1
|
Radhi N, Paul A, Muelbert M, Toldi G. Assessment of Cell Isolation From Human Milk Using Immunomagnetic Beads. J Hum Lact 2025; 41:263-271. [PMID: 39964041 PMCID: PMC11992638 DOI: 10.1177/08903344251316491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 01/13/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND There is increasing interest in better understanding the immune cell composition of human milk and how these cells interact with neonatal immune development. However, consistent methods for immune cell isolation from human milk are lacking. RESEARCH AIM Our aim was to systematically compare available cell isolation techniques to isolate T lymphocytes from human milk samples. METHODS This repeated measures study design compared three cell isolation methods using human milk samples: (1) centrifugation, (2) immunomagnetic bead isolation, and (3) a combination of both methods. We assessed the proportion and viability of CD3+, CD4+, CD25+ and regulatory T cells using flow cytometry in isolated cells to compare the performance of these isolation methods. RESULTS Immunomagnetic separation is a feasible method to isolate T lymphocytes in human milk, similar to blood. It improves target cell enrichment and cell viability compared to centrifugation, which may be an advantage when the goal is to characterize rare cell types or when cells are further used in functional assays. No excess cell activation (CD25 positivity) was observed with the use of magnetic beads. CONCLUSION Immunomagnetic separation of human milk T lymphocytes may have advantages over centrifugation depending on the intended downstream use of cells.
Collapse
Affiliation(s)
- Noor Radhi
- Department of Perinatal Science, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Ayamita Paul
- Department of Perinatal Science, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Mariana Muelbert
- Department of Perinatal Science, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Gergely Toldi
- Department of Perinatal Science, Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Naftaly S, Pery T, Mhajne R, Ashkar A, Davidovich-Pinhas M, Zinger A. Harnessing the Potential of Human Breast Milk to Boost Intestinal Permeability for Nanoparticles and Macromolecules. J Control Release 2025; 379:768-785. [PMID: 39842727 DOI: 10.1016/j.jconrel.2025.01.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
The intricate interplay between human breast milk, nanoparticles, and macromolecules holds promise for innovative nutritional delivery strategies. Compared to bovine milk and infant formula, this study explores human breast milk's role in modulating intestinal permeability and its impact on nanoparticle and macromolecule transport. Comparative analysis with bovine milk and infant formula reveals significant elevations in permeability with human breast milk, accompanied by a decrease in transepithelial electrical resistance, suggesting enhanced paracellular transport. Mechanistically, human breast milk reduces Zonula occludens-1 levels, suggesting a regulatory role in intestinal barrier function. Through in vitro and ex vivo evaluations, we aim to understand better the mechanisms behind enhanced permeability and how human breast milk affects nanoparticle physicochemical properties, potentially modulating their behavior. Specifically, human breast milk improves the intestinal permeability of liposomes in a porcine intestinal model, with associated changes in the composition of milk proteins corona related to liposome charge. These findings underscore the unexploited potential of human breast milk in facilitating transport across the intestinal barrier, offering novel avenues for human nutritional delivery and therapeutic interventions.
Collapse
Affiliation(s)
- Si Naftaly
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Topaz Pery
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Rawan Mhajne
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Areen Ashkar
- Faculty of Biotechnology and Food Engineering, Technion, Israel
| | - Maya Davidovich-Pinhas
- Faculty of Biotechnology and Food Engineering, Technion, Israel; Russell-Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Assaf Zinger
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel; Russell-Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Cardiovascular Sciences Department, Houston Methodist Academic Institute, Houston, TX 77030, United States; Neurosurgery Department, Houston Methodist Academic Institute, Houston, TX 77030, United States; Resnick Sustainability Center of Catalysis, Technion-Israel Institute of Technology, Haifa 3200003, Israel; Bruce and Ruth Rappaport Cancer Research Center, Technion-Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
3
|
Ghith A, Maleki R, Grzeskowiak LE, Amir LH, Ingman WV. Challenges and Opportunities in Quantifying Bioactive Compounds in Human Breastmilk. Biomolecules 2025; 15:325. [PMID: 40149861 PMCID: PMC11940641 DOI: 10.3390/biom15030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Breastmilk is a complex biological fluid containing over a thousand bioactive proteins, lipids, cells and small molecules that provide nutrition and immunological protection for infants and children. The composition of breastmilk is unique to each individual and can also vary within individuals according to breastfeeding duration, maternal health, time of day, and other factors. As such, the composition of breastmilk can be considered a "fingerprint" that could be interrogated to identify biomarkers of breast health and disease. However, accurate quantification of bioactive components in breastmilk remains a significant challenge. Approaches such as immunoassays and mass spectrometry have been largely applied to study blood or other biological fluids and require validation and optimisation before these techniques can be used to accurately quantify bioactive compounds in breastmilk. Development of protocols specific to breastmilk should be carried out with high precision, confidence, and sensitivity. This review explores the challenges and opportunities associated with different techniques for quantification of breastmilk bioactive components.
Collapse
Affiliation(s)
- Amna Ghith
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Reza Maleki
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| | - Luke E. Grzeskowiak
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia;
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia
| | - Lisa H. Amir
- Judith Lumley Centre, School of Nursing and Midwifery, La Trobe University, Bundoora, VIC 3086, Australia;
- Breastfeeding Service, The Royal Women’s Hospital, Parkville, VIC 3050, Australia
| | - Wendy V. Ingman
- Discipline of Surgical Specialties, Adelaide Medical School, University of Adelaide, The Queen Elizabeth Hospital, Woodville South, SA 5011, Australia; (A.G.); (R.M.)
- Robinson Research Institute, University of Adelaide, Adelaide, SA 5006, Australia
| |
Collapse
|
4
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
5
|
Komiotis C, Mavridis I. The role of stem cells in the management of neonatal posthemorrhagic hydrocephalus. Childs Nerv Syst 2024; 41:40. [PMID: 39652204 DOI: 10.1007/s00381-024-06703-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Neonatal intraventricular hemorrhage (IVH) is a common complication of prematurity as it affects 12.4% of preterm infants weighing under 1500 g. Posthemorrhagic hydrocephalus (PHH) is an important complication of neonatal IVH and can have serious long-term consequences such as cognitive impairment and cerebral palsy. The purpose of this review is to determine whether stem cell transplantation can play a role in the treatment of neonatal IVH mainly focusing on the prevention of the catastrophic sequelae of neonatal IVH, as well as to the improve outcome of these patients. METHODS A literature search was performed using the PubMed/MEDLINE and Scopus databases, and after meticulous screening, eight articles were finally selected. The authors included both animal and human studies in this narrative review. RESULTS Our review included eight articles, five animal studies and three human studies, including one phase 1 clinical trial, one pilot study, and one case report. Intraventricular transplantation of mesenchymal stem cells (MSCs) early after IVH diagnosis seems to prevent the development of PHH, improve myelination, and reduce periventricular cell death, inflammation, and reactive gliosis. It also seems to be a safe and well-tolerated procedure in preterm infants. CONCLUSION Animal and human study findings regarding stem cell transplantation in the treatment of IVH show promising results in reducing the risk of PHH. Further research with larger series is needed to better determine its safety and efficacy. Larger studies such as randomized controlled trials could establish the efficacy and tolerability of the treatment.
Collapse
Affiliation(s)
- Christodoulos Komiotis
- School of Medicine, Faculty of Health Sciences, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Mavridis
- Pediatric Neurosurgery Unit, Department of Neurosurgery, School of Medicine, Faculty of Health Sciences, Democritus University of Thrace, University General Hospital of Alexandroupolis, 68100, Alexandroupolis, Greece.
| |
Collapse
|
6
|
Surdacka LM, Jakubas A, Jagiełło J, Daniłowska K, Picheta N, Gil-Kulik P. Epigenetic and Immune Mechanisms Linking Breastfeeding to Lower Breast Cancer Rates. Med Sci Monit 2024; 30:e945451. [PMID: 39497379 PMCID: PMC11549897 DOI: 10.12659/msm.945451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/24/2024] [Indexed: 11/11/2024] Open
Abstract
This review shows how mammary stem cells (MaSCs) influence breast development, breastfeeding, and breast cancer risk. MaSCs, which can differentiate into various cell types, are vital for breast tissue health, but also disease development in breast tissue. Research shows that breastfeeding affects MaSCs, offering protection against breast cancer through various mechanisms. Hormonal changes such as increased prolactin concentration, oxytocin secretion, lower progesterone levels, and reduced exposure to estrogen during lactation promote apoptosis in potential cancer cells, boost immune surveillance, and modulate inflammation. Key findings reveal that pregnancy at an earlier age and extended breastfeeding reduce MaSC numbers, lowering cancer risk. Additionally, breastfeeding induces various epigenetic changes, such as DNA methylation and histone modification, which provide long-term protection against the development of cancer. Components of breast milk, like alpha-lactalbumin and lactoferrin, contribute by promoting cancer cell apoptosis and inhibiting tumor growth. The dual benefits of breastfeeding are reduced breast cancer risk for mothers and immunological advantages for infants. Multicenter epidemiology research has focused particular attention on longer breastfeeding duration associated with a reduced risk of triple-negative breast cancer. This review offers comprehensive evidence that breastfeeding protects against breast cancer through various biological, hormonal, and molecular mechanisms, showing the importance of promoting breastfeeding as a natural cancer prevention method. This article reviews the role of mammary stem cells in breast development, lactation, and breast cancer.
Collapse
Affiliation(s)
- Laura Maria Surdacka
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Adam Jakubas
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Jakub Jagiełło
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Karolina Daniłowska
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Natalia Picheta
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| | - Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
7
|
Schinkel ER, Nelson ER, Kim JH, Perrin MT, Dyer R, Elango R, Bode L, Dallas DC, Lueangsakulthai J, Briere CE, Taylor SN. Point-of-care human milk concentration by passive osmosis: comprehensive analysis of fresh human milk samples. J Perinatol 2024; 44:1575-1583. [PMID: 38760580 PMCID: PMC11518986 DOI: 10.1038/s41372-024-01988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
OBJECTIVE Preterm infants need enrichment of human milk (HM) for optimal growth. This study evaluated a novel, point-of-care human milk concentration (HMC) process for water removal from fresh HM samples by passive osmotic concentration. STUDY DESIGN Nineteen fresh HM samples were concentrated by incubation with the HMC devices for 3 h at 4 °C. Pre- and post-concentration HM samples were compared by HM properties for: pH, osmolality, macronutrients, enzyme activity, bioactive, and total cell viability. RESULTS Passive osmotic concentration reduced HM volume by an average of 16.3% ± 3.8% without a significant effect on pH or cell viability. Ten of the 41 HM components did not differ significantly (p > 0.05) between pre- and post-concentration samples. Twenty-three increased within the expected range by volume reduction. Six increased more than expected, two less than expected, and none decreased significantly. CONCLUSION Passive osmotic concentration of fresh HM can concentrate HM components by selective removal of water. HM osmolality and pH remained within neonatal feeding parameters.
