Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 283
Filtrar
1.
Food Funct ; 13(24): 12953-12965, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36448375

RESUMO

Necrotizing enterocolitis (NEC) is one of the most severe diseases commonly afflicting premature infants. Our previous studies suggests that human milk-derived exosomes (HM-Exos) have a potential therapeutic effect on NEC. In this study, we investigate the potentially therapeutic role of HM-Exos in an NEC animal model via comprehensive lncRNA and mRNA expression profiles. A rat model of NEC was induced through hypoxia, hypothermia and formula feeds. We extracted exosomes from the colostrum of healthy lactating mothers and identified their functions in an NEC animal model. Furthermore, high-throughput lncRNA and mRNA sequencings were explored to find the underlying mechanisms. Although both exosomes from term human breast milk (Term-Exos) and exosomes from preterm human breast milk (Pre-Exos) alleviated the severity of NEC, Pre-Exos seemed to better promote the proliferation of intestinal epithelial cells in vivo. We identified a total of 44 differentially expressed lncRNAs and 88 differentially expressed mRNAs between Term-Exos and Pre-Exos. Further GO and KEGG pathway analysis showed that the lncRNA-mRNA network of HM-Exos was associated with the JAK-STAT signaling pathway, bile secretion and the AMPK signaling pathway, which are predicted to be involved in the proliferation of cells. Therefore, this study reveals for the first time the important roles of human milk derived lncRNAs and mRNAs in protecting against necrotizing enterocolitis. These results provide new insight into the development of NEC.


Assuntos
Enterocolite Necrosante , Exossomos , Leite Humano , RNA Longo não Codificante , RNA Mensageiro , Animais , Feminino , Humanos , Ratos , Enterocolite Necrosante/prevenção & controle , Lactação , Leite Humano/citologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Modelos Animais de Doenças
2.
BMC Med ; 20(1): 388, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36316769

RESUMO

BACKGROUND: Breastmilk is a dynamic fluid whose initial function is to provide the most adapted nutrition to the neonate. Additional attributes have been recently ascribed to breastmilk, with the evidence of a specific microbiota and the presence of various components of the immune system, such as cytokines and leukocytes. The composition of breastmilk varies through time, according to the health status of mother and child, and altogether contributes to the future health of the infant. Obesity is a rising condition worldwide that creates a state of systemic, chronic inflammation including leukocytosis. Here, we asked whether colostrum, the milk produced within the first 48 h post-partum, would contain a distinct leukocyte composition depending on the body mass index (BMI) of the mother. METHODS: We collected peripheral blood and colostrum paired samples from obese (BMI > 30) and lean (BMI < 25) mothers within 48 h post-partum and applied a panel of 6 antibodies plus a viability marker to characterize 10 major leukocyte subpopulations using flow cytometry. RESULTS: The size, internal complexity, and surface expression of CD45 and CD16 of multiple leukocyte subpopulations were selectively regulated between blood and colostrum irrespective of the study groups, suggesting a generalized cell-specific phenotype alteration. In obesity, the colostrum B lymphocyte compartment was significantly reduced, and CD16+ blood monocytes had an increased CD16 expression compared to the lean group. CONCLUSIONS: This is the first characterization of major leukocyte subsets in colostrum of mothers suffering from obesity and the first report of colostrum leukocyte subpopulations in Latin America. We evidence various significant alterations of most leukocyte populations between blood and colostrum and demonstrate a decreased colostrum B lymphocyte fraction in obesity. This pioneering study is a stepping stone to further investigate active immunity in human breastmilk.


Assuntos
Colostro , Leucócitos , Leite Humano , Obesidade , Feminino , Humanos , Lactente , Recém-Nascido , Gravidez , Colostro/citologia , Estudos Transversais , Leite Humano/citologia , Mães
3.
Sci Rep ; 12(1): 17554, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266413

