1. Expression of miRNAs in the metanephros from the 17th day LP fetus compared to their expression level in the control group.

Fig 1

Reference genes U6 and U87, protein complexes composed of small nuclear RNAs (snRNAs), were used to normalize each miRNA expression. The authors established a cutoff point variation of 1.3 (upwards) or 0.65 (downwards) and data are expressed as fold change (mean ± SD, n = 4) concerning the control group. * p≤0.05: statistical significance versus NP.

Table 5. Comparison between the values obtained in the miRNA sequencing and the validation by RT-qPCR.

miRNA (17 DG) log FC Sequencing Fold Change (FC) p-value miRNA (17 DG) Fold Change (FC) log FC qPCR p-value
miR-127-3p -0.911189177 0.5317 0.01053498 miR-127-3p 0.6045 -0.7262 1.97E-08
miR-144-3p -1.024909088 0.4914 0.00754482 miR-144-3p 0.6014 -0.7335 0.0321508
miR-298-5p 0.555324736 1.4695 0.0083628 miR-298-5p 1.4317 0.5177 0.0648687
Let-7a-5p Let-7a-5p 1.8747 0.9067 0.0106146
miR-181a-5p 0.637181521 1.5553 0.00520696 miR-181a-5p 1.7645 0.8193 0.0354613
miR-181c-3p 0.40731742 1.3262 0.02187935 miR-181c-3p 1.6265 0.7018 0.0273168
miR-199a-5p -0.401388293 0.7571 0.00193047 miR-199a-5p 0.5086 -0.9755 4.551E-05

miRNA-gene targets

The expression genes of predicted targets of different miRNA such as Six-2, Bcl-2, PRDM1, cyclin A, PCNA, GDNF, Collagen 1, Caspase 3, and Bim in LP did not differ significantly from NP fetus. However, Bax, TGFβ-1 Bcl-6, c-ret, Map2k2, Ki-67, mTOR, β-catenin, ZEB1, ZEB2, and IGF1 gene expression were upregulated in the 17-DG LP group compared to age-matched controls. Conversely, c-Myc, and NOTHC1 were downregulated in maternal protein-restricted offspring (Fig 2).

Fig 2. Expression of mRNA estimated by SyBR green RT-qPCR of metanephros from the 17th day LP fetus.

Fig 2

The expression was normalized with GAPDH. The authors established a cutoff point variation of 1.3 (upwards) or 0.65 (downwards) and data are expressed as fold change (mean ± SD, n = 4) concerning the control group. * p≤0.05: statistical significance versus NP.

Fetus body mass and metanephros morphometry

The 17-DG LP body mass did not differ from the age-matched NP offspring. However, LP’s metanephros mesenchyme showed a 7.6% reduced area and a 29% reduction in the cortex thickness than the NP group (Fig 3).

Fig 3. Metanephros of the fetus with 17 DG and quantifications.

Fig 3

Comparing the HE stained micrography, we can observe the difference in the NP (A) and LP(B) metanephros size and nephrogenic cortex (NC) thickness. The differences between these parameters are statistically significant (C and D). **p<0.005; ***p<0.0001.

Immunohistochemistry

In the present study, the LP fetus showed a significant reduction (about 69%) of Six-2 cap fluorescence than NP offspring (Fig 4).

Fig 4. Immunofluorescence and immunoperoxidase for Six-2 and c-Myc in metanephros of 17DG fetus.

Fig 4

The Six-2 immunomarker cells in LP (A, a) was significantly reduced when compared to NP (B,b, C) in metanephros. Additionally, the Six-2 immunostained cells were significantly reduced in LP (E) caps (circled by black lines; F, G) when compared to NP (D). On the other hand, the c-Myc labeled area was reduced in the LP cap (K) but was the same in UB (L) when compared to NP (J). *p<0.005;**p<0.001; ***p<0.0001.

The Six-2 immunoperoxidase analysis demonstrated a reduced cell number (14%) in LP CM from associated with 28% reduced Six-2+ cells relative to the cap area compared to NP offspring (Fig 4). The present study also showed a significant percent reduction of c-Myc CM and UB immunostained cells (less 14%) in LP relative to NP offspring (Fig 4). Additionally, the percentage of Ki-67 labeled area in CM was 48% lesser in LP compared to NP fetus, while Bcl-2 and cleaved caspase-3 immunoreactivity were not different from both groups (Figs 5 and 6).

Fig 5. Immunoperoxidase for Ki-67 and Bcl-2 in metanephros of 17DG fetus.

Fig 5

The number of Ki-67+ cells was reduced in LP (B) when compared to NP (A) and was statistically significant in the caps (C). The Bcl-2 labeled area was the same in NP (D) and LP (E) in both UB and cap (F, G). ***p<0.0001.

Fig 6. Immunoperoxidase for cleaved caspase 3 in metanephros of 17DG fetus.

Fig 6

The immunostained cells were preferentially located in the ureteric epithelium. However, the quantity is not different in LP (B) than in NP (A).

The present study also showed a significant percent reduction of c-Myc CM and UB immunostained cells (less 14%) in LP relative to NP offspring (Fig 4). Additionally, the percentage of Ki-67 labeled area in CM was 48% lesser in LP compared to NP fetus, while Bcl-2 and cleaved caspase-3 immunoreactivity were not different from both groups (Figs 5 and 6). On the other hand, in LP, the CM and UB β-catenin labeled area were 154 and 85% raised, respectively, compared to that available in NP offspring (Fig 7).

Fig 7. Immunoperoxidase for b-catenin and mTor in metanephros of 17DG fetus.

Fig 7

In LP (B), the b-catenin labeled area was significantly raised in both CAP cells and UB epithelia (C, D) when compared to NP offspring (A). Also, mTor immunoreactivity occupied a more extensive area in LP (F) than in NP (E) offspring in analyzed structures (G, H). ***P<0.0001.

At the same time, mTOR immunoreactivity distribution also occupied a significantly more extensive area in LP CM (139%) and UB (104%) than in the NP fetus (Fig 7). In the LP offspring, the TGFβ-1 in UBs cells staining increased (about 30%), while in the CM, the immunostained cells were not different related to the NP group (Fig 8).

Fig 8. Immunoperoxidase for TGFβ-1, ZEB1, and ZEB2 in metanephros of 17DG fetus.

Fig 8

The area of TGFβ-1 immunoreactivity in LP (B) compared to NP offspring (A) was not different in the CAP (C) but was significantly enhanced in UB (D). ZEB1 was detected in the nuclei of CAP and other mesenchymal cells and, the LP (E) CAP occupied a more extensive area (G) than in NP (F) offspring. The ZEB2 labeled area was not different from both CAP (J) and UB (K). ***P<0.0001.

The ZEB1 metanephros-stained, located in the CM nuclei cells, enhanced 30% in LP compared to the NP fetus (Fig 8). Simultaneously, the ZEB2 immunofluorescence, although present in whole metanephros structures, was similar in both experimental groups (Fig 8). The current study, taking into account miRNA and mRNA expression and proteins immunostaining results present above may permit schedule representative pathway interactions to explain the experimental findings (Fig 9).

Fig 9. Deregulated miRNA-mRNA-protein pathways in metanephros of 17DG fetus from maternal restricted-protein intake.

Fig 9

Discussion

Knowledge about cellular and molecular mechanisms of nephrogenesis has increased [3337]. However, many regulatory factors and signaling pathways involved in renal ontogenesis remain unclear [38]. miRNAs play a crucial role in regulating gene expression during renal development [25, 3941]. To our knowledge, miRNA and mRNA expression analyses in maternal LP intake 17-DG male mesenchyme cells have not been performed.

We propose a novel molecular mechanism involved in inhibiting early nephrogenesis, resulting in a reduced number of nephrons. We used NGS to evaluate miRNA expression and found that 19 miRNAs were up- and 25 downregulated in 17-DG LP compared to NP metanephros. Among the top 10 deregulated miRNAs, we selected 7 miRNAs with biological targets involved in proliferation, differentiation, and cellular apoptosis. Both miRNA-Seq and TaqMan data analysis revealed consistent and specific changes in miRNA expression in LP animals relative to control NP age-matched animals.

The miR-181 family is composed of four highly conserved members, namely miR-181a, miR-181b, miR-181c, and miR-181d [42]. In neoplastic cells, miR-181a acts as a tumor suppressor, inhibiting cellular proliferation and migration and inducing cellular apoptosis [43]. This study revealed increased expression of miR-181a-5p in 17-DG LP relative to age-matched NP offspring. Although caspase mRNA expression was unaltered, a two-fold increase in Bax/Bcl-2 mRNA ratio in LP compared to NP offspring suggests increased apoptosis in the CM, indicating that apoptosis is regulated post-transcriptionally. Studies have shown that the BCL family promotes cytochrome release from the mitochondria and then inhibits the activation of Casp3, thereby inhibiting cellular apoptosis [44]. Li et al. used an acute lung injury model to reveal that overexpressed miR-181a is related to decreased Bcl-2 protein level; conversely, miR-181a inhibition increased Bcl-2 levels [45]. This study confirmed the results of Lv et al., who demonstrated that miR-181c regulates the expression of Six-2 expression and cell proliferation negatively, parallel to the loss of mesenchymal cells phenotype during kidney development in LP 17-DG offspring [25].

Xiang et al. demonstrated that increased miR-144 expression suppresses renal carcinoma proliferation, resulting in a shorter G2/M phase. Moreover, Xiang et al. revealed that overexpression of miR-144 inhibits mTOR gene and protein expression [46]. Nijland et al. demonstrated that an increase in mTOR signaling is crucial for determining the number of nephrons in embryos whose mothers were subjected to nutrient restriction [47]. Mammalian target of rapamycin complex 1 (mTORC1) is essential for embryo development; however, how this complex regulates the balance between growth and autophagy under physiological conditions and environmental stress remains unknown [48]. Therefore, mTOR signaling may be involved in cellular responses in animals exposed to LP intake during gestation; in the perception, induction, and termination of autophagy; and in response to intracellular nutrient availability [46]. Hypothetically, during severe protein restriction, reduced expression of miR-144-3p may be associated with increased mTOR expression, approximately 139% and 104% in CM cells and UB, respectively, to compensate for the loss of nephrons in the 17-DG LP offspring.

Chen et al. defined miR-127 as a new regulator of cell senescence through Bcl-6 [49]. Pan et al. reported that miR-127 underexpression correlates with increased cell proliferation in liver cells [50]. This study showed a decrease in cell proliferation and a significant reduction of cells positively labeled for Ki-67 in the CM of protein-restricted animals. Moreover, a reduction in nephrogenic area and proliferation in LP progeny was observed, which was consistent with the results of Menendez-Castro et al. in 8.4% protein-restricted progeny [51, 52]. Thus, increased Ki-67 and Bcl-6 mRNA expression, accompanied with reduced miR-127-3p expression in the 17-DG LP cap could be associated with counter-regulatory mechanisms to maintain proliferation.