Collapse
Affiliation(s)
- Elizabeth R Schinkel
- Mother's Milk is Best Inc., R&D, 100 Business Park Drive, Unit #5, Tyngsboro, MA, 01879, USA.
| | - Elizabeth R Nelson
- Mother's Milk is Best Inc., R&D, 100 Business Park Drive, Unit #5, Tyngsboro, MA, 01879, USA.
| | - Jae H Kim
- Perinatal Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Maryanne T Perrin
- Department of Nutrition, University of North Carolina Greensboro, Greensboro, NC, 27412, USA
| | - Roger Dyer
- Analytical Core for Metabolomics and Nutrition, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Rajavel Elango
- Analytical Core for Metabolomics and Nutrition, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC, V6H, 3V4, Canada
| | - Lars Bode
- Bode Lab, Department of Pediatrics, School of Medicine, University of California, San Diego, San Diego, CA, 92093, USA
| | - David C Dallas
- Dallas Lab, Nutrition Program, College of Health, Oregon State University, Corvallis, OR, 97331, USA
| | - Jiraporn Lueangsakulthai
- Dallas Lab, Nutrition Program, College of Health, Oregon State University, Corvallis, OR, 97331, USA
| | - Carrie-Ellen Briere
- Briere Lab, Elaine Marieb College of Nursing, University of Massachusetts Amherst, Amherst, MA, 01003, USA
- Institute for Nursing Research and Evidence-Based Practice, Connecticut Children's, Hartford, CT, 06106, US
| | - Sarah N Taylor
- Division of Neonatology, Yale School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
8
|
Buchholz MB, Scheerman DI, Levato R, Wehrens EJ, Rios AC. Human breast tissue engineering in health and disease. EMBO Mol Med 2024; 16:2299-2321. [PMID: 39179741 PMCID: PMC11473723 DOI: 10.1038/s44321-024-00112-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 08/26/2024] Open
Abstract
The human mammary gland represents a highly organized and dynamic tissue, uniquely characterized by postnatal developmental cycles. During pregnancy and lactation, it undergoes extensive hormone-stimulated architectural remodeling, culminating in the formation of specialized structures for milk production to nourish offspring. Moreover, it carries significant health implications, due to the high prevalence of breast cancer. Therefore, gaining insight into the unique biology of the mammary gland can have implications for managing breast cancer and promoting the well-being of both women and infants. Tissue engineering techniques hold promise to narrow the translational gap between existing breast models and clinical outcomes. Here, we provide an overview of the current landscape of breast tissue engineering, outline key requirements, and the challenges to overcome for achieving more predictive human breast models. We propose methods to validate breast function and highlight preclinical applications for improved understanding and targeting of breast cancer. Beyond mammary gland physiology, representative human breast models can offer new insight into stem cell biology and developmental processes that could extend to other organs and clinical contexts.
Collapse
Affiliation(s)
- Maj-Britt Buchholz
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Demi I Scheerman
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
9
|
Aydin Acar C, Pehlivanoglu S, Yesilot S, Tasdemir HI. The Effect of Breast Milk from Different Lactation Stages on in Vitro Wound Healing. Breastfeed Med 2024; 19:698-706. [PMID: 38853683 DOI: 10.1089/bfm.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Objective: Wound healing is a complex and dynamic process essential for restoring tissue integrity and homeostasis. It is thought that breast milk contributes positively to the wound healing process, thanks to the components it contains. The aim of this study is to compare the effects of breast milk on the wound healing process at different lactation stages and to evaluate the underlying mechanism(s). Materials and Methods: The effects of breast milk from different lactation stages (colostrum, transitional, and mature milk) on wound healing were determined by in vitro scratch assay in L929 fibroblast cells. 2,2-Diphenyl-1-picrylhydrazyl (DPPH), total oxidant, and antioxidant capacity were used to confirm antioxidant effects. The effect of breast milk on netrin-1 levels in L929 cells was elucidated by ELISA. Results: Breast milk at different lactation stages promoted wound healing. While the wound closure percentage was determined as 48.7% in the control group, this rate was determined to be the highest at 81.6% in the mature milk group (p:0.0002). The free radical scavenging capacity of colostrum, transitional, and mature milk with DPPH was determined as 49.69%, 60.64%, and 80.85%, respectively, depending on the lactation stages. Netrin-1 levels detected by ELISA were determined as 490.1 ± 6.5 pg/mL in the control group, while the lowest level was determined as 376.6 ± 4.5 pg/mL in mature milk (p:0.0003). Conclusions: Breast milk, especially mature milk, promoted wound healing on L929 cells by suppressing netrin-1 levels and scavenging free radicals.
Collapse
Affiliation(s)
- Cigdem Aydin Acar
- Bucak School of Health, Department of Nursing, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Suray Pehlivanoglu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey
| | - Sukriye Yesilot
- Bucak School of Health, Department of Nursing, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Halil Ibrahim Tasdemir
- Bucak School of Health, Department of Nursing, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
10
|
Goudarzi N, Shabani R, Moradi F, Ebrahimi M, Katebi M, Jafari A, Mehdinejadiani S, Vahabzade G, Soleimani M. Evaluation puramatrix as a 3D microenvironment for neural differentiation of human breastmilk stem cells. Brain Res 2024; 1836:148936. [PMID: 38649134 DOI: 10.1016/j.brainres.2024.148936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
The extracellular matrix is recognized as an efficient and determining component in the growth, proliferation, and differentiation of cells due to its ability to perceive and respond to environmental signals. Applying three-dimensional scaffolds can create conditions similar to the extracellular matrix and provide an opportunity to investigate cell fate. In this study, we employed the PuraMatrix hydrogel scaffold as an advanced cell culture platform for the neural differentiation of stem cells derived from human breastmilk to design an opportune model for tissue engineering. Isolated stem cells from breastmilk were cultured and differentiated into neural-like cells on PuraMatrix peptide hydrogel and in the two-dimensional system. The compatibility of breastmilk-derived stem cells with PuraMatrix and cell viability was evaluated by scanning electron microscopy and MTT assay, respectively. Induction of differentiation was achieved by exposing cells to the neurogenic medium. After 21 days of the initial differentiation process, the expression levels of glial fibrillary acidic protein (GFAP), microtubule-associated protein (MAP2), β-tubulin III, and neuronal nuclear antigen (NeuN) were analyzed using the immunostaining technique. The results illustrated a notable expression of MAP2, β-tubulin-III, and NeuN in the three-dimensional cell culture in comparison to the two-dimensional system, indicating the beneficial effect of PuraMatrix scaffolds in the process of differentiating breastmilk-derived stem cells into neural-like cells. In view of the obtained results, the combination of breastmilk-derived stem cells and PuraMatrix hydrogel scaffold could be an advisable preference for neural tissue regeneration and cell therapy.
Collapse
Affiliation(s)
- Nasim Goudarzi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomical Sciences, Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Ronak Shabani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Majid Katebi
- Department of Anatomy, Faculty of Medical Science, Bandarabas, Hormozgan University of Medical Sciences, Hormozgan, Iran
| | - Amir Jafari
- Laboratório de Neurofisiologia, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro.
| | - Shayesteh Mehdinejadiani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gelareh Vahabzade
- Department of Pharmacology, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoure Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Malinská N, Grobárová V, Knížková K, Černý J. Maternal-Fetal Microchimerism: Impacts on Offspring's Immune Development and Transgenerational Immune Memory Transfer. Physiol Res 2024; 73:315-332. [PMID: 39027950 PMCID: PMC11299782 DOI: 10.33549/physiolres.935296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 07/27/2024] Open
Abstract
Maternal-fetal microchimerism is a fascinating phenomenon in which maternal cells migrate to the tissues of the offspring during both pregnancy and breastfeeding. These cells primarily consist of leukocytes and stem cells. Remarkably, these maternal cells possess functional potential in the offspring and play a significant role in shaping their immune system development. T lymphocytes, a cell population mainly found in various tissues of the offspring, have been identified as the major cell type derived from maternal microchimerism. These T lymphocytes not only exert effector functions but also influence the development of the offspring's T lymphocytes in the thymus and the maturation of B lymphocytes in the lymph nodes. Furthermore, the migration of maternal leukocytes also facilitates the transfer of immune memory across generations. Maternal microchimerism has also been observed to address immunodeficiencies in the offspring. This review article focuses on investigating the impact of maternal cells transported within maternal microchimerism on the immune system development of the offspring, as well as elucidating the effector functions of maternal cells that migrate through the placenta and breast milk to reach the offspring.
Collapse
Affiliation(s)
- N Malinská
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
12
|
Jiang Q, Liu Y, Si X, Wang L, Gui H, Tian J, Cui H, Jiang H, Dong W, Li B. Potential of Milk-Derived Extracellular Vesicles as Carriers for Oral Delivery of Active Phytoconstituents. Annu Rev Food Sci Technol 2024; 15:431-454. [PMID: 38359948 DOI: 10.1146/annurev-food-072023-034354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication and have the potential to serve as in vivo carriers for delivering active molecules. The biocompatibility advantages of EVs over artificial nanocarriers create new frontiers for delivering modern active molecules. Milk is a favorable source of EVs because of its high bioavailability, low immunogenicity, and commercial producibility. In this review, we analyzed the advantages of milk-derived EVs in the oral delivery of active molecules, discussed their research progress in delivering active phytoconstituents, and summarized the necessary technologies and critical unit operations required for the development of an oral delivery system based on EVs. The review aims to provide innovative ideas and fundamental quality control guidelines for developing the next-generation oral drug delivery system based on milk-derived EVs.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Yubo Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hailong Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Hongzhou Jiang
- Anhui Ziyue Biological Technology Co., Ltd., Wuhu, China
| | - Wenjiang Dong
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| |
Collapse
|
13
|
Huang R, Chen J, Guo B, Jiang C, Sun W. Diabetes-induced male infertility: potential mechanisms and treatment options. Mol Med 2024; 30:11. [PMID: 38225568 PMCID: PMC10790413 DOI: 10.1186/s10020-023-00771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
Male infertility is a physiological phenomenon in which a man is unable to impregnate a fertile woman during a 12-month period of continuous, unprotected sexual intercourse. A growing body of clinical and epidemiological evidence indicates that the increasing incidence of male reproductive problems, especially infertility, shows a very similar trend to the incidence of diabetes within the same age range. In addition, a large number of previous in vivo and in vitro experiments have also suggested that the complex pathophysiological changes caused by diabetes may induce male infertility in multiple aspects, including hypothalamic-pituitary-gonadal axis dysfunction, spermatogenesis and maturation disorders, testicular interstitial cell damage erectile dysfunction. Based on the above related mechanisms, a large number of studies have focused on the potential therapeutic association between diabetes progression and infertility in patients with diabetes and infertility, providing important clues for the treatment of this population. In this paper, we summarized the research results of the effects of diabetes on male reproductive function in recent 5 years, elaborated the potential pathophysiological mechanisms of male infertility induced by diabetes, and reviewed and prospected the therapeutic measures.
Collapse
Affiliation(s)
- Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Jiawang Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Buyu Guo
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Chenjun Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000
| | - Weiming Sun
- The First Clinical Medical College, Lanzhou University, Lanzhou, People's Republic of China, 730000.
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
14
|
Boga B, Akbulut M, Maytalman E, Kozanoglu I. Effect of milk and whey on proliferation and differentiation of placental stromal cells. Cytotechnology 2023; 75:391-401. [PMID: 37655272 PMCID: PMC10465414 DOI: 10.1007/s10616-023-00585-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/21/2023] [Indexed: 09/02/2023] Open
Abstract
Fetal bovine serum (FBS), which is widely used in cell culture media, has the potential to cause medical and ethical problems. Here, an experimental study using milk or whey proteins containing essential nutrients and growth factors is presented to limit the use of FBS in cell culture media produced for cell and tissue regeneration. Study groups were formed by culturing human placenta mesenchymal stem cells, known to have high proliferation and differentiation capacity, with milk or whey solution at increasing concentrations, alone or in combination with FBS. Osteogenic and adipogenic differentiation capacities of proliferating cells were observed in FBS, milk or whey groups. Milk, whey or FBS groups obtained in P3 and after differentiation were separately analyzed for protein mRNA expression by reverse transcriptase-polymerase chain reaction (RT-qPCR). Fibroblast Growth Factor 2 (FGF2), Octamer-binding Transcription Factor 4 (OCT4), Bone Morphogenetic Protein 6 (BMP6), and adipogenic differentiation marker Peroxisome Proliferator-Activated Receptor Gamma (PPARG) were analysed by RT-qPCR. Proliferation was more pronounced in FBS alone and in its combinations with milk-whey compared to the groups in which only milk and whey were used. OCT4 mRNA and FGF2 mRNA expression decreased in differentiated cells. BMP6 mRNA expression increased with osteogenic and adipogenic stimuli. As expected, PPRG expression also increased with adipogenic stimulation. With this experimental study, evidence has been obtained that milk or whey can provide nutritional support to the culture media of repair cells and preserve the functional capacity of the cells, with a slightly more limited capacity than FBS. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00585-z.