RESUMO

Myocardial injury influenced by cisplatin (Cis) is a compelling reason to hunt out a treatment modality to overcome such a threat in cisplatin-treated patients. Breast Milk mesenchymal stem cells (Br-MSCs) are a non-invasive, highly reproducible source of stem cells. Herein, we investigate Br-MSCs' role in cardiotoxicity induced by cisplatin. Rats were divided into; control, Cis-treated (received 12 mg/kg single intraperitoneal injection), BrMSCs-treated (received single intraperitoneal injection of 0.5 ml sterilized phosphate-buffered saline containing 2 × 107 cells of Br-MSCs); metformin-treated (received 250 mg/kg/day orally) and BrMSCs + metformin + Cis treated groups. At the experiment end, serum creatine kinase (CK-MB) and cardiac troponin T (cTnT) activates were estimated, cardiac malondialdehyde (MDA), superoxide dismutase (SOD), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) levels were measured, cardiac expression of Bax and Bcl-2 and AMP-activated protein kinase (AMPK), as well as heart histopathology, were evaluated. Study results showed that Cis explored acute cardiotoxicity evidenced by deteriorated cardiac indices, induction of oxidative stress, and inflammation with myocardium histological alterations. Treatment with Br-MSCs restored heart function and structure deteriorated by Cis injection. The antioxidant/anti-inflammatory/anti-apoptotic results of Br-MSCs were supported by AMPK activation denoting their protective role against cisplatin-induced cardiac injury. These results were superior when metformin was added to the treatment protocol.


Assuntos
Cardiotoxicidade , Cisplatino , Células-Tronco Mesenquimais , Metformina , Humanos , Masculino , Ratos , Proteínas Quinases Ativadas por AMP/metabolismo , Anti-Inflamatórios/farmacologia , Antioxidantes/metabolismo , Apoptose , Proteína X Associada a bcl-2/metabolismo , Cardiotoxicidade/terapia , Cisplatino/toxicidade , Creatina Quinase Forma MB/metabolismo , Malondialdeído/metabolismo , Células-Tronco Mesenquimais/citologia , Metformina/farmacologia , Leite Humano/citologia , Estresse Oxidativo , Superóxido Dismutase/metabolismo , Troponina T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
Mol Nutr Food Res ; 66(19): e2200090, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35984112

RESUMO

SCOPE: Human breast milk contains a variety of cell types that have potential roles in infant immunity and development. One challenge associates with defining the purpose(s) of milk cells in the infant is a poor understanding of the effect of digestion on cell fate. METHODS AND RESULTS: This study first demonstrates that milk cell death occurs after gastric digestion in mice. Then flow cytometry and RT-PCR are used to understand the mechanism of human milk cell death and quantify live cell types before and after simulated gastric digestion. This study finds that digestion in simulated gastric fluid for 30 min reduces cell viability from 72% to 27%, with most cell death is caused by the acidic pH. The primary mechanism of cell death is caspase-mediated apoptosis. The non-cellular components of milk offer only mild protection against cell death from stomach acid. CONCLUSIONS: Gastric digestion does not select for a specific resilient cell population to survive-most cell types die in equal proportions in the gastric environment. Taken together, these results provide a foundation with which to understand the fate of human breast milk cells in the infant's intestine and beyond.


Assuntos
Digestão , Leite Humano , Estômago , Animais , Caspases/metabolismo , Feminino , Humanos , Lactente , Camundongos , Leite Humano/citologia , Leite Humano/metabolismo , Estômago/fisiologia
5.
Proc Natl Acad Sci U S A ; 119(15): e2121720119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377806

RESUMO

Human breast milk (hBM) is a dynamic fluid that contains millions of cells, but their identities and phenotypic properties are poorly understood. We generated and analyzed single-cell RNA-sequencing (scRNA-seq) data to characterize the transcriptomes of cells from hBM across lactational time from 3 to 632 d postpartum in 15 donors. We found that the majority of cells in hBM are lactocytes, a specialized epithelial subset, and that cell-type frequencies shift over the course of lactation, yielding greater epithelial diversity at later points. Analysis of lactocytes reveals a continuum of cell states characterized by transcriptional changes in hormone-, growth factor-, and milk production-related pathways. Generalized additive models suggest that one subcluster, LC1 epithelial cells, increases as a function of time postpartum, daycare attendance, and the use of hormonal birth control. We identify several subclusters of macrophages in hBM that are enriched for tolerogenic functions, possibly playing a role in protecting the mammary gland during lactation. Our description of the cellular components of breast milk, their association with maternal­infant dyad metadata, and our quantification of alterations at the gene and pathway levels provide a detailed longitudinal picture of hBM cells across lactational time. This work paves the way for future investigations of how a potential division of cellular labor and differential hormone regulation might be leveraged therapeutically to support healthy lactation and potentially aid in milk production.