Sun et al. demonstrated that overexpression of miR-199a-5p reduces cystic cell proliferation and induces apoptosis, in addition to controlling the cell cycle [53]. In this study, expression of miR-199a-5p is reduced in 17-DG LP is accompanied by the increased transcription of Ki-67, a cellular proliferation marker, and Map2k2 is associated with decreased Ki-67 reactivity in LP 17-DG metanephros. Thus, gestational undernutrition promotes differentiation through a post-transcriptional mechanism. Notably, our results reveal a repressive role of zinc-finger E-box binding homeobox 1 (ZEB1), an EMT inducer, which maintains stem cell pluripotency during embryonic stem cell differentiation. β-catenin is known to activate nuclear ZEB1 transcription resulting in ZEB1 expression [34]. The TGFβ signaling pathway, one of the best-studied pathways, can induce EMT during embryonic development. Several TGFβ like ligands are required for embryonic development. However, not all TGFβ-mediated effects on EMT depend on ZEB1/2, knockout cells can induce the expression of the mesenchymal genes fibronectin and N-cadherin. However, E-cadherin is no longer downregulated, and actin fibers are also formed [54]. Karner et al. reported that during renal development, the Wnt9b/β-catenin signaling path, expressed in both the UB and CM, is required both for nephron progenitor cell renewal and differentiation, being essential for the formation of nephrons during embryogenesis [55]. The evolutionarily conserved Wnt9b/β-catenin pathway plays a critical role in developing organs, tissues, and injury repair in pluricellular organisms. A study demonstrated that c-Myc is a transcriptional target of β-catenin, regulating the proliferation and differentiation of renal tubular epithelium [56]. The expression of β-catenin at the gene and protein level increased during the studied periods of renal development in the 17-DG LP fetus. Pan et al. reported that Myc cooperates with β-catenin to promote the renewal of nephron progenitor cells [10]. Here compared to age-matched NP offspring, LP showed lower c-Myc expression. Therefore, these animals may have a lower reserve of renewable cells necessary for proliferation and survival and may reflect the smaller number of nephrons in the LP model. Moreover, Wnt/β-catenin and Notch signals pathways may coordinate the regulation of Six-2 expression and are involved in the downregulation of Six-2 expression in nephron progenitor cells. Studies have demonstrated that low levels of β-catenin might be required to maintain Six-2 expression and CM progenitor cells in the undifferentiated state; moreover, elevated levels of β-catenin determine nephron progenitor cell fate [57, 58].Thus, we hypothesize that reduced c-Myc and Notch signaling, accompanied with increased β-catenin expression, reduced Six-2 expression by 28% in 17-DG LP offspring and correlated with early CM cell differentiation and reduced stem cell and nephron number in adulthood. Moreover, our data may sustain that, in LP offspring MM cells, the increased Let-7a-5p and β-catenin expression and reduced Notch signal may modulate c-Myc, Six-2, and Ki-67 expression, leading to a reduction in self-renewal of progenitor cells. The depletion of the remaining CM progenitor cells leads to a reduction in nephron numbers and development of arterial hypertension and renal disorders in adulthood (Fig 10).

Fig 10. The picture depicted a schematic representation of the biological response disorders in metanephros of 17DG fetus from maternal restricted-protein intake.

Fig 10

Consistent with that of Boivin et al., our results indicate that increased CM β-catenin disrupts UB growth and nephrogenesis [59]. Studies have shown that growth factor glial-derived neurotrophic factor (GDNF), a crucial regulator of UB growth, signals through the c-Ret tyrosine kinase receptor and Gfra1 co-receptor [60, 61]. In 17-DG LP offspring, a significant increase in c-Ret receptor coding mRNA would theoretically lead to a rise in UB growth. However, in this study, GDNF expression was unchanged, suggesting that despite an increase in c-Ret mRNA, UB branching was reduced. Previously, we observed a reduction of 28.3% in ureteric bud branches after 14.5 days of gestational protein restriction [4], which could be associated with a 28% reduction in Six-2 labeling of MM cells, despite no change in GDNF transcription. β-catenin probably interacts with the c-ret receptor and is transported to the UB cell’s nucleus, promoting TGFβ-1 expression in epithelial cells, inhibiting UB branching and causing premature differentiation of CM progenitor cell, as seen in 17-DG LP offspring [6264]. Therefore, GDNF may not be essential in mediating mesenchymal signals to the ureteric bud; however, the mechanism remains to be elucidated. Indeed, we have demonstrated that MM from 17-DG LP offspring showed a specific increase in Let-7 miRNA expression, resulting in significantly impaired kidney development, thereby confirming the modulatory role of these genes in the developmental timing of nephrogenesis [65].

In initial insulin-like growth factor (IGF) studies, predominant roles of IGF-1 and -2 in fetal growth were elucidated by abundant, but mostly indirect, evidence. IGFs were found to act as proliferation and differentiation factors in cultured fetal cells and preimplantation embryos. Moreover, IGFs were found to be secreted by cultured fetal cells and explants in vitro [66]. Growth factors, including IGF, can cause a partial or full epithelial–mesenchymal transition. The activation of IGF pathways results in the upregulation of EMT by inducing ZEB1 expression [67]. Although several candidate growth factors are involved in kidney development, whether they are involved in nephrogenesis is unknown. Different growth factors may be needed at different times. Some growth factors may be redundant in this context. During embryonic development, sequential rounds of EMT and MET are needed to differentiate specialized cell types and create a three-dimensional structure. In this study, mesenchymal–epithelial interconvertibility was found to maintain cell plasticity, suggesting the presence of a highly inducible system in LP conditions for the embryo. The expression of the Let-7 miRNA family has been extensively studied in various fetal tissues. The increase in Let-7 miRNA expression is related to reduced proliferation and early increase of MM cell differentiation, and consequently, decreased nephron numbers [27, 15, 6871]. Higher Let-7 expression has been demonstrated in higher organisms during the last phase of cerebral embryogenesis in rodents [72, 73]. Nagalakshmi et al. revealed that Let-7 miRNA expression changed UB epithelial cell fate from precursor to differentiated state [71]. By contrast, Yermalovich et al. demonstrated that the overexpression of Lin28b, an RNA-binding protein, is associated with suppressive Let-7 miRNAs. Although lin28 and Let-7 are known regulators of ontogenic timing in invertebrates, the role of these in mammalian organ development is not understood [65]. In this study, the increase in Let-7a-5p miRNA expression in the LP fetus could be associated with reduced CM cell proliferation, compromising nephrogenesis relative to the NP group. Thus, we hypothesize that the CM cell proliferation suppression and early cessation of nephrogenesis caused by increased Let-7 miRNA may occur directly or indirectly through the transiently reduced expression of Lin28b in 17-DG LP. This effect may significantly impair kidney development in 17-DG LP, confirming that this gene regulates developmental timing during nephrogenesis. In this study, increased Let-7a-5p miRNA expression coincides with a decrease in c-Myc expression. Myc is involved in proliferation, growth, apoptosis, and cell differentiation during renal organogenesis [7476]. In the LP 17-DG offspring, MM c-Myc gene expression was reduced, and the area of CM c-Myc immunoreactivity was 14% smaller, when compared to that in the NP offspring. Simultaneously, a 14% decrease in CM cell number was observed to decrease 48% Ki-67 immunoreactivity in LP relative to the NP offspring. Consistently, studies have shown that c-Myc plays an important role in the final phase of UB branching and in stimulating CM progenitor cell proliferation [74].

Let-7 is strongly expressed at late stages of cell differentiation; however, it is expressed at a reduced level in stem cells, maintaining them in an undifferentiated state. However, in this study, strongly expression of Let-7a-5p miRNA in the CM downregulated c-Myc expression, thereby reducing progenitor cell proliferation and early cell differentiation in the LP 17-DG offspring (Fig 10). Studies on the kidneys of c-Myc transgenic mice revealed a simultaneous decrease in c-Myc and S-ix-2 immunopositive CM cells associated with reduced stem cell proliferation [74]. This study shows a significant (28%) reduction in Six-2 positive cells, a specific renal stem cell marker, like the decrease observed in nephron numbers, in the CM of the 17-DG LP offspring compared to that in the NP offspring.

In 2009, Fogelgren et al. demonstrated that Six-2 gene expression is reduced during fetal ontogenesis when associated with decreased nephron numbers, hypertension, and chronic renal failure [77]. Thus, reduced Six-2 gene expression in CM progenitor cells indicates suppression of signal-induced differentiation during renal development in the 17-DG LP offspring. Nevertheless, redundancy should be used with caution—subtle defects in nephrogenesis may become evident with more detailed analysis or under different conditions. IGF1 mRNA levels were highest during the initial period of metanephric development, with transcripts being detected throughout the MM, whereas their levels declined during further development. However, during kidney embryogenesis, a delicate balance between nephron facilitating growth factors (IGF1) and inhibitory growth factors (TGFβ-1) regulates UB branching.

Conclusion

Although several authors have studied nephrogenesis [34, 37, 78], little is known about the mechanisms that determine nephron numbers. This study demonstrates that many MM progenitor cell miRNAs, mRNAs, and proteins are altered in the 17-DG LP offspring, which leads to reduced proliferation and early cell differentiation (Fig 11).

Fig 11. The picture depicted a schematic representation of supposed factors that evolved in a 28% reduction of the CM stem cells and nephron number in maternal restricted protein intake.

Fig 11

This delicate balance between nephron progenitor renewal and differentiation is essential for kidney development, because failure to achieve adequate numbers of nephrons is a risk factor for chronic renal disorder.

Supporting information

S1 File

(DOCX)

Acknowledgments

We thank the access to equipment and assistance provided by the National Institute of Science and Technology on Photonics Applied to Cell Biology (INFABIC) at the State University of Campinas; Data analysis was partial and generously performed in collaboration with Tao Chen, Ph.D. from the Division of Genetic and Molecular Toxicological, National Center for Toxicological Research, Jefferson, AR, USA.

List of abbreviations

Bax

Apoptosis regulator

Bcl-2

B-cell lymphoma 2

Bcl-6

B-cell lymphoma 6

Bim

or Bcl-2-like protein 11

BSA

Bovine serum albumin

cDNA

complementary deoxyribonucleic acid

CEUA/UNESP

Institutional Ethics Committee

c-Myc

regulator genes and Myc proto-oncogenes

CM

metanephros cap

c-ret

rearranged during transfection

DAB

3,3’- diaminobenzidine tetrahydrochloride

DNA

deoxyribonucleic acid

DG

days of gestation

GAPDH

Glyceraldehyde 3-phosphate dehydrogenase

GDNF

Glial cell line-derived neurotrophic factor

IGF1

Insulin-like growth factor 1

Ki-67

nuclear protein associated with cellular proliferation

let-7

lethal-7 (let-7) gene

Lin28b

Lin-28 Homolog B, suppressor of microRNA (miRNA) biogenesis

LP

gestational low-protein intake

Map2k2

mitogen-activated protein kinase kinase 2

miRNA (miR)

small non-coding RNA molecule

miRNA-Seq

miRNA transcriptome sequencing

MM

metanephric mesenchyme progenitor cells

mRNA

messenger ribonucleic acid

mTOR

mammalian target of rapamycin

mTORC1

mammalian target of rapamycin complex 1

NGS

Next Generation Sequencing

NOTCH1

single-pass transmembrane receptor protein

NP

normal protein intake

PCNA

Proliferating cell nuclear antigen

PRDM1

PR domain zinc finger protein 1

RIN

RNA Integrity Number

RT-qPCR

reverse transcription-polymerase chain reaction quantitative real-time

Six-2

SIX homeobox 2

TGFβ-1

transforming growth factor beta 1

UB

ureter bud

U6 and U87

internal reference gene

WT1

Wilms’ tumor protein

ZEB 1 and ZEB 2

Zinc finger E-box-binding homeobox 1 and 2

17-DG

17-days LP fetal kidney

Data Availability

Data are available in NCBI: (https://www.ncbi.nlm.nih.gov/sra/PRJNA694197) Further information can be found: (https://bv.fapesp.br/pt/pesquisador/671860/leticia-de-barros-sene/) (https://repositorio.unesp.br/handle/11449/148594).

Funding Statement

This work was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, 2013/12486-5 and 2014/50938-8), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, 465699/2014-6).