Collapse
Affiliation(s)
- Bircan Boga
- School of Medicine, Acıbadem University, Istanbul, Turkey
| | - Merve Akbulut
- School of Medicine, Hacettepe University, Ankara, Turkey
| | - Erkan Maytalman
- Department of Pharmacology, School of Medicine, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ilknur Kozanoglu
- Adult Bone Marrow Transplantation and Cellular Therapy Center, Başkent University, Adana, Turkey
| |
Collapse
|
15
|
Tomaszewska A, Jeleniewska A, Porębska K, Królikowska K, Rustecka A, Lipińska-Opałka A, Będzichowska A, Zdanowski R, Aleksandrowicz K, Kloc M, Kalicki B. Immunomodulatory Effect of Infectious Disease of a Breastfed Child on the Cellular Composition of Breast Milk. Nutrients 2023; 15:3844. [PMID: 37686876 PMCID: PMC10490220 DOI: 10.3390/nu15173844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Recent studies suggest that the content of immune components in milk is influenced by the mother's health and also by the infant she feeds. We aimed to evaluate the effect of a child's respiratory tract infection on the cellular composition of breast milk (neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations). Twenty-six breastfeeding mothers whose children were hospitalized for respiratory tract infections were enrolled in the study. The control group consisted of 23 mothers of healthy children. Regarding the children, baseline laboratory blood tests were performed, and nasal swabs were taken for the presence of RS virus. In the next step, milk samples were collected from the mothers to assess the cellular composition of the milk, including neutrophils, monocytes, eosinophils, lymphocytes, and their subpopulations. Significantly higher percentages of T lymphocytes (helper and cytotoxic lymphocytes) were observed in the milk of the studied mothers. There was a significantly higher percentage of milk lymphocytes in the group of affected children with confirmed RSV etiology than in children with excluded RSV etiology. A significant positive correlation was observed between the duration of infection and the percentage of milk NK cells and between milk CD19 lymphocytes and the child's serum leukocytosis. This study may provide evidence of a link between cells in breast milk and disease in the breastfed infant. The severity of the infection, its duration, and the etiological agent of the infection may affect the cellular composition of milk.
Collapse
Affiliation(s)
- Agata Tomaszewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Alicja Jeleniewska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Klaudia Porębska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Katarzyna Królikowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Rustecka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agnieszka Lipińska-Opałka
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Agata Będzichowska
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Karolina Aleksandrowicz
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (K.P.); (R.Z.); (K.A.)
| | - Małgorzata Kloc
- Transplant Immunology, The Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Bolesław Kalicki
- Department of Paediatrics, Nephrology and Allergology, Military Institute of Medicine—National Research Institute, Szaserów 128, 04-141 Warsaw, Poland; (A.J.); (K.K.); (A.R.); (A.L.-O.); (A.B.); (B.K.)
| |
Collapse
|
16
|
Khamis T, Alsemeh AE, Alanazi A, Eltaweel AM, Abdel-Ghany HM, Hendawy DM, Abdelkhalek A, Said MA, Awad HH, Ibrahim BH, Mekawy DM, Pascu C, Florin C, Arisha AH. Breast Milk Mesenchymal Stem Cells and/or Derived Exosomes Mitigated Adenine-Induced Nephropathy via Modulating Renal Autophagy and Fibrotic Signaling Pathways and Their Epigenetic Regulations. Pharmaceutics 2023; 15:2149. [PMID: 37631363 PMCID: PMC10458733 DOI: 10.3390/pharmaceutics15082149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/01/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic kidney disease (CKD), a global health concern, is highly prevalent among adults. Presently, there are limited therapeutic options to restore kidney function. This study aimed to investigate the therapeutic potential of breast milk mesenchymal stem cells (Br-MSCs) and their derived exosomes in CKD. Eighty adult male Sprague Dawley rats were randomly assigned to one of six groups, including control, nephropathy, nephropathy + conditioned media (CM), nephropathy + Br-MSCs, nephropathy + Br-MSCs derived exosomes (Br-MSCs-EXOs), and nephropathy + Br-MSCs + Br-MSCs-EXOs. Before administration, Br-MSCs and Br-MSCs-EXOs were isolated, identified, and labeled with PKH-26. SOX2, Nanog, and OCT3/4 expression levels in Br-MSCs and miR-29b, miR-181, and Let-7b in both Br-MSCs and Br-MSCs-EXOs were assayed. Twelve weeks after transplantation, renal function tests, oxidative stress, expression of the long non-coding RNA SNHG-7, autophagy, fibrosis, and expression of profibrotic miR-34a and antifibrotic miR-29b, miR-181, and Let-7b were measured in renal tissues. Immunohistochemical analysis for renal Beclin-1, LC3-II, and P62, Masson trichome staining, and histopathological examination of kidney tissues were also performed. The results showed that Br-MSCs expressed SOX2, Nanog, and OCT3/4, while both Br-MSCs and Br-MSCs-EXOs expressed antifibrotic miR-181, miR-29b, and Let-7b, with higher expression levels in exosomes than in Br-MSCs. Interestingly, the administration of Br-MSCs + EXOs, EXOs, and Br-MSCs improved renal function tests, reduced renal oxidative stress, upregulated the renal expression of SNHG-7, AMPK, ULK-1, Beclin-1, LC3, miR-29b, miR-181, Let-7b, and Smad-7, downregulated the renal expression of miR-34a, AKT, mTOR, P62, TGF-β, Smad-3, and Coli-1, and ameliorated renal pathology. Thus, Br-MSCs and/or their derived exosomes appear to reduce adenine-induced renal damage by secreting antifibrotic microRNAs and potentiate renal autophagy by modulating SNHG-7 expression.
Collapse
Affiliation(s)
- Tarek Khamis
- Department of Pharmacology and Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Amira Ebrahim Alsemeh
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Asma Alanazi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Asmaa Monir Eltaweel
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh 11481, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Heba M. Abdel-Ghany
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Doaa M. Hendawy
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Adel Abdelkhalek
- Department of Food Hygiene, Safety and Technology, Faculty of Veterinary Medicine, Badr University in Cairo, Badr City 11829, Egypt
| | - Mahmoud A. Said
- Zagazig University Hospital, Zagazig University, Zagazig 44511, Egypt
| | - Heba H. Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 12451, Egypt
| | - Basma Hamed Ibrahim
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Dina Mohamed Mekawy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt;
| | - Corina Pascu
- Faculty of Veterinary Medicine, University of Life Sciences, King Mihai I from Timisoara [ULST], Aradului St. 119, 300645 Timisoara, Romania;
| | - Crista Florin
- Department of Soil Science, Faculty of Agriculture, University of Life Sciences, King Mihai I from Timisoara [ULST], Aradului St. 119, 300645 Timisoara, Romania
| | - Ahmed Hamed Arisha
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo, Badr City 11829, Egypt
- Department of Physiology and Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
17
|
Kemeter LM, Birzer A, Heym S, Thoma-Kress AK. Milk Transmission of Mammalian Retroviruses. Microorganisms 2023; 11:1777. [PMID: 37512949 PMCID: PMC10386362 DOI: 10.3390/microorganisms11071777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
The transmission of viruses from one host to another typically occurs through horizontal or vertical pathways. The horizontal pathways include transmission amongst individuals, usually through bodily fluids or excretions, while vertical transmission transpires from mother to their offspring, either during pregnancy, childbirth, or breastfeeding. While there are more than 200 human pathogenic viruses to date, only a small number of them are known to be transmitted via breast milk, including cytomegalovirus (CMV), human immunodeficiency virus type 1 (HIV-1), and human T cell lymphotropic virus type 1 (HTLV-1), the latter two belonging to the family Retroviridae. Breast milk transmission is a common characteristic among mammalian retroviruses, but there is a lack of reports summarizing our knowledge regarding this route of transmission of mammalian retroviruses. Here, we provide an overview of the transmission of mammalian exogenous retroviruses with a focus on Orthoretrovirinae, and we highlight whether they have been described or suspected to be transmitted through breast milk, covering various species. We also elaborate on the production and composition of breast milk and discuss potential entry sites of exogenous mammalian retroviruses during oral transmission.
Collapse
Affiliation(s)
| | | | | | - Andrea K. Thoma-Kress
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (L.M.K.); (A.B.); (S.H.)
| |
Collapse
|
18
|
Gopalarethinam J, Nair AP, Iyer M, Vellingiri B, Subramaniam MD. Advantages of mesenchymal stem cell over the other stem cells. Acta Histochem 2023; 125:152041. [PMID: 37167794 DOI: 10.1016/j.acthis.2023.152041] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/13/2023]
Abstract
A stem cell is a particular group of cells that has the extraordinary potential to convert within the body into particular cell types. They are used to regenerate tissues and cells in the body that have been damaged or destroyed by the disease. Stem cells come in three different varieties: adult stem cells, embryonic stem cells and induced pluripotent stem cells (iPSCs). Embryonic stem cells have a high chance of immune rejection and also have ethical dilemmas and iPSCs have genetic instability. Adult stem cells are difficult to analyze and extract for research since they are frequently insufficient in native tissues. However, mesenchymal stem cells (MSC) one of the categories of adult stem cells are stromal cells with a variety of potentials that can differentiate into a wide range of cell types. MSCs can be transplanted into a variety of people without worrying about rejection because they have demonstrated the ability to prevent an adverse reaction from the immune system. These transplants have powerful anti-inflammatory and immunosuppressive effects and greatly enhance the body's inherent healing capacity. While MSCs do not offer treatment for illnesses, the idea behind them is to enable the body to recover sufficiently for a protracted reduction in symptoms. In many cases, this is sufficient to significantly enhance the patient's well-being. Inspite of several advantages some potential long-term concerns connected to MSC therapy are maldifferentiation, immunosuppression and cancerous tumor growth. In this review, we will compare the mesenchymal stem cells with other stem cells with respect to the source of origin, their properties and therapeutic applications, and discuss the MSC's disadvantages.
Collapse
Affiliation(s)
- Janani Gopalarethinam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Aswathy P Nair
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore 641021, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, India
| | - Mohana Devi Subramaniam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai, India.
| |
Collapse
|
19
|
Neville MC, Demerath EW, Hahn-Holbrook J, Hovey RC, Martin-Carli J, McGuire MA, Newton ER, Rasmussen KM, Rudolph MC, Raiten DJ. Parental factors that impact the ecology of human mammary development, milk secretion, and milk composition-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 1. Am J Clin Nutr 2023; 117 Suppl 1:S11-S27. [PMID: 37173058 PMCID: PMC10232333 DOI: 10.1016/j.ajcnut.2022.11.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 05/15/2023] Open
Abstract
The goal of Working Group 1 in the Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN) Project was to outline factors influencing biological processes governing human milk secretion and to evaluate our current knowledge of these processes. Many factors regulate mammary gland development in utero, during puberty, in pregnancy, through secretory activation, and at weaning. These factors include breast anatomy, breast vasculature, diet, and the lactating parent's hormonal milieu including estrogen, progesterone, placental lactogen, cortisol, prolactin, and growth hormone. We examine the effects of time of day and postpartum interval on milk secretion, along with the role and mechanisms of lactating parent-infant interactions on milk secretion and bonding, with particular attention to the actions of oxytocin on the mammary gland and the pleasure systems in the brain. We then consider the potential effects of clinical conditions including infection, pre-eclampsia, preterm birth, cardiovascular health, inflammatory states, mastitis, and particularly, gestational diabetes and obesity. Although we know a great deal about the transporter systems by which zinc and calcium pass from the blood stream into milk, the interactions and cellular localization of transporters that carry substrates such as glucose, amino acids, copper, and the many other trace metals present in human milk across plasma and intracellular membranes require more research. We pose the question of how cultured mammary alveolar cells and animal models can help answer lingering questions about the mechanisms and regulation of human milk secretion. We raise questions about the role of the lactating parent and the infant microbiome and the immune system during breast development, secretion of immune molecules into milk, and protection of the breast from pathogens. Finally, we consider the effect of medications, recreational and illicit drugs, pesticides, and endocrine-disrupting chemicals on milk secretion and composition, emphasizing that this area needs much more research attention.