Assuntos
Lactação , Leite Humano , Aleitamento Materno , Feminino , Perfilação da Expressão Gênica , Humanos , Lactação/genética , Leite Humano/citologia , Leite Humano/metabolismo , RNA-Seq , Transcriptoma
6.
Nat Commun ; 13(1): 562, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35091553

RESUMO

Under normal conditions, the most significant expansion and differentiation of the adult mammary gland occurs in response to systemic reproductive hormones during pregnancy and lactation to enable milk synthesis and secretion to sustain the offspring. However, human mammary tissue remodelling that takes place during pregnancy and lactation remains poorly understood due to the challenge of acquiring samples. We report here single-cell transcriptomic analysis of 110,744 viable breast cells isolated from human milk or non-lactating breast tissue, isolated from nine and seven donors, respectively. We found that human milk largely contains epithelial cells belonging to the luminal lineage and a repertoire of immune cells. Further transcriptomic analysis of the milk cells identified two distinct secretory cell types that shared similarities with luminal progenitors, but no populations comparable to hormone-responsive cells. Taken together, our data offers a reference map and a window into the cellular dynamics that occur during human lactation and may provide further insights on the interplay between pregnancy, lactation and breast cancer.


Assuntos
Mama/metabolismo , Perfilação da Expressão Gênica/métodos , Lactação/genética , Glândulas Mamárias Humanas/metabolismo , Leite Humano/metabolismo , Análise de Célula Única/métodos , Mama/citologia , Técnicas de Cultura de Células em Três Dimensões/métodos , Células Cultivadas , Análise por Conglomerados , Feminino , Redes Reguladoras de Genes , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/metabolismo , Glândulas Mamárias Humanas/citologia , Leite Humano/citologia , Gravidez , Células Estromais/citologia , Células Estromais/metabolismo
7.
Int J Mol Med ; 48(6)2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34651660

RESUMO

Breast milk is the ideal food for infants and undoubtedly has immediate and long­term benefits. Breast milk contains extracellular vesicles (EVs) i.e., exosomes secreted by maternal breast cells. Exosomes carry genetic material, such as long non­coding RNAs (lncRNAs), which possibly participate in cell­to­cell communications, as they are known to regulate critical gene pathways. The aim of the present study was to screen human breastmilk exosomes for their lncRNA cargo and to examine exosomal lncRNA levels associated with milk obtained from mothers that gave birth at term or prematurely (<37 weeks of gestation). Samples were collected at 3 weeks postpartum from 20 healthy, breastfeeding mothers; 10 mothers had given birth at full­term and 10 mothers preterm. Exosomal RNA was extracted from all samples and the expression of 88 distinct lncRNAs was determined using reverse transcription­quantitative PCR. A total of 13 lncRNAs were detected in ≥85% of the samples, while 31 were detected in ≥50% of the samples. Differential expression analysis of the lncRNAs between the two groups revealed ≥2­fold differences, with generally higher lncRNA concentrations found in the milk of the mothers that gave birth at term compared with those that gave birth preterm. Among these, the non­coding RNA activated at DNA damage (NORAD) was prominently detected in both groups, and its expression was significantly downregulated in the breast milk exosomes of mothers who delivered preterm. On the whole, the present study demonstrates that breast milk lncRNAs may be important factors of normal early human development. Collectively, the presence of lncRNAs in human breast milk may explain the consistent inability of researchers to fully 'humanize' animal milk.


Assuntos
Exossomos/genética , Leite Humano/citologia , RNA Longo não Codificante/genética , Adulto , Aleitamento Materno , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Leite Humano/fisiologia , Mães
8.
Gac Med Mex ; 157(2): 194-200, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34270527

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has affected all dimensions of health care, including exclusive breastfeeding assurance and its promotion. The risk of contagion and the consequences of the pandemic have raised concerns among future mothers or in those who are already breastfeeding due to the risk of possible transmission of the virus through breast milk, although active severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has not yet been detected in breast milk. The fear of contagion has favored mother-child isolation policies. So far, there is no evidence of vertical transmission, and the risk of horizontal transmission in the infant is similar to that of the general population. In infants with COVID-19, breastfeeding can even favorably change the clinical course of the disease.