References

  • 1.Langley-Evans SC. Developmental programming of health and disease. Proc Nutr Soc. 2006; 65: 97–105. 10.1079/pns2005478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lucas A. Symposium: The Effects of Childhood Diet on Adult Health and Disease Psychological Influences on Childhood Diet 1. J Nutr. 1998;128: 401S–406S. 10.1093/jn/128.2.401S [DOI] [PubMed] [Google Scholar]
  • 3.Mesquita FF, Gontijo JA, Boer PA. Expression of renin-angiotensin system signaling compounds in maternal protein-restricted rats: effect on renal sodium excretion and blood pressure. Nephrol Dial Transplant. 2010; 25: 380–388. 10.1093/ndt/gfp505 [DOI] [PubMed] [Google Scholar]
  • 4.Mesquita FF, Gontijo JAR, Boer PA. Maternal undernutrition and the offspring kidney: From fetal to adult life. Braz J Med Biol Res. 2010; 43: 1010–1018. 10.1590/s0100-879x2010007500113 [DOI] [PubMed] [Google Scholar]
  • 5.Sene LB, Mesquita FF, de Moraes LN, Santos DC, Carvalho R, Gontijo JA, Boer PA. Involvement of renal corpuscle microRNA expression on epithelial-to-mesenchymal transition in maternal low protein diet in adult programmed rats. PLoS One. 2013; 19: e71310 10.1371/journal.pone.0071310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sene LB, Rizzi VHG, Gontijo JAR, Boer PA. Gestational low-protein intake enhances whole-kidney miR-192 and miR-200 family expression and epithelial-to-mesenchymal transition in rat adult male offspring. J Exp Biol. 2018; 22: 221 10.1242/jeb.171694 [DOI] [PubMed] [Google Scholar]
  • 7.Schreuder M, Delemarre-Van De Waal H, Van Wijk A. Consequences of intrauterine growth restriction for the kidney. Kidney Blood Press Res. 2006; 29: 108–125. 10.1159/000094538 [DOI] [PubMed] [Google Scholar]
  • 8.Grobstein C. Inductive interaction in the development of the mouse metanephros. J. Exp. Zool. 1955; 130: 319–339. [Google Scholar]
  • 9.Saxén L, Sariola H. Early organogenesis of the kidney. Pediatr. Nephrol. 1987; 1: 385–392. 10.1007/BF00849241 [DOI] [PubMed] [Google Scholar]
  • 10.Pan X, Karner CM, Carroll TJ. Myc cooperates with beta-catenin to drive gene expression in the nephron progenitor cells. Development 2017; 144: 4173–4182. 10.1242/dev.153700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Phua YL, Chu JYS, Marrone AK, Bodnar AJ, Sims-Lucas S. Renal stromal miRNAs are required for normal nephrogenesis and glomerular mesangial survival. Physiol Rep. 2015; 3: 1–19. 10.14814/phy2.12537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Monk C, Spicer J, Champagne FA. Linking prenatal maternal adversity to developmental outcomes in infants: The role of epigenetic pathways. Dev. Psychopathol. 2012; 24: 1361–1376. 10.1017/S0954579412000764 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116: 281–297. 10.1016/s0092-8674(04)00045-5 [DOI] [PubMed] [Google Scholar]
  • 14.Ambros V. The functions of animal microRNAs. Nature. 2004; 431(7006): 350–355. 10.1038/nature02871 [DOI] [PubMed] [Google Scholar]
  • 15.Ambros V. MicroRNAs and Developmental Timing. Curr Opin Genet Dev. 2012; 21: 511–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bushati N, Cohen SM. microRNA functions. Annu Rev Cell Dev Biol. 2007; 23:175–205. 10.1146/annurev.cellbio.23.090506.123406 [DOI] [PubMed] [Google Scholar]
  • 17.Pillai RS, Bhattacharyya SN, Filipowicz W. Repression of protein synthesis by miRNAs: how many mechanisms? Trends in Cell Biol. 2007; 17:118–126. 10.1016/j.tcb.2006.12.007 [DOI] [PubMed] [Google Scholar]
  • 18.Nilsen TW. Mechanisms of microRNA-mediated gene regulation in animal cells. Trends Genet. 2007; 23: 243–249. 10.1016/j.tig.2007.02.011 [DOI] [PubMed] [Google Scholar]
  • 19.Chang TC, Mendell JT. microRNAs in vertebrate physiology and human disease. Annu Rev Genomics Hum Genet. 2007; 8:215–239. 10.1146/annurev.genom.8.080706.092351 [DOI] [PubMed] [Google Scholar]
  • 20.Chu C, Rana TM. Small RNAs: Regulators and Guardians of the Genome. J Cell Physiol. 2007; 213: 412–419. 10.1002/jcp.21230 [DOI] [PubMed] [Google Scholar]
  • 21.Kim V, Han J, Siomi M. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10: 126–139. 10.1038/nrm2632 [DOI] [PubMed] [Google Scholar]
  • 22.Li JY, Yong TY, Michael MZ, Gleadle JM. The role of microRNAs in kidney disease. Nephrology (Carlton). 2010; 15: 599–608. 10.1111/j.1440-1797.2010.01363.x [DOI] [PubMed] [Google Scholar]
  • 23.Chu JYS, Sims-Lucas S, Bushnell DS, Bodnar AJ, Kreidberg JA, Ho J. Dicer function is required in the metanephric mesenchyme for early kidney development. AJP: Renal Physiol. 2014; 306: F764–F772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Harvey SJ, Jarad G, Cunningham J, Goldberg S, Schermer B, Harfe BD, et al. Podocyte-Specific Deletion of Dicer Alters Cytoskeletal Dynamics and Causes Glomerular Disease. J Am Soc Nephrol. 2008; 19,: 2150–2158. 10.1681/ASN.2008020233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lv X, Mao Z, Lyu Z, Zhang P, Zhan A, Wang J, et al. miR181c promotes apoptosis and suppresses the proliferation of metanephric mesenchyme cells by targeting Six2 in vitro. Cell Biochem Funct. 2014; 32: 571–579. 10.1002/cbf.3052 [DOI] [PubMed] [Google Scholar]
  • 26.Marrone AK, Stolz DB, Bastacky SI, Kostka D, Bodnar AJ, Ho J. MicroRNA-17~92 Is Required for Nephrogenesis and Renal Function. J Am Soc Nephrol. 2014; 25:1440–52. 10.1681/ASN.2013040390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ho J, Pandey P, Schatton T, Sims-Lucas S, Khalid M, Frank MH, et al. The pro-apoptotic protein Bim is a microRNA target in kidney progenitors. J Am Soc Nephrol. 2011; 22: 1053–63. 10.1681/ASN.2010080841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Nagalakshmi VK, Ren Q, Pugh MM, Valerius MT, McMahon AP, Yu J. Dicer regulates the development of nephrogenic and ureteric compartments in the mammalian kidney. Kidney Int. 2011; 79: 317–30. 10.1038/ki.2010.385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ho J, Kreidberg J. The long and short of microRNAs in the kidney. J Am Soc Nephrol. 2012; 23: 400–4. 10.1681/ASN.2011080797 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chomczynski P, Sacchi N. The single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction: twenty-something years on Nat Protocols. 2006; 1: 581–585. 10.1038/nprot.2006.83 [DOI] [PubMed] [Google Scholar]
  • 31.Bustin SA, Benes V, Garson JA, et al. The MIQE guidelines: Minimum information for publication of quantitative real-time PCR experiments. Clin Chem 2009; 55:611–622. 10.1373/clinchem.2008.112797 [DOI] [PubMed] [Google Scholar]
  • 32.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2ΔΔC(T) Method Methods 2001;25(4):402–408. [DOI] [PubMed] [Google Scholar]
  • 33.Cerqueira DM, Bodnar AJ, Phua YL, Freer R, Hemker SL, Walensky LD, et al. Bim gene dosage is critical in modulating nephron progenitor survival in the absence of microRNAs during kidney development. FASEB J. 2017; 31: 3540–3554. 10.1096/fj.201700010R [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Constantini F, Kopan R. Patterning a complex organ: branching morphogenesis and nephron segmentation in kidney development. Dev Cell. 2010; 18: 698–712. 10.1016/j.devcel.2010.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hendry C, Rumballe B, Moritz K, Little MH. NIH Public Access. Pediatr Nephrol. 2011; 26: 1395–1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kopan R, Chen S, Little M. Nephron Progenitor Cells. Shifting the Balance of Self-Renewal and Differentiation. Curr Top Dev Biol. 2014; 107: 293–331. 10.1016/B978-0-12-416022-4.00011-1 [DOI] [PubMed] [Google Scholar]
  • 37.Little MH, Mcmahon AP. Mammalian Kidney Development: Principles, Progress, and Projections. Cold Spring Harb Perspect Biol. 2012; 4: a008300 10.1101/cshperspect.a008300 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang P, Chen Y, Yong J, Wang P, Chen Y, Yong J, et al. Dissecting the Global Dynamic Molecular Profiles of Human Fetal Kidney Development by Single-Cell Article Dissecting the Global Dynamic Molecular Profiles of Human Fetal Kidney Development by Single-Cell RNA Sequencing. Cell Reports. 2018; 24: 3554–3567. 10.1016/j.celrep.2018.08.056 [DOI] [PubMed] [Google Scholar]
  • 39.Marrone AK, Ho J. MicroRNAs: potential regulators of renal development genes that contribute to CAKUT. Pediatr Nephrol. 2014; 29: 565–74. 10.1007/s00467-013-2599-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Phua YL, Chu JYS, Marrone AK, Bodnar AJ, Sims‐Lucas S, Ho J. Renal stromal miRNAs are required for normal nephrogenesis and glomerular mesangial survival. Physiol Rep. 2015; 3: e12537 10.14814/phy2.12537 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wei J, Zhang Y, Luo Y, Wang Z, Bi S, Song D, et al. Aldose reductase regulates miR-200a-3p/141-3p to coordinate Keap1-Nrf2, Tgf1/2, and Zeb1/2 signaling in renal mesangial cells and the renal cortex of diabetic mice. Free Radic Biol Med. 2014; 67: 91–10.2. 10.1016/j.freeradbiomed.2013.10.811 [DOI] [PubMed] [Google Scholar]
  • 42.Ji J, Yamashita T, Budhu A, Forgues M, Jia H, Li C, et al. Identification of microRNA-181 by genome-wide screening as a critical player in EpCAM-positive hepatic cancer stem cells. Hepatology. 2010; 50: 472–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chen G, Zhu W, Shi D, Lv L, Zhang C, Liu P, Hu W. MicroRNA-181a sensitizes human malignant glioma U87MG cells to radiation by targeting Bcl-2. Oncol Rep. 2010; 23: 997–1003. 10.3892/or_00000725 [DOI] [PubMed] [Google Scholar]
  • 44.Scorrano L, Korsmeyer SJ. Mechanisms of cytochrome c release by proapoptotic BCL-2 family members. Biochem. Biophys. Res Commun. 2003; 304: 437–444. 10.1016/s0006-291x(03)00615-6 [DOI] [PubMed] [Google Scholar]
  • 45.Li W, Qiu X, Jiang H, Han Y, Wei D, Liu J. ScienceDirect Downregulation of miR-181a protects mice from LPS-induced acute lung injury by targeting Bcl-2. Biomed Pharmacother. 2016; 1–8. [DOI] [PubMed] [Google Scholar]
  • 46.Xiang C, Cui S, Ke Y. MiR-144 Inhibits Cell Proliferation of Renal Cell Carcinoma by Targeting mTOR. J Huazhong Univ Sci Technolog Med Sci. 2016; 36:186–192. 10.1007/s11596-016-1564-0 [DOI] [PubMed] [Google Scholar]
  • 47.Nijland MJ, Schlabritz-loutsevitch NE, Hubbard GB, Nathanielsz PW, Cox LA. Non-human primate fetal kidney transcriptome analysis indicates the mammalian target of rapamycin (mTOR) is a central nutrient-responsive pathway. J Physiol. 2007; 579: 643–656. 10.1113/jphysiol.2006.122101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gürke J, Schindler M, Pendzialek SM, Thieme R, Grybe KJ, Heller R, Spengler, et al. Maternal diabetes promotes mTORC1 downstream signaling in rabbit preimplantation embryos. Soc. Reprod. Fertil. 2016; 1–51. 10.1530/REP-15-0523 [DOI] [PubMed] [Google Scholar]
  • 49.Chen J, Wang M, Guo M, Xie Y, Cong Y. miR-127 Regulates Cell Proliferation and Senescence by Targeting BCL6. PLoS One. 2013; 8(11): e80266 10.1371/journal.pone.0080266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Pan C, Chen H, Wang L, Yang S, Fu H, Zheng Y, et al. (2012). Down-regulation of MiR-127 facilitates hepatocyte proliferation during rat liver regeneration. PLoS One. 7; 1–10. 10.1371/journal.pone.0039151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Menendez-Castro C, Hilgers KF, Amann K, Daniel C, Cordasic N, Wachtveitl R, et al. Intrauterine growth restriction leads to a dysregulation of Wilms’ tumor suppressor gene 1 (WT1) and early podocyte alterations. Nephrol Nephrol Dial Transplant. 2013; 28: 1407–17. 10.1093/ndt/gfs517 [DOI] [PubMed] [Google Scholar]
  • 52.Menendez-Castro C, Toka O, Fahlbusch F, et al. Impaired myocardial performance in a normotensive rat model of intrauterine growth restriction. Pediatr Res. 2014;25(6):697–706. [DOI] [PubMed] [Google Scholar]
  • 53.Sun H, Zhou C, Fu L. Inhibition of MiR-199a-5p Reduced Cell Proliferation in Autosomal Dominant Polycystic Kidney Disease through Targeting CDKN1C. 195–200. Med Sci Monit. 2015; 21: 195–200. 10.12659/MSM.892141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shirakihara T, Saitoh M, Miyazono K. Differential regulation of epithelial and mesenchymal markers by delta-EF1 proteins in epithelial mesenchymal transition induced by TGF-beta. Mol Biol Cell. 2007;18(9): 3533–3544. 10.1091/mbc.e07-03-0249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Karner CM, Das A, Ma Z, Self M, Chen C, Lum L, et al. Canonical Wnt9b signaling balances progenitor cell expansion and differentiation during kidney development. Development. 2011; 1257: 1247–1257. 10.1242/dev.057646 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hu MC, Rosenblum ND. Smad1, b-catenin, and Tcf4 associate in a molecular complex with the Myc promoter in dysplastic renal tissue and cooperate to control Myc transcription. Development. 2005;132: 215–225. 10.1242/dev.01573 [DOI] [PubMed] [Google Scholar]
  • 57.Cheng HT. g-Secretase activity is dispensable for mesenchyme-to-epithelium transition but required for podocyte and proximal tubule formation in developing mouse kidney. Development. 2003;130: 5031–5042. 10.1242/dev.00697 [DOI] [PubMed] [Google Scholar]
  • 58.Cheng H, Kim M, Valerius MT, Surendran K, Schuster K, Gossler A, et al. Notch2 (but not Notch1) is required for proximal fate acquisition in the mammalian nephron. Development. 2009;134: 801–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Boivin FJ, Sarin S, Evans JC, Bridgewater D. The Good and Bad of β -Catenin in Kidney Development and Renal Dysplasia. Front. Cell Dev Biol. 2015; 22: 1–10. 10.3389/fcell.2015.00081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brodbeck S, Besenbeck B, Englert C. The transcription factor Six2 activates the expression of the Gdnf gene as well as its own promoter. Mech Dev. 2004; 121: 1211–1222. 10.1016/j.mod.2004.05.019 [DOI] [PubMed] [Google Scholar]
  • 61.Davis E, Babineau D, Wang X, Zyzanski S, Abrams B, Bodnar L, Horwitz R. Short inter-pregnancy intervals, parity, excessive pregnancy weight gain, and risk of maternal obesity. Matern Child Health J. 2014; 18: 554–562. 10.1007/s10995-013-1272-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bridgewater D, Cox B, Cain J, Lau A, Athaide V, Gill PS, et al. Canonical WNT/β-catenin signaling is required for ureteric branching. Dev Biol. 2008; 317: 83–94. 10.1016/j.ydbio.2008.02.010 [DOI] [PubMed] [Google Scholar]
  • 63.Bridgewater D, Di Giovanni V, Cain JE, Cox B, Jakobson M, Sainio K, Rosenblum ND. β-Catenin Causes Renal Dysplasia via the Upregulation of Tgf 2 and Dkk1. J Am Soc Nephrol. 2011; 22: 718–731. 10.1681/ASN.2010050562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Marose TD, Merkel CE, Mcmahon AP, Carroll TJ. β-catenin is necessary to keep cells of ureteric bud/Wolffian duct epithelium in a precursor state. Dev Biol. 2009; 314: 112–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yermalovich AV, Osborne JK, Sousa P, Han A, Kinney MA, Chen MJ, et al. Lin28 and let-7 regulate the timing of cessation of murine nephrogenesis. Nat Commun. 2019; 10: 168 10.1038/s41467-018-08127-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Agrogiannis GD, Sifakis S, Patsouris ES, Konstantinidou AE. Insulin-like growth factors in embryonic and fetal growth and skeletal development. Mol Med Rep. 2014; 10(2): 579–584. 10.3892/mmr.2014.2258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ding W, You H, Dang H, LeBlanc F, Galicia V, Lu SC, et al. Epithelial-to mesenchymal transition of murine liver tumor cells promotes invasion. Hepatology. 2010; 52(3): 945–953. 10.1002/hep.23748 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bao MH, Feng X, Zhang YW, Lou XY, Cheng Y, Zhou HH. Let-7 in Cardiovascular Diseases, Heart Development, and Cardiovascular Differentiation from Stem Cells Int J Mol Sci. 2013; 14: 23086–23102. 10.3390/ijms141123086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Copley MR, Eaves CJ. Developmental changes in hematopoietic stem cell properties. Exp Mol Med. 2013; 45: e55 10.1038/emm.2013.98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Meza-Sosa KF, Pedraza-Alva G, Perez-Martinez L. microRNAs: key triggers of neuronal cell fate. Front. Cell Neurosci. 2014; 8: 1–13. 10.3389/fncel.2014.00001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nagalakshmi VK, Lindner V, Wessels A, Yu J. MicroRNA-dependent temporal gene expression in the ureteric bud epithelium during mammalian kidney development. Dev Dyn. 2015; 244: 444–456. 10.1002/dvdy.24221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schulman BRM, Esquela-kerscher A, Slack FJ. Reciprocal expression of lin-41 and the microRNAs let-7 and mir-125 During Mouse Embryogenesis. Dev Dyn. 2005; 234: 1046–1054. 10.1002/dvdy.20599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wulczyn FG, Smirnova L, Rybak A, Brandt C, Kwidzinski E, Ninnemann O, et al. Post-transcriptional regulation of the let-7 microRNA during neural cell specification. FASEB J. 2007; 21: 415–426. 10.1096/fj.06-6130com [DOI] [PubMed] [Google Scholar]
  • 74.Couillard M., and Trudel M. C-myc as a modulator of the renal stem/progenitor cell population. Dev Dyn. 2009; 238: 405–14. 10.1002/dvdy.21841 [DOI] [PubMed] [Google Scholar]
  • 75.Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM, et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet. 2008; 40: 43–50. 10.1038/ng.2007.30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P, et al. MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res. 2007; 67: 9762–9770. 10.1158/0008-5472.CAN-07-2462 [DOI] [PubMed] [Google Scholar]
  • 77.Fogelgren B, Yang S, Sharp IC, Huckstep OJ, Ma W, Somponpun SJ, et al. Deficiency in Six2 during prenatal development is associated with reduced nephron number, chronic renal failure, and hypertension in adult mice. Am J Physiol Renal Physiol. 2009; 296: F1166–F1178. 10.1152/ajprenal.90550.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mugford JW, Yu J, Kobayashi A, McMahon AP. High-resolution gene expression analysis of the developing mouse kidney defines novel cellular compartments within the nephron progenitor population. Dev Biol. 2010; 333: 312–323. [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Emmanuel A Burdmann