Collapse
Affiliation(s)
- Margaret C Neville
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA.
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, United States
| | - Jennifer Hahn-Holbrook
- Department of Psychological Sciences, University of California Merced, Merced, CA, United States
| | - Russell C Hovey
- Department of Animal Science, University of California Davis, Davis, CA, United States
| | - Jayne Martin-Carli
- Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Mark A McGuire
- Idaho Agricultural Experiment Station, University of Idaho, Moscow, ID, United States
| | - Edward R Newton
- Department of Obstetrics and Gynecology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Kathleen M Rasmussen
- Nancy Schlegel Meinig Professor of Maternal and Child Nutrition, Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Michael C Rudolph
- The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
20
|
Comparison of SOX2 and POU5F1 Gene Expression in Leukapheresis-Derived CD34+ Cells before and during Cell Culture. Int J Mol Sci 2023; 24:ijms24044186. [PMID: 36835597 PMCID: PMC9962001 DOI: 10.3390/ijms24044186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/18/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Bone marrow is an abundant source of both hematopoietic as well as non-hematopoietic stem cells. Embryonic, fetal and stem cells located in tissues (adipose tissue, skin, myocardium and dental pulp) express core transcription factors, including the SOX2, POU5F1 and NANOG gene responsible for regeneration, proliferation and differentiation into daughter cells. The aim of the study was to examine the expression of SOX2 and POU5F1 genes in CD34-positive peripheral blood stem cells (CD34+ PBSCs) and to analyze the influence of cell culture on the expression of SOX2 and POU5F1 genes. The study material consisted of bone marrow-derived stem cells isolated by using leukapheresis from 40 hematooncology patients. Cells obtained in this process were subject to cytometric analysis to determine the content of CD34+ cells. CD34-positive cell separation was conducted using MACS separation. Cell cultures were set, and RNA was isolated. Real-time PCR was conducted in order to evaluate the expression of SOX2 and POU5F1 genes and the obtained data were subject to statistical analysis. We identified the expression of SOX2 and POU5F1 genes in the examined cells and demonstrated a statistically significant (p < 0.05) change in their expression in cell cultures. Short-term cell cultures (<6 days) were associated with an increase in the expression of SOX2 and POU5F1 genes. Thus, short-term cultivation of transplanted stem cells could be used to induce pluripotency, leading to better therapeutic effects.
Collapse
|
21
|
Gil-Kulik P, Leśniewski M, Bieńko K, Wójcik M, Więckowska M, Przywara D, Petniak A, Kondracka A, Świstowska M, Szymanowski R, Wilińska A, Wiliński M, Płachno BJ, Kostuch M, Rahnama-Hezavach M, Szuta M, Kwaśniewska A, Bogucka-Kocka A, Kocki J. Influence of Perinatal Factors on Gene Expression of IAPs Family and Main Factors of Pluripotency: OCT4 and SOX2 in Human Breast Milk Stem Cells-A Preliminary Report. Int J Mol Sci 2023; 24:ijms24032476. [PMID: 36768802 PMCID: PMC9917041 DOI: 10.3390/ijms24032476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Due to their therapeutic potential, mesenchymal stem cells are the subject of intensive research on the use of their potential in the treatment of, among others, neurodegenerative diseases or immunological diseases. They are among the newest in the field of medicine. The presented study aimed to evaluate the expression of eight genes from the IAP family and the gene regulating IAP-XAF1-in stem cells derived from human milk, using the qPCR method. The relationships between the expression of genes under study and clinical data, such as maternal age, maternal BMI, week of pregnancy in which the delivery took place, bodyweight of the newborn, the number of pregnancies and deliveries, and the time elapsed since delivery, were also analyzed. The research was carried out on samples of human milk collected from 42 patients hospitalized in The Clinic of Obstetrics and Perinatology of the Independent Public Clinical Hospital No. 4, in Lublin. The conducted research confirmed the expression of the following genes in the tested material: NAIP, BIRC2, BIRC3, BIRC5, BIRC6, BIRC8, XIAP, XAF1, OCT4 and SOX2. Moreover, several dependencies of the expression of individual genes on the maternal BMI (BIRC5, XAF1 and NAIP), the time since childbirth (BIRC5, BIRC6, XAF1 and NAIP), the number of pregnancies and deliveries (BIRC2, BIRC5, BIRC6 and XAF1), the manner of delivery (XAF1 and OCT4), preterm labor (BIRC6 and NAIP) were demonstrated. Additionally, we found positive relationships between gene expression of BIRC7, BIRC8 and XAF1 and the main factors of pluripotency: SOX2 and OCT4. This work is the first to investigate the expression of genes from the IAPs family in mother's milk stem cells.
Collapse
Affiliation(s)
- Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Michał Leśniewski
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Karolina Bieńko
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Monika Wójcik
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Marta Więckowska
- Student Scientific Society of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Dominika Przywara
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Alicja Petniak
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 11 Staszica Str., 20-081 Lublin, Poland
| | - Małgorzata Świstowska
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Rafał Szymanowski
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Agnieszka Wilińska
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Mateusz Wiliński
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Bartosz J. Płachno
- Department of Plant Cytology and Embryology, Institute of Botany, Faculty of Biology, Jagiellonian University in Kraków, 9 Gronostajowa St., 30-387 Cracow, Poland
| | - Marzena Kostuch
- Department of Neonatology, Independent Public Clinical Hospital No. 4, 8 Jaczewskiego St., 20-954 Lublin, Poland
| | - Mansur Rahnama-Hezavach
- Chair and Department of Dental Surgery, Medical University of Lublin, 6 Chodzki St., 20-093 Lublin, Poland
| | - Mariusz Szuta
- Chair of Oral Surgery, Jagiellonian University Medical College, 4 Montelupich St., 31-155 Kraków, Poland
| | - Anna Kwaśniewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 11 Staszica Str., 20-081 Lublin, Poland
| | - Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, 4a Chodźki St., 20–093 Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
- Correspondence:
| |
Collapse
|
22
|
Coni P, Piras M, Piludu M, Lachowicz JI, Matteddu A, Coni S, Reali A, Fanos V, Jaremko M, Faa G, Pichiri G. Exploring cell surface markers and cell-cell interactions of human breast milk stem cells. J Public Health Res 2023; 12:22799036221150332. [PMID: 36712902 PMCID: PMC9880586 DOI: 10.1177/22799036221150332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/22/2022] [Indexed: 01/26/2023] Open
Abstract
Background Breakthrough studies have shown that pluripotent stem cells are present in human breast milk. The expression of pluripotency markers by breast milk cells is heterogeneous, relating to cellular hierarchy, from early-stage multi-lineage stem cells to fully differentiated mammary epithelial cells, as well as weeks of gestation and days of lactation. Design and methods Here, we qualitatively analyze cell marker expression in freshly isolated human breast milk cells, without any manipulation that could influence protein expression. Moreover, we use electron microscopy to investigate cell-cell networks in breast milk for the first time, providing evidence of active intercellular communication between cells expressing different cellular markers. Results The immunocytochemistry results of human breast milk cells showed positive staining in all samples for CD44, CD45, CD133, and Ki67 markers. Variable positivity was present with P63, Tβ4 and CK14 markers. No immunostaining was detected for Wt1, nestin, Nanog, OCT4, SOX2, CK5, and CD34 markers. Cells isolated from human breast milk form intercellular connections, which together create a cell-to-cell communication network. Conclusions Cells freshly isolated form human breast milk, without particular manipulations, show heterogeneous expression of stemness markers. The studied milk staminal cells show "pluripotency" at different stages of differentiation, and are present as single cells or grouped cells. The adjacent cell interactions are evidenced by electron microscopy, which showed the formation of intercellular connections, numerous contact regions, and thin pseudopods.
Collapse
Affiliation(s)
- Pierpaolo Coni
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| | - Monica Piras
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| | - Marco Piludu
- Department of Biomedical Sciences,
University of Cagliari, Cagliari, Italy
| | - Joanna Izabela Lachowicz
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy,Joanna Izabela Lachowicz, Department of
Medical Sciences and Public Health, University of Cagliari, Cittadella
Universitaria, Monserrato, Cagliari 09048, Italy.
| | - Anna Matteddu
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| | - Stefano Coni
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| | - Alessandra Reali
- Azienda Ospedaliero Universitaria di
Cagliari, Terapia Intesiva Neonatale (TIN), P.O. Duilio Casula di Monserrato,
Cagliari, Italy
| | - Vassilios Fanos
- Department of Surgical Sciences,
University of Cagliari, Cagliari, Italy
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red
Sea Research Center (RSRC), Division of Biological and Environ-mental Sciences and
Engineering (BESE), King Abdullah University of Science and Technology (KAUST),
Thuwal, Saudi Arabia
| | - Gavino Faa
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| | - Giuseppina Pichiri
- Department of Medical Sciences and
Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
23
|
Dilemmas in initiation of very preterm infant enteral feeds-when, what, how? J Perinatol 2023; 43:108-113. [PMID: 36447040 DOI: 10.1038/s41372-022-01564-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
With limited clinical evidence available to guide common nutritional decisions, significant variation exists in approaches to enteral feeding for very preterm infants, specifically when feedings are initiated, what is fed, and the method used for feedings. Preclinical studies have highlighted the benefits associated with avoiding nil per os and providing early-stage mother's own milk or colostrum. However, these recommended approaches are often mutually exclusive due to the delays in lactation associated with very preterm delivery, resulting in uncertainty regarding which approach should be prioritized. Few studies have evaluated feeding frequency in preterm infants, with limited generalizability to extremely preterm infants. Therefore, even evidence-based approaches to very preterm infant feed initiation can differ. Future research is needed to identify optimal strategies for enteral nutrition in very preterm infants, but, until then, evidence-informed approaches may vary depending on each neonatal intensive care unit's assessment of risk and benefit.
Collapse
|
24
|
Ahmadzai H, Tee LBG, Crowe A. Are active efflux transporters contributing to infant drug exposure via breastmilk? A longitudinal study. Basic Clin Pharmacol Toxicol 2022; 131:487-499. [PMID: 36130042 PMCID: PMC9827846 DOI: 10.1111/bcpt.13794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 01/12/2023]
Abstract
Although most drugs are considered safe and compatible with breastfeeding, cases of toxic drug exposure have been reported. Active efflux transporters have been implicated as a mechanism in the transfer of drugs from mother to baby via breastmilk. Using breastmilk as a source of human mammary epithelial cells, this novel longitudinal study investigated the expression of four active transporters, namely, MDR1, MRP1, MRP2 and BCRP in the lactating human breast. BCRP gene was found to be strongly overexpressed with levels peaking at 5 months postpartum, potentially indicating a time where a breastfed infant may be at risk of inadvertent exposure to BCRP substrates. Serum albumin, a major component of human breastmilk was increasingly downregulated as lactation progresses. Xanthine oxidase/dehydrogenase, an enzyme in breastmilk attributed to a reduced risk of gastroenteritis caused by Escherichia coli and Salmonella enteritides, was downregulated. Lysozyme and fatty acid synthase are progressively upregulated. This study also shows that breastmilk-derived epithelial cells, when propagated in culture, exhibit characteristics significantly different to those derived directly from breastmilk. This serves to warn that in vitro studies are not a true representation of in vivo processes in the lactating breast; hence, application of in vitro data should be conducted with caution.