La pandemia de enfermedad por coronavirus 2019 (COVID-19) ha afectado a todas las dimensiones de la atención en salud, entre ellas el aseguramiento de la lactancia materna exclusiva y su promoción. El riesgo de contagio y las consecuencias de la pandemia han provocado preocupación entre las futuras madres o las que se ya encuentran lactando debido al riesgo de una posible transmisión del virus a través de la leche materna. Aunque aún no se ha detectado el coronavirus 2 del síndrome respiratorio agudo grave (SARS-CoV-2) activo en la leche materna. El miedo al contagio ha favorecido las políticas de aislamiento madre-hijo. Hasta el momento no existe evidencia de transmisión vertical y el riesgo de transmisión horizontal en el lactante es similar al de la población general. En lactantes con COVID-19 la lactancia materna incluso puede cambiar favorablemente el curso clínico de la enfermedad.


Assuntos
Aleitamento Materno , COVID-19 , Leite Humano , Pandemias , Aleitamento Materno/psicologia , COVID-19/epidemiologia , COVID-19/transmissão , Colostro/química , Colostro/metabolismo , Transmissão de Doença Infecciosa , Feminino , Microbioma Gastrointestinal/fisiologia , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Leite Humano/química , Leite Humano/citologia , Leite Humano/metabolismo , Leite Humano/virologia , SARS-CoV-2/isolamento & purificação , Fatores de Tempo
9.
J Mammary Gland Biol Neoplasia ; 26(1): 3-8, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-34097179

RESUMO

Single cell RNA sequencing (scRNAseq) of human milk-derived cells (HMDCs) makes highly detailed analyses of the biology of human lactation possible. We explore this powerful application as an exciting tool to inspect the cellular composition of human milk. We point out some important challenges unique to this approach and highlight the importance of collaborations between biologists and well-trained bioinformaticians to utilize these data to their maximum potential. We extend this focus by discussing the first two such studies that describe HMDCs via scRNAseq and a variety of important questions in the field that warrant attention through further research. The stage is set to apply scRNAseq in human lactation biology, potentially leading to new insights regarding the molecular and cellular diversity of human secretory mammary epithelial cells.


Assuntos
Células Epiteliais/fisiologia , Lactação/fisiologia , Glândulas Mamárias Humanas/fisiologia , Leite Humano/citologia , Análise de Sequência de RNA , Análise de Célula Única/métodos , Biologia Computacional/métodos , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Leite Humano/metabolismo , Projetos de Pesquisa
10.
Gac. méd. Méx ; 157(2): 201-208, mar.-abr. 2021. tab, graf
Artigo em Espanhol | LILACS | ID: biblio-1279102

RESUMO

Resumen La pandemia de enfermedad por coronavirus 2019 (COVID-19) ha afectado a todas las dimensiones de la atención en salud, entre ellas el aseguramiento de la lactancia materna exclusiva y su promoción. El riesgo de contagio y las consecuencias de la pandemia han provocado preocupación entre las futuras madres o las que se ya encuentran lactando debido al riesgo de una posible transmisión del virus a través de la leche materna. Aunque aún no se ha detectado el coronavirus 2 del síndrome respiratorio agudo grave (SARS-CoV-2) activo en la leche materna. El miedo al contagio ha favorecido las políticas de aislamiento madre-hijo. Hasta el momento no existe evidencia de transmisión vertical y el riesgo de transmisión horizontal en el lactante es similar al de la población general. En lactantes con COVID-19 la lactancia materna incluso puede cambiar favorablemente el curso clínico de la enfermedad.


Abstract The COVID-19 pandemic has affected the health attention in all dimensions, one of them, the exclusive breastfeeding assurance and her promotion. The high risk of contagion and the pandemic consequences have raised a number of concerns in future mothers or those who are breastfeeding because of the risk of a possible transmission of the virus through breast milk. Although SARS-CoV2 has no evidence of being active on breast milk, the fear of contagion has favored mother-child isolation policies. At this point, there are no evidence of vertical transmission and the risk of horizontal transmission in the infant is similar to the general population. Breastfeeding in newborn with COVID-19, can even favorably change the clinical course of the disease.