14 Jul 2020

PONE-D-20-15837

The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses

PLOS ONE

Dear Dr. Gontijo,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process, especially those raised by reviewer 2.

Please submit your revised manuscript by Aug 28 2020 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Emmanuel A Burdmann

Academic Editor

PLOS ONE

Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. Please include further information regarding your in vivo study, per our guidelines (http://journals.plos.org/plosone/s/submission-guidelines#loc-animal-research). Specifically, please provide details regarding:

 1) Animal health monitoring, including:

     -frequency of monitoring, and

     -monitoring criteria

 2) the method of euthanasia for the rats, and

 3) the source of the mice

3. Please provide the missing information for Anti-mTOR in Table 1.

4. We note that Figures 9 and 10 appear to be similar to several general textbook mTOR diagrams. Because many pathway diagrams look similar even if they are not derived from each other, please verify that these figures are your own work and were not derived in part or full from copyrighted images. If they were derived from copyrighted images, you will need to get permissions from the copyright owner. If this is the case, please provide proof that the owner of the content (a) has given you written permission to use it, and (b) has approved of the CC BY license being applied to their content. You may have the following form completed by the owner as proof: https://journals.plos.org/plosone/s/file?id=7c09/content-permission-form.pdf. Alternatively, you may electronically request permissions electronically from the copyright owner and send us proof of approval, as long as the approval clearly shows that the owner has approved of the CC BY license being applied to their content. Please see https://journals.plos.org/plosone/s/licenses-and-copyright for more information.

5. We suggest you thoroughly copyedit your manuscript for language usage, spelling, and grammar. If you do not know anyone who can help you do this, you may wish to consider employing a professional scientific editing service.  

Whilst you may use any professional scientific editing service of your choice, PLOS has partnered with both American Journal Experts (AJE) and Editage to provide discounted services to PLOS authors. Both organizations have experience helping authors meet PLOS guidelines and can provide language editing, translation, manuscript formatting, and figure formatting to ensure your manuscript meets our submission guidelines. To take advantage of our partnership with AJE, visit the AJE website (http://learn.aje.com/plos/) for a 15% discount off AJE services. To take advantage of our partnership with Editage, visit the Editage website (www.editage.com) and enter referral code PLOSEDIT for a 15% discount off Editage services.  If the PLOS editorial team finds any language issues in text that either AJE or Editage has edited, the service provider will re-edit the text for free.

Upon resubmission, please provide the following:

  • The name of the colleague or the details of the professional service that edited your manuscript

  • A copy of your manuscript showing your changes by either highlighting them or using track changes (uploaded as a *supporting information* file)

  • A clean copy of the edited manuscript (uploaded as the new *manuscript* file)

6. Your ethics statement must appear in the Methods section of your manuscript. If your ethics statement is written in any section besides the Methods, please move it to the Methods section and delete it from any other section. Please also ensure that your ethics statement is included in your manuscript, as the ethics section of your online submission will not be published alongside your manuscript.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Partly

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: Yes

Reviewer #2: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: No

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: LB Sene et al. developed an interesting study to evaluate mechanisms responsible for renal structural changes in 17-day-old fetuses of pregnant rats fed a low-protein diet or a diet with normal protein content. The study was very well planned and had as main objective to evaluate micro RNAs and gene and protein expression of several factors. The authors observed relevant differences between the experimental and control groups. These results allows to understand some of the mechanisms responsible for the smaller number of nephrons observed in the offspring of rats subjected to malnutrition during pregnancy.

The manuscript needs a detailed correction of spelling and writing errors. As examples, I would like to draw your attention to some of the following necessary corrections.

Page 10, line 9: replace graph with figure.

Page 12, line four from bottom: replace which with with.

Page 12, line three from bottom: replace could by may

Page 13, line 4: replace "also, here, was demonstrated" by was observed.

These are just a few of the many errors in the manuscript.

In the legend of figure 1, explain the meaning of U6.

The legends of tables 1 and 2 are switched.

It would be interesting to mention some limitations of the study.

Reviewer #2: The manuscript by Sene et al analyzes the impact of maternal protein restriction on molecular aspects of renal development in mice, and reveals that such a restriction modifies the miRNA, mRNA and protein expression scenarios associated with proliferation, apoptosis and differentiation. This is a relevant and timely field of investigation and, in general, the study was adequately performed. The manuscript carries language problems, however, since the English quality is not good, particularly in the discussion. Such problems include grammatical mistakes, sentences that do not allow appropriate understanding (examples shown below in comment #3) and flaws in sentence structure that make the reading sometimes difficult. In this context, the paper should be assessed and have some portions rewritten by a native English speaker. In addition, a number of points should be addressed, clarified or modified before further evaluation. Additional analyses should be also performed for adequate interpretation of some of the data and to allow appropriate conclusions. These points are outlined in my comments below.

1. The authors have carried out the miRNA expression analyses based on statistically significant differences between the LP and NP groups, but have not established a fold-change cutoff for such analyses. They included, however, blue and red dashed lines to establish fold-change upregulation and downregulation thresholds in Figure 1. Some studies use a 2-fold-change cutoff, other studies 1.5 (less often), but such criteria are arbitrary. It is important, however, that the authors define whether they used or not a fold-change cutoff. If they did, please be clear about it and justify the decision. If they did not, please justify why they decided not to use any filtering criteria and explain what and how they defined the fold-change values associated with the blue and red up and down dashed lines.