Collapse
Affiliation(s)
- Hilai Ahmadzai
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia,Pharmacy DepartmentSir Charles Gairdner HospitalNedlandsWestern AustraliaAustralia
| | - Lisa B. G. Tee
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - Andrew Crowe
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| |
Collapse
|
25
|
Pooled evidence from preclinical and clinical studies for stem cell-based therapy in ARDS and COVID-19. Mol Cell Biochem 2022; 478:1487-1518. [PMID: 36394787 DOI: 10.1007/s11010-022-04601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022]
|
26
|
Doerfler R, Melamed JR, Whitehead KA. The Effect of Infant Gastric Digestion on Human Maternal Milk Cells. Mol Nutr Food Res 2022; 66:e2200090. [PMID: 35984112 PMCID: PMC9532377 DOI: 10.1002/mnfr.202200090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/24/2022] [Indexed: 12/30/2022]
Abstract
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.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Jilian R. Melamed
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Kathryn A. Whitehead
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA,Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| |
Collapse
|
27
|
Evaluation of the Impact of Pregnancy-Associated Factors on the Quality of Wharton's Jelly-Derived Stem Cells Using SOX2 Gene Expression as a Marker. Int J Mol Sci 2022; 23:ijms23147630. [PMID: 35886978 PMCID: PMC9317592 DOI: 10.3390/ijms23147630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 12/04/2022] Open
Abstract
SOX2 is a recognized pluripotent transcription factor involved in stem cell homeostasis, self-renewal and reprogramming. It belongs to, one of the SRY-related HMG-box (SOX) family of transcription factors, taking part in the regulation of embryonic development and determination of cell fate. Among other functions, SOX2 promotes proliferation, survival, invasion, metastasis, cancer stemness, and drug resistance. SOX2 interacts with other transcription factors in multiple signaling pathways to control growth and survival. The aim of the study was to determine the effect of a parturient’s age, umbilical cord blood pH and length of pregnancy on the quality of stem cells derived from Wharton’s jelly (WJSC) by looking at birth weight and using SOX2 gene expression as a marker. Using qPCR the authors, evaluated the expression of SOX2 in WJSC acquired from the umbilical cords of 30 women right after the delivery. The results showed a significant correlation between the birth weight and the expression of SOX2 in WJSC in relation to maternal age, umbilical cord blood pH, and the length of pregnancy. The authors observed that the younger the woman and the lower the umbilical cord blood pH, the earlier the delivery occurs, the lower the birth weight and the higher SOX2 gene expression in WJSC. In research studies and clinical applications of regenerative medicine utilizing mesenchymal stem cells derived from Wharton’s Jelly of the umbilical cord, assessment of maternal and embryonic factors influencing the quality of cells is critical.
Collapse
|
28
|
Gleeson JP, Chaudhary N, Fein KC, Doerfler R, Hredzak-Showalter P, Whitehead KA. Profiling of mature-stage human breast milk cells identifies six unique lactocyte subpopulations. SCIENCE ADVANCES 2022; 8:eabm6865. [PMID: 35767604 PMCID: PMC9242445 DOI: 10.1126/sciadv.abm6865] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Breast milk is chock-full of nutrients, immunological factors, and cells that aid infant development. Maternal cells are the least studied breast milk component, and their unique properties are difficult to identify using traditional techniques. Here, we characterized the cells in mature-stage breast milk from healthy donors at the protein, gene, and transcriptome levels. Holistic analysis of flow cytometry, quantitative polymerase chain reaction, and single-cell RNA sequencing data identified the predominant cell population as epithelial with smaller populations of macrophages and T cells. Two percent of epithelial cells expressed four stem cell markers: SOX2, TRA-1-60, NANOG, and SSEA4. Furthermore, milk contained six distinct epithelial lactocyte subpopulations, including three previously unidentified subpopulations programmed toward mucosal defense and intestinal development. Pseudotime analysis delineated the differentiation pathways of epithelial progenitors. Together, these data define healthy human maternal breast milk cells and provide a basis for their application in maternal and infant medicine.
Collapse
Affiliation(s)
- John P. Gleeson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Katherine C. Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
29
|
Avellar ACDS, Oliveira MN, Caixeta F, Souza RCVE, Teixeira A, Faria AMC, Silveira-Nunes G, Faria ES, Maioli TU. Gestational Diabetes Mellitus Changes Human Colostrum Immune Composition. Front Immunol 2022; 13:910807. [PMID: 35795656 PMCID: PMC9251182 DOI: 10.3389/fimmu.2022.910807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Breast milk is considered a complete food for babies. Up to 7 days postpartum, it is known as colostrum, rich in immunological compounds, responsible for providing nutrition and ensuring immune protection. However, some maternal factors, such as gestational diabetes mellitus (GDM), can change the concentration of bioactive compounds present in the colostrum and may affect the development of the newborn’s immune system. The effect of GDM on colostrum cytokine, chemokine, and growth factors is not well described. Thus, the present study evaluated whether the occurrence of GDM changes the concentration of biomarkers in the colostrum. A cross-sectional study was carried out on postpartum women who had healthy pregnancies and women who had been diagnosed with GDM. A sample of colostrum was collected for Luminex analysis. Our results showed that GDM mothers had higher secretion of cytokines and chemokines in the colostrum, with a higher concentration of IFN-g, IL-6, and IL-15, and a lower concentration of IL-1ra. Among growth factors, we identified a decreased concentration of GM-CSF in the colostrum of GDM mothers. Thus, the data obtained support the idea that the disease leads to immune alterations in the colostrum.
Collapse
Affiliation(s)
- Ana Carolina de Sena Avellar
- Departamento de Nutrição, Programa de Pós-graduação em Nutrição e Saúde, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana Naves Oliveira
- Departamento de Nutrição, Programa de Pós-graduação em Nutrição e Saúde, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Hospital Sofia Feldman, Belo Horizonte, Brazil
| | - Felipe Caixeta
- Programa Interunidades de Pós-Graduação em Bioinformática, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rafaela Cristina Vieira e Souza
- Departamento de Nutrição, Programa de Pós-graduação em Nutrição e Saúde, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andréa Teixeira
- Instituto de Pesquisa Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Tatiani Uceli Maioli
- Departamento de Nutrição, Programa de Pós-graduação em Nutrição e Saúde, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Tatiani Uceli Maioli,
| |
Collapse
|
30
|
Nyquist SK, Gao P, Haining TKJ, Retchin MR, Golan Y, Drake RS, Kolb K, Mead BE, Ahituv N, Martinez ME, Shalek AK, Berger B, Goods BA. Cellular and transcriptional diversity over the course of human lactation. Proc Natl Acad Sci U S A 2022; 119:e2121720119. [PMID: 35377806 PMCID: PMC9169737 DOI: 10.1073/pnas.2121720119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
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 maternal–infant 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.
Collapse
Affiliation(s)
- Sarah K. Nyquist
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Computer Science and Artificial Intelligence Laboratory, Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Patricia Gao
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Tessa K. J. Haining
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Michael R. Retchin
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yarden Golan
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California, San Francisco, CA 94143
| | - Riley S. Drake
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
| | - Kellie Kolb
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Benjamin E. Mead
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California, San Francisco, CA 94143
| | | | - Alex K. Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Program in Computational and Systems Biology, Massachusetts Institute of Technology; Cambridge, MA 02139
- Department of Chemistry and Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Division of Health Science & Technology, Harvard Medical School, Boston, MA 02115
- Department of Immunology, Massachusetts General Hospital, Boston, MA 02114
| | - Bonnie Berger
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
- Computer Science and Artificial Intelligence Laboratory, Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brittany A. Goods
- Thayer School of Engineering, Program in Quantitative Biomedical Sciences, Dartmouth College, Hanover, NH 03755
| |
Collapse
|
31
|
Zhang L, Zhou Q, Li S, Han J, Yang Y, Lee SK, Kakulas F, Cao Y. Safety and Feasibility of Using Cell-Enriched Human Milk in the Treatment of Early Necrotizing Enterocolitis. Breastfeed Med 2022; 17:326-330. [PMID: 34935478 DOI: 10.1089/bfm.2021.0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background: Necrotizing enterocolitis (NEC) is a severe intestinal disease that often occurs in preterm infants, and there currently is a lack of specific and effective therapy. Human milk is rich in cells that may become a potential NEC treatment. Research Aim: To evaluate the safety and feasibility of cell-enriched fresh human milk treatment for premature infants with stage I NEC. Materials and Methods: Infants born at <1,500 g birth weight who developed stage I NEC were enrolled. Along with routine treatment for these infants, those in the intervention group were treated with cell-enriched fresh mother's milk (1 mL/kg) once per day for seven consecutive days. The intervention feasibility and safety were monitored and evaluated as primary outcomes. Short-term outcomes, including the duration of antibiotics, days to full enteral feeding and prognosis, were investigated as secondary outcomes. Results: Forty infants were enrolled, and 20 infants were included in each group. The demographic characteristics of the two groups of infants were comparable. All infants in the intervention group completed cell-enriched fresh mother's milk feeding for 7 days without any adverse clinical issues. The infants' vital signs were within the normal range during and after the intervention. None of the enrolled patients progressed to stage II NEC or above. The time interval from milk pumping to feeding was 3.7 ± 0.5 hours. Conclusions: Using cell-enriched fresh mother's milk to treat premature infants with stage I NEC was safe and feasible.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Qi Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Shujuan Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Junyan Han
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Yang
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China.,Pediatric Research Institute, Children's Hospital of Fudan University, Shanghai, China
| | - Shoo K Lee
- Department of Pediatrics, Maternal-Infant Care Research Centre, Mount Sinai Hospital, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada.,Department of Obstetrics and Gynecology, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Foteini Kakulas
- Pathology and Laboratory Medicine, Medical School, The University of Western Australia, Crawley, Australia
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China.,NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| |
Collapse
|
32
|
de Weerth C, Aatsinki AK, Azad MB, Bartol FF, Bode L, Collado MC, Dettmer AM, Field CJ, Guilfoyle M, Hinde K, Korosi A, Lustermans H, Mohd Shukri NH, Moore SE, Pundir S, Rodriguez JM, Slupsky CM, Turner S, van Goudoever JB, Ziomkiewicz A, Beijers R. Human milk: From complex tailored nutrition to bioactive impact on child cognition and behavior. Crit Rev Food Sci Nutr 2022; 63:7945-7982. [PMID: 35352583 DOI: 10.1080/10408398.2022.2053058] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human milk is a highly complex liquid food tailor-made to match an infant's needs. Beyond documented positive effects of breastfeeding on infant and maternal health, there is increasing evidence that milk constituents also impact child neurodevelopment. Non-nutrient milk bioactives would contribute to the (long-term) development of child cognition and behavior, a process termed 'Lactocrine Programming'. In this review we discuss the current state of the field on human milk composition and its links with child cognitive and behavioral development. To promote state-of-the-art methodologies and designs that facilitate data pooling and meta-analytic endeavors, we present detailed recommendations and best practices for future studies. Finally, we determine important scientific gaps that need to be filled to advance the field, and discuss innovative directions for future research. Unveiling the mechanisms underlying the links between human milk and child cognition and behavior will deepen our understanding of the broad functions of this complex liquid food, as well as provide necessary information for designing future interventions.