Assuntos
Humanos , Feminino , Recém-Nascido , Aleitamento Materno/psicologia , COVID-19/transmissão , COVID-19/epidemiologia , Leite Humano/citologia , Leite Humano/metabolismo , Leite Humano/química , Fatores de Tempo , Colostro/metabolismo , Colostro/química , Transmissão Vertical de Doenças Infecciosas , Transmissão de Doença Infecciosa , Pandemias , Microbioma Gastrointestinal/fisiologia , SARS-CoV-2/isolamento & purificação , Leite Humano/virologia
11.
J Extracell Vesicles ; 10(5): e12071, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33732416

RESUMO

Maternal milk is nature's first functional food. It plays a crucial role in the development of the infant's gastrointestinal (GI) tract and the immune system. Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer enclosed vesicles released by cells for intercellular communication and are a component of milk. Recently, we discovered that human milk EVs contain a unique proteome compared to other milk components. Here, we show that physiological concentrations of milk EVs support epithelial barrier function by increasing cell migration via the p38 MAPK pathway. Additionally, milk EVs inhibit agonist-induced activation of endosomal Toll like receptors TLR3 and TLR9. Furthermore, milk EVs directly inhibit activation of CD4+ T cells by temporarily suppressing T cell activation without inducing tolerance. We show that milk EV proteins target key hotspots of signalling networks that can modulate cellular processes in various cell types of the GI tract.


Assuntos
Vesículas Extracelulares/metabolismo , Sistema de Sinalização das MAP Quinases , Leite Humano/citologia , Mucosa Bucal/fisiologia , Adulto , Linhagem Celular , Vesículas Extracelulares/imunologia , Feminino , Humanos , Mucosa Bucal/imunologia , Linfócitos T/imunologia , Receptor 3 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Mol Nutr Food Res ; 65(10): e2000845, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33715285

RESUMO

SCOPE: Human milk can prevent the development of necrotizing enterocolitis (NEC). Human milk is rich in cargo-carrying exosomes that participate in intercellular communication. This study investigated the effects of term and preterm human milk-derived exosomes, and elucidated their lipid expression profiles. METHODS AND RESULTS: Milk from healthy mothers is collected who have delivered full-term or preterm infants, and exosomes are isolated and quantified. Administration of term and preterm milk exosomes significantly enhances epithelial proliferation and migration in vitro, and ameliorates the severity of NEC in vivo. A total of 395 lipids are identified in term and preterm human milk-derived exosomes. Bioinformatics analysis and western blotting reveal that top 50 lipids regulate intestinal epithelial cell function via the Extracellular-Signal-Regulated Kinase/Mitogen Activated Protein Kinase (ERK/MAPK) pathway. CONCLUSION: This study reveals for the first time the lipidomic complexities in exosomes derived from preterm and term milk. The results provide novel mechanistic insight on how human milk prevents the development of NEC.


Assuntos
Exossomos/química , Exossomos/fisiologia , Lipídeos/análise , Lipídeos/fisiologia , Leite Humano/citologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Enterocolite Necrosante/prevenção & controle , Enterócitos/efeitos dos fármacos , Enterócitos/fisiologia , Exossomos/ultraestrutura , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Microscopia Eletrônica , Ratos
13.
Sci Rep ; 11(1): 572, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436707

RESUMO

Analysis of the human milk microbiome is complicated by the presence of a variable quantity of fat. The fat fraction of human milk is typically discarded prior to analysis. It is assumed that all cells are pelleted out of human milk by high speed centrifugation; however, studies of bovine milk have reported that bacteria may remain trapped within the fat fraction. Here, the bacterial DNA profiles of the fat fraction and cell pellet of human milk (n = 10) were analysed. Human and bacterial DNA was consistently recovered from the fat fraction of human milk (average of 12.4% and 32.7%, respectively). Staphylococcus epidermidis was significantly more abundant in the cell pellet compared to the fat fraction (P = 0.038), and three low-abundance species (< 5% relative abundance) were recovered from one fraction only. However, inclusion of fat reduced the efficiency of DNA extraction by 39%. Culture-based methods were used to quantify the distribution of an exogenously added strain of Staphylococcus aureus in human milk fractions. S. aureus was consistently recovered from the fat fraction (average 28.9%). Bacterial DNA profiles generated from skim milk or cell pellets are not representative of the entire human milk microbiome. These data have critical implications for the design of future work in this field.