2. Please also address the fold-change issue to the mRNA context in Figure 2.

3. Examples of inappropriate/unclear sentences:

... predicted gene expression patterns in the 17-days LP (17-DG) fetal kidney to elucidate the molecular pathways and differentiation renal cell proliferation.

Prior studies have shown that during kidney development, the miRNAs underexpression MM progenitor cells results in a premature reduction of cell proliferation and ...

In the current study, increased expression of miR-181a-5p in 17-DG LP relative to age-matched NP offspring; also, here, was demonstrated a 2-fold enhanced Bax/Bcl2 mRNA ratio …

… they also showed that enhanced miR-144 expression suppresses renal carcinoma proliferation and decreasing the G2/M phase cells ratio.

The let-7 miRNA family expression has been extensively studied in several fetal tissues and, priority is related with reduced proliferation and induced cell differentiation.

It has been shown in higher organisms, enhanced let-7 levels during embryogenesis (Schulman et al., 2005), and let-7a mature form is up-regulated during the developmental mouse brain.

How is it known that Six2 regulates transcription of GDNF (Brodbeck et al., 2004), thus, the reduction of 28% in the cells positive for Six2 could affect, in the same proportion the GDNF expression which in turn, would act in …

4. The study associates increased expression of miR 181a-5p in 17-DG LP offspring with increased Bax/Bcl2 mRNA ratio to explain increased apoptosis activity, despite no change in caspase mRNA expression. Since Bax overexpression has been shown to induce caspase-independent apoptosis and cordycepol C has been shown to induce caspase-independent apoptosis in HepG2 cells through a Bax-mediated mitochondrial pathway, I suggest to investigate caspase-independent mechanisms of apoptosis in 17-DG LP fetal kidneys.

5. Please discuss potential mechanisms relating reduced expression of miR-144-3p in 17-DG LP offspring CAP and decreased cell proliferation.

6. As pointed out by the authors, increased activity of mTOR led to nephron number reduction in fetal kidneys while hemizygous removal of mTOR also diminishes nephron population. Is there a narrow range of mTOR activity during nephrogenesis that appropriately regulates nephron number? Please discuss this issue and apply this discussion to analyze the current model.

7. Please clarify the sentence “Increased mRNA accompanied the reduction of miR 127-3p in 17-DG LP offspring for Ki67 associated with an increase of Bcl-6 in CM”.

8. Because Ki67 gene expression is increased and Ki-67 immunoreactivity is decreased in LP 17-DG metanephros, the authors state that gestational undernutrition promotes differentiation in detriment of proliferation. If so, this is a post-transcriptional mechanism. Please discuss how that may occur, cite other models in which a similar process occurred, and clarify the association with Zeb2 expression.

9. Please clarify the sentence “On the other hand, Yermalovich et al. (2019) have demonstrated the overexpression of Lin28b, an RNA-binding protein, is associated with suppressive let-7 miRNA expression elongated nephrogenesis, via the let-7 miRNAs upregulation.”

10. Since the authors hypothesize that overexpression of let-7 miRNAs, through a transient reduction of LIN28B, might decrease nephrogenesis and consequently the nephron number potentially via upregulation of Igf2, I recommend them to check Igf2 expression in the current LP model.

11. The authors state that “the current study established that the let-7 family of miRNAs promotes MYC expression through transcriptionally induced let-7 repressor, LIN28 enhancement and posttranscriptional expressed LIN28 RNA binding-protein, promoting downregulation upon LP kidney cells differentiation”, however there is no generated data on let-7 repressor and/or Lin28 that support this conclusion.

12. Notch signaling has been shown to promote nephrogenesis by downregulating Six2. In the current study, the authors show decreased Notch1 expression but unchanged Six2 expression and reduction of Six-2 positive cells in LP offspring metanephros. In this scenario, the statement “the increased let-7a-5p and β-catenin expression and reduced Notch signal modulate the c-myc, six2, and KI-67, leading to reduction of progenitor cells self-renewal in LP metanephros” should not be presented as a conclusion but instead as “suggests that the increased let-7a-5p and β-catenin expression and reduced Notch signal may modulate the c-myc, six2, and KI-67, leading to …”.

13. Given that c-Ret receptor tyrosine kinase is a major inducer of UB branching, the increase in c-Ret expression is expected to increase UB branching even if the expression of GDNF is unchanged. Moreover, despite the reduction of Six-2 positive cells, GDNF expression did not change. In this scenario, the current discussion does not seem to appropriately support the observed 28.3% reduction in UB branching.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Feb 5;16(2):e0246289. doi: 10.1371/journal.pone.0246289.r002

Author response to Decision Letter 0


6 Nov 2020

Response to Reviewers

PONE-D-20-15837

The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses

PLOS ONE

Dear Editor:

We are attaching the revised version and response to the Reviewers of the Manuscript PONE-D-20-15837. The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses submitted by Sene et al. resubmitted as a full paper. I have read and have abided by the statement of ethical standards for manuscripts submitted to the Plos One and the other information that all authors have approved the final article. This manuscript has not been submitted or published in any other journal. At first, thank you very much for the reviewer's comments, suggestions, and criticisms. Practically all sections of the manuscript were rewritten entirely, experiments are redone, the number of experiments has been increased, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected. The Introduction, Material, and Method and Discussion sections of the manuscript were revised and completely rewritten to include reviewers' suggestions and comments.

Hopefully, this edited manuscript version could now be better considered for publication by this prestigious Journal.

Sincerely yours,

José AR Gontijo, MD,

Internal Medicine Department, School of Medicine,

Campinas State University, Campinas, SP, Brazil.

E-mail: jgontijo@unicamp.br

A. Journal Requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please include further information regarding your in vivo study, per our guidelines (http://journals.plos.org/plosone/s/submission-guidelines#loc-animal-research).

Specifically, please provide details regarding:

1) Animal health monitoring, including:

-frequency of monitoring, and

-monitoring criteria

2) the method of euthanasia for the rats, and

3) the source of the mice

2. Please provide the missing information for Anti-mTOR in Table 1.

3. Your ethics statement must appear in the Methods section of your manuscript. If your ethics statement is written in any section besides the Methods, please move it to the Methods section and delete it from any other section. Please also ensure that your ethics statement is included in your manuscript, as the ethics section of your online submission will not be published alongside your manuscript.

B. Review Comments to the Author

Reviewer #1: LB Sene et al. developed an interesting study to evaluate mechanisms responsible for renal structural changes in 17-day-old fetuses of pregnant rats fed a low-protein diet or a diet with normal protein content. The study was very well planned and had as main objective to evaluate micro RNAs and gene and protein expression of several factors. The authors observed relevant differences between the experimental and control groups. These results allows to understand some of the mechanisms responsible for the smaller number of nephrons observed in the offspring of rats subjected to malnutrition during pregnancy. The manuscript needs a detailed correction of spelling and writing errors. As examples, I would like to draw your attention to some of the following necessary corrections.

Response to Reviewer 1

REF.: PONE-D-20-15837

At first, thank you very much for your criticisms. We have greatly appreciated your comments and suggestions. Practically all sections of the manuscript were rewritten entirely, experiments are redone, the number of experiments has been increased, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected. The Introduction, Material, and Method and Discussion sections of the manuscript were revised and completely rewritten to include the suggestions and comments of reviewer 1.

Major comments

Response from the authors:

1. Page 10, line 9: replace graph with figure.

R.: Regarding the suggestion brought up by Reviewer 1, the word was replaced.

2. Page 12, line four from bottom: replace which with with

R.: Regarding the suggestion brought up by Reviewer 1, the word was replaced.

3. Page 12, line three from bottom: replace could by may OK

R.: Regarding the suggestion brought up by Reviewer 1, the word was replaced.

4. Page 13, line 4: replace "also, here, was demonstrated" by was observed. OK

These are just a few of the many errors in the manuscript.

R.: Regarding the suggestion brought up by Reviewer 1, the phrase was rewitten.

5. In the legend of figure 1, explain the meaning of U6. OK

R.: Regarding the suggestion brought up by Reviewer 1, the meaning of U6 was included in the legend of Figure 1, to read… Expression of miRNAs in the metanephros from the 17th day LP fetus compared to their expression level in the control group. Reference genes U6 and U87, protein complexes composed of small nuclear RNAs (snRNAs), were used to normalize the expression of each miRNA. Data are expressed as fold change (mean ± SD, n = 4) concerning the control group. * p≤0.05: statistical significance versus NP.

And also in Methods, to read: … Based on stability analysis, the U6 snRNA, and U87 scaRNA were used as a reference gene.

Recently the improvements in high-throughput gene expression analysis have led to numerous non-protein-coding RNA (npcRNA) molecules.

Non-protein-coding RNAs are untranslated RNA molecules frequently playing regulatory roles in different developmental and cellular processes. U87 scaRNA, and U6 snRNA) possess significantly higher expression stability than the best protein-coding housekeeping RNAs. Based on our stability analysis are recommended the inclusion of U6 snRNA, and U87 scaRNA in the minimal set of reference candidates to be evaluated for normalization in any given npcRNA transcriptome analysis.

6. The legends of tables 1 and 2 are switched.

R.: Regarding the suggestion brought up by Reviewer 1, the legends of Tables 1 and 2 were switched.

Review Comments to the Author

Reviewer #2: The manuscript by Sene et al analyzes the impact of maternal protein restriction on molecular aspects of renal development in mice, and reveals that such a restriction modifies the miRNA, mRNA and protein expression scenarios associated with proliferation, apoptosis and differentiation. This is a relevant and timely field of investigation and, in general, the study was adequately performed. The manuscript carries language problems, however, since the English quality is not good, particularly in the discussion. Such problems include grammatical mistakes, sentences that do not allow appropriate understanding (examples shown below in comment #3) and flaws in sentence structure that make the reading sometimes difficult. In this context, the paper should be assessed and have some portions rewritten by a native English speaker. In addition, a number of points should be addressed, clarified or modified before further evaluation. Additional analyses should be also performed for adequate interpretation of some of the data and to allow appropriate conclusions. These points are outlined in my comments below.

Response to Reviewer 2

REF.: PONE-D-20-15837

At first, thank you very much for your criticisms. We have greatly appreciated your comments and suggestions. Practically all sections of the manuscript were rewritten entirely, experiments are redone, the number of experiments has been increased, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected. The Introduction, Material, and Method and Discussion sections of the manuscript were revised and completely rewritten to include the suggestions and comments of reviewer 1.

Major comments

Response from authors:

1. The authors have carried out the miRNA expression analyses based on statistically significant differences between the LP and NP groups, but have not established a fold-change cutoff for such analyses. They included, however, blue and red dashed lines to establish fold-change upregulation and downregulation thresholds in Figure 1. Some studies use a 2-fold-change cutoff, other studies 1.5 (less often), but such criteria are arbitrary. It is important, however, that the authors define whether they used or not a fold-change cutoff. If they did, please be clear about it and justify the decision. If they did not, please justify why they decided not to use any filtering criteria and explain what and how they defined the fold-change values associated with the blue and red up and down dashed lines.

R.: Bearing in mind the question raised by Reviewer 2, studies have used arbitrary criteria to define the upper and lower cutoff points related to gene expression. As this value is defined arbitrarily, the present study's authors established a cutoff point variation of 1.3 fold-change related to control group values. The change of the 1.3 (upwards) and 0.65 (downwards) were defined since the statistical analysis found a P-value to miRs and target mRNAs significant concerning the NP. Therefore, as we have established these cutoff points for miR, we maintained the same to validate the sequencing differences.

2. Please also address the fold-change issue to the mRNA context in Figure 2.

R.: Bearing in mind the question raised by Reviewer 2, studies have used arbitrary criteria to define the upper and lower cutoff points related to gene expression. As this value is defined arbitrarily, the present study's authors established a cutoff point variation of 1.3 fold-change related to control group values. The change of the 1.3 (upwards) and 0.65 (downwards) were defined since the statistical analysis found a P-value to miRs and target mRNAs significant concerning the NP. Therefore, as we have established these cutoff points for miR, we maintained the same to validate the sequencing differences.

3. Examples of inappropriate/unclear sentences:

3.1 ...predicted gene expression patterns in the 17-days LP (17-DG) fetal kidney to elucidate the molecular pathways and differentiation renal cell proliferation.