Collapse
Affiliation(s)
- Carolina de Weerth
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Anna-Katariina Aatsinki
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Frank F Bartol
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama, USA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California San Diego, La Jolla, California, USA
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Amanda M Dettmer
- Yale Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, College of Basic and Applied Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Meagan Guilfoyle
- Department of Anthropology, Indiana University, Bloomington, Indiana, USA
| | - Katie Hinde
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Brain Plasticity group, University of Amsterdam, Amsterdam, The Netherlands
| | - Hellen Lustermans
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
| | - Nurul Husna Mohd Shukri
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Sophie E Moore
- Department of Women & Children's Health, King's College London, St Thomas' Hospital, London, UK
- School of Hygiene and Tropical Medicine, Nutrition Theme, MRC Unit The Gambia and the London, Fajara, The GambiaBanjul
| | - Shikha Pundir
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Carolyn M Slupsky
- Department of Nutrition and Department of Food Science and Technology, University of California, Davis, California, USA
| | - Sarah Turner
- Department of Community Health Sciences, Manitoba Interdisciplinary Lactation Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Johannes B van Goudoever
- Department of Pediatrics, Amsterdam UMC, University of Amsterdam, Vrije Universiteit, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Anna Ziomkiewicz
- Department of Anthropology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Roseriet Beijers
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Medical Center, EN Nijmegen, The Netherlands
- Department of Social Development, Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Papoz A, Clément F, Laporte C, Tubbs E, Gidrol X, Pitaval A. [Generating pancreatic islets organoids: Langerhanoids]. Med Sci (Paris) 2022; 38:52-58. [PMID: 35060887 DOI: 10.1051/medsci/2021244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The extension of islet transplantation to a wider number of Type 1 diabetic patients is compromised by the scarcity of donors, the reduced ex vivo survival of pancreatic islets and the use of immunosuppressive treatments. Islets of Langerhans isolated from brain-dead donors are currently the only cell source for transplantation. Thus, it is crucial to find an alternative and an abundant source of functional insulin secreting cells not only for clinical use but also for the development of research dedicated to the screening of drugs and to the development of new therapeutic targets. Several groups around the world, including ours, develop 3D culture models as Langerhanoids that closely mimick human pancreatic islets physiology. In this review, we describe recent advances to mimic the pancreatic niche (extracellular matrix, vascularization, microfluidics) allowing better functionality of Langerhanoids.
Collapse
Affiliation(s)
- Anastasia Papoz
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Flora Clément
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Camille Laporte
- Univ. Grenoble Alpes, CEA, Leti, Division for biology and healthcare technologies, Microfluidic systems and bioengineering Lab, F-38000, Grenoble, France
| | - Emily Tubbs
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France - Univ. Grenoble Alpes, LBFA et BEeSy, Inserm U1055, F-38000, Grenoble, France
| | - Xavier Gidrol
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Amandine Pitaval
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| |
Collapse
|
34
|
Zhang T, Zhou H, Wang K, Wang X, Wang M, Zhao W, Xi X, Li Y, Cai M, Zhao W, Xu Y, Shao R. Role, molecular mechanism and the potential target of breast cancer stem cells in breast cancer development. Biomed Pharmacother 2022; 147:112616. [PMID: 35008001 DOI: 10.1016/j.biopha.2022.112616] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/01/2022] [Accepted: 01/02/2022] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is one of the most common malignant tumors in women globally, and its occurrence has surpassed lung cancer and become the biggest threat for women. At present, breast cancer treatment includes surgical resection or postoperative chemotherapy and radiotherapy. However, tumor relapse and metastasis usually lead to current therapy failure thanks to breast cancer stem cells (BCSCs)-mediated tumorigenicity and drug resistance. Drug resistance is mainly due to the long-term quiescent G0 phase, strong DNA repairability, and high expression of ABC transporter, and the tumorigenicity is reflected in the activation of various proliferation pathways related to BCSCs. Therefore, understanding the characteristics of BCSCs and their intracellular and extracellular molecular mechanisms is crucial for the development of targeted drugs for BCSCs. To this end, we discussed the latest developments in BCSCs research, focusing on the analysis of specific markers, critical signaling pathways that maintain the stemness of BCSCs,such as NOTCH, Wnt/β-catenin, STAT3, Hedgehog, and Hippo-YAP signaling, immunomicroenviroment and summarizes targeting therapy strategies for stemness maintenance and differentiation, which provides a theoretical basis for further exploration of treating breast cancer and preventing relapse derived from BCSCs.
Collapse
Affiliation(s)
- Tianshu Zhang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Huimin Zhou
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kexin Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaowei Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mengyan Wang
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wenxia Zhao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaoming Xi
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yang Li
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meilian Cai
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wuli Zhao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanni Xu
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering, Ministry of Health, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
35
|
Afify SM, Hassan G, Ishii H, Monzur S, Nawara HM, Osman A, Abu Quora HA, Sheta M, Zahra MH, Seno A, Seno M. Functional and Molecular Characters of Cancer Stem Cells Through Development to Establishment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:83-101. [PMID: 36587303 DOI: 10.1007/978-3-031-12974-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cancer stem cells (CSCs) are small subpopulation sharing similar properties like normal stem such as self-renewal and differentiation potential to direct tumor growth. Last few years, scientists considered CSCs as the cause of phenotypic heterogeneity in diverse cancer types. Also, CSCs contribute to cancer metastasis and recurrence. The cellular and molecular regulators influence on the CSCs' phenotype changing their behaviors in different stages of cancer progression. CSC markers play significance roles in cancer diagnosis and characterization. We delineate the cross-talks between CSCs and the tumor microenvironment that supports their intrinsic properties including survival, stemness, quiescence and their cellular and molecular adaptation. An insight into the markers of CSCs specific to organs is described.
Collapse
Affiliation(s)
- Said M Afify
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
- Faculty of Science, Division of Biochemistry, Chemistry Department, Menoufia University, Shebin El Koum, 32511, Egypt
| | - Ghmkin Hassan
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
- Faculty of Pharmacy, Department of Microbiology and Biochemistry, Damascus University, Damascus, 10769, Syria
| | - Hiroko Ishii
- GSP Enterprise, Inc, 1-4-38 12F Minato-Machi, Naniwaku, Osaka, 556-0017, Japan
| | - Sadia Monzur
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Hend M Nawara
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Amira Osman
- Faculty of Medicine, Department of Histology, Kafr Elsheikh University, Kafr Elsheikh, 33511, Egypt
| | - Hagar A Abu Quora
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
- Faculty of Science, Cytology, Histology and Histochemistry, Zoology Department, Menoufia University, Menoufia, 32511, Egypt
| | - Mona Sheta
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Maram H Zahra
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Akimasa Seno
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan
| | - Masaharu Seno
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, 700-8530, Japan.
| |
Collapse
|
36
|
Hong L, Zhang L, Zhou Q, Li S, Han J, Jiang S, Han X, Yang Y, Hong S, Cao Y. Impacts of Enriched Human Milk Cells on Fecal Metabolome and Gut Microbiome of Premature Infants with Stage I Necrotizing Enterocolitis: A Pilot Study. Mol Nutr Food Res 2021; 66:e2100342. [PMID: 34788490 DOI: 10.1002/mnfr.202100342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 10/08/2021] [Indexed: 11/07/2022]
Abstract
SCOPE Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in preterm infants, occurring more often in formula-fed infants than in breastfed infants. Recent animal studies have shown that cells in fresh breast milk survive in the newborns' digestive tract. However, no clinical studies have been conducted on the effects of human milk cells, and their biological roles in the infants' intestines remain unexplored. METHODS AND RESULTS Twenty premature infants are enrolled. Cells from fresh milk of their own mothers are enriched and fed to infants with Bell's Stage I NEC once a day for 7 days since the onset of NEC. Fecal samples are collected at enrollment and 2 weeks later. Fecal sphingolipids are observed to be enriched in NEC patients and positively correlated with calprotectin levels. After intervention with enriched human milk cells, inflammation-associated sphingolipids and microbiome profiles are altered and resembled those of the controls. CONCLUSION These preliminary findings reveal the potential impacts of enriched human milk cells on premature infants with Bell's Stage I NEC and provide insight into the roles of fecal sphingolipid metabolism in the neonates' intestinal inflammation. However, the limited sample size of the study indicates the need for further investigation.
Collapse
Affiliation(s)
- Luyang Hong
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Lan Zhang
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Qi Zhou
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Shujuan Li
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Junyan Han
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Siyuan Jiang
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China
| | - Xiao Han
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, 200032, China
| | - Yi Yang
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, 200032, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, 200032, China.,NHC Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, 200032, China
| |
Collapse
|
37
|
Geddes DT, Gridneva Z, Perrella SL, Mitoulas LR, Kent JC, Stinson LF, Lai CT, Sakalidis V, Twigger AJ, Hartmann PE. 25 Years of Research in Human Lactation: From Discovery to Translation. Nutrients 2021; 13:3071. [PMID: 34578947 PMCID: PMC8465002 DOI: 10.3390/nu13093071] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Researchers have recently called for human lactation research to be conceptualized as a biological framework where maternal and infant factors impacting human milk, in terms of composition, volume and energy content are studied along with relationships to infant growth, development and health. This approach allows for the development of evidence-based interventions that are more likely to support breastfeeding and lactation in pursuit of global breastfeeding goals. Here we summarize the seminal findings of our research programme using a biological systems approach traversing breast anatomy, milk secretion, physiology of milk removal with respect to breastfeeding and expression, milk composition and infant intake, and infant gastric emptying, culminating in the exploration of relationships with infant growth, development of body composition, and health. This approach has allowed the translation of the findings with respect to education, and clinical practice. It also sets a foundation for improved study design for future investigations in human lactation.
Collapse
Affiliation(s)
- Donna Tracy Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Zoya Gridneva
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Sharon Lisa Perrella
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Leon Robert Mitoulas
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
- Medela, AG, Lättichstrasse 4b, 6340 Baar, Switzerland
| | - Jacqueline Coral Kent
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Lisa Faye Stinson
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Ching Tat Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | - Vanessa Sakalidis
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| | | | - Peter Edwin Hartmann
- School of Molecular Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (Z.G.); (S.L.P.); (L.R.M.); (J.C.K.); (L.F.S.); (C.T.L.); (V.S.); (P.E.H.)
| |
Collapse
|
38
|
Khamis T, Abdelalim AF, Saeed AA, Edress NM, Nafea A, Ebian HF, Algendy R, Hendawy DM, Arisha AH, Abdallah SH. Breast milk MSCs upregulated β-cells PDX1, Ngn3, and PCNA expression via remodeling ER stress /inflammatory /apoptotic signaling pathways in type 1 diabetic rats. Eur J Pharmacol 2021; 905:174188. [PMID: 34004210 DOI: 10.1016/j.ejphar.2021.174188] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/30/2021] [Accepted: 05/12/2021] [Indexed: 01/01/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is one of the autoimmune diseases characterized by beta-cell dysfunction with serious health complications. Br-MSCs represent a novel valid candidate in regenerative medicine disciplines. Yet, the full potential of Br-MSCs in managing type 1 diabetes remains elusive. Indeed, this study was designed to explore a novel approach investigating the possible regenerative capacity of Br-MSCs in type1 diabetic islet on the level of the cellular mRNA expression of different molecular pathways involved in pancreatic beta-cell dysfunction. Sixty adult male Sprague-Dawley rats were randomly assigned into 3 groups (20 rats each); the control group, type1 diabetic group, and the type 1 diabetic Br-MSCs treated group. And, for the first time, our results revealed that intraperitoneally transplanted Br-MSCs homed to the diabetic islet and improved fasting blood glucose, serum insulin level, pancreatic oxidative stress, upregulated pancreatic mRNA expression for: regenerative markers (Pdx1, Ngn3, PCNA), INS, beta-cell receptors (IRS1, IRβ, PPARγ), pancreatic growth factors (IGF-1, VEGFβ1, FGFβ), anti-inflammatory cytokine (IL10) and anti-apoptotic marker (BCL2) too, Br-MSCs downregulated pancreatic mRNA expression for: inflammatory markers (NFKβ, TNFα, IL1β, IL6, IL8, MCP1), apoptotic markers for both intrinsic and extrinsic pathways (FAS, FAS-L, P53, P38, BAX, Caspase3), ER stress markers (ATF6, ATF3, ATF4, BIP, CHOP, JNK, XBP1) and autophagy inhibitor (mTOR). In conclusion, Br-MSCs could be considered as a new insight in beta cell regenerative therapy improving the deteriorated diabetic islet microenvironment via modulating; ER stress, inflammatory, and apoptotic signaling pathways besides, switching on the cellular quality control system (autophagy) thus enhancing beta-cell function.