Assuntos
Centrifugação/métodos , Gorduras , Leite Humano/citologia , Leite Humano/microbiologia , DNA Bacteriano/análise , Humanos , Leite Humano/química , Staphylococcus/genética , Staphylococcus/isolamento & purificação , Staphylococcus aureus/genética , Staphylococcus aureus/isolamento & purificação
14.
J Mammary Gland Biol Neoplasia ; 25(4): 367-387, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33216249

RESUMO

Cells in human milk are an untapped source, as potential "liquid breast biopsies", of material for investigating lactation physiology in a non-invasive manner. We used single cell RNA sequencing (scRNA-seq) to identify milk-derived mammary epithelial cells (MECs) and their transcriptional signatures in women with diet-controlled gestational diabetes (GDM) with normal lactation. Methodology is described for coordinating milk collections with single cell capture and library preparation via cryopreservation, in addition to scRNA-seq data processing and analyses of MEC transcriptional signatures. We comprehensively characterized 3740 cells from milk samples from two mothers at two weeks postpartum. Most cells (>90%) were luminal MECs (luMECs) expressing lactalbumin alpha and casein beta and positive for keratin 8 and keratin 18. Few cells were keratin 14+ basal MECs and a small immune cell population was present (<10%). Analysis of differential gene expression among clusters identified six potentially distinct luMEC subpopulation signatures, suggesting the potential for subtle functional differences among luMECs, and included one cluster that was positive for both progenitor markers and mature milk transcripts. No expression of pluripotency markers POU class 5 homeobox 1 (POU5F1, encoding OCT4) SRY-box transcription factor 2 (SOX2) or nanog homeobox (NANOG), was observed. These observations were supported by flow cytometric analysis of MECs from mature milk samples from three women with diet-controlled GDM (2-8 mo postpartum), indicating a negligible basal/stem cell population (epithelial cell adhesion molecule (EPCAM)-/integrin subunit alpha 6 (CD49f)+, 0.07%) and a small progenitor population (EPCAM+/CD49f+, 1.1%). We provide a computational framework for others and future studies, as well as report the first milk-derived cells to be analyzed by scRNA-seq. We discuss the clinical potential and current limitations of using milk-derived cells as material for characterizing human mammary physiology.


Assuntos
Biologia Computacional/métodos , Diabetes Gestacional/metabolismo , Lactação/fisiologia , Glândulas Mamárias Humanas/metabolismo , Leite Humano/citologia , Adulto , Diabetes Gestacional/dietoterapia , Células Epiteliais/metabolismo , Feminino , Citometria de Fluxo , Humanos , Glândulas Mamárias Humanas/citologia , Período Pós-Parto/metabolismo , Gravidez , RNA-Seq/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto , Análise de Célula Única , Células-Tronco/metabolismo
15.
Cancer Res ; 80(21): 4828-4839, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32934021

RESUMO

Radiologic techniques remain the main method for early detection for breast cancer and are critical to achieve a favorable outcome from cancer. However, more sensitive detection methods to complement radiologic techniques are needed to enhance early detection and treatment strategies. Using our recently established culturing method that allows propagation of normal and cancerous breast epithelial cells of luminal origin, flow cytometry characterization, and genomic sequencing, we show that cancer cells can be detected in breast milk. Cells derived from milk from the breast with cancer were enriched for CD49f+/EpCAM-, CD44+/CD24-, and CD271+ cancer stem-like cells (CSC). These CSCs carried mutations within the cytoplasmic retention domain of HDAC6, stop/gain insertion in MORF4L1, and deletion mutations within SWI/SNF complex component SMARCC2. CSCs were sensitive to HDAC6 inhibitors, BET bromodomain inhibitors, and EZH2 inhibitors, as mutations in SWI/SNF complex components are known to increase sensitivity to these drugs. Among cells derived from breast milk of additional ten women not known to have breast cancer, two of them contained cells that were enriched for the CSC phenotype and carried mutations in NF1 or KMT2D, which are frequently mutated in breast cancer. Breast milk-derived cells with NF1 mutations also carried copy-number variations in CDKN2C, PTEN, and REL genes. The approach described here may enable rapid cancer cell characterization including driver mutation detection and therapeutic screening for pregnancy/postpartum breast cancers. Furthermore, this method can be developed as a surveillance or early detection tool for women at high risk for developing breast cancer. SIGNIFICANCE: These findings describe how a simple method for characterization of cancer cells in pregnancy and postpartum breast cancer can be exploited as a surveillance tool for women at risk of developing breast cancer.