R.: The sentence was re-written to read: ...The current study evaluated the miRNAs and kidney predicted gene expression patterns to gestational 17-days LP (DG-17) offspring elucidates the molecular pathways involved in the proliferation and differentiation of renal embryonic/fetal cell.

3.2 Prior studies have shown that during kidney development, the miRNAs underexpression MM progenitor cells results in a premature reduction of cell proliferation and ...

R.: The sentence was re-written to read: ...Previous studies have shown that some miRNAs' underexpression in mesenchymal metanephros (MM) progenitor cells, during renal ontogenesis, results in a reduction in the cell proliferation process with early differentiation of these cells and, consequently, decreased the number of nephrons (Ho et al., 2011; Nagalakshmi et al., 2011).

3.3 In the current study, was observed increased expression of miR-181a-5p in 17-DG LP relative to age-matched NP offspring; also, here, was demonstrated a 2-fold enhanced Bax/Bcl2 mRNA ratio …

R.: The sentence was re-written to read: ...The current study showed an increased expression of miR-181a-5p in 17-DG LP relative to age-matched NP offspring. Although caspase mRNA expression was not altered, a 2-fold enhanced Bax/Bcl2 mRNA ratio in LP compared to NP offspring suggests an increased CM apoptosis activity, indicating a post-transcriptional mechanism apoptosis regulation.

3.4 … they also showed that enhanced miR-144 expression suppresses renal carcinoma proliferation and decreasing the G2/M phase of cell cycle.

R.: The sentence was re-written to read: Furthermore, studies of Xiang et al. demonstrated that miR-144 expression was decreased in kidney carcinoma cells and that enhanced miR-144 expression suppresses carcinoma proliferation, reducing the cell cycle's G2/M phase.

3.5 The let-7 miRNA family expression has been extensively studied in several fetal tissues and, priority is related with reduced proliferation and induced cell differentiation.

R.: The sentence was re-written to read: ...Besides, the expression of the let-7 miRNA family has been extensively studied in different fetal tissues. The enhanced let-7 miRNA is related to reduced proliferation and early increase of metanephric mesenchyme cells differentiation and, consequently, decreased the number of nephrons (Ho et al., 2011; Ambros, 2012; Bao et al., 2013; Copley and Eaves, 2013; Meza-Sosa et al., 2014; Nagalakshmi et al., 2015).

3.6 It has been shown in higher organisms, enhanced let-7 levels during embryogenesis (Schulman et al., 2005), and let-7a mature form is up-regulated during the developmental mouse brain.

R.: The sentence was re-written to read: ...Higher let-7 expression has been demonstrated in higher organisms during the last phase of cerebral embryogenesis in rodents (Schulman et al., 2005; Wulczyn et al., 2007).

3.7 How is it known that Six2 regulates transcription of GDNF (Brodbeck et al., 2004), thus, the reduction of 28% in the cells positive for Six2 could affect, in the same proportion the GDNF expression which in turn, would act in …

R.: The sentence was re-written to read: ...As Six2 is known to regulate GDNF transcription (Brodbeck et al., 2004), the 28% reduction in labeling for Six2 cells of the metanephric mesenchyme can affect, in the same proportion GDNF expression, which in turn would act in the decrease of 28.3% of the ramifications of the ureteric bud as previously observed (Mesquita et al., 2010).

4. The study associates increased expression of miR 181a-5p in 17-DG LP offspring with increased Bax/Bcl2 mRNA ratio to explain increased apoptosis activity, despite no change in caspase mRNA expression. Since Bax overexpression has been shown to induce caspase-independent apoptosis and cordycepol C has been shown to induce caspase-independent apoptosis in HepG2 cells through a Bax-mediated mitochondrial pathway, I suggest to investigate caspase-independent mechanisms of apoptosis in 17-DG LP fetal kidneys.

R.: We thanks reviewer 2 for suggestions for the benefit of data interpretation and clarity of Discussion. …We recognize that lack or excess of nutrients is vital during embryo/fetus development stages can have irreversible consequences. Our model shows that males subjected to gestational 6% protein restriction dams showed a ~ 28% reduction in the number of nephrons. On the 17th LP gestational day, both the number of buds in the ureter and the number of nephron-progenitor cells in the metanephrogenic Cap is 28% reduced. In that circumstance (low-protein diet) and depending on the passive exhaustion mechanism, the subpopulation of self-renewable stem cells in the CM can be early depleted. During kidney development, part of primordial cells differentiates and partly remains in divisions maintaining the necessary number of stem cells sufficient to complete the nephrogenesis. We hypothesized that the lack of maternal nutrients limits amino acids' availability for protein synthesis critical to both stem cell mitosis and differentiation of these cells. Additionally, it is plausible to assume that the apoptosis process or/and autophagy mechanisms may be acting on these primordial cells submitted to an insufficient amount of nutrients within the microenvironment formed by the CM's cluster of intensive growths and in differentiation. We also have seen that animals raised to protein restriction even during the breastfeeding period showed an additional 10% reduction in nephrons units; preliminary data from our lab assume that autophagy is also occurring. Thus, as suggest the Reviewer 2, right now, we are performing studies that aim to verify the occurrence of autophagy and involvement of apoptosis path in nephron progenitor cells of male and female metanephric mesenchyme in different intrauterine days during renal development from the offspring of mice submitted to gestational protein restriction, comparatively to their controls different periods of renal ontogenesis.

5. Please discuss potential mechanisms relating reduced expression of miR-144-3p in 17-DG LP offspring CAP and decreased cell proliferation. And, as pointed out by the authors, increased activity of mTOR led to nephron number reduction in fetal kidneys while hemizygous removal of mTOR also diminishes nephron population. Is there a narrow range of mTOR activity during nephrogenesis that appropriately regulates nephron number? Please discuss this issue and apply this discussion to analyze the current model.

R.: Regarding the comments brought up by Reviewer 2, the text was rewritten to read: ...The study of Xiang et al. demonstrated that enhanced miR-144 expression suppresses renal carcinoma proliferation, reducing the cell cycle's G2/M phase. The Xiang group also showed that overexpression of miR-144 inhibits the mTOR gene and protein expression (Xiang et al., 2016). Previously, Nijland and col. demonstrated that an increased mTOR-signaling is crucial for determining the number of nephrons in embryos whose mothers were subjected to a nutrient restriction (Nijland et al., 2007). The mammalian target of rapamycin complex 1 (mTORC1) is known to be essential for embryos development; however, how this complex regulates the balance between growth and autophagy in physiological conditions and environmental stress remains unknown (Gürke et al., 2016). Therefore, it has been suggested that mTOR signaling would undoubtedly be involved in cellular responses in maternal protein underfeeding animals and plays an intricate role in the perception, induction process, and termination of autophagy and response to intracellular nutrient availability (Xiang et al., 2016). By hypothesis, in the present severe protein-restricted study, a reduced expression of miR-144-3p may be associated with remarkably increased mTOR expression, about 139%, and 104% in CM cells and UB, as supposedly a containment mechanism to reduce the lost number of nephrons in the 17-DG LP offspring.

6. Please clarify the sentence “Increased mRNA accompanied the reduction of miR 127-3p in 17-DG LP offspring for Ki67 associated with an increase of Bcl-6 in CM”.

R.: Regarding the comments brought up by Reviewer 2, the text was rewritten to read: …Pan et col. show in liver cells that miR-127 under-expression is related to increased cell proliferation (Pan et al., 2012). Since the present study showed a decreased cell proliferation and significant reduction of cells positively labeled for Ki67 in CM in protein-restricted animals, we may infer that increased Ki67 and Bcl-6 mRNA expression accompanied by reduced miR-127-3p in the 17-DG LP cap could be associated with presumably counter-regulatory mechanisms aimed at maintaining the proliferative process.

7. Because Ki67 gene expression is increased and Ki-67 immunoreactivity is decreased in LP 17-DG metanephros, the authors state that gestational undernutrition promotes differentiation in detriment of proliferation. If so, this is a post-transcriptional mechanism. Please discuss how that may occur, cite other models in which a similar process occurred, and clarify the association with Zeb2 expression.

R.: Regarding the comments brought up by Reviewer 2, the text was rewritten to read: … The present study showed a decreased cell proliferation and significant reduction of cells positively labeled for Ki67 in CM in protein-restricted animals. The present study also demonstrates a reduced nephrogenic area and proliferation activity in LP progeny, confirming Menendez-Castro and col. studies (2013, 2014) in 8.4% protein-restricted progeny. We may infer that increased Ki67 and Bcl-6 mRNA expression accompanied by reduced miR-127-3p in the 17-DG LP cap could be associated with presumably counter-regulatory mechanisms to maintain the proliferative process. Sun et col have demonstrated that overexpression of miR-199a-5p reduces cystic cell proliferation and induces apoptosis, in addition to controlling the cell cycle (Sun et al., 2015). In the current study, expression of miR-199a-5p is reduced in the 17-DG LP cap accompanied by increased transcription of Ki67, a cellular proliferation marker, and Map2k2 is associated with decreased Ki-67 reactivity in LP 17-DG metanephros. This finding suggests that gestational undernutrition promotes differentiation in detriment of proliferation by a post-transcriptional mechanism. Surprisingly, this study showed a repressive role of zinc-finger E-box binding homeobox 1 (ZEB1) expression, a crucial inducer of EMT supposedly to maintain stem cell pluripotency on embryonic stem cell differentiation. It is known β-catenin activates nuclear ZEB1 transcription resulted in ZEB1 expression [34]. One of the best-studied pathways that were already early identified as having EMT inducing capacities during embryologic development is the TGFβ signaling pathway. Indeed, a multitude of TGFβ like ligands is necessary for proper embryonic development. However, it is clear that not all TGFβ mediated effects on EMT depend on ZEB1/2 once knocked cells can still induce the mesenchymal genes fibronectin N-cadherin. However, E-cadherin is no longer downregulated, and actin fibers are even formed (Shirakihara et al., 2007). During renal development, Karner et al. (2011) demonstrated that the Wnt9b/β-catenin signaling path, expressed in both UB and CM, is required both for nephron progenitor cell renewal and differentiation being essential for the formation of nephrons during embryogenesis. The evolutionary conserved Wnt9b/β-catenin pathway plays a critical role in developing organs, tissues, and injury repair in pluricellular organisms. The study demonstrated that c-myc is a transcriptional target of β-catenin, regulating renal tubular epithelium's proliferation and differentiation (Hu and Rosenblum, 2005). The gene and protein expression of β-catenin increases during the studied renal development periods in the 17-DG LP fetus. Recently the Pan group showed that myc cooperates with β-catenin to enhance the renewal of nephron progenitor cells (Pan et al., 2017). Here, the LP compared to age-matched NP offspring showed lower c-myc expression. So we can assume these animals may have a lower reserve of renewing cells necessary for proliferation and survival and may reflect on the smaller number of nephrons in the LP model. Also, Wnt/β-catenin and Notch signals pathways may coordinate that Six2 expression regulation and are involved in the downregulated Six2 expression in nephron progenitor cells. Have been previously demonstrated that a low level of β-catenin might be required for the maintenance of Six2 expression and the CM progenitor cell in the undifferentiated phase and, also that elevated levels of β-catenin levels also determine nephron progenitor cell fate (Cheng, 2003; Cheng et al., 2009). Thus, we may hypothesize that reduced c-myc and Notch signaling accompanied by enhanced β-catenin expression cause 28% reduced Six2 expression in the 17-DG LP offspring related to early CM cell differentiation, reduced stem cells, and nephron number adulthood. Also, our data may sustain that, in LP offspring MM cells, the increased let-7a-5p and β-catenin expression and reduced Notch signal modulate the c-myc and six2, leading to a reduction of progenitor cells self-renewal. By the way, the exhaustion of the remaining CM progenitor cell endowment, in turn, predispose reduced nephron numbers, arterial hypertension development, and renal disorders in adult age (Figure 10). In the current study, confirming Boivin et al. (2015), we may state that increased CM β-catenin might disrupt UB ramifications and nephrogenesis. Prior data observed in our Lab, showed a reduction of 28.3% in ureteric bud branches after 14.5 days of gestational protein restriction (Mesquita et al., 2010). Have been previously known that growth factor glial-derived neurotrophic factor (GDNF) encoding genes encoding, a crucial regulator of UB outgrowth acts via c-Ret tyrosine kinase receptor and Gfra1 co-receptor (Davis et al., 2015). In the 17-DG LP offspring, a significant increase in c-Ret receptor coding mRNA would theoretically lead to a rise in UB ramifications. However, in the present study, the GDNF expression was unchanged, being plausible to suppose that, despite the increase of c-Ret mRNA, the UB branching was reduced. The decrease of 28.3% ureteric bud ramifications previously observed by Mesquita et col, (2010) could be associate with a 28% reduction in labeling for Six2 cells in the MM, despite unchanged GDNF transcription. Besides, we may suppose that β-catenin interacts with the c-ret receptor and is transported to the UB cell nucleus, promoting precociously increasing TGFβ-1 expression in epithelial cells. Inhibiting UB branching and causing premature differentiation of CM progenitor cells (Bridgewater et al., 2008; Marose et al., 2009; Bridgewater et al., 2011), such as seen in 17-DG LP offspring. In this way, perhaps in this circumstance, GDNF is not essential in mediating mesenchymal signals to the ureteric bud; however, its action's exact mechanism remains to be elucidated. Indeed, we show that MM from 17-DG LP offspring showed specific enhanced let-7 miRNA expression, resulting in significantly impaired kidney development, confirming the crucial modulation role of these genes in proper developmental timing of nephrogenesis (Yermalovich et al., 2019). Therefore, we may hypothesize that up-regulated let-7 miRNAs, directly or transient by increased expression of LIN28B, controlling the early cessation of nephrogenesis. In initial IGF studies, the predominant roles of IGF-I and -II in fetal growth were elucidated by abundant but mostly indirect evidence. IGFs were shown to act as proliferation and differentiation factors in cultured fetal cells and preimplantation embryos. They were demonstrated to be secreted by cultured fetal cells and explants in vitro (Agrogiannis et al., 2014). Also, growth factors, including IGF, can cause a partial or full mesenchymal transition of the epithelial cells. Activation of IGF pathways results in EMT's upregulation by inducing ZEB1 expression (Ding et al., 2010). Although several candidate growth factors are involved in kidney development, it is unknown whether they must carry out nephrogenesis. Different growth factors may be needed at different times. Some of the growth factors may also be redundant in this particular context. During embryonic development, sequential rounds of EMT and MET are needed to differentiate specialized cell types and create the three-dimensional structure. In the present case, a mesenchymal-epithelial interconverted ability sustains cell plasticity, suggesting a highly induced phenomenon in the embryonic protein low-nutrition condition.