Collapse
Affiliation(s)
- Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt.
| | - Abdelalim F Abdelalim
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Ahmed A Saeed
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Nagah M Edress
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Alaa Nafea
- Department of Pediatrics, Faculty of Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Huda F Ebian
- Department of Clinical Pathology, Faculty of Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Reem Algendy
- Department of Milk Hygiene, Food Control Department, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Doaa M Hendawy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, 44511, Zagazig, Egypt
| | - Ahmed Hamed Arisha
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Cairo, Egypt; Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt.
| | - Somia Hassan Abdallah
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, 44511, Zagazig, Egypt
| |
Collapse
|
39
|
Silva MDB, de Oliveira RDVC, Alves DDSB, Melo ECP. The effect of risk at birth on breastfeeding duration and exclusivity: A cohort study at a Brazilian referral center for high-risk neonates and infants. PLoS One 2021; 16:e0255190. [PMID: 34358227 PMCID: PMC8346259 DOI: 10.1371/journal.pone.0255190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/11/2021] [Indexed: 11/23/2022] Open
Abstract
Background and aim Both breastfeeding and the use of human milk are strategies that provide better conformation to health throughout an individual’s life and bring countless short- and long- term benefits, which are well established in the scientific literature. For at-risk newborns (NBs), these strategies are crucial interventions to enable neonatal survival with better quality of life due to the distinctive and complex composition of human milk, which serves as personalized food-medicine-protection. However, there is limited knowledge about breastfeeding practices in high-risk NBs. The aim was to estimate the duration of EBF and to investigate the effect of risk at birth on EBF discontinuity in the first six months of life’. Methods This cohort study included 1,003 NBs from a high-risk referral center, followed up from birth to the sixth month of life, between 2017 and 2018. Correspondence and cluster analysis was used to identify neonatal risk clusters as the main exposure. The object of interest was the time until EBF discontinuity. The Kaplan-Meier methods and the Cox proportional hazards model were used to estimate the hazard ratio and 95% confidence intervals. Results The prevalence and median duration of EBF decreased proportionally in the three groups. The multiple model revealed a gradient in EBF discontinuity, which was 40% higher in risk group 1 and 111% higher in risk group 2 compared to healthy full-term NBs. Additionally, EBF during hospitalization predicted a longer median duration of this practice for high-risk NBs. Conclusion This study confirms a high proportion of high-risk NBs who have EBF discontinued before six months of life. The risk of EBF discontinuity is higher in risk groups, with a gradual effect even when adjusted by several factors. Effective interventions are needed to promote, protect, and support breastfeeding in different profiles of risk-at-birth groups.
Collapse
Affiliation(s)
- Maíra Domingues Bernardes Silva
- Human Milk Bank at the National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF) of the Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, RJ, Brazil
- * E-mail:
| | | | | | | |
Collapse
|
40
|
Transdifferentiation of goat ear fibroblasts into lactating mammary epithelial cells induced by small molecule compounds. Biochem Biophys Res Commun 2021; 573:55-61. [PMID: 34388455 DOI: 10.1016/j.bbrc.2021.07.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/26/2021] [Indexed: 02/03/2023]
Abstract
Mammary epithelial cells are the only cells in the mammary glands that are capable of lactation and they are ideal for studying cellular and molecular biology mechanisms during growth, development and lactation of the mammary glands. The limiting factors in most of the currently available mammary epithelial cells are low cell viability, transgenerational efficiency and lactation function that renders them unsuitable for subsequent studies on mammary gland's cellular and lactation mechanisms and utilizing them as bioreactors. Hence, new methods are required to obtain mammary epithelial cells with high transgenerational efficiency and lactation function. In this study, transdifferentiation of goat ear fibroblasts (GEFs) into goat mammary epithelial cells (CiMECs) was induced in only eight days by five small molecule compounds, including 500 μg/mL VPA, 10 μM Tranylcypromine, 10 μM Forskolin, 1 μM TTNPB, 10 μM RepSox. Morphological observation, marker genes comparison, specific antigen expression and comparison of gene expression levels by transcriptome sequencing between the two types of cells that led to the primary deduction that CiMECs have similar biological properties to goat mammary epithelial cells (GMECs) and comparatively more lactation capacity. Therefore, we establish a novel reprogramming route to convert fibroblasts into CiMECs under fully chemically conditions. This study is expected to provide an in vitro platform for understanding cellular mechanisms such as mammary epithelial cells' fate determination and developmental differentiation, and also to find a new way to obtain a large number of functional mammary epithelial cells in vitro.
Collapse
|
41
|
Miliku K, Moraes TJ, Becker AB, Mandhane PJ, Sears MR, Turvey SE, Subbarao P, Azad MB. Breastfeeding in the First Days of Life Is Associated With Lower Blood Pressure at 3 Years of Age. J Am Heart Assoc 2021; 10:e019067. [PMID: 34284597 PMCID: PMC8475685 DOI: 10.1161/jaha.120.019067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background Breastfeeding in infancy is associated with lower cardiovascular disease risk in adulthood; however, the amount of breastfeeding required to achieve this benefit is unknown. Methods and Results In the CHILD (Canadian Healthy Infant Longitudinal Development) Cohort Study, we analyzed 2382 children with complete data on early life feeding and blood pressure. Infant feeding was documented from hospital records in the first few days of life and reported by mothers throughout infancy. Blood pressure was measured at 3 years of age. Analyses controlled for birth weight, gestational age, socioeconomic status, maternal body mass index, and other potential confounders. We found that nearly all children (2333/2382; 97.9%) were ever breastfed, of whom 98 (4.2%) only briefly received breast milk during their birth hospitalization (“early limited breastfeeding”). At 3 years of age, blood pressure was higher in children who were never breastfed (mean systolic/diastolic 103/60 mm Hg) compared with those who were ever breastfed (99/58 mm Hg), including those who received only early limited breastfeeding (99/57 mm Hg). These differences in systolic blood pressure persisted in adjusted models (ever breastfed: −3.47 mm Hg, 95% CI, −6.14 to −0.80; early limited breastfeeding: −4.24 mm Hg, 95% CI, −7.45 to −1.04). Among breastfed children, there was no significant dose‐response association according to the duration or exclusivity of breastfeeding. Associations were not mediated by child body mass index. Conclusions Although the benefits of sustained and exclusive breastfeeding are indisputable, this study indicates any breastfeeding, regardless of duration or exclusivity, is associated with lower blood pressure at 3 years of age. Further research examining the bioactive components of early breast milk, underlying mechanisms, and long‐term associations is warranted.
Collapse
Affiliation(s)
- Kozeta Miliku
- Department of Medicine McMaster University Hamilton Canada.,Department of Pediatrics and Child Health University of Manitoba Winnipeg Canada.,Manitoba Interdisciplinary Lactation Centre (MILC) Children's Hospital Research Institute of Manitoba Winnipeg Canada
| | - Theo J Moraes
- Departments of Pediatrics Hospital for Sick Children University of Toronto Toronto Canada
| | - Allan B Becker
- Department of Pediatrics and Child Health University of Manitoba Winnipeg Canada
| | | | | | - Stuart E Turvey
- Department of Pediatrics University of British Columbia Vancouver Canada
| | - Padmaja Subbarao
- Departments of Pediatrics Hospital for Sick Children University of Toronto Toronto Canada
| | - Meghan B Azad
- Department of Pediatrics and Child Health University of Manitoba Winnipeg Canada.,Manitoba Interdisciplinary Lactation Centre (MILC) Children's Hospital Research Institute of Manitoba Winnipeg Canada
| |
Collapse
|
42
|
Kim YJ. Immunomodulatory Effects of Human Colostrum and Milk. Pediatr Gastroenterol Hepatol Nutr 2021; 24:337-345. [PMID: 34316468 PMCID: PMC8279828 DOI: 10.5223/pghn.2021.24.4.337] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023] Open
Abstract
The immune system is not fully developed in human neonates and infants; breastfeeding is important in this stage as the bioactive components of human breast milk are known to have anti-microbial, anti-inflammatory, and immunomodulatory effects, and can therefore contribute to an infant's immunity against allergies, asthma, autoimmune diseases, and inflammatory bowel disease. Herein, the positive effect on the immune system by human colostrum and milk are reviewed.
Collapse
Affiliation(s)
- Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
43
|
Rahmani-Moghadam E, Zarrin V, Mahmoodzadeh A, Owrang M, Talaei-Khozani T. Comparison of the Characteristics of Breast Milk-derived Stem Cells with the Stem Cells Derived from the Other Sources: A Comparative Review. Curr Stem Cell Res Ther 2021; 17:71-90. [PMID: 34161214 DOI: 10.2174/1574888x16666210622125309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/14/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Breast milk (BrM) not only supplies nutrition, but it also contains a diverse population of cells. It has been estimated that up to 6% of the cells in human milk possess the characteristics of mesenchymal stem cells (MSC). Available data also indicate that these cells are multipotent and capable of self-renewal and differentiation with other cells. In this review, we have compared different characteristics, such as CD markers, differentiation capacity, and morphology of stem cells, derived from human breast milk (hBr-MSC) with human bone marrow (hBMSC), Wharton's jelly (WJMSC), and human adipose tissue (hADMSC). Through the literature review, it was revealed that human breast milk-derived stem cells specifically express a group of cell surface markers, including CD14, CD31, CD45, and CD86. Importantly, a group of markers, CD13, CD29, CD44, CD105, CD106, CD146, and CD166, were identified, which were common in the four sources of stem cells. WJMSC, hBMSC, hADMSC, and hBr-MSC are potently able to differentiate into the mesoderm, ectoderm, and endoderm cell lineages. The ability of hBr-MSCs todifferentiate into the neural stem cells, neurons, adipocyte, hepatocyte, chondrocyte, osteocyte, and cardiomyocytes has made these cells a promising source of stem cells in regenerative medicine, while isolation of stem cells from the commonly used sources, such as bone marrow, requires invasive procedures. Although autologous breast milk-derived stem cells are an accessible source for women who are in the lactation period, breast milk can be considered as a source of stem cells with high differentiation potential without any ethical concern.
Collapse
Affiliation(s)
- Ebrahim Rahmani-Moghadam
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marzieh Owrang
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Talaei-Khozani
- Department of Anatomical sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
44
|
Kersin SG, Özek E. Breast milk stem cells: Are they magic bullets in neonatology? Turk Arch Pediatr 2021; 56:187-191. [PMID: 34104907 DOI: 10.5152/turkarchpediatr.2021.21006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/26/2021] [Indexed: 12/12/2022]
Abstract
Each mammal produces milk specific to its newborn that meets all nutritional needs. Breast milk is not only a secretory product but is also a complex liquid containing several components that provide enteral nutrition. The stage of lactation, the fullness of the breast, the feeding of the baby, and the health of the mother during the breastfeeding period cause differences in the composition of breast milk. Although the positive effects of breast milk on the physical and intellectual development of a child in the short and long term have been known for centuries, its mechanism has not been elucidated. Stem cells are defined as the cells that possess specific markers and have not undergone differentiation. Under suitable conditions and stimuli, they can differentiate into desired cells. The detection of stem cells, whose exact origin is not known, in breast milk and their demonstration in the baby's body have prompted the necessity of exploring the possible role of stem cells in the treatment of diseases. In this review, breast milk-derived stem cells and their possible role in neonatology are discussed.