Assuntos
Neoplasias da Mama , Carcinoma Ductal de Mama , Técnicas de Cultura de Células , Leite Humano/citologia , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/patologia , Feminino , Humanos , Células-Tronco Neoplásicas/patologia
16.
Nutrients ; 12(9)2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32854237

RESUMO

BACKGROUND: Infections are the main cause of death in preterm infants. Causative agents often descend from the intestinal flora of the infected neonate, indicating insufficient protection by the mucosal barrier. Breast milk (BM) contains different subsets of immune cells. We recently showed that BM contains significant numbers of myeloid-derived suppressor cells (MDSC)-immune cells that actively suppress pro-inflammatory immune responses-and hypothesized that the transfer of BM-MDSC may modulate the mucosal immunity of the newborn. METHODS: Percentages of MDSC in the BM from mothers of 86 preterm infants between 23 + 0 and 36 + 6 weeks of gestation during their first five postnatal weeks were analyzed by flow cytometry and correlated with maternal and infant characteristics. RESULTS: Percentages of BM-MDSC positively correlated with gestational age and postnatal age. The expression of activation markers on BM-MDSC did not change with gestational age, but it decreased with postnatal age. Mothers who received antepartum tocolytics had lower percentages of BM-MDSC, and infant's sex strongly influenced percentages of BM-MDSC. CONCLUSION: Our results point toward a role of BM-MDSC for immune regulation in the neonatal gut, making them a potential target of immune-based therapies shortly after birth.


Assuntos
Idade Gestacional , Recém-Nascido Prematuro , Leite Humano/citologia , Leite Humano/imunologia , Células Supressoras Mieloides , Contagem de Células , Parto Obstétrico , Feminino , Humanos , Lactente , Recém-Nascido , Lactação , Masculino , Células Supressoras Mieloides/citologia , Células Supressoras Mieloides/imunologia , Parto , Gravidez , Caracteres Sexuais , Tocolíticos/administração & dosagem
17.
Pediatr Surg Int ; 36(9): 999-1007, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32671487

RESUMO

Amniotic fluid and breast milk play important roles in structural development throughout fetal growth and infancy. Given their significance in physical maturation, many studies have investigated the therapeutic and protective roles of amniotic fluid and breast milk in neonatal diseases. Of particular interest to researchers are stem cells found in the two fluids. These stem cells have been investigated due to their ability to self-replicate, differentiate, reduce tissue damage, and their expression of pluripotent markers. While amniotic fluid stem cells have received some attention regarding their ability to treat neonatal diseases, breast milk stem cells have not been investigated to the same extent given the recency of their discovery. The purpose of this review is to compare the functions of amniotic fluid, breast milk, and their stem cells to provide a rationale for the use of breast milk stem cells as a therapy for neonatal diseases. Breast milk stem cells present as an important tool for treating neonatal diseases given their ability to reduce inflammation and tissue damage, as well as their multilineage differentiation potential, easy accessibility, and ability to be used in disease modelling.


Assuntos
Líquido Amniótico/citologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Doenças do Recém-Nascido/terapia , Leite Humano/citologia , Células-Tronco/citologia , Diferenciação Celular , Humanos , Recém-Nascido
18.
Lipids ; 55(6): 661-670, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32725684

RESUMO

Human milk contains oxylipins involved in infant development. Although oxylipins have been identified in whole or skim milk, their localization within human milk cream, cell, and skim fractions is not known. This study determined the distribution of free and esterified oxylipins in cream, cell, and skim fractions of human milk. Out of 72 oxylipins probed by mass-spectrometry, 42, 29, and 41 oxylipins (free or bound) were detected in cream, cell, and skim fractions, respectively. Over 90% of free and bound oxylipins were derived from linoleic acid in all milk fractions. Other oxylipins were derived from n-6 arachidonic acid and dihomo-gamma-linolenic acid, and n-3 alpha-linolenic acid, eicosapentaenoic acid, and docosahexaenoic acid. Free oxylipins were more abundant in skim milk (59.9% of total oxylipins) compared to cream and cell pellet, whereas esterified oxylipins were most abundant in milk cream and cell pellets (74.9-76.9%). The heterogenous distribution of oxylipins in different fractions of human milk may regulate the guided release of these bioactive signaling molecules within infants.