8. Please clarify the sentence “On the other hand, Yermalovich et al. (2019) have demonstrated the overexpression of Lin28b, an RNA-binding protein, is associated with suppressive let-7 miRNA expression elongated nephrogenesis, via the let-7 miRNAs upregulation.”

R.: We thanks reviewer 2 for suggestions for the benefit of data interpretation and clarity of Discussion, the text was rewritten to read…On the other hand, Yermalovich et al. (2019) have demonstrated that the overexpression of Lin28b, an RNA-binding protein, is associated with suppressive let-7 miRNAs. In contrast, kidney-specific loss of Lin28b impairs renal development. While the lin28 and let-7 genes are well-established regulators of ontogenic timing in invertebrates, the role of these in mammalian organ development was not fully understood. In the current study, the enhanced let-7a-5p miRNA expression in LP offspring could be associated with reducing CM cell proliferation, compromising the whole nephrogenesis relative to the NP group. The unprecedented CM cell proliferation suppression caused by increased let-7 miRNAs may occur directly or via the transient overexpression of Lin28b in the 17-DG LP.

9. Since the authors hypothesize that overexpression of let-7 miRNAs, through a transient reduction of LIN28B, might decrease nephrogenesis and consequently the nephron number potentially via upregulation of Igf2, I recommend them to check Igf2 expression in the current LP model.

R.: Regarding the comments brought up by Reviewer 2, suggesting includes studying expression and protein content for Igf2 in the gestational protein restriction model. Most studies reported a higher abundance of Igf2 mRNA in fetal tissues than adult tissues. This raised the suggestion that IGF-II is the IGF that mediates growth and differentiation in developing fetal tissues. However, at this time, we do not have inputs and fetal material available for these assessments. As we are continuing to evaluate this topic through studies with embryonic material, we are committed to assessing the expression of Igf2 in renal embryonic tissue in the future.

10. The authors state that “the current study established that the let-7 family of miRNAs promotes MYC expression through transcriptionally induced let-7 repressor, LIN28 enhancement and posttranscriptional expressed LIN28 RNA binding-protein, promoting downregulation upon LP kidney cells differentiation”, however there is no generated data on let-7 repressor and/or Lin28 that support this conclusion.

R.: Regarding the divergences between results obtained from gene expression and what would be expected, taking into account the miR expression that targets these mRNAs, we could explain by the fact that the miR binding to the target mRNA is in two ways: complete or partial, so if complete, there is the degradation of the target messenger, but when partial, it is not degraded (Bartel, 2013). These data may also differ since each miRNA can regulate the expression of several target mRNAs, and the presentation of each mRNA can be handled by several miRNAs (Enright et al., 2003; Van Rooiji et al., 2008).

11. Notch signaling has been shown to promote nephrogenesis by downregulating Six2. In the current study, the authors show decreased Notch1 expression but unchanged Six2 expression and reduction of Six-2 positive cells in LP offspring metanephros. In this scenario, the statement “the increased let-7a-5p and β-catenin expression and reduced Notch signal may modulate the c-myc, six2, and KI-67, leading to, leading to reduction of progenitor cells self-renewal in LP metanephros” should not be presented as a conclusion but instead as “suggests that the increased let-7a-5p and β-catenin expression and reduced Notch signal may modulate the c-myc, six2, and KI-67, leading to …”.

R.: Regarding the comments brought up by Reviewer 2, the text was rewritten to read:…Thus, we may hypothesize that reduced c-myc and Notch signaling accompanied by enhanced β-catenin expression cause 28% reduced Six2 expression in the 17-DG LP offspring related to early CM cell differentiation, reduced stem cells, and nephron number adulthood. Also, our data may sustain that, in LP offspring MM cells, suggests that the increased let-7a-5p and β-catenin expression and reduced Notch signal may modulate the c-myc, six2, and KI-67, leading to a reduction of progenitor cells self-renewal. By the way, the exhaustion of the remaining CM progenitor cell endowment, in turn, predispose reduced nephron numbers, arterial hypertension development, and renal disorders in adult age (Figure 10).

12. Given that c-Ret receptor tyrosine kinase is a major inducer of UB branching, the increase in c-Ret expression is expected to increase UB branching even if the expression of GDNF is unchanged. Moreover, despite the reduction of Six-2 positive cells, GDNF expression did not change. In this scenario, the current discussion does not seem to appropriately support the observed 28.3% reduction in UB branching.

R.: Regarding the comments brought up by Reviewer 2 for suggestions for the benefit of data interpretation and clarity of Discussion, the text was rewritten to read…However, in the present study, the GDNF expression was unchanged, being plausible to suppose that, despite the increase of c-Ret mRNA, the UB branching was reduced. Prior data observed in our Lab, showed a reduction of 28.3% in ureteric bud branches after 14.5 days of gestational protein restriction (Mesquita et al., 2010) could be associate with a 28% reduction in labeling for Six2 cells in the MM, despite unchanged GDNF transcription. Besides, we may suppose that β-catenin interacts with the c-ret receptor and is transported to the UB cell nucleus, promoting precociously increasing TGFβ-1 expression in epithelial cells. Inhibiting UB branching and causing premature differentiation of CM progenitor cells (Bridgewater et al., 2008; Marose et al., 2009; Bridgewater et al., 2011), such as seen in 17-DG LP offspring. In this way, perhaps in this circumstance, GDNF is not essential in mediating mesenchymal signals to the ureteric bud; however, its action's exact mechanism remains to be elucidated. Indeed, we show that MM from 17-DG LP offspring showed specific enhanced let-7 miRNA expression, resulting in significantly impaired kidney development, confirming the crucial modulation role of these genes in proper developmental timing of nephrogenesis (Yermalovich et al., 2019).

Attachment

Submitted filename: Response to Reviewers v.2.docx

Decision Letter 1

Emmanuel A Burdmann

23 Nov 2020

PONE-D-20-15837R1

Impact of gestational low-protein intake on embryonic kidney microRNA expression and in the nephron progenitor cells of the male offspring fetus

PLOS ONE

Dear Dr. Gontijo,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process by reviewer 2.

Please submit your revised manuscript by Jan 07 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Emmanuel A Burdmann

Academic Editor

PLOS ONE

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: The authors have significantly improved the manuscript, both scientifically and language-wise, however a number of flaws remain to be addressed. Specific comments follow below:

1. The authors were not as careful as they should be in their reply, copying and pasting their first response to reviewer 1 also as their first response to reviewer 2, including the term ”reviewer 1”.

2. It is unclear which experiments were redone; please specify them.

3. It is unclear which experiments had their numbers increased; please specify them.

4. It is true that the language quality improved, however the amount of sentence structure/clarity problems and grammatical mistakes remains above an acceptable level, both in the manuscript and in the reply to the reviewers. Please have the manuscript go through a native English speaker.

5. The authors have appropriately addressed my previous major comments 1 and 2, however such a point is unclear in the current manuscript. Please add this information to the revised version of the manuscript.

6. The modified versions of sentences 3.1 and 3.7 (re: previous major comment 3) are understandable but remain inappropriately written.

7. As a follow-up to my previous major comment 4, the authors state that they are currently performing studies to investigate autophagy and apoptosis pathways in nephron progenitor cells of metanephric mesenchyme during renal development, in the offspring of mice submitted to gestational protein restriction. Please include such data, at least part of them, in the current manuscript.

8. The authors have addressed my previous major comments 7, 11 and 12, however there are significant English problems in the corresponding texts.

9. My previous major comment 10 was not properly addressed, since the authors’ statement “the current study established that the let-7 family of miRNAs promotes MYC expression through transcriptionally induced let-7 repressor, LIN28 enhancement and posttranscriptional expressed LIN28 RNA binding-protein, promoting downregulation upon LP kidney cells differentiation” would require generation of data on Lin28 expression.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Feb 5;16(2):e0246289. doi: 10.1371/journal.pone.0246289.r004

Author response to Decision Letter 1


8 Dec 2020

Response to Reviewers

PONE-D-20-15837

The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses

Dear Editor:

We are attaching the re-revised version and response to the Reviewers of the Manuscript PONE-D-20-15837. The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses submitted by Sene et al. resubmitted as a full paper. We want to beware of the errors during the responses to the pertinent comments emanating by Reviewer 2 during the first evaluation of this manuscript. There is no justification for the carelessness that occurred. However, all occurred without bad faith and can be partially justified by the inexperience of the Author and the inattentive reading of the senior Author, who at the same time answered other criticisms from reviewers of other manuscripts sent for publication. I have read and have abided by the statement of ethical standards for documents submitted to the Plos One and the additional information that all authors have approved the final article. This manuscript has not been submitted or published in any other journal. At first, thank you very much for the reviewer's comments, suggestions, and criticisms. Practically all sections of the manuscript were rewritten entirely, experiments are redone, the number of experiments has been increased, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected. The Introduction, Material, and Method and Discussion sections of the manuscript were revised and completely rewritten to include reviewers' suggestions and comments.

Hopefully, this edited manuscript version could now be better considered for publication by this prestigious Journal.

Sincerely yours,

Patricia Aline Boer, Ph.D

Internal Medicine Department, School of Medicine,

Campinas State University, Campinas, SP, Brazil.

Reviewer 2 comments to the Author

Reviewer #2: The authors have significantly improved the manuscript, both scientifically and language-wise. However, several flaws remain to be addressed. Specific comments follow below:

1. The authors were not as careful as they should be in their reply, copying and pasting their first response to reviewer 1 as their first response to reviewer 2, including the term ”reviewer 1”.