Collapse
Affiliation(s)
- Sinem Gülcan Kersin
- Division of Neonatology, Department of Pediatrics, Marmara University School of Medicine, İstanbul, Turkey
| | - Eren Özek
- Division of Neonatology, Department of Pediatrics, Marmara University School of Medicine, İstanbul, Turkey
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Breast milk (BM) is a peculiar fluid owing unique properties and resulting the ideal food during early neonatal period. As widely known, it can improve the outcome of both neonate and lactating mother, influencing their whole life. BM is characterized by several beneficial components; among these, a great role is played by BM own and specific microbiome, deeply investigated in many studies. Moreover, the use of metabolomics in BM analysis allowed a better characterization of its metabolic pathways that vary according to lactation stage and neonatal gestational age. The aim of this review is to describe growth factors, cytokines, immunity mediators, and stem cells (SCs) contained in BM and investigate their functions and effects on neonatal outcome, especially focusing on immuno- and neurodevelopment. RECENT FINDINGS We evaluated recent and updated literature on this field. The article that we analyzed to write this review have been found in MEDLINE using breast milk-derived stem cells, biofactors, growth factors, breastfeeding-related outcomes, neurodevelopment, and neonatal immunological system as keywords. Discovering and characterizing BM components could result very useful to clarify the pathophysiology of their influence on neonatal growth and even to improve artificial formulations' composition. Moreover, since SCs abilities and their involvement in the development of several diseases, they could help to discover specific targets for new therapies. It could be useful to characterize BM-derived SC markers, properties, and variations during lactation stages, to understand their potential role in therapeutic applications, since they could be noninvasively isolated from BM. More studies will help to describe more in detail the characteristics of mother-to-child communication through breastfeeding and its potential role in the next future.
Collapse
|
46
|
Proteomic analysis of hypoxia and non-hypoxia secretome mesenchymal stem-like cells from human breastmilk. Saudi J Biol Sci 2021; 28:4399-4407. [PMID: 34354424 PMCID: PMC8324926 DOI: 10.1016/j.sjbs.2021.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction Breastmilk contains proteins and cells which have stem cell properties. The human breastmilk stem cell mimick mesenchymal stem cells and expresses pluripotency genes. The protein level of breastmilk is high in colostrum and gradually subsides in the first year of lactation. The mesenchymal stem cells from breastmilk can be an alternative source of stem cells that can potentially affect cardiovascular therapy. This study aimed to identify the proteomic analysis of secretome mesenchymal stem-like cells under hypoxia compared to non-hypoxia from human breastmilk stem cells. Material and methods The human breastmilk was collected from six healthy breastfeeding women and transported to the laboratory under aseptic conditions. The breastmilk cells were isolated then cultured. After 72 h, the human breastmilk stem cells reached confluence then cleaned up and isolated in serum-free media (spheroid) to allow serial passaging every 48 h. The acquisition stem cell was made with flow cytometry. The cells were divided into hBSC secretomes under hypoxia (A) and non-hypoxia (B) and analyzed for LC-MS to identify the peptide structure. Results The human breastmilk cells contained several mesenchymal stem-like cells in density 2.4 × 106 cell/mL for hypoxia and 2 × 106 cell/mL for non-hypoxia conditions. The human breastmilk stem cell surface markers derived from the third cell passage process were 93.77% for CD44, 98.69% for CD73, 88.45% for CD90, and 96.30% for CD105. The protein level of secretome mesenchymal stem -like cells under hypoxia was measured at 5.56 μg/mL and 4.28 μg/mL for non-hypoxia. The liquid chromatography-mass spectrometry analysis identified 130 and 59 peptides from hypoxia and non-hypoxia of the human breastmilk stem cell secretome sequentially. Some important proteomics structures were found in the hypoxic human breastmilk stem cell secretome, such as transforming growth factor-β, VE-cadherin, and caspase. Conclusion The human breastmilk cells contain mesenchymal stem-like cells and a high concentration of CD44, CD73, CD90, and CD105 as surface markers at third passage culture. The hypoxic hBSC secretome produces a higher protein level compare to non-hypoxia. The transforming growth factor -β was found in the hypoxic hBSC secretome as a modulator of VEGF-mediated angiogenesis.
Collapse
Key Words
- AFP, Alpha-Fetoprotein
- ATP, Adenosine Triphosphate
- BD, Becton Dickinson
- BMPR-II, Bone morphogenetic protein type II
- BSA, Bovine Serum Albumin
- EHD3, EH Domain-containing Protein 3
- FACS, Fluorescence-Activated Cell Sorting
- FBS, Fetal Bovine Serum
- HIF-1α, Hypoxia Inducible Factor-1α
- Hypoxia
- IGF1, Insulin-like Growth Factor 1
- LALBA, α-Lactalbumin
- LC-MS
- LC-MS, Liquid Chromatography-Mass Spectrometry
- LF, Lactoferrin
- MAPK, Mitogen-Activated Protein Kinase
- MPS, Multi Proliferative Supplement
- MPZL1, Myelin Protein Zero-like Protein 1
- MSC, Mesenchymal Stem Cell
- Mesenchymal stem-like cell
- PBS, Phosphate-buffered Saline
- SDS, Sodium Dodecyl Sulfate
- SMA, Smooth Muscle Actin
- SMAD, Signals Mothers Against the Decapentaplegic
- Secretome
- TGF-β, Transforming Growth Factor-Beta
- VEGF, Vascular Endothelial Growth Factor
- cDNA, complementary Deoxyribonucleic Acid
- hBSC
- hBSC, Human Breastmilk Stem Cell
- mRNA, messenger Ribonucleic Acid
Collapse
|
47
|
Martin Carli JF, Trahan GD, Rudolph MC. Resolving Human Lactation Heterogeneity Using Single Milk-Derived Cells, a Resource at the Ready. J Mammary Gland Biol Neoplasia 2021; 26:3-8. [PMID: 34097179 PMCID: PMC8956113 DOI: 10.1007/s10911-021-09489-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/02/2021] [Indexed: 10/21/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Jayne F Martin Carli
- Section of Nutrition, Denver Anschutz Medical Campus Department of Pediatrics, University of Colorado, Aurora, CO, 80045, USA.
| | - G Devon Trahan
- Section of Hematology, Oncology, and Bone Marrow Transplant, Denver Anschutz Medical Campus Department of Pediatrics, University of Colorado, Aurora, CO, 80045, USA
| | - Michael C Rudolph
- Department of Physiology, Oklahoma University Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
48
|
Witkowska-Zimny M, Majczyna D. How Knowledge about Stem Cells Influences Attitudes towards Breastfeeding: Case Study of Polish Women. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2382. [PMID: 33804414 PMCID: PMC7967762 DOI: 10.3390/ijerph18052382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 11/22/2022]
Abstract
Maternal breastfeeding is associated with multiple health benefits, both for the child and the mother. Since breastfeeding rates are declining, finding new, future-oriented strategies to strengthen and support mothers' positive attitudes towards breastfeeding need to be developed. In this paper, we describe how information about the presence of stem cells in breast milk can influence the willingness to breastfeed in the population of Polish pregnant women. A cross-sectional study involving a group of 150 pregnant women was conducted to assess the correlation between their knowledge about stem cells and their attitude towards breastfeeding. Among the respondents, only 6% claimed that they did not know anything about stem cells, but general knowledge about stem cells in the research group was poor. The survey results indicated that city residence, university degree, maternal experience and advanced pregnancy correlated with higher general knowledge regarding stem cells. Most respondents (77.3%) had no knowledge regarding the presence of stem cells in breast milk. Approximately two-thirds of mothers with earlier negative breastfeeding experience declared that information about the presence of stem cells in breast milk could have influenced the decision to continue and extend the time of breastfeeding. Hence highlighting the presence of stem cells in breast milk can be used to encourage breastfeeding as a unique activity.
Collapse
Affiliation(s)
- Malgorzata Witkowska-Zimny
- Department of Biophysics and Human Physiology, Medical University of Warsaw, Zwirki i Wigury 61 St, 02-091 Warsaw, Poland;
| | | |
Collapse
|
49
|
The Gut‒Breast Axis: Programming Health for Life. Nutrients 2021; 13:nu13020606. [PMID: 33673254 PMCID: PMC7917897 DOI: 10.3390/nu13020606] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The gut is a pivotal organ in health and disease. The events that take place in the gut during early life contribute to the programming, shaping and tuning of distant organs, having lifelong consequences. In this context, the maternal gut plays a quintessence in programming the mammary gland to face the nutritional, microbiological, immunological, and neuroendocrine requirements of the growing infant. Subsequently, human colostrum and milk provides the infant with an impressive array of nutrients and bioactive components, including microbes, immune cells, and stem cells. Therefore, the axis linking the maternal gut, the breast, and the infant gut seems crucial for a correct infant growth and development. The aim of this article is not to perform a systematic review of the human milk components but to provide an insight of their extremely complex interactions, which render human milk a unique functional food and explain why this biological fluid still truly remains as a scientific enigma.
Collapse
|
50
|
Pan TD, Kanaan SB, Lee NR, Avila JL, Nelson JL, Eisenberg DTA. Predictors of maternal-origin microchimerism in young women in the Philippines. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021; 174:213-223. [PMID: 33300155 PMCID: PMC11753296 DOI: 10.1002/ajpa.24191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/17/2020] [Accepted: 11/09/2020] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Microchimerism is the presence of a small quantity of cells or DNA from a genetically distinct individual. This phenomenon occurs with bidirectional maternal-fetal exchange during pregnancy. Microchimerism can persist for decades after delivery and have long-term health implications. However, little is known about why microchimerism is detectable at varying levels in different individuals. We examine the variability and the following potential determinants of maternal-origin microchimerism (MMc) in young women in the Philippines: gestational duration (in utero exposure to MMc), history of being breastfed (postpartum exposure to MMc), maternal telomere length (maternal cells' ability to replicate and persist), and participant's pregnancies in young adulthood (effect of adding fetal-origin microchimerism to preexisting MMc). MATERIALS AND METHODS Data are from the Cebu Longitudinal Health and Nutrition Survey, a population-based study of infant feeding practices and long-term health outcomes. We quantified MMc using quantitative PCR (qPCR) in 89 female participants, ages 20-22, and analyzed these data using negative binomial regression. RESULTS In a multivariate model including all predictors, being breastfed substantially predicted decreased MMc (detection rate ratio = 0.15, p = 0.007), and there was a trend of decreasing MMc in participants who had experienced more pregnancies (detection rate ratio = 0.55, p = 0.057). DISCUSSION These results might be explained by breastfeeding having lasting impact on immune regulatory networks, thus reducing MMc persistence. MMc may also decrease in response to the introduction of fetal-origin microchimerism with pregnancies experienced in adulthood.
Collapse
Affiliation(s)
- Tiffany D. Pan
- Department of Anthropology, University of Washington, Seattle, Washington
- Center for Studies in Demography and Ecology, University of Washington, Seattle, Washington
| | - Sami B. Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nanette R. Lee
- USC-Office of Population Studies Foundation, Inc., Cebu City, Philippines
- Department of Anthropology, Sociology & History, University of San Carlos, Cebu City, Philippines
| | - Josephine L. Avila
- USC-Office of Population Studies Foundation, Inc., Cebu City, Philippines
- Department of Architecture, School of Architecture, Fine Arts and Design, University of San Carlos, Cebu City, Philippines
| | - J. Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle, Washington
| | - Dan T. A. Eisenberg
- Department of Anthropology, University of Washington, Seattle, Washington
- Center for Studies in Demography and Ecology, University of Washington, Seattle, Washington
- Department of Biology, University of Washington, Seattle, Washington
| |
Collapse
|