Assuntos
Leite Humano/química , Oxilipinas/análise , Oxilipinas/química , Cromatografia Líquida de Alta Pressão , Esterificação , Feminino , Humanos , Ácido Linoleico/química , Leite Humano/citologia , Espectrometria de Massas em Tandem
19.
Front Immunol ; 11: 1035, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582165

RESUMO

Preterm infants born before 32 weeks gestational age (GA) have high rates of late onset sepsis (LOS) and necrotizing enterocolitis (NEC) despite recent improvements in infection control and nutrition. Breast milk has a clear protective effect against both these outcomes likely due to multiple mechanisms which are not fully understood but may involve effects on both the infant's immune system and the developing gut microbiota. Congregating at the interface between the mucosal barrier and the microbiota, innate and adaptive T lymphocytes (T cells) participate in this interaction but few studies have explored their development after preterm delivery. We conducted a literature review of T cell development that focuses on fetal development, postnatal maturation and the influence of milk diet. The majority of circulating T cells in the preterm infant display a naïve phenotype but are still able to initiate functional responses similar to those seen in term infants. T cells from preterm infants display a skew toward a T-helper 2(Th2) phenotype and have an increased population of regulatory cells (Tregs). There are significant gaps in knowledge in this area, particularly in regards to innate-like T cells, but work is emerging: transcriptomics and mass cytometry are currently being used to map out T cell development, whilst microbiomic approaches may help improve understanding of events at mucosal surfaces. A rapid rise in organoid models will allow robust exploration of host-microbe interactions and may support the development of interventions that modulate T-cell responses for improved infant health.


Assuntos
Recém-Nascido Prematuro/imunologia , Leite Humano/imunologia , Subpopulações de Linfócitos T/imunologia , Imunidade Adaptativa , Suscetibilidade a Doenças , Enterocolite Necrosante/imunologia , Feminino , Desenvolvimento Fetal/imunologia , Microbioma Gastrointestinal/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade Inata , Imunidade nas Mucosas , Fórmulas Infantis , Fenômenos Fisiológicos da Nutrição do Lactente/imunologia , Recém-Nascido , Leite Humano/citologia , Modelos Imunológicos , Gravidez , Sepse/imunologia
20.
Cell Transplant ; 29: 963689720912690, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32193954

RESUMO

Breastfeeding has been shown to have a protective effect on the occurrence of necrotizing enterocolitis (NEC), but the mechanism remains unclear. In the context of NEC pathogenesis, many of the protective properties of exosomes on the intestinal epithelial compartment make it an ideal therapeutic target. In the present study, our hypothesis was that intestinal stem cells (ISCs) would be protected from injury by human milk-derived exosomes (HMDEs). Human breast milk was collected, and exosomes were isolated using ExoQuick reagent. Magnetic-activated cell sorting isolation of prominin-1+ ISCs was performed from small intestines of neonatal rat. ISCs were treated with or without H2O2, and HMDEs, an equal volume of HMDE-free milk, or a control solution [phosphate-buffered solution (PBS)] was added, respectively. In the absence of HMDEs, exposure of ISCs to H2O2 led to decreased cell viability. However, addition of HMDEs to ISCs exposed to H2O2 led to significantly increased ISC viability. There was a significant upregulation of mRNA expression of Axin2, c-Myc, and Cyclin D1 genes of the Wnt/ß-catenin axis in ISCs treated with HMDEs (6.99 ± 2.34, 4.21 ± 1.68, 6.17 ± 2.22, respectively, P < 0.05 for all), as compared to control. In the presence of carnosic acid (a specific Wnt/ß-catenin signaling inhibitor), the cell viability was significantly decreased. Thus, HMDEs protect ISCs from oxidative stress injury in vitro, which were possibly mediated via the Wnt/ß-catenin signaling pathway. Our findings indicate that oral administration of HMDEs might be a promising measure in treating NEC or in preventing the development of NEC in high-risk infants when breast milk is not available.


Assuntos
Exossomos/metabolismo , Mucosa Intestinal/citologia , Leite Humano/citologia , Estresse Oxidativo/fisiologia , Células-Tronco/citologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...