2. It is unclear which experiments were redone; please specify them.

3. It is unclear which experiments had their numbers increased; please specify them.

Response from the authors:

REF.: PONE-D-20-15837

R.: 1, 2, and 3 questions.

We want to thank Reviewer 2 very much for the spending time and careful reading and to beware of the errors during the responses to the pertinent comments emanating during the first evaluation of this manuscript. There is no justification for the carelessness that occurred. However, all occurred without bad faith and can be partially justified by the inexperience of the Author and the inattentive reading of the senior Author, who at the same time answered other criticisms from reviewers of other manuscripts sent for publication. As suggested, the document was submitted for revision by a native English speaker. Among the errors that occurred during the response to Reviewer 2, the authors stated that they remade and carried out new experiments. This gross error for which we apologize was due, as stated above, to the simultaneous response of papers being reviewed simultaneously. Embarrassed, we apologize again. We have much appreciated your comments and suggestions. Practically, all manuscript sections were entirely rewritten, and many reviewer suggestions were included in that new version. The Introduction, Material, and Method and Discussion sections of the manuscript were revised and completely rewritten to include the suggestions and comments of reviewer 2.

4. It is true that the language quality improved. However, the amount of sentence structure/clarity problems and grammatical mistakes remains above an acceptable level, both in the manuscript and in reply to the reviewers. Please have the manuscript go through a native English speaker.

R.: 4 comment

As suggested, the document was submitted for revision by a native English speaker.

5. The authors have appropriately addressed my previous major comments 1 and 2, however such a point is unclear in the current manuscript. Please add this information to the revised version of the manuscript.

Response to comment 5: The changes were included in Figures 1 and 2 legends to read:

Figure 1. Expression of miRNAs in the metanephros from the 17th day LP fetus compared to their expression level in the control group. Reference genes U6 and U87, protein complexes composed of small nuclear RNAs (snRNAs), were used to normalize each miRNA expression. The authors established a cutoff point variation of 1.3 (upwards) or 0.65 (downwards), and data are expressed as fold change (mean ± SD, n = 4) concerning the control group. * p≤0.05: statistical significance versus NP.

Figure 2. Expression of mRNA estimated by SyBR green RT-qPCR of metanephros from the 17th day LP fetus. The expression was normalized with GAPDH. The authors established a cutoff point variation of 1.3 (upwards) or 0.65 (downwards), and data are expressed as fold change (mean ± SD, n = 4) concerning the control group. * p≤0.05: statistical significance versus NP.

6. The modified versions of sentences 3.1 and 3.7 (re: previous major comment 3) are understandable but remain inappropriately written.

R.: 6 comment

Sentence 3.1 was changed to read:

Authors demonstrated that gestational low-protein (LP) intake offspring presents a lower birth weight, reduced nephron numbers and renal salt excretion, arterial hypertension, and renal failure development compared to regular protein (NP) intake rats in adulthood. The current study evaluated the different miRNAs and kidney predicted target gene expression in gestational 17-days LP (DG-17) fetal metanephros seeking to elucidate some of the molecular pathways involved in the proliferation and differentiation of renal embryonic/fetal cell.

Sentence 3.7 was changed to read:

By the way, the exhaustion of the remaining CM progenitor cell endowment, in turn, predispose reduced nephron numbers, arterial hypertension development, and renal disorders in adult age (Figure 10). In the current study, confirming Boivin et al. (2015), we may state that increased CM β-catenin might disrupt UB ramifications and nephrogenesis. Have been previously known that growth factor glial-derived neurotrophic factor (GDNF) genes encoding, a crucial regulator of UB outgrowth, acts via c-Ret tyrosine kinase receptor and Gfra1 co-receptor (Brodbeck et al., 2004; Davis et al., 2015). In the 17-DG LP offspring, a significant increase in c-Ret receptor coding mRNA would theoretically lead to a rise in UB ramifications. However, in the present study, the GDNF expression was unchanged, being plausible to suppose that, despite the increase of c-Ret mRNA, the UB branching was reduced. Prior data observed in our Lab, showed a reduction of 28.3% in ureteric bud branches after 14.5 days of gestational protein restriction (Mesquita et al., 2010b) could be associate with a 28% reduction in labeling for Six2 cells in the MM, despite unchanged GDNF transcription.

7. As a follow-up to my previous major comment 4, the authors state that they are currently performing studies to investigate autophagy and apoptosis pathways in nephron progenitor cells of metanephric mesenchyme during renal development, in the offspring of mice submitted to gestational protein restriction. Please include such data, at least part of them, in the current manuscript.

R.: 7 comment

Studies are being done in a doctorate thesis in our Lab using mice from the C57BL / 6-TgCAG-RFP / EGFP / Map1lc3b1Hill / J strain, transgenic autophagy. However, the studies are ongoing, and only preliminary results have been obtained.

8. The authors have addressed my previous major comments 7, 11, and 12, however there are significant English problems in the corresponding texts.

R.: 8 comment

The grammatical and typographical errors have been edited.

9. My previous major comment 10 was not properly addressed, since the authors’ statement “the current study established that the let-7 family of miRNAs promotes MYC expression through transcriptionally induced let-7 repressor, LIN28 enhancement and posttranscriptional expressed LIN28 RNA binding-protein, promoting downregulation upon LP kidney cells differentiation” would require generation of data on Lin28 expression.

R.: 9 comment

The question raised by the reviewer is pertinent. However, we do not have enough tissue from all the animals studied to perform the analyzes. In this way, we try not to be affirmative in the discussion of the work, just carefully suggesting the possibility that, at least in part, the suppressive effects of the expression of let-7 miRNAs are indirectly due to a reduction in lin28. As we answered above, we are carrying out complementary studies on the routes shown here, so we hope in the short term to have an answer to this question.

Thus, include the text below to read:

On the other hand, Yermalovich et al. (2019) have demonstrated that the overexpression of Lin28b, an RNA-binding protein, is associated with suppressive let-7 miRNAs. While the lin28 and let-7 genes are well-established regulators of ontogenic timing in invertebrates, the role of these in mammalian organ development was not fully understood. In the current study, the enhanced let-7a-5p miRNA expression in the LP fetus could be associated with reducing CM cell proliferation, compromising the whole nephrogenesis relative to the NP group. In this way, we may hypothesize the unprecedented findings of CM cell proliferation suppression and early cessation of nephrogenesis caused by increased let-7 miRNAs may occur directly or indirectly via the transient reduced expression of Lin28b in the 17-DG LP.

Attachment

Submitted filename: Response to Reviewers v.08-12-20.docx

Decision Letter 2

Emmanuel A Burdmann

21 Dec 2020

PONE-D-20-15837R2

Impact of gestational low-protein intake on embryonic kidney microRNA expression and in the nephron progenitor cells of the male offspring fetus

PLOS ONE

Dear Dr. Gontijo,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the minor points raised during the review process.

Please submit your revised manuscript by Feb 04 2021 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org. When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: http://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols

We look forward to receiving your revised manuscript.

Kind regards,

Emmanuel A Burdmann

Academic Editor

PLOS ONE

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: Yes

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: Yes

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: My comments have been adequately addressed or justified, however two points remain to be fulfilled/considered.

1. The language quality gave one more step ahead, however it still needs improvement to reach an appropriate level. Incomplete sentences, inappropriately structured sentences and grammatical errors are still frequent, particularly in the discussion. At this point, therefore, my recommendation is to have an editing service review the manuscript.

2. The authors are presently performing studies to investigate autophagy and apoptosis pathways in nephron progenitor cells of metanephric mesenchyme during renal development in the offspring of mice submitted to gestational protein restriction. Initial data have been generated. Given the relevance of this issue to the present study and the additional time from the last revision, do the current results include any significantly robust piece of data that could expand this aspect in the present manuscript?

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #2: No

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org. Please note that Supporting Information files do not need this step.

PLoS One. 2021 Feb 5;16(2):e0246289. doi: 10.1371/journal.pone.0246289.r006

Author response to Decision Letter 2


3 Jan 2021

Response to Reviewers

PONE-D-20-15837

The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses

Dear Editor:

We are attaching the Edited version and response to the Reviewers of the Manuscript PONE-D-20-15837. The gestational low-protein intake impact in microRNA expression of the kidney progenitor cells in male offspring fetuses submitted by Sene et al. resubmitted as a full paper. Practically, all manuscript sections were rewritten entirely, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected, and the manuscript was submitted to the editing service to review. The Introduction, Material, and Method and Discussion sections of the document were revised and completely rewritten to include reviewers' suggestions and comments.

Hopefully, this edited manuscript version could now be better considered for publication by this prestigious Journal.

Sincerely yours,

Patricia Aline Boer, Ph.D

Internal Medicine Department, School of Medicine,

Campinas State University, Campinas, SP, Brazil.

Reviewer 2 comments to the Author

Reviewer #2: My comments have been adequately addressed or justified, however two points remain to be fulfilled/considered.

1. The language quality gave one more step ahead, however it still needs improvement to reach an appropriate level. Incomplete sentences, inappropriately structured sentences and grammatical errors are still frequent, particularly in the discussion. At this point, therefore, my recommendation is to have an editing service review the manuscript.

Response from Authors

R.1: Practically, all manuscript sections were rewritten entirely, and many reviewer suggestions were now included in that new version. The grammatical and typographical errors have been corrected, and the manuscript was submitted to the editing service to review. The Introduction, Material, and Method and Discussion sections of the document were revised and completely rewritten to include prior reviewers' suggestions and comments.

2. The authors are presently performing studies to investigate autophagy and apoptosis pathways in nephron progenitor cells of metanephric mesenchyme during renal development in the offspring of mice submitted to gestational protein restriction. Initial data have been generated. Given the relevance of this issue to the present study and the additional time from the last revision, do the current results include any significantly robust piece of data that could expand this aspect in the present manuscript?

R2: Dear reviewer, the authors choose to present the results on autophagy and apoptosis in a future manuscript for two main reasons: 1. it is part of a doctoral study by another student, which includes a series of other evaluations, which at this moment only presents preliminary results; 2. Studies on this topic have suffered a significant delay since the University suspended on-site activities, practically throughout the year 2020, which caused a considerable delay in obtaining the results. This delay also includes the breeding and genetic control of mice from the C57BL / 6-TgCAG-RFP / EGFP / Map1lc3b1Hill / J strain, essential for reasonable experimental control. However, the studies are ongoing, and preliminary results have been obtained.

Attachment

Submitted filename: 9Response to Reviewers v.27-12-20.docx

Decision Letter 3

Emmanuel A Burdmann

18 Jan 2021

Impact of gestational low-protein intake on embryonic kidney microRNA expression and in nephron progenitor cells of the male fetus

PONE-D-20-15837R3

Dear Dr. Gontijo,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice for payment will follow shortly after the formal acceptance. To ensure an efficient process, please log into Editorial Manager at http://www.editorialmanager.com/pone/, click the 'Update My Information' link at the top of the page, and double check that your user information is up-to-date. If you have any billing related questions, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Emmanuel A Burdmann

Section Editor

PLOS ONE

Additional Editor Comments (optional):

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #2: All comments have been addressed

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #2: (No Response)

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #2: (No Response)

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #2: (No Response)

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #2: (No Response)

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #2: (No Response)

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #2: No

Acceptance letter

Emmanuel A Burdmann

27 Jan 2021

PONE-D-20-15837R3

Impact of gestational low-protein intake on embryonic kidney microRNA expression and in nephron progenitor cells of the male fetus

Dear Dr. Gontijo:

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now with our production department.

If your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information please contact onepress@plos.org.

If we can help with anything else, please email us at plosone@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Emmanuel A Burdmann

Section Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 File

    (DOCX)

    Attachment

    Submitted filename: Response to Reviewers v.2.docx

    Attachment

    Submitted filename: Response to Reviewers v.08-12-20.docx

    Attachment

    Submitted filename: 9Response to Reviewers v.27-12-20.docx

    Data Availability Statement

    Data are available in NCBI: (https://www.ncbi.nlm.nih.gov/sra/PRJNA694197) Further information can be found: (https://bv.fapesp.br/pt/pesquisador/671860/leticia-de-barros-sene/) (https://repositorio.unesp.br/handle/11449/148594).


    Articles from PLoS ONE are provided here courtesy of PLOS

    RESOURCES